Download as pdf or txt
Download as pdf or txt
You are on page 1of 1103

Department of__ECE_______

Course Code-Course Name

Course file Index

S.No. Contents
1 Time table
2 Syllabus with course outcomes
Course plan including CO-PO Mapping and Distribution
3
of responsibilities in case of multiple instructors
4 List of Students
Any Continuous evaluation/Assessment like Quiz paper,
5
Project Names, Seminar topics etc (whatever applicable)
6 Mid Question paper with Cos
7 Solution of Mid Semester question paper
8 Award list of Mid Exam with Question wise marks
9 Analysis of Mid Exam
Analysis of continuous assessment marks and action
10
taken
11 Steps taken for advanced and slow learners
12 Best/Innovative practices followed in the course
Lab and Tutorial Assignments/Activity questions with
13
Cos.
14 Assessment methodology and Rubrics for assignment
Content beyond the curriculum like Industry Lecture,
15 Certifications, Startup activity, Case Studies, Hackathons,
Role Play etc
16 End Exam question paper
17 Solution of End Semester question paper
18 Course material
19 Handouts if any
20 Final Marks and Grade Sheet
21 Result Analysis including course attainment

Page 1 of 16
1.Timetable

Day I II III IV V VI VII VIII IX


8.30 -9.30 9.30-10.30 10.30-11.30 11.30-12.30 12.30-1.30 1.30-2.30 2.30-3.30 3.30-4.30 4.30-
5.30
Mon BMI
Tue BMI
Wed
Thu BMI
Fri BMI

Page 2 of 16
2.Syllabus

<23EC400PC713>BIOMEDICAL INSTRUMENTATION

Hours/Week Marks
Semester C
L T P CIE SEE Total
EVEN / ODD 3 1 - 4 40 60 100
Pre-requisite - COURSE TYPE:PE

COURSE OUTCOMES:
At the end of the course, the student will be able to:
1. Explain the principles and construction of artificial heart
2. Describe the tests to assess the hearing loss and development of wearable
devices for thesame.
3. Analyze and research on electrical stimulation and biofeedback techniques in
rehabilitationand physiotherapy.

UNIT I HEART LUNG MACHINE AND ARTIFICIAL HEART


Condition to be satisfied by the H/L System. Different types of Oxygenators, Pumps,
Pulsatile and Continuous Types, Monitoring Process, Shunting, The Indication for
Cardiac Transplant, Driving Mechanism, Blood Handling System, Functioning and
different types of Artificial Heart, Schematic for temporary bypass of left ventricle.

UNIT II CARDIAC ASSIST DEVICES


Assisted through Respiration, Right and left Ventricular Bypass Pump, Auxiliary
ventricle, Open Chest and Closed Chest type, Intra Aortic Balloon Pumping, Prosthetic
Cardiac valves, Principle ofExternal Counter pulsation techniques.

UNIT III ARTIFICIAL KIDNEY


Indication and Principle of Haemodialysis, Membrane, Dialysate, types of filter and
membranes, Different types of hemodialyzers, Monitoring Systems, Wearable Artificial
Kidney, Implanting Type.

UNIT IV RESPIRATORY AND HEARING AIDS


Page 3 of 16
Ventilator and its types-Intermittent positive pressure, Breathing Apparatus Operating
Sequence, Electronic IPPB unit with monitoring for all respiratory parameters. Types of
Deafness, Hearing Aids, SISI, masking techniques, wearable devices for hearing
correction.

UNIT V RECENT TRENDS


Transcutaneous electrical nerve stimulator, bio-feedback, Diagnostic and point-of-care
platforms.

TEXT BOOKS:
1. Gray E Wnek, Gray L Browlin – Encyclopedia of Biomaterials and Biomedical
Engineering–Marcel Dekker Inc New York 2004.
2. John. G . Webster – Bioinstrumentation - John Wiley & Sons (Asia) Pvt Ltd -
2004

REFERENCES:
1. Andreas.F. Von racum, “Hand book of bio material evaluation”, Mc-Millan
publishers, 1980.
2. Gray E Wnek, Gray L Browlin, “Encyclopedia of Biomaterials and Biomedical
Engineering”Marcel Dekker Inc New York 2004.

STUDIO WORK / LABORATORY EXPERIMENTS / HANDS ON ACTIVITY:


1. Design a PCB layout for any bio amplifier using suitable software tool.
2. Design of pre amplifiers to acquire bio signals along with impedance
matching circuit using suitable IC’s 2.
3. Design of ECG Amplifiers with appropriate filter to remove power line and
other artifacts. 3. Design of EMG amplifier
4. Design and study the characteristics of optical Isolation amplifier

Page 4 of 16
COURSEPLAN
For

BIOMEDICAL INSTRUMENTATION (23EC400PC713)

Course Coordinator: Dr.K RAJ KUMAR,Dr.S.MALATHY

L-T-P : 3-1-0

Credits :4

School : School of Engineering

Department : Department of ECE

Course Level : Phd

Schoolof Engineering

SR University,
Warangal

Page 5 of 16
COURSE CONTEXT

ECE
DEPARTMENT
SCHOOL Engineering

DATE THIS
DEGREE Ph.D COURSEWILL BE
EFFECTIVEFROM Aug–Jan 2024

COURSE BRIEF

BIOMEDICAL
COURSE TITLE INSTRUMENTATIO PRE-REQUISITES -
N
COURSE CODE 23EC400PC713 TOTALCREDITS 4

COURSE TYPE Professional Core L-T-PFORMAT 3-1-0

Course Articulation Matrix

PO/PSO PO PO PO PO PO PO PO PO PO PO PO PO PSO PSO

1 2 3 4 5 6 7 8 9 10 11 12 1 2
CO

CO1 3 2 3 1 2 - 2 - 3 - - 2 - -

CO2 3 3 3 1 2 - - 2 - 1 - - - -

CO3 2 2 3 2 3 - - - - - 1 - - 2

CO4 2 2 3 1 2 - 1 - - - - - 1 -

Mapping
Target Level

Page 6 of 16
Lecture Wise Plan

No. Content Planned

1 HEART LUNG MACHINE AND ARTIFICIAL HEART: Condition to be satisfied by


the H/L System. Different types of Oxygenators(50)
2 Pumps, Pulsatile and Continuous Types (50)
3 Monitoring Process, Shunting, The Indication for Cardiac Transplant (50)
4 Driving Mechanism, Blood Handling System (50)
5 Functioning and different types of Artificial Heart (50)
6 Schematic for temporary bypass of left ventricle (50)

UNIT II CARDIAC ASSIST DEVICES: Assisted through Respiration, Right and left
7 Ventricular Bypass Pump

(50)
8
Auxiliary ventricle, Open Chest and Closed Chest type(50)
9
Intra Aortic Balloon Pumping, Prosthetic Cardiac valves (50)
10
Principle ofExternal Counter pulsation techniques (50)
11
UNIT III ARTIFICIAL KIDNEY Indication and Principle of Haemodialysis (50)
12
Membrane, Dialysate, types of filter and membranes(50)
Different types of hemodialyzers (50)
13

14 Monitoring Systems (50)


15 Wearable Artificial Kidney(50)
16 Implanting Type (50)

17 UNIT IV RESPIRATORY AND HEARING AIDS Ventilator and its types-Intermittent


positive pressure (50)
18 Breathing Apparatus Operating Sequence(50)
19 Electronic IPPB unit with monitoring for all respiratory parameters (50)
20
Types of Deafness, Hearing Aids (50)

Page 7 of 16
SISI, masking techniques, wearable devices for hearing correction(50)
21

22 UNIT V RECENT TRENDS (50)


23 Transcutaneous electrical nerve stimulator (50)
24 bio-feedback (50)
25 Diagnostic and point-of-care platforms (50)

Evaluation Components

ComponentsofCourseEvaluation Percentage
MidTermExamination 20

EndTermExamination 40

Assignment 20

Certification --20

Advanced Research Topics:


Biomaterials and Regenerative Engineering

The Biomaterials Group aims to develop self-assembled, bio-integrated electronic,


synthetic, and nano scale materials for biomedical applications. Research and teaching
areas include self-assembled nano structures, bioelectronics, synthetic biology, bio-
membrane dynamics, nanotechnology, controlled drug release, cell-material interactions,
immuno engineering and computational analysis of molecular dynamics.

The Regenerative Engineering Group aims to understand the innate mechanisms of cell
protection and regeneration and establish engineering strategies for controlling protective
and regenerative processes with the ultimate goal of restoring tissue and organ function.
Research and teaching areas include genomic analysis, transport biology and engineering,
cardiovascular protective engineering, and regenerative engineering.

Page 8 of 16
4, STUDENT LIST

Department of__ECE

SCHOOL OF ENGINEERING - Ph.d

(Part Time-ECE)-STUDENT LIST

SUBJECT:BIOMEDICAL INSTRUMENTATION

S No Reg No Name

1 2305C40040 PARVATHAM SATHISH

Page 9 of 16
6. MID QUESTION PAPER

Ph.D Course work Internal Examination, December - 2023

BIOMEDICAL INSTRUMENTATION

(ECE)

Date: 18-12-2023 Max. Marks: 20:


Time: 90 min

Note: Answer all the following. 4x5

1. Classify the oxigenators and explain them. (CO1)

2. With a neat diagram illustrate the temporary bypass of left ventricle (CO2)

3. Compare open and close chest types (CO1)


4. Briefly explain the principle of external counter pulsation techniques? (CO3)

Page 10 of 16
7. SOLUTIONS-MID

1. Classify the oxygenators and explain them.

Oxygenators are medical devices used to add oxygen to the blood and remove carbon dioxide during
surgical procedures. There are two main types of oxygenators bubble oxygenators and membrane
oxygenators.

1. Bubble Oxygenators:

 Gas Exchange Mechanism:

 Oxygenation is achieved through the introduction of oxygen-enriched gas into the blood
as bubbles.

 Carbon dioxide is expelled from the blood as it comes into contact with the oxygenated
gas bubbles.

 Components:

 Reservoir: Holds the blood before it enters the oxygenation chamber.

 Gas Exchange Chamber: Facilitates the interaction between the oxygenated gas bubbles
and the blood.

 Bubble Generator: Creates bubbles for efficient gas exchange.

 Advantages:

 Simplicity: Bubble oxygenators have a straightforward design, making them easy to use.

 Cost-effectiveness: They are often more economical compared to membrane


oxygenators.

2. Membrane Oxygenators:

 Gas Exchange Mechanism:

 Oxygen and carbon dioxide traverse a semipermeable membrane to achieve gas


exchange.

 The membrane allows selective diffusion, permitting oxygen to enter the blood while
removing carbon dioxide.

 Components:

 Blood Compartment: Where the patient's blood flows, separated from the gas
compartment by the semipermeable membrane.

 Gas Compartment: Contains oxygen-enriched gas that interfaces with the blood through
the membrane.

Page 11 of 16
 Advantages:

 Efficiency: Membrane oxygenators are known for providing efficient and reliable gas
exchange.

 Reduced Trauma: They are associated with lower blood trauma compared to bubble
oxygenators.

 Versatility: Membrane oxygenators are suitable for a wide range of surgical procedures.

2. With neat diagram illustrate the temporary bypass of left ventricle.

One common method for temporarily bypassing the left ventricle is through a medical procedure known
as Extracorporeal Membrane Oxygenation (ECMO). ECMO is a life support technique that temporarily takes
over the function of the heart and lungs.

1. Cannulation:
 Two cannulas are inserted into major blood vessels. One is placed in the ascending aorta
to withdraw oxygenated blood, and the other is placed in the left atrium or left ventricle.
2. Blood Flow:
 The cannula in the ascending aorta directs blood to an external mechanical pump.
3. Mechanical Pump:
 The mechanical pump acts as an artificial heart, receiving and pumping the oxygenated
blood under pressure.
4. Membrane Oxygenator:
 The blood is then directed through a membrane oxygenator.
 Within the oxygenator, the patient's blood comes into contact with a semipermeable
membrane, allowing for the exchange of oxygen and carbon dioxide with a gas mixture,
effectively mimicking lung function.
 A post-membrane pressure monitor ensures that the blood pressure is appropriate as it
exits the oxygenator.
5. Heat Exchanger:
 The blood leaving the oxygenator passes through a heat exchanger.
 The heat exchanger regulates the blood temperature, ensuring that it matches the
normal body temperature before being returned to the patient.

Page 12 of 16
6. Warmed Water Input:
 The heat exchanger is supplied with warmed water to assist in maintaining the desired
temperature of the blood.
7. Oxygen (O2) Blender:
 An oxygen blender is employed to precisely control the amount of oxygen delivered to
the blood, optimizing oxygenation.
8. Venous Reservoir:
 Deoxygenated blood returning from the patient collects in a venous reservoir.
 The reservoir helps maintain a consistent blood flow to the oxygenator and prevents air
from entering the circulatory system.

9. Return to Circulation:
 The oxygenated and treated blood is finally returned to the patient's circulatory system
through the cannula in the left atrium or left ventricle

3. Compare open and close chest types.

Aspect Open-Chest Closed-Chest


Definition Involves making an incision in the chest, Procedures are performed without a large
often through the sternum (median incision; may involve smaller incisions or
sternotomy). alternative access points.
Use Cases Commonly used in various cardiac surgeries, Applied in minimally invasive coronary artery
including CABG, valve replacement, and bypass grafting (MICS CABG), minimally
congenital heart defect repairs. invasive valve surgery, and transcatheter
interventions.
Advantages 1. Direct access to the heart and its 1. Minimally invasive with smaller incisions.
structures. 2. Quicker recovery time.
2. Well-suited for complex cardiac surgeries. 3. Reduced postoperative complications.
Disadvantages 1. Invasive procedure with a significant 1. Limited visibility and access to the heart.
chest incision. 2. Applicability depends on the specific
2. Longer recovery time. procedure.
3. Higher risk, especially in certain patient
populations.

4. Briefly explain the principle of external counter pulsation.

External counter pulsation is based on the principle of enhancing coronary perfusion by applying external
pressure to the lower extremities in synchronization with the cardiac cycle. It involves the use of inflatable
cuffs or sleeves wrapped around the patient's legs, similar to blood pressure cuffs. The procedure typically
includes the following steps:
1. Cuff Inflation during Diastole:
 As the heart relaxes during diastole (the phase when it fills with blood), the cuffs inflate
rapidly. This increases pressure in the lower extremities.
2. Cuff Deflation during Systole:
 The cuffs deflate just before the onset of systole (the contraction phase of the heart). This
rapid deflation reduces the pressure in the cuffs, lowering resistance in the lower limbs.
3. Effect on Blood Flow:
 By inflating the cuffs during diastole, ECP increases blood flow to the coronary arteries,
enhancing perfusion to the heart muscle.

Page 13 of 16
 The rapid deflation just before systole reduces resistance in the peripheral vasculature,
easing the workload on the heart during its contraction phase.
4. Improving Oxygen Supply:
 The increased blood flow and reduced afterload (pressure the heart must work against)
contribute to better oxygen supply to the heart muscle.
5. Treatment Sessions:
 A typical course of ECP involves multiple treatment sessions over several weeks, with
each session lasting about an hour.
Clinical Applications:
 External counter pulsation is often used as a non-invasive treatment for angina (chest pain) and
can be considered in certain cases of heart failure.
 It is sometimes employed when other interventions, such as medications or invasive procedures,
are not feasible or have limitations.

Page 14 of 16
8. MID AWARD SHEET

S.NO Roll number Mid exam(20M)


Q1 Q2 Q3 Q4 Total
1 2205C40040 4 4 5 5 18

Page 15 of 16
Page 16 of 16
Proposed scheme of evaluation for Phd Scholars

Internal Exam- Assignment/Publication Certification(20 End Exam-40 Total-100 Marks


20 Marks 20 Marks hours /Publication)- Marks
20 Marks

Note: This is only proposed, guides can follow their own scheme of evaluation with proper intimation to
PhD coordinator, Head and ECE Exam cell coordinator

Note: The same publication cannot be claimed at any other stage


COURSERA SUBJECTS
PARVATHAM SATHISH,
Research scholar,ECE,SRU.
2305C40040@SRU.EDU.IN.
Under the supervision of
Sri.DR.K RAJ KUMAR ,
Assoc.Prof,ECE,SR University.
Sri.DR.s malathy ,
Asst.Prof,ECE,SR University

1.programming in python
DATE : 16 JAN 2024
GRADE: 92%
LEVEL: BEGINNER
Total Hours: 44.

MODULE 1: GET STARTED WITH THE PYTHON


Get started with the Python programming language and associated foundational
concepts.
MODULE 2 - BASIC PROGRAMMING IN PYTHON

Learn basic Python syntax, to use control flow and loops and work with functions and
data structures. You will also learn how to recognise possible errors, their causes
and how to handle them.

MODULE 3 - PROGRAMMING PARADIGMS

Learn about the paradigms of procedural programming and the associated logical
concepts. You'll explore functional and object-oriented programming, and get an
introduction to algorithms

MODULE 4 - MODULES,PACKAGES,TOOLS AND LIBRARIES


Supercharge your coding environment with popular modules libraries and tools for
Python. You'll also learn about the different types of testing and how to write a test.

MODULE-5: END OF COURSE GRADED ASSIGNMENT

Practice and reflect on the skills you learned in this course.

About this Course

In this course, you will be introduced to foundational programming skills with basic
Python Syntax. You’ll learn how to use code to solve problems. You’ll dive deep into
the Python ecosystem and learn popular modules, libraries and tools for Python.
You’ll also get hands-on with objects, classes and methods in Python, and utilize
variables, data types, control flow and loops, functions and data structures. You’ll learn
how to recognize and handle errors and you’ll write unit tests for your Python code
and practice test-driven development. By the end of this course, you will be able to: •
Prepare your computer system for Python programming • Show understanding of
Python syntax and how to control the flow of code • Demonstrate knowledge of how
to handle errors and exceptions • Explain object-oriented programming and the major
concepts associated with it • Explain the importance of testing in Python, and when to
apply particular methods This is a beginner course for learners who would like to
prepare themselves for a career in back-end development or database engineering.
To succeed in this course, you do not need prior web development experience, only
basic internet navigation skills and an eagerness to get started with coding.
2. FUNDAMENTALS OF DIGITAL IMAGE AND VIDEO PROCESSING

DATE : 16-JAN-2024
GRADE: 92%
LEVEL: BEGINNER
Total Hours: 35.

In this class you will learn the basic principles and tools used to process images and
videos, and how to apply them in solving practical problems of commercial and
scientific interests.
Digital images and videos are everywhere these days – in thousands of scientific (e.g.,
astronomical, bio-medical), consumer, industrial, and artistic applications. Moreover
they come in a wide range of the electromagnetic spectrum - from visible light and
infrared to gamma rays and beyond. The ability to process image and video signals is
therefore an incredibly important skill to master for engineering/science students,
software developers, and practicing scientists. Digital image and video processing
continues to enable the multimedia technology revolution we are experiencing today.
Some important examples of image and video processing include the removal of
degradations images suffer during acquisition (e.g., removing blur from a picture of a
fast moving car), and the compression and transmission of images and videos (if you
watch videos online, or share photos via a social media website, you use this
everyday!), for economical storage and efficient transmission. This course will cover
the fundamentals of image and video processing. We will provide a mathematical
framework to describe and analyze images and videos as two- and three-dimensional
signals in the spatial, spatio-temporal, and frequency domains. In this class not only
will you learn the theory behind fundamental processing tasks including image/video
enhancement, recovery, and compression - but you will also learn how to perform
these key processing tasks in practice using state-of-the-art techniques and tools. We
will introduce and use a wide variety of such tools – from optimization toolboxes to
statistical techniques. Emphasis on the special role sparsity plays in modern image
and video processing will also be given. In all cases, example images and videos
pertaining to specific application domains will be utilized.

3.RESEARCH METHODOLOGIES
DATE : 18-NOV-2023
GRADE: 98.80%
LEVEL: BEGINNER
Total Hours: 19.
This course focuses on research methodologies. In this vein, the focus will be
placed on qualitative and quantitative research methodologies, sampling approaches,
and primary and secondary data collection. The course begins with a discussion on
qualitative research approaches, looking at focus groups, personal interviews,
ethnography, case studies and action research. We will also discuss quantitative
research methods with a focus on experimental research design and survey
methodology. There will be an exploration of the sampling design process and different
sampling approaches, including probability and non-probability sampling as well as
sample size and non-response issues. We will look at the nature and scope of primary
and secondary data, and the importance of measurement. We will look at the role of
the Internet in market research as well as non-comparative scaling techniques. The
course ends with a discussion on different data collection approaches, with a focus on
observation, content analysis, narrative research, phenomenology, and the collection
of data using ethnography.

WEEK 1.
This week begins with a discussion on qualitative research approaches, looking at
focus groups, personal interviews, ethnography, case study and action research. The
week ends with a discussion of quantitative research methods with a focus on
experimental research design and survey methodology.
WEEK.2.
The week begins with a discussion on the sampling design process and continues with
different sampling approaches, including probability and non-probability sampling. The
week ends with a discussion on sample size and non-response issues.
WEEK.3.
The week begins with a discussion of the nature and scope of secondary data and
continues with a discussion of primary data and the importance of measurement. The
week ends with the role of the Internet in market research and a discussion about non-
comparative scaling techniques.

WEEK4.
The week starts a discussion on different data collection approaches with a focus on
observation, case study and content analysis. The week continues with a focus on
narrative research, phenomenology and the action research project. The week ends
with a discussion of collecting data using ethnography.
4.UNDERSTANDING RESEARCH METHODS
DATE : 21-NOV-2023
GRADE: 98%
LEVEL: BEGINNER
Total Hours: 19.

MODULE 1:
PURPOSE: To focus upon formulating a research question. TASK: Please compose a
brief question pertaining to your proposed research - whatever the subject - and post
it under the 'My Project' tab in the peer review exercise below. Be brief insofar as it
would fit on one metaphorical or literal 'Post-It' note. Also watch our interview videos
below. RESPONSE: Having reflected on the videos, review other students' questions.
Feeding back is an important part of the research process, so please spend some time
providing considered feedback for three or more colleagues here. OUTCOME: You will
have given considered thought to composing a research question and provided
feedback to others on theirs.
MODULE 2:
PURPOSE: To familiarise yourself with the nature and benefits of conducting a
literature review. TASK: Please read the following three pieces on literature review, in
conjunction with the interviews below. Then post your thoughts on one of the readings
to the 'My Project' tab of the peer review (200 words). RESPONSE: Provide feedback
on three or more submissions by your fellow students. OUTCOME: Upon the
completion of the full cycle of this e-tivity, you will be able to distinguish the qualities
of a literature review and begin to reflect on the value of a literature review to your own
project.
MODULE 3:
PURPOSE: To be aware of the planning and management skills that are required in
undertaking critical thinking for your research. TASK: Please watch the videos below,
and also read the chapter provided. In conjunction with your reflections on the learning
material provided and also research planning and management in general, please post
your thoughts on the chapter, in no more than 400 words, under the 'My Project' tab.
RESPONSE: Provide feedback on three or more submissions by your fellow students.
OUTCOME: Upon the completion of the full cycle of this e-tivity, you will have reflected
on the skills required to enhance your research.
MODULE 4:
PURPOSE: To reflect upon the value of a good question, and provide an outline
research proposal. TASK: In conjunction with our videos below, please go back to your
research question proposed during E-tivity 1 and consider again whether you are still
happy with it. Is the question one you are still interested in and one you think worthy
of devoting your time and energy to? If so, after reflecting on your question, please
compose an outline plan on how you would follow on from establishing your research
question. Please submit your proposal, up to 800 words. RESPONSE: Provide
feedback on three or more proposals submitted by your fellow students. OUTCOME:
Upon the completion of the full cycle of this e-tivity, you will have formulated, in draft
form, an overarching research question, and a plan of action to complete the research.
In doing this, you will have honed your writing, research and analytical skills.

Program Name: PhD -ECE Exam Type: (Regular)
Department/School of ECE(SOE)
Course Code 23EC400PC713 Course Title BIOMEDICAL INSTRUMENTATION
Year/Sem I-I Faculty Name Dr K RAJ KUMAR,Dr.S.MALATHY
Date of Exam 18-01-2024 Time 10.00 AM to 12 PM
Duration 2 Hrs Max. Marks 40

Q. No Questions Marks CO

1. Write the required condition to be satisfied by the heart and lung 8 CO1
system and explain heart & lung?
2. Explain the principle of external counter pulsation techniques? 8 CO2

3. Classify the types of hemodialyzers and explain them? 8 CO3

4. Differentiate the intermittent positive pressure of ventilator and 8 CO4


explain?

5. Explain the operation of transcutaneous electrical nerve 8 CO5


stimulator?

1. write the requried condition to be satisfied by the heart and lung system and explain heart
& lung?
Ans:-
It appears that there might be a slight confusion in your question. However, I'll provide
information on the conditions that need to be satisfied by the heart and lung systems separately,
along with brief explanations of each.

### Heart:-

**Required Conditions:-**
1. **Effective Pumping:-** The heart must effectively pump blood throughout the body to
provide oxygen and nutrients to various tissues and organs.

2. **Rhythmic Contractions:-** The heart should exhibit coordinated and rhythmic


contractions to maintain a consistent blood flow and adequate cardiac output.

3. **Valve Functionality:-** The heart valves must open and close appropriately to ensure
unidirectional blood flow and prevent backflow.

4. **Electrical Stability:-** The heart's electrical conduction system should function properly
to regulate the heart's rhythm and coordinate contractions.

## Lungs:-

**Required Conditions:-**
1. **Gas Exchange:-** The lungs must facilitate the exchange of oxygen and carbon dioxide
during respiration. Oxygen is taken in from the air, and carbon dioxide is expelled from the
body.

2. **Adequate Ventilation:-** There should be proper ventilation, involving the intake of


fresh air and the removal of stale air, to maintain the necessary oxygen and carbon dioxide
levels in the blood.

3. **Compliance of Lung Tissues:-** The lung tissues need to be compliant, allowing for
expansion during inhalation and contraction during exhalation.

4. **Surfactant Production:-** The production of pulmonary surfactant is essential for


reducing surface tension in the alveoli, preventing their collapse and facilitating efficient gas
exchange.

### Heart and Lung System (Cardiopulmonary System):-

The heart and lungs work collaboratively to ensure proper oxygenation of the blood and
circulation throughout the body.

**Interdependence:-**
 The heart pumps oxygen-depleted blood to the lungs via the pulmonary circulation.
 In the lungs, carbon dioxide is exchanged for oxygen through the process of respiration.
 Oxygen-rich blood is then pumped by the heart to the rest of the body through the
systemic circulation.

**Coordination:-**
 The heart rate and rhythm are influenced by respiratory needs, with increased heart rate
during exercise to meet the elevated oxygen demands.

**Homeostasis:-**
 The cardiopulmonary system plays a crucial role in maintaining the body's overall
homeostasis, ensuring a balance of oxygen and carbon dioxide levels in the blood.
In summary, the heart and lungs are integral components of the cardiopulmonary system,
working in concert to maintain essential physiological functions such as oxygenation,
circulation, and homeostasis in the human body. The proper functioning of both systems is
crucial for overall health and well-being.

2. Explain the principle of external counter pulsation techniques?


Ans:- External Counterpulsation (ECP) is a non-invasive medical treatment designed to
improve blood flow to the heart and alleviate symptoms of certain cardiovascular conditions.
The technique involves the use of inflatable cuffs or sleeves, which are wrapped around the
patient's legs, lower abdomen, and buttocks. Here's an explanation of the principle of External
Counterpulsation:

1. **Mechanism of Action:-**
- **Inflation and Deflation of Cuffs:-** The cuffs inflate and deflate sequentially in
synchronization with the cardiac cycle. Typically, inflation occurs during diastole (when the
heart is at rest and filling with blood), and deflation occurs just before the next systole (when
the heart contracts to pump blood).

2. **Enhancement of Blood Flow:-**


- **Diastolic Augmentation:-** During diastole, the cuffs inflate, increasing the pressure in
the lower extremities. This augments blood flow to the coronary arteries supplying the heart
muscle.
- **Systolic Reduction:-** The cuffs deflate just before the heart contracts (systole), creating
a vacuum effect that reduces resistance to blood flow. This reduction in resistance facilitates
the return of blood to the heart.

3. **Improvement of Coronary Perfusion:-**


- **Increased Oxygen Supply:-** By enhancing blood flow during diastole, ECP increases
oxygen-rich blood supply to the coronary arteries, improving oxygenation of the heart muscle.
4. **Reduction of Cardiac Workload:-**
- **Afterload Reduction:-** The deflation of cuffs just before systole reduces the workload
on the heart by lowering systemic vascular resistance. This results in decreased afterload,
making it easier for the heart to pump blood.

5. **Induction of Collateral Circulation:**


- **Stimulation of Angiogenesis:-** ECP has been suggested to promote the formation of
collateral blood vessels (angiogenesis) in the heart. These collateral vessels can provide
alternative pathways for blood flow, bypassing blocked or narrowed arteries.

6. **Clinical Applications:-**
- **Angina Pectoris:-** ECP is often used as a treatment for chronic stable angina pectoris
when other interventions are not suitable.
- **Heart Failure:-** Some studies suggest potential benefits for patients with heart failure
by improving symptoms and exercise tolerance.
- **Peripheral Artery Disease:-** ECP may also be considered for individuals with
peripheral artery disease (PAD).

It's important to note that while ECP has shown benefits for certain patients, its effectiveness
can vary, and it may not be suitable for everyone. Always consult with healthcare
professionals to determine the most appropriate treatment options based on individual health
conditions.

3. Classify the types of hemodialyzers and explain them?


Ans:- Hemodialysis is a medical procedure used to remove waste products and excess fluid
from the blood when the kidneys are unable to perform these functions adequately.
Hemodialyzers, also known as artificial kidneys or dialysis filters, play a crucial role in this
process. There are different types of hemodialyzers, classified based on their membrane
composition and design. Here are the main types:

1. **Cellulosic Membrane Hemodialyzers:**


- **Description:-** Cellulosic membranes are made from natural cellulose material, often
derived from wood pulp.
- **Characteristics:-** These membranes have a high biocompatibility, meaning they are
well-tolerated by the patient's body. They have been used traditionally in hemodialysis.
- **Advantages:-** Good removal of middle-sized molecules, cost-effective.
- **Disadvantages:-** Potential for the activation of complement and other immune
reactions.

2. **Synthetic Membrane Hemodialyzers:-**


- **Description:-** Synthetic membranes are manufactured using synthetic polymers, such
as polysulfone, polyethersulfone, or polyacrylonitrile.
- **Characteristics:-** These membranes offer better biocompatibility than some older
cellulose membranes and are known for their durability.
- **Advantages:-** Reduced risk of allergic reactions, efficient removal of small and middle-
sized molecules.
- **Disadvantages:** Can be more expensive than cellulosic membranes.

3. **High-Flux Hemodialyzers:-**
- **Description:-** High-flux hemodialyzers have membranes with larger pores, allowing for
enhanced removal of larger molecules, including beta-2 microglobulin.
- **Characteristics:-** Designed to achieve better clearance of middle-sized and larger toxins
compared to low-flux membranes.
- **Advantages:-** Improved removal of uremic toxins, especially in patients with chronic
kidney disease.
- **Disadvantages:-** May lead to higher albumin loss and increased cost.

4. **Low-Flux Hemodialyzers:**
- **Description:** Low-flux hemodialyzers have smaller pores in the membrane, allowing
for the removal of small to medium-sized molecules.
- **Characteristics:** Historically, low-flux membranes were commonly used in
hemodialysis.
- **Advantages:** Cost-effective, suitable for certain patients.
- **Disadvantages:** Less efficient in removing larger toxins, may be less effective in some
clinical situations.

5. **Highly Permeable Membrane Hemodialyzers:**


- **Description:**
These hemodialyzers have membranes designed to have higher permeability, allowing for
enhanced clearance of both small and large molecules.
- **Characteristics:** Aimed at achieving improved overall dialysis efficiency.
- **Advantages:** Enhanced clearance of a wide range of toxins.
- **Disadvantages:** Potential for increased albumin loss and higher cost.

The choice of hemodialyzer depends on various factors, including the patient's clinical
condition, the type and size of molecules to be removed, and cost considerations.
Nephrologists carefully assess individual patient needs to select the most appropriate
hemodialyzer for each case.

4. Differentiate the intermittent positive pressure of ventilator and explain?


Ans:- The terms "intermittent positive pressure ventilation" (IPPV) and "ventilator" are often
used in the context of respiratory support, particularly in cases of respiratory failure. Let's
differentiate between intermittent positive pressure ventilation and a ventilator, and then
provide an explanation of intermittent positive pressure ventilation.

1. **Ventilator:**
- **Definition:** A ventilator, also known as a mechanical ventilator or a respirator, is a
medical device designed to provide mechanical ventilation by delivering breaths to a patient
who is unable to breathe adequately on their own. It assists with the exchange of oxygen and
carbon dioxide in the lungs.
- **Function:** Ventilators can deliver breaths in different modes, including intermittent
positive pressure ventilation (IPPV), continuous positive airway pressure (CPAP), and others.
- **Use:** Ventilators are commonly used in critical care settings, such as intensive care
units (ICUs), during surgeries, or for patients with severe respiratory conditions.

2. **Intermittent Positive Pressure Ventilation (IPPV):**


- **Definition:** IPPV is a mode of mechanical ventilation where positive pressure is
applied to the airways during the inspiratory phase, forcing air into the lungs.
- **Characteristics:**
- **Intermittent:** IPPV involves periodic delivery of positive pressure breaths.
- **Positive Pressure:** Positive pressure is applied during inspiration to inflate the lungs.
- **Modes:**
- **Controlled Mode:** In this mode, the ventilator controls both the timing and volume
of breaths delivered to the patient.
- **Assist-Control Mode:** Patients can trigger additional breaths, but the ventilator
controls the volume or pressure delivered.
- **Synchronized Intermittent Mandatory Ventilation (SIMV):** Combines controlled
and spontaneous breaths, allowing patients to breathe between mechanical breaths.

- **Indications:** IPPV is used in various clinical situations, such as acute respiratory


failure, respiratory distress syndrome, and during surgeries where the patient needs assistance
or temporary control of ventilation.

- **Benefits:**
 Provides controlled and consistent ventilation.
 Assists patients who are unable to initiate or maintain adequate breathing on their own.
 Allows for adjustments in tidal volume, respiratory rate, and inspiratory time.

- **Considerations:**
- Careful monitoring is required to prevent complications, such as barotrauma (injury due
to excessive pressure) and volutrauma (injury due to excessive volume).
In summary, a ventilator is a broad term encompassing various modes of mechanical
ventilation, including intermittent positive pressure ventilation (IPPV). IPPV involves the
periodic delivery of positive pressure breaths to assist or control a patient's ventilation. The
specific mode and settings are chosen based on the patient's condition and the therapeutic
goals of respiratory support.

5. Explain the operation of transcutaneous electrical nerve stimulator?


Ans: A Transcutaneous Electrical Nerve Stimulator (TENS) is a medical device that delivers
mild electrical impulses through the skin to specific nerves, primarily for the purpose of
managing pain. TENS units are commonly used as a non-invasive and drug-free method for
pain relief. Here's an explanation of the operation of a TENS device:

1. **Electrode Placement:**
 TENS units consist of electrode pads that are attached to the skin in the area where
pain relief is desired.
 Electrodes are typically placed around or near the pain site, forming a circuit through
which the electrical impulses will travel.

2. **Electrical Impulses:**
 The TENS unit generates low-voltage electrical impulses. These impulses are delivered
through the electrodes to the underlying nerves.
 The electrical stimulation is designed to interfere with or block pain signals, providing
relief to the patient.

3. **Frequency and Intensity Settings:**


 TENS devices offer adjustable settings for both frequency and intensity.
- **Frequency:**
 Refers to the number of electrical pulses delivered per second, measured in Hertz (Hz).
Different frequencies may be used for different types of pain.
- **Intensity:**
Refers to the strength of the electrical impulses. Patients can adjust the intensity level based
on their comfort and the desired therapeutic effect.

4. **Gate Control Theory:**


 The effectiveness of TENS is often explained by the Gate Control Theory of pain.
According to this theory, the electrical impulses generated by the TENS unit stimulate
non-painful nerve fibers, which can "close the gate" to the transmission of pain signals
to the brain. Essentially, the stimulation can reduce the perception of pain.

5. **Endorphin Release:**
 TENS may also stimulate the release of endorphins, which are the body's natural
painkillers. Endorphins can help alleviate pain and induce a sense of well-being.

6. **Pulse Patterns:**
 TENS units can offer different pulse patterns, such as continuous, burst, or modulated,
providing flexibility in the type of stimulation delivered.

7. **Duration of Use:**
 TENS therapy is typically applied for short periods, ranging from 15 minutes to several
hours, depending on the patient's needs and the specific recommendations of their
healthcare provider.

It's important to note that while TENS can be effective for certain types of pain, it may not be
suitable for all conditions. It is essential to use TENS under the guidance of a healthcare
professional, who can determine the appropriate settings and ensure the device's safe and
effective use. TENS should not be used on certain areas, such as over the eyes or on the chest
if the person has a pacemaker.
.Additioral
Assignmelrls

oPersonal
Slon l,carncrs I attenlion and
counsclliog

Activities For Slow Learners

.Provis')n to
Adrrnccd I
rough MoU's
I Learners
$ith rcputed
iIlstitutions

Activities For Advanced Learners

3
Encyclopedia of
BIOMATERIALS AND
BIOMEDICAL ENGINEERING
This page intentionally left blank
Encyclopedia of
BIOMATERIALS AND
BIOMEDICAL ENGINEERING
S E C O N D E D I T I O N , V O L U M E 1

Edited by

GARY E. WNEK
Case Western Reserve University
Cincinnati, Ohio, USA

GARY L. BOWLIN
Virginia Commonwealth University
Richmond, Virginia, USA
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
© 2008 by Taylor & Francis Group, LLC
CRC Press is an imprint of Taylor & Francis Group, an Informa business

No claim to original U.S. Government works


Version Date: 20130426

International Standard Book Number-13: 978-1-4200-7803-9 (eBook - PDF)

This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been made to publish reliable data and
information, but the author and publisher cannot assume responsibility for the validity of all materials or the consequences of their use. The authors and
publishers have attempted to trace the copyright holders of all material reproduced in this publication and apologize to copyright holders if permission
to publish in this form has not been obtained. If any copyright material has not been acknowledged please write and let us know so we may rectify in any
future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,
mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in any information storage or
retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://www.copyright.com/) or contact
the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides
licenses and registration for a variety of users. For organizations that have been granted a photocopy license by the CCC, a separate system of payment
has been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identification and explanation
without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
and the CRC Press Web site at
http://www.crcpress.com
Editors
Gary E. Wnek
Case Western Reserve University
Cleveland, Ohio, U.S.A.

Gary L. Bowlin
Virginia Commonwealth University
Richmond, Virginia, U.S.A.

Editorial Advisory Board

James Anderson Arnauld Nicogossian


Case Western Reserve University, Cleveland, Ohio, George Mason University, Fairfax, Virginia,
U.S.A. U.S.A.

Deon Bezuidenhout Nikolaos Peppas


University of Cape Town, Cape Town, Purdue University, West Lafayette, Indiana,
South Africa U.S.A.

Rena Bizios Christine E. Schmidt


University of Texas at San Antonio, San Antonio, University of Texas, Austin, Texas, U.S.A.
Texas, U.S.A.
Jerome S. Schultz
Larry L. Hench University of California, Riverside, Riverside,
Imperial College London, London, United Kingdom California, U.S.A.

V. Prasad Shastri
Allan S. Hoffman
Vanderbilt University, Nashville, Tennessee, U.S.A.
University of Washington, Seattle, Washington,
U.S.A. Debra Trantolo
Cambridge Scientific, Inc., Cambridge, Massa-
Yoshito Ikada chusetts, U.S.A.
Suzuka University of Medical Science, Suzuka,
Mie, Japan Rachel L. Williams
University of Liverpool, Liverpool, United Kingdom
Robert Langer
Massachusetts Institute of Technology, Cambridge, Nicholas Ziats
Massachusetts, U.S.A. Case Western Reserve University, Cleveland, Ohio,
U.S.A.
Gerald E. Miller
Virginia Commonwealth University, Richmond,
Virginia, U.S.A.
This page intentionally left blank
List of Contributors

Soumyadipta Acharya / The University of Akron, Akron, Ohio, U.S.A.


Clayton Adam / Queensland University of Technology, Brisbane, Queensland, Australia
Babita Agrawal / University of Alberta, Edmonton, Alberta, Canada
Carlos A. Aguilar / University of Texas at Austin, Austin, Texas, U.S.A.
Juana Maria Alfaro / Cordis de Mexico, Juarez, Mexico
Heather Ambrose / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Catherine G. Ambrose / University of Texas–Houston School of Medicine, Houston, Texas, U.S.A.
Farid Amirouche / University of Illinois at Chicago, Chicago, Illinois, U.S.A.
Javier de Ana / University of Michigan, Ann Arbor, Michigan, U.S.A.
Luke Aram / DePuy Orthopedics/Johnson & Johnson Company
James H. Arps / Southwest Research Institute, San Antonio, Texas, U.S.A.
James A. Arrowood / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Tetsuo Asakura / Tokyo University of Agriculture and Technology, Koganei, Tokyo, Japan
Anthony Atala / Wake Forest Institute for Regenerative Medicine, Winston-Salem,
North Carolina, U.S.A.
Kyriacos A. Athanasiou / Rice University, Houston, Texas, U.S.A.
Jay Audett / ALZA Corporation, Mountain View, California, U.S.A.
Franc- ois A. Auger / LOEX, Hôpital du Saint-Sacrement du CHA, and Laval University, Quebec City,
Quebec, Canada
Konstantinos Avgoustakis / University of Patras, Patras, Rio, Greece
Stephen F. Badylak / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Richard Baker / Royal Children’s Hospital, Melbourne, Victoria, Australia
Deepak Banerjee / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Peter B. Barker / Johns Hopkins University School of Medicine, Baltimore, Maryland, U.S.A.
Clive M. Baumgarten / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Roger Bayston / University Hospital, Nottingham, United Kingdom
Matthew J. Beckman / Virginia Commonwealth University, and Department of Orthopedic Surgery &
Biochemistry, Richmond, Virginia, U.S.A.
Paul J. Benkeser / Georgia Institute of Technology and Emory University, Atlanta, Georgia, U.S.A.
Besim Ben-Nissan / University of Technology Sydney, New South Wales, Australia
Stanley A. Berger / University of California, Berkeley, California, U.S.A.
Earl J. Bergey / State University of New York at Buffalo, Buffalo, New York, U.S.A.
Franc- ois Berthiaume / Massachusetts General Hospital, Harvard Medical School, and the Shriners
Hospital for Children, Boston, Massachusetts, U.S.A.
Serena Best / University of Cambridge, Cambridge, U.K.
Kazuhisa Bessho / Kyoto University, Sakyo-Ku, Kyoto, Japan
Deon Bezuidenhout / University of Cape Town Medical School, and Groote Schuur Hospital,
Cape Town, South Africa
Vineet Bhandari / Yale University School of Medicine and Children’s Hospital, New Haven,
Connecticut, U.S.A.
Luc Bilodeau / Montreal Heart Institute, Montreal, Quebec, Canada
Katharina Bittner / BioSurgery European Marketing, Baxter BioScience, Munich, Germany
viii
Matthew J. Bizzarro / Yale University School of Medicine and Children’s Hospital, New Haven,
Connecticut, U.S.A.
Kirby S. Black / CryoLife, Inc., Kennesaw, Georgia, U.S.A.
Michael Blakeney / University of London, London, U.K.
Torsten Blunk / University of Regensburg, Regensburg, Germany
Aldo R. Boccaccini / Imperial College London, London, United Kingdom
Aldo Boccaccini / Imperial College London, London, United Kingdom
Ivan Bojanić / University of Zagreb, Zagreb, Croatia
Jack C. Bokros / Medical Carbon Research Institute, Austin, Texas, U.S.A.
Eugene D. Boland / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Babul Borah / Procter & Gamble Pharmaceuticals, Mason, Ohio, U.S.A.
J. T. Borenstein / Charles Stark Draper Laboratory, Cambridge, Massachusetts, U.S.A.
Adele L. Boskey / Hospital for Special Surgery, New York, New York, U.S.A.
Gary L. Bowlin / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
James R. Bowman, III / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Kellye D. Branch / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Ricardo R. Brau / Massachusetts Institute of Technology, Cambridge, Massachusetts, U.S.A.
Robert C. Bray / University of Calgary, Calgary, Alberta, Canada, and McCaig Centre for Joint
Injuries and Arthritis Research
William T. Brinkman / Emory University School of Medicine, Atlanta, Georgia, U.S.A.
William C. Broaddus / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Kelvin G. M. Brockbank / Organ Recovery Systems, Charleston, South Carolina, U.S.A.
Timothy J. Broderick / University of Cincinnati College of Medicine, Cincinnati, Ohio, U.S.A.
Anna-Liisa Brownell / Massachusetts General Hospital, Boston, Massachusetts, U.S.A.
Ian C. Bruce / McMaster University, Hamilton, Ontario, Canada
Donald M. Brunette / University of British Columbia, Vancouver, British Columbia, Canada
Gil Bub / McGill University, Montreal, Quebec, Canada
Anne-Marie Buckle / The University of Manchester, Manchester, England, U.K.
Joel D. Bumgardner / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Andrew Burd / The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
Christie D. Bures / University of Texas at Austin, Austin, Texas, U.S.A.
Elisabeth H. Burger / Vrije Universiteit, Amsterdam, The Netherlands
Karen J.L. Burg / Clemson University, Clemson, South Carolina, U.S.A.
Lori L. Burrows / Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
Sandra C. P. Cachinho / University of Liverpool, United Kingdom
Jinhua Cao / Purdue University, West Lafayette, Indiana, U.S.A.
E. L. Carniel / University of Padua, Padua, Italy
Marcus E. Carr, Jr. / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Patrick Carrier / LOEX, Hôpital du Saint-Sacrement de CHA, and Laval University, Quebec City,
Quebec, Canada
Elliot L. Chaikof / Emory University School of Medicine, and Georgia Institute of Technology, Atlanta,
Georgia, U.S.A.
Christina Chan / Massachusetts General Hospital, Harvard Medical School, and the Shriners Hospital
for Children, Boston, Massachusetts, U.S.A.
W. Chang / Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, U.S.A.
Aditya Chaubey / Clemson University, Clemson, South Carolina, U.S.A.
Babak Chehroudi / University of British Columbia, Vancouver, British Columbia, Canada
Q. Chen / The University of Wisconsin–Madison, Madison, Wisconsin, U.S.A.
Qi-Zhi Chen / Imperial College London, London, United Kingdom
ix
Shaochen Chen / University of Texas at Austin, Austin, Texas, U.S.A.
Ruizhen Rachel Chen / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
S. Chethana / Central Food Technological Research Institute, Mysore, Karnataka, India
Vikram S. Chib / Northwestern University, Chicago, Illinois, U.S.A.
Traian V. Chirila / Lions Eye Institute, Nedlands, Western Australia, Australia
Paula Chmielewski / Procter & Gamble Pharmaceuticals, Mason, Ohio, U.S.A.
Charles D. Choi / General Dynamics Advanced Information Systems, Fairfax, Virginia, and University of
Michigan, Ann Arbor, Michigan, U.S.A.
P. Christian / University of Manchester, Manchester, Greater Manchester, U.K.
Seth I. Christian / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Chih-Chang Chu / Cornell University, Ithaca, New York, U.S.A.
Paul Kim Ho Chu / City University of Hong Kong, Kowloon, Hong Kong
Hsi-Wei Chung / Mayo Clinic College of Medicine, Rochester, Minnesota, U.S.A.
Timothy A. M. Chuter / University of California San Francisco, San Francisco, California, U.S.A.
Graeme M. Clark / University of Melbourne, East Melbourne, Victoria, Australia
William R. Clark / Gambro Renal Products (Intensive Care), Indianapolis, Indiana, U.S.A.
Suzanne Conroy / AST Products, Inc., Billerica, Massachusetts, U.S.A.
Rory A. Cooper / University of Pittsburgh, and VA Pittsburgh Healthcare System, Pittsburgh,
Pennsylvania, U.S.A.
Alastair N. Cormack / New York State College of Ceramics at Alfred University, Alfred, New York, U.S.A.
Michel Cormier / ALZA Corporation, Mountain View, California, U.S.A.
Brian N. Craig / Lamar University, Beaumont, Texas, U.S.A.
Nancy A. Crigger / Baylor College of Medicine, Houston, Texas, U.S.A.
J. K. Critser / University of Missouri, Columbia, Missouri, U.S.A.
J.N. Amritha De Croos / Mount Sinai Hospital, Toronto, Ontario, Canada
M. J. Cross / Australian Institute of Musculo-Skeletal Research, Sydney, New South Wales, Australia,
University of Sydney, Sydney, New South Wales, Australia
Guy Daculsi / Nantes University, and INSERM EM 9903, INSERM, Nantes, France, France
Peter Daddona / ALZA Corporation, Mountain View, California, U.S.A.
Jawwad A. Darr / University College London, London, U.K.
Neil Davies / University of Cape Town, Cape Town, South Africa
Jeff Davis / University of South Carolina, Columbia, South Carolina, U.S.A.
Patti E. Dawson / CryoLife, Inc., Kennesaw, Georgia, U.S.A.
Richard M. Day / St. Mark’s Hospital, London, United Kingdom
Geoff Dearnaley / Southwest Research Institute, San Antonio, Texas, U.S.A.
Richard E. Debski / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
K. C. Dee / Tulane University, New Orleans, Louisiana, U.S.A.
Stephen P. Denyer / University of Brighton, Brighton, and Cardiff University, Cardiff, United Kingdom
Tejal Desai / Boston University, Boston, Massachusetts, U.S.A.
Steven Deutsch / The Pennsylvania State University, University Park, Pennsylvania, U.S.A.
Scott L. Diamond / University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A.
Robert F. Diegelmann / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
James P. Dioria / Baxter Technology Resources, Round Dake, Illinois, U.S.A.
L. Di-Silvio / University College London, Royal National Orthopaedic Hospital Trust, Middlesex, and
The Guy’s, King’s, and St. Thomas’ Hospital Medical and Dental Institute, King’s College London,
London, United Kingdom
Charles R. Doarn / Virginia Commonwealth University, Richmond, Virginia, and University of
Cincinnati, Cincinnati, Ohio, U.S.A.
P. J. Doherty / University of Liverpool, Liverpool, United Kingdom
x
A. J. Domb / The Hebrew University of Jerusalem, Jerusalem, Israel
Cristina Donini / University of Texas at Austin, Austin, Texas, U.S.A., and Plan-les-Ouates, Geneva,
Switzerland
Simon J. Doran / University of Surrey, Guildford, Surrey, and Institute of Cancer Research, Sutton,
Surrey, U.K.
Robert H. Doremus / Rensselaer Polytechnic Institute, Troy, New York, U.S.A.
Karen A. Duca / Virginia State University, Blacksburg, Virginia, U.S.A.
Thomas Dufresne / Procter & Gamble Pharmaceuticals, Mason, Ohio, U.S.A.
James C.Y. Dunn / University of California, Los Angeles, California, U.S.A.
Dominique M. Durand / Case Western Reserve University, Cleveland, Ohio, U.S.A.
Daniel Eberli / Wake Forest Institute for Regenerative Medicine, Winston Salem,
North Carolina, U.S.A.
Clive Edwards / School of Biological Sciences, The University of Liverpool, Liverpool, U.K.
Glenn A. Edwards / University of Melbourne, Werribee, Victoria, Australia
Igor R. Efimov / Case Western Reserve University, Cleveland, Ohio, U.S.A.
Jennifer H. Elisseeff / Johns Hopkins University, Baltimore, Maryland, U.S.A.
Ville Ellä / Tampere University of Technology, Institute of Biomaterials, Tampere, Finland
Özgür Erdoğan / C - ukurova University, Balcali, Adana, Turkey
Thomas Eschenhagen / University-Hospital Hamburg-Eppendorf, Hamburg, Germany
Paul G. Espy / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Anna Fallon / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Anna M. Fallon / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Sergio Fantini / Tufts University, Medford, Massachusetts, U.S.A.
Richard G. A. Faragher / University of Brighton, Brighton, United Kingdom
William J. Federspiel / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Ding-Yu Fei / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Fatiha El Feninat / Laval University, Quebec City, Quebec, Canada
John Fisher / University of Leeds, Leeds, United Kingdom
Willem Flameng / Katholieke Universiteit Leuven, Belgium
Odette Fokapu / University of Technology of Compie`gne, Compie`gne, France
Arnold A. Fontaine / The Pennsylvania State University, University Park, Pennsylvania, U.S.A.
Kevin R. Forrester / University of Calgary, Calgary, Alberta, Canada
Kenneth R. Foster / University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A.
Joanna D. Fromstein / University of Toronto, Toronto, Ontario, Canada
James G. Fujimoto / Massachusetts Institute of Technology, Cambridge, Massachusetts, U.S.A.
Hisao Fukui / Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan
Keertik S. Fulzele / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Ricky K.Y. Fu / City University of Hong Kong, Kowloon, Hong Kong
Edward P. Furlani / State University of New York at Buffalo, Buffalo, New York, U.S.A.
Franck Furno / University Hospital, Nottingham, United Kingdom, and Stockhausen GmbH and Co.
KG, Krefeld, Germany
Simon M. Gabe / St. Mark’s Hospital, London, United Kingdom
Michael Gaitan / National Institute of Standards and Technology, Gaithersburg, Maryland, U.S.A.
Bob Gale / ALZA Corporation, Mountain View, California, U.S.A.
Didier Gamet / University of Technology of Compie`gne, Compie`gne, France
Israel Gannot / National Institute of Health, NICHD, Biomedical Stochastic Physics, Bethesda,
Maryland, U.S.A., and Faculty of Engineering, Tel-Aviv University, Tel-Aviv, Israel
Dayong Gao / University of Kentucky, Lexington, Kentucky, U.S.A.
Martin Oswaldo Mendez Garcia / Polytechnic University, Milan, Italy
xi
Leslie A. Geddes / Purdue University, West Lafayette, Indiana, U.S.A.
E. Gentleman / Tulane University, New Orleans, Louisiana, U.S.A.
Lucie Germain / LOEX, Hôpital du Saint-Sacrement du CHA, and Laval University, Quebec City,
Quebec, Canada
Salem Ghrebi / University of British Columbia, Vancouver, British Columbia, Canada
Claude J. Giasson / LOEX, Hôpital du Saint-Sacrement du CHA, and Laval University, Quebec City,
Quebec, Canada
Thomas W. Gilbert / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
George T. Gillies / Virginia Commonwealth University, Richmond, and University of Virginia,
Charlottesville, Virginia, U.S.A.
David L. Glaser / University of Pennsylvania Health System, Philadelphia, Pennsylvania, U.S.A.
Julie Glowacki / Brigham and Women’s Hospital, Harvard Medical School, and Harvard School of
Dental Medicine, Boston, Massachusetts, U.S.A.
Mukul S. Goel / University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A.
Alan H. Goldstein / New York State College of Ceramics at Alfred University, Alfred, New York,
U.S.A.
Cheryl T. Gomillion / Clemson University, Clemson, South Carolina, U.S.A.
Mark Gonzalez / University of Illinois at Chicago, Chicago, Illinois, U.S.A.
Stuart B. Goodman / Stanford Medical Center, Stanford, California, U.S.A.
Richard L. Goodwin / University of South Carolina, Columbia, South Carolina, U.S.A.
J.E. Gough / University of Manchester, Manchester, Greater Manchester, U.K.
Achim Göpferich / University of Regensburg, Regensburg, Germany
Lloyd D. Graham / Riverside Life Sciences Centre, New South Wales, Australia
Edward M. Greenfield / Case Western Reserve University, Cleveland, Ohio, U.S.A.
Howard P. Greisler / Loyola University Medical Center, Maywood, Illinois, U.S.A.
Tapio Grönfors / University of Kuopio, Finland
Kelley Grorud / Edgewood College, Madison, Wisconsin, U.S.A.
Jianjun Guan / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Sylvain L. Guerin / LOEX, Hôpital du Saint-Sacrement du CHA, and Laval University, Quebec City,
Quebec, Canada
N. Gurav / University College London, Royal National Orthopaedic Hospital Trust, Middlesex,
United Kingdom
Peter J. Haar / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Mutaz B. Habal / Tampa Bay Craniofacial Center, Tampa, Florida, U.S.A.
Marie Haı̈dopoulos / Laval University, Quebec City, Quebec, Canada
Nadim James Hallab / Rush-Presbyterian-St. Lukes Medical Center, Chicago, Illinois, U.S.A.
Douglas Hamilton / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Douglas W. Hamilton / University of British Columbia, Vancouver, British Columbia, Canada
Stephen R. Hanson / Emory University, Atlanta, Georgia, U.S.A., and Oregon Health & Science
University, Beaverton, Oregon, U.S.A.
Brett M. Harnett / Virginia Commonwealth University, Richmond, Virginia, and University of
Cincinnati, Cincinnati, Ohio, U.S.A.
Alex Hartov / Dartmouth College, Hanover, New Hampshire, U.S.A.
Julie Hasenwinkel / Syracuse University, Syracuse, New York, U.S.A.
Mineo Hashizume / Nara Institute of Science and Technology, Nara, Japan
Vasif Hasirci / Middle East Technical University, Ankara, Turkey
Tomokazu Hattori / Faculty of Science and Technology, Meijo University, Tempaku-ku, Nagoya, Japan
Zhaoming He / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Sam L. Helgerson / BioScience R & D Baxter BioScience, Fremont, California, U.S.A.
Kristie A. Henchir / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
xii
Larry L. Hench / Imperial College London, London, United Kingdom
O. M. Hess / University Hospital, Bern, Switzerland
Celia R. Hicks / Lions Eye Institute, Nedlands, Western Australia, Australia
Robert S. D. Higgins / Rush-Presbyterian-St. Luke’s Medical Center, Chicago, Illinois, U.S.A.
Motohiro Hirose / National Institute of Advanced Industrial Science and Technology (AIST),
Amagasaki, Hyogo, Japan
Jeffrey W. Holmes / Columbia University, New York, New York, U.S.A.
Haoqing Hou / The University of Akron, Akron, Ohio, U.S.A.
S. M. Hudson / North Carolina State University, Raleigh, North Carolina, U.S.A.
Xin-Hua Hu / East Carolina University, Greenville, North Carolina, U.S.A.
Paul Human / University of Cape Town Medical School, and Groote Schuur Hospital,
Cape Town, South Africa
John A. Hunt / University of Liverpool, Liverpool, United Kingdom
Thomas L. Husted / University of Cincinnati College of Medicine, Cincinnati, Ohio, U.S.A.
Rejhan Idrizi / Bernard O’Brien Institute of Microsurgery, and St. Vincent’s Hospital, Melbourne,
Victoria, Australia
Yoshito Ikada / Suzuka University of Medical Science, Suzuka, Mie, Japan
Atsushi Ikeda / Nara Institute of Science and Technology, Nara, Japan
Ilko K. Ilev / Center for Devices and Radiological Health, Office of Science and Engineering
Laboratories, Division of Physics, Rockville, Maryland, U.S.A.
Eileen Ingham / University of Leeds, Leeds, United Kingdom
Koji Ioku / National Institute of Advanced Industrial Science and Technology (AIST), Amagasaki,
Hyogo, Japan
Kazuhiko Ishihara / The University of Tokyo, Tokyo, Bunkyo-ku, Japan
Atsuo Ito / National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
Alan Ivković / University of Zagreb, Zagreb, Croatia
Hiroo Iwata / Kyoto University, Kyoto, Sakyo-Ku, Japan
Joshua Jacobs / Rush-Presbyterian-St. Lukes Medical Center, Chicago, Illinois, U.S.A.
E. Duco Jansen / Vanderbilt University, Nashville, Tennessee, U.S.A.
Nicolas M. Jedynakiewicz / The University of Liverpool, Liverpool, U.K.
Jorge H. Jimenez / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Julian R. Jones / Imperial College London, London, United Kingdom
James W. Jones / Baylor College of Medicine, Houston, Texas, U.S.A.
Jeffrey I. Joseph / Jefferson Medical College of Thomas Jefferson University, Philadelphia,
Pennsylvania, U.S.A.
Martti Juhola / University of Tampere, Finland
M. R. Kaazempur-Mofrad / Massachusetts Institute of Technology, Cambridge, and Massachusetts
General Hospital, and Harvard Medical School, Boston, Massachusetts, U.S.A.
Tonya Kaltenbach / Stanford University School of Medicine, Stanford, California, U.S.A.
Rita A. Kandel / Mount Sinai Hospital, Toronto, Ontario, Canada
Leon Kane-Maguire / University of Wollongong, Wollongong, New South Wales, Australia
W. John Kao / University of Wisconsin-Madison, Madison, Wisconsin, U.S.A.
Vigneshwar Kasirajan / Medical College of Virginia, Richmond, Virginia, U.S.A.
Toshihiro Kasuga / Nagoya Institute of Technology, Gokiso-cho Showa-ku, Nagoya, Japan
Koichi Kato / Kyoto University, Sakyo-ku, Kyoto, Japan
J. Lawrence Katz / University of Missouri–Kansas City School of Dentistry, Kansas City, Missouri, and
University of Texas–Houston School of Medicine, Houston, Texas, U.S.A.
Daisuke Kawagoe / Tohoku University, Aoba-ku, Sendai, Japan
Jill S. Kawalec / Ohio College of Podiatric Medicine, Cleveland, Ohio, U.S.A.
xiii
Christopher Keen / Virginia Commonwealth University, Richmond, Virginia, U.S.A
William S. Kerwin / University of Washington, Seattle, Washington, U.S.A.
Gilson Khang / Chonbuk National University, Jeonju, Korea
Antoine E. Khoury / Hospital for Sick Children, Toronto, Ontario, Canada
Jun-ichi Kikuchi / Nara Institute of Science and Technology, Nara, Japan
Hugh Kim / University of British Columbia, Vancouver, British Columbia, Canada
Kyo-Han Kim / Kyunpook National University, Jung-gu, Daegu, South Korea
Moon Suk Kim / Korea Research Institute of Chemical Technology, Yuseong, Daejeon, Korea
Yoshihiro Kiritoshi / The University of Tokyo, Tokyo, Bunkyo-ku, Japan
Jenneke Klein-Nulend / Vrije Universiteit, Amsterdam, The Netherlands
Jonathan P. Van Kleunen / University of Pennsylvania Health System, Philadelphia,
Pennsylvania, U.S.A.
Kenneth R. Knight / Bernard O’Brien Institute of Microsurgery, and St. Vincent’s Hospital, Melbourne,
Victoria, Australia
Kenneth L. Koch / Wake Forest University School of Medicine, Winston-Salem, North Carolina, U.S.A.
Hiroko Kojima / National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba,
Ibaraki, Japan
Frank K. Ko / Drexel University, Philadelphia, Pennsylvania, U.S.A.
Claire F. Komives / San Jose State University, San Jose, California, U.S.A.
Thomas J. Koob / Shriners Hospitals for Children, Tampa, Florida, U.S.A.
Feza Korkusuz / Middle East Technical University, Ankara, Turkey
Petek Korkusuz / Hacettepe University, Sihhiye, Ankara, Turkey
Noriko Kotobuki / National Institute of Advanced Industrial Science and Technology (AIST),
Amagasaki, Hyogo, Japan
Katherine M. Kulig / Massachusetts General Hospital, Boston, Massachusetts, U.S.A.
Rakesh Kumar / University of Alberta, Edmonton, Alberta, Canada
M.N.V. Ravi Kumar / National Institute of Pharmaceutical Education and Research (NIPER),
Mohali, India
Fred M. Kusumoto / Lovelace Medical Center, Albuquerque, New Mexico, U.S.A.
Kathy E. Laber / Medical University of South Carolina, Charleston, South Carolina, U.S.A.
Roger Laham / Beth Israel Deaconess Medical Center and Harvard Medical School, Boston,
Massachusetts, U.S.A.
Andres Laib / Scanco Medical AG, Bassersdorf, Switzerland
Henry Lai / University of Washington, Seattle, Washington, U.S.A.
James C. K. Lai / College of Pharmacy and Biomedical Research Institute, Idaho State University,
Pocatello, Idaho, U.S.A.
James M. Lambert / SiTech, LLC, Richmond, Virginia, U.S.A.
Philippe Lam / Genentech, Inc., South San Francisco, California, U.S.A.
James Laredo / Loyola University Medical Center, Maywood, Illinois, U.S.A.
Rolf Larsson / University Hospital, Uppsala, Sweden
Robert A. Latour, Jr. / Clemson University, Clemson, South Carolina, U.S.A.
Jennie Baier Leach / University of Texas, Austin, Texas, U.S.A.
Benjamin Lee / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Brendan Lee / Baylor College of Medicine, Houston, Texas, U.S.A.
Hai Bang Lee / Korea Research Institute of Chemical Technology, Yuseong, Daejeon, Korea
Jin Ho Lee / Hannam University, Daejeon, Korea
Susan S. Lee / National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, and Johns
Hopkins University, Baltimore, Maryland, U.S.A.
Jae Sung Lee / Pohang University of Science and Technology, Pohang, Korea
xiv
Racquel LeGeros / New York University College of Dentistry, New York, New York, U.S.A
Nic D. Leipzig / Rice University, Houston, Texas, U.S.A.
Martin L. Lenhardt / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Hwa-Liang Leo / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Catherine A. Leonard / University of Calgary, Calgary, Alberta, Canada
Solomon W. Leung / Idaho State University, Pocatello, Idaho, U.S.A.
Andrew L. Lewis / Biocompatibles UK Limited, Farnham, Surrey, United Kingdom
Gladius Lewis / The University of Memphis, Memphis, Tennessee, U.S.A.
Baruch B. Lieber / University of Miami, Coral Gables, Florida, U.S.A.
Andrea Liebmann-Vinson / BD Technologies, Research Triangle Park, North Carolina, U.S.A.
Martin Lietz / NMI Naturwissenschaftliches und Medizinisches Institut an der Universität Tübingen,
Reutlingen, Germany
E. N. Lightfoot / Virginia State University, Blacksburg, Virginia, U.S.A.
Mengnai Li / Department of Orthopaedic Surgery, Richmond, Virginia, U.S.A.
Xingde Li / University of Washington, Seattle, Washington, U.S.A.
Steven J. Lindauer / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Christian Lindqvist / Helsinki University and University Hospital, Helsinki, Finland
Tung-Liang Lin / AST Products, Inc., Billerica, Massachusetts, U.S.A.
Huinan Liu / Purdue University, West Lafayette, Indiana, and, Brown University, Providence,
Rhode Island, U.S.A.
Jonathan T.C. Liu / Stanford University School of Medicine, Stanford, California, U.S.A.
G. A. Livesay / Tulane University, New Orleans, Louisiana, U.S.A.
Andrew W. Lloyd / University of Brighton, Brighton, United Kingdom
Laurie E. Locascio / National Institute of Standards and Technology, Gaithersburg, Maryland, U.S.A.
Ih-Houng Loh / AST Products, Inc., Billerica, Massachusetts, U.S.A.
Zerina Lokmic / Bernard O’Brien Institute of Microsurgery, and St. Vincent’s Hospital, Melbourne,
Victoria, Australia
Albert Lozano-Nieto / The Pennsylvania State University, Lehman, Pennsylvania, U.S.A.
Junfeng Lu / University of Kentucky, Lexington, Kentucky, U.S.A.
Yi Lu / University of Texas at Austin, Austin, Texas, U.S.A.
Jun Qing Lu / East Carolina University, Greenville, North Carolina, U.S.A.
Ling Ma / International Rectifier Corporation, El Segundo, California, U.S.A.
Ting Ma / Stanford Medical Center, Stanford, California, U.S.A.
S. Machan / Australian Institute of Musculo-Skeletal Research, Sydney, New South Wales, Australia
Eli E. Machtei / Rambam Medical Center and The Faculty of Medicine, Haifa, Israel
Marc Madou / University of California, Irvine, California, and Integrated Nanosystems Research
Facility (INRF), U.S.A.
Agnese Magnani / University of Siena, and University Center ‘‘Colle di Val d’Elsa,’’ Colle di Val d’Elsa
(SI), Siena, Italy
Luca T. Mainardi / Polytechnic University, Milan, Italy
Viraj P. Mane / Baylor College of Medicine, Houston, Texas, U.S.A.
Keefe B. Manning / The Pennsylvania State University, University Park, Pennsylvania, U.S.A.
Athanassios Mantalaris / Imperial College London, London, United Kingdom
Diego Mantovani / Laval University, Quebec City, Quebec, Canada
Hui Mao / Emory University School of Medicine, Atlanta, Georgia, U.S.A.
Gerard H. Markx / The University of Manchester, Manchester, England, U.K.
Erika J. Martin / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Shojiro Matsuda / Gunze Limited, Ayabe, Kyoto, Japan
Timothy M. Maul / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
xv
Sally L. McArthur / University of Sheffield, Sheffield, United Kingdom
Gretchen J. McAuliffe / Cornell University, Ithaca, New York, U.S.A.
William McCormack / Baylor College of Medicine, Houston, Texas, U.S.A.
Keith M. McLean / Commonwealth Scientific & Industrial Research Organisation (CSIRO) Molecular
Science, Clayton, Victoria, Australia
Carl McMillin / Synthetic Body Parts, Brecksville, Ohio, U.S.A.
Lito C. Mejia / EnduraTEC Systems Corporation, Minnetonka, Minnesota, U.S.A.
Ronald C. Merrell / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Aurora Messina / Bernard O’Brien Institute of Microsurgery, and St. Vincent’s Hospital, Melbourne,
Victoria, Australia
Bart Meuris / Katholieke Universiteit Leuven, Belgium
Evangelia Micheli-Tzanakou / Rutgers University, Piscataway, New Jersey, U.S.A.
Gerald E. Miller / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Tommy L. Miller / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Anthony J. Minisi / McGuire VA Medical Center, Richmond, Virginia, U.S.A.
Geraldine Mitchell / Bernard O’Brien Institute of Microsurgery, and St. Vincent’s Hospital, Melbourne,
Victoria, Australia
Teruo Miyata / Koken Co. Ltd., Toshima-ku, Tokyo, Japan
Mark P. Mooney / University of Pittsburgh and Carnegie Mellon University, Pittsburgh, Pennsylvania,
U.S.A.
Susan M. Moore / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Robert B. More / RBMore Associates, Austin, Texas, U.S.A.
Wayne Morrison / Bernard O’Brien Institute of Microsurgery, and St. Vincent’s Hospital, Melbourne,
Victoria, Australia
Alireza Moshaverinia / Ohio State University, Columbus, Ohio, U.S.A.
Shaker A. Mousa / Albany College of Medicine, and Albany College of Pharmacy, Albany, New York,
U.S.A.
Ahmed S. Mousa / Cornell University, Ithaca, New York, U.S.A.
Eike H. Mrosek / Mayo Clinic College of Medicine, Rochester, Minnesota, U.S.A.
William L. Murphy / University of Wisconsin, Madison, Wisconsin, U.S.A.
Joy L. Murray / University of Texas, Austin, Texas, U.S.A.
Karthik Nagapudi / Emory University School of Medicine, Atlanta, Georgia, U.S.A.
Kwang Woo Nam / University of Tokyo, Tokyo, Bunkyo-ku, Japan
A. N. Natali / University of Padova, Padova, Italy
E. A. Nauman / Tulane University, New Orleans, Louisiana, U.S.A.
Stephen S. Navran / Synthecon, Inc., Houston, Texas, U.S.A.
Markus Neubauer / University of Regensburg, Regensburg, Germany
Daniel Newton / Virginia Commonwealth University, Richmond, Virginia, U.S.A
S. Nguyen / Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, U.S.A.
Paul C. Nicolson / Atlanta, Georgia, U.S.A.
Alejandro Nieponice / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Mitsuo Niinomi / Toyohashi University of Technology, Tempaku-cho, Toyohashi, Japan
Vladimir P. Nikolski / Case Western Reserve University, Cleveland, Ohio, U.S.A.
Yasuharu Noishiki / Yokohama City University School of Medicine, Kanazawa-ku, Yokohama, Japan
Giandomenico Nollo / University of Trento, and FBK–Fondazione Bruno Kessler, Trento, Italy
Yukihiko Nosé / Baylor College of Medicine, Houston, Texas, U.S.A.
Robert H. Notter / University of Rochester School of Medicine, Rochester, New York, U.S.A.
Matthew O’Donnell / University of Michigan, Ann Arbor, Michigan, U.S.A.
Shawn W. O’Driscoll / Mayo Clinic College of Medicine, Rochester, Minnesota, U.S.A.
xvi
Brenda M. Ogle / Mayo Clinic, Rochester, Minnesota, U.S.A.
Hajime Ohgushi / National Institute of Advanced Industrial Science and Technology, Amagasaki City,
Hyogo, and Tohoku University, Aoba-ku, Sendai, Japan
Ruth J. Okamoto / Washington University, St. Louis, Missouri, U.S.A.
Regis J. O’Keefe / University of Rochester, Rochester, New York, USA
Hisashi Okubo / Baylor College of Medicine, Houston, Texas, U.S.A.
Eric J. Okum / Medical College of Virginia, Richmond, Virginia, U.S.A.
Joo L. Ong / University of Tennessee Health Science Center, Memphis, Tennessee, U.S.A.
Richard O. C. Oreffo / University of Southampton, Southampton, United Kingdom
Zoubeida Ounaies / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Rama Padmanabhan / ALZA Corporation, Mountain View, California, U.S.A.
Rakesh K. Pai / University of New Mexico School of Medicine, Albuquerque, New Mexico, U.S.A.
Nicki Panoskaltsis / Imperial College London, London, United Kingdom
E. N. Parish / Australian Institute of Musculo-Skeletal Research, Sydney, New South Wales, Australia
Kwideok Park / The University of Memphis, Memphis, Tennessee, U.S.A.
Sang-Won Park / Chonnam national University, Dong-Ku, Gwang-ju, South Korea
Joon B. Park / University of Iowa, Iowa City, Iowa, U.S.A.
Grace E. Park / Purdue University, West Lafayette, Indiana, U.S.A.
Robert S. Parker / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Cheryl A. Parzel / Clemson University, Clemson, South Carolina, U.S.A.
Nelesh Patel / University of Cambridge, Cambridge, U.K.
Ganapathi Patil / Central Food Technological Research Institute, Mysore, Karnataka, India
Keith D. Paulsen / Dartmouth College, Hanover, New Hampshire, U.S.A.
P. G. Pavan / University of Padova, Padova, Italy
Robert R. Pavlis / Pittsburg State University, Pittsburg, Kansas, U.S.A.
Kristin J. Pawlowski / National Aeronautics and Space Administration (NASA), Langley Research
Center, Hampton, Virginia, U.S.A.
Jon L. Pearlman / University of Pittsburgh, and VA Pittsburgh Healthcare System, Pittsburgh,
Pennsylvania, U.S.A.
Marko Pećina / University of Zagreb, Zagreb, Croatia
Nicholas A. Peppas / University of Texas at Austin, Austin, Texas, U.S.A.
Marivalda M. Pereira / Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
Brad Phipps / ALZA Corporation, Mountain View, California, U.S.A.
Dominique P. Pioletti / Swiss Federal Institute of Technology, and Hôpital Orthope´dique de la Suisse
Romande, and Bone Bioengineering Group, STI/BIO-E/CRO, Lausanne, Switzerland
Federica M. Piras / University of Siena, Siena, Italy, University Center ‘‘Colle di Val d’Elsa,’’ Colle di
Val d’Elsa (SI), Siena, Italy
Eija Marjut Pirhonen / Inion Oy Tampere, Finland
Roland N. Pittman / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Jeffrey L. Platt / Mayo Clinic, Rochester, Minnesota, U.S.A.
Churn K. Poh / University of Kentucky, Lexington, Kentucky, U.S.A.
Mariano A. Polack / Gainesville, Virginia, U.S.A.
Milos R. Popovic / University of Toronto, Toronto, Ontario, Canada
Dejan B. Popović / Aalborg University, Aalborg, Denmark, and University of Belgrade, Belgrade, Serbia
Alexandra Elizabeth Porter / University of Cambridge, Cambridge, U.K.
Sharon C. Presnell / BD Technologies, Research Triangle Park, North Carolina, U.S.A.
Robert L. Price / University of South Carolina, Columbia, South Carolina, U.S.A.
D. A. Puleo / University of Kentucky, Lexington, Kentucky, U.S.A.
Andrew J. Pullan / University of Auckland, Auckland, New Zealand
xvii
K. S. M. S. Raghavarao / Central Food Technological Research Institute, Mysore, Karnataka, India
P. Rahko / The University of Wisconsin–Madison, Madison, Wisconsin, U.S.A.
Eric J. Rainis / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Samuel C. Ramage / Department of Biochemistry, Richmond, Virginia, U.S.A.
John A. M. Ramshaw / Molecular and Health Sciences, Clayton South, Victoria, Australia
Ihtesham U. Rehman / Queen Mary University of London, London, U.K.
Bernd H. A. Rehm / Massey University, Palmerston North, New Zealand
Gregory G. Reinholz / Mayo Clinic College of Medicine, Rochester, Minnesota, U.S.A.
Darrell H. Reneker / The University of Akron, Akron, Ohio, U.S.A.
Darwin R. Reyes / National Institute of Standards and Technology, Gaithersburg, Maryland, U.S.A.
Daniel C. Richardson / Stanford University, Stanford, California, U.S.A.
Bruce W. Richman / Baylor College of Medicine, Houston, Texas, U.S.A.
J. Paul Robinson / Purdue University, West Lafayette, Indiana, U.S.A.
Michael R. Robinson / National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda,
Maryland, U.S.A.
Gerson Rosenberg / Penn State College of Medicine, Hershey, Pennsylvania, U.S.A.
Jacob Rosen / University of Washington, Seattle, Washington, U.S.A.
J.M. Rosen / Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, U.S.A.
Tobias Saam / University of Washington, Seattle, Washington, U.S.A.
E.E. Sabelman / Department of Neurology, Kaiser-Permanente Medical Center, Redwood City,
California, U.S.A.
Michael S. Sacks / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Chander Sadasivan / University of Miami, Coral Gables, Florida, U.S.A.
Yudhisthira Sahoo / State University of New York at Buffalo, Buffalo, New York, U.S.A.
Takashi Saito / Health Sciences University of Hokkaido, Ishikari-gun, Hokkaido, Japan
Christian Salesse / LOEX, Hopital du saint-Sacrement du CHA, and Lava University, Quebec City,
Quebec, Canada
Paul T. Salo / University of Calgary, Calgary, Alberta, Canada
Susan Sandeman / University of Brighton, Brighton, United Kingdom
Archit B. Sanghvi / University of Texas, Austin, Texas, U.S.A.
Ann Marie Sastry / University of Michigan, Ann Arbor, Michigan, U.S.A.
Jan C. Schagemann / Mayo Clinic College of Medicine, Rochester, Minnesota, U.S.A.
Constantin Schizas / The Whittington Hospital, University College London, London, and Institute of
Orthopaedics, Royal National Orthopaedic Hospital, Stanmore, United Kingdom
Burkhard Schlosshauer / NMI Naturwissenschaftliches und Medizinisches Institut an der Universität
Tübingen, Reutlingen, Germany
Christine E. Schmidt / University of Texas, Austin, Texas, U.S.A.
Craig L. Schmidt / Medtronic, Inc., Brooklyn Center, Minnesota, U.S.A.
Jerome S. Schultz / University of California, Riverside, California, U.S.A.
Edward M. Schwarz / University of Rochester, Rochester, New York, USA
Erik R. Scott / Medtronic, Inc., Brooklyn Center, Minnesota, U.S.A.
Thomas Seelich / BioSurgery R & D, Baxter Bio Science, Vienna, Austria
Denis Semwogerere / Emory University, Atlanta, Georgia, U.S.A.
Alexander B. G. Sevy / Massachusetts General Hospital, Boston, Massachusetts, U.S.A.
Mohsen Shahinpoor / University of New Mexico, Albuquerque, New Mexico, U.S.A.
V. Prasad Shastri / Vanderbilt University, Nashville, Tennessee, U.S.A.
Min-Shyan Sheu / AST Products, Inc., Billerica, Massachusetts, U.S.A.
Kelly J. Shields / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Michael L. Shuler / Cornell University, Ithaca, New York, U.S.A.
xviii
K. Kirk Shung / University of Southern California, Los Angeles, California, U.S.A.
Michael I. Siegel / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Olivier Siegrist / Hôpital Orthope´dique de la Suisse Romande, Lausanne, Switzerland
David G. Simpson / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
George Sines / University of California, Los Angeles, California, U.S.A.
Paul M. Skarstad / Medtronic, Inc., Brooklyn Center, Minnesota, U.S.A.
Oskar Skrinjar / Georgia Institute of Technology and Emory University, Atlanta, Georgia, U.S.A.
John D. Smart / University of Portsmouth, Portsmouth, and University of Brighton, Brighton,
United Kingdom
Mari A. Smith / Johns Hopkins University School of Medicine, Baltimore, Maryland, U.S.A.
Nadine Barrie Smith / Pennsylvania State University, University Park, Pennsylvania, U.S.A.
Daniel H. Smith / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Nicolas P. Smith / University of Auckland, Auckland, New Zealand
Theo H. Smit / Vrije Universiteit Medical Center, Amsterdam, The Netherlands
Roy M. Soetikno / Stanford University School of Medicine, Stanford, California, U.S.A.
Lorenzo Soletti / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Ying C. Song / Organ Recovery Systems, Charleston, South Carolina, U.S.A.
Louis J. Soslowsky / University of Pennsylvania Health System, Philadelphia, Pennsylvania, U.S.A.
Reiner Spaethe / BioSurgery R & D, Baxter BioScience, Vienna, Austria.
John E. Speich / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Paulette Spencer / University of Missouri–Kansas City School of Dentistry, Kansas City,
Missouri, U.S.A.
Michael J. Spivey / Cornell University, Ithaca, New York, U.S.A.
Naveen C. Srivastav / University of Alberta, Edmonton, Alberta, Canada
John J. Stankus / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Robert M. Stern / The Pennsylvania State University, University Park, Pennsylvania, U.S.A.
George D. Stetten / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Molly M. Stevens / Imperial College London, London, United Kingdom
Shih-Horng Su / AST Products, Inc., Billerica, Massachusetts, U.S.A.
Riitta Suuronen / Tampere University and University Hospital, Tampere, Finland.
Robert G. Svitek / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
M. Michael Swindle / Medical University of South Carolina, Charleston, South Carolina, U.S.A.
Michael Szycher / CardioTech International, Inc., Woburn, Massachusetts, U.S.A.
Maryam Tabrizian / McGill University, Montreal, Quebec, Canada
Nirit Tagger-Green / Rambam Medical Center and The Faculty of Medicine, Haifa, Israel
Wei Tan / University of Illinois at Urbana-Champaign, Urbana, Illinois, U.S.A.
John M. Tarbell / The Pennsylvania State University, University Park, Pennsylvania, and The City
College of New York, New York, New York, U.S.A.
Daniel A. Tatosian / Cornell University, Ithaca, New York, U.S.A.
Bill Tawil / BioSurgery marketing, Baxter BioScience, Westlake Village, California, U.S.A.
Bruce Taylor / The University of Akron, Akron, Ohio, U.S.A.
Michael J. Taylor / Organ Recovery Systems, Charleston, South Carolina, U.S.A.
Todd A. Telemeco / Shenandoah University, Winchester, Virginia, U.S.A.
Louis Terracio / New York University, New York, New York, U.S.A.
Francesco Tessarolo / University of Trento, Trento, Italy
Eser Tüfekc- i / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Bhavani P. Thampatty / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Benjamin Thierry / McGill University, Montreal, Quebec, Canada
J. Brock Thomas / University of Texas at Austin, Austin, Texas, U.S.A.
xix
T. Adam Thrasher / Toronto Rehabilitation Institute, Toronto, Ontario, Canada
Marc J. Torjman / Jefferson Medical College of Thomas Jefferson University, Philadelphia,
Pennsylvania, U.S.A.
Eleftherios Tsiridis / The Whittington Hospital, University College London, London, and Institute of
Orthopaedics, Royal National Orthopaedic Hospital, Stanmore, United Kingdom, and Boston
University School of Medicine, Boston, MA, U.S.A.
Thomas Tu / Beth Israel Deaconess Medical Center and Harvard Medical School, Boston,
Massachusetts, U.S.A.
Toshimasa Uemura / National Institute of Advanced Industrial Science and Technology (AIST),
Tsukuba, Ibaraki, Japan
Egidijus Uzgiris / Rensselaer Polytechnic Institute, Troy, New York, U.S.A.
Joseph P. Vacanti / Massachusetts General Hospital, and Harvard Medical School, Boston,
Massachusetts, U.S.A.
Bill Van Osdol / ALZA Corporation, Mountain View, California, U.S.A.
Ray Vanderby, Jr. / University of Wisconsin–Madison, Madison, Wisconsin, U.S.A.
Tomy Varghese / University of Wisconsin–Madison, Madison, Wisconsin, U.S.A.
Marcia Vasquez-Lee / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Brent Vernon / Arizona State University, Tempe, Arizona, U.S.A.
Michel Vert / Universite´ Montpellier 1, Montpellier Cedex 5, France
Ivan Vesely / The Cleveland Clinic Foundation, Cleveland, Ohio, U.S.A.
Vibeke Videm / The Norwegian University of Science and Technology, and Trondheim University
Hospital, Trondheim, Norway
Kent S. Vilendrer / Tissue Growth Technologies, Inc., Minnetonka, Minnesota, U.S.A.
Jürgen Vogt / CIBA Vision Corp., Duluth, Georgia, U.S.A.
Nicola Volpi / Biological Chemistry Section, University of Modena and Reggio Emilia, Modena,
Modena, Italy
David Vorp / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
David A. Vorp / McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh,
Pennsylvania, U.S.A.
William R. Wagner / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Gordon Wallace / University of Wollongong, Wollongong, New South Wales, Australia
B. H. Walpoth / University Hospital, Geneva, Switzerland
John R. Walsh / Organ Recovery Systems, Charleston, South Carolina, U.S.A.
Chia-Wei Wang / University of Michigan, Ann Arbor, Michigan, U.S.A.
Dong-an Wang / Johns Hopkins University, Baltimore, Maryland, U.S.A.
Paul Wang / Stanford University Medical Center, Stanford, California, U.S.A.
Yadong Wang / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Yao Wang / Pfizer, Inc., Ann Arbor, Michigan, U.S.A.
Zhengdong Wang / University of Rochester School of Medicine, Rochester, New York, U.S.A.
Thomas D. Wang / Stanford University School of Medicine, Stanford, California, U.S.A.
James H.-C. Wang / University of Pittsburgh, Pittsburgh, Pennsylvania, U.S.A.
Junji Watanabe / The University of Tokyo, Tokyo, Bunkyo-ku, Japan
Jennifer S. Wayne / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Thomas J. Webster / Purdue University, West Lafayette, Indiana, U.S.A.
Eric R. Weeks / Emory University, Atlanta, Georgia, U.S.A.
Michael Wegner / Arizona State University, Tempe, Arizona, U.S.A.
Xiaochao Wei / University of Rochester, Rochester, New York, USA
E. J. Weinberg / Charles Stark Draper Laboratory, and Massachusetts Institute of Technology,
Cambridge, Massachusetts, U.S.A.
Barbara Weiser / University of Regensburg, Regensburg, Germany
xx
Steve T. Wereley / Purdue University, West Lafayette, Indiana, U.S.A.
Jerome A. Werkmeister / Molecular and Health Sciences, Clayton South, Victoria, Australia
Jennifer L. West / Rice University, Houston, Texas, U.S.A.
Richard L. White, Jr. / Carolinas Medical Center, Charlotte, North Carolina, U.S.A.
David L. Williams / Mississippi State University, Mississippi State, Mississippi, U.S.A.
Rachel L. Williams / University of Liverpool, Liverpool, United Kingdom
Gary E. Wnek / Case Western Reserve, Cleveland, Ohio, U.S.A.
Kimberly A. Woodhouse / University of Toronto, Toronto, Ontario, Canada
Debra D. Wright / Michigan Technological University, Houghton, Michigan, U.S.A.
Daniel E. Wueste / Clemson University, Clemson, South Carolina, U.S.A.
J. H. David Wu / University of Rochester, Rochester, New York, U.S.A.
Benjamin M. Wu / University of California, Los Angeles, California, U.S.A.
Kenneth J. Wynne / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Lian Xue / Loyola University Medical Center, Illinois, U.S.A.
Hu Yang / Virginia Commonwealth University, Richmond, Virginia, U.S.A.
Yunzhi Yang / University of Tennessee Health Science Center, Memphis, Tennessee, U.S.A.
Wentao Yan / New York University, New York, New York, U.S.A.
Ioannis V. Yannas / Massachusetts Institute of Technology, Cambridge, Massachusetts, U.S.A.
Juming Yao / Tokyo University of Agriculture and Technology, Koganei, Tokyo, Japan
Martin L. Yarmush / Massachusetts General Hospital, Harvard Medical School, and the Shriners
Hospital for Children, Boston, Massachusetts, U.S.A.
Ajit Yoganathan / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Ajit P. Yoganathan / Georgia Institute of Technology, Atlanta, Georgia, U.S.A.
Michael J. Yost / University of South Carolina, Columbia, South Carolina, U.S.A.
Chun Yuan / University of Washington, Seattle, Washington, U.S.A.
Peng Yuan / National Institutes of Health Clinical Center, Bethesda, Maryland, U.S.A.
Deniz Yucel / Middle East Technical University, Ankara, Turkey
James A. Zagzebski / The University of Wisconsin–Madison, Madison, Wisconsin, U.S.A.
Pierre-Yves Zambelli / Hôpital Orthope´dique de la Suisse Romande, Lausanne, Switzerland
Conrad M. Zapanta / Penn State College of Medicine, Hershey, Pennsylvania, U.S.A.
Lyle D. Zardiackas / University of Mississippi Medical Center, Jackson, Mississippi, U.S.A.
Jing Zhang / University of Texas at Austin, Austin, Texas, U.S.A.
Peter Zilla / University of Cape Town Medical School, and Groote Schuur Hospital, Cape Town,
South Africa
Wolfram-Hubertus Zimmermann / University-Hospital Hamburg-Eppendorf, Hamburg, Germany
Contents

Volume 1
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xxix
Adhesives / J. Brock Thomas, Nicholas A. Peppas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Allografts / Kirby S. Black, Patti E. Dawson . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Alumina / Jae Sung Lee . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Angiogenesis Inhibitors / Shaker A. Mousa, Ahmed S. Mousa . . . . . . . . . . . . . . . . . . . . . . . . . . 26
Animal Models for Bone Tissue Engineering / Mark P. Mooney, Michael I. Siegel . . . . . . . . . . . 33
Animal Surrogate Systems for Toxicity Testing / Michael L. Shuler, Gretchen J. McAuliffe,
Daniel A. Tatosian . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
Antimicrobial/Antibiotic (Infection Resistance) Materials / Franck Furno, Roger Bayston . . . . . . . 61
Antimineralization Treatment / Bart Meuris, Willem Flameng . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Articular Cartilage Biomechanics / Jennifer S. Wayne . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Artificial Heart Fluid Dynamics: Positive Displacement Pumps / Steven Deutsch,
John M. Tarbell, Keefe B. Manning, Gerson Rosenberg, Arnold A. Fontaine . . . . . . . . . . . . . . 95
Artificial Muscles / Mohsen Shahinpoor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Artificial Neural Networks: An Overview / Evangelia Micheli-Tzanakou . . . . . . . . . . . . . . . . . . 119
Bioactive Glass / Marivalda M. Pereira, Larry L. Hench . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
Bioactive Materials and Scaffolds for Tissue Engineering / Qi-Zhi Chen, Aldo R. Boccaccini . . . . 142
Bioadhesion / John D. Smart . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152
Biocatalysis / Claire F. Komives, Ruizhen Rachel Chen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162
Biocompatibility Testing / Joel D. Bumgardner, Marcia Vasquez-Lee, Keertik S. Fulzele,
Daniel H. Smith, Kellye D. Branch, Seth I. Christian, David L. Williams . . . . . . . . . . . . . . . . 169
Biocomposites / Huinan Liu, Grace E. Park, Thomas J. Webster . . . . . . . . . . . . . . . . . . . . . . . . 179
Biodegradable Polymers: An Overview / Chih-Chang Chu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
Biofilms / Clive Edwards . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
Biofunctional Polymers / Jennifer L. West . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Biologic and Synthetic Apatites / Besim Ben-Nissan, Racquel LeGeros . . . . . . . . . . . . . . . . . . . . 225
Biological Adhesives from Nature / Lloyd D. Graham . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
Biological Effects of Radiofrequency Electromagnetic Field / Henry Lai . . . . . . . . . . . . . . . . . . . 254
Biomaterials Immune Response / Sandra C. P. Cachinho, John A. Hunt . . . . . . . . . . . . . . . . . . . 262
Biomaterials: Protein–Surface Interactions / Robert A. Latour, Jr. . . . . . . . . . . . . . . . . . . . . . . 270
Biomedical Ethics / Daniel E. Wueste . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
Biomimetic Materials / Jun-ichi Kikuchi, Atsushi Ikeda, Mineo Hashizume . . . . . . . . . . . . . . . . . 294
Biopotential Amplifiers / Albert Lozano-Nieto . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
Bioreactors / Lito C. Mejia, Kent S. Vilendrer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 310
Biorubber/Poly(Glycerol Sebacate) / Yadong Wang . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 327
Biosensors / Jerome S. Schultz . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 335
Biosynthesis and Applications of Alginates / Bernd H. A. Rehm . . . . . . . . . . . . . . . . . . . . . . . . 350
Biphasic Calcium Phosphate (BCP) Bioceramics: Chemical, Physical, and Biological
Properties / Guy Daculsi, Racquel LeGeros . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359
Blood-Material Interactions / Stephen R. Hanson . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 367
Blood Purification / Yukihiko Nose´, Hisashi Okubo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378
xxii
Blood Vessel Mechanics / Ruth J. Okamoto . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 392
Bone Cement / Julie Hasenwinkel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403
Bone Fracture Fixation / Eleftherios Tsiridis, Constantin Schizas . . . . . . . . . . . . . . . . . . . . . . . . 413
Bone–Implant Interface / D. A. Puleo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 423
Bone Plates and Screws, Bioabsorbable / Riitta Suuronen, Christian Lindqvist . . . . . . . . . . . . . . 432
Bone Remodeling / Elisabeth H. Burger, Jenneke Klein-Nulend, Theo H. Smit . . . . . . . . . . . . . . 439
Burn Dressing / Andrew Burd . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
Calcium Phosphate Ceramics: New Generation Produced in Japan / Atsuo Ito, Hajime Ohgushi . . . 461
Cardiac Assist Devices / Keefe B. Manning, Conrad M. Zapanta, John M. Tarbell . . . . . . . . . . . 470
Cardiac Bioelectricity / Clive M. Baumgarten . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 480
Cardiac Catheters / Tommy L. Miller, Juana Maria Alfaro, Deepak Banerjee, Benjamin Lee,
Anthony J. Minisi . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 491
Cardiac Elastography, Full-Field Development / Tomy Varghese, Q. Chen, P. Rahko,
James A. Zagzebski . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 506
Cardiac Output, Basic Measurement Principles / Leslie A. Geddes . . . . . . . . . . . . . . . . . . . . . . . 514
Cardiac Pacemaker / Rakesh K. Pai, Fred M. Kusumoto . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 532
Cardiac Patch Engineering / Michael J. Yost, Robert L. Price, David G. Simpson, Wentao Yan,
Louis Terracio . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 542
Cartilage Regeneration / Nic D. Leipzig, Kyriacos A. Athanasiou . . . . . . . . . . . . . . . . . . . . . . . 552
Cell Culture Assays / P. J. Doherty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 561
Cell-Material Interaction / Aditya Chaubey, Karen J. L. Burg . . . . . . . . . . . . . . . . . . . . . . . . . 568
Centrifugal Blood Pumps / Gerald E. Miller . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
Ceramics / Robert H. Doremus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
Ceramics in Dentistry / Nicolas M. Jedynakiewicz . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 593
Chitosan / M. N. V. Ravi Kumar, S. M. Hudson . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
Chondroitin Sulfates / Nicola Volpi . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618
Collagen / Matthew J. Beckman, Kelly J. Shields, Robert F. Diegelmann . . . . . . . . . . . . . . . . . . 628
Collagen Fixation / Thomas J. Koob . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 639
Collagen Processing / Michael J. Yost, Louis Terracio, Robert L. Price . . . . . . . . . . . . . . . . . . . 652
Composites / Chia-Wei Wang, Ann Marie Sastry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 659
Compression of Digital Biomedical Signals / Tapio Grönfors, Martti Juhola . . . . . . . . . . . . . . . . 677
Computer-Assisted Surgery / Thomas L. Husted, Timothy J. Broderick . . . . . . . . . . . . . . . . . . . 687
Conductive Polymers / Gordon Wallace, Leon Kane-Maguire . . . . . . . . . . . . . . . . . . . . . . . . . . 695
Confocal Microscopy / Denis Semwogerere, Eric R. Weeks . . . . . . . . . . . . . . . . . . . . . . . . . . . . 705
Contact Lenses: Silicone Hydrogels / Paul C. Nicolson, Jürgen Vogt . . . . . . . . . . . . . . . . . . . . . 715
Control of Movement / Dejan B. Popovic´ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 729
Controlled Release / Brent Vernon, Michael Wegner . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 739
Corneal Implants / Traian V. Chirila, Celia R. Hicks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747
Corrosion / Rachel L. Williams . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 754
Cryopreservation of Living Cells / Dayong Gao, J. K. Critser . . . . . . . . . . . . . . . . . . . . . . . . . . 762
Defibrillators / Paul Wang . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772

Volume 2
Degradable Polymer Composites / Debra D. Wright . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 779
Dendrimers for Drug Delivery / Hu Yang, W. John Kao . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789
Dental Biomechanics / A. N. Natali, P. G. Pavan, E. L. Carniel . . . . . . . . . . . . . . . . . . . . . . . . 799
Dental Implants / Nirit Tagger-Green, Eli E. Machtei . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 808
Diamond and Diamond-Like Carbons / James H. Arps, Geoff Dearnaley . . . . . . . . . . . . . . . . . . 822
xxiii
Dielectric Properties of Tissues / Kenneth R. Foster . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 832
Digital Biomedical Signal Acquisition and Processing: Basic Topics / Martin Oswaldo Mendez
Garcia, Luca T. Mainardi . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 843
Digital Biomedical Signal Processing: Advanced Topics / Luca T. Mainardi . . . . . . . . . . . . . . . . 854
Distal Protection Devices / Thomas Tu, Roger Laham . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 870
Drug Delivery, Controlled / M. N. V. Ravi Kumar, A. J. Domb . . . . . . . . . . . . . . . . . . . . . . . . 880
Elastin / William T. Brinkman, Karthik Nagapudi, Elliot L. Chaikof . . . . . . . . . . . . . . . . . . . . . 891
Elastomers, Biodegradable / John J. Stankus, Jianjun Guan, William R. Wagner . . . . . . . . . . . . . 897
Electric Cell-Substrate Impedance Sensing / Philippe Lam . . . . . . . . . . . . . . . . . . . . . . . . . . . . 908
Electrical Impedance Imaging / Keith D. Paulsen, Alex Hartov . . . . . . . . . . . . . . . . . . . . . . . . . 915
Electroactive Polymeric Materials / V. Prasad Shastri . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 926
Electrocardiography / Soumyadipta Acharya, Bruce Taylor . . . . . . . . . . . . . . . . . . . . . . . . . . . . 936
Electrogastrography / Robert M. Stern, Kenneth L. Koch . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 947
Electromyography / Didier Gamet, Odette Fokapu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 956
Electron Microscopy / Robert L. Price, Jeff Davis, Michael J. Yost, Richard L. Goodwin,
Louis Terracio . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 968
Electrospinning / Darrell H. Reneker, Haoqing Hou . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 978
Endoscopy / Jonathan T. C. Liu, Tonya Kaltenbach, Thomas D. Wang, Roy M. Soetikno . . . . . . 986
Ergonomics / Brian N. Craig . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 999
Excitable Tissue, Electrical Stimulation Of / Dominique M. Durand . . . . . . . . . . . . . . . . . . . . . 1009
Extracellular Matrix Scaffolds / Stephen F. Badylak . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1021
Eye Tracking: Characteristics and Methods / Daniel C. Richardson, Michael J. Spivey . . . . . . . . 1028
Eye Tracking: Research Areas and Applications / Daniel C. Richardson, Michael J. Spivey . . . . . 1033
Fabrics / Frank K. Ko . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1043
Fiber Optic Biosensors / Ilko K. Ilev, Israel Gannot . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1062
Fibrin / Sam L. Helgerson, Thomas Seelich, James P. DiOrio, Bill Tawil, Katharina Bittner,
Reiner Spaethe . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1072
Fibrin Sealants / Marcus E. Carr, Jr., Erika J. Martin, Heather Ambrose . . . . . . . . . . . . . . . . . 1080
Finite Element Analysis / Jennifer S. Wayne . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1091
Flow Cytometry / J. Paul Robinson . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1100
Foreign Body Response / John Hunt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1111
Functional MRI: Applications / Hui Mao . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1119
Gait Analysis / Richard Baker . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1129
Gene-Activated Matrix / Neil Davies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1142
Gene Therapy / Viraj P. Mane, William McCormack, Brendan Lee . . . . . . . . . . . . . . . . . . . . . . 1150
Glucose-Responsive Hydrogels / Nicholas A. Peppas, Christie D. Bures . . . . . . . . . . . . . . . . . . . 1163
Glucose Sensors / Jeffrey I. Joseph, Marc J. Torjman . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1174
Glues / Shojiro Matsuda, Yoshito Ikada . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1185
Glycosaminoglycans and Proteoglycans / Jan C. Schagemann, Hsi-Wei Chung,
Eike H. Mrosek, Shawn W. O’Driscoll, Gregory G. Reinholz . . . . . . . . . . . . . . . . . . . . . . . . . 1195
Gradient Surfaces: Preparation, Characterization, and Interactions with Biological
Species / Hai Bang Lee, Moon Suk Kim, Gilson Khang, Jin Ho Lee . . . . . . . . . . . . . . . . . 1210
Hard Tissue: Biomaterial Interactions / Feza Korkusuz, Petek Korkusuz . . . . . . . . . . . . . . . . . . 1229
Hard Tissue Elastic Properties / J. Lawrence Katz . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1236
Healing of Bone and Connective Tissues / William L. Murphy, Kelley Grorud,
Ray Vanderby, Jr. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1249
Hearing Aids / Ian C. Bruce . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1263
Hearing Mechanisms / Martin L. Lenhardt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1271
Heart Biomechanics / Jeffrey W. Holmes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1280
xxiv
Heart Valve, Bioprosthetic / Peter Zilla, Paul Human, Deon Bezuidenhout . . . . . . . . . . . . . . . . . 1291
Heart Valve Failure, Bioprosthetic / Peter Zilla, Paul Human, Deon Bezuidenhout . . . . . . . . . . . 1306
Heart Valve Mechanics / Ajit Yoganathan, Anna M. Fallon, Jorge H. Jimenez . . . . . . . . . . . . . . 1317
Heart Valves, Mechanical / Ajit P. Yoganathan, Zhaoming He, Hwa-Liang Leo, Anna Fallon . . . 1329
Hematopoietic Stem Cells and Assays / Athanassios Mantalaris, Nicki Panoskaltsis,
J. H. David Wu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1338
Hemocompatible Materials / Agnese Magnani, Federica M. Piras . . . . . . . . . . . . . . . . . . . . . . . 1345
Hemodynamics, Macrocirculatory / Baruch B. Lieber, Chander Sadasivan . . . . . . . . . . . . . . . . . 1356
Heparin-Binding to Improve Biocompatibility / Rolf Larsson . . . . . . . . . . . . . . . . . . . . . . . . . . . 1368
Hip Biomechanics / Farid Amirouche, Mark Gonzalez, Luke Aram . . . . . . . . . . . . . . . . . . . . . . 1377
Hip Joint: Overuse Injuries / Marko Pec´ina, Ivan Bojanic´ Alan Ivkovic´ . . . . . . . . . . . . . . . . . . . 1393
Histogenesis / Julie Glowacki . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1403
Host Reactions / Kristin J. Pawlowski . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1411
Hyaluronan / Jennie Baier Leach, Christine E. Schmidt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1421
Hybrid Vascular Prostheses / Jerome A. Werkmeister, Glenn A. Edwards,
John A. M. Ramshaw . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1432
Hydrogels / Junji Watanabe, Yoshihiro Kiritoshi, Kwang Woo Nam, Kazuhiko Ishihara . . . . . . . . 1439
Hydroxyapatite / Alexandra Elizabeth Porter, Nelesh Patel, Serena Best . . . . . . . . . . . . . . . . . . 1451
Hydroxyapatite Coatings / Yunzhi Yang, Kazuhisa Bessho, Kyo-Han Kim, Sang-won Park,
Joo L. Ong . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1464
Implant, Total Hip / Nadim James Hallab, Joshua Jacobs . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1470
Implanted Medical Devices, Power Sources For / Erik R. Scott, Craig L. Schmidt,
Paul M. Skarstad . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1484
Infection of Medical Devices / Lori L. Burrows, Antoine E. Khoury . . . . . . . . . . . . . . . . . . . . . . 1497
Inner Ear Implants / Graeme M. Clark . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1507
Insulin Delivery / Robert S. Parker . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1514
Integrins / David G. Simpson . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1524
Intellectual Property Management / Michael Blakeney . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1540
Interpenetrating Polymeric Networks / Nicholas A. Peppas, Jing Zhang . . . . . . . . . . . . . . . . . . . 1549

Volume 3
Intravascular Ultrasound / Charles D. Choi, Yao Wang, Javier de Ana, Matthew O’Donnell . . . . 1559
In Vivo Cardiovascular Models / B. H. Walpoth, O. M. Hess . . . . . . . . . . . . . . . . . . . . . . . . . . 1566
Kidneys, Artificial / Churn K. Poh, Junfeng Lu, William R. Clark, Dayong Gao . . . . . . . . . . . . . 1576
Knee Joint: Overuse Injuries / Ivan Bojanic´, Alan Ivkovic´, Marko Pec´ina . . . . . . . . . . . . . . . . . . 1597
Knee Joint Replacement / M. J. Cross, E. N. Parish, S. Machan . . . . . . . . . . . . . . . . . . . . . . . 1607
Laboratory Animals: Ethics and Regulations for Care and Use / Kathy E. Laber,
M. Michael Swindle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1616
Laser-Tissue Interaction / E. Duco Jansen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1623
Laser Transmyocardial Revascularization / James W. Jones, Nancy A. Crigger,
Bruce W. Richman . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1632
Ligament Repair: Animal Models / Robert C. Bray, Catherine A. Leonard, Kevin R. Forrester,
Paul T. Salo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1640
Liver, Bio-Artificial / Franc- ois Berthiaume, Christina Chan, Martin L. Yarmush . . . . . . . . . . . . . 1649
Lung, Artificial: Basic Principles and Current Applications / William J. Federspiel,
Kristie A. Henchir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1661
Lung, Artificial: Current Research and Future Directions / William J. Federspiel,
Robert G. Svitek . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1673
Lung Surfactants / Robert H. Notter, Zhengdong Wang . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1683
xxv
Magnetic and Electrophoretic Cell Separation / S. Chethana, Ganapathi Patil,
K. S. M. S. Raghavarao . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1695
Magnetic Nanoparticles: Structure and Bioapplications / Yudhisthira Sahoo,
Edward P. Furlani, Earl J. Bergey . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1705
Magnetic Resonance Imaging (MRI) / Simon J. Doran . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1713
Magnetic Resonance Imaging in Temperature Measurement / Nadine Barrie Smith . . . . . . . . . . . 1728
Magnetic Resonance Microscopy / Egidijus Uzgiris . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1738
Mass Transfer in Tissue Function: Roles / E. N. Lightfoot, Karen A. Duca, . . . . . . . . . . . . . . . 1749
Matrix Metalloproteinases / Todd A. Telemeco, James R. Bowman, III, David G. Simpson . . . . . 1763
Mechanical Circulatory Support / Eric J. Okum, Vigneshwar Kasirajan, Robert S. D. Higgins . . . 1775
Mechano-Regulation of Fibroblast Function / James H.-C. Wang, Bhavani P. Thampatty . . . . . . 1783
Medical Imaging, 3-D / Oskar Skrinjar, Paul J. Benkeser . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1794
Medical Robotics / John E. Speich, Jacob Rosen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1804
Melt Spinning / Eija Marjut Pirhonen, Ville Ellä . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1816
Metallic Orthopedic Devices: Failure Analysis / Lyle D. Zardiackas . . . . . . . . . . . . . . . . . . . . . 1824
Microcirculatory Oxygen Transport / Roland N. Pittman . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1842
Microcomputed Tomography and Its Applications / Thomas Dufresne, Paula Chmielewski,
Babul Borah, Andres Laib . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1850
Microelectromechanical Systems (MEMS) Manufacturing / Marc Madou . . . . . . . . . . . . . . . . . 1860
Micro-Particle Image Velocimetry in Biomedical Applications / Jinhua Cao, Steve T. Wereley . . . 1873
Microporous Materials / Julian R. Jones, Larry L. Hench . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1885
Modeling, Biomedical / Andrew J. Pullan, Nicolas P. Smith . . . . . . . . . . . . . . . . . . . . . . . . . . . 1894
Nanoscale Neurosurgery / William C. Broaddus, Peter J. Haar, George T. Gillies . . . . . . . . . . . . 1903
Nerve Guides / Burkhard Schlosshauer, Martin Lietz . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1911
Neuroprostheses / Milos R. Popovic, T. Adam Thrasher . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1924
Neutrophil-Biomaterial Interactions / Vibeke Videm . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1934
Nickel and Nickel Alloys / Rachel L. Williams . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1942
Nitric Oxide / Matthew J. Bizzarro, Vineet Bhandari . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1951
Nuclear Magnetic Resonance Spectroscopy / Mari A. Smith, Peter B. Barker . . . . . . . . . . . . . . . 1960
Ocular Implants / Andrew W. Lloyd, Susan Sandeman, Richard G. A. Faragher,
Stephen P. Denyer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1974
Ocular Implants for Drug Delivery / Susan S. Lee, Peng Yuan, Michael R. Robinson . . . . . . . . . 1981
Optical Coherence Tomography / Xingde Li, James G. Fujimoto . . . . . . . . . . . . . . . . . . . . . . . . 1996
Optical Detection of Cancers / Xin-Hua Hu, Jun Qing Lu . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2009
Optical Mapping / Igor R. Efimov, Vladimir P. Nikolski, Gil Bub . . . . . . . . . . . . . . . . . . . . . . . 2019
Optics, Biomedical / Sergio Fantini . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2029
Organic and Inorganic Matrices / Adele L. Boskey . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2039
Orthodontic Wires / Eser Tüfekc- i, Steven J. Lindauer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2054
Orthopedic Biomaterials / J. Lawrence Katz, Catherine G. Ambrose, Carl McMillin,
Paulette Spencer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2061
Orthopedic Fixation Devices / Jill S. Kawalec . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2073
Osteoclastic Bone Resorption / Samuel C. Ramage, Mengnai Li, Matthew J. Beckman . . . . . . . . 2081
Osteogenic Progenitor Cells of Bone / Richard O. C. Oreffo . . . . . . . . . . . . . . . . . . . . . . . . . . . 2091
Osteoinductive Substances and Materials / Toshimasa Uemura, Hiroko Kojima, Takashi Saito . . . 2102
Particulate Matter and Host Reactions / Edward M. Greenfield . . . . . . . . . . . . . . . . . . . . . . . . 2110
Peripheral Nerve Repair and Regeneration: Historical Perspective / E. E. Sabelman,
W. Chang, S. Nguyen, J. M. Rosen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2118
Phosphorylcholine (PC) Technology / Andrew L. Lewis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2136
Photopolymerization / Dong-an Wang, Jennifer H. Elisseeff . . . . . . . . . . . . . . . . . . . . . . . . . . . 2150
xxvi
Piezoelectric Materials / Zoubeida Ounaies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2163
Plasma Modification of Materials / Ricky K. Y. Fu, Paul Kim Ho Chu . . . . . . . . . . . . . . . . . . . 2174
Polyamides (Synthetic and Natural) / Naveen C. Srivastav, Babita Agrawal, Rakesh Kumar . . . . . 2186
Polycaprolactone / J. E. Gough, P. Christian . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2197
Poly(carbonate)urethanes / Michael Szycher . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2208
Polyelectrolyte Multilayers / Darwin R. Reyes, Laurie E. Locascio, Michael Gaitan . . . . . . . . . . 2217
Polyepoxy Compound Fixation / Yasuharu Noishiki, Teruo Miyata . . . . . . . . . . . . . . . . . . . . . . 2231
Poly(Glycolic Acid) / Eugene D. Boland, Gary E. Wnek, Gary L. Bowlin . . . . . . . . . . . . . . . . . . 2241
Poly(lactic acid)s / Michel Vert . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2249
Polylactic-Co-Glycolic Acid (PLGA) / Konstantinos Avgoustakis . . . . . . . . . . . . . . . . . . . . . . . . 2259
Polymer Foams / V. Prasad Shastri, Molly M. Stevens . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2270
Polymers / Gary E. Wnek . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2275
Polymers Used in Tissue Engineering / Vasif Hasirci, Deniz Yucel . . . . . . . . . . . . . . . . . . . . . . . 2282
Polytetrafluoroethylene / Robert R. Pavlis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2300
Polyurethane Biomaterials / Joanna D. Fromstein, Kimberly A. Woodhouse . . . . . . . . . . . . . . . . 2304
Positron Emission Tomography (PET) / Anna-Liisa Brownell . . . . . . . . . . . . . . . . . . . . . . . . . . 2314
Protein Adsorption Modeling / Alan H. Goldstein, Alastair N. Cormack . . . . . . . . . . . . . . . . . . . 2326

Volume 4
Protein and Cell Signaling with Biomaterials: Influence of Surfactants / Solomon W. Leung,
James C. K. Lai . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2337
Protein and Cell Signaling with Biomaterials: Interfacial Transport / James C. K. Lai,
Solomon W. Leung . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2346
Protein Delivery Systems / Nicholas A. Peppas, Cristina Donini . . . . . . . . . . . . . . . . . . . . . . . . 2357
Pyrolytic Carbon / Robert B. More, George Sines, Ling Ma, Jack C. Bokros . . . . . . . . . . . . . . . 2370
Rapid Prototyping / Shaochen Chen, Carlos A. Aguilar, Yi Lu . . . . . . . . . . . . . . . . . . . . . . . . . 2382
Real Time Tomographic Reflection / Vikram S. Chib, George D. Stetten . . . . . . . . . . . . . . . . . . 2391
Reconstructive Biomaterials / Mutaz B. Habal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2395
Rotary Cell Culture Systems / Stephen S. Navran . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2402
Self-Assembled Monolayers / Koichi Kato . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2409
Shape Memory Metals / Marie Haı¨dopoulos, Fatiha El Feninat, Diego Mantovani . . . . . . . . . . . 2418
Silicones / Kenneth J. Wynne, James M. Lambert . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2427
Silks / Juming Yao, Tetsuo Asakura . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2442
Small-Angle Light Scattering Methods for Soft Connective Tissue Structural
Analysis / Michael S. Sacks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2450
Spine Biomechanics / Clayton Adam . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2464
Stem Cells / Andrea Liebmann-Vinson, Sharon C. Presnell . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2473
Stent Grafts, Endovascular / Timothy A. M. Chuter . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2480
Stents / Benjamin Thierry, Luc Bilodeau, Maryam Tabrizian . . . . . . . . . . . . . . . . . . . . . . . . . . 2492
Sterilization of Biomedical Materials / Francesco Tessarolo, Giandomenico Nollo . . . . . . . . . . . . 2501
Stress Fractures / Marko Pec´ina, Ivan Bojanic´, Alan Ivkovic´ . . . . . . . . . . . . . . . . . . . . . . . . . . 2511
Supercritical Fluid Processing / Ihtesham U. Rehman, Jawwad A. Darr, Alireza Moshaverinia . . . 2522
Surface Coatings / Shih-Horng Su, Suzanne Conroy, Tung-Liang Lin, Min-Shyan Sheu,
Ih-Houng Loh . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2531
Surface Modification / Sally L. McArthur, Keith M. McLean . . . . . . . . . . . . . . . . . . . . . . . . . . 2540
Surface Topography and Cell Behavior / Douglas W. Hamilton, Salem Ghrebi, Hugh Kim,
Babak Chehroudi, Donald M. Brunette . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2551
Sutures / C. C. Chu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2562
xxvii
Telemedicine / Brett M. Harnett, Charles R. Doarn, Ronald C. Merrell . . . . . . . . . . . . . . . . . . . 2579
Tendons and Ligaments, Mechanical Testing Of / Richard E. Debski, Susan M. Moore,
Eric J. Rainis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2588
Thrombosis / Mukul S. Goel, Scott L. Diamond . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2601
Tinnitus Devices / Martin L. Lenhardt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2610
Tissue Engineering / Franc- ois A. Auger, Lucie Germain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2620
Tissue Engineering: AC Electrokinetics / Gerard H. Markx, Anne-Marie Buckle . . . . . . . . . . . . . 2628
Tissue Engineering of Bladder / Anthony Atala . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2636
Tissue Engineering of Blood Vessel / Douglas Hamilton, David Vorp . . . . . . . . . . . . . . . . . . . . . 2643
Tissue Engineering of Bone / L. Di-Silvio, N. Gurav, Eleftherios Tsiridis . . . . . . . . . . . . . . . . . . 2652
Tissue Engineering of Bone Marrow / Athanassios Mantalaris, Nicki Panoskaltsis,
J. H. David Wu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2660
Tissue Engineering of Bone Marrow, Culture Systems / Athanassios Mantalaris, Nicki
Panoskaltsis, J. H. David Wu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2670
Tissue Engineering of Breast / Cheryl T. Gomillion, Cheryl A. Parzel, Richard L. White, Jr.,
Karen J.L. Burg . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2678
Tissue Engineering of Cardiac Muscle / Wolfram-Hubertus Zimmermann, Thomas Eschenhagen . . . 2686
Tissue Engineering of Cartilage / J. N. Amritha De Croos, Rita A. Kandel . . . . . . . . . . . . . . . . . 2694
Tissue Engineering of Cornea / Lucie Germain, Claude J. Giasson, Patrick Carrier,
Sylvain L. Gue´rin, Christian Salesse, Franc- ois A. Auger . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2707
Tissue Engineering of the Esophagus / Stephen F. Badylak, Thomas W. Gilbert,
Alejandro Nieponice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2719
Tissue Engineering, Fat / Barbara Weiser, Markus Neubauer, Achim Göpferich, Torsten Blunk . . . 2725
Tissue Engineering of Heart Valves / Ivan Vesely . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2737
Tissue Engineering of the Intestine / James C. Y. Dunn, Benjamin M. Wu . . . . . . . . . . . . . . . . . 2751
Tissue Engineering of Ligament / E. Gentleman, G. A. Livesay, K. C. Dee, E. A. Nauman . . . . . . 2758
Tissue Engineering of Liver / Alexander B. G. Sevy, Katherine M. Kulig, Joseph P. Vacanti . . . . 2769
Tissue Engineering, Microscale / Wei Tan, Tejal Desai . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2779
Tissue Engineering of Microvascular Networks / J. T. Borenstein, E. J. Weinberg,
M. R. Kaazempur-Mofrad, Joseph P. Vacanti . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2792
Tissue Engineering of Pancreas / Hiroo Iwata . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2802
Tissue Engineering of Peripheral Nerve / Archit B. Sanghvi, Joy L. Murray,
Christine E. Schmidt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2811
Tissue Engineering of Rotator Cuff Tendons / Jonathan P. Van Kleunen, Louis J. Soslowsky,
David L. Glaser . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2820
Tissue Engineering Scaffolds / Eugene D. Boland, Paul G. Espy, Gary L. Bowlin . . . . . . . . . . . . 2828
Tissue Engineering of Skeletal Muscle / Christopher Keen, Daniel Newton, Todd A. Telemeco,
Gary L. Bowlin, Gary E. Wnek, Clive M. Baumgarten, David G. Simpson . . . . . . . . . . . . . . . 2838
Tissue Engineering of Skin / Ricardo R. Brau, Ioannis V. Yannas . . . . . . . . . . . . . . . . . . . . . . . 2851
Tissue Engineering of the Small Intestine / Simon M. Gabe, Richard M. Day, Aldo Boccaccini . . . 2860
Tissue Engineering of Tendons / Dominique P. Pioletti, Olivier Siegrist, Pierre-Yves Zambelli . . . 2871
Titanium and Its Alloys / Mitsuo Niinomi, Tomokazu Hattori, Toshihiro Kasuga, Hisao Fukui . . . . 2876
Transdermal Drug Delivery / Brad Phipps, Michel Cormier, Bob Gale, Bill van Osdol,
Jay Audett, Rama Padmanabhan, Peter Daddona . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2893
Transgenic and Gene-Targeted Organs / Brenda M. Ogle, Jeffrey L. Platt . . . . . . . . . . . . . . . . . 2906
Transparent Ceramics: Bioactive Materials and Tissue Engineering / Motohiro Hirose, Koji
Ioku, Noriko Kotobuki, Daisuke Kawagoe, Hajime Ohgushi . . . . . . . . . . . . . . . . . . . . . . . . . . 2916
Ultra-High Molecular Weight Polyethylene (UHMWPE) / Kwideok Park, Gladius Lewis,
Joon B. Park . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2925
Ultrasound and Tissue Interaction / K. Kirk Shung . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2933
xxviii
Ultrasound Doppler / Ding-Yu Fei, James A. Arrowood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2942
Ultrasound Therapy, Bone Healing / Özgür Erdoğan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2951
Urinary Prostheses / Daniel Eberli, Anthony Atala . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2961
Vascular Fluid Dynamics / Stanley A. Berger . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2971
Vascular Grafts / Deon Bezuidenhout, Peter Zilla . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2985
Vascular Grafts: Development Strategies / James Laredo, Lian Xue, Howard P. Greisler . . . . . . . 2997
Vascular Grafts: Host Interactions / James Laredo, Lian Xue, Howard P. Greisler . . . . . . . . . . . 3006
Vascular Imaging / Tobias Saam, William S. Kerwin, Chun Yuan . . . . . . . . . . . . . . . . . . . . . . . 3012
Vascularization of Engineered Constructs / Zerina Lokmic, Rejhan Idrizi, Aurora Messina,
Kenneth R. Knight, Wayne Morrison, Geraldine Mitchell . . . . . . . . . . . . . . . . . . . . . . . . . . . 3022
Vascular Tissue Engineering / Alejandro Nieponice, Timothy M. Maul, Lorenzo Soletti,
David A. Vorp . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3033
Vitrification / Kelvin G. M. Brockbank, John R. Walsh, Ying C. Song, Michael J. Taylor . . . . . . 3046
Wear Debris / John Fisher, Eileen Ingham . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3058
Wear Debris, Bone Resorption Animal / Xiaochao Wei, Regis J. O’Keefe, Edward M. Schwarz . . 3067
Wear Debris from Joint Arthroplasties, Biological Effects Of / Ting Ma, Stuart B. Goodman . . . 3076
Wheeled Mobility: Wheelchairs and Personal Transportation / Jon L. Pearlman,
Rory A. Cooper . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3083
Xenografts / Brenda M. Ogle, Jeffrey L. Platt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3093
Zirconia Ceramics / Mariano A. Polack . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3104
Preface

The field of biomedical engineering continues to apply engineering principles to analyze


and solve problems in life sciences and medicine, leading to improvements in the quality of
life. Biomaterials represent a central theme in a majority of the problems encountered.
These fields have evolved into a very interdisciplinary arena building on traditional
engineering principles that bridge advances in the areas of materials science, life sciences,
nanotechnology, and cell biology, to name a few. This trend of interdisciplinary research to
solve the most challenging yet compelling medical problems has been embraced in the field
and is leading to the betterment of human health. It is evident that the fields of bio-
materials and biomedical engineering are continually changing due to the rapid creation
and advancement in technology in more traditional areas as well as rapidly developing
areas (e.g., tissue engineering).
Because the scope of these medical problems is both vast and dynamic, there is a clear
need for a source of technical information that has broad coverage, is current, is written at
a level accessible to nonexperts, and has the ability to be updated on a regular basis. The
Encyclopedia of Biomaterials and Biomedical Engineering (EBBE) was born with these
considerations in mind in 2001, and its first edition appeared in 2004. Due to the success of
that endeavor and the privilege of working with experts worldwide in preparing the second
edition of the EBBE, it has blossomed into an expansive array of 298 entries. It is our hope
that we have continued to present the material in such a fashion that it conveys a balanced
overview to help stimulate further advancements in all areas of biomaterials and bio-
medical engineering.
We also believe that many thanks are in order for the efforts that led to the successful
first edition and the generation of the second edition of the EBBE. We thank all the
authors who have enthusiastically provided their time and expertise, and we also thank the
editorial board for all of their various contributions during the development of the EBBE.
Finally, we are grateful to Informa Healthcare for acquiring the EBBE, making it a
priority, and providing the excellent staff needed to ensure its continued success.
It is our continued hope that the EBBE will serve as an important and continual
resource for the many individuals whose work touches, and is touched by, biomaterials
and biomedical engineering in order to further stimulate, create, and deliver improvements
in quality of life.

Gary E. Wnek
Gary L. Bowlin
This page intentionally left blank
Encyclopedia of
BIOMATERIALS AND
BIOMEDICAL ENGINEERING
This page intentionally left blank
Adhesives
A
J. Brock Thomas
Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, U.S.A.
Nicholas A. Peppas
Departments of Chemical and Biomedical Engineering and Division of Pharmaceutics,
University of Texas at Austin, Austin, Texas, U.S.A.

INTRODUCTION synthetic materials exhibiting molecular structures


recognized by specific cellular components has the
An adhesive broadly refers to any substance that ability to afford better cell attachment. Increased cell
possesses the ability to hold two other entities together. attachment would lead to a more favorable biological
Historically, adhesives and bioadhesives have referred response effectively creating more biocompatible
to distinctly different areas of thought with very little scaffolds. On the other hand, with a complete under-
overlap and continuity. The term adhesive has been standing of the biofilm formation process, biomaterials
used to describe the particular biomaterial or system can be designed so as to decrease the propensity of
used that is responsible for adherence. Bioadhesion has microbial attachment and colonization. Here we pro-
largely remained well entrained in the field of biology vide a broad overview of the molecular and physico-
referring to cellular interactions (cytoadhesion) or to chemical characteristics involved during the formation
mucosal adhesion (mucoadhesion). Whether it is cell- of bioadhesive bonds along with a short critical review
to-cell, soft or hard tissue, or mucosal adhesion, the of the progress achieved in each of the distinct thrusts
phenomenon of bioadhesion involves the formation of of current research—hard-tissue adhesives, soft-tissue
a bond between two biological surfaces or a biological adhesives, and cytoadhesives.
surface and a synthetic surface. Therefore, bioadhesion
can be extended to include any type of adhesion pro-
cess in contact with or within the biological milieu. The MECHANISMS OF BIOADHESION
term bioadhesive refers to the particular substance,
natural or synthetic, responsible for this adhesion. Bioadhesion is truly an interfacial phenomenon and
Mucoadhesion is often used to describe bioadhesion only differs from conventional adhesion in the special
involving mucus, whereas cytoadhesion refers to cell- properties and characteristics of the substrate(s) being
to-cell bioadhesion. adhered. The mechanistic and structural analyses of
Common examples of bioadhesives include dental the phenomenon of bioadhesion have been carefully
restoration fixatives, skin adhesives, and bacterial outlined in several comprehensive reviews.[1–5] The
colonization. Much success has been achieved in hard- mechanisms of bioadhesion are often classified into
tissue adhesives with the availability of many com- chemical and physical mechanisms with the electronic
mercially available bioadhesive systems. Mucoadhesive theory and adsorption theory falling under the former
materials have been the focus of much research over mechanism, and wetting, interpenetration or diffusion,
the last couple of decades in pharmaceutical science and fracture theory falling under the latter.
research as a novel drug-delivery approach. With Electronic theory describes adhesion between two
mucoadhesive drug-delivery systems, the therapeutic different electronic structures such as a polymer and a
agent can be localized at the desired site of delivery. glycoprotein network. Upon contact of these two sys-
Peptides, proteins, or actively facilitated compounds tems, electron transfer occurs leading to a double layer
can be targeted to the upper small intestine where of electrical charge at the interface. Adsorption theory
achieving greater absorption is possible. Similar obs- describes adhesion that occurs owing to secondary
tacles can be overcome in other locations of the body, forces such as van der Waals, hydrogen bonding,
opthalmic, pulmonary, and vaginal cavities, where etc. Several occurrences of these types of forces are
mucus is present. sufficient to create a substantial adhesive bond.
The desire to create biomimetic materials has cre- The wetting theory uses a surface energy analysis to
ated interest in further understanding the mechanisms determine to what extent an adhesive or biological
involved with cellular adhesion to aid in engineering surface is capable of spreading over its corresponding
effective tissue-engineering scaffolds and biomaterials substrate to develop intimate contact. Interpenetration
resistant to biofilm formation. The ability to create theory, or diffusion theory, (Fig. 1) relies on the
Encyclopedia of Biomaterials and Biomedical Engineering DOI:10.3109/E-EBBE-120007250
Copyright r 2008 by Informa Healthcare USA, Inc. All rights reserved. 1
2 Adhesives

ESPETM DurelonTM, Caulk Tylok Plus) provided


advances in adhesion owing to the presence of
calcium–carboxylate ion interactions. However, a
major drawback to the polycarboxylate adhesives is
their poor mechanical properties. To overcome this
negative attribute and achieve better optical properties,
glass ionomer cements (GIC, GC Fuji PlusTM, and
ESPE Ketac-Cems) containing polycarboxylic acid
and aluminosilicate glass were developed.[9] These
systems were further modified to include chemical
polymerizable (Caulk Advance, 3MTM ESPETM
VitremerTM Luting), light-cured (Fuji II LC), or dual-
cured (Bisco Resinomer, Dentsply Enforce) resins,
which overcome issues of drying and limited fracture
resistance with the original GIC. Resins used alone
Fig. 1 Upon contact, the interface between two polymer (Panavia 21, J. Morita Clearfil DC Cement) have been
systems disappears and chain interpenetration or diffusion
used as adhesives that are less brittle and more fracture
occurs. This polymer interpenetration and depth is
responsible for the bioadhesion that occurs.
resistant than the originally employed zinc phosphate
cements. Mechanical interlocking and electrostatic
interactions are responsible for adhesion and rigidity
of the material. Zinc-containing materials do not
penetration depth of the polymer chains into the bio-
possess the desired optical properties, therefore
logical substrate to be sufficient enough to create a
necessitating glass ionomer systems (Fujis), which are
semipermanent adhesive bond. Finally, fracture theory
translucent.
describes adhesion as the difficulty of separation of the
Methyl methacrylate–styrene copolymer-based bone
two surfaces. In most systems, one theory alone is not
cements (SimplexTM P) have been widely used for
sufficient to describe the process of adhesion. Usually,
securing orthopedic implants to bone since the early
two or more theories more accurately portray the
1970s. Cement blends containing antibiotics such as
phenomenon that is occurring. In designing a superior
tobramycin have recently been approved for use. Fast-
bioadhesive, one must successfully employ key attri-
setting carbonated apatite (hydroxyapatite) cements
butes of these theories to develop effective bioadhesion.
(Norians SRSs) have recently been introduced to
serve as a more biocompatible material for restoration
Hard-Tissue Adhesives or augmentation of the skeletal system. These materials
harden to from dahllite, a complex of naturally
Smith[6] offers a comprehensive review on the advances, occurring apatite found in bones, which resembles
recent trends, and future direction of hard-tissue the mineral phase of new bone growth. This further
adhesives. A great deal of interest in finding alternatives remodels to bone through osteoclastic resorption and
to mechanical fixatives such as screws and pins has new bone formation. Owing to the inorganic com-
prompted the development of efficient adhesives for position similarities between bone and enamel and
skeletal and hard-tissue bonding. This area of research dentin, similar types of materials for dental restoration
has had tremendous success in developing and have also been applied as bone graft substitutes.
employing bioadhesives. Dental adhesives and sealants, Effective bioadhesives have found success in the areas
bone cements, and bone graft substitutes are areas of hard-tissue adhesives as evident by the large number
where significant progress has been achieved. One key of commercially available products in this area.
reason to achieving clinical success has been the
environment and tissue involved in these particular
bioadhesive processes. Great advances have been Soft-Tissue Adhesives
achieved in bone cements, dental cements, and enamel
and dentin adhesives—thanks in great deal to the en- Skin adhesives and tissue sealants have been the
gineering of polycarboxylate and ionomer materials traditional application of soft-tissue adhesives. Pres-
capable of achieving adhesion in these applications.[7,8] sure sensitive adhesives (PSA) have found use in the
Zinc phosphate cements containing Zn and Al attachment of medical appliances and to aid in the
(Caulk Tenacin, Mizzy, Inc. Fleck’s Cement) possess a healing process of abrasions (Band-Aids). These
high compressive strength and have been widely used bandages typically have a high tack layer composed of
since the mid-20th century in dental restoration. an adhesive derived from polyisobutylenes, acrylates,
Addition of polycarboxylates to zinc oxide (3MTM or silicone polymers. Recent advances have led to the
Adhesives 3

development of bandage systems that are more flexible


with greater adhesive capacity and able to resist water,
bacteria, and exogenous material. Semipermeable
A
membranes have been utilized in conjunction with
biological fluid absorbent materials in hydrocolloid
moisture-retentive wound dressings to promote the
wound healing process (ConvaTec). Cyanoacrylates
have been used as tissue adhesives owing to their
ability to polymerize upon contact with fluids present
at biological surfaces. Longer alkyl chain cyanoacry-
lates (2-octyl cyanoacrylate, Dermabonds) have be-
come acceptable bioadhesives owing to their slower
degradation, which provides a lower concentration of
potentially toxic degradation by-products that could
elicit an inflammatory response. Fibrin sealants have
been used in wound healing, hemostasis, and tissue
sealing and receive a biologically favorable response
because of their biomimesis of the last stages of
thrombosis (Baxter Tisseels, Ethicon CrossealTM).
The fibrin-sealant systems consist of two separate
containers of fibrinogen and thrombin and, when
combined and applied to the treatment area, are cap-
able of thrombus formation. The elastic clot formed
adheres firmly to the bleeding site and is transparent
for visual inspection. Albumin glues (Cryolifes Bio-
Glues Surgical Adhesive) have been applied at sites of
surgery to control bleeding. This two-component ad-
hesive system is composed of bovine serum albumin
(BSA) and glutaraldehyde, and when mixed the glu-
taraldehyde cross-links the BSA molecules.
A shift in focus of soft-tissue adhesives has occurred
over the last several decades to evaluate their potential
use in bioadhesive controlled drug-delivery devices.
Having the ability to prolong a therapeutic’s residence
time at a specific site of delivery will greatly enhance
the drug absorption process. These mucoadhesive
devices can protect the drug during the absorption
process in addition to protecting it on its route to
the delivery site.[10,11] Three major categories of poly-
mers have been used with some success as bio-
adhesives: hydroxyl-containing, carboxyl-containing, Fig. 2 Chemical structure of commonly employed
and charged polymers. Proposed mucoadhesives have poly(acrylic acid) (PAA)-based materials in mucoadhesive
been applied in the form of viscous liquids or pastes drug-delivery systems. 934P is cross-linked with allyl
when used to adhere a polymer to a tissue, or as sucrose, 974P with allyl pentaerythritol, and AA-1 with
films or microparticles when used in the form of divinyl glycol. A small amount of the acrylic acids are
controlled-release systems adhering to the mucus. neutralized to potassium acrylate prior to polymerization.
The two basic macromolecular structures that have
received considerable attention are polyacrylic acid
(Noveons Carbopols) and chitosan (NovamatrixTM benzene. Carbopol 971P, 974P, and 71G all possess the
ProtasanTM). same basic molecular structure, with the cross-linking
Carbopols (Fig. 2) are cross-linked polyacrylic acid density being the only difference between 971P and
microparticles that are capable of swelling upon neu- 974P. Carbopol 71G is simply compressed granules of
tralization. Five specific grades, 934P, 971P, 974P, 971P. Polycarbophil AA-1 differs in that the polymer is
71G, and Polycarbophil AA-1, have found wide cross-linked with divinyl glycol. All the later micro-
application in the scientific literature. Carbopol 934P is particles are polymerized in ethyl acetate, a benign
not widely accepted owing to its polymerization in solvent as compared to benzene. Chitosan is a linear
4 Adhesives

cationic polymer composed of D-glucosamine and N- drug delivery. Hydrogels are hydrophilic polymer
acetyl-D-glucosamine units, with the amount of either networks capable of imbibing large amounts of water.
depending on the percent deacetylation. Chitosan is However, they are insoluble because of the presence
derived from chitin, which originates from shellfish. of physical or chemical cross-links, entanglements, or
These polymers along with sodium alginate and crystalline regions. Hydrogels can be used in bio-
cellulose derivatives compose the first generation of medical applications such as drug-delivery systems,
bioadhesives owing to their ability to interact non- biosensors, contact lenses, catheters, and wound
specifically with tissue surfaces. dressings.[14] Because of the presence of certain func-
Studies show that upon application of a polymer to tional groups along the polymer chains, hydrogels are
a tissue, a process of swelling may occur.[12] Shortly often sensitive to the conditions of the surrounding
after the beginning of swelling, adhesion does occur environment.[15] For example, the swelling ratio of
but the adhesive strength is not very high. The effect these materials may be sensitive to the temperature,
of the swelling time on bioadhesion can be readily pH, or ionic strength of the swelling agent or even to
calculated by examining the time required for inter- the presence of a magnetic field or ultraviolet light
penetration (diffusion) of bioadhesive chains into the (Fig. 4).
tissue in a layer of 150–200 Å. The major emphasis Numerous experimental studies have been done to
of recent research on mucoadhesive biomaterials is test the mucoadhesive ability of various polymers.[16,17]
on the use of adhesion promoters, which would en- Among the general rules for mucin-attractive polymers
hance the adhesion between synthetic polymers and are the requirements of hydrogen-bond formation
mucus through additional polymer–biopolymer inter- ability. This is reasonable based on the chemical
penetration (Fig. 3). The use of adhesion promoters structure of mucin components.[18,19] including
such as linear or tethered polymer chains is a natural hydroxyl groups in the branched sugar chains, carb-
result of the diffusional characteristics of adhesion.[13] oxylic, acid and sulfate groups in the terminal segments
We have focused our attention on the enhancement of branched chains. Consequently, polymers such as
of mucoadhesion by tethered chains of poly(ethylene PAA are mucoadhesive; their interactions with mucus
glycol) (PEG), grafted on a polymer backbone. have been tested and the data have been widely
Mucoadhesion depends largely on the structure of cited.[20,21]
the synthetic polymer gels used in controlled release The very interaction between polymers and mucins
applications. Therefore, on the macroscopic level, we in the physiological environment requires more careful
focus on examining the effects of various preparation investigation, both theoretically and experimentally.
conditions on the structure and morphology of PAA The excess amount of water in the physiological en-
and related hydrogels. vironment (the concentration of water in the bulk
The fields of biomaterials and drug delivery have phase is around 55 M) has important implications on
evoked considerable interest and research activity in the hydrogen-binding interaction between polymer
the past 25 yr. Only in the last decade have modulated chains. For example, in aqueous solutions of PEG, the
release systems been considered for their potential polymer chains are surrounded by a hydration layer,
commercial application. Prominent among these with two to three water molecules/monomer unit.[22]
systems are ionic networks and gels used as carriers for Thus, the hydrogen-bond formation between PEG and
another polymer leads to the breakage of PEG–water

Fig. 4 Network structural changes owing to variations on


environmental pH. Higher pHs disrupt the interpolymer
Fig. 3 The free polymer chains across the hydrogel–mucus complexes and ionic moieties deprotonize leading to
interface act as adhesion promoters. The free chains are extensive swelling (left). As pH is decreased, interactions
initially in the hydrogels. When placed in contact, the between the tethered grafts with the protonized ionic
chemical potential gradient causes mutual diffusion of moieties increase leading to the formation of interpolymer
chains across the interface. complexes.
Adhesives 5

hydrogen bond, and this can only occur if there is a net


energy decrease. For example, the interaction between
PEG and poly(vinyl alcohol) (PVA) is repulsive in
A
water, though such polymers can form hydrogen-bond
complex in the dry state.[23,24] Recent studies of the
interaction between PEG and poly(methacrylic acid)
(PMAA) in water[25] showed that the PEG–PMAA
complexation diminished even when only 10% of the
acid groups in PMAA were dissociated. Therefore,
more careful analysis beyond simple consideration of
the chemical structure is needed to determine whether
there is attraction between polymers and mucins. From
an experimental point of view, the current methods for
mucoadhesion measurements do not provide direct
evidence of the nature of polymer–mucin interactions.
These methods mostly exploit homemade or modified
equipment, and there is no universally accepted
test method.[26,27] This is the reason that so few com-
parisons of intergroup data have been made, and, ex- Fig. 5 Bioadhesion measurements using a tensiometric
cept for a small number of cases, there is no consensus device such as an Instron. The adhesive is placed in contact
about which polymers are good mucoadhesives.[26,27] with the substrate to be adhered. The adhesive is removed
Moreover, different test methods involve various while measuring the force vs. elongation.
macroscopic phenomena, which are not directly related
to polymer–mucin interactions but are method- or
sample-dependent. and mucin from the current macroscopic test methods.[29]
Determination of adhesive polymer properties Therefore, despite the existence of numerous experi-
can be evaluated by using a range of experimental mental data, we still do not have enough knowledge to
techniques, leading to the determination of the bond molecularly design the next generation of mucoadhesive
strength. In vitro techniques include mostly tests by devices. We have exploited both theoretical modeling
which one can follow the destruction of the adhesive and microscopic measurement methods to provide a
bond between a polymer and the mucus or tissue clearer picture of polymer–mucus interactions.
predominantly via application of shear or peeling
forces. Often, the same tests are performed under
application of tensile forces. Cytoadhesives
For certain controlled-release systems, there are
simple experimental procedures, which can be used to The adhesion of cells has been described as a four-step
determine the adhesive force, using thin disks. The process: cell attachment, cell spreading, organization
experiment is carried out in a modified tensile tester
using appropriate cells attached to the vertical bars of
the tester. The disks or paste are placed between the
two plates of the cells and a tensile force is applied at
constant extension ratio and until the material breaks
by molecular slippage (Figs. 5 and 6). The maximum
elastic modulus is usually proportional to the adhesive
strength.
The Wilhelmy plate method has been used to test the
mucoadhesion of water-soluble polymers.[20] In this
method, the polymer-coated glass plate was tested
against mucus. However, the contact time (the hydration
time) turned out to be the essential parameter
for the result analysis: too short time would cause in-
sufficient interaction between polymers and mucus.[16]
These disadvantages are expected to cause poor
correlation with the in vivo interaction.[28] More Fig. 6 Force vs. displacement curve for bioadhesives in
importantly, it is barely possible to get an insight into contact with a biological substrate. (A) Represents the peak
molecular-level interactions between various polymers force, where (B) and (C) represent tissue detachment.
6 Adhesives

of an actin cytoskeleton, and formation of focal ad- ionizable groups, and interpenetrating structures seems
hesions.[30] Cellular adhesion is generally promoted to promote interdiffusion and therefore adhesion.
through the presence of various adhesion proteins,[31]
with fibronectin being one of the more widely recog-
nized proteins. Platelet adhesion is an integral step in
blood coagulation and blood-material interactions.[32] ARTICLES OF FURTHER INTEREST
Coagulation arises from interactions of platelet glyco-
protein Ib and exposed connective tissues such as Bioadhesion; Biofilms; Biological Adhesives from Na-
collagen, with plasma von Willebrand factor (vWF) as ture; Bone Cement; Fibrin; Fibrin Sealants; Glues; Sur-
a cofactor. Material platelet adhesion involves other face Coatings
platelet glycoproteins (IIb/IIIa) along with the coagu-
lation mechanism. This material–platelet adhesion
arises from IIb–IIIa interactions with plasma proteins REFERENCES
such as fibrinogen, vWF, fibronectin, and vitronectin.
Integrins, the family of cell-surface matrix receptors, 1. Peppas, N.A.; Buri, P.A. Surface, interface, and
serve as the mediators for cell adhesion. Arg-Gly-Asp molecular aspects of polymer bioadhesion on soft tissues.
J. Controlled Release 1985, 2, 257–275.
(RGD) has often been employed on synthetic surfaces
2. Hubbell, J.A. Biomaterials in tissue engineering. Bio-
to stimulate adhesion in tissue-engineering applications technology 1995, 13, 565–576.
so that further processes such as proliferation and 3. Peppas, N.A.; Sahlin, J.J. Hydrogels as mucoadhesive
migration will occur. This peptide attachment simu- and bioadhesive materials: a review. Biomaterials 1996,
lates the components of the extra cellular matrix 17, 1553–1561.
(ECM), thereby imparting biomimetic properties on 4. Mathiowitz, C.; Chickering, D.E.; Lehr, C.M. Bio-
the biomaterial. Aside from integrin-surface receptors, adhesive Drug Delivery Systems; Marcel Dekker: New
proteoglycans are also responsible for cell adhesion. York, 1999.
Carbohydrate-rich domains located at the cell’s surface 5. Lee, J.W.; Park, J.H.; Robinson, J.R. Bioadhesive-
have the ability to interact with carbohydrate-binding based dosage forms: the next generation. J. Pharm. Sci.
lectin macromolecules.[33] 2000, 89, 850–866.
6. Smith, D.C. Adhesives and sealants. In Biomaterials
Adhesion of bacteria (biofilm formation or bio-
Science: An Introduction to Materials in Medicine;
fouling) involves the interaction of bacterial fibers with Ratner, B.D, Hoffman, A.S., Schoen, F.J., Lemons,
fibers adsorbed to the surface being colonized.[34] J.E., Eds.; Elsevier Academic Press: San Diego, 2004;
Bacteria have often been overly simplified and de- 572–583.
scribed as a smooth colloidal particle so that DLVO 7. Scranton, A.B.; Rangarajan, B.; Klier, J. Biomedical
theory could be used, at least partially, to explain applications of polyelectrolytes. Biopolymers II Adv.
the adhesion process.[35,36] Biofouling of biomaterials Polym. Sci. 1995, 122, 1–54.
occurs owing to non-specific interactions of the bac- 8. Davidson, C.L.; Feilzer, A.J. Polymerization shrinkage
terial surface dependent on polymer conformation and and polymerization shrinkage stress in polymer-based
surface energy whereas certain bacteria (e.g., Helico- restoratives. J. Dentistry 1997, 25, 435–440.
bacter pylori) possess a surface layer of biopolymers 9. Smith, D.C. Development of glass-ionomer cement
systems. Biomaterials 1998, 19, 467–478.
capable of specific interactions with a biological
10. Lowman, A.M.; Peppas, N.A.; Morishita, M.; Nagai,
host.[37] This specific interaction is typically mediated T. Novel bioadhesive complexation networks for
by carbohydrate-binding moieties present on the bac- oral protein drug delivery. In Tailored Polymeric
terium’s surface. Bacterial colonization of the gastro- Materials for Controlled Delivery Systems; McCulloch,
intestinal tract is a result of the ability of certain I., Shalaby, S.W., Eds.; ACS Symposium Series:
bacteria’s surface binding to mucin molecules present Washington, DC, 1999; Vol. 709, 156–164.
in the mucus gel layer. 11. Lowman, A.M.; Peppas, N.A. Solute transport analysis in
pH-responsive, complexing hydrogels of poly(methacrylic
acid-g-ethylene glycol). J. Biomat. Sci. Polym. Ed. 1999,
CONCLUSIONS 10, 999–1009.
12. Mortazavi, S.A.; Smart, J.D. An investigation into
the role of water-movement and mucus gel dehydra-
Bioadhesive polymers can be used in a wide range
tion in mucoadhesion. J. Controlled Release 1993, 25,
of medical applications including cytoadhesive, muco- 197–203.
adhesive, and tissue adhesive uses. Advances in nano- 13. Peppas, N.A.; Bures, P.; Foss, A.; Huang, Y.;
structure development and nanotechnology allow us to Leobandung, W. Molecular aspects of muco- and
utilize a wide range of materials for such applications. bioadhesion: tethered structures and site-specific sur-
While PAA-based, hydrophilic polymers are excellent faces. Proceed. Intern. Symp. Recent Adv. Drug Deliv.
bioadhesive candidates, addition of tethered structures, Sys. 1999, 9, 31–34.
Adhesives 7

14. Peppas, N.A.; Brannon-Peppas, L. Drug delivery bio- 28. Lehr, C.M. Bioadhesion technologies for the delivery
materials. In Encylopedia of Materials: Science and
Technology; Elsevier: Amsterdam, 2001; 2351–2355.
of peptide and protein drugs to the gastrointestinal
tract. CRC Crit. Rev. Ther. Drug Carrier Syst. 1994,
A
15. Peppas, N.A.; Bures, P.; Leobandung, L.; Ichikawa, H. 11, 119–160.
Hydrogels in pharmaceutical formulations. Eur. J. 29. Schneider, J.; Tirrell, M. Direct measurement of mo-
Pharm. Biopharm. 2000, 50, 27–46. lecular-level forces and adhesion in biological systems.
16. Gu, J.M.; Robinson, J.R.; Leung, S.S. Binding of acrylic In Bioadhesive Drug Delivery Systems; Mathiowitz, E.,
polymers to mucin/epithelial surfaces: structure-property Chickering, D.E., Lehr, C.M., Eds.; Marcel Dekker,
relationships. CRC Crit. Rev. Ther. Drug Carrier Syst. Inc.: New York, 1999; 223–259.
1988, 5, 21–67. 30. Drotleff, S.; Lungwitz, U.; Breunig, M.; Dennis, A.;
17. Peppas, N.A.; Huang, Y.; Torres-Lugo, M.; Ward, Blunk, A.; Blunk, T.; Tessmar, J.; Gopferich, A.
J.H.; Zhang, J. Physicochemical foundations and Biomimetic polymers in pharmaceutical and bio-
structural design of hydrogels in medicine and biology. medical sciences. Eur. J. Pharm. Biopharm. 2004, 58,
Ann. Revs. Biomed. Eng. 2000, 2, 9–29. 385–407.
18. Bansil, R.; Stanley, E.; LaMont, J.T. Mucin biophysics. 31. Horbett, T.A. The role of adsorbed proteins in tissue
Ann. Rev. Physiol. 1995, 57, 635–657. response to biomaterials. In Biomaterials Science: An
19. Strous, G.J.; Dekker, J. Mucin-type glycoproteins. Introduction to Materials in Medicine; Ratner, B.D,
Crit. Rev. Biochem. Mol. Biology 1992, 27, 57–92. Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.;
20. Smart, J.D.; Kellaway, I.W.; Worthington, H.E.C. An Elsevier Academic Press: San Diego, 2004; 237–245.
in vitro investigation of mucosa-adhesive materials for 32. Hanson, S.R. Blood coagulation and blood-material
use in controlled drug delivery. J. Pharm. Pharmacol. interactions. In Biomaterials Science: An Introduction to
1984, 36, 295–299. Materials in Medicine; Ratner, B.D, Hoffman, A.S.,
21. Bures, P.; Peppas, N.A. Structural and morphological Schoen, F.J., Lemons, J.E., Eds.; Elsevier Academic
characteristics of carriers based on poly(acrylic acid). Press: San Diego, 2004; 332–338.
Polym. Prepr. 1999, 40 (1), 345–346. 33. Bies, C.; Lehr, C.M.; Woodley, J.F. Lectin-mediated
22. Kjellander, R.; Florin, E. Water structure and changes drug targeting: history and applications. Adv. Drug.
in thermal stability of the system poly(ethylene glycol)- Del. Rev. 2004, 56, 425–435.
water. J. Chem. Soc. Faraday Trans I 1981, 77, 34. Costerton, B.; Cook, G.; Shirtliff, M.; Stoodley, P.;
2053–2077. Pasmore, M. Biofilms, biomaterials, and device-related
23. Inamura, I.; Jinbo, Y. Interaction between poly(vinyl infections. In Biomaterials Science: An Introduction to
alcohol) and poly(ethylene glycol) in water studied by Materials in Medicine; Ratner, B.D, Hoffman, A.S.,
viscosity and density. Polym. J. 1991, 23, 1143–1147. Schoen, F.J., Lemons, J.E., Eds.; Elsevier Academic
24. Sawatari, C.; Kondo, T. Interchain hydrogen bonds in Press: San Diego, 2004; 345–354.
blend films of poly(vinyl alcohol) and its derivatives 35. Xu, L.C.; Vadillo-Rodriguez, V.; Logan, B.E. Resi-
with poly(ethylene oxide). Macromolecules 1999, 32, dence time, loading force, pH, and ionic strength affect
1949–1955. adhesion forces between colloids and biopolymer-
25. Zeghal, M.; Auvray, L. Structure of polymer complex coated surfaces. Langmuir 2005, 21, 7491–7500.
in water. Europhys. Lett. 1999, 45, 482–487. 36. Abu-Lail, N.I.; Camesano, T.A. Role of ionic strength
26. Tobyn, M.J.; Johnson, J.R.; Dettmar, P.W. Factors on the relationship of biopolymer conformation,
affecting in vitro gastric mucoadhesion I. Test con- DLVO contributions, and steric interactions to bio-
ditions and instrumental parameters. Eur. J. Pharm. adhesion of Pseudomonas putida KT2442. Biomacro-
Biopharm. 1995, 41, 233–241. molecules 2003, 4, 1000–1012.
27. Tobyn, M.J.; Johnson, J.R.; Dettmar, P.W. Factors 37. Walz, A.; Odenbreit, S.; Mahdavi, J.; Boren, T.; Ruhl,
affecting in vitro gastric mucoadhesion II. Physical S. Identification and characterization of binding prop-
properties of polymers. Eur. J. Pharm. Biopharm. 1996, erties of Helicobacter pylori by glycoconjugate arrays.
42, 56–61. Glycobiology 2005, 15, 700–708.
References

Adhesives

14. Peppas, N.A.; Brannon-Peppas, L. Drug delivery


biomaterials. In Encylopedia of Materials: Science and
Technology; Elsevier: Amsterdam, 2001; 2351–2355.

15. Peppas, N.A.; Bures, P.; Leobandung, L.; Ichikawa, H.


Hydrogels in pharmaceutical formulations. Eur. J. Pharm.
Biopharm. 2000, 50, 27–46.

16. Gu, J.M.; Robinson, J.R.; Leung, S.S. Binding of


acrylic polymers to mucin/epithelial surfaces:
structure-property relationships. CRC Crit. Rev. Ther. Drug
Carrier Syst. 1988, 5, 21–67.

17. Peppas, N.A.; Huang, Y.; Torres-Lugo, M.; Ward, J.H.;


Zhang, J. Physicochemical foundations and structural design
of hydrogels in medicine and biology. Ann. Revs. Biomed.
Eng. 2000, 2, 9–29.

18. Bansil, R.; Stanley, E.; LaMont, J.T. Mucin biophysics.


Ann. Rev. Physiol. 1995, 57, 635–657.

19. Strous, G.J.; Dekker, J. Mucin-type glycoproteins.


Crit. Rev. Biochem. Mol. Biology 1992, 27, 57–92.

20. Smart, J.D.; Kellaway, I.W.; Worthington, H.E.C. An in


vitro investigation of mucosa-adhesive materials for use in
controlled drug delivery. J. Pharm. Pharmacol. 1984, 36,
295–299.

21. Bures, P.; Peppas, N.A. Structural and morphological


characteristics of carriers based on poly(acrylic acid).
Polym. Prepr. 1999, 40 (1), 345–346.

22. Kjellander, R.; Florin, E. Water structure and changes


in thermal stability of the system poly(ethylene
glycol)water. J. Chem. Soc. Faraday Trans I 1981, 77,
2053–2077.

23. Inamura, I.; Jinbo, Y. Interaction between poly(vinyl


alcohol) and poly(ethylene glycol) in water studied by
viscosity and density. Polym. J. 1991, 23, 1143–1147.

24. Sawatari, C.; Kondo, T. Interchain hydrogen bonds in


blend films of poly(vinyl alcohol) and its derivatives with
poly(ethylene oxide). Macromolecules 1999, 32, 1949–1955.
25. Zeghal, M.; Auvray, L. Structure of polymer complex in
water. Europhys. Lett. 1999, 45, 482–487.

26. Tobyn, M.J.; Johnson, J.R.; Dettmar, P.W. Factors


affecting in vitro gastric mucoadhesion I. Test conditions
and instrumental parameters. Eur. J. Pharm. Biopharm. 1995,
41, 233–241.

27. Tobyn, M.J.; Johnson, J.R.; Dettmar, P.W. Factors


affecting in vitro gastric mucoadhesion II. Physical
properties of polymers. Eur. J. Pharm. Biopharm. 1996, 42,
56–61. 28. Lehr, C.M. Bioadhesion technologies for the
delivery of peptide and protein drugs to the
gastrointestinal tract. CRC Crit. Rev. Ther. Drug Carrier
Syst. 1994, 11, 119–160. 29. Schneider, J.; Tirrell, M.
Direct measurement of molecular-level forces and adhesion
in biological systems. In Bioadhesive Drug Delivery
Systems; Mathiowitz, E., Chickering, D.E., Lehr, C.M.,
Eds.; Marcel Dekker, Inc.: New York, 1999; 223–259. 30.
Drotleff, S.; Lungwitz, U.; Breunig, M.; Dennis, A.; Blunk,
A.; Blunk, T.; Tessmar, J.; Gopferich, A. Biomimetic
polymers in pharmaceutical and biomedical sciences. Eur. J.
Pharm. Biopharm. 2004, 58, 385–407. 31. Horbett, T.A. The
role of adsorbed proteins in tissue response to
biomaterials. In Biomaterials Science: An Introduction to
Materials in Medicine; Ratner, B.D, Hoffman, A.S., Schoen,
F.J., Lemons, J.E., Eds.; Elsevier Academic Press: San
Diego, 2004; 237–245. 32. Hanson, S.R. Blood coagulation
and blood-material interactions. In Biomaterials Science:
An Introduction to Materials in Medicine; Ratner, B.D,
Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.; Elsevier
Academic Press: San Diego, 2004; 332–338. 33. Bies, C.;
Lehr, C.M.; Woodley, J.F. Lectin-mediated drug targeting:
history and applications. Adv. Drug. Del. Rev. 2004, 56,
425–435. 34. Costerton, B.; Cook, G.; Shirtliff, M.;
Stoodley, P.; Pasmore, M. Biofilms, biomaterials, and
device-related infections. In Biomaterials Science: An
Introduction to Materials in Medicine; Ratner, B.D,
Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.; Elsevier
Academic Press: San Diego, 2004; 345–354. 35. Xu, L.C.;
Vadillo-Rodriguez, V.; Logan, B.E. Residence time, loading
force, pH, and ionic strength affect adhesion forces
between colloids and biopolymercoated surfaces. Langmuir
2005, 21, 7491–7500. 36. Abu-Lail, N.I.; Camesano, T.A.
Role of ionic strength on the relationship of biopolymer
conformation, DLVO contributions, and steric interactions
to bioadhesion of Pseudomonas putida KT2442.
Biomacromolecules 2003, 4, 1000–1012. 37. Walz, A.;
Odenbreit, S.; Mahdavi, J.; Boren, T.; Ruhl, S.
Identification and characterization of binding properties of
Helicobacter pylori by glycoconjugate arrays. Glycobiology
2005, 15, 700–708. A
Allografts

21. Dawson, P.E.; Schorr, A.J. In Demonstration of Superior


Cellular Viability in Cryopreserved Meniscus, 22nd Annual
Mtg. American Association of Tissue Banks, New Orleans, LA,
1998. 22. Yang, H.; Acker, J.; Chen, J.; McGann, L. In situ
assessment of cell viability. Cell Transplant 1998, 7 (5),
443–451. 23. CryoVein Saphenous Vein Allograft
Infrainguinal Bypass Clinical Experience; CryoLife, Inc.,
2001. Data on file.
Alumina

1. Hench, L.L. Ceramics, Glasses, and Glass–Ceramics. In


Biomaterials Science, An Introduction to Materials in
Medicine; Ratner, B.D., Hoffman, A.S., Schoen, F.J.,
Lemons, J.E., Eds.; Academic Press: San Diego, 1996; 73–84.

2. Hulbert, S.F. The Use of Alumina and Zirconia in


Surgical Implants. In An Introduction to Bioceramics;
Hench, L.L., Wilson, J., Eds.; World Scientific: Singapore,
1993; 25–40.

3. Heimke, G. Use of Alumina Ceramics in Medicine. In


Bioceramics; Hulbert, J.E., Hulbert, S.F., Eds.; Rose–
Hulman Institute of Technology: Terre Haute, 1992; Vol. 3,
19–30.

4. Park, J.B. Aluminum Oxide: Biomedical Applications. In


Concise Encyclopedia of Advanced Ceramic Materials; Brook,
R.J., Ed.; Pergamon Press: Oxford, 1991; 13–16.

5. Heimke, G. Aluminum Oxide. In Concise Encyclopedia of


Medical & Dental Materials Advanced; Williams, D., Ed.;
Pergamon Press: Oxford, 1990; 28–34.

6. Walter, A. On the material and tribology of alumina–


alumina couplings for hip joint prosthesis. Clin. Orthop.
1992, 282, 31–46.

7. Doerre, E.; Dawihl, W. Ceramic Hip Endoprosthesis. In


Mechanical Properties of Biomaterials; Hastings, G.W.,
Williams, D., Eds.; Wiley: New York, 1980; 113–127.

8. Park, J.B.; Lakes, R.S. Biomaterials—An Introduction,


2nd Ed.; Plenum: New York, 1992.

9. Hench, L.L. Bioceramics: From concept to clinic. J. Am.


Ceram. Soc. 1991, 74, 1487–1510.

10. McHale, J.M.; Auroux, A.; Perrotta, A.J.; Navrotsky, A.


Surface energies and thermodynamic phase stability in
nanocrystalline aluminas. Science 1997, 277, 788–791. 11.
Christel, P. Biocompatibility of surgical-grade sense
polycrystalline alumina. Clin. Orthop. 1992, 282, 10–18.
12. Toni, A.; Sudanese, A.; Terzi, S.; Tabarroni, M.;
Calista, F.; Giunti, A. Ceramics in Total Hip Arthroplasty.
In Encyclopedic Handbook of Biomaterials and
BioEngineering; Wise, D.L., Trantolo, D.J., Altobelli,
D.E., Yaszemski, M.J., Gresser, J.D., Schwarz, E.R., Eds.;
Mercel Dekker: New York, 1995; Vol. 2; 1501–1544. Part A.
13. Black, J. Biological Performance of
Materials—Fundamentals of Biocompatibility; Dekker: New
York, 1999. 14. Schackelford, J.F. Introduction to
Materials Science for Engineers, 2nd Ed.; Macmillan: New
York, 1988. 15. Christel, P.; Meunier, A.; Crolet, J.-M.;
Sedal, L.; Witvoet, J. Aluminum Oxide Ceramic–Titanium
Alloy Materials for Total Hip Replacement. In Perspectives
on Biomaterials; Lin, O.C.C., Chao, E.Y.S., Eds.; Elsevier:
Amsterdam, 1986; 277–288. 16. Bizot, P.; Nizard, R.;
Lerouge, S.; Prudhommeaux, F.; Sedel, L. Ceramic/ceramic
total hip arthroplasty. J. Orthop. Sci. 2000, 5, 622–627.
17. Al-Saffar, N.; Revell, P.A.; Kobayashi, A. Modulation
of the phenotypic and functional properties of phagocytic
macrophages by wear particles from orthopaedic implants. J.
Mater. Sci., Mater. Med. 1997, 8 , 641–648. 18. Bierbaum,
B.E.; Barsoum, W.K.; Puri, L.; Headrick, J.; Gomes, S.
Alumina–Alumina Ceramic Bearings. In Bioceramics in Hip
Joint Replacement; Willman, G., Zweymueller, K., Eds.;
Georg Thieme Verlag: Stuttgart, 2000; 22–26. 19. Yoon,
T.R.; Rowe, S.M.; Jung, S.T.; Seon, K.J.; Maloney, W.J.
Osteolysis in association with a total hip arthroplasty
with ceramic bearing surfaces. J. Bone Jt. Surg., Am. 1998,
80, 1458–1467. 20. Kaplan, E.L.; Meier, P. Nonparametric
estimation from incomplete observations. Am. Stat. Assoc.
J. 1958, 53, 457–481. 21. Clinical Performance of Skeletal
Prostheses; Hench, L.L., Wilson, J., Eds.; Chapman & Hall:
London, 1996. 22. Berry, D.J.; Harmsen, W.S.; Cabanela,
M.E.; Morrey, B.F. Twenty-five-year survivorship of two
thousand consecutive primary Charnley total hip
replacements. J. Bone Jt. Surg., Am. 2002, 84–A, 171–177.
23. Hamadouche, M.; Boutine, P.; Daussange, J.; Bolander,
M.E.; Sedel, L. Alumina-on-alumina total hip arthroplasty.
J. Bone Jt. Surg., Am. 2002, 84–A, 69–71. 24. Oonishi, H.;
Amino, H.; Ueno, M.; Yunoki, H. Concepts and Designs for
Total Hip and Knee Replacement. In Reliability and
Long-Term Results of Ceramics in Orthopaedics; Sedal, L.,
Willman, G., Eds.; Georg Thieme Verlag: Stuttgart, 1999;
7–28. 25. Oonishi, H. Knee and Ankle Joint Replacement. In
Osseo-integrated Implants, Vol. 1, Basics, Materials, and
Joint Replacements, Bioceramics; Heimke, G., Ed.; CRC
Press: Boca Raton, 1990; 171. 26. White, E.; Schors, E.C.
Biomaterials aspects of interpore-200 porous
hydroxyapatite. Dent. Clin. North Am. 1986, 30, 49–67. 27.
Yamagami, A.; Kawahara, H. Single-Crystal Alumina
(Sapphire) for Dental Implants. In Encyclopedic Handbook of
Biomaterials and BioEngineering; Wise, D.L., Trantolo,
D.J., Altobelli, D.E., Yaszemski, M.J., Gresser, J.D.,
Schwarz, E.R., Eds.; Mercel Dekker: New York, 1995; Vol. 2,
1617–1638. Part B. 28. Jahnke, K.; Schrader, M.;
Silberzahn, J. Osseo-integrated Implants in
Otorhinolaryngology. In Osseo-integrated Implants, Vol. I1,
Implants in Oral and ENT Surgery; Heimke, G., Ed.; CRC
Press: Boca Raton, 1990; 287. 29. Zollner, C.; Weerda, H.;
Strutz, J. Aluminiumoxid– Keramik als Stutzgerust in der
Trachealchirurgie. In Archives of Oto-Rhino-Laryngology;
Springer-Verlag: Dotdrecht, 1983; 214–216. Supplement II. .
30. Okudera, H.; Kobayashi, S.; Takemae, T. Neurosurgical
Applications of Alumina Ceramic Implants. In Bioceramics:
Proceedings of the 1st Int’l Symp. on Ceramics in Medicine;
Oonishi, H., Aoki, Sawai, K., Eds.; Ishiyaku EuroAmerica
Inc.: Tokyo, 1988; 320–325. 31. Polack, F.M. Clinical
results with a ceramic keratoprosthesis. Cornea 1983, 2,
185–196. 32. Buykx, W.J.; Grabaretk, E.; Reeves, K.D.;
Anderson, R.; Mathivanar, R.; Skalsky, M. Development of
Porous Ceramics for Drug Release and Other Applications. In
Bioceramics; Hulbert, J.E., Hulbert, S.F., Eds.;
Rose–Hulman Institute of Technology: Terre Haute, 1992;
Vol. 3. 33. Rack, R.; Pfaff, H.-G. A New Ceramic Material
for Orthopaedics. In Bioceramics in Hip Joint Replacement;
Willman, G., Zweymueller, K., Eds.; Georg Thieme Verlag:
Stuttgart, 2000; 141–145. A
Angiogenesis Inhibitors

15. Mousa, S. Angiogenesis, coagulation activation, and


malignant dissemination. Sem. Thromb. Haemost. 2002, 28
(1), 45–52.

16. Mousa, S.; Mohamed, S. Anti-angiogenesis efficacy of the


LMWH, tinzaparin and TFPI. Blood 1999, 94 (10), 82.
Abstract 22a.

17. Jung, S.P.; Siegrist, B.; Wade, M.R.; Anthony, C.T.;


Woltering, E.A. Inhibition of human angiogenesis with
heparin and hydrocortisone. Angiogenesis 2001, 4 (3),
175–186.

18. McCarty, O.J.T.; Mousa, S.; Bray, P.; Konstantopoulos,


K. Immobilized platelet support human colon carcinoma cell
tethering, rolling and firm adhesion under dynamic flow
conditions. Blood 2000, 96 (5), 1789–1797. 19. Kerbel,
R.S.; Yu, J.; Tran, J.; Man, S.; Viloria-Petit, A.;
Klement, G.; Coomber, B.L.; Rak, J. Possible mechanisms of
acquired resistance to anti-angiogenic drugs: Implications
for the use of combination therapy approaches. Cancer
Metastasis Rev. 2001, 20 (1–2), 79–86. 20. Kaushal, V.;
Kohli, M.; Zangari, M.; Fink, L.; Mehta, P. Endothelial
dysfunction in antiangiogenesis-associated thrombosis. J.
Clin. Oncol. 2002, 20 (13), 3042– 3043. 21. Chen, Z.;
Malhotra, P.; Thomas, G.; Ondrey, F.; Duffey, D.; Smith,
C.; Enamorada, D.; Yeh, N.; Kroog, G.; Rudy, S.; McCullagh,
L.; Mousa, S. Expression of proinflammatory and
proangiogenic cytokines in patients with head and neck
cancer. Clin. Cancer Res. 1999, (5), 1369–1379.
Animal Models for Bone Tissue Engineering

27. Davidson, M.K.; Lindsey, J.R.; Davis, J.K. Requirements


and selection of an animal model. Isr. J. Med. Sci. 1987,
23, 551–555.

28. Rodgers, J.B.; Monier-Faugere, M.C.; Malluche, H.


Animal models for the study of bone loss after cessation of
ovarian function. Bone 1993, 14 (3), 369–377.

29. Losken, A.; Mooney, M.P.; Siegel, M.I. Comparative


analysis of mandibular growth patterns in seven animal
models. J. Oral Maxillofac. Surg. 1992, 50, 490–495.

30. Losken, A.; Mooney, M.P.; Siegel, M.I. Comparative


cephalometric study of nasal cavity growth patterns in
seven animal models. Cleft Palate Craniofac. J. 1994, 31,
17–23.

31. Smith, G.C. Value of nonhuman primates in predicting


disposition of drugs in man. Ann. N.Y. Acad. Sci. 1969,
162, 600–603.

32. Reynolds, H.H. Nonhuman primates in the study of


toxicological effects on the central nervous system: a
review. Ann. N.Y. Acad. Sci. 1969, 162, 604–609.

33. Langer, R.; Vacanti, J.P. Tissue engineering. Science


1993, 260, 920–926.

34. Putnam, A.J.; Mooney, D.J. Tissue engineering using


synthetic extracellular matrices. Nat. Med. 1996, 2,
824–826.

35. Alsberg, E.; Hill, E.E.; Mooney, D.J. Craniofacial


tissue engineering. Crit. Rev. Oral Biol. Med. 2001, 12,
64–75.

36. Miller, S.C.; Bowman, B.M.; Jee, W.S. Available animal


models of osteopenia—small and large. Bone 1995, 17 (4
suppl.), 117S–123S.

37. Mosekilde, L. Assessing bone quality—animal models in


preclinical osteoporosis research. Bone 1995, 17 (4
suppl.), 343S–352S.

38. Cremieux, A.C.; Carbon, C. Experimental models of bone


and prosthetic joint infections. Clin. Infect. Dis. 1997,
25 (6), 1295–1302.
39. McCauley, L.K. Transgenic mouse models of metabolic
bone disease. Curr. Opin. Rheumatol. 2001, 13 (4), 316–325.

40. Green, D.; Walsh, D.; Mann, S.; Oreffo, R.O. The
potential of biomimesis in bone tissue engineering: lessons
from the design and synthesis of invertebrate skeletons.
Bone 2002, 30 (6), 810–815.

41. Conzemius, M.G.; Brown, T.D.; Zhang, Y.; Robinson, R.A.


A new animal model of femoral head osteonecrosis: one that
progresses to human-like mechanical failure. J. Orthop.
Res. 2002, 20 (2), 303–309.

42. Krebsbach, P.H.; Gehron-Robey, P. Dental and skeletal


stem cells: potential cellular therapeutics for
craniofacial regeneration. J. Dent. Ed. 2002, 66, 766–773.

43. Rodriguez-Merchan, E.C.; Forriol, F. Nonunion: general


principles and experimental data. Clin. Orthop. Relat. Res.
2004, 419, 4–12.

44. Blin-Wakkach, C.; Bernard, F.; Carle, G.F.


Osteopetrosis, from mouse to man. Med. Sci. (Paris) 2004,
20, 61–67.

45. Allen, M.J. Biochemical markers of bone metabolism in


animals: uses and limitations. Vet. Clin. Pathol. 2003, 32,
101–113. 46. Plaza, S.M.; Lamson, D.W. Vitamin K2 in bone
metabolism and osteoporosis. Altern. Med. Rev. 2005, 10,
24–35. 47. Salgado, A.J.; Coutinho, O.P.; Reis, R.L. Bone
tissue engineering: state of the art and future trends.
Macromol. Biosci. 2004, 4, 743–765. 48. Kruyt, M.C.; van
Gaalen, S.M.; Oner, F.C.; Verbout, A.J.; de Bruijn, J.D.;
Bhert, W.J.A. Bone tissue engineering and spinal fusion:
the potential of hybrid constructs by combining
osteoprogenitor cells and scaffolds. Biomaterials 2004, 25,
1463–1473. 49. Evans, C.H.; Ghivizzani, S.C.; Robbins, P.D.
Orthopeadic gene therapy. Clin. Orthop. Relat. Res. 2004,
429, 316–329. 50. Boyan, B.D.; Lohmann, C.H.; Romero, J.;
Schwartz, Z. Bone and cartilage tissue engineering. Clin.
Plast. Surg. 1999, 26, 629–645. 51. Goldstein, S.A.; Patil,
P.V.; Moalli, M.R. Perspectives on tissue engineering of
bone. Clin. Orthop. 1999, 367 (suppl.), S419–S423. 52.
Alsberg, E.; Anderson, K.W.; Albeiruti, A.; Rowley, J.A.;
Mooney, D.J. Engineering growing tissues. Proc. Natl. Acad.
Sci. U.S.A. 2002, 99 (19), 12,025–12,030. 53. Athanasiou,
K.A. Fundamentals of biomechanics in tissue engineering of
bone. Tissue Eng. 2000, 6 (4), 361–381. 54. Young, J.Z. The
Life of Mammals, 3rd Ed.; Clarendon Press: Oxford, 1975.
55. Torricelli, P.; Fini, M.; Giavaresi, G.; Giardino, R.
In vitro models to test orthopedic biomaterials in view of
their clinical applications in osteoporotic bone. Int. J.
Artif. Organs 2004, 27, 658–663. 56. Khan, S.N.; Lane, J.M.
Spinal fusion surgery: animal models for tissue-engineered
bone constructs. Biomaterials 2004, 25, 1475–1485. 57.
Jerome, C.P.; Peterson, P.E. Nonhuman primate models in
skeletal research. Bone 2001, 29 (1), 1–6. 58. Aufdemorte,
T.B.; Boyan, B.D.; Fox, W.C.; Miller, D. Diagnostic tools
and biologic markers: animal models in the study of
osteoporosis and oral bone loss. J. Bone. Miner. Res. 1993,
8 (suppl. 2), S529–S534. 59. Aufdemorte, T.B.; Fox, W.C.;
Miller, D.; Buffum, K.; Holt, G.R.; Carey, K.D. A non-human
primate model for the study of osteoporosis and oral bone
loss. Bone 1993, 14, 581–586. 60. Farris, E.F.; Griffith,
J.Q. The Rat in Laboratory Investigation; Hafner Press: New
York, 1949. 61. Enlow, D.H. Handbook of Facial Growth;
Saunders: New York, 1990. 62. Greene, E.C. Anatomy of the
Rat; Hafner Publishing Co.: New York, 1968. 63. Cooper, G.;
Schiller, S. Anatomy of the Guinea Pig; Harvard University
Press: Cambridge, MA, 1975. 64. Kier, E.L. Phylogenetic and
ontogenetic changes of the brain relevant to the evolution
of the skull. In Development of the Basicranium; Bosma, J.,
Ed.; DHEW/NIH Publication #76-989: Bethesda, MD, 1976;
468–499. 65. Getty, R. Sissons and Grossman’s the Anatomy
of the Domestic Animals; WB Saunders: Philadelphia, PA,
1975. 66. Merkx, M.A.; Maltha, J.C.; Freihofer, H.P.;
KuijpersJagtman, A.M. Incorporation of three types of bone
block implants in the facial skeleton. Biomaterials 1999,
20 (7), 639–645. 67. Merkx, M.A.; Maltha, J.C.; Freihofer,
H.P.; KuijpersJagtman, A.M. Incorporation of particulated
bone implants in the facial skeleton. Biomaterials 1999, 20
(21), 2029–2035. 68. Hartman, C.F.; Strauss, W.L. The
Anatomy of the Rhesus Monkey; Hafner Publishing Co.: New
York, 1933. 69. Elliot, R. Reighard and Jennings’ Anatomy
of the Cat; Holt, Rinehart, and Winston: New York, 1963.
70. Swindler, D.R.; Wood, C.D. An Atlas of Primate Gross
Anatomy: Baboon, Chimpanzee, and Man; Robert, E., Ed.;
Krieger Publishing: Malabar, FL, 1982. 71. Sirianni, J.
Nonhuman primates as models for human craniofacial growth.
In Nonhuman Primate Models for Human Growth and
Development; Watts, E.S., Ed.; A.R. Liss: New York, 1985;
95–124. 72. Enlow, D.H.; McNamara, J. The neurocranial
basis for facial form and pattern. Angle Orthod. 1973, 43,
256–271. 73. Sirianni, J.E.; Swindler, D.R. A review of the
postnatal craniofacial growth in old world monkeys and
apes. Yrbk. Phys. Anthropol. 1979, 22, 80–104. 74. Siebert,
J.R.; Swindler, D.R. Evolutionary changes in the midface
and mandible: establishing the primate form. In
Understanding Craniofacial Anomalies: The Etiopathogenesis
of Craniosynostosis and Facial Clefting; Mooney, M.P.,
Siegel, M.I., Eds.; John W. Wiley and Sons: New York, 2002;
345–378. 75. Geddes, A.D. Animal models of bone disease. In
Principles of Bone Biology; Bilezikians, J.P., Raisz, L.G.,
Rodan, G.A., Eds.; Academic Press: San Diego, 1996;
1343–1354. 76. Enlow, D.H.; Azuma, M. Functional growth
boundaries in the human and mammalian face. Birth Defect
1975, 11, 217–230. 77. Whang, K. Engineering bone
regeneration with bioabsorbable scaffolds with novel
microarchitecture. Tissue Eng. 1999, 5, 35–51. 78. Service,
R.F. Bone remodeling and repair. Science 2000, 289,
1421–1640. 79. Murphy, W.L. Sustained release of vascular
endothelial growth factor from mineralized
polylactide-coglycolic scaffolds for tissue engineering.
Biomaterials 2000, 21, 2521–2527. 80. Shea, L.D. Engineered
bone development from preosteoblast cell line on
three-dimensional scaffolds. Tissue Eng. 2000, 6, 605–617.
81. Mackenzie, D.J. Recombinant human acidic fibroblast
growth factor and fibrin carrier regenerates bone. Plast.
Reconstr. Surg. 2001, 107 (4), 989–996. 82. Hutmacher, D.W.
Scaffold design and fabrication technologies for
engineering tissues—state of the art future perspectives.
J. Biomater. Sci. Polym. Ed. 2001, 12, 102–124. 83.
Einhorn, T.; Lee, C.A. Bone regeneration: new findings and
potential clinical applications. J. Am. Acad. Orthop. Surg.
2001, 9, 157–165. 84. Jansen, J.A. Laboratory animal models
for the manufacture of tissue substitutes. Ned. Tijdschr.
Tandheelkd. 2004, 111, 490–493. 85. Schmitz, J.P.;
Hollinger, J.O. The critical size defect as an experimental
model for craniomandibulofacial nonunions. Clin. Orthop.
1986, 205, 299–308. 86. Mundell, R.; Mooney, M.P.; Siegel,
M.I.; Losken, A. Osseous guided tissue regeneration using a
collagen barrier membrane. J. Oral Maxillofac. Surg. 1993,
51, 1004–1012. 87. Mooney, M.P.; Mundell, R.D.; Stetzer,
K.; Ochs, M.W.; Milch, E.A.; Buckley, M.J.; Siegel, M.I.
The effects of guided tissue regeneration and fixation
technique on osseous wound healing in rabbit zygomatic arch
osteotomies. J. Craniofac. Surg. 1996, 7, 46–53. 88.
Stetzer, K.M.; Cooper, G.; Gassner, R.; Kapucu, R.;
Mundell, R.; Mooney, M.P. The effects of fixation type and
guided tissue regeneration on maxillary osteotomy healing
in rabbits. J. Oral Maxillofac. Surg. 2002, 60, 427–436.
89. Frame, J.W. A convenient animal model for testing bone
substitution materials. J. Oral Surg. 1980, 38, 176–180.
90. Prolo, D.J.; Pedrotti, P.W.; Burres, K.P.; Oklund, S.
Superior osteogenesis in transplanted allogenic canine
skull following chemical sterilization. Clin. Orthop. 1982,
108, 203–207. 91. Lee, J.Y.; Musgrave, D.; Pelinkovic, D.;
Fukushima, K.; Cummins, J.; Usas, A.; Robbins, P.; Fu,
F.H.; Huard, J. Effect of bone morphogenetic
protein-2-expressing muscle-derived cells on healing of
criticalsized bone defects in mice. J. Bone Joint Surg. Am.
2001, 83-A, 1032–1039. 92. Takagi, K.; Urist, M.R. The role
of bone marrow induced repair of femoral massive diaphyseal
defects. Clin. Orthop. 1982, 171, 224–231. 93. Hollinger,
J.O.; Schmitz, J.P.; Yaskovich, R.; Long, M.M.; Prasad,
K.U.; Urry, D.W. A synthetic polypentapeptide of elastin
for initiating calcification. Calcif. Tissue Int. 1988, 42,
231–236. 94. Winn, S.R.; Schmitt, J.M.; Buck, D.; Hu, Y.;
Grainger, D.; Hollinger, J.O. Tissue-engineered bone
biomimetic to regenerate calvarial critical-sized defects
in athymic rats. J. Biomed. Mater. Res. 1999, 45 (4),
414–421. 95. Clokie, C.M.; Moghadam, H.; Jackson, M.T.;
Sandor, G.K. Closure of critical sized defects with
allogenic and alloplastic bone substitutes. J. Craniofac.
Surg. 2002, 13 (1), 111–121. 96. Mooney, M.P.; Burrows,
A.M.; Smith, T.D.; Losken, H.W.; Opperman, L.A.; Dechant,
J.; Kreithen, A.M.; Kapucu, R.; Cooper, G.M.; Ogle, R.C.;
Siegel, M.I. Correction of coronal suture synostosis using
suture and dura mater allografts in rabbits with familial
craniosynostosis. Cleft Palate Craniofac. J. 2001, 38,
72–91. 97. Vesala, A.L.; Kallioinen, M.; Tormala, P.;
Kellomaki, M.; Waris, T.; Ashammakhi, N. Bone tissue
engineering: treatment of cranial bone defects in rabbits
using self-reinforced poly-L,D lactide 96/4 sheets. J.
Craniofac. Surg. 2002, 13 (5), 607–613. A

98. Costantino, P.D.; Friedman, C.D.; Jones, K.; Chow,


L.C.; Sisson, G.A. Experimental hydroxyapatite cement
cranioplasty. Plast. Reconstr. Surg. 1992, 90 (2), 174–185.

99. Urist, M.R. New advanced in bone research. West. J.


Med. 1984, 141, 71.

100. Shang, Q.; Wang, Z.; Liu, W.; Shi, Y.; Cui, L.; Cao,
Y. Tissue-engineered bone repair of sheep cranial defects
with autologous bone marrow stromal cells. J. Craniofac.
Surg. 2001, 12 (6), 586–593.

101. Hollinger, J.O.; Mark, D.E.; Bach, D.E.; Reddi, A.H.;


Seyfer, A.E. Calvarial bone regeneration using osteogenin.
J. Oral Maxillofac. Surg. 1989, 47, 1182–1186.

102. Ripamonti, U.; Ramoshebi, L.N.; Matsaba, T.; Tasker,


J.; Crooks, J.; Teare, J. Bone induction by BMPs/ OPs and
related family members in primates. J. Bone Joint Surg. Am.
2001, 83-A (suppl. 1, pt 2), S116–S127.

103. Bardach, J.; Roberts, D.M.; Klausner, E.C. Influence of


two-flap palatoplasty on facial growth in rabbits. Cleft
Palate Craniofac. J. 1979, 16, 402–411.
104. Bardach, J.; Roberts, D.M.; Yale, R.; Rosewall, D.;
Mooney, M. The influence of simultaneous cleft lip and
palate repair on facial growth in rabbits. Cleft Palate
Craniofac. J. 1980, 17 (4), 309–318.

105. Freng, A. Transversal maxillary growth in experimental


submucous mid-palatal clefts. A roentgencephalometric study
in the cat. Scand. J. Plast. Reconstr. Surg. 1979, 13 (3),
409–416.

106. Bardach, J.; Mooney, M.; Bardach, E. The influence of


two-flap palatoplasty on facial growth in beagles. Plast.
Reconstr. Surg. 1982, 69 (6), 927–936.

107. Bardach, J.; Martin, R.; Mooney, M.P.; Kelly, K.M.;


Albright, J. Bone formation in the canine palate following
partial resection. In Normal and Abnormal Bone Growth:
Basic and Clinical Research; Sarnat, B.G., Ed.; W.B.
Saunders: New York, 1985; 365–377.

108. Kawata, T.; Kohno, S.; Fujita, T.; Sugiyama, H.;


Tokimasa, C.; Kaku, M.; Tsutsui, K.; Tanne, K.
Transplantation of new autologous biomaterials into jaw
cleft. J. Int. Med. Res. 2001, 29, 287–291.

109. Takano-Yamamoto, T.; Kawakami, M.; Sakuda, M. Defects


of the rat premaxilla as a model of alveolar clefts for
testing bone-inductive agents. J. Oral Maxillofac. Surg.
1993, 51, 887–891.

110. Verwoerd, C.D.A.; Verwoerd-Verhoef, H.L.; Urbanis,


N.A.M. Skulls with facial clefts. Experimental surgery on
the facial skeleton. Acta Otolaryngol. 1976, 81, 249–256.

111. Eisbach, K.J.; Bardach, J.; Klausner, E.C. The


influence of primary unilateral cleft lip repair on facial
growth, part II: direct cephalometry of the skull. Cleft
Palate Craniofac. J. 1978, 15, 109–117.

112. Verschueren, D.; Gassner, R.; Mitchell, R.; Mooney,


M.P. Le Fort I osteotomy healing in rabbits with the use of
guided tissue regeneration (GTR). Int. J. Oral Maxillofac.
Surg. 2005, 34, 650–655.

113. El Deeb, M.; Horswell, B.; Waite, D. A primate model


for producing experimental alveolar cleft defects. J. Oral
Maxillofac. Surg. 1985, 43, 523–527.

114. Alden, T.D.; Jane, J.A., Jr.; Hudson, S.B.; Helm, G.A.
The use of bone morphogenetic protein gene therapy in
craniofacial bone repair. J. Craniofac. Surg. 2000, 11 (1),
24–30. 115. Kaban, L.B.; Glowacki, J.; Murray, J.E. Repair
of experimental bony defects in rats. Surg. Forum 1979, 30,
519–521. 116. Saadeh, P.B.; Khosla, R.K.; Mehrara, B.J.;
Steinbrech, D.S.; McCormick, S.A.; DeVore, D.P.; Longaker,
MT. Repair of a critical size defect in the rat mandible
using allogenic type I collagen. J. Craniofac. Surg. 2001,
12 (6), 573–579. 117. Kahnberg, K. Restoration of
mandibular jaw defects in the rabbit by subperiosteally
implanted Teflon s mantle leaf. Int. J. Oral Surg. 1979, 8,
449–456. 118. Hollinger, J.O.; Schmitz, J.P. Restoration of
bone discontinuities in dogs using a biodegradable implant.
J. Maxillofac. Surg, 1987, 45, 594–600. 119. Schliephake,
H.; Knebel, J.W.; Aufderheide, M.; Tauscher, M. Use of
cultivated osteoprogenitor cells to increase bone formation
in segmental mandibular defects: an experimental pilot
study in sheep. Int. J. Oral Maxillofac. Surg. 2001, 30
(6), 531–537. 120. Henkel, K.O.; Gerber, T.; Dorfling, P.;
Hartel, J.; Jonas, L.; Gundlach, K.K.; Bienengraber, V.
Stimulating regeneration of bone defects by implantation of
bioceramics and autologous osteoblast transplantation. Mund
Kiefer Gesichtschir. 2002, 6 (2), 59–65. 121. Lindsey,
W.H.; Franz, D.A.; Toung, J.S.; London, S.D.; Ogle, R.O. A
nasal critical-size defect: an experimental model for the
evaluation of facial osseous repair techniques. Arch.
Otolaryngol. Head Neck Surg. 1998, 124 (8), 912–915. 122.
Einhorn, T.A.; Lane, J.M.; Burstein, A.H.; Kopman, C.R.;
Vigorita, V.J. The healing of segmental bone defects
induced by demineralized bone matrix. J. Bone Joint Surg.
1984, 66A, 274–279. 123. Lieberman, J.R.; Daluiski, A.;
Stevenson, S.; Wu, L.; McAllister, P.; Lee, Y.P.; Kabo,
J.M.; Finerman, G.A.; Berk, A.J.; Witte, O.N. The effect of
regional gene therapy with bone morphogenetic
protein-2producing bone-marrow cells on the repair of
segmental femoral defects in rats. J. Bone Joint Surg. Am.
1999, 81 (7), 905–917. 124. Lane, J.M.; Yasko, A.W.; Tomin,
E.; Cole, B.J.; Waller, S.; Browne, M.; Turek, T.; Gross,
J. Bone marrow and recombinant human bone morphogenetic
protein-2 in osseous repair. Clin. Orthop. 1999, 361,
216–227. 125. Wheeler, D.L.; Eschbach, E.J.; Hoellrich,
R.G.; Montfort, M.J.; Chamberland, D.L. Assessment of
resorbable bioactive material for grafting of criticalsize
cancellous defects. J. Orthop. Res. 2000, 18 (1), 140–148.
126. Bruder, S.P.; Kraus, K.H.; Goldberg, V.M.; Kadiyala,
S. The effect of implants loaded with autologous
mesenchymal stem cells on the healing of canine segmental
bone defects. J. Bone Joint Surg. Am. 1998, 80 (7),
985–996. 127. Lange, T.A.; Zerwekh, J.E.; Peek, R.D.;
Mooney, V.; Harrison, B.H. Granular tricalcium phosphate in
large cancellous defects. Ann. Clin. Lab. Sci. 1986, 16,
467–472. 128. Narang, R.; Laskin, D.M. Experimental
osteogenesis at fracture sites and gaps. J. Oral Surg.
1976, 34, 225–231. 129. Landry, P.S.; Marino, A.A.;
Sadasivan, K.K.; Albright, J.A. Bone injury response. An
animal model for testing theories of regulation. Clin.
Orthop. 1996, 332, 260–273. 130. Toombs, J.P.; Wallace,
L.J.; Bjorling, D.E.; Rowland, G.N. Evaluation of Key’s
hypothesis in the feline tibia: an experimental model for
augmented bone healing studies. Am. J. Vet. Res. 1985, 46,
513–518. 131. Collier, M.A.; Brighton, C.T.; Norrdin, R.;
Twardock, A.R.; Rendano, V.T. Direct current stimulation of
bone production in the horse: preliminary study with a
‘‘gap healing’’ model. Am. J. Vet. Res. 1985, 46, 612–621.
132. Ben-fu, C.; Xue-ming, T. Ultrastructural investigation
of experimental non-union of fractures. Chin. Med. J. 1986,
99, 207–214. 133. Wheeler, D.L.; Chamberland, D.L.;
Schmitt, J.M.; Buck, D.C.; Brekke, J.H.; Hollinger, J.O.;
Joh, S.P.; Suh, K.W. Radiomorphometry and biomechanical
assessment of recombinant human bone morphogenetic protein
2 and polymer in rabbit radius ostectomy model. J. Biomed.
Mater. Res. 1998, 43 (4), 365–373. 134. Hollinger, J.O.;
Schmitt, J.M.; Buck, D.C.; Shannon, R.; Joh, S.P.; Zegzula,
H.D.; Wozney, J. Recombinant human bone morphogenetic
protein-2 and collagen for bone regeneration. J. Biomed.
Mater. Res. 1998, 43 (4), 356–364. 135. Sciadini, M.F.;
Dawson, J.M.; Johnson, K.D. Evaluation of bovine-derived
bone protein with a natural coral carrier as a bone-graft
substitute in a canine segmental defect model. J. Orthop.
Res. 1997, 15 (6), 844–857. 136. Key, J.A. The effects of
local calcium depot on osteogenesis and healing of
fractures. J. Bone Joint Surg. (Am.) 1934, 16, 176–184.
137. Nilsson, O.S.; Urist, M.R.; Dawson, E.G.; Schmalzried,
T.P.; Finerman, G.A. Bone repair induced by bone
morphogenic protein in ulnar defects in dogs. J. Bone Joint
Surg. 1986, 63B, 635–642. 138. Salkeld, S.L.; Patron, L.P.;
Barrack, R.L.; Cook, S.D. The effect of osteogenic
protein-1 on the healing of segmental bone defects treated
with autograft or allograft bone. J. Bone Joint Surg. Am.
2001, 83-A (6), 803–816. 139. Thompson D.D.;Simmons, H.A.;
Pirie, C.M.; Ke, H.Z. FDA guidelines and animal models for
osteoporosis. Bone 1995, 17 (4 suppl.), 125S–133S. 140.
Oegema, T.R.; Visco, D. Animal models of osteoarthritis. In
Animal Models in Orthopaedic Research; An, Y.H., Friedman,
R.J., Eds.; CRC Press: London, 1999; 349–367. 141. Badger,
A.M.; Bradbeer, J.N.; Votta, B.; Lee, J.C.; Adams, J.L.;
Griswold, D.E. Pharmacological profile of SB 203580, a
selective inhibitor of cytokine suppressive binding
protein/p38 kinase, in animal models of arthritis, bone
resorption, endotoxin shock and immune function. J.
Pharmacol. Exp. Ther. 1996, 279 (3), 1453–1461. 142.
Wooley, P.H. Collagen-induced arthritis in the mouse.
Methods Enzymol. 1988, 162, 361–373. 143. Breban, M. Animal
models and in vitro models for the study of
aetiopathogenesis of spondyloarthropathies. Baillieres
Clin. Rheumatol. 1998, 12 (4), 611–626. 144. Joe, B.;
Griffiths, M.M.; Remmers, E.F.; Wilder, R.L. Animal models
of rheumatoid arthritis and related inflammation. Curr.
Rheumatol. Rep. 1999, 1 (2), 139–148. 145. Phillips, T.W.;
Johnston, G.; Wood, P. Selection of an animal model for
resurfacing hip arthroplasty. J. Arthroplasty 1987, 2 (2),
111–117. 146. Mader, J.P. Animal models of osteomyelitis.
Am. J. Med. 1985, 78 (suppl. 6B), 213–217. 147. Norden,
C.W. Lessons learned from animal models of osteomyelitis.
Rev. Infect. Dis. 1988, 10, 103–110. 148. Rissing, J.P.
Animal models of osteomyelitis: knowledge, hypothesis, and
speculation. Infect. Dis. Clin. North Am. 1990, 4 (3),
377–390. 149. Cesnjaj, M.; Stavljenic, A.; Vukicevic, S. In
vivo models in the study of osteopenias. Eur. J. Clin.
Chem. Clin. Biochem. 1991, 29 (4), 211–219. 150. Black, A.;
Tilmont, E.M.; Handy, A.M.; Scott, W.W.; Shapses, S.A.;
Ingram, D.K.; Roth, G.S.; Lane, M.A. A nonhuman primate
model of age-related bone loss: a longitudinal study in
male and premenopausal female rhesus monkeys. Bone 2001, 28
(3), 295–302. 151. Draper, H.H. Advances in Nutritional
Research; Plenum Press: New York, 1985; 172–186. 152.
Riesenfeld, A.; Siegel, M.I.; Mooney, M.P.; Seroky, J.T.;
Taylor, A.B. The effects of perinatal alcohol exposure and
dietary calcium supplements on skeletal and dental growth
in rats. Acta Anat. 1991, 140, 1–7. 153. Turner, R.T.;
Aloia, R.C.; Segel, L.D.; Hannon, K.S.; Bell, N.H. Chronic
alcohol treatment results in disturbed vitamin D metabolism
and skeletal abnormalities in rates. Alcohol. Clin. Exp.
Res. 1988, 12, 159–162. 154. Yoneda, T.; Michigami, T.; Yi,
B.; Williams, P.J.; Niewolna, M.; Hiraga, T. Use of
bisphosphonates for the treatment of bone metastasis in
experimental animal models. Cancer Treat. Rev. 1999, 25
(5), 293–299. 155. Miller, D.R.; Aufdemorte, T.B.; Fox,
W.C.; Waldrop, T.C.; Mealey, B.L.; Brunsvold, M.A.
Periodontitis in the baboon: a potential model for human
disease. J. Periodontal Res. 1995, 30 (6), 404–409. 156.
Haney, J.M.; Zimmerman, G.J.; Wikesjo, U.M. Periodontal
repair in dogs: evaluation of the natural disease model. J.
Clin. Periodontol. 1995, 22 (3), 208–213. 157. Levy, B.M.
Primates in dental research. In Proceedings of the Third
Conference on Experimental Medicine and Surgery in
Primates, Medical Primatology; Goldsmith, E.I., Moor
Jankowski, J., Eds.; Karger Press: Basel, 1971; 859–869.
158. Levy, B.M. Animal model of human disease: chronic
destructive periodontitis (periodontal disease, pyorrhea
alveolaris, pyorrhea). Am. J. Pathol. 1976, 83, 637–640. A

159. Levy, B.M. Animal analogues for the study of dental


and oral diseases. Dev. Biol. Stand. 1980, 45, 51–59.

160. Dreizen, S.; Levy, B.M. Monkey models in dental


research. J. Med. Primatol. 1977, 6, 133–144.

161. Genco, C.A.; Van Dyke, T.; Amar, S. Animal models for
Porphyromonas gingivalis mediated periodontal disease.
Trends Microbiol. 1998, 6 (11), 444–449.

162. Sietsema, W.K. Animal models of cortical porosity.


Bone 1995, 17 (4 suppl.), 297S–305S.

163. Lamghari, M.; Huet, H.; Laurent, A.; Berland, S.;


Lopez, E. A model for evaluating injectable bone
replacements in the vertebrae of sheep: radiological and
histological study. Biomaterials 1999, 20, 2107–2114.

164. Miller, L.C.; Weaver, D.S.; McAlister, J.A.; Koritnik,


D.R. Effects of ovariectomy on vertebral bone in the
cynomolgus monkey (Macaca fascicularis). Calcif. Tissue
Int. 1986, 38, 62–65.

165. Mackey, M.S.; Stevens, M.L.; Ebert, D.C.; Tressler,


D.L.; Combs, K.S.; Lowry, C.K.; Smith, P.N.; McOsker, J.E.
The ferret as a small animal model with BMU-based
remodeling for skeletal research. Bone 1995, 17 (4 suppl.),
191S–196S.

166. Raab-Cullen, D.M.; Akhter, M.P.; Kimmel, D.B.; Recker,


R.R. On animal models for studying bone adaptation. Calcif.
Tissue Int. 1994, 55 (4), 317–318.

167. Kimmel, D.B. Animal models for in vivo experimentation


in osteoporosis research. In Osteoporosis; Marcus, J.,
Feldman, K.F., Kelsey, E.C., Eds.; Academic Press: San
Diego, 1996; 671–690.

168. Wronski, T.J.; Yen, C.F. The ovariectomized rat as an


animal model for postmenopausal bone loss. Cells Mater.
1991, 4 (suppl. 1), 69–74.

169. Frost, H.M.; Jee, W.S.S. On the rat model of human


osteopenia and osteoporosis. Bone Miner. 1992, 18, 227–236.
170. Kalu, D.N. The ovariectomized rat model of
postmenopausal bone loss. Bone Miner. 1991, 15, 175–192.
171. Vanderschueren, D.; Van Herck, E.; Suiker, A.M.;
Allewaert, K.; Visser, W.J.; Geusens, P.; Bouillon, R. Bone
and mineral metabolism in the adult guinea pig: long-term
effects of estrogen and androgen deficiency. J. Bone Miner.
Res. 1992, 7, 1407–1415. 172. Mosekilde, L.; Weisbrode,
S.E.; Safron, J.A.; Stills, H.F.; Jankowsky, M.L.; Ebert,
D.C.; Danielsen, C.C.; Sogaard, C.H.; Franks, A.F.;
Stevens, M.L. Calciumrestricted ovariectomized Sinclair
S-1minipigs: an animal model of osteopenia and trabecular
plate perforation. Bone 1993, 14, 379–382. 173. Kagel,
E.M.; Majeska, R.J.; Einhorn, T.A. Effects of diabetes and
steroids on fracture healing. Curr. Opin. Orthop. 1995, 6
(5), 7–13. 174. Hough, F.S. Alterations of bone and mineral
metabolism in diabetes mellitus, part I, an overview. S.
Afr. Med. J. 1987, 72 (2), 116–119. 175. Zapf, J. Growth
promotion by insulin-like growth factor I in
hypophysectomized and diabetic rats. Mol. Cell Endocrinol.
1998, 140 (1–2), 143–149. 176. Daughaday, W.H. Animal
models of abnormal GH receptor binding and postbinding
mechanisms. Pediatr. Adolesc. Endocrinol. 1992, 24,
282–287. 177. Hull, K.L.; Harvey, S. Growth hormone
resistance: clinical states and animal models. J.
Endocrinol. 1999, 163 (2), 165–172. 178. Diewert, V.M.;
Lozanoff, S. Animal models of facial clefting—experimental,
congenital, and transgenic. In Understanding Craniofacial
Anomalies: The Etiopathogenesis of Craniosynostosis and
Facial Clefting; Mooney, M.P., Siegel, M.I., Eds.; John W.
Wiley and Sons: New York, 2002; 251–272.
Animal Surrogate Systems for Toxicity
Testing

33. Glynn, S.L.; Yazdanian, M. In vitro blood-brain barrier


permeability of nevirapine compared to other antiretroviral
agents. J. Pharm. Sci. 1998, 87 (3), 306–310.

34. Ma, S.H.; Lepak, L.A.; Hussain, R.J.; Shain, W.;


Shuler, M.L. An endothelial and astrocyte co-culture model
of the blood-brain barrier utilizing an ultra-thin,
nanofabricated silicon nitride membrane. Lab On a Chip
2005, 5 (1), 74–85.

35. Mueller-Klieser, W. Three-dimensional cell cultures:


from molecular mechanisms to clinical applications. Am. J.
Physiol. Cell Physiol. 1997, 273, C1109–C1123.

36. Kunz-Schughart, L.A.; Freyer, J.P.; Hofstaedter, F.;


Ebner, R. The use of 3-D cultures for high-throughput
screening: the multicellular spheroid model. J. Biomol.
Screen. 2004, 9 (4), 273–285.

37. Kunz-Schughart, L.A.; Kreutz, M.; Knuechel, R.


Multicellular spheroids: a three dimensional in vitro
culture system to study tumor biology. Int. J. Exp. Pathol.
1998, 79, 1–23.

38. Castaneda, F.; Kinne, R.K.H. Cytotoxicity of millimolar


concentrations of ethanol on HepG2human tumor cell line
compared to normal rat hepatocytes in vitro. J. Cancer Res.
Clin. Oncol. 2000, 126, 503–510.

39. Griffith, L.G.; Naughton, G. Tissue engineering— current


challenges and expanding opportunities. Science 2002, 295,
1009–1016.

40. Schmeichel, K.L.; Bissell, M.J. Modeling tissue-specific


signaling and organ function in three dimensions. J. Cell
Sci. 2003, 116 (12), 2377–2388.

41. Shuler, M.L.; Kargi, F. Bioprocess Engineering—Basic


Concepts, 2nd Ed.; Prentice Hall PTR: Upper Saddle River,
New Jersey, 2002.

42. Choi, S.H.; Fukuda, O.; Sakoda, A.; Sakai, Y. Enhanced


cytochrome P450 capacities of Caco-2 and HepG2 cells in new
coculture system under the static and perfused conditions:
evidence for possible organto-organ interactions against
exogenius stimuli. Mat. Sci. Eng. C 2004, 24, 333–339. 43.
Kumar, S.; Wittmann, C.; Heinzel, E. Minibioreactors.
Biotechnol. Lett. 2004, 26, 1–10. 44. Casciari, J.J.;
Hollingshead, M.G.; Alley, M.C.; Mayo, J.G.; Malspeis, L.;
Miyauchi, S.; Grever, M.R.; Weinstein, J.N. Growth and
chemotherapeutic response of cells in a hollow-fiber in
vitro solid tumor model. J. Natl. Cancer Inst. 1994, 86
(24), 1846–1852. 45. Cooper, J.T.; Goldstein, S. Toxicity
testing in vitro. II. Use of a microsome—cultured human
fibroblast system to study the cytotoxicity of
cyclophosphamide. Can. J. Physiol. Pharmacol. 1976, 54 (4),
546–550. 46. Li, A.P.; Bode, C.; Sakai, Y. A novel in vitro
system, the integrated discrete multiple organ cell culture
(IdMOC) system, for the evaluation of human drug toxicity:
comparative cytotoxicity of tamoxifen towards normal human
cells from five major organs and MCF-7 adenocarcinoma breast
cancer cells. Chem. Biol. Interact. 2004, 150 (1), 129–136.
47. Sweeney, L.M.; Shuler, M.L.; Babish, J.G.; Ghanem, A. A
cell culture analogue of rodent physiology: application to
naphthalene toxicology. Toxicol. In Vitro 1995, 9 (3),
307–316. 48. Ghanem, A.; Shuler, M.L. Characterization of a
perfusion reactor utilizing mammalian cells on microcarrier
beads. Biotechnol. Prog. 2000, 16 (3), 471–479. 49. Ghanem,
A.; Shuler, M.L. Combining cell culture analogue reactor
designs and PBPK models to probe mechanisms of naphthalene
toxicity. Biotechnol. Prog. 2000, 16 (3), 334–345. 50. Sin,
A.; Chin, K.C.; Jamil, M.F.; Kostov, Y.; Rao, G.; Shuler,
M.L. The design and fabrication of threechamber microscale
cell culture analog devices with integrated dissolved
oxygen sensors. Biotechnol. Prog. 2004, 20 (1), 338–345.
51. Viravaidya, K.; Sin, A.; Shuler, M.L. Development of a
microscale cell culture analog to probe naphthalene
toxicity. Biotechnol. Prog. 2004, 20 (1), 316–323. 52.
Viravaidya, K.; Shuler, M.L. Incorporation of 3T3-L1 cells
to mimic bioaccumulation in a microscale cell culture
analog device for toxicity studies. Biotechnol. Prog. 2004,
20 (2), 590–597. 53. Khamsi, R. Labs on a chip meet the
stripped down rat. Nature 2005, 435 (7038), 12–13.
Antimicrobial/Antibiotic (Infection
Resistance) Materials

3. Cobb, D.K.; High, K.B.; Sawyer, R.G.; Sable, C.A.;


Adams, R.B.; Lindley, T.L.; Pruett, T.L.; Schwenzer, K.J.;
Farr, B.M. A controlled trial of scheduled catheter
replacement of central venous and pulmonary-artery
catheters. N. Engl. J. Med. 1992, 327, 1062–1068.

4. Nasim, A.; Thompson, M.M.; Naylor, A.R.; Bell, P.R.F.;


London, N.J.M. The impact of MRSA on vascular surgery. Eur.
J. Vasc. Endovasc. Surg. 2001, 22, 211–214.

5. Bayston, R. Biofilms and Prosthetic Devices. In Community


Structure and Cooperation in Biofilms; Allison, D.G.,
Gilbert, P., Lappin-Scott, H.M., Wilson, M., Eds.;
Cambridge University Press: Cambridge, 2000.

6. Green, R.J.; Davies, M.C.; Roberts, C.J.; Tendler,


S.J.B. Competitive protein adsorption as observed by
surface plasmon resonance. Biomaterials 1999, 20, 385–391.

7. An, Y.H.; Friedman, R.J. Concise review of mechanisms of


bacterial adhesion to biomaterial surfaces. J. Biomed.
Mater. Res. 1998, 43, 338–348.

8. Boelens, J.J.; Dankert, J.; Murk, J.L.; Weening, J.J.;


van der Poll, T.; Dingemans, K.P.; Koole, L.; Laman, J.D.;
Zaat, S.A.J. Biomaterial-associated persistence of
Staphylococcus epidermidis in pericatheter macrophages. J.
Infect. Dis. 2000, 181, 1337–1349.

9. Bayston, R.; Penny, S.R. Excessive production of mucoid


substance in Staphylococcus SII A: A possible factor in
colonisation of Holter shunts. Dev. Med. Child Neurol.,
Suppl. 1972, 27, 25–28.

10. Christensen, B.E.; Charaklis, W.G. Physical and


Chemical Properties of Biofilms; Characklis, K.C., Ed.;
Wiley & Sons: New York, 1990; 93–130. W.G.M.

11. Mack, D.; Fischer, W.; Krokotsch, A.; Leopold, K.;


Hartmann, R.; Egge, H.; Laufs, R. The intercellular adhesin
involved in biofilm accumulation of Staphylococcus
epidermidis is a linear beta-1,6-linked glucosaminoglycan:
Purification and structural analysis. J. Bacteriol. 1996,
178, 175–183.

12. Maki, D.G.; Stolz, S.M.; Wheeler, S.; Mermel, L.A.


Prevention of central venous catheter-related bloodstream
infection by use of an antiseptic-impregnated catheter—A
randomized, controlled trial. Ann. Intern. Med. 1997, 127,
257–266.

13. Raad, I.; Darouiche, R.; Hachem, R.; Sacilowski, M.;


Bodey, G. Antibiotics and prevention of microbial
colonization of catheters. Antimicrob. Agents Chemother.
1995, 39, 2397–2400.

14. Costerton, J.W.; Cook, G.; Lamont, R. The Community


Architecture of Biofilms: Dynamic, Structures and
Mechanisms. In Dental Plaque Revisited: Oral Biofilm in
Health and Disease; Newman, H.N., Wilson, M., Eds.;
Bioline: Cardiff, 1999.

15. Brown, M.R.W.; Allison, D.G.; Gilbert, P. Resistance of


bacterial biofilms to antibiotics: A growth related effect?
J. Antimicrob. Chemother. 1988, 22, 777–780.

16. Ziebuhr, W.; Dietrich, K.; Trautmann, M.; Wilhelm, M.


Chromosomal rearrangements affecting biofilm production and
antibiotic resistance in a Staphylococcus epidermidis
strain causing shunt-associated ventriculitis. Int. J. Med.
Microbiol. 2000, 290, 115–120. 17. Stoodley, P.; Wilson,
S.; Hall-Stoodley, L.; Boyle, J.D.; Lappin-Scott, H.M.;
Costerton, J.W. Growth and detachment of cell clusters from
mature mixed species biofilms. Appl. Environ. Microbiol.
2001, 67, 5608–5813. 18. Anders, C.; Gartner, R.; Steinert,
V.; Voit, B.I.; Zschoche, S. Surface modification with
hydrogels via macroinitiators for enhanced friction
properties of biomaterials. J. Macromol. Sci., Pure Appl.
Chem. 1999, A36 (7–8), 1017–1029. 19. Park, K.D.; Kim,
Y.S.; Han, D.K.; Kim, Y.H.; Lee, E.H.B.; Suh, H.; Choi,
K.S. Bacterial adhesion on PEG modified polyurethane
surfaces. Biomaterials 1998, 19 (7–9), 851–859. 20.
Schierholz, J.M.; Ko¨nig, D.P.; Beuth, J.; Pulverer, G.
Silver coating of medical devices—A review. J. Hosp.
Infect. 1998, 40, 257–262. 21. Jucker, B.A.; Harma, H.;
Hug, S.J.; Zehnder, A.B. Adsorption of bacterial surface
polysaccharides on mineral oxides is mediated by hydrogen
bonds. Colloids Surf., B 1997, 9, 331–343. 22. Gottenbos,
B.; Grijpma, D.W.; van der Mei, H.C.; Feijen, J.; Busscher,
H.J. Antimicrobials effects of positively charged surfaces
on adhering gram-positive and gram-negative bacteria. J.
Mater. Sci., Mater. Med. 1999, 10, 853–855. 23. Gorman,
S.P.; Jones, D.S.; Mawhiney, W.M.; McGovern, J.G.; Adair,
C.G. Conditioning fluid influences on the surface properties
of silicone and polyurethane peritoneal catheters:
Implications for infection. J. Mater. Sci., Mater. Med.
1997, 8, 631–635. 24. Price, C.; Waters, M.G.J.; Williams,
D.W.; Lewis, M.A.O.; Stickler, D. Surface modification of an
experimental silicone rubber aimed at reducing initial
candidal adhesion. J. Biomed. Mater. Res. 2002, 63,
122–128. 25. Bellanda, M.; Cassinelli, C.; Morra, M.
Reduced plaque accumulation on hydrocarbon thin film
deposited on restorative acrylic polymers. J. Biomed.
Mater. Res. 1997, 36, 216–222. 26. Miyayama, N.; Yoshinari,
M.; Oda, Y. Surface modification of titanium implants with
dry process-surface characterization. Jpn. J. Dent. Mater.
1999, 18, 109–121. 27. Yoshinari, M.; Oda, Y.; Kato, T.;
Okuda, K.; Hirayama, A. Influence of surface modifications to
titanium on oral bacterial adhesion in vitro. J. Biomed.
Mater. Res. 2000, 52, 388–394. 28. Choong, S.; Wood, S.;
Fry, C.; Whitfield, H. Catheter associated urinary tract
infection and encrustration. Int. J. Antimicrob. Agents
2001, 17, 305–310. 29. Angell, A.H.; Resnick, M.I. Surface
interaction between glycosaminoglycans and calcium oxalate.
J. Urol. 1989, 141 , 1255–1258. 30. Russell, J.C. Bacteria,
biofilms, and devices: The possible protective role of
phosphorylcholine materials. J. Endourol. 2000, 14 (1),
39–42. 31. Keogh, J.R.; Velander, F.F.; Eaton, J.W.
Albumin— binding surfaces for implantable devices. J.
Biomed. Mater. Res. 1992, 26, 441–456. 32. Nernst, W.Z.
Theorie der Reaktionsgeschwindigkeit in heterogenen
Systemen. Z. Phys. Chem. 1904, 47, 52–55. 33. Bustamente,
P.; Martin, A.; Gonzalez-Guisandez, M.A. Partial solubility
parameters and solvatochromic parameters for predicting the
solubility of single and multiple drugs in individual
solvents. J. Pharm. Sci. 1993, 82, 635–640. 34. Buchholz,
H.W.; Engelbrecht, E. U¨ber die Depotwirkung einiger
Antibiotika beim Vermischen mit dem Kunsthartz Palacos.
Chirurg 1970, 41, 511–515. 35. Bayston, R.; Milner, R.D.G.
The sustained release of antimicrobial drugs from bone
cement. J. Bone Jt. Surg. 1982, 64B, 460–464. 36. Flynn,
G.L. Influence of Physico-Chemical Properties of Drugs and
System of Release of Drugs from Inert Matrices. In
Controlled Release of Biologically Active Agents. Advances
in Experimental Biology and Medicine; Tanquary, C., Lacey,
R.E., Eds.; Plenum Pubs: New York, 1974; Vol. 47, 73–78.
37. von Eiff, C.; Lindner, N.; Proctor, R.A.; Winkelmann,
W.; Peters, G. Auftreten von Gentamicin-resistenten Small
Colony Variants von S. aureus nach Einsetzen von
Gentamicin-ketten bei Osteomyelitis als Mo¨gliche Ursache
fu¨r Rezidive. Z. Orthop. 1998, 136, 268–271. 38. Krajicek,
M.; Dvorak, J.; Chvapil, M. Infection-resistant synthetic
vascular substitutes. J. Cardiovasc. Surg. 1969, 10, 454.
39. Chervu, A.; Moore, W.S.; Chvapil, M.; Henderson, T.
Efficacy and duration of antistaphylococcal activity
comparing three antibiotics bonded to Dacron vascular
grafts with a collagen release system. J. Vasc. Surg. 1991,
13, 897–901. 40. Braithwaite, B.D.; Davies, B.; Heather,
B.P.; Earnshaw, J.J. Early results of a randomized trial of
rifampicinbonded grafts for extra-anatomic vascular
reconstruction. Joint Vascular Research Group. Br. J. Surg.
1998, 85, 1378–1381. 41. Widmer, A.F. Central Venous
Catheters. In CatheterRelated Infections; Seifert, H.,
Jansen, B., Farr, B., Eds.; Marcel Dekker, Inc.: New York,
1997. 42. Elliott, T.S. Role of antimicrobial central
venous catheters for the prevention of associated
infections. J. Antimicrob. Chemother. 1999, 43, 441–446.
43. Trooskin, S.Z.; Donetz, A.P.; Harvey, R.A.; Greco, R.J.
Prevention of catheter sepsis by antibiotic bonding.
Surgery 1985, 97, 547–551. 44. Jansen, B.; Jansen, S.;
Peters, G.; Pulverer, G. In vitro efficacy of a central
venous catheter (hydrocath) loaded with teicoplanin to
prevent bacterial colonisation. J. Hosp. Infect. 1992, 22,
93–107. 45. Bach, A.; Darby, D.; Bo¨ttiger, B.; Bo¨hrer,
H.; Motsch, J.; Martin, E. Retention of the antibiotic
teicoplanin on a hydromer-coated central venous catheter to
prevent bacterial colonization in postoperative surgical
patients. Intensive Care Med. 1996, 22, 1066–1069. 46.
Moss, H.A.; Tebbs, S.E.; Faroqui, M.H.; Herbst, T.; Isaac,
J.L.; Brown, J.; Elliott, T.S. A central venous catheter
coated with benzalkonium chloride for the prevention of
catheter-related microbial colonization. Eur. J.
Anaesthesiol. 2000, 17, 680–687. 47. Raad, I.; Darouiche,
R.; Dupuis, J.; Abi-Said, D.; Gabrielli, A.; Hachem, R.;
Wall, R.M.; Harris, R.; Jones, J.; Buzaid, A.; Robertson,
C.; Shenaq, S.; Curling, P.; Burke, T.; Ericsson, C.
Central venous catheters coated with minocycline and
rifampicin for the prevention of catheter-related
colonization and bloodstream infections. A randomized
double-blind trial. Ann. Intern. Med. 1997, 127, 267–274.
48. Guggenbichler, J.-P.; Bo¨swald, M.; Lugauer, S.; Krall,
T. A new technology of microdispersed silver in
polyurethane induces antimicrobial activity in central
venous catheters. Infection 1999, 27 (Suppl. 1), S16–S23.
49. Boelens, J.J.; Tan, W.-F.; Dankert, J.; Zaat, S.A.J.
Antibacterial activity of antibiotic-soaked
polyvinylpyrrolidone-grafted silicon (sic) elastomer
hydrocephalus shunts. J. Antimicrob. Chemother. 2000, 45,
221–224. 50. Boelens, J.J.; Zaat, S.A.J.; Meeldijk, J.;
Dankert, J. Subcutaneous abscess formation around catheters
induced by viable and nonviable Staphylococcus epidermidis
as well as by small amounts of bacterial cell wall
components. J. Biomed. Mater. Res. 2000, 50, 546–556. 51.
Bayston, R.; Grove, N.; Siegel, J.; Lawellin, D.; Barsham,
S. Prevention of hydrocephalus shunt catheter colonisation
in vitro by impregnation with antimicrobials. J. Neurol.
Neurosurg. Psychiatry 1989, 52, 605–609. 52. Bayston, R.;
Lambert, E. Duration of protective activity of
cerebrospinal fluid shunt catheters impregnated with
antimicrobial agents to prevent bacterial catheter
catheter-related infections. J. Neurosurg. 1997, 87,
247–251. 53. Bayston, R.; Ashraf, W.; Bhundia, C. Mode of
action of an antimicrobial shunt material. Eur. J. Pediatr.
Surg. 2002, 12, S56. 54. Rungby, J.; Hultman, P.;
Ellermann-Eriksen, S. Silver affects viability and
structure of cultured mouse peritoneal macrophages and
perioxidative capacity of whole mouse liver. Arch. Toxicol.
1987, 59, 408–412. 55. Van de Belt, H.; Neut, D.; van Horn,
J.R.; van der Mei, H.C.; Schenk, W.; Busscher, H.J. Or not
to treat? Nat. Med. 1999, 5, 358–359. 56. Avery, S.V. Metal
toxicity in yeasts and the role of oxidative stress. Adv.
Appl. Microbiol. 2001, 49, 111– 142. in press. 57. Modak,
K.; Fox, C. Binding of silver sulfadiazine in the cellular
components of Pseudomonas aeruginosa. Biochem. Pharmacol.
1973 , 22, 2392–2404. 58. Liedberg, H.; Lunderberg, T.
Silver alloy coated catheters reduce catheter-associated
bacteremia. Br. J. Urol. 1990, 65, 379–381. 59. Lundeberg,
T. Prevention of catheter-associated urinary tract
infections by use of silver-impregnated catheters. Lancet
1986, 2, 1031. 60. Riley, D.K.; Classen, D.C.; Stevens,
L.E.; Burke, J.P. A large randomised clinical trial of
silver-impregnated urinary catheter: Lack of efficacy and
staphylococcal superinfection. Am. J. Med. 1995, 98,
349–358. 61. Schierholz, J.M.; Wachol-Drebeck, L.; Lucas,
L.; Pulverer, G. Activity of silver ions in biological
fluids. Zbl. Bakteriol. 1998, 287, 411–420. 62. Johnson,
J.R.; Robert, P.L.; Olsen, R.J. Prevention of
catheters-associated urinary tract infection with a A
silver oxide-coated urinary catheter: Clinical and
microbiologic correlates. J. Infect. Dis. 1990, 162,
1145–1150.

63. Furr, J.R.; Russell, A.D.; Turner, T.D.; Andrews, A.


Antibacterial activity of Actisorb plus, Actisorb and
silver nitrate. J. Hosp. Infect. 1994, 27, 201–208.

64. Stickler, D.J. Biomaterials to prevent nosocomial


infections: Is silver the gold standard? Curr. Opin.
Infect. Dis. 2000, 13 (4), 389–393.

65. Darouiche, R.O. Anti-infective efficacy of silver-coated


medical prostheses. Clin. Infect. Dis. 1999, 29, 1371–
1377.

66. Thibon, P.; Le Coutour, X.; Leroyer, R.; Fabry, J.


Randomised multicentre trial of the effects of a catheter
coated with hydrogel and silver salts on the incidence of
hospital-acquired urinary tract infections. J. Hosp.
Infect. 2000, 45, 117–124. 67. Milder, F. Device-related
nosocomial infection: Reducing infection with antimicrobial
materials and coatings. Med. Device Technol. 1999, 10,
34–39. 68. Logghe, C.; van Ossel, C.; Hoore, C.; Ezzedine,
H.; Wauters, G.; Haxhe, J.J. Evaluation of chlorhexidine
and silver-sulfadiazine impregnated central venous
catheters for the prevention of bloodstream infection in
leukaemic patients: A randomized controlled trial. J. Hosp.
Infect. 1997, 37, 145–156. 69. Center for Devices and
Radiological Health. Potential Hypersensitivity Reactions
to Chlorhexidine-Impregnated Medical Devices; FDA Public
Health Notice, March 11, 1998.
Antimineralization Treatment

6. Kirsch, T. Determinants of pathological mineralization.


Curr. Opin. Rheumatol. 2006, 18 (2), 174–180.

7. Schoen, F.J.; Levy, R.J.; Nelson, A.C.; Bernhard, W.F.;


Nashef, A.; Hawley, M. Onset and progression of
experimental bioprosthetic heart valve calcification. Lab.
Invest. 1985, 52 (5), 523–532.

8. Valente, M.; Bortolotti, U.; Thiene, G. Ultrastructural


substrates of dystrophic calcification in porcine
bioprosthetic valve failure. Am. J. Pathol. 1985, 119 (1),
12–21.

9. Kim, K.M. Cell injury and calcification of rat aorta in


vitro. Scan. Electron Microsc. 1984, 4, 1809–1818.

10. Kim, K.M. Cells, rather than extracellular matrix,


nucleate apatite in glutaraldehyde-treated vascular tissue.
J. Biomed. Mater. Res. 2002, 59 (4), 639–645.

11. Kim, K.M. Cellular mechanism of calcification and its


prevention in glutaraldehyde treated vascular tissue. Z.
Kardiol. 2001, 90 (Suppl. 3), 99–105.

12. Schoen, F.J.; Tsao, J.W.; Levy, R.J. Calcification of


bovine pericardium used in cardiac valve bioprostheses.
Implications for the mechanisms of bioprosthetic tissue
mineralization. Am. J. Pathol. 1986, 123 (1), 134–145.

13. Anderson, H.C. Normal and abnormal mineralization in


mammals. Trans. Am. Soc. Artif. Intern. Organs 1981, 27,
702–708.

14. Levy, R.J.; Schoen, F.J.; Sherman, F.S.; Nichols, J.;


Hawley, M.A.; Lund, S.A. Calcification of subcutaneously
implanted type I collagen sponges. Effects of formaldehyde
and glutaraldehyde pretreatments. Am. J. Pathol. 1986, 122
(1), 71–82.

15. Vyavahare, N.; Ogle, M.; Schoen, F.J.; Levy, R.J.


Elastin calcification and its prevention with aluminum
chloride pretreatment. Am. J. Pathol. 1999, 155 (3),
973–982.

16. Lee, J.S.; Basalyga, D.M.; Simionescu, A.; Isenburg,


J.C.; Simionescu, D.T.; Vyavahare, N.R. Elastin
calcification in the rat subdermal model is accompanied by
up-regulation of degradative and osteogenic cellular
responses. Am. J. Pathol. 2006, 168 (2), 490–498.

17. Jian, B.; Narula, N.; Li, Q.Y.; Mohler, E.R., III;
Levy, R.J. Progression of aortic valve stenosis: TGF-beta1
is present in calcified aortic valve cusps and promotes
aortic valve interstitial cell calcification via apoptosis.
Ann. Thorac. Surg. 2003, 75 (2), 457–465.

18. LaBella, F.S.; Vivian, S.; Thornhill, D.P. Amino acid


composition of human aortic elastin as influenced by age. J.
Gerontol. 1966, 21 (4), 550–555.

19. Chen, C.C.; Boskey, A.L. Mechanisms of proteoglycan


inhibition of hydroxyapatite growth. Calcif. Tissue Int.
1985, 37 (4), 395–400.

20. Jorge-Herrero, E.; Fernandez, P.; Gutierrez, M.;


Castillo-Olivares, J.L. Study of the calcification of bovine
pericardium: Analysis of the implication of lipids and
proteoglycans. Biomaterials 1991, 12 (7), 683–689.

21. Lovekamp, J.J.; Simionescu, D.T.; Mercuri, J.J.;


Zubiate, B.; Sacks, M.S.; Vyavahare, N.R. Stability and
function of glycosaminoglycans in porcine bioprosthetic
heart valves. Biomaterials 2006, 27 (8), 1507–1518.

22. Vyavahare, N.; Ogle, M.; Schoen, F.J.; Zand, R.;


Gloeckner, D.C.; Sacks, M.; et al. Mechanisms of
bioprosthetic heart valve failure: Fatigue causes collagen
denaturation and glycosaminoglycan loss. J. Biomed. Mater.
Res. 1999, 46 (1), 44–50. 23. Mercuri, J.J.; Lovekamp,
J.J.; Simionescu, D.T.; Vyavahare, N.R.
Glycosaminoglycan-targeted fixation for improved
bioprosthetic heart valve stabilization. Biomaterials 2007,
28 (3), 496–503. 24. Mannschott, P.; Herbage, D.; Weiss,
M.; Buffevant, C. Collagen heterogeneity in pig heart
valves. Biochim. Biophys. Acta 1976, 434 (1), 177–183. 25.
Meuris, B.; Ozaki, S.; Herijgers, P.; Verbeken, E.;
Flameng, W. Influence of species, environmental factors, and
tissue cellularity on calcification of porcine aortic wall
tissue. Semin. Thorac. Cardiovasc. Surg. 2001, 13 (4 Suppl.
1), 99–105. 26. Jayakrishnan, A.; Jameela, S.R.
Glutaraldehyde as a fixative in bioprostheses and drug
delivery matrices. Biomaterials 1996, 17 (5), 471–484. 27.
Golomb, G.; Schoen, F.J.; Smith, M.S.; Linden, J.; Dixon,
M.; Levy, R.J. The role of glutaraldehyde-induced
cross-links in calcification of bovine pericardium used in
cardiac valve bioprostheses. Am. J. Pathol. 1987, 127 (1),
122–130. 28. David, T.E.; Ivanov, J.; Armstrong, S.;
Feindel, C.M.; Cohen, G. Late results of heart valve
replacement with the Hancock II bioprosthesis. J. Thorac.
Cardiovasc. Surg. 2001, 121 (2), 268–278. 29. Butany, J.;
Collins, M.J.; Nair, V.; Leask, R.L.; Scully, H.E.;
Williams, W.G.; et al. Morphological findings in explanted
Toronto stentless porcine valves. Cardiovasc. Pathol. Jan
2006, 15 (1), 41–48. 30. Fiddler, G.I.; Gerlis, L.M.;
Walker, D.R.; Scott, O.; Williams, G.J. Calcification of
glutaraldehydepreserved porcine and bovine xenograft valves
in young children. Ann. Thorac. Surg. 1983, 35 (3),
257–261. 31. Ishihara, T.; Ferrans, V.J.; Boyce, S.W.;
Jones, M.; Roberts, W.C. Structure and classification of
cuspal tears and perforations in porcine bioprosthetic
cardiac valves implanted in patients. Am. J. Cardiol. 1981,
48 (4), 665–678. 32. Thubrikar, M.J.; Deck, J.D.; Aouad,
J.; Nolan, S.P. Role of mechanical stress in calcification
of aortic bioprosthetic valves. J. Thorac. Cardiovasc.
Surg. 1983, 86 (1), 115–125. 33. Sabbah, H.N.; Hamid, M.S.;
Stein, P.D. Mechanical stresses on closed cusps of porcine
bioprosthetic valves: correlation with sites of
calcification. Ann. Thorac. Surg. 1986, 42 (1), 93–96. 34.
Vesely, I.; Krucinski, S.; Campbell, G. Micromechanics and
mathematical modeling: An inside look at bioprosthetic
valve function. J. Card. Surg. 1992, 7 (1), 85–95. 35.
Stein, P.D.; Kemp, S.R.; Riddle, J.M.; Lee, M.W.; Lewis,
J.W., Jr.; Magilligan, D.J., Jr. Relation of calcification
to torn leaflets of spontaneously degenerated porcine
bioprosthetic valves. Ann. Thorac. Surg. 1985, 40 (2),
175–180. 36. Ellsmere, J.C.; Khanna, R.A.; Lee, J.M.
Mechanical loading of bovine pericardium accelerates
enzymatic degradation. Biomaterials 1999, 20 (12),
1143–1150. 37. Smith, D.B.; Sacks, M.S.; Pattany, P.M.;
Schroeder, R. Fatigue-induced changes in bioprosthetic
heart valve three-dimensional geometry and the relation to
tissue damage. J. Heart Valve Dis. 1999, 8 (1), 25–33. 38.
Farzaneh-Far, A.; Proudfoot, D.; Shanahan, C.; Weissberg,
P.L. Vascular and valvar calcification: Recent advances.
Heart 2001, 85 (1), 13–17. 39. Srivatsa, S.S.; Harrity,
P.J.; Maercklein, P.B.; Kleppe, L.; Veinot, J.; Edwards,
W.D.; et al. Increased cellular expression of matrix
proteins that regulate mineralization is associated with
calcification of native human and porcine xenograft
bioprosthetic heart valves. J. Clin. Invest. 1997, 99 (5),
996–1009. 40. Levy, R.J.; Schoen, F.J.; Levy, J.T.; Nelson,
A.C.; Howard, S.L.; Oshry, L.J. Biologic determinants of
dystrophic calcification and osteocalcin deposition in
glutaraldehyde-preserved porcine aortic valve leaflets
implanted subcutaneously in rats. Am. J. Pathol. 1983, 113
(2), 143–155. 41. Levy, R.J.; Zenker, J.A.; Bernhard, W.F.
Porcine bioprosthetic valve calcification in bovine left
ventricleaorta shunts: Studies of the deposition of vitamin
K-dependent proteins. Ann. Thorac. Surg. 1983, 36 (2),
187–192. 42. Maranto, A.R.; Schoen, F.J. Alkaline
phosphatase activity of glutaraldehyde-treated bovine
pericardium used in bioprosthetic cardiac valves. Circ.
Res. 1988, 63 (4), 844–848. 43. Maranto, A.R.; Schoen, F.J.
Phosphatase enzyme activity is retained in glutaraldehyde
treated bioprosthetic heart valves. ASAIO Trans. 1988, 34
(3), 827–830. 44. Simionescu, A.; Simionescu, D.; Deac, R.
Biochemical pathways of tissue degeneration in
bioprosthetic cardiac valves. The role of matrix
metalloproteinases. ASAIO J. 1996, 42 (5), M561–M567. 45.
Dahm, M.; Lyman, W.D.; Schwell, A.B.; Factor, S.M.; Frater,
R.W. Immunogenicity of glutaraldehyde-tanned bovine
pericardium. J. Thorac. Cardiovasc. Surg. 1990, 99 (6),
1082–1090. 46. Rocchini, A.P.; Weesner, K.M.; Heidelberger,
K.; Keren, D.; Behrendt, D.; Rosenthal, A. Porcine
xenograft valve failure in children: an immunologic
response. Circulation 1981, 64 (2 Pt. 2), II162–II171. 47.
Levy, R.J.; Schoen, F.J.; Howard, S.L. Mechanism of
calcification of porcine bioprosthetic aortic valve cusps:
Role of T-lymphocytes. Am. J. Cardiol. 1983, 52 (5),
629–631. 48. Gong, G.; Seifter, E.; Lyman, W.D.; Factor,
S.M.; Blau, S.; Frater, R.W. Bioprosthetic cardiac valve
degeneration: role of inflammatory and immune reactions. J.
Heart Valve Dis. 1993, 2 (6), 684–693. 49. Hendriks, M.;
Everaerts, F.; Verhoeven, M. Alternative fixation of
bioprostheses. J. Long Term Eff. Med. Implants 2001, 11
(3-4), 163–183. 50. Gong, G.; Ling, Z.; Seifter, E.;
Factor, S.M.; Frater, R.W. Aldehyde tanning: The villain in
bioprosthetic calcification. Eur. J. Cardiothorac. Surg.
1991, 5 (6), 288–299. 51. Everaerts, F.; Gillissen, M.;
Torrianni, M.; Zilla, P.; Human, P.; Hendriks, M.; et al.
Reduction of calcification of carbodiimide-processed heart
valve tissue by prior blocking of amine groups with
monoaldehydes. J. Heart Valve Dis. 2006, 15 (2), 269–277.
52. Anselme, K.; Petite, H.; Herbage, D. Inhibition of
calcification in vivo by acyl azide cross-linking of a
collagen-glycosaminoglycan sponge. Matrix 1992, 12 (4),
264–273. 53. Simmons, D.M.; Kearney, J.N. Evaluation of
collagen cross-linking techniques for the stabilization of
tissue matrices. Biotechnol. Appl. Biochem. 1993, 17 (Pt.
1), 23–29. 54. Shen, S.H.; Sung, H.W.; Tu, R.; Hata, C.;
Lin, D.; Noishiki, Y.; et al. Characterization of a
polyepoxy compound fixed porcine heart valve bioprosthesis.
J. Appl. Biomater. 1994, 5 (2), 159–162. 55. Sung, H.W.;
Chang, Y.; Chiu, C.T.; Chen, C.N.; Liang, H.C. Mechanical
properties of a porcine aortic valve fixed with a naturally
occurring crosslinking agent. Biomaterials 1999, 20 (19),
1759–1772. 56. Goissis, G.; Yoshioka, S.A.; Braile, D.M.;
Ramirez, V.D. The chemical protecting group concept applied
in crosslinking of natural tissues with glutaraldehyde
acetals. Artif. Organs 1998, 22 (3), 210–214. 57. Vasudev,
S.C.; Chandy, T. Polyethylene glycol-grafted bovine
pericardium: a novel hybrid tissue resistant to
calcification. J. Mater. Sci. Mater. Med. 1999, 10 (2),
121–128. 58. Oosthuysen, A.; Zilla, P.P.; Human, P.A.;
Schmidt, C.A.; Bezuidenhout, D. Bioprosthetic tissue
preservation by filling with a poly(acrylamide) hydrogel.
Biomaterials 2006, 27 (9), 2123–2130. 59. Moore, M.A.;
Phillips, R.E. Biocompatibility and immunologic properties
of pericardial tissue stabilized by dye-mediated
photooxidation. J. Heart Valve Dis. 1997, 6 (3), 307–315.
60. Moore, M.A.; Bohachevsky, I.K.; Cheung, D.T.; Boyan,
B.D.; Chen, W.M.; Bickers, R.R.; et al. Stabilization of
pericardial tissue by dye-mediated photooxidation. J.
Biomed. Mater. Res. 1994, 28 (5), 611–618. 61. Meuris, B.;
Phillips, R.; Moore, M.A.; Flameng, W. Porcine stentless
bioprostheses: Prevention of aortic wall calcification by
dye-mediated photo-oxidation. Artif. Organs 2003, 27 (6),
537–543. 62. Schoen, F.J. Pathologic findings in explanted
clinical bioprosthetic valves fabricated from photooxidized
bovine pericardium. J. Heart Valve Dis. 1998, 7 (2),
174–179. 63. Jones, M.; Eidbo, E.E.; Hilbert, S.L.;
Ferrans, V.J.; Clark, R.E. The effects of anticalcification
treatments on bioprosthetic heart valves implanted in
sheep. ASAIO Trans. 1988, 34 (4), 1027–1030. 64. Hirsch,
D.; Drader, J.; Thomas, T.J.; Schoen, F.J.; Levy, J.T.;
Levy, R.J. Inhibition of calcification of glutaraldehyde
pretreated porcine aortic valve cusps with sodium dodecyl
sulfate: Preincubation and controlled release studies. J.
Biomed. Mater. Res. 1993, 27 (12), 1477–1484. 65. Cunanan,
C.M.; Cabiling, C.M.; Dinh, T.T.; Shen, S.H.; Tran-Hata,
P.; Rutledge, J.H., III; et al. Tissue characterization and
calcification potential of commercial bioprosthetic heart
valves. Ann. Thorac. Surg. 2001, 71 (5 Suppl.), S417–S421.
66. Flameng, W.; Meuris, B.; Yperman, J.; De, V.G.;
Herijgers, P.; Verbeken, E. Factors influencing A
calcification of cardiac bioprostheses in adolescent sheep.
J. Thorac. Cardiovasc. Surg. 2006, 132 (1), 89–98.

67. Jorge-Herrero, E.; Fernandez, P.; de la, T.N.;


Escudero, C.; Garcia-Paez, J.M.; Bujan, J.; et al.
Inhibition of the calcification of porcine valve tissue by
selective lipid removal. Biomaterials 1994, 15 (10),
815–820.

68. Jorge-Herrero, E.; Fernandez, P.; Escudero, C.; de la


Torre, N.; Zurita, M.; Ga´rcia Pa´ez, J.M.;
CastilloOlivares, J.L. Influence of stress on calcification
of delipidated bovine pericardial tissue employed in
construction of cardiac valves. J. Biomed. Mater. Res.
1996, 30 (3), 411–415.

69. Vyavahare, N.R.; Hirsch, D.; Lerner, E.; Baskin, J.Z.;


Zand, R.; Schoen, F.J.; et al. Prevention of calcification
of glutaraldehyde-crosslinked porcine aortic cusps by
ethanol preincubation: Mechanistic studies of protein
structure and water–biomaterial relationships. J. Biomed.
Mater. Res. 1998, 40 (4), 577–585.

70. Gott, J.P.; Pan, C.; Dorsey, L.M.; Jay, J.L.; Jett,
G.K.; Schoen, F.J.; et al. Calcification of porcine valves:
A successful new method of antimineralization. Ann. Thorac.
Surg. 1992, 53 (2), 207–215.

71. Gott, J.P.; Girardot, M.N.; Girardot, J.M.; Hall, J.D.;


Whitlark, J.D.; Horsley, W.S.; et al. Refinement of the
alpha aminooleic acid bioprosthetic valve anticalcification
technique. Ann. Thorac. Surg. 1997, 64 (1), 50–58.

72. Chen, W.; Kim, J.D.; Schoen, F.J.; Levy, R.J. Effect of
2-amino oleic acid exposure conditions on the inhibition of
calcification of glutaraldehyde cross-linked porcine aortic
valves. J. Biomed. Mater. Res. 1994, 28 (12), 1485–1495.

73. Webb, C.L.; Schoen, F.J.; Flowers, W.E.; Alfrey, A.C.;


Horton, C.; Levy, R.J. Inhibition of mineralization of
glutaraldehyde-pretreated bovine pericardium by AlCl 3 .
Mechanisms and comparisons with FeCl 3 , LaCl 3 , and Ga(NO
3 ) 3 in rat subdermal model studies. Am. J. Pathol. 1991,
138 (4), 971–981.

74. Levy, R.J.; Schoen, F.J.; Flowers, W.B.; Staelin, S.T.


Initiation of mineralization in bioprosthetic heart valves:
Studies of alkaline phosphatase activity and its inhibition
by AlCl 3 or FeCl 3 preincubations. J. Biomed. Mater. Res.
1991, 25 (8), 905–935.

75. Clark, J.N.; Ogle, M.F.; Ashworth, P.; Bianco, R.W.;


Levy, R.J. Prevention of calcification of bioprosthetic
heart valve cusp and aortic wall with ethanol and aluminum
chloride. Ann. Thorac. Surg. 2005, 79 (3), 897–904. 76.
Ogle, M.F.; Kelly, S.J.; Bianco, R.W.; Levy, R.J.
Calcification resistance with aluminum-ethanol treated
porcine aortic valve bioprostheses in juvenile sheep. Ann.
Thorac. Surg. 2003, 75 (4), 1267–1273. 77. Levy, R.J.;
Schoen, F.J.; Lund, S.A.; Smith, M.S. Prevention of leaflet
calcification of bioprosthetic heart valves with
diphosphonate injection therapy. Experimental studies of
optimal dosages and therapeutic durations. J. Thorac.
Cardiovasc. Surg. 1987, 94 (4), 551–557. 78. Webb, C.L.;
Benedict, J.J.; Schoen, F.J.; Linden, J.A.; Levy, R.J.
Inhibition of bioprosthetic heart valve calcification with
aminodiphosphonate covalently bound to residual aldehyde
groups. Ann. Thorac. Surg. 1988, 46 (3), 309–316. 79.
Isenburg, J.C.; Simionescu, D.T.; Vyavahare, N.R. Tannic
acid treatment enhances biostability and reduces
calcification of glutaraldehyde fixed aortic wall.
Biomaterials 2005, 26 (11), 1237–1245. 80. O’Brien, M.F.;
Goldstein, S.; Walsh, S.; Black, K.S.; Elkins, R.; Clarke,
D. The SynerGraft valve: A new acellular
(nonglutaraldehyde-fixed) tissue heart valve for autologous
recellularization first experimental studies before clinical
implantation. Semin. Thorac. Cardiovasc. Surg. 1999, 11 (4
Suppl. 1), 194–200. 81. Sharp, M.A.; Phillips, D.; Roberts,
I.; Hands, L. A cautionary case: The SynerGraft vascular
prosthesis. Eur. J. Vasc. Endovasc. Surg. 2004, 27 (1),
42–44. 82. Meuris, B.; Verbeken, E.; Flameng, W. Prevention
of porcine aortic wall calcification by acellularization:
Necessity for a non-glutaraldehyde-based fixation treatment.
J. Heart Valve Dis. 2005, 14 (3), 358–363. 83. Walles, T.;
Puschmann, C.; Haverich, A.; Mertsching, H. Acellular
scaffold implantation—No alternative to tissue engineering.
Int. J. Artif. Organs 2003, 26 (3), 225–234. 84. Neethling,
W.M.; van Rijn, R.S.; Van Den Heever, J.J.; Hough, J.;
Hodge, A.J. Evaluation of kangaroo aortic valved conduits
in a juvenile sheep model: Preliminary findings. J. Card.
Surg. 2001, 16 (5), 392–399. 85. Imai, Y.; Takanashi, Y.;
Hoshino, S.; Nakata, S. The equine pericardial valved
conduit and current strategies for pulmonary
reconstruction. Semin. Thorac. Cardiovasc. Surg. 1995, 7
(3), 157–161. 86. Herijgers, P.; Ozaki, S.; Verbeken, E.;
Van, L.A.; Meuris, B.; Lesaffre, E.; et al. Valved jugular
vein segments for right ventricular outflow tract
reconstruction in young sheep. J. Thorac. Cardiovasc. Surg.
2002, 124 (4), 798–805.
Articular Cartilage Biomechanics

11. Wayne, J.S.; Woo, S.L.-Y.; Kwan, M.K. Application of


the U-P finite element method to the study of articular
cartilage. J. Biomech. Eng. 1991, 113, 397–403.

12. Spilker, R.L.; Suh, J.K.; Mow, V.C. Effects of friction


on the unconfined compressive response of articular
cartilage: a finite element analysis. J. Biomech. Eng. 1990,
112 (2), 138–146.

13. Wu, J.; Herzog, W.; Epstein, M. Evaluation of the finite


element software ABAQUS for biomechanical modeling of
biphasic tissues. J. Biomech. 1998, 31, 165–169.

14. Mak, A.F. The apparent viscoelastic behavior of


articular cartilage—the contributions from the intrinsic
matrix viscoelasticity and interstitial fluid flows. J.
Biomech. Eng. 1986, 108 (2), 123–130.

15. Soltz, M.A.; Ateshian, G.A. A conewise linear


elasticity mixture model for the analysis of
tension-compression nonlinearity in articular cartilage. J.
Biomech. Eng. 2000, 122 (6), 576–586. 16. Soulhat, J.;
Buschmann, M.D.; Shirazi-Adl, A. A fibrilnetwork-reinforced
biphasic model of cartilage in unconfined compression. J.
Biomech. Eng. 1999, 121 (3), 340–347. 17. Wilson, W.; van
Donkelaar, C.C.; van Rietbergen, C.; Ito, K.; Huiskes, R.
Stresses in the local collagen network of articular
cartilage: a poroviscoelastic fibrilreinforced finite element
study. J. Biomech. 2004, 37 (3), 357–366. 18. Lai, W.M.;
Hou, J.S.; Mow, V.C. A triphasic theory for the swelling
and deformation behaviors of articular cartilage. J.
Biomech. Eng. 1991, 113 (3), 245–258. 19. Frank, E.H.;
Grodzinsky, A.J. Cartilage electromechanics–II. A continuum
model of cartilage electrokinetics and correlation with
experiments. J. Biomech. 1987, 20 (6), 629–639.
Artificial Heart Fluid Dynamics: Positive
Displacement Pumps

14. Yoganathan, A.P.; He, Z.; Jones, S.C. Fluid mechanics


of heart valves. Annu. Rev. Biomed. Eng. 2004, 6, 331–362.

15. National Heart, Lung, and Blood Institute Working


Group. Guidelines for blood materials interaction. NIH Pub.
85-2185.78 Natl Heart Lung Blood Inst.; Bethesda, MD: 1985.

16. Nevaril, C.; Hellums, J.; Alfrey, C.J.; Lynch, E.


Physical effects in red blood cell trauma. Am. Inst. Chem.
Eng. J. 1969, 15, 707–711.

17. Sallam, A.M.; Hwang, N.H.C. Human red blood cell


hemolysis in a turbulent shear flow: contribution of
Reynolds shear stresses. Biorheology 1984, 21, 783–797.

18. Wootton, D.M.; Ku, D.N. Fluid mechanics of vascular


systems, diseases, and thrombosis. Annu. Rev. Biomed. Eng.
1999, 1, 299–329.

19. Hubbell, J.A.; McIntire, L.V. Visualization and


analysis of mural thrombogenesis on collagen, polyurethane
and nylon. Biomaterials 1986, 7, 354–363.

20. Daily, B.B.; Pettitt, T.W.; Sutera, S.P.; Pierce, W.S.


Pierce-Donachy pediatric VAD: progress in development. Ann.
Thorac. Surg. 1996, 61, 437–443.

21. Lenker, J.A. Flow studies in artificial hearts and LVAD:


an application of flow visualization analysis. PhD thesis,
Pennsylvania State Univ; 1978.

22. Phillips, W.M.; Furkay, S.S.; Pierce, W.S. Laser


Doppler anemometer studies in unsteady ventricular flows.
ASAIO Trans. 1979, 25, 56–60.

23. Affeld, A. The state of the art of the Berlin total


artificial heart—technical aspects. In Assisted Circulation;
Unger, F., Ed.; Springer: New York, 1979; 307–333.

24. Mann, K.A.; Deutsch, S.; Tarbell, J.M.; Geselowitz,


D.B.; Rosenberg, G.; Pierce, W.S. An experimental study of
Newtonian and non-Newtonian Flow dynamics in a ventricular
assist device. J. Biomech. Eng. 1987, 109, 139–147.

25. Tarbell, J.M.; Gunshinan, J.P.; Geselowitz, D.B.;


Rosenberg, G.; Shung, K.K.; Pierce, W.S. Pulsed ultrasonic
Doppler velocity measurements inside a left ventricular
assist device. J. Biomech. Eng. 1986, 108, 232–238.

26. Jarvis, P.; Tarbell, J.M.; Frangos, J.A. An in vitro


analysis of an artificial heart. ASAIO Trans. 1991, 37,
27–32.

27. Baldwin, J.T.; Tarbell, J.M.; Deutsch, S.; Geselowitz,


D.B.; Rosenberg, G. Hot-film wall shear probe measurements
inside a ventricular assist device. J. Biomech. Eng. 1988,
110, 326–333.

28. Francischelli, D.E.; Tarbell, J.M.; Geselowitz, D.B.


Local blood residence times in the Penn State artificial
heart. Artif. Organs 1991, 15, 218–224.

29. Mussivand, T.; Navarro, R.; Chen, J.F. Flow


visualization in an artificial heart using diffuse and
planar lighting. ASAIO Trans. 1988, 34, 317–321.

30. Woodward, J.; Shaffer, F.; Schaub, R.; Lund, L.;


Borovetz, H. Optimal management of a ventricular assist
device. ASAIO J. 1992, 38, M216–M219.

31. Baldwin, J.T.; Tarbell, J.M.; Deutsch, S.; Geselowitz,


D.B. Mean flow velocity patterns within a ventricular assist
device. ASAIO Trans. 1989, 35, 429–433. 32. Baldwin, J.T.;
Deutsch, S.; Geselowitz, D.B.; Tarbell, J.M. Estimation of
Reynolds stresses within the Penn State left ventricular
assist device. ASAIO Trans. 1990, 3, M274–M278. 33.
Baldwin, J.T.; Deutsch, S.; Petrie, H.L.; Tarbell, J.M.
Determination of principal Reynolds stresses in pulsatile
flows after elliptical filtering of discrete velocity
measurements. J. Biomech. Eng. 1993, 115, 396–403. 34. Jin,
W.; Clark, C. Experimental investigation of unsteady-flow
behavior within a sac-type ventricular assist device (VAD).
J. Biomech. 1993, 26, 697–707. 35. Yoganathan, A.P.; Woo,
Y.R.; Sung, H.W. Turbulent shear stress measurements in the
vicinity of aortic heart valve prostheses. J. Biomech.
1986, 19, 422–433. 36. Maymir, J.C.; Deutsch, S.; Meyer,
R.; Geselowitz, D.B.; Tarbell, J.M. Effects of tilting disk
valve gap width on regurgitant flow through an artificial
heart mitral valve. Artif. Organs 1997, 21, 1014–1025. 37.
Maymir, J.C.; Deutsch, S.; Meyer, R.; Geselowitz, D.B.;
Tarbell, J.M. Mean velocity and Reynolds stress
measurements in the regurgitant jets of tilting disk heart
valves in an artificial heart environment. Ann. Biomed. Eng.
1988, 26, 146–156. 38. Meyer, R.S.; Deutsch, S.; Maymir,
J.C.; et al. Three component LDV measurements in the
regurgitant flow region of a Bjork-Shiley monostrut mitral
valve. Ann. Biomed. Eng. 1997, 25, 1081–1091. 39. Meyer,
R.S.; Deutsch, S.; Bachmann, C.B.; Tarbell, J.M. Laser
Doppler velocimetry and flow visualization studies in the
regurgitant leakage flow region of three mechanical heart
valves. Artif. Organs 2001, 25, 292– 299. 40. Leuer, L. In
vitro evaluation of drive parameters and valve selection
for the total artificial heart. Proceedings of International
Symposium of Artificial Organs, Biomedical Engineering and
Transplantation, Salt Lake City, Utah: 1986; Univ. Utah:
Salt Lake City, 1986. 41. Quijano, R. Edwards-Duromedics
dysfunctional analysis. Proceedings of 6th Conference on
Cardiostimulation, Monte Carlo, 1988. 42. Walker, W.
Cavitation in pulsatile blood pumps; ASME: New York, 1974;
148–150. 43. Young, F.R. Cavitation; McGraw-Hill: London,
1989. 44. Zapanta, C.M.; Stinebring, D.R.; Sneckenberger,
D.S.; et al. In vivo observation of cavitation on
prosthetic heart valves. ASAIO J. 1996, 42, M550–M555. 45.
Kafesian, R.; Howanec, M.; Ward, G.D.; Diep, L.; Wagstaff,
L.S.; Rhee, R. Cavitation damage of pyrolytic carbon in
mechanical heart valves. J. Heart Valve Dis. 1994, 3,
52–57. 46. Lamson, T.C.; Rosenberg, G.; Deutsch, S.; et al.
Relative blood damage in the three phases of a prosthetic
heart valve flow cycle. ASAIO J. 1993, 39, M626–M633. 47.
Dauzat, M.; Deklunder, G.; Aldis, A.; Rabinovitch, M.;
Burte, F.; Bret, P.M. Gas bubble emboli detected by
transcranial Doppler sonography in patients with prosthetic
heart valves: a preliminary report. J. Ultrasound Med.
1994, 13, 129–135. 48. Bachmann, C.; Kini, V.; Deutsch, S.;
et al. Mechanisms of cavitation and the formation of stable
bubbles on the Bjork-Shiley monostrut prosthetic heart
valve. J. Heart Valve Dis. 2001, 11, 105–113. 49.
Biancucci, B.; Deutsch, S.; Geselowitz, D.B.; Tarbell, J.M.
In vitro studies of gas bubble formation by mechanical
heart valves. J. Heart Valve Dis. 1999, 8, 186–196. 50.
Lin, H.Y.; Biancucci, B.; Deutsch, S.; Fontaine, A.A.;
Tarbell, J.M. Observation and quantification of gas bubble
formation on a mechanical heart valve. J. Biomech. Eng.
2000, 122, 304–309. 51. Graf, T.; Reul, H.; Detlefs, C.;
Wilmes, R.; Rau, G. Causes and formation of cavitation in
mechanical heart valves. J. Heart Valve Dis. 1994, 1,
S49–S64. 52. Chandran, K.B.; Aluri, S. Mechanical valve
closing dynamics: relationship between velocity of closing,
pressure transients, and cavitation initiation. Ann.
Biomed. Eng. 1997, 25, 926–938. 53. Bachmann, C.; Hugo, G.;
Rosenberg, G.; et al. Fluid dynamics of a pediatric
ventricular assist device. Artif. Organs 2000, 24, 362–372.
[Erratum Artif. Organs.; 24:989]. 54. Gharib, M.; Rambod,
E.; Shiota, T.; et al. Dynamic filling characteristics of
the left ventricle of the heart. Presented at 3rd
International Symposium Biofluid Mechanics, Munich, VDI
Verlag: Dusseldorf, 1994; 343–345 55. Park, Y.; Kim, S.
Development and animal study of a pediatric ventricular
assist device. Yonsei Med. J. 1988, 39, 154–158. 56.
Taenaka, Y.; Takano, H.; Noda, H.; Kinoshita, M. A
pediatric ventricular assist device: its development and
experimental evaluation of hemodynamic effects on
postoperative heart failure of congenital heart diseases.
Artif. Organs 1990, 14, 49–56. 57. Takano, H.; Nakatani, T.
Ventricular assist systems: experience in Japan with Toyobo
pump and Zeon pump. Ann. Thorac. Surg. 1996, 61, 317–322.
58. Konertz, W.; Holger, H.; Schneider, M.; et al. Clinical
experience with the MEDOSHIA–VAD system in infants and
children: a preliminary report. Ann. Thorac. Surg. 1997,
63, 1138–1144. 59. Konertz, W.; Reul, H. Mechanical
circulatory support in children. Artif. Organs 1997, 20,
657–658. 60. Hochareon, P.; Manning, K.B.; Fontaine, A.A.;
Tarbell, J.M.; Deutsch, S. Wall shear rate estimation
within the 50 cc Penn State artificial heart using particle
image velocimetry. J. Biomech. Eng. 2004, 126, 430–437. 61.
Hochareon, P.; Manning, K.B.; Fontaine, A.A.; Tarbell,
J.M.; Deutsch, S. Fluid dynamic analysis of the 50 cc Penn
State artificial heart under physiological operating
conditions using particle image velocimetry. J. Biomech.
Eng. 2004, 126, 585–593. 62. Hochareon, P.; Manning, K.B.;
Fontaine, A.A.; et al. Correlation of in vivo clot
deposition with the flow characteristics in the 50 cc Penn
State artificial heart: a preliminary study. ASIAO J. 2004,
50, 537–542. 63. Oley, L.A.; Manning, K.B.; Fontaine, A.A.;
Deutsch, S. Off design considerations of the 50 cc Penn
State ventricular assist device. Artif. Organs 2005, 29,
378–386. 64. Jin, W.; Clark, C. Experimental investigation
of the pumping diaphragm within a sac-type pneumatically
driven ventricular assist device. J. Biomech. 1994, 27,
43–55. 65. Yamanaka, H.; Rosenberg, G.; Weiss, W.J.; et al.
A multiscale surface evaluation of thombosis in left
entricular assist systems. ASAIO J. 2003, 49, 222. 66.
Reul, H. Overview of Rotary Blood Pump Designs; Presented
at Amer. Soc. Artif. Inter. Organs, 49th: Washington, DC,
2003. A
Artificial Muscles

20. Zhang, Q.M.; Bharti, V.; Zhao, X. Giant


electrostriction and relaxor ferroelectric behavior in
electronirradiated poly(vinylidene fluoride-trifluorethylene)
copolymer. Science 1998, 280, 2101–2104.

21. Perline, R.; Kornbluh, R.; Joseph, J.P.


Electrostriction of polymer dielectrics with compliant
electrodes as a means of actuation. Sens. Actuators, 4
1998, 64, 77–85.

22. Liang, C.; Rogers, C.A. One-dimensional thermometrical


constitutive relations of shape memory materials. J.
Intell. Mater. Syst. Struct., 1991, 1 (2), 207–234.

23. Liang, C.; Rogers, C.A. Design of shape memory alloy


actuators. ASME J. Mech. Des. 1992, 114, 223–230.

24. Liang, C.; Rogers, C.A.; Malafeew, E. Investigation of


shape memory polymers and their hybrid composites. J.
Intell. Mater. Syst. Struct. 1997, 8, 380.

25. Wang, G.; Shahinpoor, M. Design for shape memory alloy


rotatory joint actuators using shape memory effect and
pseudoelastic effect. Smart Mater. Technol., SPIE
Publication No. 1997, 3040, 23–30.

26. Wang, G.; Shahinpoor, M. Design, prototyping and


computer simulation of a novel large bending actuator made
with a shape memory alloy contractile wire. Smart Mater.
Struc., Int. J 1997, 6 (2), 214–221.

27. Wang, G.; Shahinpoor, M. A new design for a rotatory


joint actuator made with shape memory alloy contractile
wire. Int. J. Intell. Mater. Syst. Struct. March 1997, 8
(3), 191–279.

28. Wang, G. A General Design of Bias Force Shape Memory


Alloy (BFSMA) Actuators and An Electrically-Controlled SMA
Knee and Leg Muscle Exerciser for Paraplegics and
Quadriplegics. In Ph.D.; Department of Mechanical
Engineering, University of New Mexico: Albuquerque, NM, May
1998.

29. Zrinyi, M.; Szabo, D.; Feher, J. In Comparative Studies


of Electroand Magnetic Field Sensitive Polymer Gels,
Proceedings of the SPIE’s 6th Annual International
Symposium on Smart Structures and Materials, EAPAD Conf.,
SPIE Proc., Bar-Cohen, Y., Ed.; 1999; Vol. 3669, 406–413.
30. Dapino, M.; Flatau, A.; Calkins, F. In Statistical
Analysis of Terfenol-D Material Properties, Proceedings of
SPIE 1997 Symposium on Smart Structures and Materials, San
Diego, CA, March 1997; Vol. 3041, 256–267.

31. O’Handley, R.C. Model for strain and magnetostriction


in magnetic shape memory alloys. J. Appl. Phys. 1998, 83,
3263.

32. Sen, A.; Scheinbeim, J.I.; Newman, B.A. The effect of


plasticizer on the polarization of poly(vinylidene fluoride)
films. J. Appl. Phys. 1984, 56 (9), 2433–2439.

33. Furukawa, T.; Seo, N. Electrostriction as the origin of


piezoelectricity in ferroetectric polymers. Jpn. J. Appl.
Phys. 1990, 29 (4), 675–680.

34. Pelrine, R.; Kornbluh, R.; Pei, Q.; Joseph, J. High


speed electrically actuated elastomers with strain greater
than 100%. Science 2000, 287, 836–839.

35. Finkelmann, H.; Kock, H.J.; Rehage, G. Investigations


on liquid crystalline siloxanes: 3. Liquid crystalline
elastomer—A new type of liquid crystalline material.
Makromol. Chem., Rapid Commun. 1981, 2, 317–323. 36. Ratna,
B.R.; Selinger, J.V.; Srinivasan, A.; Hong, J.; Naciri, J.
In Namatic Elastomers as Artificial Muscles, Proceedings of
the First World Congress on Biometrics and Artificial
Muscles, Albuquerque Conventional Center, Albuquerque, New
Mexico, USA, December 9–11, 2002, 2003; 1. 37. Finkelmann,
H.; Shahinpoor, M. In Electrically-Controllable Liquid
Crystal Elastomer-Graphite Composites Artificial Muscles,
Proceeding of SPIE 9th Annual International Symposium on
Smart Structures and Materials, 2002, San Diego,
California, SPIE Publication No. 4695-53. 38. Schreyer,
H.B.; Gebhart, N.; Kim, K.J.; Shahinpoor, M. Electric
activation of artificial muscles containing
polyacrylonitrite gel fibers. Biomacromolecules J., ACS
Publications 2000, 1, 642–647. 39. Osada, Y.; Ross-Murphy,
S. Intelligent gels. Sci. Am. 1993, 268, 82–87. 40. Gong,
J.P.; Nitta, T.; Osada, Y. Electrokinetic modeling of the
contractile phenomena of polyelectrolyte gels-one
dimensional capillary model. J. Phys. Chem. 1994, 98,
9583–9587. 41. Osada, Y.; Hasebe, M. Electrically activated
mechanochemical devices using polyelectrolyte gels. Chem.
Lett. 1985, 12, 1285–1288. 42. Osada, Y.; Kishi, R.
Reversible volume change of microparticles in an electric
field. J. Chem. Soc. 1989, 85, 662–665. 43. Hirai, M.;
Hirai, T.; Sukumoda, A.; Nemoto, H.; Amemiya, Y.;
Kobayashi, K.; Ueki, T. Electrically induced reversible
structural change of a highly swollen polymer gel network.
J. Chem. Soc., Faraday Trans. 1995, 91, 473–477. 44. Hirai,
T.; Zheng, J.; Watanabe, M. In Solvent-Drag Bending Motion
of Polymer Gel Induced by an Electric Field, Proceedings of
the SPIE’s 6’h Annual International Symposium on Smart
Structures and Materials, Bar-Cohen, Y., Ed.; 1999; Vol.
3669, 209–217. 45. Oguro, K.; Kawami, Y.; Takenaka, H.
Bending of an ion-conducting polymer film-electrode
composite by an electric stimulus at low voltage. Trans. J.
Micromach. Soc. 1992, 5, 27–30. 46. Sadeghipour, K.;
Salomon, R.; Neogi, S. Development of a novel
electrochemically active membrane and smart material based
vibration sensor/damper. Smart Mater. Struc. 1992, 1 (2),
172–179. 47. Heitner-Wirguin, C. Recent advances in
perfluorinated ionomer membranes: Structure, properties and
applications. J. Membr. Sci. 1996, 120 (1), 1–33. 48. Kim,
K.J.; Shahinpoor, M. Application of Polyelectrolytes in
Ionic Polymeric Sensors, Actuators, and Artificial Muscles.
In Handbook of Polyelectrolytes; Tripathy, S., Nalwa, H.S.,
Eds.; Mindspring/Academic Press, 2002. in print, Review.
49. Kim, K.J.; Shahinpoor, M. A novel method of
manufacturing three-dimensional ionic polymer-metal
composites (IPMC’s) biomimetic sensors, actuators and
artificial muscle. Polymer 2002, 43/3, 797–802. 50. Gandhi,
M.R.; Murray, P.; Spinks, G.M.; Wallace, G.G. Mechanisms of
electromechanical actuation in polypyrrole. Synth. Met.
1995, 75, 247–256. 51. Schreyer, H.B.; Shahinpoor, M.; Kim,
K.J. In Electrical Activation of PAN-Pt Artificial Muscles,
Proceedings of SPIE/Smart Structures and
Materials/Electroactive Polymer Actuators and Devices,
Newport Beach, CA, March 1999; Vol. 3669, 192–198. 52. Hu,
X. Molecular structure of polyacrylonitrile fibers. J. Appl.
Polym. Sci. 1996, 62, 1925–1932. 53. Umemoto, S.; Okui, N.;
Sakai, T. Contraction Behavior of Poly(acrylonitrile) Gel
Fibers, Polymer Gels; DeRossi, D., et al., Eds.; Plenum
Press: New York, 1991; 257–270. 54. De Gennes, P.G.;
Okumura, K.; Shahinpoor, M.; Kim, K.J. Mechanoelectric
effects in ionic gels. Europhys. Lett. 2000, 50 (4),
513–518. 55. Takenaka, H.; Torikai, E.; Kawami, Y.;
Wakabayshi, N. Solid polymer electrolyte water
electrolysis. Int. J. Hydrogen Energy 1982, 7, 397–403. 56.
Millet, P.; Pinneri, M.; Durand, R. New solid polymer
electrolyte composites for water electrolysis. J. Appl.
Electrochem. 1989, 19, 162–166. 57. Peace, G.S. Taguchi
Methods: Hands-On Approach; Addison-Wesley Publishing
Company, Inc.: New York, 1993. 58. Adolf, D.; Shahinpoor,
M.; Segalman, D.; Witkowski, W. Electrically Controlled
Polymeric Gel Actuators. U.S. Patent #5,250,167, October
13, 1993. 59. Brock, D.L. Review of Artificial Muscle Based
on Contractile Polymers; AIMemo No. 1330, MIT, Artificial
Intelligence Lab. 1991. 60. De Rossi, D.E.; Chiarelli, P.;
Buzzigoli, G.; Domenici, C.; Lazzeri, L. Contractile
behavior of electrically activated mechanochemical polymer
actuators. Trans. Am. Soc. Artif. Intern. Organs 1986, 32,
157–163. 61. De Rossi, D.; Suzuki, M.; Osada, Y.; Morasso,
P. Pseudomuscular gel actuators for advanced robotics. J.
Intell. Mater. Syst. Struct. 1992, 3, 75–95. 62. Doi, M.;
Matsumoto, M.; Hirose, Y. Deformation of ionic polymer gels
by electric fields. Macromolecules 1992, 25, 5504–5511. 63.
Hamlen, R.P.; Kent, C.E.; Shafer, S.N. Electrolytically
activated contractile polymer. Nature 1965, 206, 1149–
1150. 64. Nemat-Nasser, S.; Li, J.Y. Electromechanical
response of ionic polymer-metal composites. J. Appl. Phys.
2000, 87 (7), 3321–3331. 65. Oguro, K.; Kawami, Y.;
Takenaka, H. Actuator Element. U.S. Patent #5,268,082,
1993. 66. Oguro, K.; Fujiwara, N.; Asaka, K.; Onishi, K.;
Sewa, S. Polymer Electrolyte Actuator with Gold Electrodes,
Proceedings of the SPIE’s 6th Annual International
Symposium on Smart Structures and Materials, SPIE Proc.,
1999; Vol. 3669, 64–71. 67. Pei, Q.; Inganas, O.;
Lundstrom, I. Bending bilayer strips built from polyaniline
for artificial electrochemical muscles. Smart Mater. Struc.
1993, 2 (1), 1–5. 68. Segalman, D.; Witkowski, W.; Adolf,
D.; Shahinpoor, M. In Electrically Controlled Polymeric
Muscles as Active Materials Used in Adaptive Structures,
Proceedings of ADPA/AIAA/ASME/SPIE Conference on Active
Materials and Adaptive Structures, Alexandria, VA, Nov.
1991. 69. Tanaka, T.; Nishio, I.; Sun, S.T.; Nishio, S.U.
Collapse of gels in an electric field. Science 1982, 218,
467–469. 70. Woojin, L. Polymer Gel Based Actuator: Dynamic
Model of Gel for Real Time Control. In Ph.D. Thesis;
Massachusetts Institute of Technology, 1996. 71.
Finkelmann, H.; Brand, H.R. Liquid crystalline elastomers—A
class of materials with novel properties. Trends Polym.
Sci. 1994, 2, 222. 72. Brand, H.R.; Finkelmann, H. Physical
Properties of Liquid Crystalline Elastomers. In Handbook of
Liquid Crystals Vol. 3: High Molecular Weight Liquid
Crystals; Demus, D., Goodby, J., Gray, G.W., Spiess, H.-W.,
Vill, V., Eds.; Wiley-VCH: Weinheim, 1998; 277–289. 73.
Brandt, H.R.; Pleiner, H. Electrohydrodynamics of nematic
liquid crystalline elastomers. Physica 1994, A208, 359. 74.
De Gennes, P.G.; Hebert, M.; Kant, R. Artificial muscles
based on nematic gels. Macromol. Symp. 1997, 113, 39. 75.
Hebert, M.; Kant, R.; de Gennes, P.G. Dynamics and
thermodynamics of artificial muscles based on nematic gels.
J. Phys., I, France 1997, 7, 909–918. 76. Shahinpoor, M.
‘‘Ionic polymer-conductor composites as biomimetic sensors,
Robotic actuators and artificial muscle—A Review.’’
Electrochimica Acta, 2003, 48 (14–16), 2343–2353. 77.
Advances in Polymer Science, Vols. 109 and 110, Responsive
Gels, Transitions I and II; Dusek, K., Ed.;
Springer-Verlag: Berlin, 1993. 78. Li, J.Y.; Nemat-Nasser,
S. Micromechanical analysis of ionic clustering in nafion
perfluorinated membrane. Mech. Mater. 2000, 32 (5), 303–314.
79. Liu, Z.; Calvert, P. Multilayer hydrogens and
musclelike actuators. Adv. Mater. 2000, 12 (4), 288–291.
80. McGehee, M.D.; Miller, E.K.; Moses, D.; Heeger, A.J.
Twenty Years of Conductive Polymers: From Fundamental
Science to Applications. In Advances in Synthetic Metal:
Twenty Years of Progress in Science and Technology; Bamier,
P., Lefrant, S., Bidan, G., Eds.; Elsevier, 1999; 98–203.
published. 81. Osada, Y.; Matsuda, A. Shape memory in
hydrogels. Nature 1995, 376, 219. 82. Osada, Y.; Okuzaki,
H.; Hori, H. A polymer gel with electrically driven
motility. Nature 1992, 355, 242–244. 83. Otero, T.F.;
Sansifiena, J.M. Soft and wet conducting polymers for
artificial muscles. Adv. Mater. 1998, 10 (6), 491–494. 84.
Roentgen, W.C. About the changes in shape and volume of
dielectrics caused by electricity. Ann. Phys. Chem. 1880, I
(1), 771–786. 85. Sacerdote, M.P. Strains in polymers due
to electricity. J. Phys. 1899, VIII (3 Series, t), 31. 86.
Shiga, T. Deformation and viscoelastic behavior of polymer
gels in electric fields. Adv. Polym. Sci. 1997, 134,
131–163. A

87. Shiga, T.; Kurauchi, T. Deformation of polyelectrolyte


gels under the influence of electric field. J. Appl. Polym.
Sci. 1990, 39, 2305–2320.

88. Shiga, T.; Hirose, Y.; Okada, A.; Kurauchi, T. Bending


of ionic polymer gel caused by swelling under sinusoidally
varying electric fields. J. Appl. Polym. Sci. 1993, 47,
113–119.

89. The Applications of Ferroelectric Polymers; Wang, T.T.,


Herbert, J.M., Glass, A.M., Eds.; Chapman and Hall: New
York, 1988. 90. Wang, Q.; Du, X.; Xu, B.; Cross, L.E.
Electromechanical coupling and output efficiency of
piezoelectric bending actuators IEEE Trans. Ultrason.
Ferroelectr. Freq. Control 1999, 46 (3), 638–646. 91.
Proceedings of the Fall MRS Symposium on Electroactive
Polymers (EAP), Warrendale, PA, Zhang, Q.M., Furukawa, T.,
Bar-Cohen, Y., Scheinbeim, J., Eds.; 1999; 1–336.
Artificial Neural Networks: An Overview

38. Graf, H.P.; Jackel, L.D. Analog Electronic Neural


Network Circuits. IEEE Circuits and Devices Magazine 1989,
July, 44–55.

39. Macq, D.; Verlcysen, M.; Jespers, P.; Legat, J. Analog


implementation of a Kohonen map with on-chip learning. IEEE
Trans. Neural Netw. 1993, 4 (3), 456–461.

40. Andreou, A.; et al. VLSI Neural Systems. IEEE Trans.


Neural Netw. 1991, 2 (2), 205–213.

41. Pandya, A.S.; Shandar, R.; Freytag, L. An SIMD


Architecture for the Alopex Neural Network. In Parallel
Architectures for Image Processing; SPIE: Portland, Oregon,
1990; 1246, 275–287.

42. Linares-Barranco, B.; Sanchez-Sinencio, E.;


RodriguezVazquez, A.; Huertas, J.L. A CMOS implementation
of Fitzhugh–Nagumo model IEEE. J. Solid-State Circuits
1991, 26, 956–965.

43. Wolpert, S.; Micheli-Tzanakou, E. An Integrated Circuit


Realization of a Neuronal Model, Proceedings of the IEEE
Northeast Bioengeering Conference; New Haven, CT, 1986;
March 13–14. 44. McCullouch, W.S.; Pitts, W. A logical
calculus of the ideas imminenet in nervous activity. Bull.
Math. Biophys. 1943, 5, 115–133. 45. Wolpert, S.;
Micheli-Tzanakou, E. A neuromime in VLSI. IEEE Trans.
Neural Netw. 1995, 6 (6), 1560–1561. 46. French, A.S.;
Stein, R.B. A flexible neuronal analog using integrated
circuits. IEEE Trans. Biomed. Eng. 1970, 17, 248–253. 47.
Nabet, B.; Pinter, R.B. Sensory Neural Networks: Lateral
Inhibition; CRC Press: Boca Raton, Florida, 1991. 48.
Wolpert, S.; Micheli-Tzanakou, E. Silicon models of lateral
inhibition. IEEE Trans. Neural Netw. 1993, 4 (6), 955–961.
49. Andreou, A.G.; Boahen, K.A.; Pouliquen, P.O.;
Pavasovic, A.; Jenkins, R.E.; Strohbehn, K. Current-mode
subthreshold MOS circuits for analog VLSI neural systems.
IEEE Trans. Neural Syst. 1991, 2 (2). 50. Mead, C.A. Analog
VLSI and Neural Systems; AddisonWesley, Boston, 1989.
Bioactive Glass

1. Yamamuro, T.; Hench, L.; Wilson, J. Handbook of


Bioactive Ceramics; CRC Press: USA, 1990; Vol. I.

2. Hench, L.L.; Wilson, J. An Introduction to Bioceramics;


World Scientific: London, 1993.

3. Hench, L.L. Bioceramics. J. Am. Ceram. Soc. 1998, 81,


1705–1728.

4. Hench, L.L.; Splinter, R.J.; Allen, W.C.; Greenlee,


T.K., Jr. Bonding mechanisms at the interface of ceramic
prosthetic materials. J. Biomed. Mater. Res. 1971, 2 (1),
117–141.

5. Li, R.; Clark, A.E.; Hench, L. An investigation of


bioactive glass powders by the sol–gel process. J. Appl.
Biomater. 1991, 2, 231–239.

6. Clark, A.E.; Pantano, C.G.; Hench, L.L. Auger


spectroscopic analysis of bioglass corrosion films. J. Am.
Ceram. Soc. 1976, 59 (1–2), 37–39.

7. Hench, L.L; Clark, A.E. Adhesion to Bone. In


Biocompatibility of Orthopedic Implants; Williams, D.F.,
Ed.; CRC Press: U.S.A., 1982; Vol. 2.

8. Kokubo, T. Surface chemistry of bioactive


glass–ceramics. J. Non-Cryst. Solids 1990, 120, 138–151.

9. Williams, D.F. The Biocompatibility and Clinical Uses of


Calcium Phosphate Ceramics. In Biocompatibility of Tissue
Analogs; Williams, D.F., Ed.; CRC Press: U.S.A., 1985; Vol.
II, 43–66.

10. Ogino, M.; Ohuchi, F.; Hench, L.L. Compositional


dependence of the formation of calcium phosphate films on
bioglass. J. Biomed. Mater. Res. 1980, 14, 55–64. 11.
Andersson, O.H.; Liu, G.; Karlsson, K.H.; Niemi, L.;
Miettinen, J.; Juhanoja, J. In vivo behavior of glasses in
the SiO 2 –Na 2 O–CaO–P 2 O 5 –Al 2 O 3 –B 2 O 3 . Syst. J.
Mater. Sci. Mater. Med. 1990, 1, 219–227. 12. Brink, M.;
Turunen, T.; Happonen, R.P.; Yli-Urpo, A. Compositional
dependence of bioactivity of glasses in the system Na 2 O–K
2 O–MgO–CaO–B 2 O 3 –P 2 O 5 –SiO 2 . J. Biomed. Mater.
Res. 1997, 37, 114–121. 13. Andersson, O.H.; Rosenqvist,
J.; Karlsson, K.H. Dissolution, leaching, and Al 2 O 3
enrichment at the surface of bioactive glasses studied by
solution analysis. J. Biomed. Mater. Res. 1993, 27,
941–948. 14. Schepers, E.J.; Ducheyne, P. Bioactive glass
particles of narrow size range for the treatment of oral
bone defects: A 1–24month experiment with several materials
and particle sizes and size ranges. J. Oral Rehabil. 1997,
24, 171–181. 15. Kaufmann, E.A.; Ducheyne, P.; Radin, S.;
Bonnell, D.A.; Composto, R. Initial events at the bioactive
glass surface in contact with protein-containing solutions.
J. Biomed. Mater. Res. 2000, 52, 825–830. 16. Pereira,
M.M.; Hench, L.L. Mechanisms of hydroxyapatite formation on
porous gel-silica substrates. J. Sol-Gel Sci. Technol.
1996, 7, 59–68. 17. Kokubo, T.; Kim, H.M.; Kawashita, M.
Novel bioactive materials with different mechanical
properties. Biomaterials 2003, 24, 2161–2175. 18.
Greenspan, D.C.; Zhong, J.P.; Wheller, D.L. Bioactivity and
Biodegradability: Melt vs. Sol–Gel Derived Bioglass s In
Vitro and In Vivo, Proceedings of the 11th International
Symposium on Ceramics in Medicine, New York, Nov. 5–8,
1998; LeGeros, R.Z., LeGeros, J.P., Eds.; World Scientific:
New York, 1998; 345–348. 19. Gross, U.; Kinne, R.; Schmitz,
H.J.; Strunz, V. The response of bone to surface active
glass/glass-ceramics. CRC Crit. Rev. Biocompat. 1988, 4, 2.
20. Yoshii, S.; Kakutani, Y.; Yamamuro, T.; Nakamura, T.;
Kitsugi, T.; Oka, M.; Kokubo, T.; Takagi, M. Strength of
bonding between A–W glass ceramic and the surface of bone
cortex. J. Biomed. Mater. Res. 1988, 22 (A), 327. 21.
Wilson, J.; Pigott, G.H.; Schoen, F.J.; Hench, L.L.
Toxicology and biocompatibility of bioglass. J. Biomed.
Mater. Res. 1981, 15, 805. 22. Wang, M.; Joseph, R.;
Bonfield, W. Hydroxyapatitepolyethylene composites for bone
substitution: Effects of ceramic particle size and
morphology. Biomaterials 1998, 19, 2357–2366. 23. Wang, M.;
Bonfield, W. Chemically coupled hydroxyapatite-polyethylene
composites: Structure and properties. Biomaterials 2001,
22, 1311–1320. 24. Peltola, M.J.; Suonpa¨a¨, J.T.;
Ma¨a¨tta¨nen, H.S.; Varpula, M.J.; Aitasalo, K.M.;
Yli-Urpo, A.; Laippala, P.J. Clinical follow-up method for
frontal sinus obliteration with bioactive glass S53P4. J.
Biomed. Mater. Res. 2001, 58, 54–60. 25. Schepers, E.J.;
Ducheyne, P.; Barbier, L.; Schepers, S. Bioactive glass
particles of narrow size range: A new material for the
repair of bone defects. Implant Dent. 1993, 2, 151–156. 26.
Wilson, J.; Douek, E.; Rust, K. Bioglass Middle Ear
Devices: Ten Year Clinical Results. In Bioceramics; Wilson,
J., Hench, L.L., Greenspan, D., Eds.; Pergamon/Elsevier:
Oxford, UK, 1995; Vol. 8, 239–246. 27. Stanley, H.R.; Hall,
M.B.; Clark, A.E.; King, J.C.; Hench, L.L.; Berte, J.J.
Using 45S5 bioglass cones as endosseous ridge maintenance
implants to prevent alveolar ridge resorptions—A 5 year
evaluation. Int. J. Oral Maxillofac. Implants 1997, 12,
95–105. 28. Jones, J.R.; Hench, L.L. Biomedical materials
for the new millennium: A perspective on the future. J.
Mater. Sci. Technol. 2001, 17, 891–900. 29. Hench, L.L.;
Polak, J.M.; Xynos, I.D.; Buttery, L.D.K. Bioactive
materials to control cell cycle. Mater. Res. Innov. 2000,
3, 313–323. 30. Sepulveda, P.; Jones, J.R.; Hench, L.L.
Bioactive sol– gel foams for tissue repair. J. Biomed.
Mater. Res. 2002, 59, 340–348. 31. Coelho, M.B.; Soares,
I.R.; Mansur, H.S.; Pereira, M.M. Effect of the type of
surfactant on bioactive glasses foam formation. Key Eng.
Mater. 2003, 240– 242, 257–260. 32. Kaufmann, E.A.B.E.;
Ducheyne, P.; Shapiro, I.M. Effect of varying physical
properties of porous, surface modified bioactive glass 45S5
on osteoblast proliferation and maturation. J. Biomed.
Mater. Res. 2000, 52, 783–796. 33. Livingston, T.;
Ducheyne, P.; Garino, J. In vivo evaluation of a bioactive
scaffold for bone tissue engineering. J. Biomed. Mater.
Res. 2002, 62, 1–13. B
Bioactive Materials and Scaffolds for
Tissue Engineering

7. Xynos, I.D.; Edgar, A.J.; Buttery, L.D.K.; Hench, L.L.;


Polak, J.M. Ionic dissolution products of bioactive glass
increase proliferation of human osteoblasts and induce
insulin-like growth factor II mRNA expression and protein
synthesis. Biochem. Biophys. Res. Commun. 2000, 276,
461–465.

8. Chen, Q.Z.; Thompson, I.D.; Boccaccini, A.R. 45S5


Bioglass s -derived glass-ceramic scaffolds for bone tissue
engineering. Biomaterials 2006, 27, 2414–2425.

9. Hench, L.L.; Wilson, J. An Introduction to Bioceramics;


World Scientific: Singapore, 1993.

10. Temenoff, J.S.; Lu, L.; Mikos, A.G. Bone tissue


engineering using synthetic biodegradable polymer
scaffolds. In Bone Engineering; Davies, J.E., Ed.; EM
Squared: Toronto, 2000; 455–462.

11. Devin, J.E.; Attawia, M.A.; Laurencin, C.T.


Threedimensional degradable porous polymer–ceramic matrices
for use in bone repair. J. Biomater. Sci. Polym. Ed. 1996,
7, 661–669.

12. Yang, X.B.B.; Webb, D.; Blaker, J.; Boccaccini, AR.;


Maquet, V.; Cooper, C.; Oreffo, R.O. C. Evaluation of human
bone marrow stromal cell growth on biodegradable
polymer/Bioglass s composites. Biochem. Biophys. Res.
Commun. 2006, 342 (4), 1098–1107.

13. Callcut, S.; Knowles, J.C. Correlation between


structure and compressive strength in a reticulate
glassreinforced hydroxyapatite foam. J. Mater. Sci. Mater.
Med. 2002, 13, 485–489.

14. Vitale-Brovarone, C.; Miola, M.; Balagna, C.; Verne, E.


3D Glass-ceramic scaffolds with antibacterial properties
for bone grafting. Chemical Eng. Journal 2008, 137,
129–136.

15. Vitale-Brovarone, C.; Verne´, E.; Robiglio, L.;


Appendino, P.; Bassi, F.; Martinasso, G.; Muzio, G.;
Canuto, R. Development of glass–ceramic scaffolds for bone
tissue engineering: Characterisation, proliferation of
human osteoblasts and nodule formation. Acta Biomaterialia
2007, 3, 199–208. 16. Wu, C.; Ramaswamy, Y.; Boughton, P.;
Zreiqat, H. Improvement of mechanical and biological
properties of porous CaSiO 3 scaffolds by poly(D,L-lactic
acid) modification. Acta Biomaterialia 2008, 4, 343–353. 17.
Miao, X.; Tan, L.P.; Tan, L.S.; Huang, X. Porous calcium
phosphate ceramics modified with PLGABioactive glass. Mater
Sci and Eng C 2007, 27, 274–280. 18. Jones, J.R.; Ahir, S.;
Hench, L.L. Large-scale production of 3D bioactive glass
macroporous scaffolds for tissue engineering. J. Sol-Gel
Sci. Tech. 2004, 29, 179–188. 19. Leong, K.F.; Cheah, C.M.;
Chua, C.K. Solid freeform fabrication of three-dimensional
scaffolds for engineering replacement tissues and organs.
Biomaterials 2003, 24, 2363–2378. 20. Chen, Q.Z.;
Boccaccini, A.R. Poly(D,L-lactic acid) coated 45S5 Bioglass
s -based scaffolds: processing and characterisation. J.
Biomed. Mater. Res. A 2006, 77A, 445–457. 21. Hutmacher,
D.W. Scaffolds in tissue engineering bone and cartilage.
Biomaterials 2000, 21, 2529–2543. 22. Guarino, V.; Causa,
F.; Ambrosio, L. Bioactive scaffolds for bone and ligament
tissue. Expert Rev. Medical Devices 2007, 4, 405–418. 23.
Khan, Y.M.; Katti, D.S.; Laurencin, C.T. Novel
polymer-synthesised ceramic composite-based system for bone
repair: an in vitro evaluation. J. Biomed. Mater. Res.
2004, 69A, 728–737. 24. Roether, J.A.; Boccaccini, A.R.;
Hench, L.L.; Maquet, V.; Gautier, S.; Jerome, R.
Development and in vitro characterisation of novel
bioresorbable and bioactive composite materials based on
polylactide foams and Bioglass s for tissue engineering
applications. Biomaterials 2002, 23, 3871–3878. 25.
Oliveira, A.L.; Mano, J.F.; Reis, R.L. Nature-inspired
calcium phosphate coatings: present status and novel
advances in the science of mimicry. Current Opi. Solid
State Mater. Sci. 2003, 7, 309–318. B
Bioadhesion

1. Smart, J.D.; Kellaway, I.W.; Worthington, H.E.C. An


in-vitro investigation of mucosa-adhesive materials for use
in controlled drug delivery. J. Pharm. Pharmacol. 1984, 36,
295–299.

2. Chen, J.L.; Cyr, G.N. Compositions Producing Adhesion


Through Hydration. In Adhesion in Biological Systems;
Manly, R.S., Ed.; Academic Press: New York, 1970; 163–181.

3. Harding, S.E.; Davis, S.S.; Deacon, M.P.; Fiebrig, I.


Biopolymer mucoadhesives. Biotechnol. Genet. Eng. Rev.
1999, 16, 41–85.

4. Lee, J.W.; Park, J.H.; Robinson, J.R. Bioadhesive-based


dosage forms: The next generation. J. Pharm. Sci. 2000, 89,
850–866. 5. Ahuja, A.; Khar, R.P.; Ali, J. Mucoadhesives
drug delivery systems. Drug Dev. Ind. Pharm. 1997, 23 (5),
489–515. 6. Smart, J.D. The Role of Water Movement and
Polymer Hydration in Mucoadhesion. In Bioadhesive Drug
Delivery Systems: Fundamentals, Novel Approaches and
Development; Mathiowitz, E., Chickering, D.E., Lehr, C.-M.,
Eds.; Marcel Decker: New York, 1999; 11–23. 7. Mortazavi,
S.A.; Smart, J.D. An in-vitro method for assessing the
duration of mucoadhesion. J. Control. Release 1994, 31,
207–212. 8. Patel, D.; Smith, A.W.; Grist, N.W.; Barnett,
P.; Smart, J.D. In-vitro mucosal model predictive of
bioadhesive agents in the oral cavity. J. Control. Release
1999, 61, 175–183. 9. Lueßen, H.L.; Rental, C.-O.; Kotze,
A.F.; Lehr, C.-M.; de Boer, A.G.; Verhoef, J.C.; Junginger,
H.E. Mucoadhesive polymers in peroral peptide delivery. IV.
Polycarbophil and chitosan are potent enhancers of peptide
transport across intestinal mucosae in vitro. J. Control.
Release 1997, 45, 15–23. 10. Bernkop-Schnurch, A.;
Scholler, S.; Biebel, R.G. Development of controlled
release systems based on thiolated polymers. J. Control.
Release 2000, 66, 39–48. 11. Shojaei, A.H.; Li, X. Novel
PEG Containing Acrylate Copolymers with Improved
Mucoadhesive Properties. In Bioadhesive Drug Delivery
Systems: Fundamentals, Novel Approaches and Development;
Mathiowitz, E., Chickering, D.E., Lehr, C.-M., Eds.; Marcel
Decker: New York, 1999; 433–458. 12. Bernkop-Schnurch, A.;
Krajicek, M.E. Mucoadhesive polymers as platforms for
peroral peptide delivery and absorption: Synthesis and
evaluation of different chitosan-EDTA conjugates. J.
Control. Release 1998, 50, 215–223. 13. Lehr, C.-M.
Lectin-mediated drug delivery: The second generation of
bioadhesives. J. Control. Release 2000, 65, 19–29. 14.
Woodley, J. Bioadhesion. New Possibilities for drug
administration. Clin. Pharmacokinet. 2001, 40 (2), 77–84.
15. Easson, J.H.; Haltner, E.; Lehr, C.-M. Bacterial
Invasion Factors and Lectins as Second Generation
Bioadesives. In Bioadhesive Drug Delivery Systems:
Fundamentals, Novel Approaches and Development; Mathiowitz,
E., Chickering, D.E., Lehr, C.-M., Eds.; Marcel Decker: New
York, 1999; 409–432. 16. Ponchel, G.; Irache, J.-M. Specific
and non-specific bioadhesive particulate systems for oral
delivery to the gastrointestinal tract. Adv. Drug Deliv.
Rev. 1998, 34, 191–219. 17. Rihova, B.; Rathi, R.C.;
Kopeckova, P.; Kopecek, J. In-vitro bioadhesion of
carbohydrate containing N(2-hydroxypropyl)methacrylamide
copolymers to the GI tract of guinea pigs. Int. J. Pharm.
1992, 87, 105–116. 18. Nagai, T.; Konishi, R.
Buccal/gingival drug delivery systems. J. Control. Release
1987, 6, 353–360. 19. Veillard, M.M.; Longer, M.A.;
Martens, T.W.; Robinson, J.R. Preliminary studies of oral
mucosal delivery of peptide drugs. J. Control. Release
1987, 6, 123–131. 20. Smart, J.D. Drug delivery using
buccal adhesive systems. Adv. Drug Deliv. Rev. 1993, 11,
253–270. 21. Kockisch, S.; Rees, G.D.; Young, S.A.;
Tsibouklis, J.; Smart, J.D. A direct-staining method to
evaluate the mucoadhesion of polymers from aqueous
dispersion. J. Control. Release 2001, 77, 1–6. 22. Wilson,
C.G. In Vivo Testing of Bioadhesion. In
Bioadhesion—Possibilites and Future Trends; Gurny, R.,
Junginger, H.E., Eds.; Wissenshaftliche
Verlagsgesellschaft: Stuttgart, 1990; 93–108. 23. Riley,
R.G.; Smart, J.D.; Tsibouklis, J.T.; Young, S.A.; Hampson,
F.; Davis, J.A.; Kelly, G.; Dettmar, P.W.; Wilber, W.R. An
in-vitro model for investigating the gastric mucosal
retention of 14 C-labelled poly (acrylic acid) dispersions.
Int. J. Pharm. 2002, 236 (1–2), 87–96. 24. Illum, L.;
Farraj, N.F.; Davis, S.S.; Johansen, B.R.; O’Hagan, D.T.
Investigation of the nasal absorption of biosynthetic human
growth hormone in sheep—Use of a bioadhesive microsphere
delivery system. Int. J. Pharm. 1990, 63, 207–211. 25.
Horstmann, M.; Muller, W.; Asmussen, B. Principles of Skin
Adhesion and Methods for Measuring Adhesion of Transdermal
Systems. In Bioadhesive Drug Delivery Systems:
Fundamentals, Novel Approaches and Development; Mathiowitz,
E., Chickering, D.E., Lehr, C.-M., Eds.; Marcel Decker: New
York, 1999; 175–196. 26. Lipman, R. Hydrocolliod PSAs: New
formulation strategies. Med. Device Diagn. Ind. Mag. 1999,
132–150. 27. Sharma, A.; Kaur, R.; Kumar, S.; Gupta, P.;
Pandov, S.; Patnaik, B.; Gupta, A. Fibrin glue versus
N-butyl–2cyanoacrylate in corneal perforations.
Ophthalmology 2003, 110, 291–298. 28. Ewoldsen, N.; Demke,
R.S. A review of orthodontic cements and adhesives. Am. J.
Orthod. Dentofac. Orthoped. 2001, 120, 45–48. 29. DiMaio,
F.R. The science of bone cement, a historical review.
Orthopedics 2002, 25, 1399–1407. 30. Moore, W.R.; Graves,
S.E.; Bain, G.I. Synthetic bone graft substitutes. Aust.
N.Z. J. Surg. 2001, 71, 354–361. B
Biocatalysis

20. Emptage, M.; Haynie, S.; Laffend, L.; Pucci, J.;


Whited, G. Genetically Engineered Escherichia coli
Containing Nonspecific Dehydratase yghD and DHA Regulon for
the Biological Production of 1,3-Propanediol with High
Titer; 2001. (E.I. du Pont de Nemours and Company: USA;
Genencor International, Inc.); PCT Int. Appl. 109 pp.

21. Zhu, M.M.; Lawman, P.D.; Cameron, D.C. Improving


1,3-propanediol production from glycerol in a metabolically
engineered Escherichia coli by reducing accumulation of
sn-glycerol-3-phosphate. Biotechnol. Prog. 2002, 18 (4),
694–699.

22. Patten, P.; Howard, R.; Stemmer, R. Applications of DNA


shuffling to pharmaceuticals and vaccines. Curr. Opin.
Biotechnol. 1991, 8, 724–733.

23. Huisman, G.W.; Gray, D. Towards novel processes for the


fine-chemical and pharmaceutical industries. Curr. Opin.
Biotechnol. 2002, 13, 1–7.

24. Joo, H.; Lin, Z.; Arnold, F.H. Laboratory evolution of


peroxide-mediated P450 hydroxylation. Nature 1999, 399,
670–673.

25. Boller, T.; Meier, C.; Menzler, S. EUPERGIT Oxirane


acrylic beads: How to make enzymes fit for biocatalysis.
Org. Process Res. Dev. 2002, 6, 509–519.

26. Tischer, W.; Wedekind, F. Immobilized enzymes: Methods


and applications. Top. Curr. Chem. 1999, 200, 95–126.

27. Gill, I.; Ballesteros, A. Bioencapsulation within


synthetic polymers (Part 2): Non-sol–gel protein–polymer
biocomposites. Trends Biotechnol. 2000, 18, 469–479.

28. LeJeune, K.E.; Russell, A.J. Covalent binding of a


nerve agent hydrolyzing enzyme within polyurethane foams.
Biotechnol. Bioeng. 1996, 51, 450–457.

29. Park; et al. Characterization of antithrombotic


activity of lumbrokinase-immobilized polyurethane valves in
the total artificial heart. Artif. Organs 1999, 23, 210–214.

30. Phaneuf, M.D.; et al. Covalent linkage of recombinant


hirudin to a novel ionic poly(carbonate) urethane polymer
with protein binding sites: Determination of surface
antithrombin activity. Artif. Organs 1998, 22, 657–665. 31.
Enzymic Reactions in Organic Medial; Koskinen, A.M.P.,
Klibanov, A.M., Eds.; Blackie: Glasgow, UK, 1996. 32. Ru,
M.T.; Wu, K.C.; Lindsay, J.P.; Dordick, J.S.; Reimer, J.A.;
Clark, D.S. Towards more active biocatalysts in organic
media: Increasing the activity of salt-activated enzymes.
Biotechnol. Bioeng. 2001, 75, 187–196. 33. DeSantis, G.;
Jones, J.B. Chemical modification of enzymes for enhanced
functionality. Curr. Opin. Biotechnol. 1999, 10, 324–330.
34. Adams, M.W.; Perler, F.B.; Kelly, R.M. Extremozymes:
Expanding the limits of biocatalysis. Bio/ Technology 1995,
13, 662–668. 35. Runnion, K.; Combie, J.; Williamson, M.
Thermally Stable Urease from Thermophilic Bacteria. In
Biocatalysis at Extreme Temperatures; Adams, M.W.W., Kelly,
R.M., Eds.; ACS Symposium Series, American Chemical
Society: Washington, DC, 1992; Vol. 498. 36. Olsen, M.J.;
Stephens, D.L.; Georgiou, G.; Iverson, B.L. A Method for
Biocatalyst Evolution Using Bacterial Surface Display. In
Book of Abstracts, 214th ACS National Meeting, Las Vegas,
NV, September 7–11, 1997; American Chemical Society:
Washington, DC. 37. Zhao, H.; Arnold, F.H. Directed
evolution converts subtilisin E into a functional
equivalent of thermitase. Prot. Eng. 1999, 12, 47–53. 38.
Song, J.K.; Rhee, J.S. Enhancement of stability and
activity of phospholipase A1 in organic solvents by
directed evolution. Biochim. Biophys. Acta 2001, 1547,
370–378. 39. Matsumura, I.; Ellington, A. In vitro
evolution of betaglucuronidase into a beta-galactosidase
proceeds through non-specific intermediates. J. Mol. Biol.
2001, 305, 331–339.
Biocompatibility Testing

25. Kimber, I.; Basketter, D.A.; Berthold, K.; Butler, M.;


Garrigue, J.L.; Lea, L.; Newsome, C.; Roggeband, R.;
Steiling, W.; Stropp, G.; Waterman, S.; Wieman, C. Skin
sensitization testing in potency and risk assessment.
Toxicol. Sci. 2001, 59, 198–208.

26. Thomas, J.A. In-Use Testing of Biomaterials in


Biomedical Devices. In Handbook of Biomaterials Evaluation:
Scientific, Technical and Clinical Testing of Implant
Materials, 2nd Ed.; von Recum, A.F., Ed.; Macmillan
Publishing Company: New York, 1999; 313–320.

27. Human Subject Research: A Handbook for Institutional


Review Boards; Greenwald, A., Ryan, M., Milvihill, J.,
Eds.; Plenum Press: New York, 1982.

28. Federal policy for the protection of human subjects,


Department of Health and Human Services regulations, Title
45 Code of Federal Regulations, part 46 (45 CFR46).

29. World Medical Association Recommendations guiding


doctors in clinical research. Br. Med. J. 1964, 2,
1119–1129.

30. Piehler, H.R. Risk–benefits of orthopaedic prosthetic


devices. Orthop. Rev. 1978, 7, 75–88.

SUGGESTED READING/ADDITIONAL

RESOURCES

American National Standard/American Dental Association


(ANSI/ADA) Specification No. 41—Biological Evaluation of
Dental Materials; American Dental Association: Chicago, IL,
2001.

Annual Book of ASTM Standards Vol. 13.01 Medical Devices;


Emergency Medical Services, ASTM International: West
Conshohocken, PA, 2002.

Black, J. Biological Performance of Materials—Fundamentals


of Biocompatibility; Marcel Dekker, Inc.: New York, 1999.

Dee, K.C.; Puleo, D.A.; Bizios, R. An Introduction to


Tissue— Biomaterial Interactions; John Wiley & Sons, Inc.:
Hoboken, NJ, 2002; 173–184.

Fundamental Aspects of Biocompatibiliy; Williams, D.F.,


Ed.; CRC Press: Boca Raton, FL, 1981; Vols. I and II.
Guidelines for Blood–Material Interactions, Publication
85–2185; National Institutes of Health: Bethesda, MD, 1985.
Handbook of Biomaterials Evaluation—Scienctific, Technical,
and Clinical Testing of Implant Materials, 2nd Ed.; von
Recum, A.F., Ed.; Macmillan Publishing Co.: New York, 1999.
Hanson, S.; Lalor, P.A.; Niemi, S.M.; Northrup, S.J.;
Ratner, B.D.; Spector, M.; Vale, B.H.; Willson, J.E.
Testing Biomaterials. In Biomaterials Science—An
Introduction to Materials in Medicine; Ratner, B.D.,
Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.; Academic
Press: San Diego, CA, 1996; 215–242. International Standard
ISO 7405—Dentistry—Preclinical Evaluation of
Biocompatibility of Medical Devices Used in Dentistry—Test
Methods for Dental Materials; International Standards
Organization: Geneva, Switzerland, 1997. Kirkpatrick, C.J.;
Bittinger, F.; Wagner, M.; Kohler, H.; van Kooten, T.G.;
Klein, C.L.; Otto, M. Current trends in biocompatibility
testing. Proc. Inst. Mech. Eng., H 1998, 212, 75–84. Mohan,
K.; Sargent, H.E. Clinical trials, an introduction. Med.
Dev. Diag. Ind. 1996, 18, 114–119. Mollnes, T.E. Complement
and biocompatibility. Vox Sang. 1998, 74 (Suppl. 2),
303–307. Technical Committee ISO/TC 94. International
Standard ISO 10993—Biological Evaluation of Medical
Devices, Part 1–17; International Standards Organization:
Geneva, Switzerland, 1997. Techniques of Biocompatibility
Testing; Williams, D.F., Ed.; CRC Press: Boca Raton, FL,
1986; Vols. I and II. Thull, R. Physiochemical principles
of tissue material interactions. Biomol. Eng. 2002, 19,
43–50. USP XXIII: The Pharmacopeia of the United States of
America, 23rd Revision Incorporating The National
Formulary, 18th Revision; The Unites States Pharmacopeial
Convention, Inc.: Washington, DC, 1995. Walum, E.;
Stenberg, K.; Jenssen, D. Understanding Cell
Toxicology—Principles and Practice; Ellis Horwood Ltd.: New
York, 1990. Wataha, J.C. Biocompatibility of Dental
Materials. In Restorative Dental Materials, 11th Ed.;
Craig, R.G., Powers, J.M., Eds.; Mosby: St. Louis, 2002;
125–163.
Biocomposites

19. Mikos, A.G. Biomaterials for Drug and Cell Delivery;


Materials Research Society: Pittsburgh, PA, 1994.

20. Liu, L.; Zhang, L.; Ren, B.; Wang, F.; Zhang, Q.
Preparation and Characterization of collagen–hydroxyapatite
composite used for bone tissue engineering scaffolds.
Artif. Cells Blood Substitutes Biotechnol. 2003, 31 (4),
435–448.

21. Du, C.; Cui, F.Z.; Zhu, X.D.; de Groot, K.


Threedimensional Nano-HAP/collagen matrix loading with
osteogenic cells in organ culture. J. Biomed. Mater. Res.
1999, 44 (4), 407–415.

22. Rho, J.Y.; Kuhn-Spearing, L.; Zioupos, P. Mechanical


properties and the hierarchical structure of bone. Med.
Eng. Phys. 1998, 20, 92–102.

23. Webster, T.J.; Ergun, C.; Doremus, R.H.; Siegel, R.W.;


Bizios, R. Specific proteins mediate enhanced osteoblast
adhesion on nanophase ceramics. J. Biomed. Mater. Res.
2000, 51 (3), 475–483.

24. Webster, T.J.; Siegel, R.W.; Bizios, R. Osteoblast


adhesion on nanophase ceramics. Biomaterials 1999, 20 (13),
1221–1227.

25. Dulgar Tulloch, A.J.; Bizios, R.; Siegel, R.W.


Nanophase alumina/poly( L-lactic acid) composite scaffolds
for biomedical applications. Mat. Res. Soc. Symp. Proc.
2003, 740, 161–166.

26. Webster, T.J.; Siegel, R.W.; Bizios, R. Nanoceramic


surface roughness enhances osteoblast and osteoclast
functions for improved orthopaedic/dental implant efficacy.
Scr. Materialia 2001, 44, 1639–1642.

27. Liu, H.; Slamovich, E.B.; Webster, T.J. Increased


osteoblast functions on poly-lactic-co-glycolic-acid with
highly dispersed nanophase titania. J. Biomed. Nanotechnol.
2005, 1, 83–89.

28. Tsang, V.L; Bhatia, S.N. Three-dimensional tissue


fabrication. Adv. Drug Deliv. Rev. 2004, 56, 1635–1647.

29. Borenstein, J.T; Terai, H.; King, K.R.; Weinberg, E.J.;


Kaazempur-Mofrad, M.R.; Vacanti, J.P. Microfabrication
technology for vascularized tissue engineering. Biomed.
Microdevices 2002, 4, 167–175.

30. Vozzi, G.; Flaim, C.; Ahluwalia, A.; Bhatia, S.N.


Fabrication of PLGA scaffolds using soft lithography and
microsyringe decomposition. Biomaterials 2003, 24,
2533–2540.

31. Leong, K.F.; Cheah, C.M.; Chua, C.K. Solid freeform


fabrication of three-dimensional scaffolds for engineering
replacement tissues and organs. Biomaterials 2003, 24,
2363–2378.

32. Yang, S.; Leong, K.F.; Du, Z.; Chua, C.K. The design of
scaffolds for use in tissue engineering: part II. Rapid
prototyping techniques. Tissue Eng. 2002, 8, 1–11.

33. Lee, G.; Barlow, J.; Fox, W.; Aufdermorte, T.


Biocompatibility of SLS-formed calcium phosphate implants.
Proceedings of Solid Freeform Fabrication Symposium,
University of Texas: Austin, TX, 1996; 15–22.

34. Hutmacher, D.W. Scaffold design and fabrication


technologies for engineering tissues—state of the art and
future perspectives. J. Biomater. Sci. Polym. Ed. 2001, 12,
107–124. 35. Zein, I.; Hutmacher, D.W.; Tan, K.C.; Teoh,
S.H. Fused deposition modeling of novel scaffold
architectures for tissue engineering applications.
Biomaterials 2002, 23, 1169–1185. 36. Cooke, M.N; Fisher,
J.P.; Dean, D.; Rimnac, C.; Mikos, A.G. Use of
stereolithography to manufacture critical-sized 3D
Bio-degradable scaffolds for bone ingrowth. J. Biomed.
Mater. Res. 2003, 64B, 65–69. 37. Park, A.; Wu, B.;
Griffith, L.G. Integration of surface modification and 3D
fabrication techniques to prepare patterned poly(
L-lactide) substrates allowing regionally selective cell
adhesion. J. Biomater. Sci. Polym. Ed. 1998, 9, 89–110. 38.
Kim, S.S.; Utsunomiya, H.; Koski, J.A.; Wu, B.M.; Cima,
M.J.; Sohn, J.; Mukai, K.; Griffith, L.G.; Vacanti, J.P.
Survival and function of hepatocytes on a novel
three-dimensional synthetic biodegradable polymer scaffold
with an intrinsic network of channels. Ann. Surg. 1998,
228, 8–13. 39. Zeltinger, J.; Sherwood, J.K.; Graham, D.A.;
Mueller, R.; Griffith, L.G. Effect of pore size and void
fraction on cellular adhesion, proliferation, and matrix
deposition. Tissue Eng. 2001, 7, 557–572. 40. Sachlos, E.;
Reis, N.; Ainsley, C.; Derby, B.; Czernuszka, J.T. Novel
collagen scaffolds with predefined internal morphology made
by solid freeform fabrication. Biomaterials 2003, 24,
1487–1497. 41. . EnvisionTec. www.envisionstec.de. 42.
Mironov, V.; Boland, T.; Trusk, T.; Forgacs, G.; Markwald,
R.R. Organ printing: computer-aided jetbased 3D tissue
engineering. Trends Biotechnol. 2003, 21, 157–161. 43.
Sciperio, I. www.sciperio.com. 44. Odde, D.J.; Renn, M.J.
Laser-guided direct writing of living cells. Biotechnol.
Bioeng. 2000, 67, 312–318. 45. Albrecht, D.R.; Sah, R.L.;
Bhatia, S.N. Dielectrophoretic cell patterning within
tissue engineering scaffolds. In Second Joint EMBS-BMES
Conference. 24th Annual International Conference of the
Engineering in Medicine and Biology Society. Annual Fall
Meeting of the Biomedical Engineering Society, IEEE,
Houston, TX, 2002, Cat. No. 02CH37392, Vol. 2, 1708–1709.
46. Landers, R.; Hubner, U.; Schmelzeisen, R.; Mulhaupt, R.
Rapid prototyping of scaffolds derived from
thermoreversible hydrogels and tailored for applications in
tissue engineering. Biomaterials 2002, 23, 4437–4447. 47.
Webster, T.J.; Smith, T.A. Increased osteoblast function on
PLGA composites containing nanophase titania. J. Biomed.
Mat. Res. 2005, 74, 677–686. 48. Palin, E.; Liu, H.;
Webster, T.J. Mimicking the nanofeatures of bone increases
bone-forming cell adhesion and proliferation.
Nanotechnology 2005, 16, 1828–1835. 49. Swaim, J.; Smith,
L.; Webster, T.J. Increased osteoblast functions on PLGA
containing HA treated with NaOH. Biomaterials, . in press.
50. Khang, D.; Webster, T.J. Aligned calcium phosphate
crystal in carbon nanofibers aligned in polymers.
International Journal of Nanomedine 2006, 1 (1), 65–72.
Biodegradable Polymers: An Overview

23. Lee, S.H.; Szinai, I.; Carpenter, K.; Katsarava, R.;


Jokhadze, G.; Chu, C.C.; Scheerder, I.D.; Hong, M.K. In
vivo biocompatibility evaluation of stents coated by a new
biodegradable elastomeric and functional polymer. Coron.
Artery Dis. 2002, 13 (4), 237–241.

24. DeFife, K.; Gopalan, S.; Price, S.; Cruz-Aranda, G.;


Chantung, R.; Turnell, W. Novel Poly(ester amide)
Copolymers Are Blood and Tissue Compatible. 30th Annual
Meeting of the Society for Biomaterials, Memphis, TN, April
27–30, 2005; 159.

25. Lee, K.H.; Chu, C.C. The effect of superoxide ions in


the degradation of five synthetic absorbable suture
materials. J. Biomed. Mater. Res. 2000, 49 (1), 25–35. 26.
Lee, K.H.; Won, C.Y.; Chu, C.C.; Gitov, I. The role of
superoxide in the biodegradation of synthetic biodegradable
polymers. J. Polym. Sci. A Polym. Chem. Ed. 1999, 37,
3558–3567. 27. Lee, K.H.; Chu, C.C. The Role of Free
Radicals in Hydrolytic Degradation of Absorbable Polymeric
Biomaterials. 5th World Biomaterials Congress, Toronto,
Canada, May 29–June 2, 1996. 28. Lee, K.H.; Chu, C.C.
Molecular design of biologically active biodegradable
polymers for biomedical applications. Macromol. Symp. 1998,
130, 71–80.
Biofilms

2. Lazazzera, B.A. Lessons fromDNAmicroarray analysis: the


gene expression profiles of biofilms. Curr. Opin. Microbiol.
2005, 8, 222–227.

3. Davey, M.E.; O’Toole, G.A. Microbial biofilms: from


ecology to molecular genetics. Microbiol. Mol. Biol. Rev.
2000, 64, 847–867.

4. Stanley, N.R.; Lazazzara, B.A. Environmental signals and


regulatory pathways that influence biofilm formation. Mol.
Microbiol. 2004, 52, 917–924.

5. Costerton, J.W.; Stewart, P.S.; Greenberg, E.P.


Bacterial biofilms: a common cause of persistent infections.
Science 1999, 284, 1318–1322.

6. Hogan, D.; Kolter, R. Why are bacteria refractory to


antimicrobials? Curr. Opin. Microbiol. 2003, 5, 472–477.

7. Harrison, D.; Ceri, H.; Stremick, C.A.; Turner, R.J.


Biofilm susceptibility to metal toxitcity. Environ.
Microbiol. 2004, 6, 1220–1227.

8. Harrison, J.J.; Ceri, H.; Stremick, C.; Turner, R.J.


Differences in biofilm and planktonic cell mediated
reduction of metalloid oxyanions. FEMS Microbiol. Lett.
2004, 235, 357–362.

9. Anderson, G.G.; Palermo, J.J.; Schilling, J.D.; Roth,


R.; Heuser, J.; Hultgren, S.J. Intracellular bacterial-like
pods in urinary tract infections. Science 2003, 301,
105–107.

10. Sauer, K.; Camper, A.K.; Ehrlich, G.D.; Costerton,


J.W.; Davies, D.G. Pseudomonas aeruginosa displays multiple
phenotypes during development as a biofilm. J. Bacteriol.
2002, 184, 1140–1154.

11. Kaplan, J.B.; Meyenhofer, M.M.; Fine, D.H. Biofilm


growth and detachment of Actinobacillus
actinomycetemcomitans. J. Bacteriol. 2003, 185, 1399–1404.

12. Auschill, T.M.; Artweiler, N.B.; Netuschil, L.; Brecx,


M.; Reich, E.; Sculean, A. Spatial distribution of vital
and dead microorganisms in dental biofilms. Arch. Oral
Microbiol. 2001, 46, 471–476.

13. Jefferson, K.K. What drives bacteria to produce a


biofilm? FEMS Microbiol. Lett. 2004, 236, 163–173.

14. Hammer, B.K.; Bassler, B.L. Quorum sensing controls


biofilm formation in Vibrio cholerae. Mol. Microbiol. 2003,
50, 101–114.

15. Dasgupta, M.K. Biofilms and infection in dialysis


patients. Semin. Dial. 2002, 15, 338–346.

16. Trautner, B.W.; Darouiche, R.O. Catheter associated


infections: pathogenesis affects prevention. Arch. Intern.
Med. 2004, 164, 842–850.

17. Hall-Stoodley, L.; Stoodley, P. Biofilm formation and


dispersal and the transmission of human pathogens. Trends
Microbiol. 2005, 13, 7–10.

18. Davey, M.E.; O’Toole, G.A. 2000 microbial biofilms: from


ecology to molecular genetics. Microbial Mol. Biol. Rev.
2000, 64, 847–867.

19. O’Toole, G.A.; Kolter, R. Flagellar and twitching


motility are necessary for Pseudomonas aeruginosa, biofilm
development. Mol. Microbiol. 1998, 30, 295–304.

20. O’Toole, G.A.; Gibbs, K.A.; Hager, P.W.; Phibbs, P.V.;


Kolter, R. The global carbon metabolism regulator Crc is a
component of a signal transduction pathway required for
biofilm development by Pseudomonas aeruginosa. J. Bacteriol.
2000, 182, 425–431.

21. Danese, P.N.; Pratt, L.A.; Dove, S.; Kolter, R. The


outer membrane protein ag43 mediates cell-cell interactions
in Escherichia coli biofilms. Mol. Microbiol. 2000, 37,
424–432. 22. Genevaux, P.; Muller, S.; Bauda, P. A rapid
screening procedure to identify mini Tn10 insertion mutants
of Escherichia coli K-12 with altered adhesion properties
FEMS Microbiol. Lett. 1996, 142, 27–30. 23. Chia, J.-S.;
Lee, Y-Y.; Huang, P.-T.; Chen, J.Y. Identification of stress
responsive genes in Streptococcus mutans by differential
display reverse transcriptionalPCR. Infect. Immun. 2001,
69, 2493–2501. 24. Svensater, G.; Welin, J.C.; Wilkins, D.;
Beighton, D.; Hamilton, I.R. Protein expression by
planktonic and biofilm cells of Streptococcus mutans. FEMS
Microbiol. Lett. 2001, 205, 139–146. 25. Zhu, M.; Takenaka,
S.; Sato, M.; Hoshino, E. Influence of starvation and biofilm
formation on acid resistance of Streptococcus mutans. Oral
Microbiol. Immunol. 2001, 16, 24–27. 26. Parsek, M.;
Greenberg, E.P. Sociobiology: the connections between
quorum sensing and biofilms. Trends Microbiol. 2005, 13,
27–33. 27. Hentzer, M.; Wu, H.; Andersen, J.B.; riedel, K.;
Rasmussen, T.B.; Bagge, N.; Kumar, N.; Schembri, M.A.;
Song, Z.; Kristofferson, P.; Manefield, M.; Costerton, J.W.;
Molin, S.; Eberl, L.; Steinberg, P.; Kjellenberg, S.;
Hoiby, N.; Givskov, M. Attenuation of Pseudomonas
aeruginosa virulence by quorum sensing inhibitors. EMBO J.
2003, 22, 3803–3815. 28. Eberl, L.; Tummler, B. Pseudomonas
aeruginosa and Burkholderia cepacia in cystic fibrosis:
genome evaluation, interactions and adaptation. Int. J.
Med. Microbiol. 2004, 294, 123–131. 29. Juhas, M.; Eberl,
L.; Tummler, B. Quorum sensing: the power of cooperation in
the world of Pseudomonas. Environ. Microbiol. 2005, 7,
459–471. 30. Withers, H.; Swift, S.; Williams, P. Quorum
sensing as an integral component of gene regulatory
networks in gramive bacteria. Curr. Opin. Microbiol. 2001,
4, 186–193. 31. Kim, E.-J.; Wang, W.; Deckwer, W.-D.; Zeng,
A.-P. Expression of the quorum sensing regulatory protein
LasR is strongly affected by iron and oxygen concentrations
in cultures of Pseudomonas aeruginosa irrespective of cell
density. Microbiol. 2005, 151, 1127–1138. 32. Peterson,
F.C.; Pecharki, D.; Scheie, A.A. Biofilm mode of growth of
Streptococcus intermedius favored by competence-stimulating
signalling peptide. J. Bacteriol. 2004, 186, 6327–6331. 33.
Aspiras, M.B.; Ellen, R.P.; Cvitkovitch, D.G. ComX activity
of Streptococcus mutans growing in biofiulms. FEMS
Microbiol. Lett. 2004, 238, 167–174. 34. Suntharalingam,
P.; Cvitkovitch, D.G. Quorum sensing in streptococcal
biofilms. Trends Microbiol. 2005, 1, 3–6. 35. Branda, S.S.;
Vik, A.; Friedman, L.; Kolter, R. Biofilms: the matrix
revisited. Trends Microbiol. 2005, 13, 20–26. 36. Beloin,
C.; Ghigo, J.-M. Finding gene expression patterns in
bacterial biofilms. Trends Microbiol. 2005, 13, 16–19. 37.
Thormann, K.M.; Saville, R.M.; Shukla, S.; Pelletier, D.A.;
Spormann, A.M. Initial phases of biofilm formation in
Shewanella oneidensis MR-1. J. Bacteriol. 2004, 186,
8096–8104. 38. Whiteley, M.; Bangera, M.G.; Bumgarner,
R.E.; Parsek, M.R.; Teitzel, G.M.; Lory, S.; Greenberg,
E.P. Gene expression in Pseudomonas aeruginosa biofilms.
Nature 2001, 413, 860–864. 39. Hancock, L.E.; Perego, M.
The Enterococcus faecalis fsr two-component system controls
biofilm development through production of gelatinase. J.
Bacteriol. 2004, 186, 5629–5639. 40. Carniol, K.; Gilmore,
M. Signal transduction, quorum sensing, extracellular
protease activity in Enterococcus faecalis biofilm
formation. J. Bacteriol. 2004, 186, 8161–8163. 41. Jones,
C.H.; Tove, C.; Bolken, K.F.; Jones, G.O.Z.; Dennis, E.H.
Conserved DegP protease in gram-positive bacteria is
essential for thermal and oxidative tolerance and full
virulence in Streptococcus pyogenes. Infect. Immun. 2001,
69, 5538–5545. 42. Diaz-Torres, M.L.; Russell, R.R. HtrA
protease and processing of extracellular proteins of
Streptococcus mutans. FEMS Microbiol. Lett. 2001, 204,
23–28. 43. Lemos, A.C.; Yi-Ywan, M.C.; Burne, R.A. Genetic
and physiologic analysis of the groE operon and role of the
HrcA repressor in stress gene regulation and acid tolerance
in Streptococcus mutans. J. Bacteriol. 2001, 183,
6074–6084. 44. Hormann, K.M.; Saville, R.M.; Shukla, S.;
Spormann, A.M. Induction of rapid detachment in Shewanella
oneidensis MR-1 biofilms. J. Bacteriol. 2005, 187,
1014–1021. 45. Kreft, J.-U. Biofilms promote altruism.
Microbiology 2004, 150, 2751–2760. 46. Webb, J.S.; Lau,
A.U.; Kjellerberg, S. Bacteriophage and phenotypic
variation in Pseudomonas aeuginosa biofilm development. J.
Bacteriol. 2004, 186, 8066–8073. 47. Baveja, J.K.; Willcox,
M.D.P.; Hume, E.B.H.; Kumar, N.; Odell, R.; Poole-Warren,
A. Furanones as potential antibacterial coatings on
biomaterials. Biomaterials 2004, 25, 5003–5012. 48.
Cicalini, S.; Palmieri, F.; Petrosillo, N. New technologies
for prevention of intravascular catheter-related
infections. Crit. Care 2004, 8, 157–162. 49. Flemming,
R.G.; Murphy, C.J.; Abrams, G.A.; Goodman, S.L.; Nealey,
P.F. Effects of synthetic microand nano-structured surfaces
on cell behaviour. Biomaterials 1999, 20, 573–588. 50.
Dalby, M.J.; Childs, S.; Riehle, M.O.; Johnstone, H.J.H.;
Affrossman, S.; Curtis, A.S.G. Fibroblast reaction to
island topography: changes in cytoskeleton and morphology
with time. Biomaterials 2003, 24, 927–935. 51. Curtis, A.;
Wilkinson, C. Nanotechniques and approaches in
biotechnology. Trends Biotechnol. 2001, 19, 97–101. 52.
Cousins, B.G.; Doherty, P.J.; Williams, R.L.; Fink, J.;
Garvey, M.J. The effect of silica nanoparticulate coatings
on cellular response. J. Mater. Sci. Mater. Med. 2004, 15,
353–357. 53. Kojic, E.M.; Darouiche, R.O. Candida
infections on medical devices. Clin. Microbiol. Rev. 2004,
17, 255–267. 54. Daniels, R.; Vanderleyden, J.; Michiels,
J. Quorum sensing and swarming migration in bacteria. FEMS
Microbiol. Rev. 2004, 28, 261–289. 55. Friedman, L.;
Kolter, R. Genes involved in matrix formation in
Pseudomonas aeruginosa PA-14 biofilms. Mol. Microbiol. 2004,
51, 675–690. 56. Stoodley, P.; Sauer, K.; Davies, D.G.;
Costerton, J.W. Biofilms as complex differentiated
communities. Ann. Rev. Microbiol. 2002, 56, 187–209. 57.
Beloin, C.; Valle, J.; Latour-Lambert, P.; Faure, P.;
Kzreminski, M.; Balestrino, D.; Haagensen, J.A.J.; Molin,
S.; Prensier, G.; Arbeille, B.; Ghigo, J.M. Global impact
of mature biofilm lifestyle on Escherichia coli K-12 gene
expression. Mol. Microbiol. 2004, 51, 659–674. 58. HauBler,
S. Biofilm formation by the small colony variant phenotype
of Pseudomonas aeruginosa. Environ. Microbiol. 2004, 6,
546–551. 59. Yarwood, J.M.; Bartels, D.J.; Volper, E.M.;
Greenberg, E.P. Quorum sensing in Staphylococcus aureus
biofilms. J. Bacteriol. 2004, 186, 1838–1850. 60. Lyte, M.;
Freeston, P.P.E.; Neal, C.P.; Olson, B.A.; Haigh, R.D.;
Brayston, R.; Williams, P.H. Stimulation of Staphylococcus
epidermidis growth and biofilm formation by catecholamine
inotropes. Lancet 2003, 361, 130–135. 61. Carmen, J.C.;
Roeder, B.L.; Nelson, J.L.; Beckstead, B.L.; Runyan, C.M.;
Schaalje, G.B.; Robison, R.A.; Pitt, W.G. Ultrasonically
enhanced vancomycin activity against Staphylococcus
epidermidis. J. Biomater. Appl. 2004, 18, 237–245. 62.
Lerebour, G.; Cupferman, S.; Bellon-Fontaine, M.N. Adhesion
of Staphylococcus aureus and Staphylococcus epidermidis to
the Episkin reconstructed epidermis model and to an inert
304 stainless steel substrate. J. Appl. Microbiol. 2004, 7,
7–16. 63. Douglas, L.J. Candida biofilm and their role in
infection. Trends Microbiol. 2003, 11, 30–36. 64. Fux,
C.A.; Costerton, J.W.; Stewart, P.S.; Stoodley, P. Survival
strategies of infectious biofilms. Trends Microbiol. 2005,
13, 34–40. 65. Marchais, V.; Kempf, M.; Licznar, P.;
Lefrancois, C.; Bouchara, J-P; Robert, R.; Cottin, J. DNA
array analysis of Candida albicans gene expression in
response to adherence on polysterene. FEMS Microbiol. Lett.
2005, 245, 25–32. 66. Keren, I.; Shah, D.; Spoering, A.;
Kaldalu N. Lewis, K. Specialised persister cells and the
mechanism of multidrug tolerance in Escherichia coli. J.
Bacteriol. 2004, 186, 8172–8180. B
Biofunctional Polymers

Fig. 3 Pretreatment of mepacrine-labeled whole blood

with NO-producing hydrogel materials significantly reduced

platelet adhesion to a model thrombogenic surface,

collagen-coated glass. A, control hydrogel; B,

NO-generating hydrogel. 8. Hern, D.L.; Hubbell, J.A.


Incorporation of adhesion peptides into nonadhesive
hydrogels useful in tissue resurfacing. J. Biomed. Mater.
Res. 1998, 39, 266–277. 9. Mann, B.K.; West, J.L. Cell
adhesion peptides alter smooth muscle cell adhesion,
proliferation, and matrix protein synthesis on modified
surfaces and in polymer scaffolds. J. Biomed. Mater. Res.
2002, 60, 86–93. 10. Hubbell, J.A.; Massia, S.P.;
Drumheller, P.D. Endothelial cell-selective tissue
engineering in the vascular graft via a new receptor.
Bio/Technology 1991, 9, 568–572. 11. Rabbani, S.A.
Metalloproteases and urokinase in angiogenesis and tumor
progression. In Vivo 1998, 12, 135–142. 12. West, J.L.;
Hubbell, J.A. Polymeric biomaterials with degradation sites
for proteases involved in cell migration. Macromolecules
1999, 32, 241–244. 13. Gobin, A.S.; West, J.L. Cell
migration through defined synthetic ECM analogs. FASEB J.
2002, 16, 751–753. 14. Barritt, D.W.; Jordan, S.C.
Anticoagulant drugs in the treatment of pulmonary embolism:
A controlled clinical trial. Lancet 1960, 1, 1309–1312. 15.
Han, D.K.; Jeong, S.Y.; Kim, Y.H. Evaluation of blood
compatibility of PEO grafted and heparin immobilized
polyurethanes. J. Biomed. Mater. Res. 1989, 23, 211–228.
16. Han, D.K.; Park, K.D.; Ahn, K.D.; Kim, Y.H. Preparation
and surface characterization of PEO-grafted and heparin
immobilized polyurethanes. J. Biomed. Mater. Res. 1989, 23,
87–104. 17. Kim, Y.J.; Kang, I.K.; Muh, M.W.; Yoon, S.C.
Surface characterization and in vitro blood compatibility
of poly(ethylene terepthalate) immobilized with insulin
and/or heparin using plasma glow discharge. Biomaterials
2000, 21, 121–130. 18. Cholakis, C.H.; Sefton, M.V. In
vitro platelet interactions with a heparin-polyvinyl
alcohol hydrogel. J. Biomed. Mater. Res. 1989, 23, 399–415.
19. Piao, A.Z.; Jacobs, H.A.; Park, K.D.; Kim, S.W. Heparin
immobilization by surface amplification. ASAIO J. 1992, 38,
M638–M643. 20. Grainger, D.; Feijen, J.; Kim, S.W.
Poly(dimethyl siloxane)–poly(ethylene oxide)–heparin block
copolymers, I: synthesis and characterization. J. Biomed.
Mater. Res. 1988, 22, 231–242. 21. Garner, B.; Georgevich,
B.; Hodgson, A.J.; Liu, L.; Wallace, G.G.
Polypyrrole-heparin composites as stimulus-responsive
substrates for endothelial cell growth. J. Biomed. Mater.
Res. 1999, 44, 121–129. 22. Seifert, B.; Groth, T.;
Hermann, K.; Romaniuk, P. Immobilization of heparin on
polylactide for application to degradable biomaterials in
contact with blood. J. Biomater. Sci., Polym. Ed. 1995, 7,
277–287. 23. Byun, Y.; Jacobs, H.A.; Kim, S.W. Heparin
surface immobilization through hydrophilic spacers:
thrombin and antithrombin III binding kinetics. J.
Biomater. Sci. Polym. Ed. 1994, 6, 1–13. 24. West, D.C.;
Hampson, I.N.; Arnold, F.; Kuman, S. Angiogenesis induced
by degradation products of hyaluronic acid. Science 1985,
228, 1324–1326. 25. Vercruysse, K.P.; Marecak, D.M.;
Marecak, J.F.; Prestwich, G.D. Synthesis and in vitro
degradation of new polyvalent hydrazide crosslinked
hydrogels of hyaluronic acid. Bioconjug. Chem. 1997, 8,
686–694. 26. Verheye, S.; Markou, C.P.; Salame, M.Y.; Wan,
B.; King, S.B.; Robinson, K.A.; Chronos, N.A.; Hanson, S.R.
Reduced thrombus formation by hyaluronic acid coating of
endovascular devices. Arterioscler. Thromb. Vasc. Biol.
2000, 20, 1168–1172. 27. Barbucci, R.; Magnani, A.;
Rappuoli, R.; Lamponi, S.; Consumi, M. Immobilization of
sulfated hyaluronan for improved biocompatibility. J.
Inorg. Biochem. 2000, 79, 119–125. 28. Ito, R.K.; Phaneuf,
M.D.; LoGerfo, F.W. Thrombin inhibition by covalently bound
hirudin. Blood Coagul. Fibrinolysis. 1991, 2, 77–81. 29.
Phaneuf, M.D.; Berceli, S.A.; Bide, M.J.; Quist, W.C.;
LoGerfo, F.W. Covalent linkage of recombinant hirudin to
poly(ethylene terepthalate) (Dacron): Creation of a novel
antithrombin surface. Biomaterials 1997, 18, 755–765. 30.
Seifert, B.; Romaniuk, P.; Groth, T. Covalent
immobilization of hirudin improves the haemocompatibility
of polylactide-polyglycolide in vitro. Biomaterials 1997,
18, 1495–1502. 31. Torchilin, V.P.; Makismenko, A.V.;
Mazaev, A.V. Immobilized enzymes for thrombolytic therapy.
Meth. Enzym. 1988, 137, 552–566. 32. Kitamoto, Y.; Tomita,
M.; Kiyama, S.; Inoue, T.; Yabushita, Y.; Sato, T.; Ryoda,
H. Antithrombotic mechanisms of urokinase immobilized
polyurethane. Thrombosis 1991, 65, 73–76. 33. Forster,
R.I.; Bernath, F. Analysis of urokinase immobilization on
the polytetrafluoroethylene vascular prosthesis. Am. J.
Surg. 1988, 156, 130–132. 34. Sugitachi, A.; Tanaka, M.;
Kawahara, T.; Kitamura, N.; Takagi, K. A new type of drain
tube. Artif. Organs 1981, 5, 69. 35. Liu, L.S.; Ito, Y.;
Imanishi, Y. Biological activity of urokinase immobilized
to cross linked poly(2-hydroxyethyl methacrylate).
Biomaterials 1991, 12, 545–549. 36. Drummond, R.K.; Peppas,
N.A. Fibrinolytic behaior of streptokinase-immobilized
poly(methacrylic acid-gethylene oxide). Biomaterials 1991,
12, 356–360. 37. Ryu, G.H.; Park, S.; Kim, M.; Han, D.K.;
Kim, Y.H.; Min, B. Antithrombogenicity of
lumbrokinase-immobilized polyurethane. J. Biomed. Mater.
Res. 1994, 28, 1069–1077. 38. Kuhl, P.R.; Griffith-Cima,
L.G. Tethered epidermal growth factor as a paradigm for
growth factor-induced stimulation from the solid phase.
Nat. Med. 1996, 2, 1022–1027. 39. Ito, Y.; Li, J.S.;
Takahasi, T.; Imanishi, Y.; Okabayashi, Y.; Kido, Y.;
Kasuga, M. Enhancement of the mitogenic effect by artificial
juxtacrine stimulation using immobilized EGF. J. Biochem.
1997, 121, 514–520. 40. Ito, Y.; Chen, G.; Imanishi, Y.
Micropatterned immobilization of epidermal growth factor to
regulate cell function. Bioconjug. Chem. 1998, 9, 277–282.
41. Bos, G.W.; Scharenborg, N.M.; Poot, A.A.; Engbers,
G.H.M.; Beugling, T.; van Aken, W.G.; Feijen, J. B
Proliferation of endothelial cells on surface-immobilized
albumin-heparin conjugate loaded with basic fibroblast
growth factor. J. Biomed. Mater. Res. 1999, 44, 330–340.

42. Doi, K.; Matsuda, T. Enhanced vascularization in a


microporous polyurethane graft impregnated with basic
fibroblast growth factor and heparin. J. Biomed. Mater. Res.
1997, 34, 361–370.

43. Wissink, M.J.B.; Beernink, R.; Poot, A.A.; Engbers,


G.H.M.; Beugling, T.; van Aken, W.G.; Feijen, J. Improved
endothelialization of vascular grafts by local release of
growth factor from heparinized collagen matrices. J.
Control. Release 2000, 64, 103–114.

44. Mann, B.K.; Schmedlen, R.H.; West, J.L. Tehtered


TGF-beta increases extracellular matrix production of
vascular smooth muscle cells. Biomaterials 2001, 22,
439–444.

45. Erdmann, L.; Campe, C.; Palms, D.; Uhrich, K. Polymer


prodrugs with pharmaceutically active degradation products.
Polym. Prepr. 1997, 41, 1048–1049.

46. Erdmann, L.; Campe, C.; Bedell, C.; Uhrich, K.


Polymeric prodrugs: Novel polymers with bioactive
components. ACS Symp. Ser. 1998, 709, 83–91.

47. Krogh-Jespersen, E.; Anastasiou, T.; Uhrich, K.


Synthesis of a novelaromatic polyanhydride containing
aminosalicylic acid. Polym. Prepr. 2000, 41, 1048–1049.

48. Feldman, P.L.; Griffith, O.W.; Stuehr, D.J. The


surprising life of nitric oxide. Chem. Eng. News 1993, 71,
26–38.
49. Maragos, C.M.; Morley, D.; Wink, D.A.; Dunams, T.M.;
Saavedra, J.E.; Hoffman, A.; Bove, A.A.; Isaac, L.; Hrabie,
J.A.; Keefer, L.K. Complexes of NO with nucleophiles as
agents for the controlled biological release of nitric
oxide—Vasorelaxant effects. J. Med. Chem. 1991, 34,
3242–3247. 50. Loscalzo, J. Nitric oxide and restenosis.
Clin. Appl. Thromb./Hemost. 1996, 2, 7–10. 51. Smith, D.J.;
Chakravarthy, D.; Pulfer, S.; Simmons, M.L.; Hrabie, J.A.;
Citro, M.E.; Saavedra, J.E.; Davies, K.M.; Hutsell, T.C.;
Mooradian, D.L.; Hanson, S.R.; Keefer, L.K. Nitric
oxide-releasing polymers containing the [N(O)NO]-group. J.
Med. Chem. 1996, 39, 1148– 1156. 52. Pulfer, S.K.; Ott, D.;
Smith, D.J. Incorporation of nitric oxide-releasing
crosslinked polyethyleneimine microspheres into vascular
grafts. J. Biomed. Mater. Res. 1997, 37, 182–189. 53.
Mowery, K.A.; Schoenfisch, M.H.; Saavedra, J.E.; Keefer,
L.K.; Meyerhoff, M.E. Preparation and characterization of
hydrophobic polymeric films that are thromboresistant via
nitric oxide release. Biomaterials 2000, 21, 9–21. 54.
Bohl, K.S.; West, J.L. Nitric oxide-generating polymers
reduce platelet adhesion and smooth muscle cell
proliferation. Biomaterials 2000, 21, 2273–2278. 55.
Hill-West, J.L.; Chowdhury, S.M.; Slepian, M.J.; Hubbell,
J.A. Inhibition of thrombosis and intimal thickening by in
situ photopolymerization of thin hydrogel barriers. Proc.
Natl. Acad. Sci. U. S. A. 1994, 91, 5967–5971.
Biologic and Synthetic Apatites

28. LeGeros, R.Z. Formation and transformation of calcium


phosphates in biological and synthetic systemsrelevance to
vascular calcification. Kardiologie 90, Suppl 3: Vascular
Calcifications: Biophysics and Biology 2001, 203–209.

29. LeGeros, R.Z.; Sakae, T.; Bautista, C. Magnesium and


carbonate in enamel and synthetic apatites. Adv. Dent. Res.
1996, 10, 225–231.

30. Okazaki, M.; LeGeros, R.Z. Crystallographic and


chemical properties of Mg-containing apatites before and
after suspension in solutions. Magnes. Res. 1992, 5,
103–108.

31. LeGeros, R.Z.; Tung, M.S. Chemical stability of


carbonate and fluoride containing apatites. Caries Res.
1983, 17, 419–429.

32. LeGeros, R.Z.; LeGeros, J.P.; Trautz, O.R.; Klein, E.


Conversion of monetite, CaHPO4, to carbonate apatite:
effect on crystallinity. Adv. X-ray Appl. 1971, 14, 57–66.

33. Margolis, H.C.; Moreno, E.C. Kinetic and thermodynamic


aspects of enamel demineralization. Caries Res. 1985, 19,
22–35.

34. LeGeros, R.Z. Chemical and crystallographic events in


caries. J. Dent. Res. 1990, 69, 567–574. Special Issue.

35. Driessens, F.C.M. In Mineral Aspects of Dentistry;


Myers, H.M., Ed.; Monographs in Oral Sciences; Basel.
Karger, 1982; Vol. 10.

36. LeGeros, R.Z. The unit-cell dimensions of human enamel


apatite: effect of chloride incorporation. Arch. Oral Biol.
1974, 20, 63–68.

37. LeGeros, R.Z.; LeGeros, J.P.; Bonel, G. Types of ‘H 2


O’ in human enamel and in precipitated apatites. Calcif.
Tissue Res. 1979, 26, 111–116.

38. LeGeros, R.Z.; LeGeros, J.P.; Trautz, O.R.; Klein, E.


Spectral properties of carbonate in carbonate-containing
apatites. Dev. Appl. Spectrosc. 1970, 7, 3–12.

39. Moreno, E.G.; Kresak, M.; Zahradnik, R.T.


Physicochemical aspects of fluoride-apatite systems relevant
to the study of dental caries. Caries Res. 1977, 11 (Suppl.
1), 142–171.

40. LeGeros, R.Z.; Silverstone, L.M.; Daculsi, G.; Kerebel,


L.M. In vitro caries-like lesion formation in F-containing
tooth enamel. J. Dent. Res. 1983, 62, 138–144.

41. Dean, H.T.; Arnold, F.A., Jr; Elvove, E. Domestic water


and dental caries. V. Additional studies of the relations
of fluoride domestic waters to dental caries experience in
4,425 white children aged 12 to 14 years, of 13 cities in 4
states. Public Health Rep. 1942, 57, 115–125.

42. LeGeros, R.Z.; Singer, L.; Ophaug, R.; Quirolgico, G.;


LeGeros, J.P. The effect of fluoride on the stability of
synthetic and biological (bone mineral) apatites. In
Osteoporosis; Menczel, J., Robin, G.C., Makin, M., Eds.;
Wiley: New York, 1982; 327–341.

43. Robinson, C.; Kirkham, J. The effect of fluoride on the


developing mineralized tissues. J. Dent. Res. 1990, 69,
184–185.

44. Pak, C.Y.C.; Sakhaee, K.; Bell, N.K.; Licata, A.;


Johnson, C.; Rubine, B. Comparison of non-randomized trials
with slow-release sodium fluoride with a randomized
placebo-controlled trial in postmenopausal osteoporosis. J.
Bone Miner. Res. 1996, 11, 160–168. 45. Kleerkoper, M.
Fluoride and the skeleton. In Principles of Bone Biology;
Bilezlkian, J.P., Raisz, L.G., Rodan, F.A., Eds.; Academic
Press: San Diego, 1996; Chapter 75, 1053–1062. 46. Ito, A.;
Nakamura, S.; Aoki, H.; Akao, M.; Traoka, K.; Tsutsumi, S.;
Onuma, K.; Tateishi, T. Hydrothermal growth of
carbonate-containing hydroxyapatite single crystals. J.
Crystal Growth 1996, 163, 311–317. 47. Hattori, T.;
Iwadate, Y. Hydrothermal preparation of calcium
hydroxyapatite powders. J. Am. Ceram. Soc. 1990, 73,
1803–1805. 48. Pena, J.; LeGeros, R.Z.; Rohanizadeh, R.;
LeGeros, J.P. CaCO 3 –CaP biphasic materials prepared by
microwave processing of natural aragonite and calcite. Key
Eng. Mater. 2001, 192–195, 267–270. 49. Lerner, E.; Sarig,
S.; Azoury, R. Enhance maturation of hydroxyapatite from
aqueous solutions using microwave irradiation. J. Mater.
Sci.: Mater. Med. 1991, 2, 138–141. 50. Milev, A.S.
Chemistry, Synthesis and Morphological Stability of sol–gel
derived Carbonate Substituted PlateLike Hydroxyapatite. PhD
Thesis, University of Technology: Sydney, 2002. 51.
Deptula, A.; Lada, W.; Olczak, T.; Sarowska, S.; LeGeros,
R.Z.; LeGeros, J.P. Complex Sol–Gel Process (CSGP)
Preparation of Calcium Phosphate Biomaterials (Powders,
Monoliths, Fibers) In Bioceramics 11, World Scientific
Publishing: Singapore, 1998; 743–746. 52. LeGeros, R.Z.;
Morales, P. Renal stone crystals grown in gel systems.
Invest. Urol. 1973, 11, 12–20. 53. Vijayraghavan, T.V.;
Bensalem, A. Electrodeposition of apatite coating on pure
titanium and titanium alloys. J. Mater. Sci. Lett. 1994, 13
(24), 1782–1785. 54. LeGeros, J.P.; Lin, S.; LeGeros, R.Z.
Electrochemically deposited CaP coating on titanium alloy.
J. Dent. Res. 2000, 79, 560. 3332 Sp.Iss,SI. 55.
Rohanizadeh, R.; LeGeros, R.Z.; Harsono, M.; BenDavid, A.
Adherent apatite coating on titanium substrate using
chemical deposition. J. Biomed. Mater. Res. 2004, 72A,
428–438. 56. Kokubo, T. Formation of biologically active
bone-like apatite on metals and polymers by a bioamimetic
process. Thermochim. Acta 1996, 280, 479–490. 57. Hayek,
E.; Newesely, H. Pentacalcium monohydroxyorthophosphate.
Inorg. Synth. 1963, 7, 63–69. 58. LeGeros, R.Z.; Go, P.;
Vandemaele, K.H.; LeGeros, D.J. Transformation of calcium
carbonates and calcium phosphate to carbonate apatites:
possible mechanism for phosphorite formation. Proceedings
of 2nd International Congress on Phosphate Compounds,
Boston, 1980, 41–53. 59. LeGeros, R.Z.; Lin, S.;
Rohanizadeh, R.; Mijares, D.; LeGeros, J.P. Biphasic
calcium phosphate bioceramics: preparation, properties and
applications. J. Mater. Sci.: Mater. Med. 2003, 14,
201–210. 60. Brown, W.E.; Chow, L.C. A new calcium
phosphate water setting cement. In Cements Research
Progress; Brown, P.W., Ed.; American Ceramic Society:
Westerville, OH, 1987. 61. Niwa, S.; LeGeros, R.Z.
Injectable calcium phosphate cements for repair of bone
defects. In Tissue Engineering and Biodegradable
Equivalents: Scientific and Clinical Applications;
Lewandrowski, K.-U., Wise, D.L., Trantolo, D.J., Gresser,
J.D., Eds.; Marcel Dekker Inc.: New York, 2002; 385–500.
62. Albee, F.H. Studies in bone growth: triple calcium
phosphate as a stimulus to osteogenesis. Ann. Surg. 1920,
71, 32–36. 63. Nery, E.B.; Lynch, K.L.; Hirthe, W.M.;
Mueller, K.H. Preliminary clinical studies of bioceramics
in periodontal osseous defects. J. Periodontol. 1978, 49,
523–527. 64. Ellinger, R.F.; Mery, E.G.; Lynch, K.L.
Histological assessment of periodontal osseous defects
following implantation of hydroxyapatite and biphasic
calcium phosphate ceramics: a case report. Int. J.
Periodontics Restorative Dent. 1986, 3, 223–233. 65.
LeGeros, R.Z.; Daculsi, G.; Nery, E.; Lynch, K.; Kerebel,
B. In vivo transformation of biphasic calcium phosphates of
varying b-TCP/HA ratios: ultrastructural characterization.
Third World Biomaterials Congress, Kyoto, Japan, 1988. 66.
LeGeros, R.Z.; Daculsi, G. In vivo transformation of
biphasic calcium phosphate ceramics: histological,
ultrastructural and physico-chemical characterization. In
Yamamuro, T., Hench, L., Wilson-Hench, J., Eds.; Handbook
in Bioactive Ceramics; CRC Press: Boca Raton, 1990; Vol. 2,
17–28. 67. Daculsi, G.; Passuti, N. Bioactive ceramics
fundamental properties and clinical applications: the
osseo-coalescence process. In Bioceramics 2; Heimcke, G.,
Oonishi, H., Eds.; Cologne: Butterworh—Heinemann, Germany,
1990; 3–10. 68. Wykrota, L.L.; Garrido, C.A.; Wykrota,
F.H.I. Clinical evaluation of biphasic calcium phosphate
ceramic used in orthopaedic lesions. In Bioceramics 11;
LeGeros, R.Z., LeGeros, J.P., Eds.; World Scientific
Publishing: Singapore, 1998; 641–644. 69. Nery, E.;
LeGeros, R.Z.; Lynch, K.L.; Lee, K. Tissue response to
biphasic calcium phosphate ceramic with different ratios of
HA/b-TCP in periodontal osseous defects. J Periodontol.
1992, 63, 729–735. 70. Roy, D.L.; Linnehan, S.K.
Hydroxylapatite formed from coral skeletal carbonate by
hydrothermal exchange. Nature 1974, 247, 220–222. 71.
LeGeros, R.Z.; Bautista, C.; LeGeros, J.P.; Vijaraghavan,
T.V.; Retino, M. Comparative properties of bioactive bone
graft materials. In Bioceramics 8; Sedel, L., Ed.; Pergamon
Press: London, 1995; 1–7. 72. Berndt, C.C.; Haddad, G.N.;
Farmer, A.J.D.; Gross, K.A. Review: thermal spraying for
bioceramic applications. Mater. Forum 1990, 14, 161–173.
73. LeGeros, R.Z.; LeGeros, J.P.; Kim, Y.; Kijkowska, R.;
Zheng, R.; Bautista, C.; Wong, J.L. Calcium phosphates in
plasma-sprayed HA coatings. Ceram. Trans. 1995, 48,
173–189. 74. Lin, S.; LeGeros, R.Z.; LeGeros, J.P. Adherent
octacalcium phosphate coating on titanium alloy using a
modulated electrochemical deposition method. J. Biomed.
Mater. Res. 2003, 66A, 820–828. 75. Salgado, T.; LeGeros,
J.P.; Wang, J. Effect of alumina and apatitic abrasives on
Ti alloy substrates. In Bioceramics 11; Ohgushi, H., Ed.;
World Scientific Publishing: Singapore, 1998; 683–686. 76.
Doyle, C. Bioactive composites in orthopedics. In Yamamuro,
T., Henth, L., Wilson-Hench, J., Eds.; Handbook of
Bioactive Ceramics; CRC Press: Boca Raton 1990; Vol. 2,
195–207. 77. Okazaki, M.; Ohmae, H.; Hino, T.
Insolubilization of apatite-collagen composites by UV
irradiation. Biomaterials 1989, 10, 564–568. 78. Ohgushi,
H.; Caplan, A.I. Stem cell technology and bioceramics: from
cell to gene engineering. J. Biomed. Mater. Res. Appl.
Biomater. 1999, 48, 913–927. 79. Toquet, J.; Rohanizadeh,
R.; Guicheux, J.; Daculsi, G. Osteogenic potential in vitro
of human bone marrow cells cultured on macroporous biphasic
calcium phosphate ceramic. J. Biomed. Mater. Res. 1999, 44,
98–109. 80. LeGeros, R.Z. Properties of osteoconductive
biomaterials: calcium phosphates. Clin. Orthopaed Related
Res. 2002, 395, 81–98. 81. Klawitter, J.J. A Basic
Investigation of Bone Growth in Porous Materials. PhD
Thesis, Clemson University: Clemson, 1979. 82. Hubbard, W.
Physiological calcium phosphate as orthopedic implant
material. PhD Thesis. Marquette University: Milwaukee,
1974. 83. Harada, Y.; Want, J.-T.; Doppalppudi, V.A.;
Willis, A.A.; Goldring, S.R. Differential effects of
different forms of hydroxyapatite and
hydroxyapatite/tricalcium phosphate particles on human
monocyte/macrophages in vitro. J. Biomed. Mater. Res. 1996,
31, 19–26. 84. Hench, L.L. Bioceramics: from concept to
clinic. J. Am. Ceram. Soc. 1994, 74, 1487–1510. 85. Osborn,
J.F.; Newesely, H. The material science of calcium
phosphate ceramic. Biomaterials 1980, 1, 108–111. 86.
Heughebaert, M.; LeGeros, R.Z.; Gineste, M.; Guilhem, M.;
Bonel, G. Physico-chemical characterization of deposits
associated with HA ceramics implanted in nonosseous sites.
J. Biomed. Mater. Res. 1988, 22, 257–268. 87. LeGeros,
R.Z.; Daculsi, G.; Orly, I. Substrate surface dissolution
and interfacial biological mineralization. In The Bone
Biomaterials Interface; Davies, J.E., Ed.; University of
Toronto Press: Toronto, 1991; 76–88. 88. Urist, M.R. Bone
formation by autoinduction. Science 1965, 150, 893–898. 89.
Ripamonti, U.; Ma, S.; Reddi, A.H. The critical role of
geometry of porous hydroxyapatite delivery system induction
of bone by osteogenin, a bone morphogenetic protein. Matrix
1992, 12 (3), 202–212. 90. Le Nihouannen, D.; Daculsi, G.;
Saffarzadeh, A.; Gauthier, O.; Delplace, S.; Pilet, P.;
Layrolle, P. Ectopic bone formation by microporous calcium
phosphate ceramic particles in sheep muscles. Bone 2005, 36
(6), 1086–1093. B
Biological Adhesives from Nature

14. Olivieri, M.P.; Wollman, R.M.; Alderfer, J.L. Nuclear


magnetic resonance spectroscopy of mussel adhesive protein
repeating peptide segment. J. Pept. Res. 1997, 50 (6),
436–442.

15. Kanyalkar, M.; Srivastava, S.; Coutinho, E.


Conformation of a model peptide of the tandem repeat
decapeptide in mussel adhesive protein by NMR and MD
simulations. Biomaterials 2002, 23 (2), 389–396.

16. Waite, J.H.; Qin, X. Polyphosphoprotein from the


adhesive pads ofMytilus edulis. Biochemistry 2001, 40 (9),
2887–2893.

17. Naldrett, M.J.; Kaplan, D.L. Characterization of


barnacle (Balanus eburneus and B. crenatus) adhesive
proteins. Mar. Biol. 1997, 127 (4), 629–635.

18. Wiegemann, M. Adhesion in blue mussels (Mytilus edulis)


and barnacles (genus Balanus): mechanisms and technical
applications. Aquat. Sci. 2005, 67 (2), 166–176.

19. Kaplan, D.L.; Gatenholm, P.; Berglin, K.M.; Platko,


J.D.; Pepper, L.R.; Ngangan, A.V. Barnacle adhesion
proteins. International Patent WO 03/093413, Apr 24, 2003.

20. Naldrett, M.J. The importance of sulphur cross-links


and hydrophobic interactions in the polymerization of
barnacle cement. J. Mar. Biol. Ass. U.K. 1993, 73 (3),
689–702.

21. Kamino, K.; Inoue, K.; Maruyama, T.; Takamatsu, N.;


Harayama, S.; Shizuri, Y. Barnacle cement proteins.
Importance of disulfide bonds in their insolubility. J.
Biol. Chem. 2000, 275 (35), 27360–27365.

22. Kamino, K. Novel barnacle underwater adhesive protein


is a charged amino acid-rich protein constituted by a
Cys-rich repetitive sequence. Biochem. J. 2001, 356 (2),
503–507.

23. Barnacle Adhesion Protein Gene—For Production of


Adhesive Protein. Japanese Patent JP7,265,081, Mar 31, 1994
(Kaiyo Biotechnology Kenkyusho KK).

24. Kamino, K.; Odo, S.; Maruyama, T. Cement proteins of


the acorn barnacle, Megabalanus rosa. Biol. Bull. 1996, 190
(3), 403–409.
25. Barnacle Second Adhesive Protein Gene—Providing Protein
Useful as Starting Material for Adhesive or as Substrate in
Cell Culture. Japanese Patent JP9,047,288, Aug 9, 1995
(Kaiyo Biotechnology Kenkyusho KK).

26. New Barnacle Gene That Encodes the Fourth Adhesive


Protein—Which May Be Used as a Bioadhesive. Japanese Patent
JP10,327,867, May 30, 1997 (Kaiyo Biotechnology Kenkyusho
KK).

27. Barnacle Third Adhesion Protein Gene—Useful for


Producing Protein Recombinantly Used as Raw Material for
Adhesives. Japanese Patent JP9,299,089, May 17, 1996 (Kaiyo
Biotechnology Kenkyusho KK).

28. Nakano, M.; Shen, J.R.; Urushida, Y.; Mori, Y.; Kamino,
K. Peptide-based material design from barnacle underwater
adhesive proteins. In Transactions; 7th World Biomaterials
Congress, Sydney, Australia, May 17–21, 2004; Australian
Society for Biomaterials Inc.: Melbourne, Victoria, 2004;
1649.

29. Flammang, P.; Miche, A.; Cauwenberge, A.V.; Alexandre,


H.; Jangoux, M. A study of the temporary adhesion of the
podia in the sea star Asterias rubens (Echinodermata,
Asteroidea) through their footprints. J. Exp. Biol. 1998,
201 (16), 2383–2395. 30. Santos, R.; Haesaerts, D.;
Jangoux, M.; Flammang, P. Comparative histological and
immunohistochemical study of sea star tube feet
(Echinodermata, Asteroidea). J. Morphol. 2005, 263 (3),
259–269. 31. Santos, R.; Gorb, S.; Jamar, V.; Flammang, P.
Adhesion of echinoderm tube feet to rough surfaces. J. Exp.
Biol. 2005, 208 (13), 2555–2567. 32. DeMoor, S.; Waite,
J.H.; Jangoux, M.; Flammang, P. Characterization of the
adhesive from Cuvierian tubules of the sea cucumber
Holothuria forskali (Echinodermata, Holothuroidea). Mar.
Biotechnol. 2003, 5 (1), 45–57. 33. Smith, A.M.; Quick,
T.J.; St. Peter, R.L. Differences in the composition of
adhesive and non-adhesive mucus from the limpet Lottia
limatula. Biol. Bull. 1999, 196 (1), 34–44. 34. Smith,
A.M.; Morin, M.C. Biochemical differences between trail
mucus and adhesive mucus from marsh periwinkle snails.
Biol. Bull. 2002, 203 (3), 338–346. 35. Jones, I.;
Lindberg, C.; Jakobsson, S.; Hellqvist, A.; Hellman, U.;
Borg, B.; Olsson, P.E. Molecular cloning and
characterization of spiggin. J. Biol. Chem. 2001, 276 (21),
17857–17863. 36. Olsson, P-E.; Jaas, J.; Jones, I.;
Jakobsson, S.; Borg, B. Adhesive Protein and Uses Thereof.
U.S. Patent 2004/ 0072183, Apr 15, 2004. 37. Sadler, J.E.
Biochemistry and genetics of vonWillebrand factor. Annu.
Rev. Biochem. 1998, 67, 395–424. 38. Combie, J. Natural
polymer with adhesive properties produced by bacteria.
Adhesives & Sealants Industry Website, Cover Story, Feb
2003; http://www.adhesives
mag.com/CDA/ArticleInformation/coverstory/BNPCov
erStoryItem/0,2103,101154,00.html (accessed Jan 2005). 39.
Haag, A.P.; Maier, R.M.; Combie, J.; Geesey, G.G. Int. J.
Adhes. Adhes. 2004, 24 (6), 495–502. 40. Remaut, H.;
Waksman, G. Structural biology of bacterial pathogenesis.
Curr. Opin. Struct. Biol. 2004, 14 (2), 161–170. 41.
Wagner, V.T.; Brian, L.; Quatrano, R.S. Role of a
vitronectin-like molecule in embryo adhesion of the brown
alga Fucus. Proc. Natl. Acad. Sci. USA 1992, 89 (8),
3644–3648. 42. Levi, B.; Friedlander, M. Identification of
two putative adhesive polypeptides in Caulerpa prolifera
rhizoids using an adhesion model system. J. Appl. Phycol.
2004, 16 (1), 1–9. 43. Stanley, M.S.; Callow, M.E.; Callow,
J.A. Monoclonal antibodies to adhesive cell coat
glycoproteins secreted by zoospores of the green alga
Enteromorpha. Planta 1999, 210 (1), 61–71. 44. Callow,
J.A.; Crawford, S.A.; Higgins, M.J.; Mulvaney, P.;
Wetherbee, R. The application of atomic force microscopy to
topographical studies and force measurements on the
secreted adhesive of the green alga Enteromorpha. Planta
2000, 211 (5), 641–647. 45. Vreeland, V.; Waite, J.H.;
Epstein, L. Polyphenols and oxidases in substratum adhesion
by marine algae and mussels. J. Phycol. 1998, 34 (1), 1–8.
46. Berglin, M.; Delage, L.; Potin, P.; Vilter, H.; Elwing,
H. Enzymatic cross-linking of a phenolic polymer extracted
from the marine alga Fucus serratus. Biomacromolecules
2004, 5 (6), 2376–2383. 47. Scholtmeijer, K.; Wessels,
J.G.; Wo¨sten, H.A. Fungal hydrophobins in medical and
technical applications. Appl. Microbiol. Biotechnol. 2001,
56 (1–2), 1–8. 48. Wessels, J.G.H. Fungi in their own
right. Fungal Gen. Biol. 1999, 27 (2–3), 134–145. 49. Kwon,
Y.H.; Epstein, L. Involvement of the 90 kDa glycoprotein in
adhesion of Nectria haematococca macroconidia. Physiol.
Mol. Plant Pathol. 1997, 51 (5), 287–303. 50. Sugui, J.A.;
Leite, B.; Nicholson, R.L. Partial characterization of the
extracellular matrix released onto hydrophobic surfaces by
conidia and conidial germlings of Colletotrichum
graminicola. Physiol. Mol. Plant Pathol. 1998, 52 (6),
411–425. 51. Vardar-U¨nlu¨, G. Mannoprotein adhesin of
Candida albicans germ tubes. Turk. J. Med. Sci. 1998, 28
(5), 469–474. 52. Staab, J.F.; Bahn, Y.S.; Tai, C.H.; Cook,
P.F.; Sundstrom, P. Expression of transglutaminase
substrate activity on Candida albicans germ tubes through a
coiled, disulfide-bonded N-terminal domain of Hwp1 requires
C-terminal glycosylphosphatidylinositol modification. J.
Biol. Chem. 2004, 279 (39), 40737–40747. 53. Hamwood, T.E.;
Cribb, B.W.; Halliday, J.A.; Kearn, G.C.; Whittington, I.D.
Preliminary characterisation and extraction of anterior
adhesive secretion in monogenean (platyhelminth) parasites.
Folia Parasitol. 2002, 49 (1), 39–49. 54. Yamamoto, H.
Adhesive Composition with Potential use as Bioadhesive in
Humans. Japanese Patent JP7,188,640, Jul 19, 1991. 55.
Shen, X.; Belcher, A.M.; Hansma, P.K.; Stucky, G.D.; Morse,
D.E. Molecular cloning and characterization of lustrin A, a
matrix protein from shell and pearl nacre of Haliotis
rufescens. J. Biol. Chem. 1997, 272 (51), 32472–32481. 56.
Waite, J.H.; Jensen, R.A.; Morse, D.E. Cement precursor
proteins of the reef-building polychaete Phragmatopoma
californica (Fewkes). Biochemistry 1992, 31 (25),
5733–5738. 57. Stewart, R.J.; Weaver, J.C.; Morse, D.E.;
Waite, J.H. The tube cement of Phragmatopoma californica: a
solid foam. J. Exp. Biol. 2004, 207 (26), 4727–4734. 58.
Kiel, E.; Ro¨der, T. Gelelectrophoretic studies on labial
gland secretions of immature blackflies (Simuliidae,
Diptera). Limnologica 2002, 32 (3), 201–205. 59.
Nokhbatolfoghahai, M.; Downie, J.R. Larval cement gland of
frogs: comparative development and morphology. J. Morphol.
2005, 263 (3), 270–283. 60. Evans, C.M.; Brodie, E.D.
Adhesive strength of amphibian skin secretions. J.
Herpetol. 1994, 28 (4), 499–502. 61. Williams, T.A.;
Anthony, C.D. Technique to isolate salamander granular
gland products with a comment on the evolution of
adhesiveness. Copeia 1994, (2), 540–541. 62. Tyler, M.J.;
Ramshaw, J.A.M. An Adhesive Derived From Amphibian Skin
Secretions. International Patent WO 02/22756, Sep 18, 2001.
63. Graham, L.D.; Glattauer, V.; Huson, M.G.; Maxwell,
J.M.; Knott, R.B.; White, J.W.; Vaughan, P.R.; Peng, Y.;
Tyler, M.J.; Werkmeister, J.A.; Ramshaw, J.A.
Characterization of a protein-based adhesive elastomer
secreted by the Australian frog Notaden bennetti.
Biomacromolecules. Available online at http://dx.doi.
org/10.1021/bm050335e. 64. Graham, L.D.; Glattauer, V.;
Peng, Y.Y.; Vaughan, P.R.; Werkmeister, J.A.; Tyler, M.J.;
Ramshaw, J.A. An adhesive secreted by Australian frogs of
the genus Notaden. In Biological Adhesives; Smith, A.M.,
Callow, J.A., Eds.; Springer-Verlag: Berlin, in press 65.
Nuttall, P.A.; Paesen, G.C. Tissue Cement Proteins Produced
by Blood-Feeding Ectoparasites and Related Polynucleotides.
International Patent WO 99/ 24567, Nov 12, 1998. 66.
Trimnell, A.R.; Paesen, G.C.; Nuttall, P.A. Vaccine
Comprising a Tick Cement Protein. International Patent WO
01/80881, Apr 25, 2001. 67. Mulenga, A.; Sugimoto, C.;
Sako, Y.; Ohashi, K.; Musoke, A.; Shubash, M.; Onuma, M.
Molecular characterization of a Haemaphysalis longicornis
tick salivary gland-associated 29-kilodalton protein and
its effect as a vaccine against tick infestation in
rabbits. Infect. Immun. 1999, 67 (4), 1652–1658. 68.
Bishop, R.; Lambson, B.; Wells, C.; Pandit, P.; Osaso, J.;
Nkonge, C.; Morzaria, S.; Musoke, A.; Nene, V. A cement
protein of the tick Rhipicephalus appendiculatus, located
in the secretory e cell granules of the type III salivary
gland acini, induces strong antibody responses in cattle.
Int. J. Parasitol. 2002, 32 (7), 833–842. 69. Guilfoile,
P.G.; Packila, M. Identification of four genes expressed by
feeding female Ixodes scapularis, including three with
sequence similarity to previously recognized genes. Exp.
Appl. Acarol. 2004, 32 (1–2), 103–110. 70. Beckendorf,
S.K.; Kafatos, F.C. Differentiation in the salivary glands
of Drosophila melanogaster: characterization of the glue
proteins and their developmental appearance. Cell 1976, 9
(3), 365–373. 71. Korge, G. Larval saliva in Drosophila
melanogaster: production, composition, and relationship to
chromosome puffs. Dev. Biol. 1977, 58 (2), 339–355. 72.
Swida, U.; Lucka, L.; Kress, H. Glue protein genes in
Drosophila virilis: their organization, developmental
control of transcription and specific mRNA degradation.
Development 1990, 108 (2), 269–280. 73. Lanio, W.; Swida,
U.; Kress, H. Molecular cloning of the Drosophila virilis
larval glue protein gene Lgp-3 and its comparative analysis
with other Drosophila glue protein genes. Biochem. Biophys.
Acta 1994, 1219 (2), 576–580. 74. Kress, H. Biochemical and
ontogenetic aspects of glycoprotein synthesis in Drosophila
virilis salivary glands. Dev. Biol. 1982, 93 (1), 231–239.
75. Ramesh, S.R.; Kalisch, W.E. Glue proteins in Drosophila
nasuta. Biochem. Genet. 1988, 26 (7–8), 527–541. B

76. Lung, O.; Wolfner, M.F. Identification and


characterization of the major Drosophila melanogaster
mating plug protein. Insect Biochem. Mol. Biol. 2001, 31
(6–7), 543–551.

77. Graham, L.D. Unpublished data.

78. Baer, B.; Maile, R.; Schmid-Hempel, P.; Morgan, E.D.;


Jones, G.R. Chemistry of a mating plug in bumblebees. J.
Chem. Ecol. 2000, 26 (8), 1869–1875.

79. Zhu, L.J.; Arai, M.; Hirabayashi, K. Sol-gel transition


of sericin. Nippon Sanshigaku Zasshi 1996, 65 (4), 270–274.

80. Takasu, Y.; Yamada, H.; Tsubouchi, K. Isolation of


three main sericin components from the cocoon of the
silkworm, Bombyx mori. Biosci. Biotechnol. Biochem. 2002,
66 (12), 2715–2718.
81. Huang, J.; Valluzzi, R.; Bini, E.; Vernaglia, B.;
Kaplan, D.L. Cloning, expression, and assembly of
sericinlike protein. J. Biol. Chem. 2003, 278 (46), 46117–
46123.

82. Bullough, P.A.; Tulloch, P.A. High-resolution spotscan


electron microscopy of microcrystals of an alphahelical
coiled-coil protein. J. Mol. Biol. 1990, 215 (1), 161–173.

83. Kenchington, W.; Flower, N.E. Studies on insect fibrous


proteins: the structural protein of the ootheca in the
praying mantis, Sphodromantis centralis (Rehn). J. Microsc.
1969, 89 (2), 263–281.

84. Rudall, K.M. Protein ribbons and sheets. In Lectures on


the Scientific Basis of Medicine; Athlone Press: London,
1959; Vol. 5, 217–230.

85. Yago, M.; Sato, H.; Oshima, S.; Kawasaki, H. Enzymatic


activities involved in the oothecal sclerotization of the
praying mantid, Tenodera aridifolia Sinensis (Saussure).
Insect Biochem. 1990, 20 (7), 745–750.

86. Hinton, H.E. Biology of Insect Eggs; Pergamon Press:


Oxford, U.K., 1981; Vol. I, 193–196, 200, and 634.

87. Li, D.; Graham, L.D. Unpublished data.

88. Tillinghast, E.K. Selective removal of glycoproteins


from the adhesive spiral of the spiders orb web.
Naturwissenschaften 1981, 68, 526–527.

89. Dreesbach, K.; Uhlenbruck, G.; Tillinghast, E.K.


Carbohydrates of the trypsin soluble fraction of the orb
web of Argiope trifasciata. Insect Biochem. 1983, 13 (6),
627–631.

90. Vollrath, F.; Tillinghast, E.K. Glycoprotein glue


beneath a spider web’s aqueous coat. Naturwissenschaften
1991, 78 (12), 557–559.

91. Benkendorff, K.; Beardmore, K.; Gooley, A.A.; Packer,


N.H.; Tait, N.N. Characterisation of the slime gland
secretion from the peripatus, Euperipatoides kanangrensis
(Onychophora: Peripatopsidae). Comp. Biochem. Physiol. B.
1999, 124 (4), 457–465.

92. Kolsch, G. The ultrastructure of glands and the


production and function of the secretion in the adhesive
capture apparatus of Stenus species (Coleoptera:
Staphylinidae). Can. J. Zool. 2000, 78 (3), 465–475.

93. Benthien, J.P.; Russlies, M.; Behrens, P. Investigating


the effects of bone cement, cyanoacrylate glue and marine
mussel adhesive protein fromMytilus edulis on human
osteoblasts and fibroblasts in vitro. Ann. Anat. 2004, 186
(5–6), 561–566. 94. Schnurrer, J.; Lehr, C.M. Mucoadhesive
properties of the mussel adhesive protein. Int. J. Pharm.
1996, 141 (1–2), 251–256. 95. Chivers, R.A.; Wolowacz, R.G.
The strength of adhesive-bonded tissue joints. Int. J.
Adhes. Adhes. 1997, 17 (2), 127–132. 96. Benedict, C.V.;
Picciano, P.T. Adhesives Derived from Polyphenolic
Proteins. U.S. Patent 5,015,677, Jun 27, 1988. 97. Ninan,
L.; Monahan, J.; Stroshine, R.L.; Wilker, J.J.; Shi, R.
Adhesive strength of marine mussel extracts on porcine
skin. Biomaterials 2003, 24 (22), 4091–4099. 98. Yamamoto,
H.; Sakai, Y.; Ohkawa, K. Synthesis and wettability
characteristics of model adhesive protein sequences
inspired by a marine mussel. Biomacromolecules 2000, 1 (4),
543–551. 99. Hwang, D.S.; Yoo, H.J.; Jun, J.H.; Moon, W.K.;
Cha, H.J. Expression of functional recombinant mussel
adhesive protein Mgfp-5 in Escherichia coli. Appl. Environ.
Microbiol. 2004, 70 (6), 3352–3359. 100. Hwang, D.S.; Gim,
Y.; Cha, H.J. Expression of functional recombinant mussel
adhesive protein type 3A in Escherichia coli. Biotechnol.
Prog. 2005, 21 (3), 965–970. 101. Benedict, C.V.; Picciano,
P.T. Method for Making Dopa-Containing Bioadhesive Proteins
from TyrosineContaining Proteins. European Patent
EP242,656, Apr 2, 1987. 102. Salerno, A.J.; Goldberg, I.
Cloning, expression, and characterization of a synthetic
analog to the bioadhesive precursor protein of the sea
mussel Mytilus edulis. Appl. Microbiol. Biotechnol. 1993,
39 (2), 221–226. 103. Kitamura, M.; Kawakami, K.; Nakamura,
N.; Tsumoto, K.; Uchiyama, H.; Ueda, Y.; Kumagai, I.;
Nakaya, T. Expression of a model peptide of a marine mussel
adhesive protein in Escherichia coli and characterization
of its structural and functional properties. J. Polym. Sci.
A. 1999, 37 (6), 729–736. 104. McQueen Mason, S.; Filatov,
V. Transgenic Cells Expressing Adhesive Proteins.
International Patent WO 03/082912, Mar 24, 2003. 105.
Kieliszewski, M.J.; Lamport, G.T.A. Extensin— repetitive
motifs, functional sites, posttranslational codes, and
phylogeny. Plant J. 1994, 5 (2), 157–172. 106. Tatehata,
H.; Mochizuki, A.; Kawashima, T.; Yamashita, S.; Yamamoto,
H. Model polypeptide of mussel adhesive protein. I.
Synthesis and adhesive studies of sequential polypeptides
(X-Tyr-Lys) n and (Y-Lys) n . J. Appl. Polym. Sci. 2000, 76
(6), 929–937. 107. Tatehata, H.; Mochizuki, A.; Ohkawa, K.;
Yamada, M.; Yamamoto, H. Tissue adhesive using synthetic
model adhesive proteins inspired by the marine mussel. J.
Adhes. Sci. Technol. 2001, 15 (9), 1003–1013. 108. Huang,
K.; Lee, B.P.; Ingram, D.R.; Messersmith, P.B. Synthesis
and characterization of self-assembling block copolymers
containing bioadhesive end groups. Biomacromolecules 2002,
3 (2), 397–406. 109. Lee, B.P.; Huang, K.; Nunalee, F.N.;
Shull, K.R.; Messersmith, P.B. Synthesis of
3,4-dihydroxyphenylalanine (Dopa) containing monomers and
their co-polymerization with PEG-diacrylate to form
hydrogels. J. Biomater. Sci. Polym. Ed. 2004, 15 (4),
449–464. 110. Zanchetta, P.; Lagarde, N.; Guezennec, J. A
new bone-healing material: a hyaluronic acid-like bacterial
exopolysaccharide. Calcif. Tissue Int. 2003, 72 (1), 74–79.
111. Guezennec, J.; Zanchetta, P.; Durand, P. Use of a
Polysaccharide Excreted by the Vibrio diabolicus Species in
Bone Repair. International Patent WO 02/ 02051, Jul 4,
2001. 112. Haber, M. Plant Proteins. U.S. Patent 5,859,198,
Aug 31, 1995. 113.
http://algal-adhesives.ocean.org.il/index.html (accessed
Mar 2005). 114. http://www.algawish.com/index.html
(accessed Mar 2005). 115. Szomor, Z.L.; Appleyard, R.;
Tyler, M.J.; Murrell, G.A.C. Meniscal repair with a frog
glue. In Abstracts, Pre-Olympic Congress on Sports Medicine
and Physical Education and International Congress on Sport
Science, Brisbane, Australia, Sep 7–13, 2000; National
Sport Information Centre: Canberra, Australia, 2000; 297.
http://www.ausport.gov.au/fulltext/2000/preoly/abs297. htm
(accessed Nov 2004). 116. Szomor, Z.L.; Appleyard, R.;
Tyler, M.J.; Murrell, G.A.C. Meniscal repair with a new
biologic glue. Abstracts, 47th Annual Meeting, Orthopaedic
Research Society, San Francisco, CA, Feb 25–28, 2001;
Orthopaedic Research Society: Illinois, 2001; Session 8,
0047. 117. Crisp, D.J.; Walker, G.; Young, G.A.; Yule, A.B.
Adhesion and substrate choice in mussels and barnacles. J.
Colloid Interface Sci. 1985, 104 (1), 40–50. 118. Yamamoto,
H. Synthesis and adhesive studies of marine polypeptides.
J. Chem. Soc. Perkin Trans. 1987, 1 (3), 613–618. 119.
Yule, A.B.; Walker, G. Adhesion in barnacles. In Crustacean
Issues, Biology of Barnacles; Southward, A.J., Ed.;
Balkema: Rotterdam, 1987; 389–402. 120. Flammang, P.;
Ribesse, J.; Jangoux, M. Biomechanics of adhesion in sea
cucumber Cuvierian tubules (Echinodermata, Holothuroidea).
Integr. Comp. Biol. 2002, 42 (6), 1107–1115. 121. Flammang,
P.; Walker, G. Measurement of the adhesion of the podia in
the asteroid Asterias rubens (Echinodermata). J. Mar. Biol.
Assoc. U.K. 1997, 77 (4), 1251–1254. 122. Grenon, J-F.;
Walker, G. The tenacity of the limpet Patella vulgata L.:
an experimental approach. J. Exp. Mar. Biol. Ecol. 1981,
54, 277–308. 123. Roscoe, D.T.; Walker, G. Observations on
the adhesion of the calcareous tube of Pomatoceros
lamarckii and the holdfast of Laminaria digitata.
Biofouling 1995, 9 (1), 39–50. 124. Berglin, M.; Gatenholm,
P. The nature of bioadhesive bonding between barnacles and
fouling-release silicone coatings. J. Adhes. Sci. Technol.
1999, 13 (6), 713–727. 125. McDermott, M.K.; Chen, T.;
Williams, C.M.; Markley, K.M.; Payne, G.F. Mechanical
properties of biomimetic tissue adhesive based on the
microbial transglutaminase-catalyzed crosslinking of
gelatin. Biomacromolecules 2004, 5 (4), 1270–1279. 126.
Pa´ez, J.M.G.; Herrero, E.J.; Rocha, A.; Maestro, M.;
Castillo-Olivares, J.L.; Millan, I.; Sanmartin, A.C.;
Cordon, A. Comparative study of the mechanical behaviour of
a cyanoacrylate and a bioadhesive. J. Mater. Sci. Mater.
Med. 2004, 15 (2), 109–115. 127. Edlund, A.F.; Koehl,
M.A.R. Adhesion and reattachment of compound ascidians to
various substrata: weak glue can prevent tissue damage. J.
Exp. Biol. 1998, 201 (16), 2397–2402. B
Biological Effects of Radiofrequency
Electromagnetic Field

43. Lai, H.; Horita, A.; Guy, A.W. Acute low-level


microwave exposure and central cholinergic activity:
studies on irradiation parameters. Bioelectromagnetics
1988, 9, 355–362.

44. Lai, H.; Carino, M.A.; Horita, A.; Guy, A.W. Acute
low-level microwave exposure and central cholinergic
activity: a dose-response study. Bioelectromagnetics 1989,
10, 203–209.

45. Arber, S.L.; Lin, J.C. Microwave-induced changes in


nerve cells: effects of modulation and temperature.
Bioelectromagnetics 1985, 6, 257–270.

46. Frey, A.H.; Feld, S.R. Avoidance by rats of


illumination with low power nonionizing electromagnetic
radiation. J. Comp. Physiol. Psychol. 1975, 89, 183–188.

47. Sanders, A.P.; Joines, W.T.; Allis, J.W. Effect of


continuous-wave, pulsed, and sinusoidal-amplitudemodulated
microwaves on brain energy metabolism. Bioelectromagnetics
1985, 6, 89–97.

48. Baranski, S. Histological and histochemical effects of


microwave irradiation on the central nervous system of
rabbits and guinea pigs. Am. J. Physiol. Med. 1972, 51,
182–190.

49. Takashima, S.; Onaral, B.; Schwan, H.P. Effects of


modulated RF energy on the EEG of mammalian brain. Radiat.
Environ. Biophys. 1979, 16, 15–27.

50. Johnson, R.B.; Spackman, D.; Crowley, J.; Thompson, D.;


Chou, C.K.; Kunz, L.L.; Guy, A.W. Effects of Long-Term
Low-Level Radiofrequency Radiation Exposure on Rats, Vol.
4. Open field Behavior and Corticosterone; Brooks AFB: San
Antonio, TX, 1983; USAF SAM-TR83-42; Report of USAF School
of Aerospace Medicine.

51. Lai, H.; Horita, A.; Chou, C.K.; Guy, A.W. Effects of
low-level microwave irradiation on hippocampal and frontal
cortical choline uptake are classically conditionable.
Pharmacol. Biochem. Behav. 1987, 27, 635–639.

52. Lai, H.; Carino, M.A.; Horita, A.; Guy, A.W. Single vs.
repeated microwave exposure: effects on benzodiazepine
receptors in the brain of the rat. Bioelectromagnetics
1992, 13, 57–66.

53. Di Carlo, A.; White, N.; Guo, F.; Garrett, P.;


Litovitz, T. Chronic electromagnetic field exposure
decreases HSP70 levels and lowers cytoprotection. J. Cell.
Biochem. 2002, 84, 447–454.

54. Dumansky, J.D.; Shandala, M.G. The biologic action and


hygienic significance of electromagnetic fields of super high
and ultra high frequencies in densely populated areas,
Biologic Effects and Health Hazard of Microwave Radiation:
Proceedings of an International Symposium; Czerski, P., et
al., Ed.; Polish Medical Publishers: Warsaw, 1974.

55. de Lorge, J.O. Operant behavior and colonic temperature


of Macaca mulatta exposed to radiofrequency fields at and
above resonant frequencies. Bioelectromagnetics 1984, 5,
233–246.

56. Persson, B.R.R.; Salford, L.G.; Brun, A. Blood-brain


barrier permeability in rats exposed to electromagnetic
fields used in wireless communication. Wireless Network
1997, 3, 455–461. 57. Lai, H.; Carino, M.A.; Horita, A.;
Guy, A.W. Single vs. repeated microwave exposure: effects
on benzodiazepine receptors in the brain of the rat.
Bioelectromagnetics 1992, 13, 57–66. 58. Lai, H.; Horita,
A.; Chou, C.K.; Guy, A.W. A review of microwave irradiation
and actions of psychoactive drugs. IEEE Eng. Med. Biol.
1987, 6, 31–36. 59. D’Inzeo, G.; Bernardi, P.; Eusebi, F.;
Grassi, F.; Tamburello, C.; Zani, B.M. Microwave effects on
acetylcholine-induced channels in cultured chick myotubes.
Bioelectromagnetics 1988, 9, 363–372. 60. Johnson, C.C.;
Guy, A.W. Nonionizing electromagnetic wave effect in
biological materials and systems. Proc. IEEE 1971, 60,
692–718. 61. Seaman, R.L.; Wachtel, H. Slow and rapid
responses to CW and pulsed microwave radiation by
individual Aplysia pacemakers. J. Microw. Power 1978, 13,
77–86. 62. Wachtel, H.; Seaman, R.; Joines, W. Effects of
low-intensity microwaves on isolated neurons. Ann. N.Y.
Acad. Sci. 1975, 247, 46–62. 63. Chou, C.K.; Guy, A.W.;
Galambos, R. Auditory perception of radiofrequency
electromagnetic fields. J. Acoust. Soc. Am. 1982, 71,
1321–1334. 64. Lebovitz, R.M. Detection of weak
electromagnetic radiation by the mammalian
vestibulocochlear apparatus. Ann. N.Y. Acad. Sci. 1975,
247, 182–193. 65. Guy, A.W.; Chou, C.K.; McDougall, J.A. A
quarter century of in vitro research: a new look at
exposure methods. Bioelectromagnetics 1999, (suppl. 4),
21–39. 66. Diem, E.; Schwarz, C.; Adlkofer, F.; Jahn, O.;
Rudiger, H. Non-thermal DNA breakage by mobile-phone
radiation (1800MHz) in human fibroblasts and in transformed
GFSH-R17 rat granulosa cells in vitro. Mutat. Res. 2005,
583, 178–183. 67. Lai, H.; Singh, N.P. Singleand
double-strand DNA breaks in rat brain cells after acute
exposure to radiofrequency electromagnetic radiation. Int.
J. Radiat. Biol. 1996, 69, 513–521. 68. Malyapa, R.S.;
Ahern, E.W.; Straube, W.L.; Moros, E.G.; Pickard, W.F.;
Roti Roti, J.L. Measurement of DNA damage after exposure to
2450MHz electromagnetic radiation. Radiat. Res. 1997, 148,
608–617. 69. Cassel, J.C.; Cosquer, B.; Galani, R.; Kuster,
N. Whole-body exposure to 2.45GHz electromagnetic fields
does not alter radial-maze performance in rats. Behav.
Brain Res. 2004, 155, 37–43. 70. Cobb, B.L.; Jauchem, J.R.;
Adair, E.R. Radial arm maze performance of rats following
repeated low level microwave radiation exposure.
Bioelectromagnetics 2004, 25, 49–57. 71. Lai, H.; Horita,
A.; Guy, A.W. Microwave irradiation affects radial-arm maze
performance in the rat. Bioelectromagnetics 1994, 15,
95–104. 72. National Radiological Protection Board (NRPB).
Mobile phones and health 2004. Doc. NRPB 2004, 15 (5),
1–114. 73. Kues, H.A.; Monahan, J.C.; D’Anna, S.A.; McLeod,
D.S.; Lutty, G.A.; Koslov, S. Increased sensitivity of the
non-human primate eye to microwave radiation following
ophthalmic drug pretreatment. Bioelectromagnetics 1992, 13,
379–393. 74. Anderson, L.E.; Morris, J.E.; Sasser, L.B.;
Loscher, W. Effects of 50or 60-hertz, 100microT magnetic
field exposure in the DMBA mammary cancer model in
Sprague-Dawley rats: possible explanations for different
results from two laboratories. Environ. Health Perspect.
2002, 108, 797–802. 75. Fedrowitz, M.; Kamino, K.; Loscher,
W. Significant differences in the effects of magnetic field
exposure on 7,12-dimethylbenz(a)anthracene-induced mammary
carcinogenesis in two substrains of Sprague-Dawley rats.
Cancer Res. 2004, 64, 243–251. B
Biomaterials Immune Response

10. Lee, K.A.; Klasing, K.C. A role for immunology in


invasion biology. Trends Ecol. Evol. 2004, 19 (10),
523–529.

11. Pier, G.B.; Lyczak, J.B.; Wetzler, L.M. Immunology,


Infection, and Immunity; ASM Press: Washington, 2004.

12. Kao, W.J. Evaluation of protein-modulated macrophage


behavior on biomaterials: Designing biomimetic materials
for cellular engineering. Biomaterials 1999, 20 (23–24),
2213–2221.

13. Parham, P. The Immune System; Garland Science: New


York, USA, 2004.

14. Muno, D.; Kominami, E.; Mizuochi, T. Generation of both


MHC class Iand class II-restricted antigenic peptides from
exogenously added ovalbumin in murine phagosomes. FEBS
Lett. 2000, 478 (1–2), 178–182.

15. Doherty, T.M. T-cell regulation of macrophage function.


Curr. Opin. Immunol. 1995, 7 (3), 400–404.

16. Perry, V.H. A revised view of the central nervous


system microenvironment and major histocompatibility
complex class II antigen presentation. J. Neuroimmunol.
1998, 90 (2), 113–121.

17. Boes, M.; Ploegh, H.L. Translating cell biology in


vitro immunity in vivo. Nature 2004, 430 (8), 264–271.

18. King, C.A.; Wills, M.R. Immunology II: Acquired


immunity. Surgery 2005, 23 (9), 319–323.

19. Roy, C.R. Professional secrets. Nature 2003, 425 (25),


351–352.

20. Rodriguez, A.; Regnault, A.; Kleijmeer, M.;


RicciardiCastagnoli, P.; Amigorena, S. Selective transport
of internalized antigens to the cytosol for MHC class I
presentation in dendritic cells. Nat. Cell Biol. 1999, 1
(6), 362–368.

21. Sirica, A.E. Cellular and Molecular Pathogenesis;


Lippincott-Raven: Philadelphia, 1996.

22. Ignatowicz, L.; Kappler, J.; Marrack, P. The repertoire


of T cells shaped by a single MHC/peptide ligand. Cell
1996, 84 (4), 521–529.

23. Carmichael, A.; Wills, M. The immunology of infection.


Medicine 2005, 33 (3), 3–9.

24. Yewdell, J.W.; Bennink, J.R. The binary logic of


antigen processing and presentation to T cells. Cell 1990,
62 (2), 203–206.

25. Cone, R.E.; Malley, A. Soluble, antigen-specific T-cell


proteins: T-cell-based humoral immunity? Immunol. Today
1996, 17 (7), 318–322.

26. Bachmann, M.F.; Ohashi, P.S. The role of T-cell


receptor dimerization in T-cell activation. Rev. Immunol.
Today 1999, 20 (12), 568–575.

27. Patel, D.M.; Mannie, M.D. Intercellular exchange of


Class II major histocompatibility complex/peptide complexes
is a conserved process that requires activation of T cells
but is constitutive in other types of antigen presenting
cells. Cell. Immunol. 2001, 214 (2), 165–172.

28. Choudhuri, K.; Kearney, A.; Bakker, T.R.; Merwe,


P.A.V.D. Immunology: How do T cells recognize antigen?
Curr. Biol. 2005, 15 (10), R382–R385.

29. Guo, W.H.; Chan, K.L.; Dallman, M.; Tam, P.K.H. Role of
CD4+ and CD8+ T cells in early and late acute rejection of
small bowel allograft. J. Pediatr. Surg. 2001, 36 (2),
352–356. 30. Moalem, G.; Tracey, D.J. Immune and
inflammatory mechanisms in neuropathic pain. Brain Res. Rev.
2006, 51 (2), 240–264. 31. Robey, E.A. Immunology guide for
a cell-fate decision. Nature 2005, 433 (24), 813–814. 32.
Romagnani, S. The Th1/Th2 paradigm. Immunol. Today 1997, 18
(6), 263–266. 33. Frossi, B.; Carli, M.D.; Piemonte, M.;
Pucillo, C. Oxidative microenvironment exerts an opposite
regulatory effect on cytokine production by Th1 and Th2
cells. Mol. Immunol. 2008, 45 (1), 58–64. 34.
Furuzawa-Carbadella, J.; Vargas-Rojas, M.I.; Cabral, A.R.
Autoimmune inflammation from the Th17 perspective.
Autoimmun. Rev. 2007, 6 (3), 169–175. 35. Horiuchi, Z.;
Kawano, Y.; Yamasaki, K.; Minohara, M.; Furue, M.;
Taniwaki, T.; Miyazaki, T.; Kira, J.-I. Th1 dominance in
HAM/TSP and the optico-spinal form of multiple sclerosis
versus Th2 dominance in mite antigen-specific IgE myelitis.
J. Neurol Sci. 2000, 172 (1), 17–24. 36. Hofstetter, H.H.;
Ibrahim, S.M.; Koczan, D.; Kruse, N.; Weishaupt, A.; Toyka,
K.V.; Gold, R. Therapeutic efficacy of IL-17 neutralization
in murine experimental autoimmune encephalomyelitis. Cell.
Immunol. 2005, 237 (2), 123–130. 37. Schmidt-Weber, C.B.;
Akdis, M.; Akdis, C.A. TH17 cells in the big picture of
immunology. J. Allergy Clin. Immunol. 2007, 120 (2),
247–254. 38. Bettelli, E.; Oukka, M.; Kuchroo, V.K. TH-17
cells in the circle of immunity and autoimmunity. Nat.
Immunol. 2007, 8 (4), 345–350. 39. Wilson, N.J.; Bonidace,
K.; Chan, J.R.; McKenzie, B.S.; Blumenschein, W.M.;
Mattson, J.D.; Basham, B.; Smith, K.; Chen, T.; Morel, F.;
Lecron, J.-C.; Kastelein, R.A.; Cua, D.J.; McClanahan,
T.K.; Bowman, E.P.; Malefyt, R.D.W. Development, cytokine
profile and function of human interleukin 17producing helper
T cells. Nat. Immunol. 2007, 8 (9), 950–957. 40. Bettelli,
E.; Korn, T.; Kuchroo, V.K. Th17: The third member of the
effector T cell trilogy. Curr. Opin. Immunol. 2007, 19 (6),
652–657. 41. Aggarwal, S.; Gurney, A.L. IL-17: Prototype
member of an emerging cytokine family. J. Leukocyte Biol.
2002, 71 (1), 1–8. 42. Stockinger, B.; Veldhoen, M.
Differentiation and function of Th17 T cells. Curr. Opin.
Immunol. 2007, 19 (3), 281–286. 43. Belich, M.P.;
Trowsdale, J. Proteasome and class I antigen processing and
presentation. Mol. Biol. Rep. 1995, 21 (1), 53–56. 44.
Modo, M.; Mellodew, K.; Rezaie, P. In vitro expression of
major histocompatibility class I and class II antigens by
conditionally immortalized murine neural stem cells.
Neurosci. Lett. 2003, 337 (2), 85–88. 45. Brown, R.E.;
Roser, B.; Singh, P.B. Class I and Class II regions of the
major histocompatibility complex both contribute to
individual odors in congenic inbred strains of rats. Behav.
Genet. 1989, 19 (5), 659– 674. B 46. Buckley, R.H.
Transplantation immunology: Organ and bone marrow. J.
Allergy Clin. Immunol. 2003, 11 (2), S733–S744. 47.
Maiorana, A.; Cesinaro, A.M.; Fano, R.A.; Collina, G.
Expression of MHC class I and class II antigens in primary
breast carcinomas and synchronous nodal metastases. Clin.
Exp. Metast. 1995, 12 (1), 43–48. 48. Peters, V.B.;
Sperber, K.E. The effect of viruses on the ability to
present antigens via the major histocompatibility complex.
Microbes Infect. 1999, 1 (4), 335–345. 49. Scha¨ffer, M.;
Bongartz, M.; Hoffmann, W.; Viebahn, R.
MHC-Class-II-deficiency impairs wound healing. J. Surg. Res.
2007, 138 (1), 100–105. 50. Maffei, A.; Harris, P.E.
Peptides Bound to major histocompatibility complex
molecules. Peptides 1998, 19 (1), 179–198. 51. Moss, D.J.;
Khanna, R. Major histocompatibility complex: From genes to
function. Immunol. Today 1999, 20 (4), 165–167. 52.
Germain, R.N. MHC-dependent antigen processing and peptide
presentation: Providing ligands for T lymphocyte
activation. Cell 1994, 76 (2), 287–299. 53. Kittur, D.S.;
Wilasrusmee, C.; Han, W.-F.; Xu, R.; Burdick, J.F.; Adler,
W. Locally derived cytokines and upregulation of MHC Class
II genes in allografts. J. Heart Lung Transplant. 2002, 21
(8), 882–889. 54. Ting, J.P.-Y.; Trowsdale, J. Genetic
control of MHC Class II expression. Cell 2002, 109 (2),
S21–S33. 55. Williams, D.B.; Vassilakos, A.; Suh, W.-K.
Peptide presentation by MHC class I molecules. Trends in
Cell Biol. 1996, 6 (7), 267–273. 56. Faulkner, L.;
Borysiewicz, L.K.; Man, S. The use of human leucocyte
antigen class I transgenic mice to investigate human immune
function. J. Immunol. Methods 1998, 221 (1–2), 1–16. 57.
Toumbis, C.A.; Kronick, J.L.; Wooley, P.H.; Nasser, S.
Total joint arthroplasty and the immune response. Semin.
Arthritis Rheum. 1997, 127 (1), 44–47. 58. Case, C.P.;
Langkamer, V.G.; James, C.; Palmer, M.R.; Kemp, A.J.; Heap,
P.F.; Solomon, L. Widespread dissemination of metal debris
from implants. Journal of Bone and Joint Surgery [Br] 1994,
76B (5), 701–712. 59. DiCarlo, E.F.; Bullough, P.G. The
biologic responses to orthopaedic implants and their wear
debris. Clinical Materials 1992, 9 (3–4), 235–260. 60.
Arora, A.; Song, Y.; Chun, L.; Huie, P.; Trindade, M.;
Smith, R.L.; Goodman, S. The role of the TH1 and TH2 immune
responses in loosening and osteolysis of cemented total hip
replacements. Journal of Biomedical Materials Research
2003, 64A (4), 693–697. 61. Goodman, S.B. Wear particles,
periprosthetic osteolysis and the immune system.
Biomaterials 2007, 28 (34), 5044–5048. 62. Hallab, N.J.;
Mikecz, K.; Vermes, C.; Skipor, A.; Jacobs, J.J.
Differential lymphocyte reactivity to serum-derived
metal-protein complexes produced from cobalt-based and
titanium-based implant alloy degradation. Journal of
Biomedical Materials Research 2001, 56 (3), 427–436. 63.
Hercus, B.; Revell, P. Phenotypic characteristics of T
lymphocytes in the interfacial tissue of aseptically
loosened prosthetic joints. Journal of Materials Science:
Materials in Medicine 2001, 12 (10–12), 1063– 1067. 64.
Lalor, P.; Revell, P. T lymphocytes and titanium aluminium
vanadium (TiAlV) alloy: evidence for immunological events
associated with debris deposition. Clinical Materials 1993,
12 (1), 57–62. 65. Wooley, P.H.; Nasser, S.; Fitzgerald,
R.H. The Immune Response to Implant Materials in Humans.
Clinical Orthopaedics and Related Research 1996, 32 6 (3),
63–70. 66. Santavirta, S.; Konttinen, Y.; Bergroth, V.;
Gro¨nblad, M. Lack of immune response to methyl
methacrylate in lymphocyte cultures. Acta Orthopaedica
Scandinavica 1991, 62 (1), 29–32. 67. Doorn, P.F.;
Campbell, P.A.; Worrall, J.; Benya, P.D.; McKellop, H.A.;
Amstutz, H.C. Metal wear particle characterization from
metal on metal total hip replacements: transmission
electron microscopy study of periprosthetic tissues and
isolated particles. Journal of Biomedical Materials
Research 1998, 42 (1), 1998. 68. Haddad, F.S.; Cobb, A.G.;
Bentley, G.; Levell, N.J.; Dowd, P.M. Hypersensitivity in
aseptic loosening of total hip replacements. The role of
constituents of bone cement. The Journal of Bone and Joint
Surgery [BR] 1996, 78B (4), 546–549. 69. Methe, H.;
Edelman, E.R. Tissue Engineering of Endothelial Cells and
the Immune Response. Transplantation Proceedings 2006, 38
(10), 3293–3299. 70. Granchi, D.; Ciapetti, G.; Savarino,
L.; Stea, S.; Filippini, F.; Sudanese, A.; Rotini, R.;
Giunti, A. Expression of the CD69 activation antigen on
lymphocytes of patients with hip prosthesis. Biomaterials
2000, 21 (20), 2059–2065. 71. Trindade, M.C.D.; Lind, M.;
Sun, D.; Schurman, D.J.; Goodman, S.B.; Smith, R.L. In
vitro reaction to orthopaedic biomaterials by macrophages
and lymphocytes isolated from patients undergoing revision
surgery. Biomaterials 2001, 22 (3), 253–259. 72. Baldwin,
L.; Flanagan, B.F.; McLaughlin, P.J.; Parkinson, R.W.;
Hunt, J.A.; Williams, D.F. A study of tissue interface
membranes from revision accord knee arthroplasty: the role
of T lymphocytes. Biomaterials 2002, 23 (14), 3007–3014.
73. Voggenreiter, G.; Leiting, S.; Brauer, H.; Leiting, P.;
Majetschak, M.; Bardenheuer, M.; Obertacke, U.
Immuno-inflammatory tissue reaction to stainless-steel and
titanium plates used for internal fixation of long bones.
Biomaterials 2003, 24 (2), 247–254. 74. Valle´s, G.;
Gonza´lez-Melendi, P.; Gonza´lez-Carrasco, J.L.; Saldanˇa,
L.; Sa´nchez-Sabate´, E.; Munuera, L.; Vilaboa, N.
Differential inflammatory macrophage response to rutile and
titanium particles. Biomaterials 2006, 27 (30), 5199–5211.
75. Luyn, M.J.A.v.; Plantinga, J.A.; Brouwer, L.A.; Khouw,
I.M.S.L.; Leij, L.F.M.H.d.; Wachem, P.B.v. Repetitive
subcutaneous implantation of different types of
(biodegradable) biomaterials alters the foreign body
reaction. Biomaterials 2001, 22 (11), 1385–1391.
Biomaterials: Protein–Surface Interactions

1. Brandon, C.; Tooze, J. The building blocks. In


Introduction to Protein Structure, 2nd Ed.; Garland
Publishing: New York, 1999; 3–12.

2. Voet, D.; Voet, J.G.; Pratt, C.W. Fundamentals of


Biochemistry; John Wiley & Sons Inc.: New York, 2002. 3.
Norde, W.; Haynes, C.A. Reversibility and the mechanism of
protein adsorption. In Proteins at Interfaces II.
Fundamentals and Applications; Horbett, T.A., Brash, J.L.,
Eds.; American Chemical Society: Washington, DC, 1995;
26–40. 4. Wertz, C.F.; Santore, M.M. Fibrinogen adsorption
on hydrophilic and hydrophobic surfaces: geometrical and
energetic aspects of interfacial relaxations. Langmuir
2002, 18, 706–715. 5. Adamson, A.W. The solid–liquid
interface—adsorption from solution. In Physical Chemistry
of Surfaces, 5th Ed.; JohnWiley & Sons: New York, 1990;
Chap. XI, 421–435. 6. Latour, R.A.; Rini, C.J. Theoretical
analysis of adsorption thermodynamics for hydrophobic
peptide residues on SAM surfaces of varying functionality.
J. Biomed. Mater. Res. 2002, 60, 564–577. 7. Wertz, C.F.;
Santore, M.M. Effect of surface hydrophobicity on
adsorption and relaxation kinetics of albumin and
fibrinogen: single-species and competitive behavior.
Langmuir 2001, 17, 3006–3016. 8. Talbot, J.; Tarjus, G.;
Van Tassel, P.R.; Viot, P. From car parking to protein
adsorption: an overview of sequential adsorption processes.
Colloids Surf. A. 2000, 165, 287–324. 9. Malmsten, M.
Formation of adsorbed protein layers. J. Colloid Interface
Sci. 1998, 207, 186–199. 10. Wertz, C.F.; Santore, M.M.
Adsorption and relaxation kinetics of albumin and fibrinogen
on hydrophobic surface: single-species and competitive
behavior. Langmuir 1999, 15, 8884–8894. 11. Haynes, C.A.;
Norde, W. Globular proteins at solid/ liquid interfaces.
Colloids Surf. B 1994, 2, 517–566. 12. Nakanishi, K.;
Sakiyama, T.; Imamura, K. On the adsorption of proteins on
solid surfaces, a common but very complicated phenomenon.
J. Biosci. Bioeng. 2001, 91, 233–244. 13. Agnihotri, A.;
Siedlecki, C.A. Time-dependent conformational changes in
fibrinogen measured by atomic force microscopy. Langmuir
2004, 20, 8846–8852. 14. Fournier, R.L. Solute transport in
biological systems. In Basic Transport Phenomena in
Biomedical Engineering; Taylor & Francis: Philadelphia, PA,
1999; 27–28. 15. Dee, K.C.; Puleo, D.A.; Bizios, R.
Protein–surface interactions. In Tissue-Biomaterials
Interactions; John Wiley & Sons Inc.: Hoboken, NJ, 2002;
Chap. 3, 45–49. 16. Slack, S.M.; Horbett, T.A. The Vroman
effect: a critical review. In Proteins at Interfaces II.
Fundamentals and Applications; Horbett, T.A., Brash, J.L.,
Eds.; American Chemical Society: Washington, DC, 1995;
112–128. 17. Vroman, L.; Adams, A.L. Findings with the
recording ellipsometer suggesting rapid exchange of specific
plasma proteins at liquid/solid interfaces. Surf. Sci.
1969, 16, 438–446. 18. Turbill, P.; Beugeling, T.; Poot,
A.A. Competitive adsorption of proteins during exposure of
human blood plasma to glass and polyethylene. In Proteins
at Interfaces II. Fundamentals and Applications; Horbett,
T.A., Brash, J.L., Eds.; American Chemical Society:
Washington, DC, 1995; 150–162. 19. Warkentin, P.H.;
Lundstrom, I.; Tengvall, P. Protein– protein interactions
affecting proteins at surfaces. In Proteins at Interfaces
II. Fundamentals and Applications; Horbett, T.A., Brash,
J.L., Eds.; American Chemical Society: Washington, DC,
1995; 163–180. 20. Jung, S.-Y.; Lim, S.-M.; Albertorio, F.;
Kim, G.; Gurau, M.C.; Yang, R.D.; Holden, M.A.; Cremer,
P.S. The Vroman effect: a molecular level description of
fibrinogen displacement. J. Am. Chem. Soc. 2003, 125,
12782–12786. 21. Lee, W.-K.; McGuire, J.; Bothwell, M.K.
Competitive adsorption of bacteriophage T4 lysozyme
stability variants at hydrophilic glass surfaces. J.
Colloid Interface Sci. 2004, 269, 251–254. 22. Morra, M. On
the molecular basis of fouling resistance. J. Biomater.
Sci. Polym. Ed. 2000, 11, 547–569. 23. Kjellander, R.;
Florin, E. Water structure and changes in thermal stability
of the system poly(ethylene oxide)water. J. Chem. Soc. Far.
Trans. 1981, 1, 2053–2077. 24. Prime, K.L.; Whitesides,
G.M. Adsorption of proteins onto surfaces containing
end-attached oligo(ethylene oxide): a model system using
self-assembled monolayers. J. Am. Chem. Soc. 1993, 115,
10714–10721. 25. Li, L.; Chen, S.; Zheng, J.; Ratner, B.D.;
Jiang, S. Protein adsorption on oligo(ethylene
glycol)-terminated alkanethiolate self-assembled
monolayers: the molecular basis of nonfouling behavior. J.
Phys. Chem. B. 2005, 109, 2934–2941. 26. Herrwerth, S.;
Eck, W.; Reinhardt, S.; Grunze, M. Factors that determine
the protein resistance of oligoether self-assembled
monolayers—internal hydrophilicity, terminal
hydrophilicity, and lateral packing density. J. Am. Chem.
Soc. 2003, 125, 9359–9366. 27. Ostuni, E.; Chapman, R.G.;
Holmlin, R.E.; Takayama, S.; Whitesides, G.M. A survey of
structure–property relationships of surfaces that resist
the adsorption of protein. Langmuir 2001, 17, 5605–5620.
28. Chapman, R.G.; Ostuni, E.; Yan, L.; Whitesides, G.M.
Preparation of mixed self-assembled monolayers (SAMs) that
resist adsorption of proteins using the reaction of amines
with a SAM that presents interchain carboxylic anhydride
groups. Langmuir 2000, 16, 6927–6936. 29. Ulman, A.;
Eilers, J.E.; Tillman, N. Packing and molecular orientation
of alkanethiol monolayers on gold surfaces. Langmuir 1989,
5, 1147–1152. 30. Ishihara, K.; Iwasaki, Y. Reduced protein
adsorption on novel phospholipid polymers. J. Biomater.
Appl. 1998, 13, 111–127. 31. Feng, W.; Brash, J.; Zhu, S.
Atom-transfer radical grafting polymerization of
2-methacryloyloxyethyl phosphorylcholine from silicon wafer
surfaces. J. Polym. Sci. Pt. A: Polym. Chem. 2004, 42,
2931–2942. 32. Shaw, W.J.; Long, J.R.; Campbell, A.A.;
Stayton, P.S.; Drobny, G.P. A solid-state NMR study of
dynamics in a hydrated salivary peptide adsorbed to
hydroxyapatite. J. Am. Chem. Soc. 2000, 122, 7118–7119. 33.
Bower, P.V.; Louie, E.A.; Long, J.R.; Stayton, P.S.;
Drobny, G.P. Solid-state NMR structural studies of peptides
immobilized on gold nanoparticles. Langmuir 2005, 21,
3002–3007. 34. Vermeer, A.W.P.; Norde, W. CD spectroscopy
of proteins adsorbed at flat hydrophilic quartz and
hydrophobic Teflon surfaces. J. Colloid Interface Sci. 2000,
225, 394–397. 35. Engel, M.F.M.; van Mierlo, C.P.M.;
Visser, A.J.W.G. Kinetic and structural characterization of
adsorptioninduced unfolding of bovine a-lactalbumin. J.
Biol. Chem. 2002, 277, 10,922–10,930. 36. Hylton, D.M.;
Shalaby, S.W.; Latour, R.A. Direct correlation between
adsorption-induced changes in protein structure and
platelet adhesion. J. Biomed. Mater. Res. 2005, 73A,
349–358. 37. Lundqvist, M.; Sethson, I.; Jonsson, B.-H.
Protein adsorption onto silica nanoparticles:
conformational changes depend on the particles’ curvature
and the protein stabilitiy. Langmuir 2004, 20, 10639–10647.
38. Engel, M.F.M.; Visser, A.J.W.G.; van Mierlo, C.P.M.
Conformation and orientation of a protein folding
intermediate trapped by adsorption. Proc. Natl. Acad. Sci.
U.S.A. 2004, 101, 11,316–11,321. 39. Tidwell, C.D.;
Castner, D.G.; Golledge, S.L.; Ratner, B.D.; Meyer, K.;
Hagenhoff, B.; Benninghoven, A. Static time-of-flight
secondary ion mass spectrometry and x-ray photoelectron
spectroscopy characterization of adsorbed albumin and
fibronectin films. Surf. Interface Anal. 2001, 31, 724–733.
40. Wagner, M.S.; Castner, D.G. Analysis of adsorbed
proteins by static time-of-flight secondary ion mass
spectrometry. Appl. Surf. Sci. 2004, 231–232, 366–376. 41.
Wagner, M.S.; Graham, D.J.; Ratner, B.D.; Castner, D.G.
Maximizing information obtained from secondary ion mass
spectra of organic thin films using multivariate analysis.
Surf. Sci. 2004, 570, 78–97. 42. Matthews, B.W. Studies on
protein stability with T4 lysozyme. Adv. Prot. Chem. 1995,
46, 249–278. 43. Bower, C.K.; Xu, Q.; McGuire, J. Activity
losses among T4 lysozyme variants after adsorption to
colloidal silica. Biotechnol. Bioeng. 1998, 58, 658–662.
44. Michael, K.E.; Keselowsky, B.G.; Vernekar, V.N.;
Meredith, C.A.; Latour, R.A.; Garcia, A.J.
Adsorptioninduced conformational changes in fibronectin due
to interactions with well-defined surface chemistries.
Langmuir 2003, 19, 8033–8040. 45. Keselowsky, B.G.;
Collard, D.M.; Garcia, A.J. Surface chemistry modulates
fibronectin conformation and directs integrin binding and
specificity to control cell adhesion. J. Biomed. Mater. Res.
2003, 66A, 247–259. 46. Hu, W.J.; Eaton, J.W.; Tang, L.
Molecular basis of biomaterial-mediated foreign body
reactions. Blood 2001, 98, 1231–1238. 47.
http://www.pdb.org/. 48. Latour, R.A. Molecular modeling of
biomaterial surfaces. Curr. Opin. Solid State Mater. Sci.
1999, 4, 413–417. 49. Agashe, M.; Raut, V.; Stuart, S.J.;
Latour, R.A. Molecular simulation to characterize the
adsorption behavior of a fibrinogen gamma-chain fragment.
Langmuir 2005, 21, 1103–1117. 50. Sun, Y.; Latour, R.A.
Prediction of the orientations of adsorbed protein using an
empirical energy function with implicit solvation. Langmuir
2005, 21, 5616–5626. 51. Zheng, J.; Li, L.; Chen, S.;
Jiang, S. Molecular simulation study of water interactions
with oligo (ethylene B glycol)-terminated alkanethiol
self-assembled monolayers. Langmuir 2004, 20, 8931–8938.

52. Zhou, J.; Zheng, J.; Jiang, S.Y. Molecular simulation


studies of the orientation and conformation of cytochrome c
adsorbed on self-assembled monolayers. J. Phys. Chem. B.
2004, 108, 17,418–17,424.

53. Kreuzer, H.J.; Wang, R.L.C.; Grunze, M. Effect of


stretching on the molecular conformation of oligo (ethylene
oxide): a theoretical study. New J. Phys. 1999, 1,
21.1–21.16.

54. Wang, R.L.C.; Kreuzer, H.J.; Grunze, M. The interaction


of oligo(ethylene oxide) with water: a quantum mechanical
study. Phys. Chem. Chem. Phys. 2000, 2, 3613–3622.

55. Castner, D.G.; Ratner, B.D. Biomedical surface science:


foundations to frontiers. Surf. Sci. 2002, 500, 28–60.

56. Ratner, B.D.; Bryant, S.J. Biomaterials: where we have


been and where we are going. Annu. Rev. Biomed. Eng. 2004,
6, 41–75.

FURTHER READINGS

Brash, J.L.; Horbett, T.A., Eds. Proteins at Interfaces.


Physiochemical and Biochemical Studies; American Chemical
Society: Washington, DC, 1987. Brash, J.L.; Wojciechowski,
P.W., Eds. Interfacial Phenomena and Bioproducts; Marcel
Dekker, Inc.: New York, 1996. Dee, K.C.; Puleo, D.A.;
Bizios, R. Protein–surface interactions. In
Tissue–Biomaterial Interactions; John Wiley & Sons:
Hoboken, NJ, 1999; 37–52. Gray, J.J. The interaction of
proteins with solid surfaces. Curr. Opin. Struct. Biol.
2004, 14, 110–115. Hlady, V.; Buijs, J. Protein adsorption
on solid surfaces. Curr. Opin. Biotechnol. 1996, 7, 72–77.
Hoffman, A.S. Non-fouling surface technologies. J.
Biomater. Sci. Polym. Ed. 1999, 10, 1011–1014. Horbett,
T.A. The role of adsorbed proteins in tissue response to
biomaterials. In Biomaterials Science— An Introduction to
Materials in Medicine, 2nd Ed.; Ratner, B.D., Hoffman,
A.S., Schoen, F.J., Lemons, J.E., Eds.; Elsevier Academic
Press: New York, 2004; 237–246. Horbett, T.A.; Brash, J.L.,
Eds. Proteins at Interfaces II. Fundamentals and
Applications; American Chemical Society: Washington, DC,
1995. Malmsten, M. Biopolymers at Interfaces; Marcel
Dekker, Inc.: New York, 1998. Ramsden, J.J. Puzzles and
paradoxes in protein adsorption. Chem. Soc. Rev. 1995, 24,
73–78.
Biomedical Ethics

1. http://www.npr.org/programs/morning/features/2002/
jul/tuskegee/index.html (accessed June 2005).

2. New York Times. 26 July 1972, 1, 8.

3. http://www.tuskegee.edu/Global/Story.asp?s=1207598
(accessed June 2005).

4. http://www.cdc.gov/nchstp/od/tuskegee/time.htm (accessed
June 2005). 5.
http://webk.ask.com/redir?u=http%3a%2f%2fwww.kn.
pacbell.com%2fwired%2fBHM%2ftuskegee_quest.html (accessed
June 2005). 6.
Wysiwyg//24http://www.britannica.com/magazine/
print?/ebsco-id=11382 (accessed April 2001). 7. Jones, J.
Bad Blood: The Tuskegee Syphilis Experiment: A Tragedy of
Race and Medicine; The Free Press: New York, 1981. 8.
Pellegrino, E. The Nazi doctors and Nuremberg: some moral
lessons revisited. Ann. Intern. Med. 1997, 127 (4),
307–308. 9. http://www.cdc.gov/od/ads/ethcodes/belm-eng.pdf
(accessed June 5, 2005). 10.
http://www.georgetown.edu/research/nrcbl/nrc/
archives/ncphsguide.pdf (accessed June 2005). 11.
Guidelines for the Conduct of Research Involving Human
Subjects at the National Institutes of Health; 00-4783;
United States Department of Health and Human Services,
Public Health Services, National Institutes of Health:
Washington, DC, 2004; 1–24. 12. Beauchamp, T.L.; Childress,
J.F. Principles of Biomedical Ethics, 2nd Ed.; Oxford
University Press: New York, 1983. 13. Hart, H.L.A. The
Concept of Law; Oxford University Press: New York, 1961.
14. Gert, B. The Moral Rules; Harper and Row: New York,
1973. 15. Frankena, W.K. Ethics, 2nd Ed.; Prentice Hall:
Englewood Cliffs, New Jersey, 1973. 16.
http://www.indiana.edu/Bancmed/oath.htm (accessed June
2005). B
Biomimetic Materials

1. Bruice, T.C.; Benkovic, S.J. Bioorganic Mechanisms;


Breslow, R., Karplus, M., Eds.; W.A. Benjamin Inc.: New
York, 1966; Vol. 2.

2. Jencks, W.P. Catalysis in Chemistry and Enzymology;


McGraw-Hill: New York, 1969.

3. Bender, M.L. Mechanisms of Homogeneous Catalysis from


Protons to Proteins; Wiley-Interscience: New York, 1971.

4. Fendler, J.H.; Fendler, E.J. Catalysis in Micellar and


Macromolecular Systems; Academic Press: New York, 1975.

5. Breslow, R.; Dong, S.D. Biomimetic reactions catalyzed


by cyclodextrins and their derivatives. Chem. Rev. 1998, 98
(5), 1997–2011.

6. Murakami, Y.; Kikuchi, J.; Hisaeda, Y.; Hayashida, O.


Artificial enzymes. Chem. Rev. 1996, 96 (2), 721–758.

7. Lehn, J.-M. Supramolecular chemistry—Scope and


perspectives. Molecules, supramolecules, and molecular
devices. Angew. Chem., Int. Ed. Engl. 1988, 27 (1), 89–112.

8. Calixarenes: A Versatile Class of Macrocyclic Compounds;


Vecens, J., Bo¨hmer, V., Eds.; Kluwer Academic Publishers:
Dordrecht, 1991.

9. Kondo, H. Towards Design of Artificial Enzymes and


Receptors Through Site-Directed Mutagenesis of Proteins. In
Comprehensive Supramolecular Chemistry; Atwood, J.L.,
Davies, J.E.D., MacNicol, D.D., Vo¨gtle, F., Lehn, J.-M.,
Murakami, Y., Eds.; Pergamon: Oxford, 1996; Vol. 4,
528–547.

10. Wang, L.; Schultz, P.G. Expanding the genetic code.


Chem. Commun. 2002, (1), 1–11.

11. Feiters, M.C. Supramolecular Catalysis. In


Comprehensive Supramolecular Chemistry; Atwood, J.L.,
Davies, J.E.D., MacNicol, D.D., Vo¨gtle, F., Lehn, J.-M.,
Reinhoudt, D.N., Eds.; Pergamon: Oxford, 1996; Vol. 10,
268–360.

12. Qi, D.; Tann, C.-M.; Haring, D.; Distefano, M.D.


Generation of new enzymes via covalent modification of
existing proteins. Chem. Rev. 2001, 101 (10), 3081–3111.
13. Hayashi, T.; Hisaeda, Y. New functionalization of
myoglobin by chemical modification of heme-propionates. Acc.
Chem. Res. 2002, 35 (1), 35–43.

14. Wulff, G. Enzyme-like catalysis by molecularly


imprinted polymers. Chem. Rev. 2002, 102 (1), 1–27.

15. Astruc, D.; Chardac, F. Dendritic catalysts and


dendrimers in catalysis. Chem. Rev. 2001, 101 (9),
2991–3023. 16. Kunitake, T. Synthetic Bilayer Membranes:
Molecular Design and Molecular Organization. In
Comprehensive Supramolecular Chemistry; Atwood, J.L.,
Davies, J.E.D., MacNicol, D.D., Vo¨gtle, F., Lehn, J.-M.,
Sauvage, J.-P., Hosseini, M.W., Eds.; Pergamon: Oxford,
1996; Vol. 9, 351–406. 17. Ariga, K.; Kunitake, T.
Molecular recognition at airwater and related interfaces:
Complementary hydrogen bonding and multisite interaction.
Acc. Chem. Res. 1998, 31 (6), 371–378. 18. Ulman, A.
Formation and structure of self-assembled monolayers. Chem.
Rev. 1996, 96 (4), 1533–1554. 19. Sackmann, E. Supported
membranes: Scientific and practical applications. Science
1996, 271 (5245), 43–48. 20. Murakami, Y.; Kikuchi, J.
Supramolecular Assemblies Formed with Synthetic Peptide
Lipids. Functional Models of Biomembranes and Enzymes. In
Bioorganic Chemistry Frontiers; Dugas, H., Ed.;
Springer-Verlag: Berlin, 1991; Vol. 2, 73–113. 21. Mueller,
A.; O’Brein, D.F. Supramolecular materials via
polymerization of mesophases of hydrated amphiphiles. Chem.
Rev. 2002, 102 (3), 727–757. 22. Kikuchi, J.; Ariga, K.;
Sasaki, Y. Molecular Recognition and Functional Connection
in Lipid Membranes. In Advances in Supramolecular
Chemistry; Gokel, G.W., Ed.; Cerberus Press: South Miami,
2001; 131–173. 23. Nielsen, P.E. Peptide nucleic acid. A
molecule with two identities. Acc. Chem. Res. 1999, 32 (7),
624–630. 24. Kool, E.T. Replacing the nucleobases in DNA
with designer molecules. Acc. Chem. Res. 2002, 35 (11),
936–943. 25. Wang, Q.; Dordick, J.S.; Linhardt, R.J.
Synthesis and application of carbohydrate-containing
polymers. Chem. Mater. 2002, 14 (8), 3232–3244. 26.
Lundquist, J.J.; Toone, E.J. The cluster glycoside effect.
Chem. Rev. 2002, 102 (2), 555–578. 27. Akiyoshi, K.;
Sasaki, Y.; Kuroda, K.; Ueminami, A.; Sunamoto, J.
Controlled Association of Macromolecules in Water:
Hydrophobized Polymers and Proteins. In Molecular
Interactions and Time-Space Organization in Macromolecular
Systems; Morishima, Y., Norisue, T., Tashiro, K., Eds.;
Springer-Verlag: Berlin, 1999; 151–158. 28. Ringsdorf, H.;
Schlarb, B.; Venzmer, J. Molecular architecture and
function of polymeric oriented systems: Models for the
study of organization, surface recognition, and dynamics of
biomembranes. Angew. Chem., Int. Ed. Engl. 1988, 27 (1),
113–158. 29. Decher, G. Layered Nanoarchitectures Via
Directed Assembly of Anionic and Cationic Molecules. In
Comprehensive Supramolecular Chemistry; Atwood, J.L.,
Davies, J.E.D., MacNicol, D.D., Vo¨gtle, F., Lehn, J.-M.,
Sauvage, J.-P., Hosseini, M.W., Eds.; Pergamon: Oxford,
1996; Vol. 9, 507–528.
Biopotential Amplifiers

11. Nagel, J.H. Biopotential amplifiers. In The Biomedical


Engineering Handbook; Bronzino, J.D., Ed.; CRC Press: Boca
Raton, FL, 1995.

12. Ott, H.W. Noise Reduction Techniques in Electronic


Systems; Wiley: New York, 1988. 13. Pallas-Areny, R.;
Webster, J.G. Sensors and Signal Conditioning; John Wiley &
Sons: New York, 1991. 14. Kimmel, W.D.; Gerke, D.D.
Electromagnetic Compatibility in Medical Equipment; IEEE
Press: New York, 1995. B
Bioreactors

41. Ingber, D.E. Mehanical and Chemical Determinants of


Tissue Development. In Principles of Tissue Engineering;
Lanza, R., Langer, R., Vacanti, J., Eds.; Academic Press:
San Diego, 2002; 101–109. Chapter 9.

42. Vorp, D.A.; Severyn, D.A.; Steed, D.L.; Webster, M.W. A


device for the application of cyclic twist and extension on
perfused vascular segments. Am. J. Physiol. 1996, 270, H
787–H795. (Heart Circ. Phisiol. 39).

43. Banes, A.J.; Link, G.W., Jr; Gilbert, J.W.; Tran Son
Tay, R.; Monbureau, O. Culturing cells in a mechanically
active environment: The Flexercell Strain Unit can apply
cyclic or static tension or compression to cells in
culture. Am. Biotechnol. Lab. May 1990, 8 (7), 12–22.

44. Kasra, M.; Goel, V.; Martin, J.; Wang, S.-T.; Choi, W.;
Buckwalter, J. Effect of Dynamic Hydrostatic Pressure on
Rabbit Intervertebral Disc Cells. In Journal of Orthopedic
Research; Elsevier, 8 February 2003. DTD 4.3.1 8 pp.

45. Vilender, K. Bioreactor with Plurality of Chambers for


Conditioning Intravascular Tissue Engineered Medical
Products. Patent Pending: US Patent Application Serial
Number 10/371175, November 27, 2002.

46. MCAllister; Todd, N.; L’Heurex, N. Cytograph Tissue


Engineering, Inc., La Jolla CA (US). Improved Tissue
Engineered Blood Vessels and Methods and Apparatus for
their Manufacture; World Intellectual Property
Organization. WO 01/37884 A2 published May 31, 2001. 47.
Dimitrijevich, D.S.; Dodd, C.W.; Anderson, W.J.; Schwarz,
R.P. Two Chamber Cell Culture Vessel; Word Intellectual
Property Organization. WO 02/38735 A2, published May 16,
2002. FURTHER READING Huard, J.; Li, Y.; Peng, H.R.; Fu,
F.H. Gene therapy and tissue engineering for sports
medicine. J. Gene Med. Feb 2003, 5 (2), 93–108. Kuriwaka,
M.; Ochi, M.; Uchio, Y.; Maniwa, S.; Adachi, N.; Mori, R.;
Kawasaki, K.; Kataoka, H. Optimum combination of monolayer
and three-dimensional cultures for cartilage-like tissue
engineering. Tissue Eng. Feb 2003, 9 (1), 41–49. Sah, R.;
Kim, Y.; Doong, JY.; Grodzinsky, A.J.; Plass, A.H.; Sandy,
J.D. BioSynthetic response of cartilage explants to dynamic
compression. J. Orthop. Res. 1989, 7, 619–636. Toyoda, T.;
Matsumoto, H.; Fujikwas, K.; Saito, S.; Inoue, K. Tensile
load and the metabolism of anterior cruciate ligament
cells. Clin. Orthop. Relat. Res. 1998, 353, 247–255. Woo,
SL-Y.; Young, E.P.; Kwan, M.K. Fundamental Studies in Knee
Ligament Mechanics. In Knee Ligaments: Structure, Function,
Injury, and Repair; Daniel, D.M., Akeson, W.H., O’Connor,
Eds.; Raven Press: New York, NY, 1990; 115–134.
Biorubber/Poly(Glycerol Sebacate)

1. Peppas, N.A.; Langer, R. New challenges in biomaterials.


Science 1994, 263 (5154), 1715–1720.

2. Langer, R. Biomaterials: Status, Challenges, and


Perspectives. AIChE J. 2000, 46 (7), 1286–1289.

3. Hench, L.L.; Polak, J.M. Third-generation biomedical


materials. Science 2002, 295, 1014–1007.

4. Langer, R. Drug delivery and targeting. Nature 1998, 392


(6679 Suppl.), 5–10.

5. Liu, G.; Hinch, B.; Beavis, A.D. Mechanisms for the


transport of a,o-dicarboxylates through the mitochondrial
inner membrane. J. Biol. Chem. 1996, 271 (41), 25338–25344.

6. Grego, A.V.; Mingrone, G. Dicarboxylic acids, an


alternate fuel substrate in parenteral nutrition: An
update. Clin. Nutr. 1995, 14 (3), 143–148.

7. Tamada, J.; Langer, R. The development of polyanhydrides


for drug delivery applications. J. Biomater. Sci., Polym.
Ed. 1992, 3 (4), 315–353.

8. Nagata, M.; Machida, T.; Sakai, W.; Tsutsumi, N.


Synthesis, characterization, and enzymatic degradation of
network aliphatic copolyesters. J. Polym. Sci., A, Polym.
Chem. 1999, 37, 2005–2011.

9. Dupont-Gillain, C.C.; Nysten, B.; Rouxhet, P.G. Collagen


adsorption on poly(methyl methacrylate): Netlike structure
formation upon drying. Polym. Int. 1999, 48 (4), 271–276.

10. Sperling, L.H. Introduction to Physical Polymer


Science; John Wiley & Sons: New York, 1992.

11. Yamaguchi, S. Analysis of stress-strain curves at fast


and slow velocities of loading in vitro in the transverse
section of the rat incisor periodontal ligament following
the administration of beta-aminopropionitrile. Arch. Oral
Biol. 1992, 37 (6), 439–444.

12. Komatsu, K.; Chiba, M. The effect of velocity of


loading on the biomechanical responses of the periodontal
ligament in transverse sections of the rat molar in vitro.
Arch. Oral Biol. 1993, 38 (5), 369–375.

13. Chiba, M.; Komatsu, K. Mechanical responses of the


periodontal ligament in the transverse section of the rat
mandibular incisor at various velocities of loading in
vitro. J. Biomech. 1993, 26 (4–5), 561–570.

14. Nagdi, K. Rubber as an Engineering Material: Guideline


for Users; Hanser: Munich, 1993. 15. Fratzl, P.; Misof, K.;
Zizak, I.; Rapp, G.; Amenitsch, H.; Bernstorff, S.
Fibrillar structure and mechanical properties of collagen.
J. Struct. Biol. 1998, 122 (1–2), 119–122. 16. Misof, K.;
Rapp, G.; Fratzl, P. A new molecular model for collagen
elasticity based on synchrotron x-ray scattering evidence.
Biophys. J. 1997, 72 (3), 1376–1381. 17. Wang, J.L.;
Parnianpour, M.; Shirazi-Adl, A.; Engin, A.E. Failure
criterion of collagen fiber: Viscoelastic behavior simulated
by using load control data. Theor. Appl. Fract. Mech. 1997,
27 (1), 1–12. 18. Lee, M.C.; Haut, R.C. Strain rate effects
on tensile failure properties of the common carotid artery
and jugular veins of ferrets. J. Biomech. 1992, 25 (8),
925–927. 19. Haut, R.C. The effect of a lathyritic diet on
the sensitivity of tendon to strain rate. J. Biomech. Eng.
1985, 107 (2), 166–174. 20. Yoon, J.J.; Park, T.G.
Degradation behaviors of biodegradable macroporous
scaffolds prepared by gas foaming of effervescent salts. J.
Biomed. Mater. Res. 2001, 55 (3), 401–408. 21. Kranz, H.;
Ubrich, N.; Maincent, P.; Bodmeier, R. Physicomechanical
properties of biodegradable poly(D,L-lactide) and
poly(D,L-lactide-co-glycolide) films in the dry and wet
states. J. Pharm. Sci. 2000, 89 (12), 1558–1566. 22. Wang,
Y.; Kim Yu, M.; Langer, R. In vivo degradation
characteristics of poly(glycerol sebacate). J. Biomed.
Mater. Res. 2003, 66A (1), 192–197. 23. Lu, L.; Peter,
S.J.; Lyman, M.D.; Lai, H.L.; Leite, S.M.; Tamada, J.A.;
Uyama, S.; Vacanti, J.P.; Langer, R.; Mikos, A.G. In vitro
and in vivo degradation of porous
poly(dl-lactic-co-glycolic acid) foams. Biomaterials 2000,
21 (18), 1837–1845. 24. Vert, M.; Li, S.; Garreau, H. More
about the degradation of LA/GA-derived matrixes in aqueous
media. J. Control. Release 1991, 16 (1–2), 15–26. 25.
Kenley, R.A.; Lee, M.O.; Mahoney, T.R., II; Sanders, L.M.
Poly(lactide-co-glycolide) decomposition kinetics in vivo
and in vitro. Macromolecules 1987, 20 (10), 2398–2403. 26.
Wang, Y.; Ameer, G.; Sheppard, B.; Langer, R. A tough
biodegradable elastomer. Nat. Biotechnol. 2002, 20,
602–606. 27. Cadee, J.A.; Brouwer, L.A.; den Otter, W.;
Hennink, W.E.; Van Luyn, M.J.A. A comparative
biocompatibility study of microspheres based on crosslinked
dextran or poly(lactic-co-glycolic)acid after subcutaneous
injection in rats. J. Biomed. Mater. Res. 2001, 56 (4),
600–609. 28. van der Elst, M.; Klein, C.P.A.T.; de
BlieckHogervorst, J.M.; Patka, P.; Haarman, H.J.T.M. Bone
tissue response to biodegradable polymers used for intra
medullary fracture fixation: A long-term in vivo study in
sheep femora. Biomaterials 1999, 20 (2), 121–128.
Biosensors

1. Clark, L.C., Jr. Monitoring and control of blood tissue


PO 2 tensions. Trans. Am. Soc. Artif. Intern. Organs 1956,
2, 41–48.

2. Clark, L.C., Jr.; Lyons, C. Electrode systems for


continuous monitoring in cardiovascular surgery. Ann. N.Y.
Acad. Sci. 1962, 102, 29–45. 3. Lubbers, D.W.; Opitz, D.
Quantitative fluorescence photometry with biological fluids
and gases. Arch. Exp. Med. Biol. 1976, 75, 6511. 4.
Peterson, J.I.; Goldstein, S.R.; Fitzgerald, R.J.; Ruckold,
D.R. Fiberoptic pH probe for physiological use. Anal. Chem.
1980, 52, 864–869. 5. Kulp, T.J.; Camins, I.; Angel, S.M.;
Munkholm, C.; Walt, D.R. Polymer immobilized enzyme
optrodes for the detection of penicillin. Anal. Chem. Dec.
15, 1987, 59 (24), 2849–2853. 6. Fischer, U.; Rebrin, K.;
vonWoedtke, T.; Abel, P. Clinical usefulness of the glucose
concentration in the subcutaneous tissue—Properties and
pitfalls of electrochemical biosensors. Horm. Metab. Res.
1994, 26, 515–522. 7. Armour, J.C.; Lucisano, J.Y.; McKean,
B.D.; Gough, D.A. Application of chronic intravascular
blood glucose sensor in dogs. Diabetes 1990, 39, 1519–1526.
8. Cass, A. Biosensors. A Practical Approach; IRL Press at
Oxford University Press: Oxford, 1990; 271 pp. 9. Berson,
S.A.; Yalow, R.S. General principles of radioimmunoassay.
Clin. Chim. Acta 1968, 22, 51–69. 10. Liu, B.
Characterization of Immunosensor Binding. Ph.D. Thesis;
University of Michigan; 1986. 11. Schultz, J.S.; Mansouri,
S.; Goldstein, I.J. Affinity sensor: A new technique for
developing implantable sensors for glucose and other
metabolites. Diabetes Care 1982, 5, 245–253. 12.
Ballerstadt, R.; Schultz, J.S. A fluorescence affinity hollow
fiber sensor for continuous transdermal glucose monitoring.
Anal. Chem. 1999, 72, 4185–4192. 13. Tyagi, S.; Kramer,
F.R. Molecular beacons: Probes that fluoresce upon
hybridization. Nat. Biotechnol. 1996, 14, 303–308. 14.
Fodor, S.P.A.; Need, J.L.; Pirrung, M.C.; Stryer, L.; Lu,
A.T.; Solas, D. Light directed, spatially addressable
parallel chemical synthesis. Science 1991, 251, 767–773.
15. Cornell, B.A.; Braach-Maksvytis, V.L.B.; King, L.G.;
Osman, P.D.J.; Raguse, B.; Wieczorek, L.; Pace, R.J. A
biosensor that uses ion-channel switches. Nature 1997, 387,
580–583. 16. Pancrazio, J.J.; Whelan, D.A.; Borkholder,
W.M.A.; Stenger, D.A. Development and application of
cellbased biosensors. Ann. Biomed. Eng. 1999, 27, 697–711.
17. Selifonova, O.; Burlage, R.S.; Barkay, T.
Bioluminescent sensors for detection of bioavailable Hg(II)
in the environment. Appl. Environ. Microbiol. 1993, 59,
3083–3090. 18. Liedberg, B.; Nylander, C.; Lundstrom, I.
Biosensing with surface plasmon resonance—how it all
started. Biosens. Bioelectron. 1995 Fall, 10 (8), i–ix. 19.
Holtz, J.H.; Asher, S.A. Polymerized colloidal crystal
hydrogel films as intelligent chemical sensing materials.
Nature 1997, 389, 829–832. FURTHER READING Buck, R.P.; et
al. Biosensor Technology. Fundamentals and Applications;
Harold Dekker: New York, 1990; 419 pp. Hall, A.H.
Biosensors; Open University Press: Buckingham, 1990; 351
pp. Janata, I. Principles of Chemical Sensors; Plenum
Press: New York, 1989; 317 pp. Kress-Rogers, E. Handbook of
Biosensors and Electronic Noses; CRC Press: New York, 1996;
695 pp. Taylor, R.F.; Schultz, J.S. Handbook of Chemical
and Biological Sensors; Institute of Physics Press:
Philadelphia, 1996; 604 pp. Turner, A.P.F.; Karube, I.;
Wilson, G.S. Biosensors. Fundamentals and Applications;
Oxford Science Publications: Oxford, 1987; 770 pp. B
Biosynthesis and Applications of Alginates

9. Ross, P.; Mayer, R.; Benziman, M. Cellulose biosynthesis


and function in bacteria. Microbiol. Rev. 1991, 55, 35–58.

10. Chu, L.; May, T.B.; Chakrabarty, A.M.; Misra, T.K.


Nucleotide sequence and expression of the algE gene
involved in alginate biosynthesis by Pseudomonas
aeruginosa. Gene 1991, 107, 1–10.

11. Rehm, B.H.; Boheim, G.; Tommassen, J.; Winkler, U.K.


Overexpression of algE in Escherichia coli: subcellular
localization, purification, and ion channel properties. J.
Bacteriol. 1994, 176, 5639–5647.

12. Rehm, B.H.; Grabert, E.; Hein, J.; Winkler, U.K.


Antibody response of rabbits and cystic fibrosis patients to
an alginate-specific outer membrane protein of a mucoid
strain of Pseudomonas aeruginosa. Microb. Pathog. 1994, 16,
43–51.

13. Rehm, B.H. The Azotobacter vinelandii gene algJ encodes


an outer-membrane protein presumably involved in export of
alginate. Microbiology 1996, 142 (pt 4), 873–880.

14. Nyvall, P.; Corre, E.; Boisset, C.; Barbeyron, T.;


Rousvoal, S.; Scornet, D.; Kloareg, B.; Boyen, C.
Characterization of mannuronan C-5-epimerase genes from the
brown alga Laminaria digitata. Plant Physiol. 2003, 133,
726–735.

15. Franklin, M.J.; Chitnis, C.E.; Gacesa, P.; Sonesson,


A.; White, D.C.; Ohman, D.E. Pseudomonas aeruginosa AlgG is
a polymer level alginate C5-mannuronan epimerase. J.
Bacteriol. 1994, 176, 1821–1830.

16. Franklin, M.J.; Ohman, D.E. Identification of algI and


algJ in the Pseudomonas aeruginosa alginate biosynthetic
gene cluster which are required for alginate O acetylation.
J. Bacteriol. 1996, 178, 2186–2195.

17. Schiller, N.L.; Monday, S.R.; Boyd, C.M.; Keen, N.T.;


Ohman, D.E. Characterization of the Pseudomonas aeruginosa
alginate lyase gene (algL): cloning, sequencing, and
expression in Escherichia coli. J. Bacteriol. 1993, 175,
4780–4789.

18. Nivens, D.E.; Ohman, D.E.; Williams, J.; Franklin, M.J.


Role of alginate and its O acetylation in formation of
Pseudomonas aeruginosa microcolonies and biofilms. J.
Bacteriol. 2001, 183, 1047–1057.

19. Bjerkan, T.M.; Lillehov, B.E.; Strand, W.I.;


SkjakBraek, G.; Valla, S.; Ertesvag, H. Construction and
analyses of hybrid Azotobacter vinelandii mannuronan C-5
epimerases with new epimerization pattern characteristics.
Biochem. J. 2004, 381, 813–821.

20. Ertesvag, H.; Hoidal, H.K.; Schjerven, H.; Svanem,


B.I.; Valla, S. Mannuronan C-5-epimerases and their
application for in vitro and in vivo design of new
alginates useful in biotechnology. Metab. Eng. 1999, 1,
262–269.

21. Valla, S.; Li, J.; Ertesvag, H.; Barbeyron, T.;


Lindahl, U. Hexuronyl C5-epimerases in alginate and
glycosaminoglycan biosynthesis. Biochimie 2001, 83,
819–830.

22. Campos, M.; Martinez-Salazar, J.M.; Lloret, L.; Moreno,


S.; Nunez, C.; Espin, G.; Soberon-Chavez, G.
Characterization of the gene coding for GDP-mannose
dehydrogenase (algD) from Azotobacter vinelandii. J.
Bacteriol. 1996, 178, 1793–1799. 23. Hoidal, H.K.; Glaerum
Svanem, B.I.; Gimmestad, M.; Valla, S. Mannuronan C-5
epimerases and cellular differentiation of Azotobacter
vinelandii. Environ. Microbiol. 2000, 2, 27–38. 24. Rehm,
B.H.; Ertesvag, H.; Valla, S. A new Azotobacter vinelandii
mannuronan C-5-epimerase gene (algG) is part of an alg gene
cluster physically organized in a manner similar to that in
Pseudomonas aeruginosa. J. Bacteriol. 1996, 178, 5884–5889.
25. Gimmestad, M.; Sletta, H.; Ertesvag, H.; Bakkevig, K.;
Jain, S.; Suh, S.J.; Skjak-Braek, G.; Ellingsen, T.E.;
Ohman, D.E.; Valla, S. The Pseudomonas fluorescens AlgG
protein, but not its mannuronan C-5-epimerase activity, is
needed for alginate polymer formation. J. Bacteriol. 2003,
185, 3515–3523. 26. Jain, S.; Franklin, M.J.; Ertesvag, H.;
Valla, S.; Ohman, D.E. The dual roles of AlgG in
C-5-epimerization and secretion of alginate polymers in
Pseudomonas aeruginosa. Mol. Microbiol. 2003, 47,
1123–1133. 27. May, T.B.; Shinabarger, D.; Maharaj, R.;
Kato, J.; Chu, L.; DeVault, J.D.; Roychoudhury, S.;
Zielinski, N.A.; Berry, A.; Rothmel, R.K.; Misra, K.T.;
Chakrabarty, A.M. Alginate synthesis by Pseudomonas
aeruginosa: a key pathogenic factor in chronic pulmonary
infections of cystic fibrosis patients. Clin. Microbiol.
Rev. 1991, 4, 191–206. 28. Schmitt-Andrieu, L.; Hulen, C.
Alginates of Pseudomonas aeruginosa: a complex regulation
of the pathway of biosynthesis. C.R. Acad Sci III 1996,
319, 153–160. 29. Schurr, M.J.; Yu, H.; Martinez-Salazar,
J.M.; Boucher, J.C.; Deretic, V. Control of AlgU, a member
of the sigma E-like family of stress sigma factors, by the
negative regulators MucA and MucB and Pseudomonas
aeruginosa conversion to mucoidy in cystic fibrosis. J.
Bacteriol. 1996, 178, 4997–5004. 30. Schurr, M.J.; Martin,
D.W.; Mudd, M.H.; Hibler, N.S.; Boucher, J.C.; Deretic, V.
The algD promoter: regulation of alginate production by
Pseudomonas aeruginosa in cystic fibrosis. Cell. Mol. Biol.
Res. 1993, 39, 371–376. 31. Baynham, P.J.; Wozniak, D.J.
Identification and characterization of AlgZ, an
AlgT-dependent DNA-binding protein required for Pseudomonas
aeruginosa algD transcription. Mol. Microbiol. 1996, 22,
97–108. 32. Cochran, W.L.; Suh, S.J.; McFeters, G.A.;
Stewart, P.S. Role of RpoS and AlgT in Pseudomonas
aeruginosa biofilm resistance to hydrogen peroxide and
monochloramine. J. Appl. Microbiol. 2000, 88, 546–553. 33.
Firoved, A.M.; Boucher, J.C.; Deretic, V. Global genomic
analysis of AlgU (sigma(E))-dependent promoters (sigmulon)
in Pseudomonas aeruginosa and implications for inflammatory
processes in cystic fibrosis. J. Bacteriol. 2002, 184,
1057–1064. 34. Firoved, A.M.; Deretic, V. Microarray
analysis of global gene expression in mucoid Pseudomonas
aeruginosa. J. Bacteriol. 2003, 185, 1071–1081. 35. Keith,
L.M.; Bender, C.L. AlgT (sigma22) controls alginate
production and tolerance to environmental stress in
Pseudomonas syringae. J. Bacteriol. 1999, 181, 7176–7184.
36. Mathee, K.; McPherson, C.J.; Ohman, D.E.
Posttranslational control of the algT (algU)-encoded
sigma22 for expression of the alginate regulon in
Pseudomonas aeruginosa and localization of its antagonist
proteins MucA and MucB (AlgN). J. Bacteriol. 1997, 179,
3711–3720. 37. Wu, W.; Badrane, H.; Arora, S.; Baker, H.V.;
Jin, S. MucA-mediated coordination of type III secretion
and alginate synthesis in Pseudomonas aeruginosa. J.
Bacteriol. 2004, 186, 7575–7585. 38. Yorgey, P.; Rahme,
L.G.; Tan, M.W.; Ausubel, F.M. The roles of mucD and
alginate in the virulence of Pseudomonas aeruginosa in
plants, nematodes and mice. Mol. Microbiol. 2001, 41,
1063–1076. 39. Boucher, J.C.; Schurr, M.J.; Yu, H.; Rowen,
D.W.; Deretic, V. Pseudomonas aeruginosa in cystic fibrosis:
role of mucC in the regulation of alginate production and
stress sensitivity. Microbiology 1997, 143 (pt 11),
3473–3480. 40. Deretic, V.; Dikshit, R.; Konyecsni, W.M.;
Chakrabarty, A.M.; Misra, T.K. The algR gene, which
regulates mucoidy in Pseudomonas aeruginosa, belongs to a
class of environmentally responsive genes. J. Bacteriol.
1989, 171, 1278–1283. 41. Deretic, V.; Konyecsni, W.M.
Control of mucoidy in Pseudomonas aeruginosa:
transcriptional regulation of algR and identification of the
second regulatory gene, algQ. J. Bacteriol. 1989, 171,
3680–3688. 42. Mohr, C.D.; Hibler, N.S.; Deretic, V. AlgR,
a response regulator controlling mucoidy in Pseudomonas
aeruginosa, binds to the FUS sites of the algD promoter
located unusually far upstream from the mRNA start site. J.
Bacteriol. 1991, 173, 5136–5143. 43. Zielinski, N.A.;
Chakrabarty, A.M.; Berry, A. Characterization and
regulation of the Pseudomonas aeruginosa algC gene encoding
phosphomannomutase. J. Biol. Chem. 1991, 266, 9754–9763.
44. Carterson, A.J.; Morici, L.A.; Jackson, D.W.; Frisk,
A.; Lizewski, S.E.; Jupiter, R.; Simpson, K.; Kunz, D.A.;
Davis, S.H.; Schurr, J.R.; Hassett, D.J.; Schurr, M.J. The
transcriptional regulator AlgR controls cyanide production
in Pseudomonas aeruginosa. J. Bacteriol. 2004, 186,
6837–6844. 45. Lizewski, S.E.; Lundberg, D.S.; Schurr, M.J.
The transcriptional regulator AlgR is essential for
Pseudomonas aeruginosa pathogenesis. Infect. Immun. 2002,
70, 6083–6093. 46. Lizewski, S.E.; Schurr, J.R.; Jackson,
D.W.; Frisk, A.; Carterson, A.J.; Schurr, M.J.
Identification of AlgRregulated genes in Pseudomonas
aeruginosa by use of microarray analysis. J. Bacteriol.
2004, 186, 5672–5684. 47. Penaloza-Vazquez, A.; Fakhr,
M.K.; Bailey, A.M.; Bender, C.L. AlgR functions in algC
expression and virulence in Pseudomonas syringae pv.
syringae. Microbiology 2004, 150, 2727–2737. 48. Chitnis,
C.E.; Ohman, D.E. Genetic analysis of the alginate
biosynthetic gene cluster of Pseudomonas aeruginosa shows
evidence of an operonic structure. Mol. Microbiol. 1993, 8,
583–593. 49. Lloret, L.; Barreto, R.; Leon, R.; Moreno, S.;
MartinezSalazar, J.; Espin, G.; Soberon-Chavez, G. Genetic
analysis of the transcriptional arrangement of Azotobacter
vinelandii alginate biosynthetic genes: identification of
two independent promoters. Mol. Microbiol. 1996, 21 ,
449–457. 50. Vazquez, A.; Moreno, S.; Guzman, J.; Alvarado,
A.; Espin, G. Transcriptional organization of the
Azotobacter vinelandii algGXLVIFA genes: characterization
of algF mutants. Gene 1999, 232, 217–222. 51. Thomas, S.
Alginate dressings in surgery and wound management—part 1.
J. Wound Care 2000, 9, 56–60. 52. Malmstrom, A.; Fransson,
L.A. Biosynthesis of dermatan sulfate. I. Formation of
L-iduronic acid residues. J. Biol. Chem. 1975, 250,
3419–3425. 53. Ertesvag, H.; Hoidal, H.K.; Hals, I.K.;
Rian, A.; Doseth, B.; Valla, S. A family of modular type
mannuronan C-5epimerase genes controls alginate structure
in Azotobacter vinelandii. Mol. Microbiol. 1995, 16,
719–731. 54. Svanem, B.I.; Skjak-Braek, G.; Ertesvag, H.;
Valla, S. Cloning and expression of three new Aazotobacter
vinelandii genes closely related to a previously described
gene family encoding mannuronan C-5-epimerases. J.
Bacteriol. 1999, 181, 68–77. 55. Ertesvag, H.; Doseth, B.;
Larsen, B.; Skjak-Braek, G.; Valla, S. Cloning and
expression of an Azotobacter vinelandii mannuronan
C-5-epimerase gene. J. Bacteriol. 1994, 176, 2846–2853. 56.
Ertesvag, H.; Hoidal, H.K.; Skjak-Braek, G.; Valla, S. The
Azotobacter vinelandii mannuronan C-5-epimerase AlgE1
consists of two separate catalytic domains. J. Biol. Chem.
1998, 273, 30927–30932. 57. Svanem, B.I.; Strand, W.I.;
Ertesvag, H.; Skjak-Braek, G.; Hartmann, M.; Barbeyron, T.;
Valla, S. The catalytic activities of the bifunctional
Azotobacter vinelandii mannuronan C-5-epimerase and
alginate lyase AlgE7 probably originate from the same
active site in the enzyme. J. Biol. Chem. 2001, 276,
31542–31550. 58. Campa, C.; Holtan, S.; Nilsen, N.;
Bjerkan, T.M.; Stokke, B.T.; Skjak-Braek, G. Biochemical
analysis of the processive mechanism for epimerization of
alginate by mannuronan C-5 epimerase AlgE4. Biochem. J.
2004, 381, 155–164. 59. Donati, I.; Draget, K.I.; Borgogna,
M.; Paoletti, S.; Skjak-Braek, G. Tailor-made alginate
bearing galactose moieties on mannuronic residues:
selective modification achieved by a chemoenzymatic
strategy. Biomacromolecules 2005, 6, 88–98. 60. Donati, I.;
Holtan, S.; Morch, Y.A.; Borgogna, M.; Dentini, M.;
Skjak-Braek, G. New hypothesis on the role of alternating
sequences in calcium-alginate gels. Biomacromolecules 2005,
6, 1031–1040. 61. King, A.; Strand, B.; Rokstad, A.M.;
Kulseng, B.; Andersson, A.; Skjak-Braek, G.; Sandler, S.
Improvement of the biocompatibility of
alginate/poly-L-lysine/alginate microcapsules by the use of
epimerized alginate as a coating. J. Biomed. Mater. Res. A
2003, 64, 533–539. 62. Pandey, R.; Khuller, G.K.
Chemotherapeutic potential of alginate-chitosan
microspheres as anti-tubercular drug carriers. J.
Antimicrob. Chemother. 2004, 53, 635–640. 63. Kulseng, B.;
Skjak-Braek, G.; Ryan, L.; Andersson, A.; King, A.;
Faxvaag, A.; Espevik, T. Transplantation of alginate
microcapsules: generation of antibodies against B alginates
and encapsulated porcine islet-like cell clusters.
Transplantation 1999, 67, 978–984.

64. Rokstad, A.M.; Kulseng, B.; Strand, B.L.; SkjakBraek,


G.; Espevik, T. Transplantation of alginate microcapsules
with proliferating cells in mice: capsular overgrowth and
survival of encapsulated cells of mice and human origin.
Ann. N.Y. Acad. Sci. 2001, 944, 216–225.

65. Rokstad, A.M.; Strand, B.; Rian, K.; Steinkjer, B.;


Kulseng, B.; Skjak-Braek, G.; Espevik, T. Evaluation of
different types of alginate microcapsules as bioreactors
for producing endostatin. Cell Transplant. 2003, 12,
351–364.
66. Strand, B.L.; Morch, Y.A.; Syvertsen, K.R.; Espevik,
T.; Skjak-Braek, G. Microcapsules made by enzymatically
tailored alginate. J. Biomed. Mater. Res. A 2003, 64,
540–550. 67. Schneider, S.; Feilen, P.J.; Brunnenmeier, F.;
Minnemann, T.; Zimmermann, H.; Zimmermann, U.; Weber, M.M.
Long-term graft function of adult rat and human islets
encapsulated in novel alginate-based microcapsules after
transplantation in immunocompetent diabetic mice. Diabetes
2005, 54, 687–693. 68. Soon-Shiong, P.; Heintz, R.E.;
Merideth, N.; Yao, Q.X.; Yao, Z.; Zheng, T.; Murphy, M.;
Moloney, M.K.; Schmehl, M.; Harris M., et al. Insulin
independence in a type 1 diabetic patient after
encapsulated islet transplantation. Lancet 1994, 343,
950–951.
Biphasic Calcium Phosphate (BCP)
Bioceramics: Chemical, Physical, and
Biological Properties

16. Daculsi, G.; Bagot D’arc, M.; Corlieu, P.; Gersdorff,


M. Ann. Orol. Rhinol. Laryngol. 1992, 101, 669.

17. Trecant, M.; Delecrin, J.; Royer, J.; Goyenvalle, E.;


Daculsi, G. Clin. Mater. 1994, 15, 233.

18. Gouin, F.; Delecrin, J.; Passuti, N.; Touchais, S.;


Poirier, P.; Bainvel, J.V. Chir. Orthop. 1995, 81, 59.

19. Ransford, P.; Passuti, N.; Chopin, D.; Morin, C. J.


Bone Joint Surg. Br. 1998, 80, 13.

20. Cavagna, R.; Daculsi, G.; Bouler, J.-M. Longterm


Effects Med. Impl. 1999, 9, 403.

21. Soares, E.J.C.; Franca, V.P.; Wykrota, L.; Stumpf, S.


Bioceramics 11; LeGeros, R.Z., LeGeros, J.P., Eds.; World
Scientific: Singapore, 1998; 633 pp.

22. Wykrota, L.L.; Garrido, C.A.; Wykrota, F.H.I.


Bioceramics 11; LeGeros, R.Z., LeGeros, J.P., Eds.; World
Scientific: Singapore, 1998; 641 pp.

23. Malard, O.; Guicheux, J.; Bouler, J.M.; Gauthier, O.;


Beauvillain de Montreuil, C.; Aguado, E.; Pilet, P.;
LeGeros, R.; Daculsi, G. Bone 2005, 36, 323–330.

24. Daculsi, G. Biomaterials 1998, 19, 1473–1478.

25. Daculsi, G.; Laboux, O.; Malard, O.; Weiss, P. J.


Mater. Sci. Mater. Med. 2003, 14, 195–200.

26. LeGeros, R.Z.; Lin, S.; Rohanizadeh, R.; Mijares, D.;


LeGeros, J.P. J. Mater. Sci. Mat. Med. 2003, 14, 201–210.

27. Hench, L.L.; Splinter, R.J.; Allen, W.C.; Greelee, T.K.


J. Biomed. Mater. Res. 1971, 2, 117–141.

28. LeGeros, R.Z. Calcium phosphates in oral biology and


medicine. In Monographs in Oral Sciences; Myers, H., Ed.;
S. Karger: Basel, 1991; Vol. 15.

29. Basle, M.; Chappard, D.; Grizon, F.; Filmon, R.;


Daculsi, D.; Rebel, A. Calcif. Tissue Int. 1993, 53, 348.

30. Gauthier, O.; Bouler, J.M.; Aguado, E.; Pilet, P.;


Daculsi, G. Biomaterials 1998, 11, 133.

31. Osborne, J.; Newesely, H. Biomaterials 1980, 1, 108.

32. Heughebaert, M.; LeGeros, R.Z.; Gineste, M.; Guilhem,


A. J. Biomed. Mat. Res. 1988, 22, 257–268.

33. Daculsi, G.; LeGeros, R.Z.; Heugheaert, M.; Barbieux,


I. Calcif. Tissue Int. 1990, 46, 20–27.

34. LeGeros, R.Z.; Daculsi, G.; Orly, I.; LeGeros, J.P. The
Bone Biomaterial Interface; Davies, J.E.D., Ed.; University
of Toronto Press, 1991; 76–88.

35. LeGeros, R.Z. Prog. Cryst. Growth Charact. 1981, 4, 1.

36. Bouler, J.M.; LeGeros, R.Z.; Daculsi, G. J. Biomed.


Mater. Res. 2000, 51, 680.

37. LeGeros, R.Z.; Zheng, R.; Kijkowska, R.; Fan, D.;


LeGeros, J.P. Characterization, Performance of Calcium
Phosphate Coatings for Implants; Horowitz, E., Parr, J.E.,
Eds.; American Society For Testing Materials Stp:
Philadelphia, 1994; Vol. 1, 198,43 pp.

38. Bouler, J.M.; Trecant, M.; Delecrin, J.; Royer, J.;


Passuti, N.; Daculsi, G. J. Biomed. Mater. Res. 1996, 32,
603.

39. Hubbard, W.G. Ph.D. Thesis. Marquette University:


Milwaukee, 1974.

40. Schmitt, M. Ph.D. Thesis. Universite De Nantes: Nantes,


2000.

41. Driessen, A.A.; Klein, C.P.A.T.; De Groot, K.


Biomaterials 1982, 3, 113.

42. Rohanizadeh, R.; Padrines, M.; Bouler, J.M.;


Couchourel, G.; Fortun, Y.; Daculsi, G. J. Biomed. Mater.
Res. 1998, 42, 530. 43. Gauthier, O.; Bouler, J.M.; Aguado,
E.; LeGeros, R.Z.; Pilet, P.; Daculsi, G. J. Mat. Sci. Mat.
Med. 1999, 10, 99. 44. LeGeros, R.Z. Clin. Mater. 1993, 14,
65. 45. Soueidan, A.; Gan, O.I.; Bouler, J.M.; Gouin, F.;
Daculsi, G. Cells Mater. 1995, 5, 31. 46. Benahmed, M.;
Bouler, J.M.; Heymann, D.; Gan, O.; Daculsi, G.
Biomaterials 1996, 17, 2173. 47. Yamada, S.; Heymann, D.;
Bouler, J.M.; Daculsi, G. Biomaterials 1997, 18, 1037. 48.
Chang, Y.L.; Stanford, C.M.; Keller, J.C. J. Biomed. Mater.
Res. 2000, 52, 270. 49. Yuan, H.; Kurashina, K.; Joost de
Bruijn, D.; Li, Y.; de Groot, K.; Zhang, X. Biomaterials
1999, 20, 1799–1806. 50. Kuboki, Y.; Takita, H.; Kobayashi,
D. J. Biomed. Mater. Res. 1998, 3, 190. 51. Ripamonti, U. J
Bone Joint Surg. Am. 1991, 73, 692–703. 52. Ripamonti, U.
Biomaterials. 1996, 17, 31–35. 53. Ripamonti, U.; Crooks,
J.; Kirbride, A. South Africa J. Sci. 1999, 95, 335–343.
54. Daculsi, G.; Corre, P.; Malard, O.; Legeros, R.;
Goyenvalle, E. Bioceramics 18, key engineering, 2005. 55.
Daculsi, G.; Weiss, P.; Delecrin, J.; Grimandi, G.;
Passuti, N.; Guerin, F. Compositions pour Biomate´riaux.
Patent 94-01-414, 235 1994. 56. Daculsi, G.; Weiss, P.;
Bouler, J.M.; Gauthier, O.; Aguado, E. Bone 1999, 25,
59–61. 57. Millot, F.; Grimandi, G.; Weiss, P.; Daculsi, G.
Cells Mat. 1999, 9, 21–30. 58. Constanz, B.R.; Ison, I.C.;
Fulmer, M.T.; Poser, R.D.; Smith, S.T.; VanWagoner, M.;
Ross, J.; Goldstein, S.A. Science 1995, 267, 1796–1799. 59.
Niwa, S.; LeGeros, R.Z. Tissue Engineering and
Biodegradable Equivalents. Scientific and Clinical
Applications; Lewandrowski, K.-U., Wise, D.L., Trantolo,
D.J., Gresser, J.D., Eds.; Marcel Dekker: New York, 2002;
395 pp. 60. LeGeros, R.Z. Clin. Orthopaed. Rel. Res. 2002,
395, 81. 61. Reddi, A.H. Tissue Eng. 2000, 6, 351. 62.
Barrere, F.; van der Valk, C.M.; Dalmeijer, R.A.; Meijer,
G.; van Blitterswijk, C.A.; de Groot, K.; Layrolle, P. J.
Biomed. Mater. Res. 2003, 66A, 779. 63. Habibovic, P.; van
der Valk, C.M.; van Blitterswijk, C.A.; De Groot, K.;
Meijer, G. J. Mater. Sci. Mater. Med. 2004, 15, 373–380.
64. Habibovic, P.; Li, J.; van der Valk, C.M.; Meijer, G.;
Layrolle, P.; van Blitterswijk, C.A.; de Groot, K.
Biomaterials 2005, 26, 23. 65. Le Nihouhannen, D.; Daculsi,
G.; Gauthier, O.; Saffarzadeh, A.; Delplace, S.; Pilet, P.;
Layrolle, P. Bone 2005, 36, 1086–1096. 66. Daculsi, G.;
Layrolle, P. Key Eng. Mat. 2004, 254–256, 1005. 67. De
Groot, J. Tissue Eng. 1998, 4, 337. 68. Diaz-Flores, L.;
Gutierrez, R.; Lopez-Alonso, A.; Gonzalez, R.; Varela, H.
Clin. Orthop. 1992, 6, 280. 69. Steitz, S.A.; Speer, M.Y.;
Curinga, G.; Yang, H.Y.; Haynes, P.; Aebersold, R.;
Schinke, T.; Karsenty, G.; Giachelli, C.M. Circ. Res. 2001,
89, 1147–1154. Phosphate (BCP)
Blood-Material Interactions

5. Bennett, B.; Booth, N.A.; Ogston, D. Potential


Interactions Between Complement, Coagulation, Fibrinolysis,
Kinin-Forming, and Other Enzyme Systems. In Haemostasis and
Thrombosis, 2nd Ed.; Bloom, A.L., Thomas, D.P., Eds.;
Churchill Livingstone: New York, 1987; 267–282.

6. Hanson, S.R.; Griffin, J.H.; Harker, L.A.; Kelly, A.B.;


Esmon, C.T.; Gruber, A. Antithrombotic effects of
thrombin-induced activation of endogenous protein C in
primates. J. Clin. Invest. 1993, 92, 2003–2012.

7. Coughlin, S.R. Protease-activated receptors in vascular


biology. Thromb. Haemost. 2001, 86 (1), 298–307.

8. Hanson, S.R.; Harker, L.A.; Ratner, B.D.; Hoffmann, A.S.


In vivo evaluation of artificial surfaces using a nonhuman
primate model of arterial thrombosis. J. Lab. Clin. Med.
1980, 95, 289–304.

9. Platelet Kinetics; Paulus, J.M., Ed.; American Elsevier:


New York, 1971; 1–360.

10. Slack, S.M.; Cui, Y.; Turitto, V.T. The effects of flow
on blood coagulation and thrombosis. Thromb. Haemost. 1993,
70 (1), 129–134.

11. Turitto, V.T.; Weiss, H.J. Red cells: Their dual role
in thrombus formation. Science 1980, 207, 541–544. 12.
Brodbeck, W.G.; Nakayama, Y.; Matsuda, T.; Colton, E.;
Ziats, N.P.; Anderson, J.M. Biomaterial surface chemistry
dictates adherent monocyte/macrophage cytokine expression
in vivo. Cytokine 2002, 18 (6), 311–319. 13. Shive, M.S.;
Brodbeck, W.G.; Colton, E.; Anderson, J.M. Shear stress and
material surface effects on adherent human monocyte
apoptosis. J. Biomed. Mater. Res. 2002, 60 (1), 148–158.
14. Salzman, E.W.; Merrill, E.D. Interaction of Blood with
Artificial Surfaces. In Haemostasis and Thrombosis, 2nd Ed.;
Colman, R.W., Hirsh, J., Marder, V.J., Salzman, E.W., Eds.;
J.B. Lippincott: Philadelphia, 1987; 1335– 1347. 15. Yu,
J.; Lamba, N.M.; Courtney, J.M. Polymeric biomaterials:
Influence of phosphorylcholine polar groups on protein
adsorption and complement activation. Int. J. Artif. Organs
1994, 17 (9), 499–504. 16. Verheye, S.; Markou, C.P.;
Salame, M.Y.; Wan, B.; King, S.B.; Robinson, K.A.; Chronos,
N.A.; Hanson, S.R. Reduced thrombus formation by hyaluronic
acid coating of endovascular devices. Arterioscler. Thromb.
Vasc. Biol. 2000, 20, 1168–1172. B
Blood Purification

Fig. 21 Filtrate flux is represented as a function of the

transmembrane pressure for dialysis, hemofiltration, and

plasma separation. The shaded regions represent normal

operating ranges. Filtration flux is equal to the number

of milliliters per minute transferred per meter squared

(ml/min/m 2 ). (From Ref. [2].)


Blood Vessel Mechanics

36. Taber, L.A.; Humphrey, J.D. Stress-modulated growth,


residual stress, and vascular heterogeneity. J. Biomech.
Eng. 2001, 123 (6), 528–535.

37. Liu, S.Q.; Fung, Y.C. Indicial functions of arterial


remodeling in response to locally altered blood pressure.
Am. J. Physiol. 1996, 270 (4 Pt 2), H1323–H1333.

38. Gleason, R.L.; Taber, L.A.; Humphrey, J.D. A 2-D model


of flow-induced alterations in the geometry, structure, and
properties of carotid arteries. J. Biomech. Eng. 2004, 126
(3), 371–381.

39. Bunce, D.F.M. Atlas of Arterial Histology; Warren H.


Green: St. Louis, MO, 1974.

40. Nichols, W.W.; O’Rourke, M.F.; Hartley, C. McDonald’s


Blood Flow in Arteries: Theoretical, Experimental, and
Clinical Principles, 4th Ed.; Oxford University Press: New
York, 1998.

41. O’Rourke, M.F.; Staessen, J.A.; Vlachopoulos, C.; et


al. Clinical applications of arterial stiffness; definitions
and reference values. Am. J. Hypertens. 2002, 15 (5),
426–444.

42. Fridez, P.; Makino, A.; Miyazaki, H.; et al. Short-term


biomechanical adaptation of the rat carotid to acute
hypertension: contribution of smooth muscle. Ann. Biomed.
Eng. 2001, 29 (1), 26–34.

43. Davis, M.J.; Gore, R.W. Length–tension relationship of


vascular smooth muscle in single arterioles. Am. J.
Physiol. 1989, 256 (3 Pt 2), H630–H640.

44. Wesly, R.L.; Vaishnav, R.N.; Fuchs, J.C.; et al. Static


linear and nonlinear elastic properties of normal and
arterialized venous tissue in dog and man. Circ. Res. 1975,
37 (4), 509–520. 45. Ben Driss, A.; Benessiano, J.;
Poitevin, P.; et al. Arterial expansive remodeling induced
by high flow rates. Am. J. Physiol. 1997, 272 (2 Pt 2),
H851–H858. 46. Saini, A.; Berry, C.; Greenwald, S. Effect
of age and sex on residual stress in the aorta. J. Vasc.
Res. 1995, 32 (6), 398–405. 47. Bruel, A.; Oxlund, H.
Changes in biomechanical properties, composition of
collagen and elastin, and advanced glycation endproducts of
the rat aorta in relation to age. Atherosclerosis 1996, 127
(2), 155–165. 48. Chau, A.H.; Chan, R.C.; Shishkov, M.; et
al. Mechanical analysis of atherosclerotic plaques based on
optical coherence tomography. Ann. Biomed. Eng. 2004, 32
(11), 1494–1503. 49. Vande Geest, J.P.; Sacks, M.S.; Vorp,
D.A. The effects of aneurysm on the baxial mechanical
behavior of human abdominal aorta. J. Biomech. 2006, 39
(7), 1324–1334. 50. Sims, T.J.; Rasmussen, L.M.; Oxlund,
H.; et al. The role of glycation cross-links in diabetic
vascular stiffening. Diabetologia 1996, 39 (8), 946–951.
51. Liu, S.Q.; Fung, Y.C. Changes in the rheological
properties of blood vessel tissue remodeling in the course
of development of diabetes. Biorheology 1992, 29 (5–6),
443–457. 52. Burton, A.C. Relation of structure to function
of the tissues of the wall of blood vessels. Physiol. Rev.
1954, 34 (4), 619–642.
Bone Cement

42. Vazquez, B.; Roman, J.S.; Deb, S.; Bonfield, W.


Application of long chain amine activator in conventional
acrylic bone cement. J. Biomed. Mater. Res. 1998, 43 (2),
131–139.

43. Vazquez, B.; Deb, S.; Bonfield, W.; Roman, J.S.


Characterization of new acrylic bone cements prepared with
oleic acid derivatives. J. Biomed. Mater. Res. 2002, 63
(2), 88–97. 44. Hasenwinkel, J.M.; Lautenschlager, E.P.;
Wixson, R.L.; Gilbert, J.L. Effect of initiation chemistry
on the fracture toughness, fatigue strength, and residual
monomer content of a novel high-viscosity, two-solution
acrylic bone cement. J. Biomed. Mater. Res. 2002, 59,
411–421. 45. Hasenwinkel, J.M.; Lautenschlager, E.P.;
Wixson, R.L.; Gilbert, J.L. A novel high viscosity, two
solution acrylic bone cement: Effect of chemical
composition on properties. J. Biomed. Mater. Res. 1999, 47
(1), 36–45.
Bone Fracture Fixation

1. Brinker, M. Basic Sciences. In Review of Orthopaedics,


3rd Ed.; Miller, M., Brinker, M., Eds.; W.B. Saunders:
Philadelphia, 2000; pp. 1–22, 119–127.

2. Claiborne Christian General Principles of Fracture


Treatment. In Operative Orthopaedics, 9th Ed.; Canale, T.,
Ed.; Mosby: St. Louis, 1998; Vol. 3, 2011–2015, 2018–2021.
3. Harkess, J.; Ramsey, W.; Harkess, J. Principles of
Fracture and Dislocations. In Rockwood and Green’s
Fractures in Adults, 3rd Ed.; Rockwood, C., Jr., Green, P.,
Bucholz, R., Eds.; J.B. Lippincott Company: Philadephia,
1991; Vol. 1, 119–121. 4. Allen, C.W.; Piotrowski, G.;
Burstein, A.H.; Frankel, V.H. Biomechanical principles of
intramedullary fixation. Clin. Ortop. 1968, 60, 13–20. 5.
Rhinelander, F.W. Effects of medullary nailing on the
normal blood supply of diaphyseal cortex. Clin. Ortop.
1998, 350, 5–17. 6. Perren, S.M. The biomechanics and
biology of internal fixation using plates and nails.
Orthopedics 1989, 12 (1), 21–34. 7. Tydings, J.D.; Martino,
L.J.; Kircher, M.; Alfred, R.; Lozman, J. The
osteoinductive potential of intramedullary canal bone
reamings. Curr. Surg. 1986, 43 (2), 121–124. 8. Giannoudis,
P.V.; Pape, H.C.; Cohen, A.P.; Krettek, C.; Smith, R.M.
Systemic effects of femoral nailing. Clin. Ortop. 2002,
404, 378–386. 9. Nepola, J.V. External Fixation. In
Rockwood and Green’s Fractures in Adults, 4th Ed.;
Rockwood, C., Jr., Green, P., Bucholz, R., Heckman, J.D.,
Eds.; J.B. Lippincott-Raven Publishers: Philadephia, 1996;
Vol. 1, 229–231, 235–240. 10. Behrems, F. A primer of
fixator devices and configurations. Clin. Ortop. 1989, 241,
5–15.
Bone–Implant Interface

1. Osborn, J.F.; Newesley, H. Dynamic Aspects of the


Implant–Bone Interface. In Dental Implants—Materials and
Systems; Heimke, G., Ed.; Carl Hanser: Munich, 1980;
111–123.

2. Roberts, W.E. Bone tissue interface. J. Dent. Educ.


1988, 52, 804–809.

3. Bra˚nemark, P.-I. Introduction to Osseointegration. In


Tissue-Integrated Prostheses; Bra˚nemark, P.-I., Zarb,
G.A., Albrektsson, T., Eds.; Quintessence: Chicago, 1985;
11–76.

4. Bra˚nemark, P.I.; Hansson, B.O.; Adell, R.; Breine, U.;


Lindstrom, J.; Hallen, O.; Ohman, A. Osseointegrated
implants in the treatment of the edentulous jaw. Experience
from a 10-year period. Scand. J. Plast. Reconstr. Surg.,
Suppl. 1977, 16, 1–132.

5. Steflik, D.E.; Sisk, A.L.; Parr, G.R.; Lake, F.T.; Hanes,


P.J.; Berkerey, D.J.; Brewer, P. Transmission electron and
high-voltage electron microscopy of osteocyte cellular
processes extending to the dental implant surface. J.
Biomed. Mater. Res. 1994, 28, 1095–1107.

6. Dorr, L.D.; Bloebaum, R.; Emmanual, J.; Meldrum, R.


Histologic, biochemical, and ion analysis of tissue and
fluids retrieved during total hip arthroplasty. Clin.
Orthop. 1990, 261, 82–95.

7. Jacobs, J.J.; Skipor, A.K.; Patterson, L.M.; Hallab,


N.J.; Paprosky, W.G.; Black, J.; Galante, J.O. Metal
release in patients who have had a primary total hip
arthroplasty. A prospective, controlled, longitudinal
study. J. Bone Jt. Surg., Am. 1998, 80, 1447–1458.

8. Thompson, G.J.; Puleo, D.A. Ti–6Al–4V ion solution


inhibition of osteogenic cell phenotype as a function of
differentiation timecourse in vitro. Biomaterials 1996, 17,
1949–1954.

9. Nanci, A.; McKee, M.D.; Zalzal, S.; Sakkal, S.


Ultrastructural and Immunocytochemical Analysis of the
Tissue Response to Metal Implants in the Rat Tibia. In
Biological Mechanisms of Tooth Eruption, Resorption and
Replacement by Implants; Davidovitch, Z., Mah, J., Eds.;
Harvard Society for the Advancement of Orthodontics:
Boston, 1998; 487–500.
10. Hench, L.L.; Splinter, R.J.; Allen, W.C.; Greenlee,
T.K., Jr. Bonding mechanisms at the interface of ceramic
prosthetic materials. Biomed. Mater. Symp. 1972, 2,
117–141. 11. Neo, M.; Kotani, S.; Nakamura, T.; Yamamuro,
T.; Ohtsuki, C.; Kokubo, T.; Bando, Y. A comparative study
of ultrastructures of the interfaces between four kinds of
surface-active ceramic and bone. J. Biomed. Mater. Res.
1992, 26, 1419–1432. 12. Xynos, I.D.; Hukkanen, M.V.;
Batten, J.J.; Buttery, L.D.; Hench, L.L.; Polak, J.M.
Bioglass 45S5 stimulates osteoblast turnover and enhances
bone formation In vitro: Implications and applications for
bone tissue engineering. Calcif. Tissue Int. 2000, 67,
321–329. 13. Jarcho, M. Calcium phosphate ceramics as hard
tissue prosthetics. Clin. Orthop. 1981, 157, 259–278. 14.
Kim, Y.H.; Kim, V.E. Early migration of uncemented porous
coated anatomic femoral component related to aseptic
loosening. Clin. Orthop. 1993, 295, 146–155. 15. Jasty, M.;
Maloney, W.J.; Bragdon, C.R.; O’Connor, D.O.; Haire, T.;
Harris, W.H. The initiation of failure in cemented femoral
components of hip arthroplasties. J. Bone Jt. Surg., Br.
1991, 73, 551–558. 16. Kwong, L.M.; Jasty, M.; Mulroy,
R.D.; Maloney, W.J.; Bragdon, C.; Harris, W.H. The
histology of the radiolucent line. J. Bone Joint Surg. Br.
1992, 74, 67–73. 17. Eriksson, R.A.; Albrektsson, T. The
effect of heat on bone regeneration: An experimental study
in the rabbit using the bone growth chamber. J. Oral
Maxillofac. Surg. 1984, 42, 705–711. 18. Wykman, A.G.M.
Acetabular cement temperature in arthroplasty. Acta Orthop.
Scand. 1992, 63, 543–544. 19. Cochran, D.L.; Schenk, R.K.;
Lussi, A.; Higginbottom, F.L.; Buser, D. Bone response to
unloaded and loaded titanium implants with a sandblasted
and acid-etched surface: A histometric study in the canine
mandible. J. Biomed. Mater. Res. 1998, 40, 1–11. 20. Urban,
R.M.; Jacobs, J.J.; Sumner, D.R.; Peters, C.L.; Voss, F.R.;
Galante, J.O. The bone-implant interface of femoral stems
with non-circumferential porous coating. J. Bone Joint
Surg. Am. 1996, 78, 1068–1081. 21. Klawitter, J.J.;
Hulbert, S.F. Application of porous ceramics for the
attachment of load bearing internal orthopedic
applications. Biomed. Mater. Symp. 1972, 2, 161–229. 22.
Bobyn, J.D.; Pilliar, R.M.; Cameron, H.U.; Weatherly, G.C.
The optimum pore size for the fixation of poroussurfaced
metal implants by the ingrowth of bone. Clin. Orthop. 1980,
150, 263–270. 23. Lee, T.M.; Wang, B.C.; Yang, Y.C.; Chang,
E.; Yang, C.Y. Comparison of plasma-sprayed hydroxyapatite
coatings and hydroxyapatite/tricalcium phosphate composite
coatings: In vivo study. J. Biomed. Mater. Res. 2001, 55,
360–367. 24. Dhert, W.J. Retrieval studies on calcium
phosphatecoated implants. Med. Prog. Technol. 1994, 20,
143– 154. 25. Geesink, R.G.; Hoefnagels, N.H. Six-year
results of hydroxyapatite-coated total hip replacement. J.
Bone Jt. Surg., Br. 1995, 77, 534–547. 26. Bauer, T.W.;
Taylor, S.K.; Jiang, M.; Medendorp, S.V. An indirect
comparison of third-body wear in retrieved
hydroxyapatite-coated, porous, and cemented femoral
components. Clin. Orthop. 1994, 298, 11–18. 27. Bloebaum,
R.D.; Beeks, D.; Dorr, L.D.; Savory, C.G.; DuPont, J.A.;
Hofmann, A.A. Complications with hydroxyapatite particulate
separation in total hip arthroplasty. Clin. Orthop. 1994,
298, 19–26. 28. Noble, P.C.; Alexander, J.W.; Lindahl,
L.J.; Yew, D.T.; Granberry, W.M.; Tullos, H.S. The anatomic
basis of femoral component design. Clin. Orthop. 1988, 235,
148–165. 29. Bobyn, J.D.; Pilliar, R.M.; Cameron, H.U.;
Weatherly, G.C. Osteogenic phenomena across endosteal bone–
implant spaces with porous surfaced intramedullary
implants. Acta Orthop. Scand. 1981, 52, 145–153. 30. Adell,
R. Long-Term Treatment Results. In TissueIntegrated
Prostheses; Bra˚nemark, P.-I., Zarb, G.A., Albrektsson, T.,
Eds.; Quintessence: Chicago, 1985; 175–186. 31.
Raab-Cullen, D.M.; Thiede, M.A.; Petersen, D.N.; Kimmel,
D.B.; Recker, R.R. Mechanical loading stimulates rapid
changes in periosteal gene expression. Calcif. Tissue Int.
1994, 55, 473–478. 32. Szmukler-Moncler, S.; Salama, H.;
Reingewirtz, Y.; Dubruille, J.H. Timing of loading and
effect of micromotion on bone–dental implant interface:
Review of experimental literature. J. Biomed. Mater. Res.
1998, 43, 192–203. 33. Mouzin, O.; Soballe, K.; Bechtold,
J.E. Loading improves anchorage of hydroxyapatite implants
more than titanium implants. J. Biomed. Mater. Res. 2001,
58, 61–68. B
Bone Plates and Screws, Bioabsorbable

11. Bergsma, E.J.; Rozema, F.R.; Bos, R.R.M.; De Brujin,


W.C. Foreign body reactions to resorbable poly ( L-lactide)
bone plates ans screws used for the fixation on unstable
zygomatic fractures. J. Oral Maxillofac. Surg. 1993, 51,
666–670.

12. To¨rma¨la¨, P.; Rokkanen, P.; Laiho, J.; Tamminma¨ki,


M.; Vainionpa¨a¨, S. Material for Osteosynthetic Devices.
U.S. Pat. 4,743,257, May 10, 1988.

13. Maurer, P.; Holweg, S.; Knoll, W.D.; Schubert, J. Study


by finite element method of the mechanical stress of
selected biodegradable osteosynthesis screws in sagittal
ramus osteotomy. Br. J. Oral Maxillofac. Surg. 2002, 40,
76–83.

14. Cutright, D.E.; Hunsuck, E.E.; Beasley, J.D. Fracture


reduction using a biodegradable material, polylactic acid.
J. Oral Surg. 1971, 29, 393–397.

15. Suuronen, R. Comparison of absorbable self-reinforced


polyL-lactide screws and metallic screws in the fixation of
mandibular condyle osteotomies: An experimental study in
sheep. J. Oral Maxillofac. Surg. 1991, 49, 989–995.

16. Suuronen, R.; Pohjonen, T.; Vasenius, J.; Vainionpa¨a¨,


S. Comparison of absorbable self-reinforced multilayer
polyL-lactide and metallic plates for the fixation of
mandibular body osteotomies: An experimental study in
sheep. J. Oral Maxillofac. Surg. 1992, 50, 255–262.

17. Peltoniemi, H.H.; Tulamo, R.M.; Toivonen, T.;


Hallikainen, D.; To¨rma¨la¨, P.; Waris, T. Biodegradable
semirigid plate and miniscrew fixation in experimental
calvarial osteotomies: A comparative study with rigid
titanium fixation. J. Neurosurg. 1999, 90, 910–917.

18. Bos, R.R.M.; Boering, G.; Rozema, F.R.; Leenslag, J.W.;


Pennings, A.J.; Verwey, A.B. Resorbable poly( Llactide)
plates and screws for the fixation of zygomatic fractures.
J. Maxillofac. Surg. 1987, 45, 751–753.

19. Fuente del Campo, A.; Pohjonen, T.; To¨rma¨la¨, P.;


Waris, T. Fixation of horizontal maxillary osteotomies with
biodegradable self-reinforced absorbable polylactide
plates: Preliminary results. Eur. J. Plast. Surg. 1996, 19,
7–9.
20. Suuronen, R.; Haers, P.; Lindqvist, C.; Sailer, H. An
update on bioresorbable plates in maxillofacial surgery:
Invited review. Facial Plast. Surg. 1999, 15, 61–72.

21. Turvey, T.A.; Bell, R.B.; Tejera, T.J.; Proffitt, W.R.


The use of self-reinforced biodegradable bone plates and
screws in orthognathic surgery. J. Oral Maxillofac. Surg.
2002, 60, 59–65. 22. Laine, P.; Kontio, R.; Lindqvist, C.;
Suuronen, R. Are there any complications with bioabsorbable
fixation devices? 10 years of experience in orthognathic
surgery. Int. J. Oral Maxillofac. Surg. accepted 2003. 23.
Kallela, I.; Iizuka, T.; Salo, A.; Lindqvist, C. Lag screw
fixations of anterior mandibular fractures using
biodegradable polylactide screws: Preliminary report. J.
Oral Maxillofac. Surg. 1999, 57, 113–118. 24. Yerit, K.C.;
Enislidis, G.; Schopper, C.; Turhani, D.; Wanschitz, F.;
Wagner, A.; Watzinger, F.; Ewers, R. Fixation of mandibular
fractures with biodegradable plates and screws. Oral Surg.
Oral Med. Oral Pathol. Oral Radiol. Endo. 2002, 94,
294–300. 25. Suuronen, R.; Laine, P.; Salo, A.; Kontio, R.;
Lindqvist, C. Bioabsorbable miniplates in the fixation of
access osteotomies. Int. J. Oral Maxillofac. Surg. 2003,
32, S74. 26. Ninkovic, M.; Rumer, A.; Kofler, A. Preliminary
Clinical Experience with Biodegradable Plate and Screw
Fixation: The European Union Demonstration Project
BMH4-98-3892. In Treatment of Craniofacial Syndromes with
Biodegradable Plates, Screws and Wire [Abstract 6],
Proceedings of the 2nd Symposium on Biodegradable Materials
in Craniofacial Surgery, Saariselka¨, Finland, April 8–11,
1999. 27. Spanio, S.; Ashammakhi, N.; Iloma¨ki, J.;
Va¨limaa, T.; Rainer, C.; Waris, T.; To¨rma¨la¨, P.;
Nincovic, M. Use of new bioabsorbable tacks and a
tackshooter in cranial bone osteofixation saves operative
time. J. Craniofac. Surg. 2002, 13, 693–696. 28. Bos,
R.R.M.; Rozema, F.R.; Boering, G.; Nijenhuis, A.J.;
Pennings, A.J.; Verwey, A.B. Bio-absorbable plates and
screws for internal fixation of mandibular fractures. A
study in six dogs. Int. J. Oral Maxillofac. Surg. 1989, 18,
356–369. 29. Gerlach, K.L.; Krause, H.R.; Eitenmu¨ller, J.
Use of Absorbable Osteosynthesis Material for Mandibular
Fracture Treatment of Dogs. In Biomaterials and Clinical
Applications; Pizzoferrato, A., Marchetti, P.G.,
Ravagliori, A., Lee, A.J.C., Eds.; Elsevier: Amsterdam,
1987; 459–464. 30. To¨rma¨la¨, P.; Vasenius, J.;
Vainionpa¨a¨, S.; Laiho, J.; Pohjonen, T.; Rokkanen, P.
Ultra-high-strength absorbable self-reinforced
polyglycolide (SR-PGA) rods for internal fixation of bone
fractures. In vitro and in vivo study. J. Biomed. Mater.
Res. 1991, 25, 1–22.
Bone Remodeling

6. Smit, T.H.; Burger, E.H. Is BMU-coupling a


strainregulated phenomenon? A finite element analysis. J.
Bone Miner. Res. 2000, 15, 301–307.

7. Smit, T.H.; Huyghe, J.M.; Cowin, S.C. Estimation of the


poro-elastic parameters of cortical bone. J. Biomech. 2002,
35, 829–835.

8. Smit, Th.H.; Burger, E.H.; Huyghe, J.M. A case for


strain-induced fluid flow as regulator of BMU-coupling and
osteonal alignment. J. Bone Miner. Res. 2002, 17,
2021–2029.

9. Burger, E.H.; Klein-Nulend, J.; Smit, T.H.


Strain-derived canalicular fluid flow regulates osteoclast
activity in a remodelling osteon—A proposal. J. Biomech.
2003, 36, 1453–1459.

10. Skerry, T.M.; Bitensky, L.; Chayen, J.; Lanyon, L.E.


Early strain-related changes in enzyme activity in
osteocytes following bone loading in vivo. J. Bone Miner.
Res. 1989, 4, 783–788.

11. Lean, J.M.; Jagger, C.J.; Chambers, T.J.; Chow, J.W.


Increased insulin-like growth factor I mRNA expression in
rat osteocytes in response to mechanical stimulation. Am.
J. Physiol. 1995, 268, E318–E327.

12. Burger, E.H.; Klein-Nulend, J. Mechanotransduction in


bone—Role of the lacunocanalicular network. FASEB J. 1999,
13, S101–S112.

13. Ajubi, N.E.; Klein-Nulend, J.; Nijweide, P.J.;


VrijheidLammers, T.; Alblas, M.J.; Burger, E.H. Pulsating
fluid flow increases prostaglandin production by cultured
chicken osteocytes—A cytoskeleton-dependent process.
Biochem. Biophys. Res. Commun. 1996, 225, 62–68.

14. Klein-Nulend, J.; Van der Plas, A.; Semeins, C.M.;


Ajubi, N.E.; Frangos, J.A.; Nijweide, P.J.; Burger, E.H.
Sensitivity of osteocytes to biomechanical stress in vitro.
FASEB J. 1995, 9, 441–445.

15. Pitsillides, A.A.; Rawlinson, S.C.F.; Suswillo, R.F.L.;


Bourrin, S.; Zaman, G.; Lanyon, L.E. Mechanical
strain-induced NO production by bone cells—A possible role
in adaptive bone (re)modeling. FASEB J. 1995, 9, 1614–1622.
16. Burr, D.B.; Milgrom, C.; Fyhrie, D.; Forwood, M.;
Nyska, M.; Finestone, A.; Hoshaw, S.; Saiag, E.; Simkin, A.
In vivo measurement of human tibial strains during vigorous
activity. Bone 1996, 18, 405–410.

17. Burger, E.H.; Veldhuijzen, J.P. Influence of Mechanical


Factors on Bone Formation, Resorption, and Growth in vitro.
In Bone; Hall, B.K., Ed.; CRC Press: Boca Raton, FL, USA,
1993; Vol. 7, 37–56.

18. Weinbaum, S.; Cowin, S.C.; Zeng, Y. A model for the


excitation of osteocytes by mechanical loading-induced bone
fluid shear stresses. J. Biomech. 1994, 27, 339–360.

19. Cowin, S.C.; Weinbaum, S.; Zeng, Y. A case for bone


canaliculi as the anatomical site of strain generated
potentials. J. Biomech. 1995 , 28, 1281–1296.

20. Turner, C.H.; Forwood, M.R.; Otter, M.W.


Mechanotransduction in bone: Do bone cells act as sensors
of fluid flow? FASEB J. 1994, 8, 875–878.

21. Turner, C.H.; Owan, I.; Takano, Y. Mechanotransduction


in bone: Role of strain rate. Am. J. Physiol. 1995, 269,
E438–E442. 22. Owan, I.; Burr, D.B.; Turner, C.H.; Qui, J.;
Tu, Y.; Onyia, J.E.; Duncan, R.L. Mechanotransduction in
bone: Osteoblasts are more responsive to fluid forces than
mechanical strain. Am. J. Physiol. 1997, 273, C810–C815.
23. Weinbaum, S.; Guo, P.; You, L. A new view of
mechanotransduction and strain amplification in cells with
microvilli and cell processes. Biorheology 2001, 38,
119–142. 24. Klein-Nulend, J.; Semeins, C.M.; Ajubi, N.E.;
Nijweide, P.J.; Burger, E.H. Pulsating fluid flow increases
nitric oxide (NO) synthesis by osteocytes but not
periosteal fibroblasts—Correlation with prostaglandin
upregulation. Biochem. Biophys. Res. Commun. 1995, 217,
640–648. 25. Klein-Nulend, J.; Helfrich, M.H.; Sterck,
J.G.H.; MacPherson, H.; Joldersma, M.; Ralston, S.H.;
Semeins, C.M.; Burger, E.H. Nitric oxide response to shear
stress by human bone cell cultures is endothelial nitric
oxide synthase dependent. Biochem. Biophys. Res. Commun.
1998, 250, 108–114. 26. McAllister, T.N.; Frangos, J.A.
Steady and transient fluid shear stress stimulate NO release
in osteoblasts through distinct biochemical pathways. J.
Bone Miner. Res. 1999, 14, 930–936. 27. Westbroek, I.;
Ajubi, N.E.; Alblas, M.J.; Semeins, C.M.; Klein-Nulend, J.;
Burger, E.H.; Nijweide, P.J. Differential stimulation of
prostaglandin G/H synthase2 in osteocytes and other
osteogenic cells by pulsating fluid flow. Biochem. Biophys.
Res. Commun. 2000, 268, 414–419. 28. Imai, S.; Kaksonen,
M.; Raulo, E.; Kinnunen, T.; Fages, C.; Meng, X.; Lakso,
M.; Rauvala, H. Osteoblast recruitment and bone formation
enhanced by cell-matrix associated heparin-binding
growth-associated molecule (HB-GAM). J. Cell Biol. 1998,
143, 1113–1128. 29. Bronckers, A.L.J.J.; Goei, S.W.; Luo,
G.; Karsenty, G.; D’Souza, R.N.; Lyaruu, D.M.; Burger, E.H.
DNA fragmentation during bone formation in neonatal rodents
assessed by transferase-mediated end labeling. J. Bone
Miner. Res. 1996, 11, 1281–1291. 30. Dimmeler, S.; Zeiher,
A.M. Nitric oxide and apoptosis: Another paradign for the
double-edged role of nitric oxide. Nitric Oxide 1997, 1,
275–281. 31. Rossig, L.; Haendeler, J.; Hermann, C.;
Malchow, P.; Urbich, C.; Zeiher, A.M.; Dimmeler, S. Nitric
oxide down-regulates MKP-3mRNA levels: Involvement in
endothelial cell protection from apoptosis. J. Biol. Chem.
2000, 275, 2552–2557. 32. Zaman, G.; Pitsillides, A.A.;
Rawlinson, S.C.; Suswillo, R.F.; Mosley, J.R.; Cheng, M.Z.
Mechanical strain stimulates nitric oxide production by
rapid activation of endothelial nitric oxide synthase in
osteocytes. J. Bone Miner. Res. 1999, 14, 1123–1131. 33.
Pavalko, F.M.; Gerard, R.L.; Ponik, S.M.; Gallagher, P.J.;
Jin, Y.; Norvell, S.M. Fluid shear stress inhibits
TNF-alpha-induced apoptosis in osteoblasts: A role for fluid
shear stress-induced activation of caspase-3. J. Cell.
Physiol. 2003, 194, 194–205. 34. Elmardi, A.S.;
Katchburian, M.V.; Katchburian, E. Electron microscopy of
developing calvaria reveals images that suggest that
osteoclasts engulf and destroy osteocytes during bone
resorption. Calcif. Tissue Int. 1990, 46, 239–245. 35.
MacIntyre, I.; Zaidi, M.; Alam, A.S.M.T.; Datta, H.K.;
Moonga, B.S.; Lidbury, P.S.; Hecker, M.; Vane, J.R.
Osteoclast inhibition: An action of nitric oxide not
mediated by cyclic GMP. Proc. Natl. Acad. Sci. U. S. A.
1991, 88, 2936–2940. 36. Bentolilla, V.; Boyce, T.M.;
Fyhrie, D.B.; Drumb, R.; Skerry, T.M.; Schaffler, M.B.
Intracortical remodeling in adult rat long bones after
fatigue loading. Bone 1998, 23, 275–281. 37. Verborgt, O.;
Gibson, G.J.; Schaffler, M.B. Loss of osteocyte integrity in
association with microdamage and bone remodeling after
fatigue in vivo. J. Bone Miner. Res. 2000, 15, 60–67. 38.
Burr, D.B.; Martin, R.B. Calculating the probability that
microcracks initiate resorption spaces. J. Biomech. 1993,
26, 613–616. 39. Burr, D.B.; Forwood, M.R.; Fyhrie, D.P.;
Martin, R.B.; Schaffler, M.B.; Turner, C.H. Bone microdamage
and skeletal fragility in osteoporotic and stress
fractures. J. Bone Miner. Res. 1997, 12, 6–15. B
Burn Dressing

17. Cuono, C.B.; Langdon, R.; Birchall, N.; Barttelbort,


S.; McGuire, J. Composite autologous–allogeneic skin
replacement: Development and clinical application. Plast.
Reconstr. Surg. 1987, 80, 626–637.

18. Rheinwald, J.G.; Green, H. Serial cultivation of


strains of human epidermal keratinocytes: The formation of
keratinizing colonies from single cells. Cell 1975, 6, 331.

19. Boyce, S.T. Design principles for composition and


performance of cultured skin substitutes. Burns 2001, 27,
523–533.

20. Navarro, F.A.; Stoner, M.L.; Lee, H.B.; Park, C.S.;


Wood, F.M.; Orgill, D.P. Melanocyte repopulation in
full-thickness wounds using a cell spray apparatus. J. Burn
Care Rehabil. 2001, 22, 41–46.

21. Klasen, H.J. A historical review of the use of silver


in the treatment of burns. II. Renewed interest for silver.
Burns 2000, 26, 131–138.

22. Klasen, H.J. Historical review of the use of silver in


the treatment of burns. I. Early uses. Burns 2000, 26,
117–130. 23. Boeckx, W.; Focquet, M.; Cornelissen, M.;
Nuttin, B. Bacteriological effect of cerium-flamazine cream
in major burns. Burns Incl. Therm. Inj. 1985, 11, 337–342.
24. Yin, H.Q.; Langford, R.; Burrell, R.E. Comparative
evaluation of the antimicrobial activity of ACTICOAT
antimicrobial barrier dressing. J. Burn Care Rehabil. 1999,
20, 195–200. 25. Chu, C.S.; McManus, A.T.; Matylevich,
N.P.; Mason, A.D., Jr.; Pruitt, B.A. Enhanced survival of
autoepidermal–allodermal composite grafts in allosensitized
animals by use of silver–nylon dressings and direct
current. J. Trauma 1995, 39, 273–278. 26. Lawrence, F.C.
Dressings for Burns. In Principles and Practice of Burns
Management; Settle, J.A.D., Ed.; Churchill Livingstone:
London, 1996; 259–269. 27. Winter, G.D. Formation of the
scab and rate of epithelialization of superficial wounds in
the domestic pig. Nature 1962, 193, 293–294. 28. Martin, P.
Wound healing—Aiming for perfect skin regeneration. Science
1997, 276, 75–81.
Calcium Phosphate Ceramics: New
Generation Produced in Japan

7. Sogo, T.; Uchida, S.; Saito, S.; Ikeuchi, M.; Bando, K.


Treatment with Biopex s for calcaneus fracture (in
Japanese). In Transactions of the 3rd Symposium on
Bioactive Paste, The 3rd Symposium on Bioactive Paste,
Tokyo, Nov 30, 2002; Organizing Committee of the 3rd
Symposium on Bioactive Paste: Tokyo, 2003; 6-II-1, 95–97.

8. Takemasa, R.; Yamamoto, H. Clinical application of


calcium phosphate cement for repair of vertebral fractures
in the osteoporotic spine (in Japanese). Orthop. Surg.
Traumatol. 2002, 45 (10), 989–1001.

9. Hashimoto, S.; Kawashima, M.; Sakuragi, T.; Sato, K.


Clinical results of augmentation of femoral neck fractures
and intertrochanteric fractures with bioactive
calcium-phosphate bone cement (in Japanese). Orthop. Surg.
Traumatol. 2002, 45 (10), 975–981.

10. Moriwaki, T.; Tahara, Y.; Takayama, T.; Kodera, M.;


Satomi, K.; Ishi, Y.; Bando, K. Clinical applications of
calcium phosphate paste to revision surgery of hip
arthroplasty (in Japanese). In Transactions of the 3rd
Symposium on Bioactive Paste, The 3rd Symposium on
Bioactive Paste, Tokyo, Nov 30, 2002; Organizing Committee
of the 3rd Symposium on Bioactive Paste: Tokyo, 2003; 4–4,
75–77.

11. Nakano, M.; Hirano, N.; Matsuura, K.; Watanabe, H.;


Kitagawa, H.; Ishihara, H.; Kawaguchi, Y. Percutaneous
transpedicular vertebroplasty with calcium phosphate cement
in the treatment of osteoporotic vertebral compression and
burst fractures. J. Neurosurg. Spine 2002, 97 (3), 287–293.

12. Hoshikawa, A.; Fukui, N.; Fukuda, A.; Sawamura, T.;


Hattori, M.; Nakamura, K.; Oda, H. Quantitative analysis of
the resorption and osteoconduction process of a calcium
phosphate cement and its mechanical effect for screw
fixation. Biomaterials 2003, 24 (27), 4967–4975.

13. Itoh, S.; Kikuchi, M.; Koyama, Y.; Matsumoto, H.N.;


Takakuda, K.; Shinomiya, K.; Tanaka, J. Development of a
novel biomaterial, hydroxyapatite/collagen (HAp/ Col)
composite for medical use. Biomed. Mater. Eng. 2005, 15
(1–2), 29–41. 14. Urist, M.R. Bone: formation by
autoinduction. Science 1965, 150 (698), 893–899. 15. Noshi,
T.; Yoshikawa, T.; Dohi, Y.; Ikeuchi, M.; Horiuchi, K.;
Ichijima, K.; Sugimura, M.; Yonemasu, K.; Ohgushi, H.
Recombinant human bone morphogenetic protein-2 potentiates
the in vivo osteogenic ability of
marrow/hydroxyapatitecomposites. Artif. Organs 2001, 25
(3), 201–208. 16. Takaoka, K.; Nakamura, H.; Yoshikawa, H.;
Masuhara, K.; Tsuda, T.; Ono, K. Ectopic bone induction on
and in porous hydroxyapatite combined with collagen and
bone morphogenetic protein. Clin. Orthop. Relat. Res. 1988,
234, 250–254. 17. Ohgushi, H.; Okumura, M.; Tamai, S.;
Shors, E.C.; Caplan, A.I. Marrow cell induced osteogenesis
in porous hydroxyapatite and tricalcium phosphate: a
comparative histomorphometric study of ectopic bone
formation. J. Biomed. Mater. Res. 1990, 24 (12), 1563–1570.
18. Ohgushi, H.; Dohi, Y.; Yoshikawa, T.; Tamai, S.;
Tabata, S.; Okunaga, K.; Shibuya, T. Osteogenic
differentiation of cultured marrow stromal stem cells on
the surface of bioactive glass ceramics. J. Biomed. Mater.
Res. 1996, 32 (3), 341–348. 19. Caplan, A.I. Mesenchymal
stem cells. J. Orthop. Res. 1991, 9 (5), 641–650. 20.
Maniatopoulos, C.; Sodek, J.; Melcher, A.H. Bone formation
in vitro by stromal cells obtained from marrow of young
adult rats. Cell. Tissue Res. 1988, 254 (2), 317–330. 21.
Yoshikawa, T.; Ohgushi, H.; Tamai, S.; Ichijima, K. Self
setting hydroxyapatite cement as a career for boneforming
cells. Biomed. Mater. Eng. 1996, 6 (5), 345–351. 22.
Yoshikawa, T.; Ohgushi, H.; Akahane, M.; Tamai, S.;
Ichijima, K. Analysis of gene expression in osteogenic
cultured marrow/hydroxyapatite construct implanted at
ectopic sites: a comparison with the osteogenic ability of
cancellous bone. J. Biomed. Mater. Res. 1998, 41 (4),
568–573. 23. Yoshikawa, T.; Ohgushi, H.; Uemura, T.;
Nakajima, H.; Ichijima, K.; Tamai, S.; Tateishi, T. Human
marrow cells-derived cultured bone in porous ceramics.
Biomed. Mater. Eng. 1998, 8 (5–6), 311–320. 24. Ohgushi,
H.; Caplan, A.I. Stem cell technology and bioceramics: from
cell to gene engineering. J. Biomed. Mater. Res. 1999, 48
(6), 913–927. C in Japan
Cardiac Assist Devices

14. Samuels, L.E.; Holmes, E.C.; Thomas, M.P.; Entwistle,


J.C.; Morris, R.J.; Narula, J.; Wechsler, A.S. Management
of acute cardiac failure with mechanical assist: Experience
with the ABIOMED BVS 5000. Ann. Thorac. Surg. 2001, 71
(suppl. 3), S67–S72.

15. Lavee, J.; Paz, Y. Mechanical alternatives to the human


heart: paracoporeal assist systems. Isr. Med. Assoc. J.
2002, 4, 125–130.

16. Poirier, V.L. Worldwide experience with the TCI


HeartMate system: Issues and future perspective. Thorac.
Cardiovasc. Surg. 1999, 47 (suppl. 2), 316–320.

17. Mehta, S.M.; Pae, W.E., Jr.; Rosenberg, G.; Snyder,


A.J.; Weiss, W.J.; Lewis, J.P.; Frank, D.J.; Thompson,
J.J.; Pierce, W.S. The LionHeart LVD-2000: A completely
implanted left ventricular assist device for chronic
circulatory support. Ann. Thorac. Surg. 2001, 71 (suppl.
3), S156–S161.

18. Wheeldon, D.R.; LaForge, D.H.; Lee, J.; Jansen, P.G.;


Jassawalla, J.S.; Portner, P.M. Novacor left ventricular
assist system long-term performance: Comparison of clinical
experience with demonstrated in vitro reliability. ASAIO J.
2002, 48, 546–551.

19. Frazier, O.H.; Rose, E.A.; Oz, M.C.; Dembitsky, W.;


McCarthy, P.; Radovancevic, B.; Poirier, V.L.; Dasse, K.A.
Multicenter clinical evaluation of the HeartMate vented
electric left ventricular assist system in patients
awaiting heart transplantation. J. Thorac. Cardiovasc.
Surg. 2001, 122, 1186–1195.

20. Rose, E.A.; Gelijns, A.C.; Moskowitz, A.J.; Heitjan,


D.F.; Stevenson, L.W.; Dembitsky, W.; Long, J.W.; Ascheim,
D.D.; Tierney, A.R.; Levitan, R.G.; Watson, J.T.; Meier,
P.; Ronan, N.S.; Shapiro, P.A.; Lazar, R.M.; Miller, L.W.;
Gupta, L.; Frazier, O.H.; Desvigne-Nickens, P.; Oz, M.C.;
Poirier, V.L. Randomized evaluation of mechanical
assistance for the treatment of congestive heart failure
(REMATCH) study group. Long-term mechanical left
ventricular assistance for end-stage heart failure. N.
Engl. J. Med. 2001, 345, 1435–1443.

21. Nose, Y.; Yoshikawa, M.; Murabayashi, S.; Takano, T.


Development of rotary blood pump technology: Past, present,
and future. Artif. Organs 2000, 24, 412–420.
22. Nose, Y.; Kawahito, K.; Nakazawa, T. Can we develop a
nonpulsatile permanent rotary blood pump? Yes, we can.
Artif. Organs 1996, 20, 467–474.

23. Lynch, M.F.; Peterson, D.; Baker, V. Centrifugal blood


pumping for open heart surgery. Minn. Med. 1978, 61,
536–538.

24. Noon, G.P.; Lafuente, J.A.; Irwin, S. Acute and


temporary ventricular support with BioMedicus centrifugal
pump. Ann. Thorac. Surg. 1999, 68, 650–654.

25. Noon, G.P.; Lafuente, J.A.; Irwin, S. Acute and


temporary ventricular support with BioMedicus centrifugal
pump. Ann. Thorac. Surg. 1999, 68, S133–S138.

26. Pennington, D.G.; Swartz, M.T.; Lohmann, D.P.; McBride,


L.R. Cardiac assist devices. Surg. Clin. North Am. 1998,
78, 691–704.

27. Curtis, J.J.; Walls, J.T.; Demmy, T.L.; Boley, T.M.;


Schmaltz, R.A.; Goss, C.F.; Wagner-Mann, C.C. Clinical
experience with the Sarns centrifugal pump. Artif. Organs
1993, 17, 630–633. 28. Tansley, G.; Vidakovic, S.; Reizes,
J. Fluid dynamic characteristics of the VentrAssist rotary
blood pump. Artif. Organs 2000, 24, 483–487. 29. Allaire,
P.E.; Wood, H.G.; Awad, R.S.; Olsen, D.B. Blood flow in a
continuous flow ventricular assist device. Artif. Organs
1999, 23, 769–773. 30. Curtis, J.J.; Wagner-Mann, C.
Extracorporeal Support: Centrifugal Pumps. In Cardiac
Assist Devices; Goldstein, D.J., Oz, M.C., Eds.; Futura
Publishing Company: Armonk, NY, 2000; 215–233. 31.
Mizuguchi, K.; Damm, G.; Benkowsky, R.; Aber, G.; Bacak,
J.; Svjkovsky, P.; Glueck, J.; Takatani, S.; Nose, Y.;
Noon, G.P.; DeBakey, M.E. Development of an axial flow
ventricular assist device: In vitro and in vivo evaluation.
Artif. Organs 1995, 19, 653–659. 32. Jarvik, R.K. System
considerations favoring rotary artificial hearts with blood
immersed bearings. Artif. Organs 1995, 19, 565–570. 33.
Macris, M.P.; Parnis, S.M.; Frazier, O.H.; Fuqua, J.M.,
Jr.; Jarvik, R.K. Development of an implantable ventricular
assist system. Ann. Thorac. Surg. 1997, 63, 367–370. 34.
Frazier, O.H.; Myers, T.J.; Gregoric, I.D.; Khan, T.;
Delgado, R.; Croitoru, M.; Miller, K.; Jarvik, R.; Westaby,
S. Initial clinical experience with the Jarvik 2000
implantable axial-flow flow ventricular assist system.
Circulation 2002, 105, 2855–2860. 35. Wernicke, J.T.;
Meier, D.; Mizuguchi, K.; Damm, G.; Aber, G.; Benkowski,
R.; Nose, Y.; Noon, G.P.; DeBakey, M.E. A fluid dynamic
analysis using flow visualization of the Baylor/NASA
implantable axial flow pump for design improvement. Artif.
Organs 1995, 19, 161–177. 36. Wieselthaler, G.M.; Schima,
H.; Hiesmayr, M.; Pacher, R.; Laufer, G.; Noon, G.P.;
DeBakey, M.E.; Wolner, E. First clinical experience with
the DeBakey VAD continuous-axial-flow pump for bridge to
transplantation. Circulation 2000, 101, 356–359. 37. Noon,
G.P.; Morley, D.; Irwin, S.; Benkowski, R. Development and
clinical application of the MicroMed DeBakey VAD. Curr.
Opin. Cardiol. 2000, 15, 166–171. 38. Noon, G.P.; Morley,
D.N.; Irwin, S.; Abdelsayed, S.V.; Benkowski, R.J.; Lynch,
B.E. Clinical experience with the MicroMed DeBakey
ventricular assist device. Ann. Thorac. Surg. 2001, 71
(suppl. 3), S133–S138. 39. Arabia, F.A.; Copeland, J.G.;
Smith, R.G.; Banchy, M.; Foy, B.; Kormos, R.; Tector, A.;
Long, J.; Dembitsky, W.; Carrier, M.; Keon, W.; Pavie, A.;
Duveau, D. CardioWest total artificial heart: A
retrospective controlled study. Artif. Organs 1999, 23,
204–207. 40. DeVries, W.C. The permanent artificial heart.
four case reports. JAMA 1988, 259, 849–859. 41. Weiss,
W.J.; Rosenberg, G.; Snyder, A.J.; Pierce, W.S.; Pae, W.E.;
Kuroda, H.; Rawhouser, M.A.; Felder, G.; Reibson, J.D.;
Cleary, T.J.; Ford, S.K.; Marlotte, J.A.; Nazarian, R.A.;
Hicks, D.L. Steady state hemodynamic and energetic
characterization of the Penn State/3M health care total
artificial heart. ASAIO J. 1999, 45, 189–193. 42. Kung,
R.T.; Yu, L.S.; Ochs, B.D.; Parnis, S.M.; Macris, M.P.;
Frazier, O.H. Progress in the development of the ABIOMED
total artificial heart. ASAIO J. 1995, 41, M245–M248. 43.
Dowling, R.D.; Gray, L.A.; Etoch, S.W.; Laks, H.; Marelli,
D.; Samuels, L.; Entwistle, J.; Couper, G.; Vlahakes, G.J.;
Frazier, O.H. The AbioCor implantable replacement heart.
Ann. Thorac. Surg. 2003, 75, S93–S99. 44. Rosenberg, G.;
Phillips, W.M.; Landis, D.L.; Pierce, W.S. Design and
evaluation of the Pennsylvania State University mock
circulatory system. ASAIO J. 1981, 4, 41–49. 45. Rosenberg,
G. Artificial Heart and Circulatory Assist Devices. In The
Biomedical Engineering Handbook; Bronzino, J.D., Ed.; CRC
Press, Inc.: Boca Raton, FL, 1995; 1839–1870. 46. Baldwin,
J.T.; Deutsch, S.; Geselowitz, D.B.; Tarbell, J.M. LDA
measurements of mean velocity and reynolds stress fields
within an artificial heart ventricle. J. Biomech. Eng. 1994,
116, 190–200. 47. Hochareon, P.; Manning, K.B.; Fontaine,
A.A.; Deutsch, S.; Tarbell, J.M. In Development of High
Resolution Particle Image Velocimetry for Use in Artificial
Heart Research, Second Joint Meeting of the IEEE-EMBS and
BMES, Houston, TX, Oct. 2002. 48. Manning, K.B.; Miller,
G.E. Flow through an outlet cannula of a rotary ventricular
assist device. Artif. Organs 2002, 26, 714–723. 49. Lee,
J.; Miller, P.J.; Chen, H.; Conley, M.G.; Carpenter, J.L.;
Wihera, J.C.; Jassawalla, J.S.; Portner, P.M. Reliability
model from the in vitro durability tests of a left
ventricular assist system. ASAIO J. 1999, 45, 595–601. 50.
Ducko, C.T.; McGregor, M.L.; Rosenberg, G.; Pierce, W.S.
The effect of valve type and drive line dP/dt on hemolysis
in the pneumatic ventricular assist device. Artif. Organs
1994, 8, 454–460. 51. Wampler, R.K.; Moise, J.C.; Frazier,
O.H.; Olsen, D.B. In vivo evaluation of a peripheral
vascular access axial flow blood pump. ASAIO Trans. 1988,
34, 450–454. 52. Weiss, W.J.; Rosenberg, G.; Snyder, A.J.;
Donachy, J.; Reibson, J.; Kawaguchi, O.; Sapirstein, J.S.;
Pae, W.E.; Pierce, W.S. A completely implanted left
ventricular assist device: Chronic in vivo testing. ASAIO
J. 1993, 39, M427–M432. 53. Mehta, S.M.; Aufiero, T.X.; Pae,
W.E., Jr.; Miller, C.A.; Pierce, W.S. Results of mechanical
ventricular assistance for the treatment of post cardiotomy
cardiogenic shock. ASAIO J. 1996, 42, 211–218. C
Cardiac Bioelectricity

13. Makielski, J.; Fozzard, H.A. Ion Channels and Cardiac


Arrhythmias in Heart Disease. In Handbook of Physiology,
Section 2: The Cardiovascular System, Vol. 1: The Heart;
Page, E., Fozzard, H.A., Solaro, J., Eds.; Oxford: New
York, 2001; 709–740.

14. Noble, D. Ionic Mechanisms of Cardiac Electrical


Activity. In Cardiac Electrophysiology: From Cell to
Bedside, 2nd Ed.; Zipes, D.P., Jalife, J., Eds.; Saunders:
Philadelphia, 1995; 305–313.

15. Rudy, Y. The Cardiac Ventricular Action Potential. In


Handbook of Physiology, Section 2: The Cardiovascular
System, Vol. 1: The Heart; Page, E., Fozzard, H.A., Solaro,
J., Eds.; Oxford: New York, 2001; 531–547. 16. Boyett,
M.R.; Hono, H.; Kodama, I. The sinoatrial node, a
heterogeneous pacemaker structure. Cardiovasc. Res. 2000,
47, 658–687. 17. Accili, E.A.; Proenza, C.; Baruscotti, M.;
DiFrancesco, D. From funny current to HCN channels: 20
years of excitation. News Physiol. Sci. 2002, 17, 32–37.
18. Antzelevitch, C.; Dumaine, R. Electrical Heterogeneity
in the Heart: Physiological, Pharmacological, and Clinical
Implications. In Handbook of Physiology, Section 2: The
Cardiovascular System, Vol. 1: The Heart; Page, E.,
Fozzard, H.A., Solaro, J., Eds.; Oxford: New York, 2001;
654–692.
Cardiac Catheters

3. Cournand, A. Cardiac catheterization: Development of the


technique, its contributions to experimental medicine, and
its initial application in man. Acta Med. Scand., Suppl.
1975, 579, 1.

4. Forssmann, W. Experiments on Myself: Memoirs of a


Surgeon in Germany; St. Martin’s Press: New York, 1974.

5. Cournand, A.F.; Ranges, H.S. Catheterization of the


right auricle in man. Proc. Soc. Exp. Biol. Med. 1941, 45,
462.

6. Cournand, A.F.; Riley, R.L.; Breed, E.S.; Baldwin, E.F.;


Richards, D.W. Measurement of cardiac output in man using
the technique of catheterization of the right auricle. J.
Clin. Invest. 1945, 24, 106.

7. Brannon, E.S.; Weens, H.S.; Warren, J.V. Atrial septal


defect: Study of hemodynamics by the technique of right
heart catheterization. Am. J. Med. Sci. 1945, 210, 480.

8. Hellems, H.K.; Haynes, F.W.; Dexter, L. Pulmonary


‘‘capillary’’ pressure in man. J. Appl. Physiol. 1949, 2,
24.

9. Zimmerman, H.A.; Scot, R.W.; Becker, N.D.


Catheterization of the left side of the heart in man.
Circulation 1950, 1, 357.

10. Seldinger, S.J. Catheter replacement of the needle in


percutaneous arteriography. Acta Radiol. 1953, 39, 368.

11. Sones, F.M., Jr.; Shirey, E.K.; Proudfit, W.L.; Wescott,


R.N. Cine coronary arteriography. Circulation 1959, 20,
773.

12. Sones, F.M., Jr.; Shirey, E.K. Cine coronary


arteriography. Mod. Concepts Cardiovasc. Dis. 1962, 31,
735.

13. Kohchi, K.; Takebayashi, S.; Block, P.C.; Hiroki, T.;


Nobuyoshi, M. Arterial changes after percutaneous
transluminal coronary angioplasty: Results at autopsy. J.
Am. Coll. Cardiol. 1987, 10, 592–599.

14. Dotter, C.T.; Judkins, M.P. Transluminal treatment of


arteriosclerotic obstruction: Description of a new
technique and preliminary report of its application.
Circulation 1964, 30, 654.

15. Swan, H.J.C.; Ganz, W.; Forrester, J.; et al.


Catheterization of the heart in man with use of a flow
directed balloon-tipped catheter. N. Engl. J. Med. 1970,
283, 447.

16. Gru¨ntzig, A.; et al. Coronary transluminal


angioplasty. Circulation 1977, 56 (Suppl II), 319. (abst).

17. Gru¨ntzig, A.; Senning, A.; Siegenthaler, W.E.


Nonoperative dilatation of coronary artery stenosis;
percutaneous transluminal coronary angioplasty. N. Engl. J.
Med. 1979, 301, 61. 18. Grines, C.L.; Browne, K.F.; Marco,
J.; et al. A comparison of immediate angioplasty with
thrombolytic therapy for acute myocardial infarction. N.
Engl. J. Med. 1998, 328, 673. 19. Tiefenbrunn, A.J.;
Chandra, N.C.; French, W.J.; et al.Clinical experience with
primary percutaneous transluminal coronary angioplasty
compared with alteplase (recombinant tissue-type
plasminogen activator) in patients with acute myocardial
infarction: A report from the Second National Registry of
Myocardial Infarction (NRMI-2). J. Am. Coll. Cardiol. 1998,
31, 1240. 20. Botnar, R.M.; Stuber, M.; Danias, P.G.; et
al. Improved coronary artery definition with T2-weighted,
free-breathing, three dimensional coronary MRA. Circulation
1999, 99, 3139. 21. Achenbach, S.; Moshage, W.; Ropers, D.;
et al. Value of electron-beam computed tomography for the
noninvasive detection of high-grade coronary-artery
stenosis and occlusions. N. Engl. J. Med. 1998, 339, 1964.
22. Noto, T.J.; Johnson, L.W.; Krone, R.; et al. Cardiac
catheterization 1990: A report of the registry of the
Society for Cardiac Angiography and Interventions. Cathet.
Cardiovasc. Diagn. 1991, 24, 75. 23. Katzen, B.T.
Interventional radiology: Past, present and future. Med.
Mundi 2002, 40 (3), 151–155. 24. Payne, M. Charles Theodore
Dotter: The father of intervention. Texas Heart Inst. J.
2001, 28, 28–38. 25. Anonymous Portraits in radiology: T.
Charles, Dotter, MD. Appl. Radiol. 1981, 10 (28), 116.
(Jan–Feb). 26. Rosch, J.; Abrams, H.L.; Cook, W. Memorials:
Charles Theodore Dotter, 1920–1985. Am. J. Roentgenol.
1985, 144, 1321–1323. 27. Physics, Techniques and
Procedures. In The Encyclopaedia of Medical Imaging;
Pettersson, H., Ed.; Isis Medical Medica, 1998; Vol 1. s.v.
‘‘Catheter’’. 28. The Cardiac Catheterization Handbook, 3rd
Ed.; Kern, M.J., Ed.; Mosby, Inc.: St. Louis, 1999. 29.
Invasive Cardiology: Principles and Techniques; Bashore,
T.M., Ed.; B.C. Decker: Toronto, 1990. 30. Grossman’s
Cardiac Catheterization, Angiography, and Intervention, 6th
Ed.; Baim, D.S., Grossman, W., Eds.; Lippincott, Williams,
and Wilkins: Philadelphia, 2000. 31. Diagnostic and
Therapeutic Cardiac Catheterization, 3rd Ed.; Pepine, C.J.,
Ed.; Williams and Wilkins: Baltimore, 1998. C
Cardiac Elastography, Full-Field
Development

15. McDicken, W.N.; Sutherland, G.R.; Moran, C.M.; Gordon,


L.N. Colour Doppler velocity imaging of the myocardium.
Ultrasound Med. Biol. 1992, 18 (6–7), 651–654.

16. Sutherland, G.R.; Stewart, M.J.; Groundstroem, K.W.;


Moran, C.M.; Fleming, A.; Guell-Peris, F.J.; Riemersma,
R.A.; Fenn, L.N.; Fox, K.A.; McDicken, W.N. Color Doppler
myocardial imaging: a new technique for the assessment of
myocardial function. J. Am. Soc. Echocardiogr. 1994, 7 (5),
441–458.

17. Fleming, A.D.; Xia, A.; McDicken, W.N.; Sutherland,


G.R.; Fenn, L. Myocardial velocity gradients detected by
Doppler imaging. Br. J. Radiol. 1994, 67 (799), 679–688.

18. Kanai, H.; Hasegawa, H.; Chubachi, N.; Koiwa, Y.;


Tanaka, M. Noninvasive evaluation of local myocardial
thickening and its color-coded imaging. IEEE Trans.
Ultrason. Ferroelectric. Freq. Control 1997, 44, 752–768.

19. Hashimoto, I.; Li, X.; Hejmadi Bhat, A.; Jones, M.;
Zetts, A.D.; Sahn, D.J. Myocardial strain rate is a
superior method for evaluation of left ventricular
subendocardial function compared with tissue Doppler
imaging. J. Am. Coll. Cardiol. 2003, 42 (9), 1574–1583.

20. Urheim, S.; Edvardsen, T.; Torp, H.; Angelsen, B.;


Smiseth, O.A. Myocardial strain by Doppler
echocardiography. Validation of a new method to quantify
regional myocardial function. Circulation 2000, 102 (10),
1158–1164.

21. Prinzen, F.W.; Hunter, W.C.; Wyman, B.T.; McVeigh, E.R.


Mapping of regional myocardial strain and work during
ventricular pacing: experimental study using magnetic
resonance imaging tagging. J. Am. Coll. Cardiol. 1999, 33
(6), 1735–1742.

22. Zabalgoitia, M.; Rahman, S.N.; Haley, W.E.; Yarows, S.;


Krause, L.; Anderson, L.C.; Oraby, M.A.; Amarena, J. Effect
of regression of left ventricular hypertrophy from systemic
hypertension on systolic function assessed by midwall
shortening (HOT echocardiographic study). Am. J. Cardiol.
2001, 88 (5), 521–525. 23. Ophir, J.; Cespedes, I.;
Ponnekanti, H.; Yazdi, Y.; Li, X. Elastography: a
quantitative method for imaging the elasticity of
biological tissues. Ultrason. Imaging 1991, 13 (2),
111–134. 24. O’Donnell, M.; Skovoroda, A.R.; Shapo, B.M.;
Emelianov, S.Y. Internal displacement and strain imaging
using ultrasonic speckle tracking. IEEE Trans. Ultrason.
Ferroelec. Freq. Control 1994, 41 (3), 314–325. 25.
Varghese, T.; Ophir, J.; Konofagou, E.; Kallel, F.;
Righetti, R. Tradeoffs in elastographic imaging. Ultrason.
Imaging 2001, 23 (4), 216–248. 26. Konofagou, E.E.;
D’Hooge, J.; Ophir, J. Myocardial elastography—a
feasibility study in vivo. Ultrasound Med. Biol. 2002, 4,
475–482. 27. Varghese, T.; Zagzebski, J.A.; Rahko, P.;
Breburda, C.S. Ultrasonic imaging of myocardial strain
using cardiac elastography. Ultrason. Imaging 2003 , 25
(1), 1–16. 28. Varghese, T.; Breburda, C.S.; Zagzebski,
J.A.; Rahko, P. Method and Apparatus for Cardiac
Elastography. U.S. Patent 6,749,571, United States:
Wisconsin Alumni Research Foundation, University of
Wisconsin– Madison, 2004. 29. Konofagou, E.; Ophir, J. A
new elastographic method for estimation and imaging of
lateral displacements, lateral strains, corrected axial
strains, and Poisson’s ratios in tissues. Ultrasound Med.
Biol. 1998, 24 (8), 1183–1199. 30. Techavipoo, U.; Chen,
Q.; Varghese, T.; Zagzebski, J.A. Estimation of
displacement vectors and strain tensors in elastography
using angular insonifications. IEEE Trans. Med. Imaging
2004, 23 (12), 1479–1489. FURTHER READING Konofagou, E.E.;
D’hooge, J.; Ophir, J. Myocardial elastography—a
feasibility study in vivo. Ultrasound Med. Biol. 2002, 28
(4), 475–482. Varghese, T.; Zagzebski, J.A.; Rahko, P.;
Breburda, C.S. Ultrasonic imaging of myocardial strain
using cardiac elastography. Ultrason Imaging 2003, 25 (1),
1–16. C
Cardiac Output, Basic Measurement
Principles

Fig. 15 The relationship between impedance cardiac output

(solid line) and dye-cardiac output in exercising subjects.

Source: Redrawn by permission from American Physiological

Society (see Ref. [21]).

Fig. 16 The relationship between dye-cardiac output and

impedance cardiac output (corrected) in subjects exercising

on a bicycle ergometer.

Source: Redrawn by permission from American

Physiological Society (see Ref. [21]). Fig. 17 The


correction factor (F ) reported by Mohapatra et al. [22]
for correcting impedance cardiac output values. 2. Altman,
P.L.; et al. Handbook of Circulation, NRC. National Academy
of Science, Contract AF 33 (616)-972, 1960. 3. Fick, A.
Uber die messung des blutstroms in den hertzventrikle.
Verhandl. d. phys. Med. Ges zu Wurzburg 1870, 2, XVI. [See
Hoff, 1965]. 4. Kovach, J.C.; Paulos, P.; Arabadjis, C. J.
Thorac. Surg. 1955, 29, 552–554. 5. Stewart, G.N. The
output of the heart in dogs. Am. J. Physiol. 1921, 57,
27–50. 6. Connolly, D.C.; Wood, E.H. Simultaneous
Measurement of the appearance and disappearance of T-1824
(Evans Blue) in blood and tissue after intravenous
injection in man. J. Appl. Physiol. 1954, 7, 73–83. 7. Fox,
I.J. Indicators and detectors for circulatory dilution
studies and their application to organ or regional bloodflow
determination. Circ. Res. 1962, X (3), 447–471. 8. Wheeler,
H.O.; Cranston, W.I.; Meltzer, J.I. Hepatic uptake and
biliary excretion of indocyanine green in the dog. Proc.
Soc. Exp. Biol. Med. 1958, 99, 11–14. 9. Taylor, S.H.;
Thorp, J.M. Properties and biological behavior of coomassie
blue. Brit. Heart J. 1959, 21, 492–496. 10. Swan, H.; Ganz,
W.; Forrester, X. Catherterization of the heart in man with
use of a flow-through balloontipped catheter. N. Engl. J.
Med. 1970, 283, 447–451. 11. Fegler, G. Measurement of
cardiac output in anesthetized animals by a thermodilution
method. Q. J. Exp. Physiol. 1954, 39, 153–164. 12. Goodyer,
V.N.; Huvos, A.; Eckhardt, W.F.; Ostberg, R.H. Thermal
dilution curves in the intact animal. Circ. Res. 1959, 7,
432–441. 13. Ganz, W.; Donoso, R.; Marcus, H.; Forester,
J.S.; Swan, H.J.C. A new technique for measurement of
cardiac output by thermodilution in man. Am. J. Cardiol.
1971, 27, 392–396. 14. Forrester, J.S.; Ganz, W.; Diamond,
G.; McHugh, T.; Chanette, D.W.; Swan, H.J.C. Thermodilution
cardiac output determination with a single flow-directed
catheter. Am. Heart J. 1972, 83, 306–311. 15. Weissel,
R.D.; Berger, R.L.; Hechtman, H.B. Measurement of cardiac
output by thermodilution. N. Engl. J. Med. 1975, 292,
682–684. 16. Grubbs, D.S.; Geddes, L.A.; Voorhees, W.D.
Rightside cardiac output determined with a newly developed
catheter-tip resistivity probe using saline indicator. Jpn.
Heart J. 1984, 25 (1), 105–111. 17. Geddes, L.A.; Babbs,
C.F. A new technique for repeated measurements of cardiac
output during cardiopulmonary resuscitation. Crit. Care
Med. 1980, 8, 131–133. 18. Chinard, F.P.; Enns, T.; Nolan,
M.F. Indicator-dilution studies with diffusible indicators.
Circ. Res. 1962, 10, 473–490. 19. Kubicek, W.G.; Kanegris,
J.N.; Patterson, R.P.; Witsoe, D.A.; Mattson, R.H.
Development and evaluation of an impedance cardiac output
system. Aerosp. Med. 1966, 37, 1208–1212. 20. Geddes, L.A.;
Baker, L.E. Principles of Applied Biomedical
Intrumentation; Wiley: New York, 1975. 21. Denniston, J.;
Maher, J.T.; Reeves, F.T.; Cruz, J.C.; Gymerman, A.;
Grover, R.F. Measurement of cardiac output by electrical
impedance at rest and during exercise. J. Appl. Physiol.
1976, 40, 91–95. 22. Mohaptra, S.N.; Costeloe, K.L.; Hill,
D.W. Blood resistivity and its implication for calculation
of cardiac output by the thoracic electrical impedance
technique. Intens. Care Med. 1977, 3, 1–5. 23. Aldrete, A.;
Brown, C.; Daily, J.; et al. Pacemaker malfunction due to
microcurrent injection from a bioimpedance noninvasive
cardiac output monitor. J. Clin. Monit. 1995, 11 (2),
131–133. C
Cardiac Pacemaker

1. Elmqvist, R.; Senning, A. In Implantable Pacemaker for


the Heart, Medical Electronics: Proceedings of the Second
International Conference on Medical Electronics, Paris,
June 24–27, 1959; Smyth, C.M., Ed.

2. Kusumoto, F.M.; Goldschlager, N. Device therapy for


cardiac arrhythmias. JAMA 2002, 287, 1848–1852.

3. Kusumoto, F.M. Arrhythmias. In Cardiovascular


Pathophysiology, 1st Ed.; Kusumoto, F.M., Ed.; Blackwell
Science: Malden, MA, 1999; 54.

4. Keith, A.; Flack, M.W. The form and nature of the


muscular connections between the primary divisions of the
vertebrate heart. J. Anat. Physiol. Lond. 1907, 41,
172–189.

5. Holden, W.; McAnulty, J.H.; Rahimtoola, S.H.


Characterisation of heart rate response to exercise in the
sick sinus syndrome. Br. Heart J. 1978, 40, 923–930.

6. Mond, H.G. Pacing Leads. In Cardiac Pacing for the


Clinician, 1st Ed.; Kusumoto, F.M., Goldschlager, N.F.,
Eds.; Lippincott Williams and Wilkins: Philadelphia, PA,
2001; 20–24.

7. Lloyd, M.A.; Hayes, D.L.; Friedman, P.A. Clinically


Relevant Basics of Pacing and Defibrillation. In Cardiac
Pacing and Defibrillation: A Clinical Approach, 1st Ed.;
Lloyd, M.A., Hayes, D.L., Friedman, P.A., Eds.; Futuraco
Publishing: Armonk, NY, 2000; 11–12.

8. Tyers, G.F.; Brownlee, R.R. Power pulse generators,


electrodes, and longevity. Prog. Cardiovasc. Dis. 1981, 23,
421–434.

9. Wiens, R.; Lafia, P.; Marder, C.M.; Evans, R.G.; Kennedy,


H.L. Chronotropic incompetence in clinical exercise
testing. Am. J. Cardiol. 1984, 54, 74–82.

10. Gregaratos, G.; Abrams, J.; Epstein, A.E.; Freedman,


R.A.; Hayes, D.L.; Hlatky, M.A.; Kerber, R.E.; Naccarelli,
G.V.; Schoenfeld, M.H.; Silka, M.J.; Winters, S.L.;
Gibbons, R.J.; Antman, E.M.; Alpert, J.S.; Hiratzka, L.F.;
Faxon, D.P.; Jacobs, A.K.; Fuster, V.; Smith, S.C.
ACC/AHA/NASPE 2002 guidelines for implantation of cardiac
pacemakers and antiarrhythmia devices: Summary article.
Circulation 2002, 106, 2145–2161. 11. Dreifus, L.S.;
Michelson, E.L.; Kaplinsky, E. Bradyarrhythmias: Clinical
significance and management. J. Am. Coll. Cardiol. 1983, 1,
327–338. 12. Lamas, G.A.; Pashos, C.L.; Normand, S.L.T.;
McNeil, B. Permanent pacemaker selection and subsequent
survival in elderly medicare pacemaker recipients.
Circulation 1995, 91, 1063–1069. 13. Kusumoto, F.M.;
Goldschlager, N.F. Cardiac pacing. N. Engl. J. Med. 1996,
334, 89–97. 14. Connolly, S.J.; Sheldon, R.; Roberts, R.;
Gent, M. The North American vasovagal pacemaker study: A
randomized trial of permanent cardiac pacing for the
prevention of vasovagal syncope. J. Am. Coll. Cardiol.
1999, 33, 16–20. 15. Sutton, R.; Brignole, M.; Menozzi, C.;
Raviele, A.; Alboni, P.; Giani, P.; Moya, A. Dual-chamber
pacing in the treatment of neurally mediated tilt-positive
cardioinhibitory syncope. Circulation 2000, 102, 294–299.
16. Kenny, R.A.; Richardson, D.A.; Steen, N.; Bexton, R.S.;
Shaw, F.E.; Bond, J. Carotid Sinus Syndrome: A modifiable
risk factor for nonaccidental falls in older adults (SAFE
PACE). J. Am. Coll. Cardiol. 2001, 38, 1491–1496. 17.
Cazeau, S.; Leclerq, C.; Lavergne, T.; Walker, S.; Varma,
C.; Linde, C.; Garrigue, S.; Kappenberger, L.; Haywood,
G.A.; Santini, M.; Bailleul, C.; Daubert, J.C. Effects of
multisite biventricular pacing in patients with heart
failure and intraventricular conduction delay. New Engl. J.
Med. 2001, 344, 873–880. 18. Abraham, W.T.; Fisher, W.G.;
Smith, A.L.; Delurgio, D.B.; Leon, A.R.; Loh, E.; Kocovic,
D.Z.; Packer, M.; Clavell, A.L.; Hayes, D.L.; Ellestad, M.;
Trupp, R.J.; Underwood, J.; Pickering, F.; Truex, C.;
McAtee, P.; Messenger, J. Cardiac resynchronization in
chronic heart failure. New Engl. J. Med. 2002, 346,
1845–1852. 19. Saksena, S. In DAPPAF Trial Data, Oral
Presentation at the American College of Cardiology Meeting,
Orlando, FL, March 18, 2001. C
Cardiac Patch Engineering

1. Cotran, R.; Kumar, V.; Collins, T. The Heart. In


Pathological Basis of Disease, 6th Ed.; W.B. Saunders Co.:
Philadelphia, PA, 1999; 543–599.

2. Chiu, R.C.-J. Therapeutic cardiac angiogenesis and


myogenesis: The promises and challenges on a new frontier.
J. Thorac. Cardiovasc. Surg. 2001, 122 (5), 851–852.

3. Marelli, D.; Desrosiers, C.; El-Alfy, M.; Kao, R.L.;


Chiu, R.C. Cell transplantation for myocardial repair: An
experimental approach. Cell Transplant. 1992, (1), 383–390.
4. Taylor, D.A.; Atkins, B.Z.; Hungspreugs, P.; Jones,
T.R.; Reedy, M.C.; Hutchenson, K.A.; Glower, D.D.; Kraus,
W.E. Delivery of primary autologous skeletal myoblasts into
rabbit heart by coronary infusion: A potential approach to
myocardial repair. Proc. Assoc. Am. Physicians 1997, 109,
245–253. 5. Soonpaa, M.H.; Koh, G.Y.; Klug, M.G.; Field,
L.G. Formation of nascent intercalated disks between
grafted cardiomyocytes and host myocardium. Science 1994,
264, 98–101. 6. Koh, G.Y.; Soonpaa, M.H.; Klug, M.G.;
Field, L. Long-term survival of AT-1 cardiomyocyte grafts
in syngenic myocardium. Am. J. Physiol. 1993, 264,
H1727–H1733. (Heart Circ Physiol 33). 7. Klung, M.;
Soonpaa, M.; Koh, G.; Field, L. Genetically selected
cardiomyocytes from differentiating embryonic stem cells
form stable intracardiac grafts. J. Clin. Invest. 1996, 98,
216–224. 8. Taylor, D.A.; Atkins, B.Z.; Hungspreugs, P.;
Jones, T.R.; Reddy, M.C.; Hutcheson, K.A.; Glower, D.D.;
Kraus, W.E. Regenerating functional myocardium: Improved
performance after skeletal myoblast transplantation. Nat.
Med. 1998, 4 (8), 929–933. 9. Li, R.-K.; Weisel, R.D.;
Mickle, D.A.; Jia, Z-Q.; Kim, E.J.; Sakai, T.; Tomita, S.;
Schwartz, L.; Iwanocko, M.; Husain, M.; Cusimaro, R.J.;
Burns, R.J.; Yau, T.M. Autologous porcine heart cell
transplantation improved heart function after a myocardial
infarction. J. Thorac. Cardiovasc. Surg. 2000, 119, 62–68.
10. Sakai, T.; Li, R.-K.; Weisel, R.D.; Mickle, D.A.; Kim,
E.J.; Tomita, S.; Jia, Z.-Q.; Yau, T.M. Autologous heart
cell transplantation improves function after myocardial
injury. Ann. Thorac. Surg. 1999, 68, 2074–2081. 11.
Reincke, H.; Zhang, M.; Bartosek, T.; Murry, C.E. Survival,
integration, and differentiation of cardiomyocyte grafts. A
study in normal and injured rat hearts. Circulation 1999,
100, 193–202. 12. Simpson, D.G.; Terracio, L.; Terracio,
M.; Price, R.L.; Turner, D.C.; Borg, T.K. Modulation of
cardiac myocyte phenotype in vitro by the composition and
orientation of the extracellular matrix. J. Cell. Physiol.
1994, 161, 89–105. 13. McDevitt, T.C.; Angello, J.C.;
Whitney, M.L.; Reinecke, H.; Hauschka, S.D.; Murry, C.E.;
Stayton, P.S. In vitro generation of differentiated cardiac
myofibers on micropatterned laminin surfaces. J. Biomed.
Mater. Res. 2002, 60, 472–479. 14. Bursac, N.; Papadaki,
M.; Cohen, R.J.; Schoen, F.J.; Eisenberg, S.R.; Carrier,
R.; Vunjak-Novakovic, G.; Freed, L. Cardiac muscle tissue
engineering: Toward an in vitro model for
electrophysiological studies. Am. J. Physiol. 1999, 277,
433–444. (Heart Cir Physiol 4e). 15. Carrier, R.L.;
Papdaki, M.; Rupnick, M.; Schoen, F.J.; Bursac, N.; Langer,
R.; Freed, L.E.; Vanjak-Novakovic, G. Cardiac tissue
engineering: Cell seeding, cultivation parameters, and
tissue construct characterization. Biotechnol. Bioeng.
1999, 64, 580–589. 16. Freed, L.E.; Vunjak-Novakovic, G.
Tissue Culture Bioreactors: Chondrogenesis as a Model
System. In Principles of Tissue Engineering; RG Landes:
Austin, TX, 1997; 151–165. 17. Freed, L.E.;
Vunjak-Novakovic, G. Cultivation of cellpolymer tissue
constructs in simulated microgravity. Biotech.
Bioengineering. 1995, 46, 306–315. 18. Freed, L.E.;
Vunjak-Novakovic, G. Microgravity tissue engineering. In
Vitro Cell. Dev. Biol., Anim. 1997, 33, 381–385. 19.
Jankowski, R.J.; Wagner, W.R. Directions in cardiovascular
tissue engineering. Clin. Plast. Surg. 1999, 26 (4),
605–616. 20. Rubin, K.; Gullberg, D.; Tomasini-Johansson,
B.; Reed, R.K.; Ryden, C.; Borg, T.K.; Part, H. Molecular
Components. In Structure and Function of the Extracellular
Matrix of Connective Tissues; Comer, W.D., Ed.; Harwood
Academic Press: New York, 1995. 21. Borg, T.K.; Lill, P.H.;
Johnson, L.D. Specific attachment of collagen to cardiac
myocytes: In vivo and in vitro. Dev. Biol. 1983, 97,
417–423. 22. Borg, T.K.; Ranson, W.F.; Moslehy, F.A.
Structural basis of ventricular stiffness. Lab. Invest.
1981, 44, 49–54. 23. Weber, K.T.; Kalil, J.E.; Janicki,
J.S. Myocardial collagen remodeling in pressure overload
hypertrophy: A case for interstitial heart disease. Am. J.
Hypertens. 1989, 2, 93. 24. Weber, K.T.; Pick, R.; Janicld,
J.S. Inadequate Collagen tethers in dilated cardiopathy.
Am. J. Heart. 1988, 116, 1641. 25. Weber, K.T.; Brilla,
C.G. Pathological hypertrophy and cardiac interstitium.
Circulation 1991, 86 (6), 1849–1865. 26. Borg, T.K.;
Terracio, L. Attachment Substrates for Heart Muscle Cells.
In Cell Culture Techniques in Cardiovascular Research;
Piper, M., Ed.; Springer-Verlag: Berlin, 1989. 27. Ross,
R.S.; Borg, T.K. Integrins and the myocardium. Circ. Res.
2001 Jun 8, 88 (11), 1112–1119. [Review] [102 refs]
[Journal Article. Review. Review, Tutorial]. 28. Hynes,
R.O. Integrins: Versatility, modulation, and signaling in
cell adhesion. Cell 1992, 69 (1), 11–25. 29. Borg, T.K.;
Nakagawa, M.; Carver, W.; Terracio, L. Extracellular
Matrix, Receptors and Heart Development. In Developmental
Mechanisms of Heart Disease; Clark, E.B., Markwald, R.R.,
Takao, A., Eds.; Futura Publ. Co.: Armonk, NY, 1995. 30.
Simpson, D.G.; Sharp, W.W.; Borg, T.K.; Price, R.L.;
Terracio, L.; Samarel, A.M. Mechanical regulation of
cardiac myocyte protein turnover of a myofibrillar
structure. Am. J. Physiol. 1996, 270 (4 pt 1), C1075–C1087.
31. Sharp, W.W.; Simpson, D.G.; Borg, T.K.; Samarel, A.M.;
Terracio, L. Contractile activity and external mechanical
tension affect focal adhesion formation by cultured
neonatal rat cardiac myocytes. Am. J. Physiol. 1997, 273 (2
Pt 2), H546–H556. 32. Tingstrom, A.; Heldin, C.H.; Rubin,
K. Regulation of fibroblast mediated collagen gel
contraction by PDGF, IL-1, and TGFB I. J. Cell. Sci. 1992,
102 (pt 2), 315–322. 33. Lundgren, E.; Gullberg, D.; Rubin,
K.; Borg, T.K.; Terracio, M.J.; Terracio, L. In vitro
studies on adult cardiac myocytes: Attachment and
biosynthesis of collagen type IV and laminin. J. Cell.
Physiol. 1988, 136, 43–53. 34. Izumo, S.; Lompre, A.M.;
Matsuoka, R.; Koren, G.; Schwartz, K.; Ginard-Nadal, B.;
Mahdavi, V. Myosin heavy chain messenger RNA and Protein
isoform transitions during Cardiac Hypertrophy. Interaction
between hemodynamic and thyroid-induced signals. J. Clin.
Invest. 1987, 79, 977–979. 35. Samarel, A.M.; Spragia,
M.L.; Maloney, V.; Kamal, S.A.; Englmann, G.L. Contractile
arrest accelerates myosin heavy chain degradation in
neonatal rat heart cells. Am. J. Physiol. 1992, 263 (3 Pt
1), C642– C652. 36. Terracio, L.; Peters, W.; Durig, B.;
Miller, B.; Borg, K.; Borg, T.K. Cellular Hypertrophy Can
be Induced by Cyclical Stretch In Vitro. In UCLA Symposium
on Molecular and Cellular Biology; Skalak, R., Fox, C.F.,
Eds.; Alan R Liss, Inc.: New York, NY, 104, 51–56. 37.
Shiraishi, I.; Simpson, D.G.; Fu, L.; Zhao, Z.; Majewski,
M.; Borg, T.K.; Terracio, L. Inhibition of vinculin with
antisense ODNS in fetal mouse cardiac myocytes. I. Mol.
Cell. Cardiol. 1996, 29, 2041– 2052. 38. Yost, M.J.;
Simpson, D.G.; Wrona, K.; Ridley, S.; Ploehn, H.; Borg, T.;
Terracio, L. Design and construction of a uniaxial cell
stretcher. Am. J. Physiol. Heart Circ. Physiol. 2000, 279,
H3124–H3130. 39. Simpson, D.G.; Reaves, T.A.; Shih, D.;
Burgess, W.; Borg, T.K.; Terracio, L. Cardiac integrins:
The ties that bind. Cardiovasc. Pathol. 1998, 7, 135–143.
40. Akins, R.E.; Schroedi, N.A.; Gonda, S.R.; Hartzell,
C.R. Neonatal rat heart cells cultured in simulated
microgravity. In Vitro 1997, 33, 337–343. 41. Molnar, G.;
Schroedl, N.A.; Gonda, S.R.; Hartzell, C.R. Skeletal muscle
satellite cells cultured in simulated microgravity. In
vitro, Animal 1997, 33 (5), 386. C
Cartilage Regeneration

6. Muir, H. The chondrocyte, architect of cartilage.


Biomechanics, structure, function and molecular biology of
cartilage matrix macromolecules. Bioessays 1995, 17 (12),
1039–1048.

7. Hu, J.C.Y.; Athanasiou, K.A. The Role of Mechanical


Forces in Tissue Engineering of Articular Cartilage. In
Functional Tissue Engineering; Guilak, F., Butler, D.L.,
Mooney, D., Eds.; Springer Verlag: New York, 2003; 227–243.

8. Buckwalter, J.A.; Mankin, H.J. Articular cartilage:


Tissue design and chondrocyte-matrix interactions. Instr.
Course Lect. 1998, 47, 477–486.

9. Kadiyala, S.; Young, R.G.; Thiede, M.A.; Bruder, S.P.


Culture expanded canine mesenchymal stem cells possess
osteochondrogenic potential in vivo and in vitro. Cell
Transplant. 1997, 6 (2), 125–134.

10. Coutts, R.D.; Sah, R.L.; Amiel, D. Effects of growth


factors on cartilage repair. Instr. Course Lect. 1997, 46,
487–494.

11. Darling, E.M.; Athanasiou, K.A. Articular cartilage


bioreactors and bioprocesses. Tissue Eng, 2003, 9 (1),
9–26.

12. Mow, V.C.; Kuei, S.C.; Lai, W.M.; Armstrong, C.G.


Biphasic creep and stress relaxation of articular cartilage
in compression: Theory and experiments. J. Biomech. Eng.
1980, 102 (1), 73–84.

13. Athanasiou, K.A.; Rosenwasser, M.P.; Buckwalter, J.A.;


Malinin, T.I.; Mow, V.C. Interspecies comparisons of in
situ intrinsic mechanical properties of distal femoral
cartilage. J. Orthop. Res. 1991, 9 (3), 330–340.

14. Athanasiou, K.A.; Agarwal, A.; Dzida, F.J. Comparative


study of the intrinsic mechanical properties of the human
acetabular and femoral head cartilage. J. Orthop. Res.
1994, 12 (3), 340–349.

15. Grodzinsky, A.J.; Levenston, M.E.; Jin, M.; Frank, E.H.


Cartilage tissue remodeling in response to mechanical
forces. Annu. Rev. Biomed. Eng. 2000, 2, 691–713.

16. Hunziker, E.B. Articular cartilage repair: Basic


science and clinical progress. A review of the current
status and prospects. Osteoarthr. Cartil. 2002, 10 (6),
432–463.

17. Buckwalter, J.A. Articular cartilage: Injuries and


potential for healing. J. Orthop. Sports Phys. Ther. 1998,
28 (4), 192–202.

18. LeBaron, R.G.; Athanasiou, K.A. Ex vivo synthesis of


articular cartilage. Biomaterials 2000, 21 (24), 2575–2587.

19. Niederauer, M.Q.; Wilkes, R.P.; Niederauer, G.M.;


Cristante, S.; Cline, S.F.; Tynmann, R.A. Hand-Held
Materials Tester. US Patent 5,904,658, 1997.

20. Darling, E.M.; Athanasiou, K.A. Bioactive Scaffold


Design for Articular Cartilage. In Biomedical Technology
and Devices Handbook; Moore, J., Zouridakis, G., Eds.; CRC
Press: New York, 2003; Ch21 (1–16).

21. Coutts, R.D.; Healey, R.M.; Ostrander, R.; Sah, R.L.;


Goomer, R.; Amiel, D. Matrices for cartilage repair. Clin.
Orthop. 2001, 391 (Suppl), S271–279. 22. Lu, L.;
Valenzuela, R.G.; Yaszemski, M.J. Articular cartilage
engineering. J. Regenerative Med. 2000, 2, 99–114. 23. van
Susante, J.L.C.; Pieper, J.; Buma, P.; van Kuppevelt, T.H.;
van Beuningen, H.; van Der Kraan, P.M.; Veerkamp, J.H.; van
den Berg, W.B.; Veth, R.P.H. Linkage of chondroitin-sulfate
to type I collagen scaffolds stimulates the bioactivity of
seeded chondrocytes in vitro. Biomaterials 2001, 22 (17),
2359–2369. 24. Nehrer, S.; Breinan, H.A.; Ramappa, A.;
Shortkroff, S.; Young, G.; Minas, T.; Sledge, C.B.; Yannas,
I.V.; Spector, M. Canine chondrocytes seeded in type I and
type II collagen implants investigated in vitro. J. Biomed.
Mater. Res. 1997, 38 (2), 95–104. 25. Freed, L.E.;
Hollander, A.P.; Martin, I.; Barry, J.R.; Langer, R.;
Vunjak-Novakovic, G. Chondrogenesis in a
cell-polymer-bioreactor system. Exp. Cell. Res. 1998, 240
(1), 58–65. 26. Kohn, J.; Langer, R. Bioresorbable and
bioerodable materials. In Biomaterials Science; Ratner,
B.D., Hoffman, A.S., Shoen, F.J., Lemons, J.E., Eds.;
Academic Press: New York, 1996; 64–84. 27. Middleton, J.C.;
Tipton, A.J. Synthetic biodegradable polymers as orthopedic
devices. Biomaterials 2000, 21 (23), 2335–2346. 28.
Athanasiou, K.A.; Korvick, D.; Schenck, R.C., Jr.
Biodegradable implants for the reatment of ostechondral
defects in a goat model. Tissue Eng. 1997, 3 (4), 363–373.
29. Athanasiou, K.A.; Singhal, A.R.; Agrawal, C.M.; Boyan,
B.D. In vitro degradation and release characteristics of
biodegradable implants containing trypsin inhibitor. Clin.
Orthop. 1995, 315, 272–281. 30. LeBaron, R.G.; Athanasiou,
K.A. Extracellular matrix cell adhesion peptides:
functional applications in orthopedic materials. Tissue
Eng. 2000, 6 (2), 85–103. 31. Blunk, T.; Sieminski, A.L.;
Gooch, K.J.; Courter, D.L.; Hollander, A.P.; Nahir, A.M.;
Langer, R.; VunjakNovakovic, G.; Freed, L.E. Differential
effects of growth factors on tissue-engineered cartilage.
Tissue Eng. 2002, 8 (1), 73–84. 32. Zhu, C.; Bao, G.; Wang,
N. Cell mechanics: Mechanical response, cell adhesion, and
molecular deformation. Annu. Rev. Biomed. Eng. 2000, 2,
189–226. 33. Shieh, A.C.; Athanasiou, K.A. Biomechanics of
single chondrocytes and osteoarthritis. Crit. Rev. Biomed.
Eng, 2002, 30 (4–6), 307–343. 34. Hall, A.C.; Horwitz,
E.R.; Wilkins, R.J. The cellular physiology of articular
cartilage. Exp. Physiol. 1996, 81 (3), 535–545. 35.
Pazzano, D.; Mercier, K.A.; Moran, J.M.; Fong, S.S.;
DiBiasio, D.D.; Rulfs, J.X.; Kohles, S.S.; Bonassar, L.J.
Comparison of chondrogensis in static and perfused
bioreactor culture. Biotechnol. Prog. 2000, 16 (5),
893–896.
Cell Culture Assays

1. Paul, J. Cell and Tissue Culture, 3rd Ed.; Churchill


Livingstone Press: Edinburgh, 1975.

2. Harrison, R.G. Observations on the living developing


nerve fibre. Proc. Soc. Exp. Biol. N.Y. 1907, 4, 140–143.

3. Carrell, A. On the permanent life of tissues outside the


organism. J. Exp. Med. 1912, 15, 516–528.

4. Hayflick, L.; Moorhead, P.S. The serial cultivation of


human diploid cell strains. Exp. Cell Res. 1961, 25,
585–621.

5. Freshney, R.I. Culture of Animal Cells, a Manual of


Basic Technique, 4th Ed.; Wiley-Liss: New York, 2000.

6. Williams, D.F. Progress in Biomedical Engineering,


Definitions in Biomaterials; Elsevier: Amsterdam, 1987; Vol.
4, 54.

7. Rosenbluth, S.A.; Weddington, G.R.; Guess, W.L.; Autian,


J. Tissue culture method for screening toxicity of plastic
materials to be used in medical practice. J. Pharm. Sci.
1965, 54, 156–159.

8. Guess, W.L.; Rosenbluth, S.A.; Schmidt, B.; Autian, J.


Agar diffusion method for toxicity screening of plastics on
cultured cell monolayers. J. Pharm. Sci. 1965, 54,
1545–1547. 9. Autian, J. Toxicological evaluation of
biomaterials: Primary acute toxicity screening program.
Artif. Organs 1977, 1, 53–60. 10. ISO 10993 The Biological
Evaluation of Medical Devices, Part 5, Tests for
Toxicity—In vitro Methods; International Standards
Organisation 1995. ISO 10993-5. 11. Chauvel-Lebret, D.J.;
Auroy, P.; Tricot-Doleux, S.; Bonnaure-Mallet, M.
Evaluation of the SCGE assay to assess the genotoxicity of
biomaterials. Biomaterials 2001, 22, 1795–1807. 12. ISO
10993 The Biological Evaluation of Medical Devices, Part 4,
Selection of Tests for Interactions with Blood;
International Standards Organisation 1995. 10993-4. 13.
Renvoize, C.; Biola, A.; Pallardy, M.; Breard, J.
Apoptosis: Identification of dying cells. Cell Biol.
Toxicol. 1998, 14, 111–120. 14. Ferro, M.; Doyle, A.
Standardisation for in vitro testing. Cell Biol. Toxicol.
2001, 17, 205–212. 15. Kirkpatrick, C.J.; Mittermayer, C.
Theoretical and practical aspects of testing potential
biomaterials in vitro. J. Mater. Sci., Mater. Med. 1990, 1,
9–13. 16. Ciapetti, G.; Cenni, E.; Pratelli, L.;
Pizzoferrato, A. In vitro evaluation of cell/biomaterial
interaction by MTT assay. Biomaterials 1993, 14, 359–364.
17. Freshney, I. Application of cell cultures to
toxicology. Cell Biol. Toxicol. 2001, 17 , 213–230. 18.
Ames, B.N. The Detection of Chemical Mutagens with Enteric
Bacteria. In Chemical Mutagens: Principles and Methods for
Their Detection; Hollaender, Ed.; Plenum: New York, 1971;
267–282. 19. Kirkpatrick, C.J.; Bittinger, F.; Wagner, M.;
Kohler, H.; van Kooten, T.G.; Klein, C.L.; Otto, M. Current
trends in biocompatibility testing. Proc. Inst. Mech. Eng.
1998, 212 Part H, 75–84. 20. Meyle, J.; Gultig, K.; Brich,
M.; Hammerle, H.; Nisch, W. Contact guidance of fibroblasts
on biomaterials surfaces. J. Mater. Sci., Mater. Med. 1994,
5, 463–466. 21. Flemming, R.G.; Murphy, C.J.; Abrams, G.A.;
Goodman, S.L.; Nealy, P.F. Effects of synthetic microand
nano-structured surfaces on cell behaviour. Biomaterials
1999, 20, 573–588. 22. Pizzoferatto, A.; Ciapetti, G.;
Stea, S.; Cenni, E.; Arciola, C.R.; Grachi, D.; Savarino,
L. Cell culture methods for testing biocompatibility. Clin.
Mater. 1994, 15, 173–190. 23. Hoffman, R.M. The
3-dimensional question—Can clinically relevant tumour drug
resistance be measured in vitro? Cancer Metastasis Rev.
1994, 13, 169–173. 24. Cenni, E.; Granchi, D.; Ciapetti,
G.; Verri, E.; Cavadagena, D.; Gamberini, S.; DiLeo, A.;
Pizzoferatto, A. Cytokine production and adhesive protein
expression by endothelial cells after contact with
polyethylene terephthalate. Biomaterials 1996, 17,
2071–2076. 25. Stacey, G.; MacDonald, C. Immortalisation of
primary cells. Cell Biol. Toxicol. 2001, 17, 231–246. 26.
Jones, B.; Stacey, G. Safety considerations for in vitro
toxicology testing. Cell Biol. Toxicol. 2001, 17, 247–270.
C
Cell-Material Interaction

9. von Recum, A.F.; Shannon, C.E.; Cannon, C.E.; Long,


K.J.; van Kooten, T.G.; Meyle, J. Surface roughness,
porosity, and texture as modifiers of cellular adhesion.
Tissue Eng. 1996, 2 (4), 241–253.

10. Kaelble, D.H.; Moacanin, J. A surface energy analysis


of bioadhesion. Polymer 1977, 18 (5), 475–482.

11. Sugimoto, Y. Effect of adhesion and locomotion of mouse


fibroblasts by their interacting with differently charged
substrates. Exp. Cell Res. 1981, 135 (1), 39–45.

12. Folkman, J.; Moscona, A. Role of cell shape in growth


control. Nature 1978, 273 (5661), 345–349.

13. Gugala, Z.; Gogolewski, S. Attachment, growth, and


activity of rat osteoblasts on polylactide membranes
treated with various low temperature radiofrequency
plasmas. J. Biomed. Mater. Res. Part A. 2006, 76A (2),
288–299.

14. Janorkar, A.V.; Fritz, E.W.; Burg, K.J.L.; Metters,


A.T.; Hirt, D.E. Grafting amine-terminated branched
architectures from poly(L-lactide) film surfaces for
improved cell attachment. J. Biomed. Mater. Res. Part B.
Appl. Biomater. 2007, 81B, 142–152.

15. Atthoff, B.; Hilborn, J. Protein adsorption onto


polyester surfaces: Is there a need for surface activation?
J. Biomed. Mater. Res. Part B. Appl. Biomater. 2007, 80
(1), 121–130.

16. Liu, X.; Won, Y.; Ma, P.X. Surface modification of


interconnected porous scaffolds. J. Biomed. Mater. Res. A
2005, 74 (1), 84–91.

17. Narotam, P.K.; Jose, S.; Nathoo, N.; Taylon, C.; Vora,
Y. Collagen matrix (DuraGen) in dural repair: Analysis of a
new modified technique. Spine 2004, 29 (24), 2868–2669.

18. Hsu, S.-H.; Chang, S.-H.; Yen, H.J.; Whu, S.W.; Tsai,
C.-L.; Chen, D.C. Evaluation of biodegradable polyesters
modified by type II collagen and arg-gly-asp as tissue
engineering scaffolding materials for cartilage
regeneration. Artif. Organs 2006, 30 (1), 42–55.

19. Zhang, L.; Hum, M.; Wang, M.; Li, Y.; Chen, H.; Chu,
C.; Jiang, H. Evaluation of modifying collagen matrix with
RGD peptide through periodate oxidation. J. Biomed. Mater.
Res. Part A 2005, 73A (4), 468–475.

20. Ye, Q.; Zund, G.; Benedikt, P.; Jockenhoevel, S.;


Hoerstrup, S.P.; Sakyama, S.; Hubbell, J.A.; Turina, M.
Fibrin gel as a three dimensional matrix in cardiovascular
tissue engineering. Eur. J. Cardiothorac. Surg. 2000, 17
(5), 587–591. 21. Siggelkow, W.; Gescher, D.M.; Siggelkow,
A.; Klee, D.; Malik, E.; Rath, W.; Faridi, A. In vitro
analysis of modified surfaces of silicone breast implants.
Int. J. Artif. Organs 2004, 27 (12), 1100–1108. 22.
Hegewald, A.A.; Ringe, J.; Bartel, J.; Kruger, I.; Notter,
M.; Barnewitz, D.; Kaps, C.; Sittinger, M. Hyaluronic acid
and autologous synovial fluid induce chondrogenic
differentiation of equine mesenchymal stem cells: A
preliminary study. Tissue Cell. 2004, 36 (6), 431–438. 23.
Rowley, J.A.; Mooney, D.J. Alginate type and RGD density
control myoblast phenotype. J. Biomed. Mater. Res. 2002, 60
(2), 217–223. 24. Itoh, D.; Yoneda, S.; Kuroda, S.; Kondo,
H.; Umezawa, A.; Ohya, K.; Ohyama, T.; Kasugai, S.
Enhancement of osteogenesis on hydroxyapatite surface
coated with synthetic peptide (EEEEEEEPRGDT) in vitro. J.
Biomed. Mater. Res. 2002, 62 (2), 292–298. 25. Fussell,
G.W.; Cooper, S.L. Endothelial cell adhesion on
RGD-containing methacrylate terpolymers. J. Biomed. Mater.
Res. A 2004, 70 (2), 265–273. 26. Folch, A.; Toner, M.
Microengineering of cellular interactions. Annu. Rev.
Biomed. Eng. 2000, 2 (1), 227–256. 27. Campbell, C.E.; von
Recum, A.F. Microtopography and soft tissue response. J.
Invest. Surg. 1989, 2 (1), 51–74. 28. Clark, P.; Connolly,
P.; Curtis, A.S.; Dow, J.A.; Wilkinson, C.D. Topographical
control of cell behavior: II. Multiple grooved substrata.
Development 1990, 108 (4), 635–644. 29. Matsuzaka, K.;
Walboomers, F.; de Ruijter, A.; Jansen, J.A. Effect of
microgrooved poly-l-lactide (PLA) surfaces on
proliferation, cytoskeletal organization, and mineralized
matrix formation of rat bone marrow cells. Clin. Oral
Implants Res. 2000, 11 (4), 325–333. 30. Lossdorfer, S.;
Schwartz, Z.; Wang, L.; Lohmann, C.H.; Turner, J.D.;
Wieland, M.; Cochran, D.L.; Boyan, B.D. Microrough implant
surface topographies increase osteogenesis by reducing
osteoclast formation and activity. J. Biomed. Mater. Res. A
2004, 70 (3), 3361–3369. 31. Boyan, B.D.; Bonewald, L.F.;
Paschalis, E.P.; Lohmann, C.H.; Rosser, J.; Cochran, D.L.;
Dean, D.D.; Schwartz, Z.; Boskey, A.L. Osteoblast-mediated
mineral deposition in culture is dependent on surface
microtopography. Calcif. Tissue Int. 2002, 71 (6), 519–529.
C
Centrifugal Blood Pumps

18. Takano, T.; Schulte-Eistrup, S.; Kawahito, S.; Maeda,


T.; Nonaka, K.; Linneweber, J.; Glueck, J.; Fujisawa, A.;
Makinouchi, K.; Yokokawa, M.; Nose, Y. Inlet port
positioning for a miniaturized centrifugal blood pump.
Artif. Organs Jan 2002, 26 (1), 45–48.

19. Nishimura, I.; Ichikawa, S.; Mikami, M.; Ishitoya, H.;


Motomura, T.; Kawamura, M.; Linneweber, J.; Glueck, J.;
Shinohara, T.; Nose, Y.; Nishimura, I. Evaluation of
floating impeller phenomena in a gyro centrifugal pump.
ASAIO J. Nov–Dec 2003, 49 (6), 744–747.

20. Miller, G.E.; Etter, B.D.; Dorsi, J.M. A multiple disk


centrifugal pump as a blood flow device. IEEE Trans. Biomed.
Eng. Feb 1990, 37 (2), 157–163.

21. Miller, G.E.; Sidhu, A.; Fink, R.; Etter, B.D.


Evaluation of a multiple disk centrifugal pump as an
artificial ventricle. Artif. Organs Jul 1993, 17 (7),
590–592.

22. Miller, G.E.; Madigan, M.; Fink, R. A preliminary flow


visualization study in a multiple disk centrifugal
artificial ventricle. Artif. Organs Jul 1995, 19 (7),
680–684.

23. Manning, K.B.; Miller, G.E. Shaft/shaft-seal interface


characteristics of a multiple disk centrifugal blood pump.
Artif. Organs Jun 1999, 23 (6), 552–558.

24. Miller, G.E.; Fink, R. Analysis of optimal design


configurations for a multiple disk centrifugal blood pump.
Artif. Organs Jun 1999, 23 (6), 559–565.

25. Manning, K.B.; Miller, G.E. Flow through an outlet


cannula of a rotary ventricular assist device. Artif.
Organs Aug 2002, 26 (8), 714–723.

26. Rice, W. An analytical and experimental investigation


of multiple disk pumps and compressors. J. Eng. Power 1963,
85, 191–200. 27. Brieter, M.; Pohlhausen, K. Laminar Flow
Between Two Parallel Rotating Disks. In Aeronatical
Research Laboratory Report, WPAFB; U.S. Air Force, 1962.
Project No. 7071, Task No. 70437. 28. Hassinger, S.H.;
Kehrt, L.G. Investigation of a shearforce pump. J. Eng.
Power July 1963, 85, 201–207. 29. Tesla, N. Turbine. US
Patent No. 1,061,142, May, 1913. 30. Noon, G.P.; Ball,
J.W., Jr.; Papaconstantinou, H.T. Clinical experience with
BioMedicus centrifugal ventricular support in 172
patitents. Artif. Organs Jul 1995, 19 (7), 756–760. 31.
Noon, G.P.; Lafuente, J.A.; Irwin, S. Acute and temporary
ventricular support with BioMedicus centrifugal pump. Ann.
Thorac. Surg. Aug 1999, 68 (2), 650–654. 32. Reiss, N.;
El-Banayosy, A.; Mirow, N.; Minami, K.; Korfer, R.
Implantation of the Biomedicus centrifugal pump in
post-transplant right heart failure. J. Cardiovasc. Surg.
Oct 2000, 41 (5), 691–694. 33. Ochiai, Y.; Golding, L.A.;
Massiello, A.L.; Medvedev, A.L.; Gerhart, R.L.; Chen, J.F.;
Takagai, M.; Fukamachi, K. In vivo hemodynamic performance
of the Cleveland Clinic CorAide blood pump in calves. Ann.
Thorac. Surg. Sep 2001, 72 (3), 747–752. 34. Ochiai, Y.;
Golding, L.A.; Massiello, A.L.; Medvedev, A.L.; Horvath,
D.J.; Gerhart, R.L.; Chen, J.F.; Krogulecki, A.Y.;
Takagaki, M.; Doi, K.; Howard, M.W.; Fukamachi, K.
Cleveland Clinic CorAide blood pump circulatory support
without anticoagulation. ASAIO J. May–Jun 2002, 48 (3),
249–252. 35. Fukamachi, K.; Ochiai, Y.; Doi, K.; Massiello,
A.L.; Medvedev, A.L.; Horvath, D.J.; Gerhart, R.L.; Chen,
J.F.; Krogulecki, A.Y.; Takagaki, M.; Howard, M.W.; Kopcak,
M.W., Jr.; Golding, L.A. Chronic evaluation of the
Cleveland Clinic CorAide left ventricular assist system in
calves. Artif. Organs Jun 2002, 26 (6), 529–533.
Ceramics

31. Heimann, R.B.; Vu, T.A. Effect of CaO on thermal


decomposition during sintering of composite hydroxyapatite
zirconia mixtures for monolithic bioceramic implants. J.
Matls. Sci.: Letters 1997, 16, 437–439.

32. Rao, R.R.; Kannan, T.S. Synthesis and sintering of


hydroxyapatite zirconia composites. Matls. Sci. and Eng. C
2002, 20, 187–193.

33. Kim, H.W.; Noh, Y.J.; Koh, Y.H.; Kim, H.E.; Kim, H.M.
Effect of CaF 2 on densification and properties of
hydroxyapatite-zirconia composites for biomedical
applications. Biomaterials 2002, 23, 4113–4121.

34. Wu, J.; Yeh, F. Sintering of hydroxyapatite-zirconia


composite materials. J. Matls. Sci. 1988, 23, 3771–3777.

35. Ahn, E.S.; Gleason, N.J.; Nakahira, A.; Ying, J.Y.


Nanostructure processing of hydroxyapatite based
bioceramics. Nano Letters 2001, 1–3, 149–153.

36. Cook, S.D.; Kay, J.F.; Thomas, K.A.; Jarcho, M.


Interface mechanics and histology of titanium and
hydroxylapatite coated metal implants. J. Biomed. Mater.
Res. 1984, 18, 719–726.

37. Ducheyne, P.; Healy, K.E. The effect of plasma sprayed


calcium phosphate ceramic coatings on the metal ion release
from porous titanium and cobalt-chromium alloys. J. Biomed.
Mater. Res. 1988, 22, 1137–1163.

38. Ji, H.; Ponton, C.B.; Marquise, P.M. Microstructural


characterization of hydroxylapatite coating on titanium. J.
Mater. Sci.: Mater. Med. 1992, 3, 283–287.

39. Park, E.; Condrate, R.A.; Hoelzer, D.T.; Fischman, G.S.


Interfacial characterization of plasma-spray coated calcium
phosphate on Ti-6A1-4V. J. Mater. Sci.: Mater. Med. 1998,
9, 643–649.

40. VanDijk, K.; Schaeken, H.G.; Wolke, J.G.C.; Jansen,


J.A. Influence of annealing temperature on RFmagnetron
sputtered calcium phosphate coatings. Biomaterials 1996,
17, 405–410.

41. Liu, X.; Weng, J.; Tong, W.; Zuo, C; Zhang, X.; Wang,
P.; Liu, Z. Characterization of a hydroxylapatite film with
mixed interface by Ar + ion beam enhanced deposition.
Biomaterials 1997, 18, 1487–1493. 42. Brendel, T.; Engel,
A.; Russel, C. Hydroxylapatite coating by polymeric route.
J. Mater. Sci.: Mater. Med. 1992, 3, 175–179. 43. Li, T.;
Lee, J.; Kobayashi, T.; Aoki, H. Hydroxylapaite coating by
dipping method. J. Mat. Sci.: Mat. Med. 1996, 7, 355–357.
44. Russell, S.W.; Luptak, K.A.; Suchicital, C.T.A.;
Alford, T.L.; Pizzicano, V.B. Chemical and structural
evaluation of sol–gel derived hydroxylapatite thin films
under rapid thermal processing. J. Am. Ceram. Soc. 1996,
79, 837–842. 45. Kim, C.S.; Ducheyne, P. Compositional
variations in the surface and interface of calcium
phosphate ceramic coatings on Ti and Ti-6A1-4V due to
sintering and immersion. Biomaterials 1991, 12, 461–469.
46. Hero, H.; Wie, H.; Jorgensen, R.B.; Ruyter, I.E.
Hydroxylapatite coating on Ti by hot isostatic pressing. J.
Biomed. Mater. Res. 1994, 28, 343–348. 47. Garcia, R;
Doremus, R.H. Electron microscopy of the
bone-hydroxylapatite interface from a human dental implant.
J. Mater. Sci.: Mater. Med. 1992, 3, 154–156. 48. Ergun, C;
Doremus, R.H.; Lamford, W.A. Hydroxylapatite and titanium:
interfacial reactions. J. Biomed. Matls. Res. 2003,
336–343. 49. Sarikaya, M.; Tamerler, C.; Jen, A.K.Y.;
Schulten, K.; Baneyx, F. Molecular biomimetics:
nanotechnology through biology. Nature Materials, 2003, 2,
577–585. 50. Brown, S.; Sarikaya, M.; Johnson, E. Genetic
analysis of crystal growth. J. Mol. Biol. 2000, 299,
725–732. 51. Beverly, K.C. Quantum dot artificial solids:
understanding the static and dynamic role of size and
packing disorder. Proc. Natl. Acad. Sci. USA 2002, 99,
6456–6459. 52. Kempa, K. Photonic crystals based on
periodic arrays of aligned carbon nanotubes. Nano Letters
2003, 3, 13–18. 53. Knoblauch, M. ATP-independent
contractile proteins from plants. Nature Materials, 2003,
2, 800–803. 54. Mavroidis, C.; Dubey, A. Biomimetics: from
pulses to motors. Nature Materials 2003, 2, 573–574.
Ceramics in Dentistry

1. Paul, S.J.; Peter, A.; Rodoni, L.; Pietrobon, N.


Conventional visual vs spectrophotometric shade taking for
porcelain-fused-to-metal crowns, a clinical comparison.
Int. J. Periodontics Restorative Dent. 2004, 24 (3),
222–231.

2. Lundgren, D.; Laurell, L. Occlusal forces in


prosthetically restored dentitions, a methodological study.
J. Oral Rehabil. 1984, 11 (1), 29–37. 3. International
Standards Organisation, ISO Standard 6872, Dental Ceramics,
1997. 4. Burke, F.J.; Fleming, G.J.; Nathanson, D.;
Marquis, P.M. Are adhesive technologies needed to support
ceramics? An assessment of the current evidence
(1988–2002). J. Adhes. Dent. 2002, 4, 7–22. 5. Fradeani,
M.; Redemagni, M.; Corrado, M. Porcelain laminate veneers,
6to 12-year clinical evaluation—a retrospective study. Int.
J. Periodontics Restorative Dent. 2005, 25 (1), 9–17. 6.
Weinstein, M.; Katz, S.; Weinstein, A.B. Fused
Porcelain-To-Metal Teeth. US Patent 3,052,982, Sept. 11,
1962; Porcelain-Covered Metal-Reinforced Teeth. US Patent
3,052,983, Sept. 11, 1962. 7. Grossman, D.G.; Johnson, J.L.
Glass Ceramic Compositions for Dental Constructs. U.S.
Patent 4,652,312, March 24, 1987. 8. Martin, N. Interfaces
in Dental CAD-CAM Restorations. Ph.D. thesis, The
University of Liverpool, 1996. 9. Grossman, D.G. Structure
and physical properties of Dicor/MGC glass-ceramic. In
International Symposium on Computer Restorations. Moermann,
W.H., Ed.; Quintessence: Berlin, 1991; Vol. 103, 115. 10.
Hoeland, W.; Frank, M. Material Science of Empress
Glass-Ceramics, Ivoclar-Vivadent Report No. 10; July 1994,
3–8. 11. Raigrodski, A.J.; Chiche, G.J. The safety and
efficacy of anterior ceramic fixed partial dentures, a review
of the literature. J. Prosthet. Dent. 2001, 86 (5),
520–525. 12. Luthy, H.; Filser, F.; Loeffel, O.;
Schumacher, M.; Gauckler, L.J.; Hammerle, C.H. Strength and
reliability of four-unit all-ceramic posterior bridges.
Dent. Mater. 2005, 21 (5), 476–482. 13. Devigus, A.;
Lombardi, G. Shading Vita YZ substructures, influence on
value and chroma, Part I. Int. J. Comput. Dent. 2004, 7
(3), 293–301. 14. Schuh, C.; Kinast, E.J.; Mezzomo, E.;
Kapczinski, M.P. Effect of glazed and polished surface
finishes on the friction coefficient of two low-fusing
ceramics. J. Prosthet. Dent. 2005, 93 (3), 245–252. 15.
Balkaya, M.C.; Cinar, A.; Pamuk, S. Influence of firing
cycles on the margin distortion of 3 all-ceramic crown
systems. J. Prosthet. Dent. 2005, 93 (4), 346–355. 16.
Fradeani, M.; D’Amelio, M.; Redemagni, M.; Corrado, M.
Five-year follow-up with Procera all-ceramic crowns.
Quintessence Int. 2005, 36 (2), 105–113. 17. Jedynakiewicz,
N.M.; Martin, N. The Cerec method, science, research and
clinical application. Compend. Contin. Educ. Dent. 2001, 22
(6), 7–13. 18. Martin, N.; Jedynakiewicz, N.M. Clinical
performance of Cerec ceramic inlays, a systematic review.
Dent. Mater. 1999, 15, 54–61. 19. Guler, A.U.; Yilmaz, F.;
Ural, C.; Guler, E. Evaluation of 24-hour shear bond
strength of resin composite to porcelain according to
surface treatment. Int. J. Prosthodont. 2005, 18 (2),
156–160. 20. Bottino, M.A.; Valandro, L.F.; Scotti, R.;
Buso, L. Effect of surface treatments on the resin bond to
zirconium-based ceramic. Int. J. Prosthodont. 2005, 18 (1),
60–65.
Chitosan

Table 3 Applications of chitinous materials in food

technology

Area of application Examples

Antimicrobial agent Bactericidal Fungicidal Measure of mold


contamination in agricultural commodities

Edible films Controlled moisture transfer between food and


surrounding environment Controlled release of antimicrobial
substances Controlled release of nutrients, flavors, and
drugs Reduction of oxygen partial pressure Controlled rate
of respiration Temperature control Controlled enzymatic
browning in fruits Reverse osmosis membranes

Additive Clarification and deacidification of fruits and


beverages Natural flavor extender Texture controlling agent
Emulsifying agent Food mimetic Thickening and stabilizing
agent Color stabilization

Nutritional quality Dietary fiber Hypocholesterolemic effect


Livestock and fish food additive Reduction of lipid
absorption Production of single cell protein Antigastritis
agent Infant feed ingedient

Recovery of solid

materials from food

processing waste Affinity flocculation Fractionation of agar

Purification of water Recovery of metal ions, pesticides,


phenols, and PCBs Removal of dyes 4. Applications of Chitin
and Chitosan; Goosen, M.F.A., Ed.; Technomic Publishing Co.
Inc.: Lancaster, 1997; 1–336. 5. Skaugrud, O.; Hagen, A.;
Borgersen, B.; Dornish, M. Biomedical applications of
alginate and chitosan. Biotechnol. Genet. Eng. Rev. 1999,
16, 23–40. 6. Hirano, S. Chitin biotechnology applications.
Biotechnol. Annu. Rev. 1996, 2, 237–258. 7. Singla, A.K.;
Chawla, M. Chitosan: Some pharmaceutical and biological
aspects—An update. J. Pharm. Pharmacol. 2001, 53 (8),
1047–1067. 8. Genta, I.; Perugini, P.; Pavanetto, F.;
Modena, T.; Muzzarelli, R.A.A.; Conti, B. Chitosan
Crosslinking for Pharmaceutical Purposes: An Overview. In
Chitosan in Pharmacy and Chemistry; Muzzarelli, R.A.A.,
Muzzarelli, C., Eds.; Atec, Italy, 2002; 11–20. 9.
Muzzarelli, R.; Biagini, G.; Pugnaloni, A.; Filippini, O.;
Baldassarre, V.; Castaldini, C.; Rizzoli, C. Reconstruction
of parodontal tissue with chitosan. Biomaterials 1989, 53
(2), 131–140. 10. Rathke, T.D.; Hudson, S.M. Review of
chitin and chitosan as fibre and film formers. JMS Rev.
Macromol. Chem. 1994, C34, 375. 11. Kurita, K. Chemical
Modifications of Chitin and Chitosan. In Chitin in Nature
and Technology; Muzzarelli, R.A.A., Jeuniaux, C., Gooday,
G.W., Eds.; Plenum Press: New York, 1986; 287–293. 12.
Dodane, V.; Vilivalam, V.D. Pharmaceutical applications of
chitosan. Pharm. Sci. Technol. Today 1998, 6, 246–253. 13.
Sawayanagi, Y.; Nambu, N.; Nagai, T. Directly compressed
tablets containing chitin or chitosan in addition to
lactose or potato starch. Chem. Pharm. Bull. 1982, 30,
2935–2940. 14. Ritthidej, G.C.; Chomto, P.; Pummangura, S.;
Menasveta, P. Chitin and chitosan as disintigrants in
paracetamol tablets. Drug Dev. Ind. Pharm. 1994, 20,
2109–2134. 15. Illum, L. Chitosan and its use as a
pharmaceutical excipient. Pharm. Res. 1998, 15 (9),
1326–1331. 16. Sawayanagi, Y.; Nambu, N.; Nagai, T.
Enhancement of dissolution properties of griseofulvin from
ground mixtures with chitin and chitosan. Chem. Pharm.
Bull. 1982, 30, 4464–4467. 17. Dutta, P.D.; Ravi Kumar,
M.N.K.; Dutta, J. Chitin and chitosan for versatile
applications. J. Macromol. Sci., Part C, Polym. Rev. 2002,
C42 (3), 307–354. 18. Liu, W.G.; Yao, K.D. Chitosan and its
derivatives—A promising non-viral vector for gene
transfection. J. Control. Release 2002, 83, 1–11. 19.
Borchard, G. Chitosans for gene delivery. Adv. Drug Deliv.
Rev. 2001, 52, 145–150. 20. Paul, W.; Sharma, C.P.
Chitosan, a drug carrier for the 21st century: A review.
STP Pharma Sciences 2000, 10 (1), 5–22. 21. Felt, O.; Buri,
P.; Gurny, R. Chitosan: A unique polysaccharide for drug
delivery. Drug Dev. Ind. Pharm. 1998, 24 (11), 979–993. 22.
Bernkop-Schnurch, A. Chitosan and its derivatives:
Potential excipients for peroral peptide delivery systems.
Int. J. Pharm. 2000, 194, 1–13. 23. Cascone, M.G.;
Maltinti, S. Hydrogels based on chitosan and dextran as
potential drug delivery systems. J. Mater. Sci., Mater.
Med. 1999, 10 (5), 301–307. 24. Zhang, M.; Kohr, E.;
Hirano, S. Hydrogels of Chitin and Chitosan. In Food
Hydrocolloids: Proc. Int. Conf. Ind. Exhib.; Plenum: New
York, 1993; 65–70. 25. Kas, H.K. Chitosan: Properties,
preparations and application to microparticulate systems.
J. Microencapsul 1997, 14 (6), 689–711. 26. Janes, K.A.;
Calvo, P.; Alonso, M.J. Polysaccharide colloidal particles
as delivery systems for macromolecules. Adv. Drug Deliv.
Rev. 2001, 47, 83–97. 27. Risbud, M.V.; Bhonde, R.R.
Polyacrylamide–chitosan hydrogels: In vitro
biocompatibility and sustained antibiotic release studies.
Drug Deliv. 2000, 7, 69–75. 28. Chenite, A.; Chaput, C.;
Wang, D.; Combes, C.; Buschmann, M.D.; Hoemann, C.D.;
Leroux, J.C.; Atkinson, B.L.; Binette, F.; Selmani, A.
Novel injectable neutral solutions of chitosan form
biodegradable gels in situ. Biomaterials 2000, 21,
2155–2161. 29. Ruel-Garie`py, E.; Chenite, A.; Chaput, C.;
Guirguis, S.; Leroux, J.-C. Characterization of
thermosensitive chitosan gels for the sustained delivery of
drugs. Int. J. Pharm. 2000, 203, 89–98. 30. Chenite, A.;
Buschmann, M.; Wang, D.; Chaput, C.; Kandani, N.
Rheological characterization of thermogelling
chitosan/glycerol–phosphate solutions. Carbohydr. Polym.
2001, 46, 39–47. 31. Jarry, C.; Chaput, C.; Chenite, A.;
Renaud, M.-A.; Buschmann, M.; Leroux, J.-C. Effects of
steam sterilization on thermogelling chitosan-based gels.
J. Biomed. Mater. Res. 2001, 58, 127–135. 32. Molinaro, G.;
Leroux, J.-C.; Damas, J.; Adam, A. Biocompatibility of
thermosensitive chitosan-based hydrogels: An in vivo
experimental approach to injectable biomaterials.
Biomaterials 2002, 23 (13), 2717–2722. 33. Ruel-Garie`py,
E.; Leclair, G.; Hildgen, P.; Gupta, A.; Leroux, J.C.
Thermosensitive chitosan-based hydrogel containing
liposomes for the delivery of hydrophilic molecules. J.
Control. Release 2002, 82, 373–383. 34. Martin, L.; Wilson,
C.G.; Koosha, F.; Tetley, L.; Gray, A.I.; Senel, S.;
Uchegbu, I.F. The release of model macromolecules may be
controlled by the hydrophobicity of palmitoyl glycol
chitosan hydrogels. J. Control. Release 2002, 80, 87–100.
35. Martin, L.; Wilson, C.G.; Koosha, F.; Uchegbu, I.F.
Sustained buccal delivery of the hydrophobic drug
denbufylline using physically cross-linked palmitoyl glycol
chitosan hydrogels. Eur. J. Pharm. Biopharm. 2003, 55,
35–45. 36. Lee, K.Y.; Kim, J.-H.; Kwon, I.C.; Jeong, S.Y.
Selfaggregates of deoxycholic acid-modifed chitosan as a
novel carrier of adriamycin. Colloid Polym. Sci. 2000, 278,
1216–1219. 37. Janes, K.A.; Fresneau, M.P.; Marazuela, A.;
Fabra, A.; Alonso, M.J. Chitosan nanoparticles as delivery
systems for doxorubicin. J. Control. Release 2001, 73,
255–267. 38. De Campos, A.M.; Sa´nchez, A.; Alonso, M.J.
Chitosan nanoparticles: A new vehicle for the improvement
of the C delivery of drugs to the ocular surfaces
application to cyclosporin A. Int. J. Pharm. 2001, 224,
159–168.

39. Felt, O.; Gurny, R.; Buri, P.; Baeyens, V. Delivery of


antibiotics to the eye using a positively charged
polysaccharide as vehicle. AAPS Pharm. Sci. 2001, 3 (4),
1–7.

40. Ko, J.A.; Park, H.J.; Hwang, S.J.; Park, J.B.; Lee,
J.S. Preparation and characterization of chitosan
microparticles intended for controlled drug delivery. Int.
J. Pharm. 2002, 249, 165–174.

41. Gonza´lez-Rodrı`guez, M.L.; Holgado, M.A.;


Sa´nchezLafuente, C.; Rabasco, A.M.; Fini, A.
Alginate/chitosan particulate systems for sodium diclofenac
release. Int. J. Pharm. 2002, 232, 225–234.

42. Shikata, F.; Tokumitsu, H.; Ichikawa, H.; Fukumori, Y.


In vitro cellular accumulation of gadolinium incorporated
into chitosan nanoparticles designed for neutron-capture
therapy of cancer. Eur. J. Pharm. Biopharm. 2002, 53,
57–63.

43. Banerjee, T.; Mitra, S.; Singh, A.K.; Sharma, R.K.;


Maitra, A. Preparation, characterization and
biodistribution of ultrafine chitosan nanoparticles. Int. J.
Pharm. 2002, 243, 93–105.

44. Thanou, M.; Verhoef, J.C.; Junginger, H.E. Chitosan and


its derivatives as intestinal absorption enhancers. Adv.
Drug Deliv. Rev. 2001, 50, S91–S101.

45. van der Lubben, I.M.; Verhoef, J.C.; Borchard, G.;


Junginger, H.E. Chitosan and its derivatives in mucosal
drug and vaccine delivery. Eur. J. Pharm. Sci. 2001, 14,
201–207.

46. Thanou, M.; Kotze´, A.F.; Scharringhausen, T.; LueXen,


H.L.; de Boer, A.G.; Verhoef, J.C.; Junginger, H.E. Effect
of degree of quaternization of N-trimethyl chitosan
chloride for enhanced transport of hydrophilic compounds
across intestinal Caco-2 cell monolayers. J. Control.
Release 2000, 64, 15–25.

47. Thanou, M.; Florea, B.I.; Langemey¨er, M.W.E.; Verhoef,


J.C.; Junginger, H.E. N-trimethylated chitosan chloride
(TMC) improves the intestinal permeation of the peptide
drug buserelin in vitro (Caco-2 cells) and in vivo (rats).
Pharm. Res. 2000, 17, 27–31.

48. Thanou, M.; Verhoef, J.C.; Marbach, P.; Junginger, H.E.


Intestinal absorption of octreotide: N-trimethyl chitosan
(TMC) ameliorates the permeability and absorption
properties of the somatostatin analogue in vitro and in
vivo. J. Pharm. Sci. 2000 , 89, 951–957.

49. Thanou, M.; Verhoef, J.C.; Verheijden, J.H.M.;


Junginger, H.E. Intestinal absorption of octreotide using
N-trimethyl chitosan (TMC): Studies in pigs. Pharm. Res.
2001, 18, 823–828.

50. Vandenberg, G.W.; Drolet, C.; Scott, S.L.; de la Nou¨e,


J. Factors affecting protein release from alginate–chitosan
coacervate microcapsules during production and
gastric/intestinal simulation. J. Control. Release 2001,
77, 297–307.

51. Huang, Y.C.; Yeh, M.K.; Chiang, C.H. Formulation


factors in preparing BTM–chitosan microspheres by spray
drying method. Int. J. Pharm. 2002, 242, 239–242.
Chondroitin Sulfates

1. Volpi, N., Ed.; Analytical techniques to evaluate the


structure and function of natural polysaccharides,
glycosaminoglycans. Res. Signpost, India 2002.

2. Sugahara, K; Mikami, T.; Uyama, T.; Mizuguchi, S.;


Nomura, K.; Kitagawa, H. Recent advances in the structural
biology of chondroitin sulfate and dermatan sulfate. Curr.
Opin. Struct. Biol. 2003, 13, 612–620.

3. Hardingham, T.E.; Fosang, A.J. Proteoglycans: many forms


and many functions. FASEB J. 1992, 6, 861–870.

4. Jackson, R.L.; Busch, S.J.; Cardin, A.D.


Glycosaminoglycans: molecular properties, protein
interactions, and role in physiological processes. Physiol.
Rev. 1991, 71, 481–539.

5. Casu, B.; Choay, J.; Ferro, D.R.; Gatti, G.; Jacquinet,


J.C.; Petitou, M.; Provasoli, A.; Ragazzi, M.; Sinay, P.;
Torn, G. Controversial glycosaminoglycan conformations.
Nature 1986, 322, 215–216.

6. Karamanos, N.K.; Syrokou, A.; Vanky, P.; Nurminen, M.;


Hjerpe, A. Determination of 24 variously sulfated
galactosaminoglycanand hyaluronan-derived disaccharides by
high-performance liquid chromatography. Anal. Biochem.
1994, 221, 189–199. 7. Poole, A.R. Proteoglycans in health
and disease: structures and functions. Biochem. J. 1986,
236, 1–14. 8. Silbert, J.E. Metabolism and
structure-function relationship of connective tissue
glycosaminoglycans and proteoglycans. Curr. Organic Chem.
2004, 8, 395–411. 9. Volpi, N. The pathobiology of
osteoarthritis and the rationale for using the chondroitin
sulfate for its treatment. Curr. Drug. Targets-Immune
Endocr. Metab. Disord. 2004, 4, 119–127. 10. Volpi, N.
Chondroitin sulfate for the treatment of osteoarthritis.
Curr. Med. Chem. Anti-inflammatory Anti-allergy Agents.
2005, 4, 221–234. 11. Akiyama, H.; Sakai, S.; Linhardt,
R.J.; Goda, Y.; Toida, T.; Maitani, T. Chondroitin sulfate
structure affects its immunological activities on murine
splenocytes sensitized with ovalbumin. Biochem. J. 2004,
382, 269–278. 12. Trowbridge, J.M.; Gallo, R.L. Dermatan
sulfate: new functions from an old glycosaminoglycan.
Glycobiology 2002, 12, 117R–125R. 13. Linhardt, R.J.;
Hileman, R.E. Dermatan sulfate as a potential therapeutic
agent. Gen. Pharmacol. 1995, 26, 443–451. 14. Rebaudi, A.;
Silvestrini, P.; Trisi, P. Use of a resorbable
hydroxyapatite-collagen chondroitin sulfate material on
immediate postextraction sites: a clinical and histologic
study. Int. J. Periodontics Restorative Dent. 2003, 23,
371–379. 15. Kirker, K.R.; Luo, Y.; Nielson, J.H.; Shelby,
I; Prestwich, G.D. Glycosaminoglycan hydrogel films as
biointeractive dressings for wound healing. Biomaterials
2002, 23 , 3661–3671. C
Collagen

55. Weadock, K.; Olson, R.M.; Silver, F.H. Evaluation of


collagen crosslinking techniques. Biomater. Med. Dev.
Artif. Organs 1984, 11, 293–318.

56. Kato, Y.; Silver, F. Formation of continuous collagen


fibres: Evaluation of biocompatibility and mechanical
properties. Biomaterials 1990, 11, 169–175.

57. Weadock, K.S.; Miller, E.J.; Bellincampi, L.D.;


Zawadsky, J.P.; Dunn, M.G. Physical crosslinking of
collagen fibers: Comparison of ultraviolet irradiation and
dehydrothermal treatment. J. Biomed. Mater. Res. 1995, 29,
1373–1379.

58. Nehrer, S.; Breinan, H.A.; Ramappa, A.; Hsu, H.P.;


Minas, T.; Shortkroff, S.; Sledge, C.B.; Yannas, I.V.;
Spector, M. Chondrocyte-seeded collagen matrices implanted
in a chondral defect in a canine model. Biomaterials 1998,
19, 2313–2328.

59. Kato, Y.; Christiansen, D.; Hahn, R.; Shieh, S.;


Goldstein, J.; Silver, F. Mechanical properties of collagen
fibres: A comparison of reconstituted and rat tail tendon
fibres. Biomaterials 1989, 10, 38–42.

60. Cavallaro, J.; Kemp, P.; Kraus, K. Collagen fabrics as


biomaterials. Biotechnol. Bioeng. 1993, 43, 781–791.

61. Petite, H.; Frei, V.; Huc, A.; Herbage, D. Use of


diphenylphosphorylazide for cross-linking collagenbased
biomaterials. J. Biomed. Mater. Res. 1994, 28, 159–165.

62. Gustavson, K.H. The Chemistry and Reactivity of


Collagen; Academic Press: New York, 1956.

63. Fielding, A.M. Preparation of Neutral Salt Soluble


Collagen. In The Methodology of Connective Tissue Research;
Hall, D.A., Ed.; Joynson–Bruvvers: Oxford, 1976; 9–12. 64.
Trelstad, R.L. Immunology of Collagens. In Immunochemistry
of the Extracellular Matrix; Furthmayer, H., Ed.; CRC
Press: Boca Raton, FL, 1982; Vol. 1, 32–39. 65.
Chandrakasan, G.; Torchia, D.A.; Piez, K.A. Preparation of
intact monomeric collagen from rat tail tendon and skin and
the structure of the nonhelical ends in solution. J. Biol.
Chem. 1976, 251, 6062–6067. 66. Piez, K.A. Molecular and
Aggregate Structures of the Collagens. In Extracellular
Matrix Biochemistry; Piez, K.A., Reddi, A.H., Eds.;
Elsevier: New York, 1984; 1–40. 67. Hall, B.K. Cartilage;
Academic Press: New York, 1983. 68. Lee, C.H.; Singla, A.;
Lee, Y. Biomedical applications of collagen. Int. J. Pharm.
2001, 221, 1–22. 69. Nimni, M.E.; Harkness, R.D. Molecular
Structures and Functions of Collagen. In
Collagen–Biochemistry; Nimni, M.E., Ed.; CRC Press: Boca
Raton, FL, 1988; 1–79. 70. Chvapil, M.; Kronentahl, R.L.;
van Winkle, W., Jr. Medical and Surgical Applications of
Collagen. In International Review of Connective Tissue
Research; Hall, D.A., Jackson, D.S., Eds.; Academic Press:
New York, 1973; 1–61. 71. Gorham, S.D. Collagen. In
Biomaterials; Byrom, V., Ed.; Stockton Press: New York,
1991; 55–122. 72. Thies, C. Microcapsules as drug delivery
devices. Crit. Rev. Biomed. Eng. 1982, 8, 335–383.
Collagen Fixation

18. Park, S.N.; Lee, H.J.; Lee, K.H.; Suh, H. Biological


characterization of EDC-cross-linked collagen hyaluronic
acid matrix in dermal tissue restoration. Biomaterials
2003, 24, 1631–1641.

19. Daamen, W.F.; van Moerkerk, H.T.; Hafmans, T.;


Buttafoco, L.; Poot, A.A.; Veerkamp, J.H.; van Kuppevelt,
T.H. Preparation and evaluation of molecularly-defined
collagen-elastin-glycosaminoglycan scaffolds for tissue
engineering. Biomaterials 2003, 24, 4001–4009.

20. Petit, H.; Rault, I.; Huc, A.; Menasche, P.; Herbage,
D. Use of acyl azide method for cross-linking collagen-rich
tissues such as pericardium. J. Biomed. Mater. Res. 1990,
24, 179–187.

21. Petit, D.; Duval, J.-L.; Frei, V.; Addul-Malik, N.;


SigotLuizard, M.-F.; Herbage, D. Cytocompatibility of calf
pericardium treated by glutaraldehyde and by the acyl azide
methods in an organotypic culture model. Biomaterials 1995,
16, 1003–1008.

22. Weadcock, K.S.; Miller, E.J.; Bellincampi, L.D.;


Zawadsky, J.P.; Dunn, M.G. Physical crosslinking of
collagen fibers: Comparison of ultraviolet irradiation and
dehydrothermal treratment. J. Biomed. Mater. Res. 1995, 29,
1373–1379.

23. Wang, M.-C.; Pins, G.D.; Silver, F.H. Collagen fibers


with improved strength for the repair of soft tissue
injuries. Biomaterials 1994, 15, 507–512.

24. Girton, T.S.; Oegema, T.R.; Tranquillo, R.T. Exploiting


glycation to stiffen and strengthen tissue equivalents for
tissue engineering. J. Biomed. Mater. Res. 1999, 46, 87–92.

25. Girton, T.S.; Oegema, T.R.; Grassl, E.D.; Isenberg,


B.C.; Tranquillo, R.T. Mechanisms of stiffening and
strengthening in media-equivalents fabricated using
glycation. J. Biomech. Eng. 2000, 122, 216–223.

26. Ohan, M.P.; Weadcoak, K.S.; Dunn, M.G. Synergistic


effects of glucose and ultraviolet irradiation on the
physical properties of collagen. J. Biomed. Mater. Res.
2002, 60, 384–391. 27. Sung, H.-W.; Huang, R.-N.; Huang,
L.L.H.; Tsai, C.-C.; Chiu, C.-T. Feasibility study of a
natural crosslinking reagent for biological tissue fixation.
J. Biomed. Mater. Res. 1998, 42, 560–567. 28. Sung, H.-W.;
Chang, Y.; Chiu, C.-T.; Chen, C.-N.; Liang, H.-C.
Crosslinking characteristics and mechanical properties of a
bovine pericardium fixed with a naturally occurring
crosslinking agent. J. Biomed. Mater. Res. 1999, 47,
116–126. 29. Huang, L.L.H.; Sung, H.-W.; Tsai, C.-C.;
Huang, D.-M. Biocompatibility study of a biological tissue
fixed with a naturally occurring crosslinking reagent. J.
Biomed. Mater. Res. 1998, 42, 568–576. 30. Koob, T.J.
Biomimetic approaches to tendon repair. Comp. Biochem.
Physiol., Part A 2002, 133, 1171–1192. 31. Koob, T.J.;
Hernandez, D.J. Material properties of polymerized
NDGA-collagen composite fibers: Development of biologically
based tendon constructs. Biomaterials 2002, 23, 203–212.
32. Koob, T.J.; Willis, T.A.; Hernandez, D.H.
Biocompatibility of NDGA polymerized fibers. I. In vitro
evaluation of cytotoxicity with tendon fibroblasts. J.
Biomed. Mater. Res. 2001, 56, 31–39. 33. Koob, T.J.;
Willis, T.A.; Qiu, Y.S.; Hernandez, D.H. Biocompatibility
of NDGA polymerized fibers. II. Attachment, replication and
migration of tendon fibroblasts. J. Biomed. Mater. Res.
2001, 56, 40–48. 34. Weadcock, K.; Olson, R.M.; Silver,
F.H. Evaluation of collagen crosslinking techniques.
Biomater. Med. Dev. Artif. Organs 1983–1984, 11, 293–318.
35. Rault, I.; Herbage, D.; Abdul-Mulak, N.; Huc, A.
Evaluation of different chemical methods for crosslinking
collagen gel, films and sponges. J. Mater. Sci., Mater. Med.
1996, 7, 215–221. 36. Pachence, J.M. Collagen-based devices
for soft tissue repair. J. Biomed. Mater. Res. 1996, 33,
35–40. 37. Lee, C.H.; Singla, A.; Lee, Y. Biomedical
applications of collagen. Int. J. Pharm. 2001, 221, 1–22.
38. Ramshaw; Werkmeister, J.A.; Glattauer, V. Collagenbased
biomaterials. Biotechnol. Genet. Eng. Rev. 1995, 13,
335–382. 39. Bailey, A.J. The fate of collagen implants in
tissue defects. Wound Repair Regen. 2000, 8, 5–12. C
Collagen Processing

1. The Olde Hide House. History of Leather; 2003.


www.leathertown.com. Ontario, Canada.

2. Heidemann, E. The Chemistry of Tanning. In Collagen;


Nimni, M.E., Ed.; CRC Press: Boca Raton, FL, 1988; Vol. 3,
39–62.

3. Miller, E.J.; Rhodes, R.K. Preparation and


Characterization of the Different Types of Collagen.
InMethods in Enzymology; Cunningham, L.W., Frederiksen,
D.W., Eds.; Academic Press: New York, 1982; Vol. 82, 33–64.

4. Wallace, D.G.; Thompson, A. Description of collagen


fibril formation by a theory of polymer crystallization.
Biopolymers 1983, 22, 1793–1811.

5. Wood, G.C. The precipitation of collagen fibers from


solution. Biochem. J. 1960, 75, 598–605.

6. Cassel, J.M.; Mandelkern, L.; Roberts, D.F. The kinetics


of the heat precipitation of collagen. J. Am. Leather Chem.
Assoc. 1962, 57, 556–575.

7. Cassel, J.M. Collagen aggregation phenomena. Biopolymers


1966, 4, 989–997.

8. Yannas, I.V. Collagen and gelatin in the solid state. J.


Macromol. Sci., Rev. Macromol. Chem. 1972, C7 (1), 49–104.

9. Comper, W.D.; Vies, A. The mechanism of nucleation for


in vitro collagen fibril formation. Biopolymers 1977, 16,
2113–2131.

10. Trelstad, R.L.; Birk, D.E.; Silver, F.H. Collagen


fibrillogenesis in tissues, in solution and from modeling: A
synthesis. J. Invest. Dermatol. 1982, 79, 109s–112s.

11. Silver, F.H.; Birk, D.E. Kinetic analysis of collagen


fibrillogenesis: I. Use of turbidity–time data. Collagen
Relat. Res. 1983, 3, 393–405.

12. Birk, D.E.; Silver, F.H. Collagen fibrillogenesis in


vitro: Comparison of types I, II, and III. Arch. Biochem.
Biophys. 1984, 235, 178–185.

13. Birk, D.E.; Silver, F.H. Kinetic analysis of collagen


fibrillogenesis: II. Corneal and scleral type I collagen.
Collagen Relat. Res. 1984, 4, 265–277. 14. Holmes, D.F.;
Capaldi, M.J.; Chapman, J.A. Reconstitution of collagen
fibrils in vitro; the assembly process depends on the
initiating procedure. Int. J. Macromol. 1986, 8, 161–166.
15. Yannas, I.V.; Lee, E.; Orgill, D.P.; Skrabut, E.M.;
Murphy, G.F. Synthesis and characterization of a model
extracellular matrix that induces partial regeneration of
adult mammalian skin. Proc. Natl. Acad. Sci. 1989, 86,
933–937. 16. Birk, D.E.; Fitch, J.M.; Babiarz, J.P.; Doane,
K.J. Collagen fibrillogenesis in vitro: Interaction of types
I and V collagen regulates fibril diameter. J. Cell. Sci.
1990, 95, 649–657. 17. Veis, A.; Payne, K. Collagen
Fibrillogenesis. In Collagen; Nimni, M.E., Ed.; CRC Press:
Boca Raton, FL, 1988; Vol. 1, 113–137. 18. Birk, D.E.;
Nurminskaya, M.V.; Zycband, E.I. Collagen fibrillogenesis in
situ: Fibril segments undergo postdepositional modifications
resulting in linear and lateral growth during matrix
development. Dev. Dyn. 1995, 202, 229–243. 19. Obrink, B.;
Wasteson, A. Nature of the interaction of chondriotin
4-sulfate and chondriotin sulfateproteoglycan with
collagen. Biochem. J. 1971, 121, 227–233. 20. Obrink, B.A.
Study of the interactions between monomeric tropocollagen
and glycosaminoglycans. Eur. J. Biochem. 1973, 33, 387–400.
21. Vogel, K.G.; Paulsson, M.; Heinegard, D. Specific
inhibition of type I and type II collagen fibrillogenesis by
the small proteoglycan of tendon. Biochem. J. 1984, 223,
587–597. 22. Reed, R.K.; Aukland, K. Transcapillary fluid
balance in immature rats. Interstitial fluid pressure, serum
and interstitial protein concentration, and colloid osmotic
pressure. Microvasc. Res. 1977, 14, 37–43. 23. Aukland, K.;
Reed, R.K. Interstitial-lymphatic mechanisms in the control
of extracellular fluid volume. Physiol. Rev. 1993, 73 (1),
1–78. 24. Zurovsky, Y.; Mitchell, G.; Hattingh, J.
Composition and viscosity of interstitial fluid of rabbits.
Exp. Physiol. 1995, 80, 203–207. 25. Borg, T.K.; Rubin, K.;
Carver, W.; Samuel, A.; Terracio, L. The cell biology of
the cardiac interstitium. TCM 1996, 6 (2), 65–70.
Composites

23. Dewey, J.M. The elastic constants of materials loaded


with non-rigid fillers. J. Appl. Phys. 1947, 18 (6),
578–581.

24. Landauer, R. The electrical resistance of binary


metallic mixtures. J. Appl. Phys. 1952, 23 (7), 779–784.

25. Hershey, A.V. The elasticity of an isotropic aggregate


of anisotropic cubic crystals. J. Appl. Mech., Trans. ASME
1954, 21 (3), 236–240.

26. Brown, W.F. Solid mixture permittivities. J. Chem.


Phys. 1955, 23 (8), 1514–1517.

27. Eshelby, J.D. The determination of the elastic field of


an ellipsoidal inclusion, and related problems. Proc. R.
Soc. Lond., A Math. Phys. Sci. 1957, 241 (1226), 376–396.

28. Kerner, E.H. The electrical conductivity of composite


media. Proc. Phys. Soc. Lond., B 1956, 69 (8), 802–807.

29. Van Der Pol, C. On the rheology of concentrated


dispersions. Rheol. Acta 1958, 1, 198.

30. Meredith, R.E.; Tobias, C.W. Resistance to potential


flow through a cubical array of spheres. J. Appl. Phys.
1960, 31 (7), 1270–1273.

31. Hashin, Z. The elastic moduli of heterogeneous


materials. J. Appl. Mech., Trans. ASME 1962, 29 (3),
143–150.

32. Hashin, Z.; Shtrikman, S. A variational approach to the


theory of the elastic behaviour of polycrystals. J. Mech.
Phys. Solids 1962, 10 (4), 343–352.

33. Keller, J.B. Theorem on conductivity of composite


medium. J. Math. Phys. 1964, 5 (4), 548.

34. Hashin, Z.; Rosen, B.W. The elastic moduli of


fiberreinforced materials. J. Appl. Mech., Trans. ASME 1964,
31, 233.

35. Hill, R. Theory of mechanical properties of


fibrestrengthened materials. 1. Elastic behaviour. J. Mech.
Phys. Solids 1964, 12 (4), 199–212.

36. Hill, R. A self-consistent mechanics of composite


materials. J. Mech. Phys. Solids 1965, 13 (4), 213.

37. Budiansk, B. On elastic moduli of some heterogeneous


materials. J. Mech. Phys. Solids 1965, 13 (4), 223.

38. Hashin, Z. Viscoelastic fiber reinforced materials. AIAA


J. 1966, 4 (8), 1411.

39. Russel, W.B.; Acrivos, A. Effective moduli of


composite-materials—Slender rigid inclusions at dilute
concentrations. Zeit. Angew. Math. Phys. 1972 , 23 (3),
434–463.

40. Russel, W.B. Effective moduli of


composite-materials—Effect of fiber length and geometry at
dilute concentrations. Zeit. Angew. Math. Phys. 1973, 24
(4), 581–600.

41. Mendelson, K.S. Theorem on effective conductivity of a


two-dimensional heterogeneous medium. J. Appl. Phys. 1975,
46 (11), 4740–4741.

42. Doyle, W.T. Clausius-mossotti problem for cubic arrays


of spheres. J. Appl. Phys. 1978, 49 (2), 795–797.

43. Mcphedran, R.C.; Mckenzie, D.R. Conductivity of


lattices of spheres. 1. Simple cubic lattice. Proc. R. Soc.
Lond., A Math. Phys. Eng. Sci. 1978, 359 (1696), 45–63.

44. Landauer, R. Stability in dissipative steady-state.


Phys. Today 1978, 31 (11), 23–29.

45. Perrins, W.T.; Mckenzie, D.R.; Mcphedran, R.C.


Transport-properties of regular arrays of cylinders. Proc.
R. Soc. Lond., A Math. Phys. Eng. Sci. 1979, 369 (1737),
207–225. 46. Christensen, R.M.; Lo, K.H. Solutions for
effective shear properties in 3 phase sphere and cylinder
models. J. Mech. Phys. Solids 1979, 27 (4), 315–330. 47.
Mclachlan, D.S. Measurement and analysis of a model
dual-conductivity medium using a generalized
effective-medium theory. J. Phys. C. Solid State Phys.
1988, 21 (8), 1521–1532. 48. Bao, K.D.; Axell, J.;
Grimvall, G. Electrical-conduction in checkerboard
geometries. Phys. Rev., B 1990, 41 (7), 4330–4333. 49.
Guedes, J.M.; Kikuchi, N. Preprocessing and postprocessing
for materials based on the homogenization method with
adaptive finite-element methods. Comput. Methods Appl. Mech.
Eng. 1990, 83 (2), 143–198. 50. Gu, G.Q.; Liu, Z.R.
Thermal-conductivity of periodic composite media with
spherical inclusions. Commun. Theor. Phys. 1991, 15 (2),
141–148. 51. Gu, G.Q. Calculation methods for effective
constants of periodic composite media. J. Phys., D. Appl.
Phys. 1993, 26 (9), 1371–1377. 52. Christensen, R.M.
Effective properties of compositematerials containing
voids. Proc. R. Soc. Lond., A Math. Phys. Eng. Sci. 1993,
440 (1909), 461–473. 53. Lu, S.Y. The effective
thermal-conductivities of composites with 2-d arrays of
circular and square cylinders. J. Compos. Mater. 1995, 29
(4), 483–506. 54. Batchelor, G.K. Transport properties of
2-phase materials with random structure. Annu. Rev. Fluid
Mech. 1974, 6, 227–255. 55. Whitney, J.M.; Mccullough, R.L.
Micro-Models for Predicting Viscoelastic Behavior. In
Delaware Composites Design Encyclopedia; Whitney, J.M.,
Mccullough, R.L., Eds.; Technomic Pub. Co.: Lancaster, PA,
USA, 1990; Vol. 2, 189–201. 56. Sastry, A.M. 2.17
Impregnation and Consolidation Phenomena. In Comprehensive
Composite Materials; Kelly, A., Zweben, C.H., Eds.;
Elsevier: Amsterdam, 2000; Vol. II, 609–622. 57. Fung, Y.C.
Introduction to Bioengineering; World Scientific: Singapore,
2001. 58. Torquato, S. Random Heterogeneous Materials:
Microstructure and Macroscopic Properties; Springer: New
York, 2002. 59. Tsai, S.W.; Wu, E.M. General theory of
strength for anisotropic materials. J. Compos. Mater. 1971,
5, 58. (JAN). 60. Tsai, S.W.; Hahn, H.T. Introduction to
Composite Materials; Technomic Pub. Co.: Westport, CT,
1980. 61. Sastry, A.M.; Phoenix, S.L. Load redistribution
near nonaligned fiber breaks in a 2-dimensional
unidirectional composite using break-influence
superposition. J. Mater. Sci. Lett. 1993, 12 (20),
1596–1599. 62. Sastry, A.M.; Phoenix, S.L.; Schwartz, P.
Analysis of interfacial failure in a composite microbundle
pullout experiment. Compos. Sci. Technol. 1993, 48 (1–4),
237–251. 63. Sastry, A.M.; Phoenix, S.L. Shielding and
magnification of loads in elastic, unidirectional
composites. SAMPE J. 1994, 30 (4), 61–67. 64. Beyerlein,
I.J.; Phoenix, S.L.; Sastry, A.M. Comparison of shear-lag
theory and continuum fracture mechanics for modeling fiber
and matrix stresses in an elastic cracked composite lamina.
Int. J. Solids Struct. 1996, 33 (18), 2543–2574. 65.
Phoenix, S.L. Statistical Strength Theory for Fibrous
Composite Materials. In Comprehensive Composite Materials;
Kelly, A., Zweben, C.H., Eds.; Elsevier: Amsterdam, 2000;
Vol. I, 559. 66. Reddy, J.N. Mechanics of Laminated
Composite Plates: Theory and Analysis; CRC Press: Boca
Raton, 1997. 67. Hyer, M.W. Stress Analysis of
Fiber-Reinforced Materials; WCB McGraw-Hill: Boston, 1998.
Especially Chapter 3. 68. Reuss, A. Berechnung der
fliessgrenze kristallen von mischkristallen auf grund der
plastizitatsbedingung fur einkristalle. Math. Mech. Z.
Angew. 1929, 9, 49. 69. Voigt, W. Lehrbuch der
Kristallphysik; B.G. Teubner: Leipzig, 1910. 70. Boucher,
S. On the effective moduli of isotropic twophase elastic
composites. J. Compos. Mater. 1974, 8 (82). 71. Hershey,
A.V. The plasticity of an isotropic aggregate of
anisotropic face-centered cubic crystals. J. Appl. Mech.,
Trans. ASME 1954, 21 (3), 241–249. 72. Kro¨ner, E.
Berechnung der elastischen konstanten des vielkristalls aus
den konstanten des einkristalls. Zeit. Phys. 1958, 151 (4),
504–518. 73. Hashin, Z.; Shtrikman, S. A variational
approach to theory of effective magnetic permeability of
multiphase materials. J. Appl. Phys. 1962, 33 (10), 3125.
74. Hashin, Z.; Shtrikman, S. On some variational
principles in anisotropic and nonhomogeneous elasticity. J.
Mech. Phys. Solids 1962, 10 (4), 335–342. 75. Hashin, Z.;
Shtrikman, S. A variational approach to the theory of the
elastic behaviour of multiphase materials. J. Mech. Phys.
Solids 1963, 11 (2), 127–140. 76. Mccullough, R.L.
Micro-Models for Composite Materials. In Delaware
Composites Design Encyclopedia; Whitney, J.M., Mccullough,
R.L., Eds.; Technomic Pub. Co.: Lancaster, PA, USA, 1990;
2, 49–142. 77. http://www.matweb.com (2003). 78. Kriz,
R.D.; Stinchcomb, W.W. Elastic-moduli of transversely
isotropic graphite fibers and their composites. Exp. Mech.
1979, 19 (2), 41–49. 79. http://structures.ucsd.edu (2003).
80. Bergman, D.J. Analytical properties of the complex
effective dielectric constant of a composite medium with
applications to the derviation of rigrous bounds and to
percolation problems. 81. Lurie, K.A.; Cherkaev, A.V. Exact
estimates of conductivity of composites formed by 2
isotropically conducting media taken in prescribed
proportion. Proc. R. Soc. Edinb., Sect. A, Math. 1984, 99,
71–87. 82. Milton, G.W.; Kohn, R.V. Variational bounds on
the effective moduli of anisotropic composites. J. Mech.
Phys. Solids 1988, 36 (6), 597–629. 83. Gibiansky, L.V.;
Torquato, S. Link between the conductivity and
elastic-moduli of composite-materials. Phys. Rev. Lett.
1993, 71 (18), 2927–2930. 84. Torquato, S.; Gibiansky,
L.V.; Silva, M.J.; Gibson, L.J. Effective mechanical and
transport properties of cellular solids. Int. J. Mech. Sci.
1998, 40 (1), 71–82. 85. Lu, W.T.; Carlsson, L.A.;
Andersson, Y. Micro model of paper. 1. Bounds on elastic
properties. Tappi J. 1995, 78 (12), 155–164. 86. Lu, W.T.;
Carlsson, L.A. Micro-model of paper. 2. Statistical
analysis of the paper structure. Tappi J. 1996, 79 (1),
203–210. 87. Lu, W.T.; Carlsson, L.A. Influence of
viscoelastic behavior on curl of paper. Mech. Time-Depend.
Mater. 2001, 5 (1), 79–100. 88. Ostoja-Starzewski, M.;
Sheng, P.Y.; Jasiuk, I. Damage patterns and constitutive
response of random matrixinclusion composites. Eng. Fract.
Mech. 1997, 58 (5– 6), 581–606. 89. Cheng, X.; Sastry, A.M.
On transport in stochastic, heterogeneous fibrous domains.
Mech. Mater. 1999, 31 (12), 765–786. 90. Wang, C.W.;
Berhan, L.; Sastry, A.M. Structure, mechanics and failure
of stochastic fibrous networks: Part I—Microscale
considerations. J. Eng. Mater. Technol. Trans. ASME 2000,
122 (4), 450–459. 91. Wang, C.W.; Sastry, A.M. Structure,
mechanics and failure of stochastic fibrous networks: Part
II— Network simulations and application. J. Eng. Mater.
Technol. Trans. ASME 2000, 122 (4), 460–468. 92. Berhan,
L.; Yi, Y.B.; Sastry, A.M. Effect of nanorope waviness on
the effective moduli of nanotube sheets. J. Appl. Phys.
2004, 95 (8), 4335–4345. 93. Berhan, L.; Yi, Y.B.; Sastry,
A.M.; Munoz, E.; Selvidege, M.; Baughman, R. Mechanical
properties of nanotube sheets: Alteration in joint
morphology, and achievable moduli in manufacturable
materials. J. Appl. Phys. 2004, 95 (9), 5027–5034. 94. Yi,
Y.B.; Berhan, L.; Sastry, A.M. Statistical geometry of
random fibrous networks, revisited: Waviness, dimensionality
and percolation. J. Appl. Phys. 2004. Accepted. 95. Wang,
C.W.; Cook, K.A.; Sastry, A.M. Conduction in multiphase
particulate/fibrous networks simulations and experiments on
li-ion anodes. J. Electrochem. Soc. 2003, 150 (3),
A385–A397. 96. Sastry, A.M.; Cheng, X.; Wang, C.W.
Mechanics of stochastic fibrous networks. J. Thermoplast.
Compos. Mater. 1998, 11 (3), 288–296. 97. Cheng, X.; Wang,
C.W.; Sastry, A.M.; Choi, S.B. Investigation of failure
processes in porous battery substrates: Part II—Simulation
results and comparisons. J. Eng. Mater. Technol. Trans.
ASME 1999, 121 (4), 514–523. 98. Wang, C.W.; Cheng, X.;
Sastry, A.M.; Choi, S.B. Investigation of failure processes
in porous battery substrates: Part I—Experimental findings.
J. Eng. Mater. Technol. Trans. ASME 1999, 121 (4), 503–513.
99. Nielaba, P.; Privman, V. Multilayer adsorption with
increasing layer coverage. Phys. Rev., A 1992, 45 (8),
6099–6102. C

100. Deckmyn, P.; Davies, G.A.; Bell, D.J. Properties of


percolating clusters on finite lattices applied to model
filtration processes. Appl. Math. Model. 1995, 19 (5),
258–269.

101. Datta, S.; Redner, S. Gradient and percolative


clogging in depth filtration. Int. J. Mod. Phys. C 1998, 9
(8), 1535–1543.

102. Cheng, X.; Sastry, A.M.; Layton, B.E. Transport in


stochastic fibrous networks. J. Eng. Mater. Technol. Trans.
ASME 2001, 123 (1), 12–19.

103. Mohanty, S.; Sharma, M.M. A Monte-Carlo RSRG method


for the percolation conduction properties of correlated
lattices. Phys. Lett., A 1991, 154 (9), 475–481.

104. Kirkpatrick, S. Percolation and conduction. Rev. Mod.


Phys. 1973, 45 (4), 574–588.

105. Pike, G.E.; Seager, C.H. Percolation and conductivity—


Computer study. 1. Phys. Rev., B 1974, 10 (4), 1421–1434.

106. Yi, Y.B.; Sastry, A.M. Analytical approximation of the


two-dimensional percolation threshold for fields of
overlapping ellipses. Phys. Rev., E 2002, 66 (6). Art.
no.-066130. 107. Yi, Y.B.; Sastry, A.M. Analytical
approximation of the percolation threshold for overlapping
ellipsoids of revolution. Proc. R. Soc. Lond., A 2004,
accepted. 108. Haan, S.W.; Zwanzig, R. Series expansions in
a continuum percolation problem. J. Phys., A, Math. Gen.
1977, 10 (9), 1547–1555. 109. Coniglio, A.; Deangelis, U.;
Forlani, A.; Lauro, G. Distribution of physical clusters.
J. Phys., A, Math. Gen. 1977, 10 (2), 219–228. 110.
Coniglio, A.; Deangelis, U.; Forlani, A. Pair connectedness
and cluster size. J. Phys., A, Math. Gen. 1977, 10 (7),
1123–1139. 111. Hill, T.L. Molecular clusters in imperfect
gases. J. Chem. Phys. 1955, 23 (4), 617–622. 112. Drory, A.
Theory of continuum percolation. I. General formalism.
Phys. Rev., E 1996, 54 (6), 5992–6002. 113. Quintanilla,
J.; Torquato, S. Clustering properties of d-dimensional
overlapping spheres. Phys. Rev., E 1996, 54 (5), 5331–5339.
114. Quintanilla, J.; Torquato, S. Clustering in a
continuum percolation model. Adv. Appl. Probab. 1997, 29
(2), 327–336.
Compression of Digital Biomedical Signals

31. Al-Fahoum, A.S. Quality assessment of ECG compression


techniques using a wavelet-based diagnostic tool. IEEE
Trans. Inf. Technol. Biomed. 2006, 10 (1), 182–191.

32. Stern, R.; Ray, W.; Quigley, K. Psychophysiological


Recording, 2nd ed. Ed.; Oxford University Press: New York,
2001.

33. Farina, D.; Merletti, R. Comparison of algorithms for


estimation of EMG variables during voluntary isometric
contractions. J. Electromyogr. Kinesiol. 2000, 10 (5),
337–349.

34. Cuerrero, A.; Mailhes, C. On the choice of an


electromyogram data compression method, Proceedings of the
19th Annual International Conference of the IEEE
Engineering in Medicine and Biology Society: Chicago, IL,
1997; 1558–1561.

35. Wellig, P.; Cheng, Z.; Semling, M.; Moschytz, G.S.


Electromyogram data compression using single-tree and
modified zero-tree wavelet encoding, Proceedings of the 20th
Annual International Conference of the IEEE Engineering in
Medicine and Biology Society, Oct. 29– Nov. 1, 1998; 3,
1303–1306.

36. Berger, P.A.; Nascimento, F.A.O.; Carmo, J.C.; Rocha,


A.F.; dos Santos, I. Algorithm for compression of EMG
signals, Proceedings of the 25th Annual International
Conference of the IEEE Engineering in Medicine and Biology
Society, Sept. 17–21, 2003; 2, 1299–1302.

37. Chan, A.; Lovely, D.; Hudgins, B. Errors associated


with the use of adaptive differential pulse code modulation
in the compression of isometric and dynamic myo-electric
signals. Med. Biol. Eng. Comp. 1997, 36, 215–219.

38. Carotti, E.; De Martin, J.C.; Merletti, R.; Farina, D.


Compression of surface EMG signals with algebraic code
excited linear prediction. Med. Eng. Phys. 2007, 29 (2),
253–258.

39. Gro¨nfors, T.; Pa¨ivinen, N. Comparison of vector


quantization methods for medical fidelity preserving lossy
compression of EMG signals. International Conference on
Computional Intelligence for Modelling Control and
Automation CIMCA 2005, Wien: Austria, Nov. 28–30, 2005.
40. Gro¨nfors, T.; Reinikainen, M.; Sihvonen, T. Vector
quantization as a method for integer EMG signal
compression. J. Med. Eng. Techn. 2006, 30 (1), 41–52. 41.
Tossavainen, T.; Juhola, M. Lossless compression of
otoneurological eye movement signals. J. Clin. Monit.
Comput. 2002, 17, 393–402. 42. Tossavainen, T.; Juhola, M.;
Gro¨nfors, T. Lossy compression of eye movement and
auditory brainstem response signals. Comput. Methods
Programs Biomed. 2003, 72, 43–54. 43. Juhola, M.;
Tossavainen, T.; Aalto, H. Influence of lossy compression on
eye movement signals. Comput. Biol. Med. 2004, 34, 221–239.
44. Jewett, D.; Williston, J. Auditory-evoked far fields
averaged from the scalp of humans. Brain 1971, 94, 681–696.
45. Antoniol, G.; Tonella, P. EEG data compression
techniques. IEEE Trans. Biomed. Eng. 1997, 44 (2), 105–
114. 46. Magotra, N.; Mandyam, G.; Mingui, S.; McCoy, W.
Lossless compression of electroencephalographic (EEG) data,
IEEE International Symposium on Circuits and Systems,
‘‘Connecting the World,’’ 1996; 2, 313–315. 47. Yi, K.C.;
Sun, M.; Li, C.C.; Sclabassi, R.J. A lossless compression
algorithm for multichannel EEG, Proceedings of the First
Joint BMES/EMBS Conference, 1999; 1, 429. 48. Sriraam, N.;
Kannan, R.; Eswaran, C. Lossless compression of EEG data
using neural network predictors, Proceedings of the 9th
International Conference on Neural Information Processing
(ICONIP ‘OZ), 2002; 4, 2046–2048. 49. Folkers, A.; Mosch,
F.; Malina, T.; Hofmann, U.G. Realtime bioelectrical data
acquisition and processing from 128 channels utilizing the
wavelet-transformation. Neurocomputing 2003, 52–54,
247–254. 50. Cinkler, J.; Kong, X.; Memon, N. Lossless and
nearlossless compression of EEG signals. Conference Record
of the 31st Asilomar Conference on Signals, Systems, &
Computers, 1997; 2. 51. Memon, N.; Kong, X.; Cinkler, J.
Context-based lossless and near-lossless compression of EEG
signals. IEEE Trans. Inf. Technol. Biomed. 1999, 3 (3),
231–238. 52. Mitra, S.K.; Sarbadhikari, S.N. Iterative
function system and genetic algorithm based EEG
compression. Med. Eng. Phys. 1997, 19, 605–617. 53.
Sijercˇic´, Z.; Agarwal, G.C.; Anderson, C.W. EEG signal
compression with ADPCM subband coding. IEEE 39th Midwest
Symposium on Circuits and Systems, 1996; 2, 695–698.
Computer-Assisted Surgery

12. Kypson, A.P.; Chitwood, W.R., Jr. Robotic mitral valve


surgery. Am. J. Surg. 2004, 188 (Suppl. 4A), 83S–88S.

13. Chitwood, W.R., Jr.; Nifong, L.W. Minimally invasive


videoscopic mitral valve surgery: the current role of
surgical robotics. J. Card. Surg. 2000, 15, 61–75.

14. Mohr, F.W.; Falk, V.; Diegeler, A.; Walther, T.;


Gummert, J.F.; Bucerius, J.; Jacobs, S.; Autschbach, R.
Computer-enhanced ‘‘robotic’’ cardiac surgery: experience
in 148 patients. J. Thorac. Cardiovasc. Surg. 2001, 121,
842–853. 15. Kypson, A.P.; Nifong, L.W.; Chitwood, W.R.,
Jr. Robotic mitral valve surgery. Semin. Thorac.
Cardiovasc. Surg. 2003, 15, 121–129. 16. Smith, J.A., Jr.
Robotically assisted laparoscopic prostactomy: an
assessment of its contemporary role in the surgical
management of localized prostate cancer. Am. J. Surg. 2004,
188 (Suppl. 4A), 63S–67S. 17. Hanly, E.J.; Talamini, M.A.
Robotic abdominal surgery. Am. J. Surg. 2004, 188 (Suppl.
4A), 19S–26S. 18. Tewari, A.; Menon, M. Vattikuti institute
prostatectomy: surgical technique and current results.
Curr. Urol. Rep. 2003, 4, 119–123. 19. Camarillo, D.B.;
Krummel, T.M.; Salisbury, J.K., Jr. Robotic technology in
surgery: past, present, and suture. Am. J. Surg. 2004, 188
(Suppl 4A), 2S–157S.
Conductive Polymers

1. Lavine, L.S.; Grodzinsky, A.J. Current concepts review


electrical stimulation of repair of bone. J. Bone Jt. Surg.
1987, 69-A (4), 626–630.

2. Karba, R.; Semrov, D.; Vodovnik, L.; Benko, H.; Savrin,


R. DC electrical stimulation for chronic wound healing
enhancement. Part 1. Clinical study and determination of
electrical field distribution in the numerical wound model.
Bioelectrochem. Bioenerg. 1997, 43, 265–270.

3. Kerns, J.M.; Fakhouri, A.J.; Weinrib, H.P.; Freeman,


J.A. Electrical stimulation of nerve regeneration in the
rat: The early effects evaluated by a vibrating probe and
electron microscopy. Neuroscience 1991, 40 (1), 93–107.

4. Skotheim, T.A.; Elsenbaumer, R.L.; Reynolds, J.R.


Handbook of Conducting Polymers, 2nd Ed.; Marcel Dekker:
New York, 1998.

5. Wallace, G.G.; Teasdale, P.R.; Spinks, G. Conductive


electroactive polymers: Intelligent material systems;
Technomic Publ. Co.: Lancaster, 1997.

6. Krische, B.; Zagorska, M. The polythiophene paradox.


Synth. Met. 1989, 28 (1–2), C263–C268.

53. Cui, X.; Hetke, J.F.; Wiler, J.A.; Anderson, D.J.;


Martin, D.C. Electrochemical deposition and
characterization of conducting polymer polypyrrole/PSS on
multichannel neural probes. Sens. Actuators, A 2001, 93,
8–18.

54. Wong, J.Y.; Langer, R.; Ingber, D.E. Electrically


conducting polymers can noninvasively control the shape and
growth of mammalian cells. Proc. Natl. Acad. Sci. 1994, 91,
3201–3204.

55. Garner, B.; Hodgson, A.J.; Wallace, G.G.; Underwood,


P.A. Human endothelial cell attachment to and growth on
polypyrrole-heparin is vitronectin dependent. J. Mater.
Sci., Mater. Med. 1999, 10, 19–27.

56. Kotwal, A.; Schmidt, C.E. Electrical stimulation alters


protein adsorption and nerve cell interactions with
electrically conducting biomaterials. Biomaterials 2001,
22, 1055–1064.

57. Hodgson, A.J.; Gilmore, K.; Small, C.; Wallace, G.G.;


MacKenzie, I.L.; Aoki, T.; Ogata, N. Reactive
supramolecular assemblies of mucopolysaccharide,
polypyrrole and protein as controllable biocomposites for a
new generation of ‘intelligent biomaterials’. Supramol.
Sci. 1994, 1, 77–83.

58. Hodgson, A.J.; John, M.J.; Campbell, T.; Georgevich,


A.; Woodhouse, S.; Aoki, T.; Ogata, N.; Wallace, G.G.
Integration of biocomponents with synthetic structuresuse
of conducting polymer polyelectrolyte composites. SPIE Int.
Soc. Opt. Eng. 1996, 2716, 164–176.

59. Schmidt, C.E.; Shastri, V.R.; Vacanti, J.P.; Langer, R.


Stimulation of neurite outgrowth using an electrically
conducting polymer. Proc. Natl. Acad. Sci. 1997, 94,
8948–8953.

60. Chen, S.J.; Wang, D.Y.; Yuan, C.W.; Wang, X.D.; Zhang,
P.Y.; Gu, X.S. Template synthesis of the polypyrrole tube
and its bridging in vivo sciatic nerve regeneration. Mater.
Sci. Lett. 2000, 19, 2157–2159. 61. Shastri, V.R.; Schmidt,
C.E.; Kim, T.H.; Vacanti, J.P.; Langer, R. Polypyrrole—a
potential candidate for stimulated nerve regeneration.
Mater. Res. Soc. Symp. Proc. 1996, 414, 113–118. 62.
Garner, B.; Georgevich, A.; Liu, L.; Hodgson, A.J.;
Wallace, G.G. Polypyrrole-heparin composites as
stimulus-responsive substrates for endothelial cell growth.
J. Biomed. Mater. Res. 1999, 44, 121–129. 63. De Giglio,
E.; Sabbatini, L.; Zambonin, P.G. Development and
analytical characterization of cystein-grafted polypyrrole
films electrosynthesized on Pt-and Ti-substrates as
precursors of bioactive interfaces. Biomater. Sci. Polym.
Ed. 1999, 10 (8), 845–858. 64. De Giglio, E.; Sabbatini,
L.; Colucci, S.; Zambonin, G. Synthesis, analytical
characterization, and osteoblast adhesion properties on
RGD-grafted polypyrrole coatings on titanium substrates.
Biomater. Sci. Polym. Ed. 2000, 11 (10), 1073–1083. 65.
Cui, X.; Lee, V.A.; Raphael, Y.; Wiler, J.A.; Hetke, J.F.;
Anderson, D.J.; Martin, D.C. Surface modification of neural
recording electrodes with conducting polymer/biomolecule
blends. Biomed. Mater. Res. 2001, 56, 261–272. 66. Rivers,
T.J.; Hudson, T.W.; Schmidt, C.E. Synthesis of a novel,
biodegradable electrically conducting polymer for
biomedical applications. Adv. Funct. Mater. 2002 , 12 (1),
33–37. 67. Zhang, Z.; Roy, R.; Dugre´, F.J.; Tessier, D.;
Dao, L.H. In vitro biocompatibility study of electrically
conductive polypyrrole-coated polyester fabrics. Biomed.
Mater. Res. 2001, 57 (1), 63–71. 68. Boeckl, M.S.; Baas,
T.; Fujita, A.; Hwang, K.-O.; Bramblett, A.L.; Ratner,
B.D.; Rogers, J.W.; Sasaki, T. Template-assisted
nano-patterning of solid surfaces. Biopoly 1998, 47,
185–193. 69. Innis, P.C.; Wallace, G.G. Inherently
conducting polymer nanostructures. J. Nanoscie.
Nanotechnol. in press.
Confocal Microscopy

1. Minsky, M. Memoir on inventing the confocal microscope.


Scanning 1988, 10, 128–138.

2. Wilson, T.; Carlini, A.R. Three dimensional imaging in


confocal imaging systems with finite-sized detectors. J.
Microsc. 1988, 141, 51–66.

3. Sheppard, C.J.R.; Shotton, D.M. Image formation in the


confocal laser scanning microscope. In Confocal Laser
Scanning Microscopy; Springer-Verlag, New York Inc.: New
York, 1997; 15–31.

4. Guilbault, G. General aspects of luminescence


spectroscopy. In Practical Fluorescence, 2nd Ed.;
Guilbault, G., Ed.; Marcel Dekker, Inc.: New York, 1990;
1–40.

5. Atkins, P. Spectroscopy 2: electronic transitions. In


Physical Chemistry, 5th Ed.; W. H. Freeman and Company: New
York, 1994; 590–622.

6. Tsien, R.Y.; Waggoner, A. Fluorophores for confocal


microscopy. In Handbook of Biological Confocal Microscopy,
2nd Ed.; Pawley, J.B., Ed.; Plenum Press: New York, 1995;
267–280.

7. Sheppard, C.J.R.; Shotton, D.M. Introduction. In


Confocal Laser Scanning Microscopy; Springer-Verlag New
York Inc.: New York, 1997; 1–13.

8. Porter, T.; Duff, T. Compositing digital images. Comput.


Graphics 1984, 18 (3), 253–259.

9. Inoue´, S.; Spring, K.R. Microscope image formation. In


Video Microscopy the Fundamentals; Plenum Press: New York,
1997; 13–118.

10. Keller, H.E. Objective lenses for confocal microscopy.


In Handbook of Biological Confocal Microscopy, 2nd Ed.;
Pawley, J.B., Ed.; Plenum Press: New York, 1995; 111–126.

11. Wilson, T. The role of the pinhole in confocal imaging


system. In Handbook of Biological Confocal Microscopy, 2nd
Ed.; Pawley, J.B., Ed.; Plenum Press: New York, 1995;
167–182.

12. Pawley, J. Fundamental limits in confocal microscopy.


In Handbook of Biological Confocal Microscopy, 2nd Ed.;
Pawley, J.B., Ed.; Plenum Press: New York, 1995; 19–38.

13. Becker, P.L. Quantitative fluorescence measurements. In


Fluorescence Imaging Spectroscopy and Microscopy; Wang,
X.F., Herman, B., Eds.; John Wiley & Sons, Inc.: New York,
1996; 1–29.

14. Sheppard, C.J.R.; Shotton, D.M. Confocal fluorescence


microscopy. In Confocal Laser Scanning Microscopy;
Springer-Verlag New York Inc.: New York, 1997; 61–70.

15. Chen, H.; Swedlow, J.R.; Grote, M.; Sedat, J.W.; Agard,
D.A. The collection, processing, and display of digital
three-dimensional images of biological specimens. In
Handbook of Biological Confocal Microscopy, 2nd Ed.;
Pawley, J.B., Ed.; Plenum Press: New York, 1995; 197–210.
16. Song, L.; Varma, C.A.; Verhoeven, J.W.; Tanke, H.J.
Influence of the triplet excited state on the photobleaching
kinetics of fluorescein in microscopy. Biophys. J. 1996, 70,
2959–2968. 17. Lemasters, J.J. Confocal microscopy of
single living cells. In Fluorescence Imaging Spectroscopy
and Microscopy; Wang, X.F., Herman, B., Eds.; John Wiley &
Sons, Inc.: New York, 1996; 157–177. 18. Axelrod, D.;
Koppel, D.E.; Schlessinger, J.; Webb, W.W. Mobility
measurement by analysis of fluorescence photobleaching
recovery kinetics. Biophys. J. 1976 , 16, 1055–1069. 19.
Teraski, M.; Dailey, M.E. Confocal microscopy of living
cells. In Handbook of Biological Confocal Microscopy, 2nd
Ed.; Pawley, J.B., Ed.; Plenum Press: New York, 1995;
327–346. 20. Stelzer, E.H. The intermediate optical system
of laser-scanning confocal microscopes. In Handbook of
Biological Confocal Microscopy, 2nd Ed.; Pawley, J.B., Ed.;
Plenum Press: New York, 1995; 139–154. 21. Tsien, R.Y.;
Bacskai, B.J. Video-rate confocal microscopy. In Handbook
of Biological Confocal Microscopy, 2nd Ed.; Pawley, J.B.,
Ed.; Plenum Press: New York, 1995; 459–478. 22. Iyer, V.;
Losavio, B.E.; Saggau, P. Compensation of spatial and
temporal dispersion for acousto-optic multiphoton
laser-scanning microscopy. J. Biom. Opt. 2003, 8 (3),
460–471. 23. Xiao, G.Q.; Corle, T.R.; Kino, G.S. Real-time
confocal scanning optical microscope. Appl. Phys. Lett.
1988, 53 (8), 716–718. 24. Kino, G.S. Intermediate optics
in Nipkow disk microscopes. InHandbook of Biological
Confocal Microscopy, 2nd Ed.; Pawley, J.B., Ed.; Plenum
Press: New York, 1995; 155–165. 25. Diaspro, A.; Sheppard,
C. Two-photon microscopy: basic principles and
architectures. In Confocal and Two-Photon Microscopy
Foundations, Applications and Advances; Diaspro, A., Ed.;
Wiley-Liss, Inc.: New York, 2002; 39–73. 26. Denk, W.;
Svoboda, K. Photon upmanship: why multiphoton imaging is
more than a gimmick. Neuron 1997, 18, 351–357. 27. Denk,
W.; Piston, D.W.; Webb, W.W. Two-photon molecular
excitation in laser-scanning microscopy. In Handbook of
Biological Confocal Microscopy, 2nd Ed.; Pawley, J.B., Ed.;
Plenum Press: New York, 1995; 445–458.
Contact Lenses: Silicone Hydrogels

13. Schein, O.; Glynn, R.; Poggio, E.; Seddon, J.; Kenyon,
K. The relative risk of ulcerative keratitis among users of
daily-war and extended-wear soft contact lenses. N. Engl.
J. Med. 1989, 321, 773–778.

14. F.D.A. Practitioner Letter, Ulcerative keratitis and


contact lens wear, May 30, 1989.

15. Schein, O.; Buehler, P.; Stamler, J.; Verdier, D.;


Katz, J. The impact of overnight wear on the risk of
contact lens associated ulcerative keratitis. Arch.
Ophthalmol. 1994, 112, 186–190.

16. Hamano, H.; Watanabe, K.; Hamano, T.; Mitsunaga, S.;


Katani, S.; Okada, A. A study of complications induced by
conventional and disposable contact lenses. CLAO J. 1994,
20, 103–108.

17. Boswall, G.; Ehlers, W.; Luistro, A.; Worrall, M.;


Donshik, P. A comparison of conventional and disposable
extended wear contact lenses. CLAO J. 1993, 19, 158–165.

18. U.S.P. Dictionary of USAN and International Drug Names;


U.S. Pharmacopeia: Rockville, MD, 2000; 259.

19. Professional Edition; Tyler’s Quarterly Soft Contact


Lens Parameter Guide; Tyler’s Quarterly, Inc.; Little Rock,
2000; Vol. 18, 29.

20. Holden, B.; Mertz, G. Critical oxygen levels to avoid


corneal edema for daily and extended wear contact lenses.
Invest. Ophthalmol. Vis. Sci. 1984, 25, 1161–1167.

21. Lai, Y. The role of bulky polysiloxanyl methacrylates


in oxygen-permeable hydrogel materials. J. Appl. Polym.
Sci. 1996, 56, 317–324.

22. Robertson, R.; et al. US Patent 5,070,169, 1991.

23. Kunzler, J.; Ozark, R. US Patent 5,321,108, 1994.

24. Vanderlaan, D.; et al. PCT Application WO 01/27174,


2001.

25. Chang, H. US Patent 4,182,822, 1980.

26. Mueller, K.; Kleiner, E. US Patent 4,136,250, 1979.


27. Tanaka, K.; Takahashi, K.; Kanada, M.; Kanome, S.;
Nakajima, T. US Patent 4,139,513, 1979.

28. Deichert, W.; Su, K.; van Buren, M. US Patent


4,153,641, 1979.

29. Keogh, P.; Ku¨nzler, J.; Niu, G. US Patent 4,259,467,


1981.

30. Rice, D.; Ihlenfeld, J. US Patent 4,440,918, 1984.

31. Rice, D.; Ihlenfeld, J. US Patent 4,818,801, 1989.

32. Isaacson, W. President of 3M Vision Care, Minneapolis,


MN, private communication.

33. Salame, M.; Steingeiser, S. Barrier polymers. Polym.


Plast. Technol. Eng. 1977, 8, 155–175.

34. Molock, F. US Patent 5,039,769, 1991.

35. Su, K.; Robertson, R. US Patent 4,740,533, 1988.

36. Alli, A.; et al. PCT Application WO 01/30867, 2001.

37. Vanderlaan, D.; et al. PCT Application WO 01/27174,


2001.

38. McCabe, K.; et al. US Patent Application 2004/6822016,


2004.

39. Mueller, K.; Heiber, S.; Plankl, W. US Patent


4,486,577, 1984.

40. Lai, Y; Baccei, L. US Patent 5,034,461, 1991.

41. Goldenberg, M. US Patent 4,929,692, 1990.

42. Hirt, T. US Patent 6,039,913, 2000.

43. Keogh, P.; Ku¨nzler, J.; Niu, G. US Patent 4,260,725,


1981. 44. LeBoeuf, A. US Patent 4,261,875, 1981. 45.
LeBoeuf, A. US Patent 4,294,974, 1981. 46. Ku¨nzler, J.;
Ozark, R. Hydrogels based on hydrophilic side-chain
siloxanes. J. Appl. Polym. Sci. 1995, 55, 611–619. 47. CIBA
Vision; unpublished results. 48. Bambury, R.; Seelye, D. US
Patent 5,070,215, 1991. 49. Lai, Y. US Patent 5,310,779,
1994. 50. Ichihara, M. US Patent Application 2004/0014898,
2004. 51. Nicolson, P.; Baron, R.; Chabrecek, P.; et al. US
Patent 5,760,100, 1998. 52. Gido, S. Connectivity of
domains in microphase separated polymer materials. In New
Polymer Materials; Kourgic-Karasz L.S., MacKnight, W.J.,
Mortuscelli, E., Eds.; A.C.S. Symposium Series; Oxford
University Press: Oxford, London, 2005; 309–325. 53.
Nicolson, P.; et al. US Patent 5,776,999, 1998. 54.
Nicolson, P.; et al. US Patent 5,789,461, 1998. 55.
Nicolson, P.; et al. US Patent 5,848,811, 1998. 56.
Nicolson, P.; et al. US Patent 5,965,631, 1999. 57.
Domschke, A.; Lohmann, D.; Winterton, L. On-eye mobility of
soft oxygen permeable contact lenses. Proceedings of the
ACS Spring National Meeting, San Francisco, PMSE; 1997. 58.
Tighe, B. Silicone hydrogel materials—how do they work?. In
Silicone Hydrogels: the Rebirth of Continuous Wear Contact
Lenses; Sweeney, D., Ed.; Butterworth, Heinemann: Oxford,
2000; 1–21. 59. Takayanagi, M.; Harima, H.; Iwata, Y. J.
Soc. Mater. Sci. Jpn. 1963, 12, 389. 60. Marcussen, L. In
Thermal Conductivity; Ashworth, T., Smith, D., Eds.;
Plenum: New York, 1985; 585. 61. Nicolson, P. Continuous
wear contact lens surface chemistry and wearability. Eye
Contact Lens 2003, 29, S30–S32. 62. Lai, Y.; et al. US PCT
Application WO 92/09644, 1992. 63. Lai, Y. PCT Application
WO 92/09421, 1992. 64. Vanderlaan, D.; et al. US Patent US
6,087,415, 2000. 65. McGee, J.; et al. US Patent 6,599,559,
2003. 66. Winterton, L.; et al. US Patent 6451871, 2002.
67. Turner, D.; et al. US Application 2003/005242, 2003.
68. Maiden, A.; et al. US Patent 6,367,929, 2002. 69.
Jones, L.; Tighe, B. Silicone hydrogel contact lens
materials update—Part 1, On the Silicone Hydrogels web site
at www.siliconehydrogels.org/editorials/index.asp, July 22,
2004. 70. Jones, L.; Dumbleto, K. Silicone hydrogel contact
lenses, Part 1: Evolution and current status, Optometry
Today, 2002, September 20, 26–31. 71. American Medical
Association (USAN) USAN Assigned to Hydrophilic Contact
Lens Materials, available at
http://www.ama-assn.org/ama1/pub/upload/mm/ 365/galyfilcon_
a.doc, 2005. 72. American Medical Association (USAN) USAN
Assigned to Hydrophilic Contact lens Materials, available
at http://www.ama-assn.org/ama1/pub/upload/mm/365/
senofilcon_ a.doc, 2005. 73. Jones, L.; Tighe, B. Silicone
hydrogel contact lens materials update—Part 2, On the
Silicone hydrogels, Web site available at
www.siliconehydrogels.org/editorials/ index.asp, 2005.
Control of Movement

1. Popovic´, D.B.; Sinkjær, T. Control of Movement for the


Physically Disabled; Springer: London, 2000.

2. Kandel, E.R., Schwartz, J.H., Jessel, T.M., Eds.;


Principles of Neural Science, 4th Ed.; Appleton and Lange:
Norwalk Connecticut, 1991.

3. Biomechanics and Neural Control of Posture and Movement;


Winters, J.M., Crago, P.E., Eds.; SpringerVerlag: New York,
2000.

4. Georgopoulos, A.P. On reaching. Annu. Rev. Neurosci.


1986, 9, 147–170.

5. Bernstein, N.A. The Coordination and Regulation of


Movements; Pergamon Press: London, 1967 (Original work in
1926–1935.).

6. Soechting, J.F. Elements of coordinated arm movements in


three-dimensional space. In Perspectives on the
Coordination of Movement, Wallace, S.A., Ed.; Amsterdam:
North-Holland, 1989; 47–83.

7. Hogan, N. An organization principle for a class of


voluntary movements. J. Neurosci. 1984, 4, 2745–2754. 8.
Crago, P.E. Creating neuromuscular models. In Biomechanics
and Neural Control of Posture and Movement; Winters, J.M.,
Crago, P.E., Eds.; Springer: London, 2000; 119–133. 9.
Kearney, R.E.; Kirsch, R.F. System identification and
neuromuscular modeling. In Biomechanics and Neural Control
of Posture and Movement; Springer Verlag: New York, 2000;
134–147. 10. Brown, I.E.; Loeb, G.E. A reductionist
approach to creating and using neuromuscular models. In
Biomechanics and Neural Control of Posture and Movement;
Winters, J.M., Crago, P.E., Eds.; Springer: London, 2000;
148–163. 11. Zajac, F.E. Muscle and tendon: properties,
models, scaling, and application to biomechanics and motor
control. Crit. Rev. Biomed. Eng. 1989, 17 (4), 359–411. 12.
Hill, T.L. The heat of shortening and the dynamic constants
of muscle. Proc. R. Soc. Lond. Biol. 1938, 126, 135–195.
13. Winters, J.M. Hill-based muscle models: a systems
engineering prospective. In Multiple Muscle Systems:
Biomechanics and Movement Organization; Winters, J.M., Woo,
S.L.-Y., Eds.; Springer Verlag: New York, 1990; 66–93. 14.
Popovic´, D.B. Finite state model of locomotion for
functional electrical stimulation systems. Prog. Brain Res.
1993, 97, 397–407. 15. Popovic´, D.B.; Stein, R.B.;
Og˘uzto¨reli, M.N.; Lebiedowska, M.; Jonic´, S. Optimal
control of walking with functional electrical stimulation:
a computer simulation study. IEEE Trans. Rehabil. Eng.
1999, TRE-7 (1), 69–79. 16. Tomovic´, R.; Popovic´, D.B.;
Stein, R.B. Nonanalytical Methods for Motor Control; World
Science: Singapore, 1995. 17. Antsaklis, P.J.; Nerode, A.
Hybrid control systems: an introductory discussion to the
special issue. IEEE Autom. Control 1988, AU-43, 457–459.
18. Lima, P.U.; Saridis, G.N. Intelligent controllers as
hierarchical stochastic automata. IEEE Trans. Syst. Man.
Cybern. 1999, SMC (B)-29 (2), 151–163. 19. Antsaklis, P.;
Kohn, W.; Nerode, A.; Sastry, S. Eds.; Hybrid Systems II,
Lecture Notes in Computer Science, No. 999 Ed.; Springer
Verlag: New York, 1995. 20. Branicki, M.S. Multiple
Liapunov functions and other analysis tool for switched and
hybrid systems. IEEE Trans. Autom. Control. 1998, AU-43,
475–482. 21. Popovic´, M.B. Control of neural prostheses
for grasping and reaching. Med. Eng. Phys. 2003, 25 (1),
41–50. 22. Popovic´, D.B.; Popovic´, M.B. Tuning of a
nonanalytic hierarchical control system for reaching with
FES. IEEE Trans. Biomed. Eng. 1998, BME-45, 203–212.
Controlled Release

11. Dash, A.K.; Cudworth, G.C. Therapeutic applications of


implantable drug delivery systems. J. Pharmacol. Toxicol.
Methods 1998, 40 (1), 1–12.

12. Selam, J.L.; Charles, M.A. Devices for insulin


administration. Diabetes Care 1990, 13 (9), 955–979.

13. Skitzki, J.; Chang, A. Hepatic artery chemotherapy for


colorectal liver metastases: Technical considerations and
review of clinical trials. Surg. Oncol. 2002, 11 (3),
123–135.

14. Rise, M. Instrumentation for neuromodulation. Arch.


Med. Res. 2000, 31 (3), 237–247.

15. Schade, D.S.; Eaton, R.P.; Edwards, W.S.; Doberneck,


R.C.; Spencer, W.J.; Carlson, G.A.; Bair, R.E.; Love, J.T.;
Urenda, R.S.; Gaona, J.I. A remotely programmable insulin
delivery systemsuccessful short-term implantation in man.
JAMA J. Am. Med. Assoc. 1982, 247 (13), 1848–1853.

16. Theeuwes, F.; Yum, S. Principles of the design and


operation of generic osmotic pumps for the delivery of
semisolid or liquid drug formulations. Ann. Biomed. Eng.
1976, 4, 343–353.

17. Makhija, S.; Vavia, P. Controlled porosity osmotic


pump-based controlled release system of pseudoephedrineI.
Cellulose acetate as a semipermeable membrane. J. Control.
Release 2003, 89 (1), 5–18.

18. Wong, P.S.L.; Gupta, S.K.; Stewart, B.E. Osmotically


Controlled Tablets. In Modified-Release Drug Delivery
Technology; Rathbone, M.J., Hadgraft, J., Roberts, M.S.,
Eds.; Marcel Dekker, Inc.: New York, 2003; 101–114.

19. Uchiyama, T.; Watanabe, J.; Ishihara, K.


Pressureinduced change in permeation of insulin through a
polymer alloy membrane for an implantable insulin pump. J.
Membr. Sci. 2002, 210 (2), 423–431.

20. Langer, R. Controlled releasea new approach to drug


delivery. Technol. Rev. 1981, 83 (5), 26–34.

21. Brannon-Peppas, L. Polymers in controlled drug


delivery. Med. Plast. Biomater. Mag. 1997, November, 34

22. Ron, E.; Langer, R. Erodible Systems. In Treatise on


Controlled Drug Delivery; Kydonieus, A., Ed.; Marcel
Dekker, Inc.: New York, 1992; 199–224.

23. Fu, J.; Fiegel, J.; Krauland, E.; Hanes, J. New


polymeric carriers for controlled drug delivery following
inhalation or injection. Biomaterials 2002, 23 (22),
4425–4433.

24. Lupron Depot TAP Pharmaceuticals, www.lupron.com


Accessed Sept. 9, 2003.

25. Wang, L.; Kleiner, L.; Venkatraman, S. Structure


formation in injectable poly(lactide-co-glycolide) depots.
J. Control. Release 2003, 90 (3), 345–354. 26. Greenwald,
R.B.; Choe, Y.H.; McGuire, J.; Conover, C.D. Effective drug
delivery by PEGylated drug conjugates. Adv. Drug Deliv.
Rev. 2003, 55 (2), 217–250. 27. Sivin, I.; Diaz, S.; Holma,
P.; Alvarezsanchez, F.; Robertson, D.N.; Stern, J.
Long-term experience with norplant-Tm in international
clinic trials. Contracept. Deliv. Syst. 1982, 3 (3-4), 90.
28. Kim, S. Temperature Sensitive Polymers for Delivery of
Macromolecular Drugs. In Advanced Biomaterials in
Biomedical Engineering and Drug Delivery Systems; Ogata,
N., Kim, S., Feijen, J., Eds.; Springer: Tokyo, 1996;
126–133. 29. Korsmeyer, R.W.; Gurny, R.; Doelker, E.; Buri,
P.; Peppas, N.A. Mechanisms of solute release from porous
hydrophilic polymers. Int. J. Pharm. 1983, 15 (1), 25–35.
30. Saltzman, W.M. 2001. Drug Delivery: Engineering
Principles for Drug Therapy; Oxford University Press: New
York, 2001; 235–279. 31. Vallbacka, J.J.; Nobrega, J.N.;
Sefton, M.V. Tissue engineering as a platform for
controlled release of therapeutic agents: Implantation of
microencapsulated dopamine producing cells in the brains of
rats. J. Control. Release 2001, 72 (1–3), 93–100. 32.
Soon-Shiong, P. Treatment of type I diabetes using
encapsulated islets. Adv. Drug Deliv. Rev. 1999, 35 (2– 3),
259–270. 33. Vernon, B.; Kim, S.W.; Bae, Y.H. Insulin
release from islets of Langerhans entrapped in a
poly(N-isopropylacrylamide-co-acrylic acid) polymer gel. J.
Biomater. Sci. Polym. Ed. 1999, 10 (2), 183–198. 34. Code
of Federal Regulations. Title 21, 2003; Vol. 1. 21CFR3. 35.
Food and Drug Administration Intercenter agreement between
the center for drug evaluation and research and the center
for devices and radiological health. HFA305. Food Drug
Admin. 1991. 36. Medical Device User Fee and Modernization
Act of 2002 PL 107-250; Office of the Federal Register
(OFR), National Archives and Records Administration (NARA),
2002; 403. 37. Marroum, P.J. Regulatory Issues Relating to
ModifiedRelease Drug Formulations. In Modified-Release Drug
Delivery Technology; Rathbone, M.J., Hadgraft, J., Roberts,
M.S., Eds.; Marcel Dekker, Inc.: New York, 2003; 943–975.
Corneal Implants

1. Stulting, R.D.; Lahners, W.J.; Carr, J.D. Advances in


refractive surgery: 1975 to the present. Cornea 2000, 19
(5), 741–753.

2. Waring, G.O. Making sense of ‘keratospeak’: A


classification of refractive corneal surgery. Arch.
Ophthalmol. 1985, 103 (10), 1472–1477.

3. Chirila, T.V.; Hicks, C.R. The origins of the artificial


cornea: Pellier de Quengsy and his contribution to the
modern concept of keratoprosthesis. Gesnerus 1999, 56
(1–2), 96–106.

4. Belau, P.G.; Dyer, J.A.; Ogle, K.N.; Henderson, J.W.


Correction of ametropia with intracorneal lenses. Arch.
Ophthalmol. 1964, 72 (10), 541–547.

5. Brown, S.I.; Dohlman, C.H. A buried corneal implant


serving as a barrier to fluid. Arch. Ophthalmol. 1965, 73
(5), 635–639.

6. Mester, U.; Roth, K.; Dardenne, M.U. Trial with


2-hydroxy-ethyl-methacrylate lenses as keratophakia
material [Ger.]. Ber. Dtsch. Ophthalmol. Ges. 1974, 72,
326–329.

7. McCarey, B.E. Current status of refractive surgery with


synthetic intracorneal lenses. Refract. Corneal Surg. 1990,
6 (1), 40–46.

8. McCarey, B.E.; van Rij, G.; Beekhuis, W.H.; Waring, G.O.


Hydrogel keratophakia: A freehand pocket dissection in the
monkey model. Br. J. Ophthalmol. 1986, 70 (3), 187–191. 9.
Binder, P.S. Hydrogel implants for the correction of
myopia. Curr. Eye Res. 1983, 2 (7), 435–441. 10. Choyce,
D.P. The correction of refractive errors with polysulfone
corneal inlays: A new frontier to be explored? Trans.
Ophthalmol. Soc. U. K. 1985, 104 (3), 332–342. 11.
Kirkness, C.M.; Steele, A.D.M.; Garner, A. Polysulfone
corneal inlays. Adverse reactions: A preliminary report.
Trans. Ophthalmol. Soc. U. K. 1985, 104 (3), 343–350. 12.
Ismail, M.M. Correction of hyperopia with intracorneal
implants. J. Cataract Refract. Surg. 2002, 28 (3), 527–
530. 13. Werblin, T.P. Lamellar refractive surgery: Where
have we been and where are we going? Refract. Corneal Surg.
1989, 5 (3), 167–176. 14. Thompson, K.P.; Hanna, K.;
Waring, G.O.; Gipson, I.; Liu, Y.; Gailitis, R.P.;
Johnson-Wint, B.; Green, K. Current status of synthetic
epikeratoplasty. Refract. Corneal Surg. 1991, 7 (3),
240–248. 15. Thompson, K.P.; Hanna, K.D.; Waring, G.O.
Emerging technologies for refractive surgery: Laser
adjustable synthetic epikeratoplasty. Refract. Corneal
Surg. 1989, 5 (1), 46–48. 16. Xie, R.Z.; Stretton, S.;
Sweeney, D.F. Artificial cornea: Towards a synthetic onlay
for correction of refractive error. Biosci. Rep. 2001, 21
(4), 513–536. 17. Evans, M.D.M.; McLean, K.M.; Hughes,
T.C.; Sweeney, D.F. A review of the development of a
synthetic corneal onlay for refractive correction.
Biomaterials 2001, 22 (24), 3319–3328. 18. Burris, T.E.;
Ayer, C.T.; Evensen, D.A.; Davenport, J.M. Effects of
intrastromal corneal ring size and thickness on corneal
flattening in human eyes. Refract. Corneal Surg. 1991, 7
(1), 46–50. 19. Hoh, H.; Rehfeldt, K.; Reiss, G. Correction
of myopia with the intracorneal ring [Ger.]. Ophtalmologe
1999, 96 (11), 717–723. 20. Simon, G.; Parel, J.-M.; Lee,
W.; Kervick, G.N. Gel injection adjustable keratoplasty.
Graefe’s Arch. Clin. Exp. Ophthalmol. 1991, 229, 418–424.
21. Legeais, J.-M.; Parel, J.-M.; Savoldelli, M.; Villain,
F.L.; D’Hermies, F.; Renard, G.; Pouliquen, Y. Reticulated
polyethylene oxide for gel injection adjustable
keratoplasty. Biocompatibility in critical situation [Fr.].
J. Franc. Ophtalmol. 1997, 20 (1), 31–36. 22. Day, R.
Artificial cornea implants. Trans. Am. Ophthalmol. Soc.
1957, 55, 455–475. 23. Hicks, C.R.; Fitton, J.H.; Chirila,
T.V.; Crawford, G.J.; Constable, I.J. Keratoprosthesis:
Advancing towards a true artificial cornea. Surv.
Ophthalmol. 1997, 42 (2), 175–189. 24. Chirila, T.V.;
Hicks, C.R.; Dalton, P.D.; Vijayasekaran, S.; Lou, X.;
Hong, Y.; Clayton, A.B.; Ziegelaar, B.W.; Fitton, J.H.;
Platten, S.; Crawford, G.J.; Constable, I.J. Artificial
cornea. Prog. Polym. Sci. 1998, 23 (3), 447–473. 25.
Mannis, M.J.; Dohlman, C.H. The Artificial Cornea: A Brief
History. In Corneal Transplantation: A History in Profiles;
Mannis, M.J., Mannis, A.A., Eds.; Hirschberg History of
Ophthalmology—The Monographs, J.P. Wayenborgh: Oostende,
Belgium, 1999; Vol. 6, 321–335. 26. Cardona, H.
Keratoprosthesis: Acrylic optical cylinder with supporting
intralamellar plate. Am. J. Ophthalmol. 1962, 54 (2),
284–294. 27. Dohlman, C.H.; Doane, M.G. Some factors
influencing outcome after keratoprosthesis surgery. Cornea
1994, 13 (3), 214–218. 28. Chirila, T.V. Modern artificial
corneas: The use of porous polymers. Trends Polym. Sci.
1994, 2 (9), 296–300. 29. Kain, H.L. A new concept for
keratoprosthesis [Ger.]. Klin. Mon.bl. Augenheilkd. 1990,
197, 386–392. 30. Trinkaus-Randall, V.; Wu, X.Y.; Tablante,
R.; Tsuk, A. Implantation of a synthetic cornea: Design,
development and biological response. Artif. Organs 1997, 21
(11), 1185–1191. 31. Legeais, J.-M.; Renard, G. A second
generation of artificial cornea (Biokpro II). Biomaterials
1998, 19 (16), 1517–1522. 32. Caldwell, D.R. The soft
keratoprosthesis. Trans. Am. Ophthalmol. Soc. 1997, 95,
751–802. 33. Chirila, T.V.; Vijayasekaran, S.; Horne, R.;
Chen, Y.-C.; Dalton, P.D.; Constable, I.J.; Crawford, G.J.
Interpenetrating polymer network (IPN) as a permanent joint
between the elements of a new type of artificial cornea. J.
Biomed. Mater. Res. 1994, 28 (6), 745–753. 34. Chirila,
T.V. An overview of the development of artificial corneas
with porous skirts and the use of PHEMA for such an
application. Biomaterials 2001, 22 (24), 3311–3317. 35.
Crawford, G.J.; Hicks, C.R.; Lou, X.; Vijayasekaran, S.;
Tan, D.; Mulholland, B.; Chirila, T.V.; Constable, I.J. The
Chirila keratoprosthesis: Phase I human clinical trials.
Ophthalmology 2002, 109 (5), 883–889. 36. Hicks, C.R.;
Crawford, G.J.; Lou, X.; Tan, D.T.; Snibson, G.R.; Sutton,
G.; Downie, N.; Werner, L.; Chirila, T.V.; Constable, I.J.
Corneal replacement using a synthetic hydrogel cornea,
AlphaCort: Device, preliminary outcomes and complications.
Eye 2003, 17 (3), 385–392. 37. Kim, M.K.; Lee, J.L.; Wee,
W.R.; Lee, J.H. Seoul-type keratoprosthesis: Preliminary
results of the first 7 human cases. Arch. Ophthalmol. 2002,
120 (6), 761–766. C
Corrosion

1. Williams, D.F.; Williams, R.L. Degradative Effects of


the Biological Environment on Metals and Ceramics. In
Biomaterials Science: An Introduction to Materials in
Medicine; Ratner, B., Hoffman, A., Lemons, J., Schoen, F.,
Eds.; Academic Press: San Diego, 1996; 260–267.

2. Bundy, K.J. Corrosion and other electrochemical aspects


of biomaterials. Crit. Rev. Biomed. Eng. 1994, 22 (3/4),
139–251.

3. Jacobs, J.J.; Gilbert, J.L.; Urban, R.M. Current


concepts review corrosion of metal orthopaedic implants. J.
Bone Jt. Surg. 1998, 80-A, 268–282.

4. Cook, S.D.; Tomas, K.A.; Harding, A.F.; Collins, C.L.;


Haddad, R.J.; Milicic, M.; Fischer, W.L. The in vivo
performance of 250 internal fixation devices; a follow up
study. Biomaterials 1987, 8, 177–184.

5. Seah, K.H.W.; Thampuran, R.; Teoh, S.H. The influence of


pore morphology on corrosion. Corr. Sci. 1998, 40, 547–556.

6. Khan, M.A.; Williams, R.L.; Williams, D.F. Conjoint


corrosion and wear in titanium alloys. Biomaterials 1999,
20 (8), 765–772.

7. Okazaki, Y. Effect of friction on anodic polarization


properties of metallic biomaterials. Biomaterials 2002, 23,
2071–2077. 8. Gilbert, J.L.; Buckley, C.A.; Jacobs, J.J. In
vivo corrosion of modular hip prosthesis components in
mixed and similar metal combinations. The effect of
crevice, stress, motion and alloy coupling. J. Biomed.
Mater. Res. 1993, 27, 1533–1544. 9. Goldberg, J.R.;
Gilbert, J.L. In vitro corrosion testing of modular hip
tapers. J. Biomed. Mater. Res. (Appl. Biomater.) 2003, 64B,
73–78. 10. Fonseca, C.; Barbosa, M.A. Corrosion behaviour
of titanium in biofluids containing H 2 O 2 studied by
electrochemical impedance spectroscopy. Corr. Sci. 2001,
43, 547–559. 11. Shih, C.-C.; Lin, S.-J.; Chung, K.-H.;
Chen, Y.-L.; Su, Y.-Y. Increased corrosion resistance of
stent materials by converting current surface film of
polycrystalline oxide into amorphous oxide. J. Biomed.
Mater. Res. 2000, 52, 323–332. 12. Laurent, F.; Grosgogeat,
B.; Reclaru, L.; Dalard, F.; Lissac, M. Comparison of
corrosion behaviour in presence of oral bacteria.
Biomaterials 2001, 22, 2273–2282. 13. Cai, Z.; Shafer, T.;
Watanabe, I.; Nunn, M.E.; Okabe, T. Electrochemical
characterization of cast titanium alloys. Biomaterials
2003, 24, 213–218. 14. Lo´pez, M.F.; Gutie´rrez, A.;
Jime´nez, J.A. In vitro corrosion behaviour of titanium
alloys without vanadium. Electrochim. Acta 2002, 47,
1359–1364. 15. Lo´pez, M.F.; Jime´nez, J.A.; Gutie´rrez, A.
Corrosion study of surface-modified vanadium-free titanium
alloys. Electrochim. Acta 2003, 48, 1395–1401. 16.
Trepanier, C.; Tabrizian, M.; Yahia, L’H.; Bilodeau, L.;
Piron, D. Effect of modification of oxide layer on NiTi
stent corrosion resistance. J. Biomed. Mater. Res. 1998,
43, 433–440. 17. Pan, J.; Thierry, D.; Leygraf, C.
Electrochemical impedance spectroscopy study of the passive
oxide film on titanium for implant applications.
Electrochim. Acta 1996, 41, 1143–1153. 18. Garcı´a-Alonso,
M.C.; Saldan˜a, L.; Valle´s, G.; Gonza´lez-Carrasco, J.L.;
Gonza´lez-Cabrero, J.; Martı´nez, M.E.; Gil-Garay, E.;
Munuera, L. In vitro corrosion behaviour and osteoblast
response of thermally oxidies Ti6Al4V alloy. Biomaterials
2003, 24, 19–26. 19. Hiromoto, S.; Noda, K.; Hanawa, T.
Development of electrolytic cell with cell-culture for
metallic biomaterials. Corr. Sci. 2002, 44, 955–965. 20.
Hanawa, T. Evaluation techniques of metallic biomaterials
in vitro. Sci. Technol. Adv. Mater. 2002, 3, 289–295. 21.
Khan, M.A.; Williams, R.L.; Williams, D.F. The corrosion
behaviour of Ti-6Al-4V, Ti-6Al-7Nb and Ti13Nb-13Zr in
protein solutions. Biomaterials 1999 , 20 (7), 631–637. 22.
Williams, D.F. Physiological and microbiological corrosion.
Crit. Rev. Biocompat. 1985, 1 (1), 1–24. 23. Hanawa, T. In
vivo metallic biomaterials and surface modification. Mater.
Sci. Eng. 1999, A267, 260–266. 24. Steinemann, S.G. Metal
implants and surface reactions. Injury 1996, 27 (S-3),
16–22. 25. Bu¨dinger, L.; Hertl, M. Immunological
mechanisms in hypersensitivity reactions to metal ions: An
overview. Allergy 2000, 55, 108–115. 26. Corrosion and
Degradation of Implant Materials; Fraker, A.C., Griffith,
C.D., Eds.; ASTM S.T.P. No. 859, American Society for
Testing and Materials: Philadelphia, 1985. 27. Long, M.;
Rack, H.J. Titanium alloys in total joint replacement—A
materials science perspective. Biomaterials 1998, 19,
1621–1639. 28. Sittig, C.; Textor, M.; Spencer, N.D.;
Wieland, M.; Vallotton, P.-H. Surface characterization of
implant materials c.p.Ti, Ti-6Al-7Nb and Ti-6Al-4V with
different pretreatments. J. Mater. Sci., Mater. Med. 1999,
10, 5–46. 29. Aronsson, B.-O.; Lausmaa, J.; Kasemo, B. Glow
discharge plasma treatment for surface cleaning and
modification of metallic biomaterials. J. Biomed. Mater.
Res. 1997, 35, 49–73. 30. Thierry, B.; Tabrizian, M.;
Savadogo, O.; Yahia, L’H. Effects of sterilization
processes on NiTi alloy: Surface characterisation. J.
Biomed. Mater. Res. 2000, 49, 88–98. 31. Rondelli, G.;
Vicentini, B. Localized corrosion behaviour in simulated
human body fluids of commercial Ni-Ti orthodontic wires.
Biomaterials 1999, 20, 785–792. 32. Disegi, J.A.; Eschbach,
L. Stainless steel in bone surgery. Injury 2000, 31 (Suppl.
4), 2–6. 33. Walczak, J.; Shahgaldi, F.; Heatley, F. In
vivo corrosion of 316L stainless-steel hip implants:
Morphology and elemental compositions of corrosion
products. Biomaterials 1998, 19, 229–237. 34. Textor, M.;
Sittig, C.; Frauchiger, V.; Tosatti, S.; Brunette, D.M.
Properties and Biological Significance of Natural Oxide
Films on Titanium and Its Alloys. In Titanium in Medicine;
Brunette, D.M., Tengvall, P., Textor, M., Thomsen, P.,
Eds.; Springer: Berlin, 2001; 171–230. 35. Tengvall, P.;
Lundstro¨m, I.; Sjo¨kvist, L.; Elwing, H.; Bjursten, L.
Titanium-hydrogen peroxide interactions: Model studies of
the influence of the inflammatory response on titanium
implants. Biomaterials 1989, 10, 166–175. 36. Hanawa, T.;
Asami, K.; Asaoka, K. Repassvation of titanium and surface
oxide film regeneration in simulated bioliquid. J. Biomed.
Mater. Res. 1998, 40, 530–538. 37. Ong, J.L.; Lucas, L.C.
Auger electron spectroscopy and its use for the
characterization of titanium and hydroxyapatite surfaces.
Biomaterials 1998, 19, 455–464. 38. Esposito, M.; Lausmaa,
J.; Hirsch, J.-M.; Thomsen, P. Surface analysis of failed
oral titanium implants. J. Biomed. Mater. Res. (Appl.
Biomater.) 1999, 48, 559–568. 39. Ryha¨nen, J.; Niemi, E.;
Serlo, W.; Niemela¨, E.; Sandvik, P.; Pernu, H.; Salo, T.
Biocompatibility of nickeltitanium shape memory metal and
its corrosion behaviour in human cell cultures. J. Biomed.
Mater. Res. 1997, 35, 451–457. 40. Thierry, B.; Tabrizian,
M.; Trepanier, C.; Savadogo, O.; Yahia, L’H. Effect of
surface treatment and sterilization processes on the
corrosion behaviour of NiTi shape memory alloy. J. Biomed.
Mater. Res. 2000, 51, 685–693. 41. Ryha¨nen, J.;
Kallioninen, M.; Serlo, W.; Pera¨ma¨ki, P.; Jumila, J.;
Sandvik, P.; Niemela¨, E.; Tuukkanen, J. Bone healing and
mineralization, implant corrosion, and trace metals after
nickel-titanium shape memory metal intramedullary fixation.
J. Biomed. Mater. Res. 1999, 47, 472–480. C
Cryopreservation of Living Cells

10. Farrant, J. Mechanisms of Injury and Protection in


Living Cells and Tissues at Low Temperatures. In Current
Trends in Cryobiology; Smith, A.U., Ed.; Plenum: New York,
1970; 139–152.

11. Farrant, J. Water transport and cell survival in


cryobiological procedures. Philos. Trans. R. Soc. Lond., B
1977, 278, 291–306.

12. Fujikawa, S. Freeze-fracture and etching studies on


membrane damage on human erythrocytes caused by formation
of intracellular ice. Cryobiology 1980, 17, 351–362.

13. Mazur, P. Freezing and Low Temperature Storage of


Living Cells. In Proceedings of the Workshop on Basic
Aspects of Freeze Preservation of Mouse Strains; Muhlbock,
O., Ed.; , 1976; 1–12. Jackson Laboratory: Bar Harbor,
Gustav Fisher Verlag, Stuttgart. .

14. Mazur, P. The role of intracellular freezing in the


death of cells cooled at superoptimal rates. Cryobiology
1977, 14, 251–272.

15. Mazur, P. Freezing of living cells: Mechanisms and


implications. Am. J. Physiol. 1984, M7, C125–C142. (Cell
PhysioL.16).

16. Rapatz, G.; Nath, J.; Luyet, B. Electron microscope


study of erythrocytes in rapidly frozen mammalian blood.
Biodynamica 1963, 9, 83–94. 17. Steponkus, P.L.; Wiest,
S.C. Freeze-Thaw Induced Lesions in the Plasma Membrane. In
Low Temperature Stress in Crop Plants: The Role of the
Membrane; Lyons, M., Graham, D.G., Raison, J.K., Eds.;
Academic Press: New York, 1979; 231–253. 18. Stowell, R.E.;
Young, D.E.; Arnold, E.A.; Tromp, B.F. Structural, chemical
and functional alterations in mammalian nucleus following
different conditions of freezing, storage and thawing.
FASEB Fed. Proc. 1965, 24 (Suppl. 15), S115–S141. 19.
Trump, B.F.; Young, D.F.; Arnold, E.A.; Stowell, R.E.
Effects of freezing and thawing on the structure, chemical
constitution and function of cytoplasmic structures. FASEB
Fed. Proc. 1965, 24 (Suppl. 15), S144–S167. 20. Lovelock,
J.E. The denaturation of lipid-protein complexes as a cause
of damage by freezing. Proc. R. Soc. Lond., B 1957, 147,
427–434. 21. Levitt, J.A. Sulfhydryl disulphide hypotheses
of frost injury and resistance in plants. J. Theor. Biol.
1962, 3, 355. 22. Karow, A.M.; Gilbert, W.B., Jr.; Black,
J.B. Effects of temperature, potassium and sugar on human
spermatozoa motility: A cell preservation model from
reproductive medicine. Cryobiology 1992, 29, 250–254. 23.
Mazur, P.; Leibo, S.P.; Chu, E.H.Y. A two-factor hypothesis
of freezing injury. Exp. Cell Res. 1972, 71, 345–355. 24.
Meryman, H.T. The Exceeding of a Minimum Tolerable Cell
Volume in Hypertonic Suspension as a Cause of Freezing
Injury. In The Frozen Cell; Wolstenhoime O.E.W., O’Connor,
M., Eds.; Ciba Foundation Symposium, Churchill: London,
1970; 51–64. 25. Williams, R.J. The mechanisms of
cryoprotection in the intestinal mollusk Mylilus.
Cryobiology 1979, 4, 250. 26. Fishbein, W.M.; Winkert, J.W.
Parameters of biological freezing damage in simple
solutions: Catalase 11 demonstration of an optimum recovery
cooling rate curve in a membraneless system. Cryobiology
1978, 15, 168. 27. Pegg, D.E.; Diaper, M.P. The mechanism
of injury to slowly-frozen erythrocytes. Biophys. J. 1988,
54, 471– 488. 28. Mazur, P.; Cole, K.W. Influence of cell
concentration on the contribution of unfrozen fraction and
salt concentration to the survival of slowly frozen human
erythrocytes. Cryobiology 1985, 22, 509–536. 29. Levin,
R.L.; Cravalho, E.G.; Huffins, C.G. A membrane model
describing the effect of temperature on the water
conductivity of erythrocyte membranes at subzero
temperatures. Cryobiology 1976, 13, 415–429. 30. McGrath,
J.J. Preservation of Biological Material by Freezing and
Thawing. In Heat Transfer in Medicine and Biology; Shitzer,
A., Eberhart, R.C., Eds.; Plenum: New York, 1985. 31.
McGrath, J.J. Membrane Transport Properties. In Low
Temperature Biotechnology: Emerging Applications and
Engineering Contributions; Mcgrath, J.J., Diller, K.R.,
Eds.; A.S.M.E. 1988; 273–330. BED, HTDVol. 10; HTDVol. 98.
32. Gao, D.Y.; Liu, C.; Benson, C.; Liu, J.; Lin, S.;
Critser, E.S.; Critser, J.K. Theoretical and Experimental C
Analyses on the Optimal Experimental Design for
Determination of Hydraulic Conductivity of Cell Membrane.
In Advances in Heat and Mass Transfer in Biological System;
Hayes, L., Roemer, R.B., Eds.; ASME Press, HTD: New York,
1994; Vol. 288, 151–158.

33. Gao, D.Y.; Benson, C.T.; Liu, C.; McGrath, J.J.;


Critser, J.S.; Critser, J.K. Development of a novel
microperfusion chamber for determination of cell membrane
transport properties. Biophys. J. 1996, 71, 443–450.

34. Devireddy, R.V.; Raha, D.; Bischof, J.C. Measurement of


water transport during freezing in cell suspensions using a
differential scanning calorimeter. Cryobiology 1998, 36,
124–155.

35. Polge, C.; Lovelock, J.E. Preservation of bull sperm at


70C. Vet. Rec. 1952, 64, 296–297.

36. Woods, E.; Zieger, M.; Gao, D.Y.; Critser, J.K.


Equations for obtaining melting points for the ternary
system ethylene glycol/sodium chloride/water and their
application to cryopreservation. Cryobiology 1999, 38,
403–407.

37. Gao, D.Y.; Liu, J.; Liu, C.; McGann, L.E.; Watson,
P.F.; Kleinhans, F.W.; Mazur, P.; Critser, E.S.; Critser,
J.K. Prevention of osmotic injury to human spermatozoa
during addition and removal of glycerol. Hum. Reprod. 1995,
10, 1109–1122.

38. Mazur, P.; Schneider, U. Osmotic response of


preimplantation mouse and bovine embryos and their
cryobiological implications. Cell Biophys. 1986, 8,
259–285.

39. Schneider, U.; Mazur, P. Osmotic consequences of


cryoprotectant permeability and its relation to the
survival of frozen-thawed embryos. Theriogenology 1988, 21,
68–79.

40. Takahashi, T.; Inada, S.; Pommier, C.G.; O’Shea, J.J.;


Brown, E.J. Osmotic stress and the freeze-thaw cycle cause
shedding of Fc and C3b receptors by human polymorphonuclear
leukocytes. J. Immunol. 1985, 34, 4062.

41. Fahy, G.M.; MacFarlane, D.R.; Angell, C.A.; Meryman,


H.T. Vitrification as an approach to cryopreservation.
Cryobiology 1984, 21, 407–426.

42. Fahy, G.M. The relevance of cryoprotectant ‘‘toxicity’’


to cryobiology. Cryobiology 1986, 23 (1), 1–13.

43. Mahadevan, M. Cryobiological and Biochemical Studies of


Human Semen. In Ph.D. Thesis; Monash University: Melbourne,
Victoria, Australia, 1980.

44. Leibo, S.P.; Farrant, J.; Mazur, P.; Hanna, M.G.;


Smith, L.H. Effects of freezing on marrow stem cell
suspensions: Interactions of cooling and warming rates in
the presence of PVP, sucrose or glycerol. Cryobiology 1970,
6, 315–332.

45. Johnson, M.H.; Pickering, S.J. The effects of


dimethylsulfoxide on the microtubular system of the mouse
oocyte. Development 1987, 100, 313–324.
46. Joly, C.; Behini, O.; Boulekbache, H.; Testart, J.;
Maro, B. Effects of 1,2-propanediol on the cytoskeletal
organizaton of the mouse oocyte. Hum. Reprod. 1992 , 7,
374–378.

47. Watson, P.F. The Preservation of Semen in Mammals. In


Oxford Reviews of Reproductive Biology; Finn, C.A., Ed.;
Oxford Univ. Press: London, 1979; 283–350. 48. Sherman,
J.K. Improved methods of preservation of human spermatozoa
by freezing and freezing-drying. Fertil. Steril. 1964, 14,
49–64. 49. Critser, J.K.; Huse-Benda, A.R.; Aaker, D.V.;
Arneson, B.W.; Ball, G.D. Cryopreservation of human
spermatozoa. III. The effect of cryoprotectants on
motility. Fertil. Steril. 1998, 50, 314–320. 50. Berndtson,
W.E.; Foote, R.H. Bovine sperm cell volume at various
intervals after addition of glycerol of 5C. Cryobiology
1972, 9 (1), 29–33. 51. Berndtson, W.E.; Foote, R.H. The
freezabilily of spermatozoa after minimal prefreezing
exposure to glycerol or lactose. Cryobiology 1972, 9 (1),
57–60. 52. Sherman, J.K. Synopsis of the use of frozen
human semen since 1964: State of the art of human semen
banking. Fertil. Steril. 1973, 24, 397–416. 53. Karow,
A.M.; Gilbert, W.B., Jr.; Black, J.B. Effects of
temperature, potassium and sugar on human spermatozoa
motility: A cell preservation model from reproductive
medicine. Cryobiology 1992, 29, 250–254. 54. Critser, J.K.;
Arneson, B.W.; Aaker, D.V.; HuseBenda, A.R.; Ball, G.D.
Factors affecting the cryosurvival of mouse two-cell
embryos. J. Reprod. Fertil. 1988, 82 (1), 27–33. 55. Foote,
R.H. Semen quality from the bull to the freezer: An
assessment. Theriogenology 1975, 3, 219–235. 56. Graham,
E.G. Fundamentals of the Preservation of Spermatozoa. In
The Integrity of Frozen Spermatozoon; National Academy of
Sciences: Washington, DC, 1978; 4–44. 57. Watson, P.F.
Recent developments and concepts in the cryopreservation of
spermatozoa and the assessment of their post-thawing
function. Reprod. Fertil. Dev. 1995, 7, 871–891. 58.
Armitage, W.J.; Mazur, P. Osmotic tolerance of human
granulocytes. Am. J. Physiol. 1984, 247, C373– C381. 59.
McGann, L.E.; Yang, H.; Walterson, M. Manifestations of
cell damage after freezing and thawing. Cryobiology 1988,
25, 178–185. 60. Yudin, A.L.; Gottlieb, W.; Meizel, S.
Ultrastructural studies of the early events of the human
sperm acrosome reaction as initiated by human follicular
fluid. Gamete Res. 1988, 20, 11–24. 61. Mazur, P.; Cole,
K.W. Roles of unfrozen fraction, salt concentration, and
changes in cell volume in the survival of slowly frozen
human erythrocytes. Cryobiology 1989, 26, 1–29. 62.
Critser, J.K.; Huse-Benda, A.R.; Aaker, D.V.; Arneson,
B.W.; Ball, G.D. Cryopreservation of human spermatozoa. 1.
Effects of holding procedure and seeding on motility,
fertilizability, and acrosome reaction. Fertil. Steril.
1987, 47, 656–663. 63. Critser, J.K.; Arneson, B.W.; Aaker,
D.V.; Ball, G.D. Cryopreservation of human spermatozoa. II.
Post-thaw chronology of motility and of zona-free hamster
ova penetration. Fertil. Steril. 1987, 47, 980–984. 64.
Wheeler, M.B.; Seidel, G.E. Time course of in vitro
capacitation of frozen and unfrozen bovine spermatozoa.
Intl. Embryo Trans. Soc. 1986, 216. (ABSTRACT).

65. Pitt, R.E.; Steponkus, P.L. Quantitative analysis of


the probability of intracellular ice formation during
freezing of isolated protoplasts. Cryobiology 1989, 24,
44–63.

66. Rubinsky, B.; Pegg, D. A mathematical model for the


freezing process in biological tissue. Proc. R. Soc. 1988,
234, 343–358.

67. Bischof, J.; Hunt, C.J.; Rubinsky, B.; Burgess, A.;


Pegg, D.E. The effect of cooling rate and glycerol
concentration on the structure of the frozen kidney:
Assessment by cryo-scanning electron microscopy.
Cryobiology 1990, 27, 301–310. 68. Rubinsky, B.; Arav, A.;
DeVries, A.L. The cryoprotective effect of antifreeze
glycopeptides from antarctic fishes. Cryobiology 1992, 229,
69–72. 69. Baust, J.M.; Hollister, W.; Van Buskirk, R.;
Baust, J.G. Cryopreservation outcome is enhanced by
intracellulartype media and inhibition of apoptosis.
Cryobiology 1998, 37 (4), 410–411. C
Defibrillators

Moss, A.J.; Zareba, W.; Hall, W.J.; Klein, H.; Wilber,


D.J.; Cannom, D.S.; Daubert, J.P.; Higgins, S.L.; Brown,
M.W.; Andrews, M.L. The multicenter automatic defibrillator
implantation trial II investigators. Prophylactic
implantation of a defibrillator in patients with myocardial
infarction and reduced ejection fraction. N. Engl. J. Med.
2002, 346 (12), 877–883.
Degradable Polymer Composites

1. Ratner, B.D.; Hoffman, A.S.; Scheon, F.J.; Lemons, J.E.


Biomaterials Science: An Introduction to Materials in
Medicine; Academic Press: San Diego, 1996.

2. Whang, K.; Goldstick, T.K.; Healy, K. A biodegradable


polymer scaffold for delivery of osteotropic factors.
Biomaterials 2000, 21, 2545–2551.

3. Evans, G.; Brandt, K.; Niederbichler, A.; Chauvin, P.;


Hermann, S.; Bogle, M.; Otta, L.; Wang, B.; Patrick, C.W.,
Jr. Clinical long-term in vivo evaluation of poly(
L-lactide acid) porous conduits for peripheral nerve
regeneration. J. Biomater. Sci., Polym. Ed. 2000, 11 (8),
869–878.

4. Sarmiento, A.; Mullis, D.L.; Latta, L.L.; Tarr, R.R.;


Alvarez, R. A quantitative comparative analysis of fracture
healing under the influence of compression plating vs.
closed weight-bearing treatment. Clin. Orthop. 1980, 149,
232–239.

5. Claes, L. The mechanical and morphological properties of


bone beneath internal fixation plates of differing rigidity.
J. Orthop. Res. 1989, 7 (2), 170–177.

6. Beaupre´, G.S.; Csongradi, J.J. Refracture risk after


plate removal in the forearm. J. Orthop. Trauma 1996, 10
(2), 87–92.

7. Bo¨stman, O.M. Refracture after removal of a condylar


plate from the distal third of the femur. J. Bone Jt. Surg.
1990, 72 (7), 1013–1018.

8. McAuliffe, J.A. Forearm fixation. Hand Clin. 1997, 13


(4), 689–701.

9. Lovell, M.E.; Galasko, C.S.; Wright, N.B. Removal of


orthopedic implants in children: Morbidity and
postoperative radiologic changes. J. Pediatr. Orthop., Part
B 1999, 8 (2), 144–146.

10. Juutilainen, T.; Pa¨tia¨la¨, H.; Ruuskanen, M.;


Rokkanen, P. Comparison of costs in ankle fractures treated
with absorbable or metallic fixation devices. Arch. Orthop.
Trauma. Surg. 1997, 116 (4), 204–208.

11. Middleton, J.C.; Tipton, A.J. Synthetic biodegradable


polymers as orthopedic devices. Biomaterials 2000, 21,
2335–2346.

12. Hirashima, Y.; Ishiguro, N.; Kondo, S.; Iwata, H.


Osteoclast induction from bone marrow cells is due to
pro-inflammatory mediators from macrophages exposed to
polyethylene particles: A possible mechanism of osteolysis
in failed THA. J. Biomed. Mater. Res., Appl. Biomater.
2001, 56, 177–183.

13. Bo¨stman, O.M. Intense granulomatous inflammatory


lesions associated with absorbable internal fixation devices
made of polyglycolide in ankle fractures. Clin. Orthop.
1992, 278, 193–199.

14. Bo¨stman, O.; Pihlajama¨ki, H.K. Clinical


biocompatibility of biodegradable orthopaedic implants for
internal fixation: A review. Biomaterials 2000, 21,
2615–2621.

15. Pemberton, D.J.; McKibbin, B.; Savage, R.; Tayton, K.;


Stuart, D. Carbon-fibre reinforced plates for problem
fractures. J. Bone Jt. Surg. 1992, 74 (1), 88–92.

16. Tayton, K.; Johnson-Nurse, C.; McKibbin, B.; Bradley,


J.; Hastings, G. The use of semi-rigid
carbon-fibre-reinforced plastic plates for fixation of human
fractures. Results of preliminary trials. J. Bone Jt. Surg.
1982, 64 (1), 105–111. 17. Ali, M.S.; French, T.A.;
Hastings, G.W.; Rae, T.; Rushton, N.; Ross, E.R.;
Wynn-Jones, C.H. Carbon fibre composite bone plates.
Development, evaluation and early clinical experience. J.
Bone Jt. Surg. 1990, 72 (4), 586–591. 18. Brown, S.A.;
Hastings, R.S.; Mason, J.J.; Moet, A. Characterization of
short-fibre reinforced thermoplastics for fracture fixation
devices. Biomaterials 1990, 11 (8), 541–547. 19. Jockisch,
K.A.; Brown, S.A.; Bauer, T.W.; Merritt, K. Biological
response to chopped-carbon-fiber-reinforced peek. J. Biomed.
Mater. Res. 1992, 26 (2), 133–146. 20. Bonner, M.J.; Hine,
P.J.; Ward, I.M. Hot compaction of crosslinked high modulus
polyethylene fibers and fabrics. Plast. Rubber Compos.
Process. Technol. 1998, 27 (2), 58–64. 21. Hine, P.J.;
Ward, I.M.; Olley, R.H.; Bassett, D.C. The hot compaction
of high modulus melt-spun polyethylene fibres. J. Mater.
Sci. 1993, 28, 316–324. 22. Kabeel, M.A.; Bassett, D.C.;
Olley, R.H.; Hine, P.J.; Ward, I.M. Compaction of
high-modulus melt-spun polyethylene fibres at temperatures
above and below the optimum. J. Mater. Sci. 1994, 29,
4694–4699. 23. Kabeel, M.A.; Bassett, D.C.; Olley, R.H.;
Hine, P.J.; Ward, I.M. Differential melting in compacted
highmodulus melt-spun polyethylene fibres. J. Mater. Sci.
1995, 30, 601–606. 24. Morye, S.S.; Hine, P.J.; Duckett,
R.A.; Carr, D.J.; Ward, I.M. Comparison of the properties
of hot compacted gelspun polyethylene fibre composites with
conventional gel-spun polyethylene fibre composites.
Compos., Part A Appl. Sci. Manuf. 1999, 30 (5), 649–660.
25. Olley, R.H.; Bassett, D.C.; Hine, P.J.; Ward, I.M.
Morphology of compacted polyethylene fibres. J. Mater. Sci.
1993, 28, 1107–1112. 26. Tissington, B.; Pollard, G.; Ward,
I.M. A study of the influence of fibre/resin adhesion on the
mechanical behaviour of ultra-high-modulus polyethylene
fibre composites. J. Mater. Sci. 1991, 26, 82–92. 27. Ward,
I.M. A review of recent developments in the processing and
properties of oriented polyethylene. Plast. Rubber Compos.
Process. Appl. 1993, 19, 7–13. 28. Woods, D.W.; Ward, I.M.
Study of the interlaminar shear strength of unidirectional
high-modulus polyethylene fibre composites. Polymer 1994,
29, 2572– 2578. 29. Rasburn, J.; Hine, P.J.; Ward, I.M.;
Olley, R.H.; Bassett, D.C.; Kabeel, M.A. The hot compaction
of polyethylene terephthalate. J. Mater. Sci. 1995, 30,
615–622. 30. Abo El-Maaty, M.I.; Bassett, D.C.; Olley,
R.H.; Hine, P.J.; Ward, I.M. The hot compaction of
polypropylene fibres. J. Mater. Sci. 1996, 31, 1157–1163.
31. Wright, D.D.; Lautenschlager, E.P.; Gilbert, J.L.
Interfacial properties of self-reinforced composite
poly(methyl methacrylate). J. Biomed. Mater. Res. 1998, 43
(2), 153–161. 32. Wright, D.D.; Lautenschlager, E.P.;
Gilbert, J.L. Bending and fracture toughness of woven
self-reinforced composite poly(methyl methacrylate). J.
Biomed. Mater. Res. 1997, 36 (4), 441–453. 33. Andriano,
K.P.; Pohjonen, T.; To¨rma¨la¨, P. Processing and
characterization of absorbable polylactide polymers for use
in surgical implants. J. Appl. Biomater. 1994, 5, 133–140.
34. Ashammakhi, N.; Rokkanen, P. Absorbable polyglycolide
devices in trauma and bone surgery. Biomaterials 1997, 18
(1), 3–9. 35. Ashammakhi, N.; Peltoniemi, H.; Waris, E.;
Suuronen, R.; Serlo, W.; Kelloma¨ki, M.; To¨rma¨la¨, P.;
Waris, T. Developments in craniomaxillofacial surgery: Use
of self-reinforced bioabsorbable osteofixation devices.
Plast. Reconstr. Surg. 2001, 108 (1), 167–180. 36.
Hirvensalo, E.; Bo¨stman, O.; To¨rma¨la¨, P.; Vainionpa¨a¨,
S.; Rokkanen, P. Chevron osteotomy fixed with absorbable
polyglycolide pins. Foot Ankle 1991, 11 (4), 212–218. 37.
Jukkala-Partio, K.; Partio, E.K.; Hirvensalo, E.; Rokkanen,
P. Absorbable fixation of femoral head fractures. A
prospective study of six cases. Ann. Chir. Gynaecol. 1998,
87 (1), 44–48. 38. Jukkala-Partio, K.; Laitinen, O.;
Partio, E.K.; Vasenius, J.; Vainionpa¨a¨, S.; Pohjonen, T.;
To¨rma¨la¨, P.; Rokkanen, P. Comparison of the fixation of
subcapital femoral neck osteotomies with absorbable
self-reinforced polyL-lactide lag-screws or metallic screws
in sheep. J. Orthop. Res. 1997, 15 (1), 124–127. 39.
Juutilainen, T.; Hirvensalo, E.; Majola, A.; Partio, E.K.;
Pa¨tia¨la¨, H.; Rokkanen, P.; Kinnunen, J. Bone mineral
density in fractures treated with absorbable or metallic
implants. Ann. Chir. Gynaecol. 1997, 86 (1), 51–55. 40.
Kellomaki, M.; Paasimaa, S.; To¨rma¨la¨, P. Pliable
polylactide plates for guided bone regeneration:
Manufacturing and in vitro. Proc. Inst. Mech. Eng., Part H
J. Eng. Med. 2000, 214 (6), 615–629. 41. Koskikare, K.;
Hirvensalo, E.; Patiala, H.; Rokkanen, P.; Pohjonen, T.;
To¨rma¨la¨, P.; Lob, G. Intraosseous plating with
absorbable self-reinforced polyL-lactide plates in the
fixation of distal femoral osteotomies on rabbits. J.
Biomed. Mater. Res. 1996, 30 (4), 417–421. 42. Majola, A.;
Vainionpa¨a¨, S.; Vihtonen, K.; Mero, M.; Vasenius, J.;
To¨rma¨la¨, P.; Rokkanen, P. Absorption, biocompatibility,
and fixation properties of polylactic acid in bone tissue:
An experimental study in rats. Clin. Orthop. Relat. Res.
1991, 268, 260–269. 43. Ma¨kela¨, P.A.; Ruuskanen, M.;
Ashammakhi, N.; Kallioinen, M.; Pohjonen, T.; Serlo, W.;
To¨rma¨la¨, P.; Waris, T. Comparison of self-reinforced
polyL-lactide and steel wire in fixation of sternotomy in
rabbits. Ann. Chir. Gynaecol. 1999, 88 (4), 318–321. 44.
Niiranen, H.; To¨rma¨la¨, P. Bioabsorbable polymer plates
coated with bioactive glass spheres. J. Mater. Sci., Mater.
Med. 1999, 10 (12), 707–710. 45. Partio, E.K.; Hirvensalo,
E.; Bo¨stman, O.; Rokkanen, P. A prospective controlled
trial of the fracture of the humeral medial epicondyle–how
to treat? Ann. Chir. Gynaecol. 1996, 85 (1), 67–71. 46.
Peltoniemi, H.H.; Tulamo, R.M.; Toivonen, T.; Hallikainen,
D.; To¨rma¨la¨, P.; Waris, T. Biodegradable semirigid plate
and miniscrew fixation compared with rigid titanium fixation
in experimental calvarial osteotomy. J. Neurosurg. 1999, 90
(5), 910–917. 47. Rokkanen, P. Current clinical use of
absorbable fracture fixation devices. Ann. Chir. Gynaecol.
1991, 80 (3), 243–244. 48. Rokkanen, P.U.; Bo¨stman, O.M.;
Hirvensalo, E.; Makela, E.A.; Partio, E.K.; Patiala, H.;
Vainionpa¨a¨, S.; Vihtonen, K.; To¨rma¨la¨, P.
Bioabsorbable fixation in orthopaedic surgery and
traumatology. Biomaterials 2000, 21 (24), 2607–2613. 49.
Saikku-Backstrom, A.; Tulamo, R.M.; Pohjonen, T.;
To¨rma¨la¨, P.; Raiha, J.E.; Rokkanen, P. Material
properties of absorbable self-reinforced fibrillated
poly-96L/4 D-lactide (SR-PLA96) rods; A study in vitro and
in vivo. J. Mater. Sci., Mater. Med. 1999, 10 (1), 1–8. 50.
Sinisaari, I.; Pa¨tia¨la¨, H.; Bo¨stman, O.; Ma¨kela¨,
E.A.; Hirvensalo, E.; Partio, E.K.; To¨rma¨la¨, P.;
Rokkanen, P. Metallic or absorbable implants for ankle
fractures: A comparative study of infections in 3,111
cases. Acta Orthop. Scand. 1996, 67 (1), 16–18. 51.
To¨rma¨la¨, P.; Vasenius, J.; Vainionpa¨a¨, S.; Laiho, J.;
Pohjonen, T.; Rokkanen, P. Ultra-high-strength absorbable
self-reinforced polyglycolide (SR-PGA) composite rods for
internal fixation of bone fractures: In vitro and in vivo
study. J. Biomed. Mater. Res. 1991, 25 (1), 1–22. 52.
To¨rma¨la¨, P. Biodegradable self-reinforced composite
materials; Manufacturing structure and mechanical
properties. Clin. Mater. 1992, 10, 29–34. 53. Tuompo, P.;
Partio, E.; Rokkanen, P. Bioabsorbable fixation in the
treatment of proximal tibial osteotomies and fractures. A
clinical study. Ann. Chir. Gynaecol. 1999, 88 (1), 66–72.
54. Vasenius, J.; Vainionpa¨a¨, S.; Vihtonen, K.; Mero, M.;
Makela, A.; To¨rma¨la¨, P.; Rokkanen, P. Histomorphological
study on self-reinforced polyglycolide (SR-PGA)
osteosynthesis implants coated with slowly absorbable
polymers. J. Biomed. Mater. Res. 1990, 24 (12), 1615– 1635.
55. Ferguson, S.; Wahl, D.; Gogolewski, S. Enhancement of
the mechanical properties of polylactides by solidstate
extrusion. II. Poly( L-lactide), poly(L/D-lactide), and
poly( L/DL-lactide). J. Biomed. Mater. Res. 1996, 30,
543–551. 56. Serlo, W.; Kaarela, O.I.; Peltoniemi, H.H.;
Merikanto, J.; Ashammakhi, N.A.; Lassila, K. Use of
self-reinforced polylactide osteosynthesis devices in
craniofacial surgery: A long-term follow-up study. Scand.
J. Plast. Reconstr. Surg. Hand Surg. 2001, 35 (3), 285–
292. 57. McVicar, I.; Hatton, P.V.; Brook, I.M.
Self-reinforced polyglycolic acid membrane: A bioresorbable
material for orbital floor repair. Initial clinical report.
Br. J. Oral Maxillofac. Surg. 1995, 33 (4), 220–223. 58.
Pakkanen, M.; Salisbury, A.V.; Ersek, R.A. Biodegradable
positive fixation for the endoscopic brow lift. Plast.
Reconstr. Surg. 1996, 98 (6), 1087–1091. 59. Ambrosio,
A.M.; Sahota, J.S.; Khan, Y.; Laurencin, C.T. A novel
amorphous calcium phosphate polymer D ceramic for bone
repair: I. Synthesis and characterization. J. Biomed.
Mater. Res. 2001, 58 (3), 295–301.

60. Devin, J.E.; Attawia, M.A.; Laurencin, C.T.


Threedimensional degradable porous polymer-ceramic matrices
for use in bone repair. J. Biomater. Sci., Polym. Ed. 1996,
7 (8), 661–669.

61. Gomes, M.E.; Reis, R.L.; Cunha, A.M.; Blitterswijk,


C.A.; de Bruijn, J.D. Cytocompatibility and response of
osteoblastic-like cells to starch-based polymers: Effect of
several additives and processing conditions. Biomaterials
2001, 22 (13), 1911–1917.

62. Ignatius, A.A.; Betz, O.; Augat, P.; Claes, L.E. In


vivo investigations on composites made of resorbable
ceramics and poly(lactide) used as bone graft substitutes.
J. Biomed. Mater. Res. 2001, 58 (6), 701–709.

63. Marra, K.G.; Szem, J.W.; Kumta, P.N.; DiMilla, P.A.;


Weiss, L.E. In vitro analysis of biodegradable polymer
blend/hydroxyapatite composites for bone tissue
engineering. J. Biomed. Mater. Res. 1999, 47 (3), 324– 335.

64. Murphy, W.L.; Kohn, D.H.; Mooney, D.J. Growth of


continuous bonelike mineral within porous
poly(lactideco-glycolide) scaffolds in vitro. J. Biomed.
Mater. Res. 2000, 50 (1), 50–58.

65. Schliephake, H.; Kage, T. Enhancement of bone


regeneration using resorbable ceramics and a polymerceramic
composite material. J. Biomed. Mater. Res. 2001, 56 (1),
128–136.

66. Ignatius, A.A.; Augat, P.; Claes, L.E. Degradation


behavior of composite pins made of tricalcium phosphate and
poly( L,DL-lactide). J. Biomater. Sci., Polym. Ed. 2001, 12
(2), 185–194.

67. Kasuga, T.; Ota, Y.; Nogami, M.; Abe, Y. Preparation


and mechanical properties of polylactic acid composites
containing hydroxyapatite fibers. Biomaterials 2001, 22 (1),
19–23.

68. Vaz, C.M.; Reis, R.L.; Cunha, A.M. Use of coupling


agents to enhance the interfacial interactions in
starchEVOH/hydroxylapatite composites. Biomaterials 2002,
23 (2), 629–635.

69. Lin, F.H.; Chen, T.M.; Lin, C.P.; Lee, C.J. The merit
of sintered PDLLA/TCP composites in management of bone
fracture internal fixation. Artif. Organs 1999, 23 (2),
186–194.

70. Andriano, K.P.; Daniels, A.U.; Heller, J.


Biocompatibility and mechanical properties of a totally
absorbable composite material for orthopaedic fixation
devices. J. Appl. Biomater. 1992, 3 (3), 197–206.

71. Andriano, K.P.; Daniels, A.U.; Smutz, W.P.; Wyatt, R.W.


Preliminary biocompatibility screening of several
biodegradable phosphate fiber reinforced polymers. J. Appl.
Biomater. 1993, 4 (1), 1–12.

72. Furukawa, T.; Matsusue, Y.; Yasunaga, T.; Nakagawa, Y.;


Shikinami, Y.; Okuno, M.; Nakamura, T. Bone bonding ability
of a new biodegradable composite for internal fixation of
bone fractures. Clin. Orthop. 2000, 379, 247–258. 73.
Mendes, S.C.; Reis, R.L.; Bovell, Y.P.; Cunha, A.M.; van
Blitterswijk, C.A.; de Bruijn, J.D. Biocompatibility
testing of novel starch-based materials with potential
application in orthopaedic surgery: A preliminary study.
Biomaterials 2001, 22 (14), 2057–2064. 74. Knowles, J.C.;
Hastings, G.W.; Ohta, H.; Niwa, S.; Boeree, N. Development
of a degradable composite for orthopaedic use: In vivo
biomechanical and histological evaluation of two bioactive
degradable composites based on the polyhydroxybutyrate
polymer. Biomaterials 1992, 13 (8), 491–496. 75. Ignatius,
A.A.; Ohnmacht, M.; Claes, L.E.; Kreidler, J.; Palm, F. A
composite polymer/tricalcium phosphate membrane for guided
bone regeneration in maxillofacial surgery. J. Biomed.
Mater. Res. 2001, 58 (5), 564–569. 76. Nguyen, T.P.;
Dupraz, A. Spectroscopic studies of a multiphasic
polymer-ceramic mixture material. J. Biomater. Sci., Polym.
Ed. 1996, 8 (2), 141–149. 77. Qiu, Q.Q.; Ducheyne, P.;
Ayyaswamy, P.S. New bioactive, degradable composite
microspheres as tissue engineering substrates. J. Biomed.
Mater. Res. 2000, 52 (1), 66–76. 78. Tracy, B.M.; Doremus,
R.H. Direct electron microscopy studies of the
bone-hydroxylapatite interface. J. Biomed. Mater. Res.
1984, 18, 719–726. 79. Neo, M.; Nakamura, T.; Ohtsuki, C.;
Kokubo, T.; Yamamuro, T. Apatite formation on three kinds
of bioactive material at an early stage in vivo. A
comparative study by transmission electron microscopy. J.
Biomed. Mater. Res. 1993, 27, 999–1006. 80. Neo, M.;
Kotani, S.; Nakamura, T.; Yamamuro, T.; Ohtsuki, C.;
Kokubo, T.; Bando, Y. A comparative study of
ultrastructures of the interfaces between four kinds of
surface-active ceramic and bone. J. Biomed. Mater. Res.
1992, 26, 1419–1432. 81. Kangasniemi, I.M.; Va¨ha¨talo, K.;
Happonen, R.P.; YliUrpo, A.; de Groot, K. In vivo reactions
of Ca,P particle containing surface reactive glasses. J.
Biomed. Mater. Res. 1994, 28 (9), 993–1002. 82. Hench, L.L.
Bioactive ceramics. Ann. N.Y. Acad. Sci. 1988, 523, 54–71.
83. Ishaug, S.L.; Crane, G.M.; Miller, M.J.; Yasko, A.W.;
Yaszemski, M.J.; Mikos, A.G. Bone formation by
three-dimensional stromal osteoblast culture in
biodegradable polymer scaffolds. J. Biomed. Mater. Res.
1997, 36 (1), 17–28. 84. Cui, D.; Gao, H.A.B. Advance and
prospect of bionanomaterials. Biotechnol. Prog. 2003, 19
(3), 683–692. 85. Du, C.; Cui, F.Z.; Zhu, X.D.; de Groot,
K.A.D.o. Three-dimensional nano-HAp/collagen matrix loading
with osteogenic cells in organ culture. J. Biomed. Mater.
Res. 1999, 44 (4), 407–415. 86. Du, C.; Cui, F.Z.; Feng,
Q.L.; Zhu, X.D.; de Groot, K.A.D.o. Tissue response to
nano-hydroxyapatite/collagen composite implants in marrow
cavity. J. Biomed. Mater. Res. 1998, 42 (4), 540–548.
Dendrimers for Drug Delivery

13. Hawker, C.; Frechet, M.J. A new convergent approach to


monodisperse dendritic macromolecules. J. Chemical Soc.
Chem. Commun. 1990, (15), 1010–1013.

14. Ihre, H.; Hult, A.; Frechet, J.M.J.; Gitsov, I.


Doublestage convergent approach for the synthesis of
functionalized dendritic aliphatic polyesters based on
2,2bis(hydroxymethyl)propionic acid. Macromolecules 1998,
31 (13), 4061–4068.

15. L’Abbe, G.; Forier, B.; Dehaen, W. A fast double-stage


convergent synthesis of dendritic polyethers. Chem. Commun.
(Cambridge) 1996, (18), 2143–2144.

16. Wooley, K.L.; Hawker, C.J.; Frechet, J.M.J.


Hyperbranched macromolecules via a novel double-stage
convergent growth approach. J. Am. Chem. Soc. 1991, 113
(11), 4252–4261.

17. Kawaguchi, T.; Walker, K.L.; Wilkins, C.L.; Moore, J.S.


Double exponential dendrimer growth. J. Am. Chem. Soc.
1995, 117 (8), 2159–2165.

18. Zeng, F.; Zimmerman, S.C. Rapid synthesis of dendrimers


by an orthogonal coupling strategy. J. Am. Chem. Soc. 1996,
118 (22), 5326–5327.

19. Zimmerman, S.C.; Zeng, F.; Reichert, D.E.; Kolotuchin,


S.V. Self-assembling dendrimers. Science 1996, 271 (5252),
1095–1098.

20. de Brabander-van den Berg, E.M.M.; Meijer, E.W.


Poly(propylenimine) dendrimers: large-scale synthesis via
heterogeneously catalyzed hydrogenation. Angewandte Chemie.
1993, 105 (9), 1370–1372; Angew. Chem. Int. Ed. Engl. 1993,
32 (9),1308–1311.

21. Hawker, C.J.; Lee, R.; Frechet, J.M.J. One-step


synthesis of hyperbranched dendritic polyesters. J. Am.
Chem. Soc. 1991, 113 (12), 4583–4588.

22. Uhrich, K.E.; Boegeman, S.; Frechet, J.M.J.; Turner,


S.R. The solid-phase synthesis of dendritic polyamides.
Polymer Bulletin (Berlin, Germany) 1991, 25 (5), 551–558.

23. Launay, N.; Caminade, A.-M.; Majoral, J.-P. Synthesis


and reactivity of unusual phosphorus dendrimers a useful
divergent growth approach up to the seventh generation. J.
Am. Chem. Soc. 1995, 117 (11), 3282–3283.

24. Salamonczyk, G.M.; Kuznikowski, M.; Skowronska, A. A


divergent synthesis of thiophosphate-based dendrimers.
Tetrahedron Lett. 2000, 41 (10), 1643–1645.

25. Zhang, W.; Simanek, E.E. Dendrimers based on melamine


divergent and orthogonal, convergent syntheses of a G3
dendrimer. Organic Lett. 2000, 2 (6), 843–845.

26. Morikawa, A.; Kakimoto, M.; Imai, Y. Convergent


synthesis of siloxane starburst dendrons and dendrimers via
hydrosilylation. Macromolecules 1992, 25 (12), 3247–3253.

27. Shao, J.; Tam, J.P. Unprotected peptides as building


blocks for the synthesis of peptide dendrimers with oxime,
hydrazone, and thiazolidine linkages. J. Am. Chem. Soc.
1995, 117 (14), 3893–3899.

28. Jansen, J.F.G.A.; de Brabander van den Berg, E.M.M.;


Meijer, E.W. Encapsulation of guest molecules into a
dendritic box. Science (Washington, DC) 1994, 266 (5188),
1226–1229.

29. Hawker, C.J.; Wooley, K.L.; Frechet, J.M.J.


Unimolecular micelles and globular amphiphiles: dendritic
macromolecules as novel recyclable solubilization agents.
J. Chem. Soc. Perkin Trans. 1. 1993, 12, 1287–1297. 30.
Liu, M.; Frechet, J.M.J. Preparation, MALDI-TOF analysis,
and micelle-like behavior of alkyl-modified poly(propylene
imine) dendrimers. Polym. Bull. (Berlin) 1999, 43 (4–5),
379–386. 31. Heise, A.; Hedrick, J.L.; Frank, C.W.; Miller,
R.D. Starlike block copolymers with amphiphilic arms as
models for unimolecular micelles. J. Am. Chem. Soc. 1999,
121 (37), 8647–8648. 32. Liu, H.; Jiang, A.; Guo, J.;
Uhrich, K.E. Unimolecular micelles: synthesis and
characterization of amphiphilic polymer systems. J. Polym.
Sci. Part A: Polym. Chem. 1999, 37 (6), 703–711. 33.
Milhem, O.M.; Myles, C.; McKeown, N.B.; Attwood, D.;
D’Emanuele, A. Polyamidoamine starburst dendrimers as
solubility enhancers. Int. J. Pharm. 2000, 197 (1–2),
239–241. 34. Roberts, J.C.; Bhalgat, M.K.; Zera, R.T.
Preliminary biological evaluation of polyamidoamine (PAMAM)
starburst dendrimers. J. Biomed. Mater. Res. 1996, 30 (1),
53–65. 35. Li, J.; Piehler, L.T.; Qin, D.; Baker, J.R.,
Jr.; Tomalia, D.A.; Meier, D.J. Visualization and
characterization of poly(amidoamine) dendrimers by atomic
force microscopy. Langmuir 2000, 16 (13), 5613–5616. 36.
Bhalgat, M.K.; Roberts, J.C. Molecular modeling of
polyamidoamine (PAMAM) starburst dendrimers. Eur. Polym. J.
2000, 36 (3), 647–651. 37. Kojima, C.; Kono, K.; Maruyama,
K.; Takagishi, T. Synthesis of polyamidoamine dendrimers
having poly(ethylene glycol) grafts and their ability to
encapsulate anticancer drugs. Bioconjugate Chem. 2000, 11
(6), 910–917. 38. Sideratou, Z.; Tsiourvas, D.; Paleos,
C.M. Solubilization and release properties of pegylated
diaminobutane poly(propylene imine) dendrimers. J. Colloid
Interface Sci. 2001, 242 (1), 272–276. 39. Yang, H.;
Morris, J.J.; Lopina, S.T. Polyethylene
glycol-polyamidoamine dendritic micelle as solubility
enhancer and the effect of the length of polyethylene
glycol arms on the solubility of pyrene in water. J.
Colloid Interface Sci. 2004, 273 (1), 148–154. 40. Gitsov,
I.; Zhu, C. Amphiphilic hydrogels constructed by
poly(ethylene glycol) and shape-persistent dendritic
fragments. Macromolecules 2002, 35 (22), 8418–8427. 41. Wu,
X.-Y.; Huang, S.-W.; Zhang, J.-T.; Zhuo, R.-X. Preparation
and characterization of novel physically cross-linked
hydrogels composed of poly(vinyl alcohol) and
amine-terminated polyamidoamine dendrimer. Macromolecular
Bioscience 2004 , 4 (2), 71–75. 42. El Ghzaoui, A.;
Gauffre, F.; Caminade, A.-M.; Majoral, J.-P.;
Lannibois-Drean, H. Self-assembly of water-soluble
dendrimers into thermoreversible hydrogels and macroscopic
fibers. Langmuir 2004, 20 (21), 9348–9353. 43. Braun, C.S.;
Vetro, J.A.; Tomalia, D.A.; Koe, G.S.; Koe, J.G.; Middaugh,
C.R. Structure/function relationships of polyamidoamine/DNA
dendrimers as gene delivery vehicles. J. Pharm. Sci. 2005,
94 (2), 423–436. 44. Luo, D.; Haverstick, K.; Belcheva, N.;
Han, E.; Saltzman, W.M. Polyethylene glycol-conjugated
PAMAM dendrimer for biocompatible, high-efficiency DNA
delivery. Macromolecules 2002, 35 (9), 3456–3462. 45. Kim,
T.I.; Seo, H.J.; Choi, J.S.; Jang, H.S.; Baek, J.U.; Kim,
K.; Park, J.S. PAMAM-PEG-PAMAM: novel triblock copolymer as
a biocompatible and efficient gene delivery carrier.
Biomacromolecules 2004, 5 (6), 2487–2492. 46. Schatzlein,
A.G.; Zinselmeyer, B.H.; Elouzi, A.; Dufes, C.; Chim,
Y.T.A.; Roberts, C.J.; Davies, M.C.; Munro, A.; Gray, A.I.;
Uchegbu, I.F. Preferential liver gene expression with
polypropylenimine dendrimers. J. Contro. Release 2005, 101
(1–3), 247–258. 47. Bielinska, A.U.; Kukowska-Latallo,
J.F.; Baker, J.R., Jr. The interaction of plasmid DNA with
polyamidoamine dendrimers: mechanism of complex formation
and analysis of alterations induced in nuclease sensitivity
and transcriptional activity of the complexed DNA. Biochim.
Biophys. Acta 1997, 1353 (2), 180–190. 48. Veronese, F.M.;
Morpurgo, M. Bioconjugation in pharmaceutical chemistry. IL
Farmaco. 1999, 54 (8), 497–516. 49. Quintana, A.; Raczka,
E.; Piehler, L.; Lee, I.; Myc, A.; Majoros, I.; Patri,
A.K.; Thomas, T.; Mule, J.; Baker, J.R., Jr. Design and
function of a dendrimer-based therapeutic nanodevice
targeted to tumor cells through the folate receptor. Pharm.
Res. 2002, 19 (9), 1310–1316. 50. Kobayashi, H.; Reijnders,
K.; English, S.; Yordanov, A.T.; Milenic, D.E.; Sowers,
A.L.; Citrin, D.; Krishna, M.C.; Waldmann, T.A.; Mitchell,
J.B.; Brechbiel, M.W. Application of a macromolecular
contrast agent for detection of alterations of tumor vessel
permeability induced by radiation. Clin. Cancer. Res. 2004,
10 (22), 7712–7720. 51. Ong, K.K.; Jenkins, A.L.; Cheng,
R.; Tomalia, D.A.; Durst, H.D.; Jensen, J.L.; Emanuel,
P.A.; Swim, C.R.; Yin, R. Dendrimer enhanced immunosensors
for biological detection. Analytica Chimica Acta 2001, 444
(1), 143–148. 52. Yang, H.; Lopina, S.T. Penicillin
V-conjugated PEGPAMAM star polymers. J. Biomater. Sci.
Polym. Ed. 2003, 14 (10), 1043–1056. 53. Zanini, D.; Park,
W.K.C.; Roy, R. Synthesis of novel dendritic glycosides.
Tetrahedron Lett. 1995, 36 (41), 7383–7386. 54. Woller,
E.K.; Cloninger, M.J. The lectin-binding properties of six
generations of mannose-functionalized dendrimers. Org.
Lett. 2002, 4 (1), 7–10. 55. Shaunak, S.; Thomas, S.;
Gianasi, E.; Godwin, A.; Jones, E.; Teo, I.; Mireskandari,
K.; Luthert, P.; Duncan, R.; Patterson, S.; Khaw, P.;
Brocchini, S. Polyvalent dendrimer glucosamine conjugates
prevent scar tissue formation. Nature Biotechnol. 2004, 22
(8), 977–984. 56. Bracci, L.; Falciani, C.; Lelli, B.;
Lozzi, L.; Runci, Y.; Pini, A.; De Montis, M.G.;
Tagliamonte, A.; Neri, P. Synthetic peptides in the form of
dendrimers become resistant to protease activity. J. Biol.
Chem. 2003, 278 (47), 46,590–46,595. 57. Spetzler, J.C.;
Tam, J.P. Unprotected peptides as building blocks for
branched peptides and peptide dendrimers. Int. J. Pept.
Protein Res. 1995, 45 (1), 78–85. 58. Sadler, K.; Tam
James, P. Peptide dendrimers: applications and synthesis.
J. biotechnol. 2002, 90 (3–4), 195–229. 59. Marano, R.J.;
Wimmer, N.; Kearns, P.S.; Thomas, B.G.; Toth, I.; Brankov,
M.; Rakoczy, P.E. Inhibition of in vitro VEGF expression
and choroidal neovascularization by synthetic dendrimer
peptide mediated delivery of a sense oligonucleotide. Exp.
Eye Res. 2004, 79 (4), 525–535. 60. Santhakumaran, L.M.;
Thomas, T.; Thomas, T.J. Enhanced cellular uptake of a
triplex-forming oligonucleotide by nanoparticle formation
in the presence of polypropylenimine dendrimers. Nucleic
Acids Res. 2004, 32 (7), 2102–2112. 61. Vandamme, T.F.;
Brobeck, L. Poly(amidoamine) dendrimers as ophthalmic
vehicles for ocular delivery of pilocarpine nitrate and
tropicamide. J. Control Release 2005, 102 (1), 23–38. 62.
Choi, Y.; Thomas, T.; Kotlyar, A.; Islam, M.T.; Baker,
J.R., Jr. Synthesis and functional evaluation of
DNAassembled polyamidoamine dendrimer clusters for cancer
cell-specific targeting. Chem. Biol. 2005, 12 (1), 35–43.
63. Li, Y.; Tseng, Y.D.; Kwon, S.Y.; D’Espaux, L.; Bunch,
J.S.; McEuen, P.L.; Luo, D. Controlled assembly of
dendrimer-like DNA. Nat. Mater. 2004, 3 (1), 38–42. 64.
Esfand, R.; Tomalia, D.A. Poly(amidoamine) (PAMAM)
dendrimers: from biomimicry to drug delivery and biomedical
applications. Drug Discovery Today 2001, 6 (8), 427–436.
65. Forssen, E.; Willis, M. Ligand-targeted liposomes. Adv.
Drug Delivery Rev. 1998, 29 (3), 249–271. 66. Spragg, D.D.;
Alford, D.R.; Greferath, R.; Larsen, C.E.; Lee, K.-D.;
Gurtner, G.C.; Cybulsky, M.I.; Tosi, P.F.; Nicolau, C.;
Gimbrone, M.A., Jr. Immunotargeting of liposomes to
activated vascular endothelial cells: a strategy for
site-selective delivery in the cardiovascular system. Proc.
Natl. Acad. Sci. USA 1997, 94 (16), 8795–8800. 67.
Vandorpe, J.; Schacht, E.; Dunn, S.; Hawley, A.; Stolnik,
S.; Davis, S.S.; Garnett, M.C.; Davies, M.C.; Illum, L.
Long circulating biodegradable poly(phosphazene)
nanoparticles surface modified with
poly(phosphazene)poly(ethylene oxide) copolymer.
Biomaterials 1997, 18 (17), 1147–1152. 68. Malik, N.;
Wiwattanapatapee, R.; Klopsch, R.; Lorenz, K.; Frey, H.;
Weener, J.W.; Meijer, E.W.; Paulus, W.; Duncan, R.
Dendrimers: relationship between structure and
biocompatibility in vitro, and preliminary studies on the
biodistribution of 125I-labeled polyamidoamine dendrimers
in vivo. J. Control. Release 2000, 65 (1–2), 133–148. 69.
Chen, H.T.; Neerman, M.F.; Parrish, A.R.; Simanek, E.E.
Cytotoxicity, hemolysis, and acute in vivo toxicity of
dendrimers based on melamine, candidate vehicles for drug
delivery. J. Am. Chem. Soc. 2004, 126 (32), 10,044–10,048.
70. Wiwattanapatapee, R.; Carreno-Gomez, B.; Malik, N.;
Duncan, R. Anionic PAMAM dendrimers D rapidly cross adult
rat intestine in vitro: a potential oral delivery system?
Pharmaceutical Res. 2000, 17 (8), 991– 998.

71. Jevprasesphant, R.; Penny, J.; Attwood, D.; D’Emanuele,


A. Transport of dendrimer nanocarriers through epithelial
cells via the transcellular route. J. Control. Release
2004, 97 (2), 259–267.

72. Kannan, S.; Kolhe, P.; Raykova, V.; Glibatec, M.;


Kannan, R.M.; Lieh-Lai, M.; Bassett, D. Dynamics of
cellular entry and drug delivery by dendritic polymers into
human lung epithelial carcinoma cells. J. Biomater. Sci.
Polym. Ed. 2004, 15 (3), 311–330. 73. Thomas, T.P.; Patri,
A.K.; Myc, A.; Myaing, M.T.; Ye, J.Y.; Norris, T.B.; Baker,
J.R., Jr. In vitro targeting of synthesized
antibody-conjugated dendrimer nanoparticles.
Biomacromolecules 2004, 5 (6), 2269–2274. 74. Florence,
A.T.; Sakthivel, T.; Toth, I. Oral uptake and translocation
of a polylysine dendrimer with a lipid surface. J. Control.
Release 2000, 65 (1–2), 253–259.
Dental Biomechanics

1. Berkovitz, B.K.B.; Moxham, B.J.; Newmann, H.N. The


Periodontal Ligament in Health and Disease; MosbyWolfe:
London, 1995.

2. Mu¨hlemann, H.R. Ten years of tooth mobility


measurements. J. Periodontol. 1960, 31, 110–122.

3. Roberts, W.E.; Huja, S.; Roberts, J.A. Bone modelling:


biomechanics, molecular mechanisms, and clinical
perspectives. Semin. Orthod. 2004, 10 (2), 123–161.

4. Schroeder, H.E. Orale Strukturbiologie; Georg Thieme


Verlag: Stuttgard, 1987.

5. Nishihira, M.; Yamamoto, K.; Sato, Y.; Ishikawa, H.;


Natali, A.N. Mechanics of periodontal ligament. In Dental
Biomechanics; Natali, A.N., Ed.; Taylor & Francis: London,
2003; 20–34.

6. Dorow, C.; Krstin, N.; Sander, F.G. Experiments to


determine the material properties of the periodontal
ligament. J. Orofac. Orthop. 2002, 63, 94–104.

7. Maurel, W.; Wu, Y.; Magnenat Thalmann, N.; Thalmann, D.


Biomechanical Models for Soft Tissue Simulation; Springer:
Berlin, 1998.

8. Pini, M.; Zysset, Ph.; Botsis, J.; Contro, R. Tensile


and compressive behaviour of the bovine periodontal
ligament. J. Biomech. 2004, 37 (1), 111–119.

9. Dorow, C.; Krstin, N.; Sander, F.G. Determination of the


mechanical properties of the periodontal ligament in a
uniaxial tensional experiment. J. Orofac. Orthop. 2003, 64,
100–107.

10. Parfitt, G.J. Measurement of the physiological mobility


of individual teeth in an axial direction. J. Dent. Res.
1960, 39 (3), 608–618.

11. Mu¨hlemann, H.R. The role of interdental contact points


and of activation on tooth mobility. J. Periodontol. 1955,
25, 125–137.

12. Dorow, C.; Schneider, J.; Sander, F.G. Finite element


simulation of in vivo tooth mobility in comparison with
experimental results. J. Mech. Med. Biol. 2003, 3 (1),
79–94.
13. Commer, P.; Bourauel, C.; Maier, K.; Ja¨ger, A.
Construction and testing of a computer-based intraoral
laser scanner for determining tooth positions. Med. Eng.
Phys. 2000, 22, 625–635.

14. Andersen, K.; Mortensen, H.T.; Pedersen, E.H.; Melsen,


B. Determination of stress levels and profiles in the
periodontal ligament by means of an improved
threedimensional finite element model for various types of
orthodontic and natural force. J. Biomed. Eng. 1991, 13,
293–303.

15. Natali, A.N.; Pavan, P.G.; Scarpa, C. Numerical


analysis of tooth mobility: formulation of a non-linear
constitutive law for the periodontal ligament. Dent. Mater.
2004, 20 (7), 623–629. 16. Pietrzak, G.; Curnier, A.;
Botsis, J.; Scherrer, S.; Wiskott, A.; Belser, U. A
nonlinear elastic model of the periodontal ligament and its
numerical calibration for the study of tooth mobility.
Comput. Methods Biomech. Biomed. Eng. 2002, 5 (2), 91–100.
17. Natali, A.N.; Pavan, P.G.; Carniel, E.L.; Dorow, C. A
transversally isotropic elasto-damage constitutive model
for the periodontal ligament. Comput. Methods Biomech.
Biomed. Eng. 2003, 6, 329–336. 18. Natali, A.N.; Pavan,
P.G.; Carniel, E.L.; Dorow, C. Visco-elastic response of
the periodontal ligament: an experimental-numerical
approach. J. Connect. Tissue Res. 2004, 45, 222–230. 19.
Natali, A.N.; Pavan, P.G.; Schrefler, B.A.; Secchi, S. A
multi-phase formulation for biomechanical analysis of
periodontal ligament. Meccanica 2002, 15, 345–352. 20.
Ross, G.G.; Lear, C.S.; DeCou, R. Modelling the lateral
movement of teeth. J. Biomech. 1976, 9, 723–734. 21.
Kawarizadeh, A.; Bourauel, C.; Zhang, D.; Go¨tz, W.;
Ja¨ger, A. Correlation of stress and strain profiles and the
distribution of osteoclastic cells induced by orthodontic
loading in rat. Eur. J. Oral Sci. 2004, 112, 140–147. 22.
Albrektsson, T.; Sennerby, L. State of the art in oral
implants. J. Clin. Periodontol. 1991, 18, 474–481. 23.
Sahin, S.; Cehre, M.C. The significance of passive framework
fit in implant prosthodontics: current status. Implant Dent.
2001, 10, 895–92. 24. Kasemo, B. Biocompatibility of
titanium implants: surface science aspects. J. Prosthet.
Dent. 1983, 2, 335–380. 25. Bra˚nemark, P.I.; Zarb, G.;
Albrektsson, T. Tissue Integrated Prostheses; Quintessence:
Chicago, 1985. 26. Klein, C.P.A.T.; Patka, P.; Wolke,
J.G.C.; de BlieckHogervost, J.M.A.; de Groot, K.
Plasma-sprayed coatings of tetracalcium-phosphate,
hydroxyapatite and TCP on titanium alloy: an interface
study. J. Biomed. Mater. Res. 1991, 25, 53–65. 27. Brunski,
J.B. Biomechanical forces affecting the bone-dental implant
interface. Dent. Mater. 1992, 10, 153–201. 28.
Merickse-Stern, R.; Piotti, M.; Sirtes, G. 3-D in vivo
force measurements on mandibular implants supporting
overdentures. Clin. Oral Implants Res. 1996, 7, 387–396.
29. Rangert, B.; Krough, P.H.; Langer, B.; Van Roekel, N.
Bending overload and implant fracture: a retrospective
clinical analysis. Int. J. Oral Maxillofac. Implants 1995,
10, 326–334. 30. Hart, R.T. Bone modeling and remodeling:
theories and computation. In Bone Mechanics Handbook;
Cowin, S.C., Ed.; CRC Press: Boca Raton, 2001; 1–42. 31.
Hoshaw, S.J.; Brunski, J.B.; Cochran, G.V.B. Mechanical
loading of Bra˚nemark implants affects interfacial bone
modelling and remodelling. Int. J. Oral Maxillofac.
Implants 1994, 9, 345–360. 32. Soncini, M.; Pietrabissa,
R.; Natali, A.N.; Pavan, P.G.; Williams, K.R. Testing the
reliability of dental implant devices. In Dental
Biomechanics; Natali, A.N., Ed.; Taylor & Francis: London,
2003; 111–131. 33. Carr, A.B.; Gerard, D.A.; Larsen, P.E.
The response of bone in primates around unloaded dental
implants supporting prostheses with different levels of fit.
J. Prosthet. Dent. 1996, 76, 500–509. 34. Natali, A.N.;
Pavan, P.G. Numerical approach to dental biomechanics. In
Dental Biomechanics; Natali, A.N., Ed.; Taylor & Francis:
London, 2003; 211–239. 35. Rodriguez, A.; Aquilino, S.;
Lund, P.; Ryther, J.; Southard, T. Evaluation of strain at
the terminal abutment site of a fixed mandibular prosthesis
during cantilever loading. Int. J. Prosthodont. 1993, 2,
93–102. 36. Chang, M.C.; Ko, C.C.; Liu, C.C.; Douglas,
W.H.; DeLong, R.; Seong, W.J.; Hodges, J.; An, K.N.
Elasticity of alveolar bone near dental implant-bone
interfaces after one month’s healing. J. Biomech. 2003, 36,
1209–1214. 37. Bra˚nemark, R.; Ohrnell, L.O.; Nilsson, P.;
Thomsen, P. Biomechanical characterization of
osseointegration during healing: an experimental in vivo
study in the rat. Biomaterials 1997, 18, 969–978. 38.
Natali, A.N.; Hart, R.T.; Pavan, P.G.; Knets, I. Mechanics
of bone tissue. In Dental Biomechanics; Natali, A.N., Ed.;
Taylor & Francis: London, 2003; 1–19. 39. Natali, A.N.;
Pavan, P.G. A comparative analysis based on different
strength criteria for evaluation of risk factor for dental
implants. Comput. Methods Biomech. Biomed. Eng. 2002, 5,
511–523. 40. Hansson, S.; Norton, M. The relation between
surface roughness and interfacial shear strength for
bone-anchored implants. A mathematical model. J. Biomech.
1999, 32, 829–83. D
Dental Implants

1. Anjard, R. Mayan dental wonders. Oral Implant 1981, 9,


423.

2. Proussaefs, P. Evaluation of two Vitallium blade-form


implants retrieved after 13–21 years of function: A
clinical report. J. Prosthet. Dent. 2002, 87, 412–415.

3. Bodine, R.L. Forty years of experience with


subperiosteal implant denture in 41 edentulous patients. J.
Prosthet. Dent. 1996, 75 (1), 33–44. Jan.

4. Branemark, P.-I.; Adell, R.; Breine, U.; Hansson, B.O.;


Lindstrom, J.; Ohlsson, A. Intra-osseous anchorage of
dental prostheses. I. Experimental studies. Scand. J.
Plast. Reconstr. Surg. 1969, 3, 81–100.

5. Branemark, P.-I.; Hansson, B.O.; Adell, R.; Breine, U.;


Lindstrom, J.; Hallen, O.; Ohman, A. Osseointegrated
implants in the treatment of the edentulous jaw. Scand. J.
Plast. Reconstr. Surg. 1977, 16 (Suppl.).

6. Adell, R.; Lekholm, U.; Rocklar, B.; Branemark, P.-I. A


15-year study of osseointegrated implants in the treatment
of edentulous jaw. Int. J. Oral. Surg. 1981, 10, 387–416.

7. Lindhe. Textbook, pp. 852–853.

8. Schroeder, W.; van der Zypen, E.; Stich, H.; Sutter, F.


The reactions of bone, connective tissue and epithelium to
endosteal implants with titanium-sprayed surfaces. J.
Maxillofac. Surg. 1981, 9, 15–25.

9. Ericsson, I.; Nilner, K. Early functional loading using


Branemark dental implants. Int. J. Periodontics Restor.
Dent. 2002, 22, 9–19.

10. Pierrisnard, L.; Hure, G.; Berquins, M.; Chappard, D.


Two dental implants designed for immediate loading: A finite
element analysis. Int. J. Oral Maxillofac. Implants 2002,
17, 353–362. 11. Andersen, E.; Haanaes, H.R.; Knusten, B.M.
Immediate loading of single tooth ITI implants in the
anterior maxilla: A prospective 5-year pilot study. Clin.
Oral Implants Res. 2002, 13, 281–287. 12. Albrektsson, T.;
Zarb, G.A.; Warthington, P.; et al. The long term efficacy
of currently used dental implants: A review and proposed
criteria of success. Int. J. Oral Maxillofac. Implants
1986, 1, 1–25. 13. Zarb, G.A.; Albrektsson, T.
Osseointegration—A requiem for the periodontal ligament? An
editorial. Int. J. Periodontics Restor. Dent. 1995, 11,
88–91. 14. Albrektsson, T.; Branemark, P.-I.; Hansson,
H.-A.; Lindstrom, J. Osseointegrated titanium implants.
Requirements for ensuring a long lasting, direct bone
anchorage in man. Acta Orthop. Scand.52, 155–170. 15.
Schenk, K.; Buser, D. Implant dentistry. Periodontology
2000, 17, 1998. 16. Binon, P.P. Implants and components:
Entering the new millenium. JOMI 2000, 15, 76–94. 17.
Sykaras, N.; Iacopino, A.M.; et al. JOMI 2000, 15, 675–690.
18. Strong, J.T.; Misch, C.E.; Nalluri, P. Functional
surface area: Thread form parameter optimization for
implant body design. Compendium 1998, 19 (3), 4–9. Special
issue. 19. Bobyn, J.D.; Pilliar, R.M.; Cameron, H.U.;
Weatherly, G.C. The optimum pore size for the fixation of
poroussurfaces metal implants by the ingrowth of bone.
Clin. Orthop. 1980, 150, 263–270. 20. Krauser, J.T.
Hydroxyl apatite coated dental implants. Biologic rationale
and surgical technique. Dent. Clin. North Am. 1989, 33 (4),
879–903. Oct. 21. Lee, J.J.; Rouhfar, L.; Beirne, O.R.
Survival of the hydroxyapatite coated dental implants: A
meta analytic review. J. Oral Maxillofac. Surg. 2000, 58,
1372–1379. 22. Lazzara, R.J.; Testori, T.; Trisi, P.;
Porter, S.S.; Weinstein, R.L. A human histologic analysis
of osseotite and machined surface using implants with two
opposing surfaces. Int. J. Periodontics Restor. Dent. 1999,
19 (2), 117–129. 23. London, R.M.; Roberts, F.A.; Baker,
D.A.; Rohrer, M.D.; O’Neal, R.B. Histologic comparison of a
thermal dual-etched implant surface to machined, TPS and HA
surfaces: Bone contact in-vivo in rabbits. Int. J. Oral
Maxillofac. Implants 2002, 17, 369–376. 24. Wong, M.;
Eulenberger, J.; Schenk, R.; Hunziker, E. Effect of surface
topology on the osseointegration of implant materials in
trabecular bone. J. Biomed. Mater. Res. 1995, 29,
1567–1576. 25. Ann. Periodontol. 1996. 26. # Year results
from the VA multicenter prospective implant study. Ann.
Periodontol. December 2000, 5 (1). 27. Raadsheer, M.C.; van
Eijden, T.M.G.J.; van Ginkel, F.C.; Prahl-Andersen, B.
Contribution of jaw muscle size and craniofacial morphology
to human bite force magnitude. J. Dent. Res. 1999, 78,
31–42. 28. Mericske-Stern, R.; Zarb, G.A. In-vivo
measurements of some functional aspects with mandibular
fixed prostheses supported by implants. Clin. Oral Implants
Res. 1996, 7, 153–161. 29. Campelo, L.D.; Camara, J.R.D.
Flapless implant surgery: A 10 year clinical retrospective
analysis. Int. J. Oral Maxillofac. Implants 2002, 17,
271–276. 30. Binderman, I.; Bahar, H.; Yaffe, A. Strain
relaxation of fibroblasts in the marginal periodontium is
the common trigger for alveolar bone resorption: A novel
hypothesis. J. Periodontol. 2002, 73, 1210–1215. 31. Gaggl,
A.; Schultes, G. Assessment of accuracy of navigated
implant placement in the maxila. Int. J. Oral Maxillofac.
Implants 2002, 17, 263–270. D
Diamond and Diamond-Like Carbons

1. Fu, Y.; Yan, B.; Loh, N.L. Effects of pretreatments and


interlayers on the nucleation and growth of diamond
coatings on titanium substrates. Surf. Coat. Technol. 2000,
130, 173–185.

2. Rats, D.; Vandenbulcke, L.; Boher, C.; Farges, G.


Tribological study of diamond coatings on titanium alloys.
Surf. Coat. Technolnol. 1997, 94–95, 555–560.

3. Dearnaley, G.

4. Seal, M. The Current Status of CVD Diamond Applications


and Prospects for the Future. In Applications of Diamond
Films and Related Materials; Feldman, A., Tzeng, Y.,
Yarbrough, W.A., Yoshikawa, M., Murakawa, M., Eds.;
National Institute of Standards and Technology: Washington,
DC, 1995; 3–10.

5. Liu, Y.; Erdemir, A.; Meletis, E.I. A study of the wear


mechanisms of diamond-like carbon films. Surf. Coat.
Technolnol. 1996, 82, 48–56.

6. Wei, Q.; Sankos, J.; Narayan, J. Structure and


properties of novel functional diamond-like carbon coatings
produced by laser ablation. Surf. Coat. Technol. 2001,
146–147, 250–257.

7. Fountzoulas, C.G.; Kattamis, T.Z.; Demaree, J.D.;


Hirvonen, J.K. Silicon-Containing DLC Coatings Formed by
Ion Beam Assisted Deposition and Properties of a New
Category of Hard, Wear Resistant Thin Films. In
Applications of Diamond Films and Related Materials;
Feldman, A., Tzeng, Y., Yarbrough, W.A., Yoshikawa, M.,
Murakawa, M., Eds.; National Institute of Standards and
Technology: Washington, DC, 1995; 907–910.

8. Xu, S.; Tay, B.K.; Tan, H.S.; Zhong, L.; Tu, Y.Q.;
Silva, S.R.P.; Milne, W.I. Properties of carbon ion
deposited tetrahedral amorphous films as a function of ion
energy. J. Appl. Phys. 1996, 79, 7234–7240.

9. Anders, S.; Anders, A.; Brown, I.G.; Wei, B.;


Komvopoulos, K.; Ager, J.W.; Yu, K.M. Effect of vacuum arc
deposition parameters on the properties of amorphous carbon
thin films. Surf. Coat. Technol. 1994, 68–69, 388–393. 10.
Peng, X.L.; Barber, Z.H.; Clyne, T.W. Surface roughness of
DLC films prepared using various techniques. Surf. Coat.
Technolnol. 2001, 138, 23–32. 11. Erdemis, A.; Nilufer,
J.B.; Eryilmaz, O.C.; Beschliesser, M.; Fenske, G.R.
Friction and wear performance of DLC films grown in various
source gas plasmas. Surf. Coat. Technol. 1999, 120–121,
589–593. 12. Liu, Y.; Meletis, E.I. Evidence of
graphitization of DLC films during sliding wear. J. Mater.
Sci. 1997, 32, 3491–3495. 13. Jiang, J.; Zhang, S.; Arnell,
R.D. The effect of relative humidity on wear of a
diamond-like carbon coating. Surf. Coat. Technol. 2003,
167, 221–225. 14. Saikko, V.; Ahlroos, T.; Calonius, O.;
Kera¨nen, J. Wear simulation of total hip prostheses with
polyethylene against CoCr, alumina and DLC. Biomaterials
2001, 22, 1507–1514. 15. Sheeja, D.; Tay, B.K.; Lau, S.P.;
Nung, L.N. Tribological characterisation of DLC coatings on
Co–Cr–Mo alloy for orthopaedic applications. Surf. Coat.
Technol. 2001, 146–147 , 410–416. 16. Xu, T.; Pruitt, L.
Diamond-like carbon coatings for orthopaedic applications.
J. Mater. Sci.: Mater. Med. 1999, 10, 83–90. 17. On˜ate,
J.I.; Comin, M.; Braceras, I.; Garcia, A.; Viviente, J.L.;
Brizuela, M.; Garagorri, N.; Peris, J.L.; Alava, J.I. Wear
reduction effect on UHMWPE by application of hard coatings
and ion implantation on Co– Cr alloy, as measured in a knee
wear simulator machine. Surf. Coat. Technol. 2001, 142–144,
1056–1062. 18. Lappalainen, R.; Anttila, A.; Heinonen, H.
Diamondcoated total hip replacements. Clin. Orthop. Relat.
Res. 1998, 352, 118–127. 19. Affatato, S.; Frigo, M.; Toni,
A. An invitro investigation of DLC as a femoral head
coating. J. Biomech. Mater. Res. (Appl. Biomater.) 2000,
41, 221–226. 20. Thomson, L.A.; Law, F.C.; Rushton, N.;
Franks, J. Biocompatibility of a DLC coating. Biomaterials
1991, 12, 37–40. 21. Allen, M.; Law, F.; Rushton, N. The
effects of DLC coatings on macrophages, fibroblasts and
osteoblastlike cells in vitro. Clin. Mater. 1994, 17, 1–10.
22. Lu, L.; Jones, M.W.; Wu, R.L.C. DLC as a biocompatible
material for cell culture and medical application. Biomed.
Mater. Eng. 1993, 3, 223–228. 23. Butter, R.S.; Lettington,
A.H. DLC for biomedical applications. J. Chem. Vapor Depos.
1995, 3, 182–192. 24. Steffen, H.J.; Schmidt, J.;
Gonzalez-Elipe, A. Biocompatible surfaces by immobilization
of heparin on DLC films deposited on various substrates.
Surf. Interface Anal. 2000, 29, 386–391. 25. Allen, M.;
Myer, B.; Rushton, N. In vitro and in vivo investigations
into the biocompatibility of DLC coatings for orthopedic
applications. J. Biomed. Mater. Res. 2001, 58, 319–328. 26.
Gutensohn, K.; Beyrhien, C.; Brockmann, M.; Bau, J.;
Keuhnl, P. Surface Coating of Stents with DLC: Reduction of
Thrombogeneticity. In 10th Congress on Thrombosis; Porto,
May 2000. Abstract No. 24. 27. Evans, E.A.; Hafeli, U.;
Wusinke, R.; Morrison, P.W. Mater. Res. Soc. Symp. Proc.
2000, 593, 433–438. 28. Alanazi, A.; Nojiri, C.; Noguchi,
T.; Kido, T.; Komatsu, Y.; Hirakuri, K.; Funakubo, A.;
Sakai, K.; Fukui, Y. Improved blood compatibility of DLC
coated polymeric material. ASAIO J. 2000, 440–443. 29.
Elinson, V.M.; Sleptsov, V.V.; Laymin, A.N.; Potraysay,
V.V.; Kostaychenko, L.N.; Moussina, A.D. Barrier properties
of carbon films deposited on polymer-based devices in
aggressive environments. Diamond and Related Materials
1999, 8, 2103–2108. 30. McLaughlin, J.A.; Meenan, B.;
Maguire, P.; Jamieson, N. Properties of DLC thin film
coatings on stainless steel medical guidewires. Diamond and
Related Materials 1996, 5, 486–491. 31. Schwank, M.;
Mu¨lder, U.; Hauert, R.; Rossi, R.; Volkert, M.;
Wintermantel, E. Production of a microelectrode for
intracellular measurements based on a Pt/Ir needle
insulated with amorphous hydrogenated carbon. Sens.
Actuators 1999, B56, 6–14. D
Dielectric Properties of Tissues

5. Foster, K.R.; Schwan, H.P. Dielectric properties of


tissues—a review. In Handbook of Biological Effects of
Electromagnetic Radiation, 2nd Ed.; Polk, C., Postow, E.,
Eds.; CRC Press: Boca Raton, 1995; 25–102.

6. Schwan, H.P. Electrical properties of tissue and cell


suspensions. In Advances in Biological and Medical Physics;
Lawrence, J.H., Tobias, C.A., Eds.; Academic Press: New
York, 1957; 147–209.

7. Cole, K.S.; Cole, R.H. Dispersion and absorption in


dielectrics: alternating current characteristics. J. Chem.
Phys. 1941, 9, 341–351.

8. Debye, P. Polar Molecules; Chemical Catalog Co.: New


York, 1929.

9. Oncley, J.L. In Proteins, Amino Acids, and Peptides;


Cohn, E.J., Edsall, J.T., Eds.; Reinhold: New York, 1943; .
chap 22, 543–568. 10. Schwarz, G. A theory of the low
frequency dielectric dispersion of colloidal particles in
electrolyte solution. J. Phys. Chem. 1962, 66, 2636–2642.
11. Dukhin, S.S.; Shilov, V.N. Dielectric Phenomena and the
Double Layer in Disperse Systems and Polyelectrolytes;
Greenberg, P., Ed.; John Wiley & Sons: New York, 1974. 12.
Mandel, M.; Odijk, T. Dielectric properties of
polyelectrolyte solutions. Ann. Rev. Phys. Chem. 1984, 35,
75–108. 13. Gabriel, S.; Lau, R.W.; Gabriel, C. The
dielectric properties of biological tissues: II.
Measurements in the frequency range 10Hz to 20GHz. Phys.
Med. Biol. 1996, 41, 2251–2269. 14. Raicu, V.; Sato, T.;
Raicu, G. Non-Debye dielectric relaxation in biological
structures arises from their fractal nature. Phys. Rev. E.
2001, 64, 21916–21926.
Digital Biomedical Signal Acquisition and
Processing: Basic Topics

1. Webster, J. Medical Instrumentation: Application and


Design, 3rd Ed.; Wiley: New York, NY, U.S.A., 1998.

2. Cobbold, R. Transducers for Biomedical Measurements;


Wiley & Sons: New York, U.S.A., 1998. 3. Lynn, P.; Fouerst,
W. Introductory Digital Signal Processing with Computer
Application, 2nd Ed.; Wiley & Sons: Chichester, England,
1997. 4. Oppenheim, A.; Schafer, R. Discrete-Time Signal
Processing, 2nd Ed.; Oppenheim, A., Ed.; Prentice-Hall: New
Jersey, U.S.A., 1999. 5. Haddad, R.; Parsons, T. Digital
Signal processing: Theory, Applications and Hardware;
Computer Science Press: New York, U.S.A., 1991. 6. Simson,
M. Use of signals in the terminal QRS complex to identify
patients with ventricular tachycardia after myocardial
infarction. Circulation 1981, 64 (2), 235–241. 7. Tompkins,
W. Biomedical Digital Signal Processing; Tompkins, W., Ed.;
Prentice-Hall: New Jersey, U.S.A., 1993. 8. Rangayyan, R.
Biomedical Signal Analysis: A CaseStudy Approach; Metin,
A., Ed.; IEEE press Wiley & Sons: New Jersey, U.S.A., 2002.
9. Challis, R.; Kitney, R. The design of digital filters for
biomedical signals processing. J. Biomed. Eng. 1982, 4,
267–278. 10. Bianchi, A.; Mainardi, L.; Cerutti, S. Signal
processing. In Noninvasive Electrocardiology: Clinical
Aspects of Holter Monitoring; Moss, A., Stern, S., Eds.; W.
B. Saunders Company Ltd: London, England, 1996; 11–35. 11.
Mainardi, L.; Bianchi, A.; Cerutti, S. Digital biomedical
signal acquisition and processing. In The Biomedical
Engineering Handbook, 2nd Ed.; Bronzino, J., Ed.; CRC Press
and IEEE Press: Florida, U.S.A., 2000; Vol. 53, 1–25. 12.
Gevins, A.S.; Remond, A. Handbook of Electrophysiology and
Clinical Neurophysiology; Elsevier: Amsterdam, 1987. 13.
Holm, M.; Pehrson, S.; Ingemansson, M.; Sornmo, L.;
Johansson, R.; Sandhall, L.; Sunemark, M.; Smideberg, B.;
Olsson, C.; Olsson, S.B. Non-invasive assessment of atrial
cycle length during atrial fibrilation in man: introducing,
validating and illustrating a new ECG method. Cardiovasc.
Res. 1998, 38, 69–81. 14. Kay, S. Modern Spectral
Estimation: Theory and Application; Signal Processing
Series; Oppenheim, A., Ed.; Prentice-Hall: New Jersey,
U.S.A., 1988. 15. Marple, S. Digital Spectral Analysis with
Applications; Oppenheim, A., Ed.; Signal Processing Series;
PrenticeHall: New Jersey, U.S.A., 1987. 16. Cohen, A.
Biomedical Signal Processing; CRC Press: Boca Raton, FL,
1986. 17. Zetterberg, L. Estimation of parameters for a
linear difference equation with application to EEG
analysis. Mathematical Biosci. 1969, 5, 227–275. 18.
Malliani, A. Principles of Cardiovascular Neural Regulation
in Health and Disease; Kluwer Academic Publishers:
Boston/Dordrecht/London, 2000. 19. Vanhamme, L.; Sundin,
T.; Ven Hecke, P.; Van Huffel, S. MR spectroscopy
quantitation: a review of timedomain methods. NMR Biomed.
2001, 14, 233–246. 20. Serby, H.; Yom-Tov, E.; Inbar, G.F.
An improved P300-based brain-computer interface. IEEE
Trans. Neural Syst. Rehabil. Eng. 2005, 13, 89–98. Topics
Digital Biomedical Signal Processing:
Advanced Topics

16. Jalaleddine, S.; Hutchens, C.; Strattan, R.; Coberly,


W. ECG data compression techniques—a unified approach. IEEE
Trans. Biomed. Eng. 1990, 37, 329–343.

17. Lu, Z.; Kim, D.Y.; Pearlman, W.A. Wavelet compression


of ECG signals by the set partitioning in hierarchical
trees algorithm. IEEE Trans. Biomed. Eng. 2000, 47,
849–855.

18. Khan, Y.U.; Gotman, J. Wavelet based automatic seizure


detection in intracerebral electroencephalogram. Clin.
Neurophysiol. 2003, 114, 898–908.

19. Bertrand, O.; Bohorquez, J.; Pernier, J. Time–frequency


digital filtering based on an invertible wavelet transform:
an application of evoked potentials. IEEE Trans. Biomed.
Eng. 1994, 41, 45–69.

20. Bartnik, E.A.; Blinowska, K.J.; Durka, P.J. Single


evoked potential reconstruction by means of wavelet
transform. Biolog. Cybern. 1992, 67, 175–181.

21. Causevic, E.; Morley, R.E.; Wickerhauser, M.V.;


Jacquin, A.E. Fast wavelet estimation of weak biosignals.
IEEE Trans. Biomed. Eng. 2005, 52, 1021–1032.

22. Hlawatsch, F.; Boudreaux-Bartels, G.F. Linear and


quadratic time–frequency signal representations. IEEE SP
1992, 21–67.

23. Flandrin, P. Some features of time–frequency


representations of multicomponent signals, acoustics,
speech, and signal processing. IEEE Int. Conf. ICASSP’84
1984, 9, 266–269.

24. Williams, W.J. Reduced interference distribution:


biological application and interpretations. Proc. IEEE
1996, 84, 1264–1280.

25. Mainardi, L.T.; Bianchi, A.M.; Cerutti, S.


Time–frequency and time-varying analysis for assessing the
dynamic responses of cardiovascular control. Crit. Rev.
Biomed. Eng. 2002, 30, 175–217.

26. Soderstrom, T.; Stoica, P. System Identification;


Prentice Hall International: London, U.K., 1989.
27. Morgan, N.H.; Gevins, A.S. Wigner distributions of
human event-related brain potentials. IEEE Trans. Biomed.
Eng. 1986, 33, 66–70.

28. Williams, W.J.; Shevrin, H.; Marshall, R.E. Information


modeling and analysis of event related potentials. IEEE
Trans. Biomed. Eng. 1987, 51, 737–743.

29. Zaveri, H.P.; Williams, W.J.; Iasemidis, L.D.;


Sackellares, J.C. Time–frequency representation of
electrocorticograms in temporal lobe epilepsy. IEEE Trans.
Biomed. Eng. 1992, 39, 502–509.

30. Nayak, A.; Roy, R.J.; Sharma, A. Time–frequency


spectral representation of the EEG as an aid in the
detection of depth of anesthesia. Ann. Biomed. Eng. 1994,
22, 501–513.

31. Novak, P.; Li, Z.; Novak, V.; Hatala, R. Time–frequency


mapping of the QRS complex in normal subjects and in
postmyocardial infarction patients. J. Electrocardiol.
1994, 27, 49–60.

32. Chouvarda, I.; Maglaveras, N.; Boufidou, A.; Mohlas, S.;


Louridas, G. Wigner-Ville analysis and classification of
electrocardiograms during thrombolysis. Med. Biol. Eng.
Comput. 2003, 41, 609–617.

33. Zhao, Y.; Atlas, L.E.; Marks, R.J., II. The use of
cone-shaped kernels for generalized time–frequency
representations of nonstationary signals. IEEE Trans. ASSP
1990, 38, 1084–1091. 34. Stridh, M.; Sornmo, L.; Meurling,
C.J.; Olsson, S.B. Characterization of atrial fibrillation
using the surface ECG: time-dependent spectral properties.
IEEE Trans. Biomed. Eng. 2001, 48, 19–27. 35. Novak, V.;
Novak, P.; Kus, T.; Nadeau, R. Slow cardiovascular rhythms
in tilt and syncope. J. Clin. Neurophysiol. 1995, 12,
64–71. 36. Knaflitz, M.; Bonato, P. Time–frequency method
applied to muscle fatigue assessment during dynamic
contractions. J. Electromyogr. Kinesiol. 1999, 9, 337–350.
37. Lin, Z.; Chen, J.D. Time–frequency representation of
the electrograstrogram-application of the exponential
distribution. IEEE Trans. Biomed. Eng. 1994, 41, 267–275.
38. Petrucci, E.; Mainardi, L.T.; Balian, V.; Ghiringhelli,
S. Assessment of heart rate variability changes during
dipyridamole infusion and dipyridamole-induced myocardial
ischemia: a time variant spectral approach. J. Am. Coll.
Cardiol. 1996, 28, 924–934. 39. Furlan, R.; Piazza, S.;
Dell’Orto, S.; Barbic, F.; Bianchi, A.; Mainardi, L.;
Cerutti, S.; Pagani, M.; Malliani, A.; et al. Cardiac
autonomic patterns preceding occasional vasovagal reactions
in healthy humans. Circulation 1998, 31(98), 1756–1761. 40.
Muthuswamy, J.; Thakor, N.V. Spectral analysis methods for
neurological signals. J. Neurosci. Methods 1998, 31 (83),
1–14. 41. Chen, J. A computerized data analysis system for
electrogastrogram. Comput. Biol. Med. 1992, 22, 45–57. 42.
Duche`ne, J.; Devedeux, D.; Mansour, S.; Marque, C.
Analizing uterine EMG: tracking instantaneous burst
frequency. IEEE Eng. Med. Biol. 1995, 41, 1867–1880. 43.
Nikias, C.L.; Mendel, J.M. Signal processing with
higherorder spectra. IEEE Signal Process. 1993, 64 (2),
10–37. 44. Mendel, J.M. Tutorial on higher-order statistics
(spectra) in signal processing and system theory:
theoretical results and some applications. Proc. IEEE 1991,
79 (3), 278–305. 45. Rampil, I.J. A primer for EEG signal
processing in anesthesia. Anesthesiology 1998 , 89,
980–1020. 46. Glass, P.S.A.; Bloom, M.; Kearse, L.A., Jr.;
et al. Bispectral analysis measures sedation and memory
effects of propofol, midazolam, isoflurane and anfentanil in
healthy volunteers. Anesthesiology 1997, 86, 836–847. 47.
Zhang, J.W.; Zheng, C.X.; Xie, A. Bispectrum analysis of
focal ischemic cerebral EEG signal using third-order
recursion method. IEEE Trans. Biomed. Eng. 2000, 47,
352–359. 48. Akgul, T.; Mingui, S.; Sclahassi, R.J.; et al.
Characterization of sleep spindles using higher order
statistics and spectra. IEEE Trans. Biomed. Eng. 2000, 47,
997–1009. 49. Hadjileontiadis, L.J.; Panas, S.M.
Higher-order statistics: a robust vehicle for diagnostic
assessment and characterisation of lung sounds. Technol.
Health Care 1997, 5, 359–374. 50. Goldberger, A.L.; Amaral,
L.A.N.; Hausdorff, J.M.; Ivanov, P.Ch.; Peng, C.-K.; Eugene
Stanley, H. Fractal dynamics in physiology: alterations
with disease and aging. PNAS 2002, 99 (S1), 2466–2472. 51.
Peng, C.K.; Havlin, S.; Stanley, H.E.; Goldberger, A.L.
Quantification of scaling exponents and crossover phenomena
in nonstationary heartbeat time series. Chaos 1995, 5,
82–87. 52. Pincus, S.M. Approximate entropy as a measure of
system complexity. Proc. Natl Acad. Sci. U.S.A. 1991, 88,
2297–2301. 53. Porta, A.; Baselli, G.; Liberati, D.;
Montano, N.; Cogliati, C.; Gnecchi-Ruscone, T.; Malliani,
A.; Cerutti, S. Measuring regularity by means of a
corrected conditional entropy in sympathetic outflow. Biol.
Cybern. 1998, 78, 71–78. 54. Peng, C.K.; Buldyrev, S.V.;
Goldberger, A.L.; Havlin, S.; Mantegna, R.N.; Simons, M.;
Stanley, H.E. Statistical properties of DNA sequences.
Physica A 1995, 221, 180–192. 55. Hausdorff, J.M.;
Mitchell, S.L.; Firtion, R.; Peng, C.K.; Cudkowicz, M.E.;
Wei, J.Y.; Goldberger, A.L. Altered fractal dynamics of
gait: reduced stride-interval correlations with aging and
huntington’s disease. J. Appl. Physiol. 1997, 82, 262–269.
56. Bhattacharya, J.; Edwards, J.; Mamelak, A.N.; Schuman,
E.M. Long-range temporal correlations in the spontaneous
spiking of neurons in the hippocampal–amygdala complex of
humans. Neuroscience 2005, 131, 547–555. 57. Tapanainen,
J.M.; Thomsen, P.E.; Kober, L.; TorpPedersen, C.;
Makikallio, T.H.; Still, A.M.; Lindgren, K.S.; Huikuri,
H.V. Fractal analysis of heart rate variability and
mortality after an acute myocardial infarction. Am. J.
Cardiol. 2002, 90, 347–352. 58. Ho, K.K.; Moody, G.B.;
Peng, C.K.; Mietus, J.E.; Larson, M.G.; Levy, D.;
Goldberger, A.L. Predicting survival in heart failure case
and control subjects by use of fully automated methods for
deriving nonlinear and conventional indices of heart rate
dynamics. Circulation 1997, 96, 842–848. 59. Penzel, T.;
Kantelhardt, J.W.; Grote, L.; Peter, J.H.; Bunde, A.
Comparison of detrended fluctuation analysis and spectral
analysis for heart rate variability in sleep and sleep
apnea. IEEE Trans. Biomed. Eng. 2003, 50, 1143–1151. 60.
Zhang, X.S.; Roy, R.J.; Jensen, E.W. EEG complexity as a
measure of depth of anesthesia for patients. IEEE Trans.
Biomed. Eng. 2001, 48, 1424–1433. 61. Bruhn, J.; Ropcke,
H.; Rehberg, B.; Bouillon, T.; Hoeft, A.
Electroencephalogram approximate entropy correctly
classifies the occurrence of burst suppression pattern as
increasing anesthetic drug effect. Anesthesiology 2000, 93,
981–985. 62. Koskinen, M.; Seppanen, T.; Tong, S.; Mustola,
S.; Thakor, N.V. Monotonicity of approximate entropy during
transition from awareness to unresponsiveness due to
propofol anesthetic induction. Trans. Biomed. Eng. 2006,
53, 669–675. 63. Pikkujamsa, S.M.; Makikallio, T.H.;
Sourander, L.B.; et al. Cardiac interbeat interval dynamics
from childhood to senescence: comparison of conventional
and new measures based on fractals and chaos theory.
Circulation 1999, 100, 393–399. 64. Lange, S.; Van Leeuwen,
P.; Geue, D.; Hatzmann, W.; Gro¨nemeyer, D. Influence of
gestational age, heart rate, gender and time of day on
fetal heart rate variability. Med. Biol. Eng. Comput. 2005,
43, 481–486. 65. Moorman, J.R.; Lake, D.E.; Griffin, M.P.
Heart rate characteristics monitoring for neonatal sepsis.
IEEE Trans. Biomed. Eng. 2006, 53, 126–132. 66. Vikman, S.;
Makikallio, T.H.; Yli-Mayry, S.; Pikkuja¨msa¨, S.;
Koivisto, A.-M.; Reinikainen, P.; Juhani Airaksinen, K.E.;
Huikuri, H.V. Altered complexity and correlation properties
of R–R interval dynamics before the spontaneous onset of
paroxysmal atrial fibrillation. Circulation 1999, 100,
2079–2084. 67. Hornero, R.; Aboy, M.; Abasolo, D.; McNames,
J.; Goldstein, B. Interpretation of approximate entropy:
analysis of intracranial pressure approximate entropy
during acute intracranial hypertension. IEEE Trans. Biomed.
Eng. 2005, 52, 1671–1680. 68. Akay, M.; Sekine, N.
Investigating the complexity of respiratory patterns during
recovery from severe hypoxia. J. Neural. Eng. 2004, 1,
16–20. 69. Porta, A.; Guzzetti, S.; Montano, N.; Pagani,
M.; Somers, V.; Malliani, A.; Baselli, G.; Cerutti, S.
Information domain analysis of cardiovascular variability
signals: evaluation of regularity, synchronisation and
co-ordination. Med. Biol. Eng. Comput. 2000, 38, 180–188.
70. Mainardi, L.T.; Porta, A.; Calcagnini, G.; Bartolini,
P.; Michelucci, A.; Cerutti, S. Linear and non-linear
analysis of atrial signals and local activation period
series during atrial fibrillation episodes. Med. Biol. Eng.
Comput. 2001, 39, 249–254. D
Distal Protection Devices

6. Koch, K.C.; vom Dahl, J.; Kleinhans, E.; Koch, K.C.; vom
Dahl, J.; Kleinhans, E.; Klues, H.G.; Radke, P.W.;
Ninnemann, S.; Schulz, G.; Buell, U.; Hanrath, P. Influence
of a platelet GPIIb/IIIa receptor antagonist on myocardial
hypoperfusion during rotational atherectomy as assessed by
myocardial Tc-99m sestamibi scintigraphy. J. Am. Coll.
Cardiol. 1999, 33, 998–1004.

7. Ambrosio, G.; Weissman, H.F.; Mannisi, J.A.; Becker,


L.C. Progressive impairment of regional myocardial
perfusion after initial restoration of postischemic blood
flow. Circulation 1989, 80, 1846–1861.

8. Hong, M.K.; Mehran, R.; Dangas, G.; Mintz, G.S.; Lansky,


A.J.; Pichard, A.D.; Kent, K.M.; Satler, L.F.; Stone, G.W.;
Leon, M.B. Creatine kinase-MB enzyme elevation following
successful saphenous vein graft intervention is associated
with late mortality. Circulation 1999, 100, 2400–2405.

9. Califf, R.M.; Abdelmeguid, A.E.; Kuntz, R.E.; Popma,


J.J.; Davidson, C.J.; Cohen, E.A.; Kleiman, N.S.; Mahaffey,
K.W.; Topol, E.J.; Pepine, C.J.; Lipicky, R.J.; Granger,
C.B.; Harrington, R.A.; Tardiff, B.E.; Crenshaw, B.S.;
Bauman, R.P.; Zuckerman, B.D.; Chaitman, B.R.; Bittl, J.A.;
Ohman, E.M. Myonecrosis after revascularization procedures.
J. Am. Coll. Cardiol. 1998, 31, 241–251.

10. Lincoff, A.M.; Califf, R.M.; Topol, E.J. Platelet


glycoprotein IIb/IIIa receptor blockade in coronary artery
disease. J. Am. Coll. Cardiol. 2000, 35, 1103–1115.

11. Mathew, V.; Grill, D.E.; Scott, C.G.; Grantham, J.A.;


Ting, H.H.; Garratt, K.N.; Holmes, D.R., Jr. The influence
of abciximab use on clinical outcome after aortocoronary
vein graft interventions. J. Am. Coll. Cardiol. 1999, 34,
1163–1169.

12. Mak, K.H.; Challapalli, R.; Eisenberg, M.J.; Anderson,


K.M.; Califf, R.M.; Topol, E.J. Effect of platelet
glycoprotein IIb/IIIa receptor inhibition on distal
embolization during percutaneous revascularization of
aortocoronary saphenous vein grafts. EPIC Investigators.
Evaluation of IIb/IIIa platelet receptor antagonist 7E3 in
preventing ischemic complications. Am. J. Cardiol. 1997,
80, 985–988.

13. Ellis, S.G.; Lincoff, A.M.; Miller, D.; Tcheng, J.E.;


Kleiman, N.S.; Kereiakes, D.; Califf, R.; Topol, E.J.
Reduction in complications of angioplasty with abciximab
occurs largely independently of baseline lesion morphology.
J. Am. Coll. Cardiol. 1998, 32, 1619–1623.

14. Liu, M.W.; Douglas, J.S., Jr.; Lembo, N.J.; King, S.B.,
III. Angiographic predictors of a rise in serum creatinine
kinase (distal embolization) after balloon angioplasty of
saphenous vein coronary artery bypass grafts. Am. J.
Cardiol. 1993, 72, 514–517.

15. Motwani, J.G.; Topol, E.J. Aortocoronary saphenous vein


graft disease: Pathogenesis, predisposition, and
prevention. Circulation 1998, 97 (9), 916–931.

16. Roubin, G.S.; New, G.; Iyer, S.S.; Vitek, J.J.;


Al-Mubarak, N.; Liu, M.W.; Yadav, J.; Gomez, C.; Kuntz,
R.E. Immediate and late clinical outcomes of carotid artery
stenting in patients with symptomatic and asymptomatic
carotid artery stenosis: A 5-year prospective analysis.
Circulation 2001, 103, 532–537. 17. Jordan, W.D.;
Voellingeer, D.C.; Doblar, D.D.; Plyushcheva, N.P.; Fisher,
W.S.; McDowell, H.A. Microemboli detected by transcranial
Doppler monitoring in patients during carotid angioplasty
versus carotid endarterectomy. Cardiovasc. Surg. 1999, 7,
33–38. 18. Rapp, J.H.; Pan, X.M.; Sharp, F.R.; Shah, D.M.;
Wille, G.A.; Velez, P.M.; Troyer, A.; Higashida, R.T.;
Saloner, D. Atheroemboli to the brain: Size threshold for
causing acute neuronal cell death. J. Vasc. Surg. 2000, 32,
68–76. 19. Ackerstaff, R.G.; Moons, K.G.; van de Vlasakker,
C.J.; Moll, F.L.; Vermeulen, F.E.; Algra, A.; Spencer, M.P.
Association of intraoperative transcranial doppler
monitoring variables with stroke from carotid
endarterectomy. Stroke 2000, 31, 1817–1823. 20. Grube, E.;
Schofer, J.; Webb, J.; Schuler, G.; Colombo, A.; Sievert,
H.; Gerckens, U.; Stone, G.W.; for the SAFE Trial Study
Group. Evaluation of a balloon occlusion and aspiration
system for protection from distal embolization during
stenting in saphenous vein grafts. Am. J. Cardiol. 2002, 89
(8), 941–945. 21. Baim, D.S.; Wahr; George, B.; Leon, M.B.;
Greenberg, J.; Cutlip, D.E.; Kaya, U.; Popma, J.J.; Ho,
K.K.L.; Kuntz, R.E.; on behalf of the SAFER Trial
Investigators. Randomized trial of a distal embolic
protection device during percutaneous intervention of
saphenous vein aorto-coronary bypass grafts. Circulation
2002, 105, 1285–1290. 22. Carrozza, J. In Early Clinical
Trial Results with the Triactiv System, 13th Annual
Symposium on Transcatheter Cardiovascular Therapeutics,
Washington, DC, September 11–16, 2001. 23. Grube, E.;
Gerckens, U.; Yeung, A.C.; Rowold, S.; Kirchhof, N.;
Sedgewick, J.; Yadav, J.S.; Stertzer, S. Prevention of
distal embolization during coronary angioplasty in
saphenous vein grafts and native vessels using porous filter
protection. Circulation 2001, 104 (20), 2436–2441. 24.
Al-Mubarak, N.; Colombo, A.; Gaines, P.A.; Iyer, S.S.;
Corvaja, N.; Cleveland, T.J.; Macdonald, S.; Brennan, C.;
Vitek, J.J. Multicenter evaluation of carotid artery
stenting with a filter protection system. J. Am. Coll.
Cardiol. 2002, 39 (5), 841–846. 25. Grube, E.; Hauptman,
K.; Reifart, N.; Tully, G.; McColl, M. The FilterWire EX
embolic protection device for use during percutaneous
coronary artery and saphenous vein graft interventions. Am.
J. Cardiol. 2001, 88 (Suppl. 5A), 1G–147G. 26. Ohki, T.;
Parodi, J.; Veith, F.J.; Bates, M.; Bade, M.; Chang, D.;
Mehta, M.; Rabin, J.; Goldstein, K.; Harvey, J.; Lipsitz,
E. Efficacy of a proximal occlusion catheter with reversal
of flow in the prevention of embolic events during carotid
artery stenting: An experimental analysis. J. Vasc. Surg.
2001, 33, 504–509. 27. Parodi, J.C.; La Mura, R.; Ferreira,
L.M.; Mendez, M.V.; Cersosimo, H.; Schonholz, C.; Garelli,
G. Initial evaluation of carotid angioplasty and stenting
with three different cerebral protection devices. J. Vasc.
Surg. 2000, 32, 1127–1136. D
Drug Delivery, Controlled

Table 1 Controlled-release products in market

Technology Product/drug Application Organization

Liposome-based formulations Evacet (Doxorubicin) Breast


cancer and other cancers Elan with acquisition of Liposome
Inc.

One-yearly drug implant Leuprolide (Viadur) Prostate cancer


ALZA

Intradose injectable gel Cisplatin/ epinephrine Recurrent


or refractory head and neck cancer Matrix Pharmaceuticals
Inc.

Biodegradable microsphere for

sustained release Medisorb Delivery of peptides and small


molecules Alkermes Inc.

Sustained-release injections ProLease Delivery of fragile


proteins and small bioactive molecules Alkermes Inc.

Time release oral drug release Pulsincap Drug release at


predetermined time or location in the GI-tract RP scherer
corporation

Oral controlled-release system

to control the release of a

specific drug Geomatrix Predetermined therapeutic objective


for a drug SkyePharma 4. Hejazi, R.; Amiji, M.
Chitosan-based gastrointestinal delivery systems. J.
Control. Release 2003, 89 (2), 151–165. 5. Duvvuri, S.;
Majumdar, S.; Mitra, A.K. Drug delivery to the retina:
Challenges and opportunities. Expert Opin. Biol. Ther.
2003, 3 (1), 45–56. 6. Uhrich, K.E.; Cannizzaro, S.M.;
Langer, R.S.; Shakesheff, K.M. Polymeric systems for
controlled drug release. Chem. Rev. 1999, 99, 3181–3198. 7.
Ravi Kumar, M.N.V.; Kumar, N.; Domb, A.J.; Arora, M.
Pharmaceutical polymeric controlled drug delivery systems.
Adv. Polym. Sci. 2002, 160, 45–117. 8. Luo, Y.; Prestwich,
G.D. Cancer-targeted polymeric drugs. Curr. Cancer Drug
Targets 2002, 2 (3), 209–226. 9. Qiu, Y.; Park, K.
Environment-sensitive hydrogels for drug delivery. Adv.
Drug Deliv. Rev. 2001, 53 (3), 321–339. 10. Panyam, J.;
Labhasetwar, V. Biodegradable nanoparticles for drug and
gene delivery to cells and tissue. Adv. Drug Deliv. Rev.
2003, 55 (3), 329–347. 11. Edlund, E.; Albertsson, A.-C.
Degradable polymer microspheres for controlled drug
delivery. Adv. Polym. Sci. 2002, 157, 67–112. 12.
Panchagnula, R. Transdermal delivery of drugs. Indian J.
Pharmacol. 1997, 29 (3), 140–156. 13. Ritschel, W.A.
Biopharmaceutic and pharmacokinetic aspects in the design
of controlled-release peroral drug delivery systems. Drug
Dev. Ind. Pharm. 1989, 15 (6–7), 1073–1103. 14. Rubinstein,
A.; Robinson, J.R. Controlled drug delivery. Prog. Clin.
Biochem. Med. 1987, 4, 71–107. 15. Vyas, S.P.; Khar, R.K.
Targeted and Controlled Drug Delivery: Novel Carrier
Systems; CBS: New Delhi, 2002. 16. Witt, C.; Kissel, T.
Morphological characterization of microspheres, films and
implants prepared from poly(lactide-co-glycolide) and ABA
triblock copolymers: Is the erosion controlled by
degradation, swelling or diffusion? Eur. J. Pharm.
Biopharm. 2001, 51 (3), 171–181. 17. Rothen-Weinhold, A.;
Gurny, R.; Orsolini, P.; Heimgartner, F. Implants for
controlled release of pharmaceutically active principles
and method for making same. PCT Int. Appl. 1998. 26 pp. WO
9855101. 18. Ravi Kumar, M.N.V. Nano and microparticles as
controlled drug delivery devices. J. Pharm. Pharm. Sci.
2000, 3 (2), 234–258. 19. Jain, R.A. The manufacturing
techniques of various drug loaded biodegradable
poly(lactide-co-glycolide) (PLGA) devices. Biomaterials
2000, 21 , 2475–2490. 20. Torchilin, V.P. Polymer-coated
long-circulating microparticulate pharmaceuticals. J.
Microencapsul. 1998, 15 (1), 1–19. 21. Palmieri, G.F.;
Bonacucina, G.; Di Martino, P.; Martelli, S. Spray-drying
as a method for microparticulate controlled release systems
preparation: Advantages and limits. I. Water-soluble drugs.
Drug Dev. Ind. Pharm. 2001, 27 (3), 195–204. 22. Johnson,
O.L.; Jaworowicz, W.; Cleland, J.L.; Bailey, L.; Charnis,
M.; Duenas, E.; Wu, C.; Shepard, D.; Magil, S.; Last, T.;
Jones, A.J.; Putney, S.D. The stabilization and
encapsulation of human growth hormone into biodegradable
microspheres. Pharm. Res. 1997, 14 (6), 730–735. 23. Jeong,
B.; Bae, Y.H.; Kim, S.W. In situ gelation of PEG-PLGA-PEG
triblock copolymer aqueous solutions and degradation
thereof. J. Biomed. Mater. Res. 2000, 50 (2), 171–177. 24.
Chenite, A.; Chaput, C.; Wang, D.; Combes, C.; Buschmann,
M.D.; Hoemann, C.D.; Leroux, J.C.; Atkinson, B.L.; Binette,
F.; Selmani, A. Novel injectable neutral solutions of
chitosan form biodegradable gels in situ. Biomaterials
2000, 21 (21), 2155–2161. 25. Ruel-Gariepy, E.; Chenite,
A.; Chaput, C.; Guirguis, S.; Leroux, J.-C.
Characterization of thermosensitive chitosan gels for the
sustained delivery of drugs. Int. J. Pharm. 2000, 203
(1–2), 89–98. 26. Chenite, A.; Buschmann, M.; Wang, D.;
Chaput, C.; Kandani, N. Rheological characterization of
thermogelling chitosan/glycerol-phosphate solutions.
Carbohydr. Polym. 2001, 46 (1), 39–47. 27. Brodbeck, K.J.;
Pushpala, S.; McHugh, A.J. Sustained release of human
growth hormone from PLGA solution depots. Pharm. Res. 1999,
16 (12), 1825–1829. 28. Agu, R.U.; Ugwoke, M.I.; Armand,
M.; Kinget, R.; Verbeke, N. Respir. Res. 2001, 2, 198–209.
29. Singh, R.; Matharu, P.; Lalla, J.K. A
diffusioncontrolled drug delivery system for theophylline.
Drug Dev. Ind. Pharm. 1994, 20 (7), 1225–1238. 30. Heller,
J. Controlled drug release from monolithic systems. Fidia
Res. Ser. 1987, 11 (Ophthalmic Drug Delivery), 179–189. 31.
Laurencin, C.T.; Koh, H.J.; Neenan, T.X.; Allcock, H.R.;
Langer, R. Controlled release using a new bioerodible
polyphosphazene matrix system. J. Biomed. Mater. Res. 1987,
21 (10), 1231–1246. 32. Zuleger, S.; Lippold, B.C. Polymer
particle erosion controlling drug release. I. Factors
influencing drug release and characterization of the release
mechanism. Int. J. Pharm. 2001, 217 (1–2), 139–152. 33.
Heller, J.; Domb, A.J.; Kumar, N. Polyanhydrides and
poly(ortho esters). Adv. Drug Deliv. Rev. 2002, 54 (7),
887–888. 34. Shenderova, A.; Burke, T.G.; Schwendeman, S.P.
The acidic microclimate in poly(lactide-co-glycolide)
microspheres stabilizes camptothecins. Pharm. Res. 1999, 16
(2), 241–248. 35. Siepmann, J.; Peppas, N.A. Modeling of
drug release from delivery systems based on hydroxypropyl
methylcellulose (HPMC). Adv. Drug Deliv. Rev. 2001, 48
(2–3), 139–157. 36. Ritger, P.L.; Peppas, N.A. A simple
equation for description of solute release. II. Fickian and
anomalous release from swellable devices. J. Control.
Release 1987, 5 (1), 37–42. 37. Batycky, R.P.; Hanes, J.;
Langer, R.; Edwards, D.A. A theoretical model of erosion
and macromolecular drug release from biodegrading
microspheres. J. Pharm. Sci. 1997, 86 (12), 1464–1477. 38.
Singh, P.; Sihorkar, V.; Mishra, V.; Saravanababu, B.;
Venkatesan, N.; Vyas, S.P. Osmotic pumps. From present view
to newer perspectives in pharmaceutical industry. East.
Pharm. 1999, 42 (503), 39–46. D

39. Ehrenfreund-Kleinman, T.; Azzam, T.; Falk, R.;


Polacheck, I.; Golenser, J.; Domb, A.J. Synthesis and
characterization of novel water soluble amphotericin
B-arabinogalactan conjugates. Biomaterials 2002, 23 (5),
1327–1335.

40. Irwin, W.J.; Belaid, K.A. Drug-delivery by


ion-exchange. Part I: Ester prodrugs of propranolol. Drug
Dev. Ind. Pharm. 1987, 13 (9–11), 1231–2017.

41. Mehvar, R. Dextrans for targeted and sustained delivery


of therapeutic and imaging agents. J. Control. Release
2000, 69 (1), 1–25.

42. Kost, J.; Noecker, R.; Kunica, E.; Langer, R.


Magnetically controlled release systems: Effect of polymer
composition. J. Biomed. Mater. Res. 1985, 19 (8), 935–940.

43. Saslawski, O.; Couvrer, P.; Peppas, N. Proceedings of


15th International Symposium on Controlled Release of
Bioactive Materials. Basel, Heller, J., Harris, F.,
Lohmann, H., Merkle, H., Robinson, J., Eds.; 1988, 26.

44. Edelman, E.R.; Brown, L.; Taylor, J.; Langer, R. In


vitro and in vivo kinetics of regulated drug release from
polymer matrixes by oscillating magnetic fields. J. Biomed.
Mater. Res. 1987, 21 (3), 339–353.

45. Suzuki, A.; Tanaka, T. Photoinduced phase transition of


gels. Nature 1990, 346, 345–347.

46. Kost, J.; Leong, K.; Langer, R. Ultrasound-enhanced


polymer degradation and release of incorporated substances.
Proc. Natl. Acad. Sci. U. S. A. 1989, 86 (20), 7663–7666.

47. Miyazaki, S.; Yokouchi, C.; Takada, M. External control


of drug release: Controlled release of insulin from a
hydrophilic polymer implant by ultrasound irradiation in
diabetic rats. J. Pharm. Pharmacol. 1988, 40 (10), 716–717.

48. Grimshaw, P.E.; Grodzinsky, A.J.; Yarmush, M.L.;


Yarmush, D.M. Dynamic membranes for protein transport:
Chemical and electrical control. Chem. Eng. Sci. 1989, 44
(4), 827–840.

49. Kwon, I.C.; Bae, Y.H.; Kim, S.W. Electrically erodible


polymer gel for controlled release of drugs. Nature 1991,
354 (6351), 291–293.

50. Cohen, S.; Yoshioka, T.; Lucarelli, M.; Hwang, L.H.;


Langer, R. Controlled delivery systems for proteins based
on poly(lactic/glycolic acid) microspheres. Pharm. Res.
1991, 8 (6), 713–720.

51. Okada, H. Oneand three-month release injectable


microspheres of the LH-RH superagonist leuprorelin acetate.
Adv. Drug Deliv. Rev. 1997, 28, 43–70. 52. Davies, P.H.;
Stewart, S.E.; Lancranjan, L.; Sheppard, M.C.; Stewart,
P.M. Long-term therapy with longacting octreotide
(Sandostatin–LAR) for the management of acromegaly. Clin.
Endocrinol. 1998, 48 (3), 311–316. 53. Schrier, J.A.;
DeLuca, P.P. Recombinant human bone morphogenetic protein-2
binding and incorporation in PLGA microsphere delivery
systems. Pharm. Dev. Technol. 1999 , 4 (4), 611–621. 54.
Pistel, K.F.; Bittner, B.; Koll, H.; Winter, G.; Kissel, T.
Biodegradable recombinant human erythropoietin loaded
microspheres prepared from linear and starbranched block
copolymers: Influence of encapsulation technique and polymer
composition on particle characteristics. J. Control.
Release 1999, 59 (3), 309–325. 55. Lam, X.M.; Duenas, E.T.;
Cleland, J.L. Encapsulation and stabilization of nerve
growth factor into poly(lacticco-glycolic) acid
microspheres. J. Pharm. Sci. 2001, 90 (9), 1356–1365. 56.
Regar, E.; Sianos, G.; Serruys, P.W. Stent development and
local drug delivery. Br. Med. Bull. 2001, 59, 227–248. 57.
Lewis, A.L.; Cumming, Z.L.; Goreish, H.H.; Kirkwood, L.C.;
Tolhurst, L.A.; Stratford, P.W. Crosslinkable coatings from
phosphorylcholine-based polymers. Biomaterials 2001, 22
(2), 99–111. 58. van der Giessen, W.J.; Lincoff, A.M.;
Schwartz, R.S.; van Beusekom, H.M.; Serruys, P.W.; Holmes,
D.R., Jr.; Ellis, S.G.; Topol, E.J. Marked inflammatory
sequelae to implantation of biodegradable and
nonbiodegradable polymers in porcine coronary arteries.
Circulation 1996, 94 (7), 1690–1697. 59. Hietala, E.-M.;
Salminen, U.-S.; Stahls, A.; Valimaa, T.; Maasilta, P.;
Tormala, P.; Nieminen, M.S.; Harjula, A.L.J. Biodegradation
of the copolymeric polylactide stent: Long-term follow-up
in a rabbit aorta model. J. Vasc. Res. 2001, 38 (4),
361–369. 60. Harper, E.; Dang, W.; Lapidus, R.G.; Garver,
R.I., Jr. Enhanced efficacy of a novel controlled release
paclitaxel formulation (PACLIMER delivery system) for
local-regional therapy of lung cancer tumor nodules in
mice. Clin. Cancer Res. 1999, 5 (12), 4242– 4248. 61.
Paavola, A.; Tarkkila, P.; Xu, M.; Wahlstrom, T.; Yliruusi,
J.; Rosenberg, P. Controlled release gel of ibuprofen and
lidocaine in epidural use—Analgesia and systemic absorption
in pigs. Pharm. Res. 1998, 15 (3), 482–487.
Elastin

30. Villani, V.; Tamburro, A. Conformational chaos of an


elastin-related peptide in aqueous solution. Ann. N.Y.
Acad. Sci. 1997, 879, 284–287.

31. Urry, D.W.; Luan, C.H.; Peng, S.Q. Molecular Biophysics


of Elastin Structure, Function and Pathology. In Symposium
on the Molecular Biology and Pathology of Elastic Tissues;
Chadwick, D.J.P.S., Ed.; John Wiley and Sons Ltd.: Windsor
Golf Club, Nairobi, Kenya, 1994; Vol. 192, 4–22.

32. Urry, D.W. Physical chemistry of biological free energy


transduction as demonstrated by elastic proteinbased
polymers. J. Phys. Chem., B 1997, 101, 11007– 11028.

33. Urry, D.W. Molecular machines: How motion and other


functions of living organisms can result from reversible
chemical changes. Angew. Chem., Int. Ed. Engl. 1993, 32,
819–841.

34. Bressan, G.M.; Castellani, I.; Giro, G.M.; et al.


Banded fibers in tropoelastin coacervates at physiologic
temperatures. J. Ultrastruct. Res. 1983, 82, 335–340.

35. Long, M.M.; Rapka, R.S.; D.V.; et al. Spectroscopic and


electron micrographic studies on the repeat tetrapeptide of
tropoelastin: Val-Pro-Gly-Gly. Arch. Biochem. Biophys.
1980, 201, 445–452.

36. Pasquali-Ronchetti, I.; Fornieri, C.; Baccarani-Contri,


M.; Volpin, D. The ultrastructure of elastin revealed by
freeze-fracture electron microscopy. Micron 1979, 10,
89–99.

37. Sage, H.; Gray, W.R. Studies on the evolution of


elastin-II. Histology. Comp. Biochem. Physiol. 1980, 66B,
13–22.

38. Pasquali-Ronchetti, I.B.-C.M.; Young, R.D.; Vogel, A.;


Steinmann, B.; Royce, P.M. Ultrastructural analysis of skin
and aorta from a patient with Menkes disease. Exp. Mol.
Pathol. 1994, 61, 36–57.

39. Quaglino, D.; Fornieri, C.; Nanney, L.; Davidson, J.


Extracellular matrix modifications in rat tissues of
different ages. Correlations between elastin and collagen
type I mRNA expression and lysyl oxidase activity. Matrix
1993, 13, 481–490.
40. Vitellaro-Zuccarello, L.; Cappelletti, S.; Dal
PozzoRossi, V.; Sari-Gorla, M. Sterological analysis of
collagen and elastic fibers in the normal human dermis:
Variability with age, sex and body region. Anat. Rec. 1994,
238, 153–162.

41. Pasquali-Ronchetti, I.; Fornieri, C.; Baccarani-Contri,


M.; Quaglino, D. Ultrastructure of Elastin. In Symposium on
the Molecular Biology and Pathology of Elastic Tissues;
Chadwick, D.J.P.S., Ed.; John Wiley and Sons Ltd.: Windsor
Golf Club, Nairobi, Kenya, 1994; Vol. 192, 31–42. 42.
Nidhi, N.; Chilkoti, A. Interfacial phase transition of an
environmentally responsive elastin biopolymer adsorbed on
functionalized gold nanoparticles studied by colloidal
surface plasmon resonance. J. Am. Chem. Soc. 2001, 123,
8197–8202. 43. Rama Rao, G.; Balamurugan, S.; Meyer, D.;
Hybrid bioinorganic smart membranes that incorporate
protein based molecular switches. Langmuir 2002, 18, 1819–
1824. 44. Betre, H.; Setton, L.; Meyer, D.; Chilkoti, A.
Characterization of a genetically engineered elastin-like
polypeptide for cartilaginous tissue repair.
Biomacromolecules 2002, 3, 910–916. 45. Hyun, J.; Chilkoti,
A. Micropatterning biological molecules on a polymer
surface using elastomeric microwells. J. Am. Chem. Soc.
2001, 123, 6934–6944. 46. Lillie, M.; Gosline, J. The
viscoelastic basis for the tensile strength of elastin.
Int. J. Biol. Macromol. 2002, 30 (2), 119–127. 47. Urry,
D.; Parker, T.; Reid, M.; Gowda, D. J. Bioact. Compat.
Polym. 1991, 6, 263–282. 48. McMillan, R.; Lee, T.;
Conticello, V. Rapid assembly of synthetic genes encoding
protein polymers. Macromolecules 1999, 32 (11), 3643–3648.
49. McPherson, D.T.X.J.; Urry, D.W. Product purification by
reversible phase transition following Escherichia coli
expression of genes encoding up to 251 repeats of the
elastomeric pentapeptide GVGVP. Protein Expr. Purif. 1996,
7, 51–57. 50. Nagapudi, K.B.W.; Huang, L.; McMillan, R.A.;
Conticello, V.P.; Chaikof, E.L. Photomediated solid-state
crosslinking of an elastin-mimetic recombinant protein
polymer. Macromolecules 2002, 35, 1730–1737. 51. Huang, L.;
McMillan, R.; Apkarian, R.; et al. Generation of synthetic
elastin-mimetic small diameter fibers and fiber networks.
Macromolecules 2000, 33, 2989–2997. 52. Panitch, A.;
Yamaoka, T.; Fournier, M.; Design and biosynthesis of
elastin-like artificial extracellular matrix proteins
containing periodically spaced fibronectin CS5 domains.
Macromolecules 1999, 32 (5), 1701–1703. 53. Nicol, A.;
Gowda, D.; Parker, T.; Urry, D. Elastomeric
polytetrapeptide matrices: Hydrophobicity dependence of
cell attachment from adhesive (GGIP)n to nonadhesive
(GGAP)n even in serum. J. Biomed. Mater. Res. 1993, 27,
801–810. 54. Megeed, Z.; Cappello, J.; Ghandehari, H.
Genetically engineered silk-elastinlike protein polymers
for controlled drug delivery. Adv. Drug Deliv. Rev. 2002,
54 (8), 1075–1092. 55. Wright, E.; Conticello, V.
Self-assembly of block copolymers derived from
elastin-mimetic polypeptide sequences. Adv. Drug Deliv.
Rev. 2002, 54, 1057–1074.
Elastomers, Biodegradable

1. Cai, Q.; Bei, J.Z.; Wang, S.G. Synthesis and


characterization of polycaprolactone
(B)-poly(lactide-co-glycolide) (A) ABA block copolymer.
Polym. Adv. Technol. 2000, 11 (4), 159–166.

2. Kwon, I.K.; Park, K.D.; Choi, S.W.; Lee, S.H.; Lee,


E.B.; Na, J.S.; Kim, S.H.; Kim, Y.H. Fibroblast culture on
surface-modified poly(glycolide-co-epsilon-caprolactone)
scaffold for soft tissue regeneration. J. Biomater. Sci.,
Polym. Ed. 2001, 12 (10), 1147–1160.

3. Cai, Q.; Bei, J.Z.; Wang, S.G. Synthesis and degradation


of a tri-component copolymer derived from glycolide,
L-lactide, and epsilon-caprolactone. J. Biomater. Sci.,
Polym. Ed. 2000, 11 (3), 273–288.

4. Mohammadi-Rovshandeh, J.; Farnia, S.M.F.; Sarbolouki,


M.N. Synthesis and characterization of novel ABA triblock
copolymers from L-lactide, glycolide, and PEG. J. Appl.
Polym. Sci. 1999, 74 (8), 2004–2009.

5. Hiki, S.; Miyamoto, M.; Kimura, Y. Synthesis and


characterization of hydroxy-terminated
[RS]-poly(3-hydroxybutyrate) and its utilization to block
copolymerization with L-lactide to obtain a biodegradable
thermoplastic elastomer. Polymer 2000, 41 (20), 7369–7379.

6. Wang, Y.D.; Ameer, G.A.; Sheppard, B.J.; Langer, R. A


tough biodegradable elastomer. Nat. Biotechnol. 2002, 20
(6), 602–606.

7. Storey, R.F.; Herring, K.R.; Hoffman, D.C.


Hydroxyterminated poly(e-caprolactone-co-d-valerolactone)
oligomers: Synthesis, characterization, and polyurethane
network formation. J. Polym. Sci., Polym. Chem. Edn. 1991,
29, 1759–1777. 8. Storey, R.F.; Hickey, T.P. Degradable
polyurethane networks based on D, L-lactide, glycolide,
e-caprolactone, and trimethylene carbonate homopolyester
and copolyester triols. Polymer 1994, 35 (4), 830–838. 9.
Albertsson, A.C.; Varma, I.K. Aliphatic polyesters:
Synthesis, properties and applications. Adv. Polym. Sci.
2002, 157, 1–40. 10. Sudesh, K.; Doi, Y. Molecular design
and biosynthesis of biodegradable polyesters. Polym. Adv.
Technol. 2000, 11, 865–872. 11. Abe, H.; Matsubara, I.;
Doi, Y.; Hori, Y.; Yamaguchi, A. Physical-properties and
enzymatic degradability of poly(3-hydroxybutyrate)
stereoisomers with different stereoregularities.
Macromolecules 1994, 27 (21), 6018–6025. 12. Dobrzynski, P.
Synthesis of biodegradable copolymers with low-toxicity
zirconium compounds. II. Copolymerization of glycolide with
e-caprolactone initiated by zirconium(IV) acetylacetonate
and zirconium(IV) chloride. J. Polym. Sci., Polym. Chem.
2002, 40 (10), 1379–1394. 13. Hiljanen-Vainio, M.P.; Orava,
P.A.; Seppa¨la¨, J.V. Properties of e-caprolactone/
DL-lactide (e-CL/DL/LA) copolymers with a minor e-CL
content. J. Biomed. Mater. Res. 1997, 34, 39–46. 14.
Lofgren, A.; Albertsson, A.C.; Dubois, P.; Jerome, R.;
Teyssie, P. Synthesis and characterization of biodegradable
homopolymers and blockpolymers and block-copolymers and
block-copolymers based on 1,5-Dioxepan-2-one.
Macromolecules 1994, 27 (20), 5556–5562. 15. Stridsberg,
K.; Albertsson, A.C. Controlled ring-opening polymerization
of L-lactide and 1,5-dioxepan-2-one forming a triblock
copolymer. J. Polym. Sci., Polym. Chem. 2000, 38 (10),
1774–1784. 16. Pego, A.P.; Poot, A.A.; Grijpma, D.W.;
Feijen, J. Copolymers of trimethylene carbonate and
e-caprolactone for porous nerve guides: Synthesis and
properties. J. Biomater. Sci., Polym. Ed. 2001, 12 (1),
35–53. 17. Kim, J.H.; Lee, J.H. Preparation and
chain-extension of P(LLA-b-TMC-b-LLA) triblock copolymers
and their elastomeric properties. Macromol. Res. 2002, 10
(2), 54–59. 18. Lin, S. Lactide/Caprolactone Polymer,
Method of Making the Same, Composites Thereof, and
Prostheses Produced Therefrom. US Patent 4,643,734,
February 17, 1987. 19. Lofgren, A.; Renstad, R.;
Albertsson, A.C. Synthesis and characterization of a new
degradable thermoplastic elastomer based on
1,5-dioxepan-2-one and e-caprolactone. J. Appl. Polym. Sci.
1995, 55 (11), 1589–1600. 20. Bruin, P.; Veenstra, G.J.;
Nijenhuis, A.J.; Pennings, A.J. Design and synthesis of
biodegradable poly(esterurethane) elastomer networks
composed of non-toxic building blocks. Makromol. Chem.,
Rapid Commun. 1988, 9, 589–594. 21. Bruin, P.; Smedinga,
J.; Pennings, A.J. Biodegradable lysine diisocyanate-based
poly(glycolide-co-e-caprolactone)-urethane network in
artificial skin. Biomaterials 1990, 11, 291–295. 22. Storey,
R.F.; Wiggins, J.S.; Puckett, A.D. Hydrolyzable
poly(ester-urethane) networks from L-lysine diisocyanate
and D,L-lactide/e-caprolactone homoand copolyester triols.
J. Polym. Sci., Polym. Chem. 1994, 32, 2345–2363. 23. Guan,
J.; Sacks, M.S.; Beckman, E.J.; Wagner, W.R. Synthesis,
characterization, and cytocompatibility of elastomeric,
biodegradable poly(ester-urethane)ureas based on
poly(caprolactone) and putrescine. J. Biomed. Mater. Res.
2002, 61, 493–503. 24. Spaans, C.J.; de Groot, J.H.;
Belgraver, V.W.; Pennings, A.J. A new biomedical
polyurethane with a high modulus based on
1,4-butanediisocyanate and e-caprolactone. J. Mater. Sci.,
Mater. Med. 1998, 9, 675–678. 25. Spaans, C.J.; de Groot,
J.H.; Dekens, F.G.; Pennings, A.J. High molecular weight
polyurethanes and a polyurethane urea based on
1,4-butanediisocyanate. Polym. Bull. 1998, 41, 131–138. 26.
Kylma¨, J.; Seppa¨la¨, J.V. Synthesis and characterization
of a biodegradable thermoplastic poly(ester-urethane)
elastomer. Macromolecules 1997, 30, 2876–2882. 27.
Lendlein, A.; Neuenschwander, P.; Suter, U.W.
Tissuecompatible multiblock copolymers for medical
applications, controllable in degradation rate and
mechanical properties. Macromol. Chem. Phys. 1998, 199,
2785–2796. 28. Saad, G.R.; Lee, Y.J.; Seliger, H. Synthesis
and characterization of biodegradable poly(ester-urethanes)
based on bacterial poly(R-3-hydroxybutyrate). J. Appl.
Polym. Sci. 2002, 83, 703–718. 29. Gorna, K.; Polowinski,
S.; Gogolewski, S. Synthesis and characterization of
biodegradable poly(e-caprolactone urethane)s. I. Effect of
the polyol molecular weight, catalyst, and chain extender
on the molecular and physical characteristics. J. Polym.
Sci., Part A, Polym. Chem. 2002, 40, 156–170. 30. Gorna,
K.; Gogolewski, S. Biodegradable polyurethanes for
implants. II. In vitro degradation and calcification of
materials from poly(e-caprolactone)poly(ethylene oxide)
diols and various chain extenders. J. Biomed. Mater. Res.
2002, 60, 592–606. 31. de Groot, J.H.; de Vrijer, R.;
Wildeboer, B.S.; Spaans, C.S.; Pennings, A.J. New
biomedical polyurethane ureas with high tear strengths.
Polym. Bull. 1997, 38, 211–218. 32. Lamba, N.M.K.;
Woodhouse, K.A.; Cooper, S.L. Polyurethanes in Biomedical
Applications; CRC Press: New York, 1998. 33. Schoental, R.
Carcinogenic and chronic effects of 4,4diamino-diphenyl
methane, an epoxy resin hardener. Nature 1968, 219, 1968.
34. National Toxicology Program Technical Report on the
Carcinogenesis Bioassay of 4,4u-Methylenedianiline
Dihydrochloride, NTP-81-143;NIH Publication No. 82-2504,
National Institutes of Health: Bethesda, MD, 1982. 35.
Cohn, D.; Stern, T.; Gonza´lez, M.F.; Epstein, J.
Biodegradable poly(ethylene oxide)/poly(e-caprolactone)
multiblock copolymers. J. Biomed. Mater. Res. 2002, 59,
273–281. 36. de Groot, J.H.; Spaans, C.J.; Dekens, F.G.;
Pennings, A.J. On the role of aminolysis and
transesterification in the synthesis of e-caprolactone and
L-lactide based polyurethanes. Polym. Bull. 1998, 41,
299–306. 37. Skarja, G.A.; Woodhouse, K.A. In vitro
degradation and erosion of degradable, segmented
polyurethanes containing an amino acid-based chain
extender. J. Biomater. Sci., Polym. Ed. 2001, 12 (8),
851–873. 38. Saad, B.; Hirt, T.D.; Welti, M.; Uhlschmid,
G.K.; Neuenschwander, P.; Suter, U.W. Development of
degradable polyesterurethanes for medical applications: In
vitro and in vivo evaluations. J. Biomed. Mater. Res. 1997,
36, 65–74. 39. Lendlein, A.; Colussi, M.; Neuenschwander,
P.; Suter, U.W. Hydrolytic degradation of phase-segregated
multiblock copoly(ester urethane)s containing weak links.
Macromol. Chem. Phys. 2001, 202, 2702–2711. 40. Lendlein,
A.; Langer, R. Biodegradable, elastic shapememory polymers
for potential biomedical applications. Science 2002, 296,
1673–1676. 41. Skarja, G.A.; Woodhouse, K.A. Synthesis and
characterization of degradable polyurethane elastomers
containing an amino acid-based chain extender. J. Biomater.
Sci., Polym. Ed. 1998, 9 (3), 271–295. 42. Skarja, G.A.;
Woodhouse, K.A. Structure-property relationships of
degradable polyurethane elastomers containing an amino
acid-based chain extender. J. Appl. Polym. Sci. 2000, 75
(12), 1522–1534. 43. Agrawal, C.M.; Ray, R.B. Biodegradable
polymeric scaffolds for musculoskeletal tissue engineering.
J. Biomed. Mater. Res. 2001, 55, 141–150. 44. van Tienen,
T.G.; Heijkants, R.G.J.C.; Buma, P.; de Groot, J.H.;
Pennings, A.J.; Veth, R.P.H. Tissue ingrowth and
degradation of two biodegradable porous polymers with
different porosities and pore sizes. Biomaterials 2002, 23
(8), 1731–1738. 45. Borkenhagen, M.; Stoll, R.C.;
Neuenschwander, P.; Suter, U.W.; Aebischer, P. In vivo
performance of a new biodegradable polyeste urethane system
used as a nerve guidance channel. Biomaterials 1998, 19,
2155–2165. 46. Butler, D.L.; Goldstein, S.A.; Guilak, F.
Functional tissue engineering: The role of biomechanics. J.
Biomech. Eng. 2000, 122, 570–575. 47. Hoerstrup, S.P.;
Zund, G.; Sodian, R.; Schnell, A.M.; Grunenfelder, J.;
Turina, M.I. Tissue engineering of small caliber vascular
grafts. Eur. J. Cardio-thorac. Surg. 2001, 20, 164–169. 48.
Davisson, T.; Kunig, S.; Chen, A.; Sah, R.; Ratcliffe, A.
Static and dynamic compression modulate matrix metabolism
in tissue engineered cartilage. J. Orthop. Res. 2002, 20,
842–848. E
Electric Cell-Substrate Impedance Sensing

1. Giaever, I.; Keese, C.R. Use of electric fields to


monitor the dynamical aspect of cell behavior in tissue
culture. IEEE Trans. Biomed. Eng. 1986, 33, 242–247.

2. Macdonald, J.R. Fundamentals of Impedance Spectroscopy.


In Impedance Spectroscopy; John Wiley & Sons: New York,
1987; 1–26.

3. Giaever, I.; Keese, C.R. Monitoring fibroblast behavior


with an applied electric field. Proc. Natl. Acad. Sci. U. S.
A. 1984, 81, 3761–3764.

4. Giaever, I.; Keese, C.R. Fractal motion of mammalian


cells. Physica, D 1989, 39, 128–133.

5. Wegener, J.; Keese, C.R.; Giaever, I. Electric


cellsubstrate impedence sensing (ECIS) as a noninvasive
means to monitor the kenetics of cell spreading to
artificial surfaces. Exp. Cell Res. 2000, 259, 158–166.

6. Charboneau, A.L.; Singh, V.; Yu, T.X.; Newsham, I.F.


Suppression of growth and increased cellular attachment
after expression of DAL-1 in MCF-7 breast cancer cells.
Int. J. Cancer 2002, 100 (2), 181–188.

7. Schmidt, M.H.H.; Chen, B.; Randazzo, L.M.; Bogler, O.


SETA/CIN85/Ruk and its binding partner ALP1 associated with
diverse cytoskeletal elements, including FAKs, and
modulated cell adhesion. J. Cell. Sci. 2003, 166,
2845–2855.

8. Sawhney, R.S.; Zhou, G.K.; Humphrey, L.E.; Ghosh, P.;


Kreisberg, J.I.; Brattain, M.G. Differences in sensitivity
of biological functions mediated by epidermal growth factor
receptor activation with respect to endogenous and
exogenous ligands. J. Biol. Chem. 2002, 277, 75–85. 9. Lo,
C.M.; Keese, C.R.; Giaever, I. Monitoring motion of
confluent cells in tissue culture. Exp. Cell Res. 1993, 204,
102–109. 10. Lo, C.M.; Keese, C.R.; Giaever, I. pH Changes
in pulsed CO 2 incubators cause periodic change in cell
morphology. Exp. Cell Res. 1994, 213, 391–397. 11.
Tiruppathi, C.; Malik, A.B.; Del Vecchio, P.J.; Keese,
C.R.; Giaever, I. Electrical method for detection of
endothelial cell shape change in real time. Proc. Natl.
Acad. Sci. U. S. A. 1992, 89, 7919–7923. 12. Lo, C.M.;
Keese, C.R.; Giaever, I. Cell-substrate contact: Another
factor may influence transepithelial electrical resistance
of cell layers cultured on permeable filters. Exp. Cell Res.
1999, 250, 576–580. 13. Giaever, I.; Keese, C.R.
Micromotion of mammalian cells measured electrically. Proc.
Natl. Acad. Sci. U. S. A. 1991, 88, 7896–7900. 14. Giaever,
I.; Keese, C.R. Correction. Proc. Natl. Acad. Sci. U. S. A.
1993, 90, 1634. 15. Lo, C.M.; Keese, C.R.; Giaever, I.
Impedance analysis of MDCK cells measured by electric
cell-substrate impedance sensing. Biophys. J. 1995, 69,
2800–2807. 16. Burns, A.R.; Bowden, R.A.; MacDonell, S.D.;
Walker, D.C.; Odebunmi, T.O.; Donnachie, E.M.; Simon, S.I.;
Entman, M.L.; Smith, C.W. Analysis of tight junctions
during neutrophil transendothelial migration. J. Cell. Sci.
2000, 113, 45–75. 17. Keese, C.R.; Bhawe, K.; Wegener, J.;
Giaever, I. Realtime impedance assay to follow the invasive
activities of metastatic cells in culture. BioTechniques
2002, 33, 842–850. 18. Giaever, I.; Keese, C.R. Toxic?
Cells can tell. Chemtech 1992, 116–125. 19. Keese, C.;
Karra, N.; Dillon, B.; Goldberg, A.; Giaever, I.
Cell–substratum interactions as a predictor of
cytotoxicity. In Vitro Mol. Toxicol. 1998, 11 (2), 183–192.
20. Xiao, C.; Lachance, B.; Sunahara, G.; Luong, J.
Assessment of cytotoxicity using electric cell-substrate
impedance sensing: Concentration and time response function
approach. Anal. Chem. 2002, 74 (22), 5748– 5753. 21.
Hadjout, N.; Laevsky, G.; Knecht, D.A.; Lynes, M.A.
Automated real-time measurement of chemotactic cell
motility. BioTechniques 2001, 31 (5), 1130–1138. 22. Ghosh,
P.M.; Keese, C.R.; Giaever, I. Monitoring
electropermeabilization in the plasma membrane of adherent
mammalian cells. Biophys. J. 1993, 64, 1602– 1609. 23.
Wegener, J.; Keese, C.R.; Giaever, I. Recovery of adherent
cells after in situ electroporation monitored
electronically. BioTechniques 2002, 33 (2), 348–357.
Electrical Impedance Imaging

15. Somersalo, E.; Cheney, M.; Isaacson, D. Existence and


uniqueness for electrode models for electric current
computed tomography. SIAM J. Appl. Math. 1992, 52 (4),
1023–1040.

16. Cook, R.D.; Saulnier, G.J.; Gisser, D.; Goble, J.C.;


Newell, J.C.; Isaacson, D. ACT3: A high-speed,
highprecision electrical impedance tomograph. IEEE Trans.
Biomed. Eng. 1994, 41 (8), 713–722.

17. Hartov, A.; Kerner, T.E.; Markova, M.T.; Osterman,


K.S.; Paulsen, K.D. Dartmouth’s next generation EIS system:
Preliminary hardware considerations. Physiol. Meas. 2001,
22, 25–30.

18. Hartov, A.; Mazzarese, R.; Reiss, F.; Kerner, T.;


Osterman, S.; Williams, D.; Paulsen, K.D. A multichannel
continuously-selectable multi-frequency electrical
impedance spectroscopy measurement system. IEEE Trans. Med.
Imag. 2000, 47 (1), 49–58.

19. Hartov, A.; Demidenko, E.; Soni, N.; Markova, M.;


Paulsen, K. Using voltage sources as current drivers for
electrical impedance tomography. Meas. Sci. Technol. 2002,
13, 1425–1430.

20. IEC International Standard. Medical Electrical


Equipment. Part 1: General Requirements for Safety; 1993.
Document reference number IEC 601-1-2.

21. Association for the Advancement of Medical


Instrumentation. American National Standard. Safe Current
Limits for Electromedical Apparatus; Dec. 2 1993. ANSI/
AAMI ES1-1993, Revision of ANSI/AAMI ES1-1985.

22. Barber, D.C.; Brown, B.H. Imaging spatial distribution


of resistivity using applied potential tomography.
Electron. Lett. 1983, 19 (22), 933–935. 23. Yorkey, J.Y.;
Webster, J.G.; Tompkins, W.J. Comparing reconstruction
algorithms for electrical impedance tomography. IEEE Trans.
Biomed. Eng. 1987, BME-34 (11), 843–852. 24. Barber, D.C.;
Brown, B.H.; Avis, N.J. Image reconstruction in electrical
impedance tomography using filtered back-projection. Proc.
14th Annu. Int. Conf. IEEE-EMBS 1994, 14, 1691–1692. 25.
Vauhkonen, P.J.; Vauhkonen, M.; Savolainen, T.; Kaipio,
J.P. Three-dimensional electrical impedance tomography
based on the complete electrode model. IEEE Trans. Biomed.
Eng. 1999, 46 (9), 1150–1160. 26. Paulsen, K.D.; Jiang, H.
An enhanced electrical impedance imaging algorithm for
hyperthermia applications. Int. J. Hypertherm. 1997, 13
(5), 459–480. 27. Polydorides, N.P.; Lionheart, W.R.B. A
MATLAB toolkit for three dimensional electrical impedance
tomography: A contribution to the EIDORS project. Meas.
Sci. Technol. 2002, in press. 28. Vauhkonen, M.; Lionheart,
W.R.B.; Heikkinen, L.M.; Vauhkonen, P.J.; Kaipio, J.P. A
Matlab package for the EIDORS project to reconstruct
two-dimensional EIT images. Physiol. Meas. 2001, 22,
107–111. 29. Mueller, J.L.; Siltanen, S.; Isaacson, D. A
direct reconstruction algorithm for electrical impedance
tomography. IEEE Trans. Med. Imag. 2002, 21 , 555–559. 30.
Hartov, A.; Kerner, T.E.; Paulsen, K.D. Simulation of error
propagation in finite element image reconstruction for
electrical impedance tomography. Meas. Sci. Technol. 2001,
12, 1040–1049. E
Electroactive Polymeric Materials

24. Schuhmann, W.; Ohara, T.; Heller, A.; Schmidt, H.-L.


Electron transfer between glucose oxidase and electrodes
via redox mediators bound with flexible chains to the enzyme
surface. J. Am. Chem. Soc. 1991, 113, 1394.

25. Foulds, N.; Lowe, C. Immobilization of glucose oxidase


in ferrocene modified pyrrole polymers. Anal. Chem. 1988,
60, 2473.

26. Willner, I.; Willner, B. Electrical communication of


redox proteins by means of electron relay-tethered polymers
in photochemical, electrochemical and photoelectrochemical
systems. React. Polym. 1994, 22, 267.

27. Khan, G.; Ohwa, M.; Wernet, W. Design of stable charge


transfer complex electrode for a third generation
amperometric glucose sensor. Anal. Chem. 1996, 68, 2939.

28. Schuhmann, W.; Huber, J.; Kranz, C.; Wohlscjalger, H.


Conducting polymer based amperometric enzyme electrode:
Towards the development of miniaturized reagentless
biosensors. Synth. Met. 1994, 61, 31.

29. Nishizawa, M.; Matsue, T.; Uchida, I. Penciliin sensor


based on a microarray electrode coated with pH responsive
polypyrrole. Anal. Chem. 1992, 64, 2462.

30. Livache, T.; Roget, A.; Dejean, E.; Barthet, C.; Bidan,
G.; Te´oule, R. Preparation of a DNA matrix via an
electrochemically directed polymerization of pyrrole and
oligonucleotides bearing a pyrrole group. Nucleic Acids
Res. 1994, 22, 2915.

31. Livache, T.; Roget, A.; Dejean, E.; Barthet, C.; Bidan,
G.; Te´oule, R. Biosensing effects in functionalized
electroconducting conjugated polymer layers: Addressable
DNA matrix for the detection of gene mutations. Synth. Met.
1995, 71, 2143.

32. Yamato, H.; Ohwa, M.; Wernet, W. Stability of


polypyrrole and poly(3,4,-ethydioxythiophene) for biosensor
application. J. Electroanal. Chem. 1995, 397, 163.

33. Parente, A.; Marques, E.; Azevedo, W.; Diniz, F.; Melo,
E.; Lime Filho, J. Glucose biosensor using glucose oxidase
immobilized in polyaniline. Appl. Biochem. Biotechnol.
1992, 37, 267.
34. Miller, L.L.; Zhou, Q.-X. Poly(N-methypyrrolylium)
poly(styrenesulfonate). A conductive, electrically
switchable cation exchanger that cathodically binds and
anodically releases dopamine. Macromolecules 1987, 20,
1594.

35. Zhou, Q.-X.; Miller, L.L.; Valentine, J.R.


Electrochemically controlled binding and release of
protonated dimethyldopamine and other cations from poly
(N-methy pyrrole)/polyanion composite redox polymers. J.
Electroanal. Chem. 1989, 261, 147.

36. Prezyna, L.A.; Qiu, Y.-J.; Reynolds, J.R.; Wnek, G.E.


Interaction of cationic polypeptides with electroactive
polypyrrole/poly(styrenesulfonate) and
poly(N-methylpyrrole)/poly(styrenesulfonate) films.
Macromolecules 1991, 24, 2224.

37. Wong, J.; Ingber, D.E.; Langer, R. Electrically


conductive polymers can noninvasively control cell shape.
Proc. Natl. Acad. Sci. U. S. A. 1994, 91, 3201. 38.
Shastri, V.R.; Wnek, G.E. Effect of dopant ion on cell
growth on polypyrrole thin films. Polym. Prepr. 1993, 34
(2), 70. 39. Shastri, V.R. Evaluation of Polypyrrole ‘‘Thin
Films’’ as Substratum for Mammalian Cell Culture, Doctoral
Dissertation; Rensselaer Polytechnic Institute: Troy, NY,
1995. 40. Schmidt, C.E.; Shastri, V.R.; Vacanti, J.P.;
Langer, R. Stimulation of neurite outgrowth using
electrically conducting polymer. Proc. Natl. Acad. Sci. U.
S. A. 1997, 94, 8948. 41. Shastri, V.R.; Schmidt, C.E.;
Kim, T-H.; Vacanti, J.P.; Langer, R. Polypyrrole-A
potential candidate for stimulated nerve regeneration. Mat.
Res. Soc. Symp. Proc. 1996, 414, 113. 42. Collier, J.H.;
Camp, J.P.; Hudson, T.W.; Schmidt, C.E. Synthesis and
characterization of polypyrrole-hyaluronic acid composite
biomaterials for tissue engineering applications. J.
Biomed. Mat. Res. 2000, 50, 574. 43. Shastri, V.P.; Rahman,
N.; Martin, I.; Langer, R. Applications of conductive
polymers in bone regeneration. Mat. Res. Soc. Symp. Proc.
1999, 550, 215. 44. Zelikin, A.; Lynn, D.M.; Farhadi, J.;
Martin, I.; Shastri, V.P.; Langer, R. Erodible conducting
polymers for potential biomedical applications. Angew.
Chem. Int. Ed. 2002, 41 (1), 141. 45. Rivers, T.J.; Hudson,
T.W.; Schmidt, C.E. Synthesis of a novel, biodegradable
electrically conducting polymer for biomedical
applications. Adv. Funct. Mat. 2002, 12 (1), 33. 46. Lines,
M.E.; Glass, A.M. Principles and Applications of
Ferroelectric Related Materials; Clarendon Press: Oxford,
1977. 47. Kawai, H. The piezoelectricity of
poly(vinylidinefluoride). Jpn. J. Appl. Phys. 1969, 8, 975.
48. Shamos, M.H.; Lavine, L.S. Piezoelectricity as a
fundamental property of biological tissue. Nature 1967,
213, 267. 49. Fakuda, E. Piezoelectricity of biopolymers.
Biorheology 1995, 32 (6), 593. 50. Konikoff, J.J. Origins
of osseous bioelectric potentials. A review. Ann. Clin.
Lab. Sci. 1975, 5 (5), 330. 51. Kepler, R.G.; Anderson,
R.A. Ferroelectric polymers. Adv. Phys. 1992, 41 , 1. 52.
Lovinger, A.J. Poly(Vinylidinefluoride), in Developments in
Crystalline Polymers. In Applied Science; Bassett, D.C.,
Ed.; Kluwer Academic Publishers, 1982; 195. 53.
Dvey-Aharon, H.; Sluckin, T.J.; Taylor, P.L. Kink
propagation as a model for poling in
poly(vinylidinefluoride). Phys. Rev., B. 1980, 21, 3700. 54.
Soballe, K.; Overgaard, S.; Hansen, E.S.;
BrokstedtRasmussen, H.; Lind, M.; Bunger, C.J. A review of
ceramic coatings for implant fixation. J. Long-Term Eff.
Med. Implants 1999, 9 (1–2), 131. 55. Wahlstrom, O.
Stimulation of fracture healing with electromagnetic fields
of extremely low frequency. Clin. Orthop. 1984, 186, 293.
56. Weiss, D.S.; Kirsner, R.; Eaglstein, W.H. Electrical
stimulation and wound healing. Arch. Dermatol. 1990, 126,
222. 57. Alvarez, O.M.; Mertz, P.M.; Smerbeck, R.V.;
Eaglstein, W.H. The healing of superficial skin wounds is
stimulated by external electric fields. J. Invest. Dermatol.
1983, 81, 144. 58. Aebischer, P.; Valentini, R.; Dario, P.;
Domencini, C.; Galletti, P. Piezoelectric guidance channels
enhance regeneration in the mouse sciatic nerve after
axotomy. Brain Res. 1987, 436, 165. 59. Valentini, R.;
Vargo, T.; Gardella, J.; Aebischer, P. Electrically charged
polymeric substrates enhance nerve fiber outgrowth in vitro.
Biomaterials 1992, 13, 183. 60. Tanaka, T. Collapse of gels
and the critical endpoint. Phys. Rev. Lett. 1978, 40, 820.
61. Tanaka, T.; Fillmore, D.J. Kinetics of swelling of
gels. J. Chem. Phys. 1979, 70, 1214. 62. Tanaka, T.;
Nishio, I.; Sun, S-T.; Ueno-Nishio, S. Collapse of gels
under an electric field. Science 1982, 218, 467. 63. Osada,
Y.; Ross-Murphy, S. Intelligent gels. Sci. Am. 1993, 268,
82. 64. Osada, Y.; Hasebe, M. Electrically activated
mechanochemical devices using polyelectrolyte gels. Chem.
Lett. 1985, 1285–1288, 1985. 65. Gong, J.P.; Nitta, T.;
Osada, Y. Electrokinetic modeling of the contractile
Phenomena of polyelectrolyte gels-one dimensional capillary
model. J. Phys. Chem. 1994, 98, 9583. 66. Osada, Y.; Gong,
J.P.; Sawahata, K.J. Synthesis, mechanism, and application
of electro-driven chemomechanical system using polymer
gels. J. Macromol. Sci., Chem. 1991, A28, 1189. 67. Osada,
Y.; Okuzaki, H.; Hori, H. A polymer gel with electrically
driven motility. Nature 1992, 355, 242. 68. Kwon, I.C.;
Bae, Y.H.; Kim, S.W. Electrically erodible polymer gel for
controlled release of drugs. Nature 1991, 354, 291. 69.
Alexiou, C.; Arnold, W.; Klein, R.J.; Parak, F.G.; Hulin,
P.; Bergemann, C.; Erhardt, W.; Wagenpfeil, S.; Lu¨bbe,
A.S. Locoregional cancer treatment with magnetic drug
targeting. Cancer Res. 2000, 60, 6641. 70. Lee, S.W.; Mao,
C.; Flynn, C.E.; Belcher, A.M. Ordering of quantum dots
using genetically engineered viruses. Science 2002, 296,
892. 71. Santini, J.T.; Cima, M.J.; Langer, R. A
controlledrelease microchip. Nature 1999, 397, 335. 72.
Lahann, J.; Mitragotri, S.; Tran, T.-H.; Kaido, H.;
Sundaram, J.; Choi, I.S.; Hoffer, S.; Somorjai, G.; Langer,
R. A reversibly switching surface. Science 2003, 299, 371.
E
Electrocardiography

6. Rawlings, C.A. Traditional Electrocardiographic


Voltages. In Electrocardiography, 1st Ed.; Spacelabs, Inc.:
Redmond, Washington, 1991; 27–47.

7. Rideout, V.C. Simulation of a Complete Cardiovascular


Loop. In Mathematical and Computer Modeling of
Physiological Systems, 1st Ed.; Prentice Hall: New Jersey,
1991; 90–105.

8. Guyton, A.C.; Hall, J. The Normal Electrocardiogram. In


Textbook of Medical Physiology, 9th Ed.; W.B. Saunders
Company: Philadelphia, 1996; 129–134. 9. Thaler, M.S. The
Only EKG Book You’ll Ever Need, 2nd Ed.; J.B. Lippincott
Co: Philadelphia, 1995; 10–233. 10. Guyton, A.C.; Hall, J.
Electrocardiographic Interpretation of Cardiac Muscle and
Coronary Abnormalities: Vectorial Analysis. In Textbook of
Medical Physiology, 9th Ed.; W.B. Saunders Company:
Philadelphia, 1996; 135–148.
Electrogastrography

1. Alvarez, W.C. New methods of studying gastric


peristalsis. J. Am. Med. Assoc. 1922, 22, 1281–1284.

2. Davis, R.C.; Garafolo, L.; Gault, F.P. An exploration of


abdominal potentials. J. Comparative Physiol. Psychol.
1957, 50, 519–523.

3. Davis, R.C.; Garafolo, L.; Kveim, K. Conditions


associated with gastrointestinal activity. J. Comparative
Physiol. Psychol. 1959, 52, 466–475.

4. Smout, A.J.P.M.; van der Schee, E.J.; Grashuis, J.L.


What is measured in electrogastrography? Digestive Dis.
Sci. 1980, 25, 179–187.

5. Koch, K.L.; Stewart, W.R.; Stern, R.M. Effect of barium


meals on gastric electromechanical activity in man. A
fluoroscopic-electrogastrographic study. Digestive Dis. Sci.
1987, 32, 1217–1222.

6. Koch, K.L.; Stern, R.M. Handbook of Electrogastrography;


Oxford University Press; New York, 2004.

7. Harm, D.L.; Sandoz, G.R.; Stern, R.M. Changes in gastric


myoelectric activity during space flight. Digestive Dis.
Sci. 2002, 47, 1737–1745.

8. Chen, J.D.Z.; Zou, X.P.; Lin, X.M.; Ouyang, S.; Liang,


J.Detection of gastric slow wave propagation from the
cutaneous electrogastrogram. Am. J. Physiol. 1996, 277,
G424–G430.

9. McNearney, T.; Lin, X.; Shrestha, J.; Lisse, J.; Chen,


J.D.Z. Characterization of gastric myoelectrical rhythms in
patients with systemic sclerosis using multichannel surface
electrogastrography. Digestive Dis. Sci. 2002, 47, 690–698.

10. Stern, R.M.; Stacher, G. Recording the


electrogastrogram from parts of the body surface distant
from the stomach. Psychophysiology 1982, 19, 350.

11. Chen, J. Signal processing and analysis.


Neurogastroenterology 2000, 3, 104–112.

12. Stern, R.M.; Koch, K.L.; Muth, E.R. Gastrointestinal


system. In Handbook of Psychophysiology, 2nd Ed.; Cacioppo,
J.T., Tassinary, L.G., Berntson, G.G., Eds.; Cambridge
University Press: New York, 2000; 294–314.
13. Van der Schee, E.J.; Smout, A.J.P.M.; Grashuis, J.L.
Application of running spectral analysis to
electrogastrographic signals recorded from dog and man. In
Motility in the Digestive Tract; Wienbeck, Ed.; Raven
Press: New York, 1982; 241–250.

14. Smout, A.J.P.M.; Jebbink, H.J.A.; Samsom, M.


Acquisition and analysis of electrogastrographic data: The
Dutch experience. In Electrogastrography; Chen, J.Z.,
McCallum, R.W., Eds.; Raven Press: New York, 1994; 3–30. E
Electromyography

1. Liddell, E.G.; Sherrington, C.S. Recruitment and some


other factors of reflex inhibition. Proc. R. Soc. London
1925, Ser. B 97 (686), 488–518. 2. Burke, R.E. Motor units:
Anatomy, physiology and functional organization. In
Handbook of Physiology: The Nervous System; Brookhart,
J.M., Mountcastle, V.B., Eds.; American Physiological
Society: Bethesda, MD, 1981; Vol. 2, 345–422. 3. Buchthal,
F.; Erminio, F.; Rosenfalck, P. Motor unit territory in
different human muscles. Acta Physiol. Scand. 1959, 45,
72–87. 4. Miller-Larsson, A. A model of spatial
distribution of muscle fibers of a motor unit in normal
human limb muscles. Electromyogr. Clin. Neurophysiol. 1980,
20, 281–298. 5. Hermens, H.J.; Freriks, B.; Merletti, R.;
Ha¨gg, G.; Stegeman, D.F.; Blok, J.; Rau, G.;
Disselhorst-Klug, C. SENIAM 8: European Recommendations for
Surface Electromyography, Roessingh Research and
Development; Enschede, The Netherlands, 1999. 6. Henneman,
E. Relations between size of neurons and their
susceptibility to discharge. Science 1957, 126, 1345–1346.
7. Radicheva, N.; Gerilovsky, L.; Gydikov, A. Effect of
short interstimulus intervals on the intra and
extracellular action potentials of isolated frog muscle
fibers. Acta Physiol. Pharmacol. Bulgar. 1986, 12 (1),
26–35. 8. Ekstedt, J. Human single muscle fiber action
potentials. Acta Physiol. Scand. Suppl. 1964, 226, 1–96. 9.
Basmajian, J.V.; De Luca, C.J. Muscles Alive: Their
Functions Revealed by Electromyography, 5th Ed.; Williams &
Wilkins: Baltimore, 1985. 10. Bretschneider, F.; de Weille,
J.R. Introduction to Electrophysiological Methods and
Instrumentation; Academic Press: London, 2006. 11. Desmedt,
J.E. Computer-aided electromyograph. In Progress in
Clinical Neurophysiology; Desmedt, J.E., Ed.; Karger:
Basel, 1983; Vol. 10. 12. Weitkunat, R. Digital biosignal
processing. In Techniques in the Behavioral and Neural
Sciences; Huston, J.P., Ed.; Elsevier: Du¨sseldorf, 1991;
Vol. 5. 13. Duchene, J.; Goubel, F. Surface electromyogram
during voluntary contraction: Processing tools and related
to physiological events. CRC Crit. Rev. Biomed. Eng. 1993,
21, 313–397. 14. Farina, D.; Merletti, R.; Enoka, R.M. The
extraction of neural strategies from the surface EMG. J.
Appl. Physiol. 2004, 96 (4), 1486–1495. 15. Milner-Brown,
H.S.; Stein, R.B.; Yemm, R. The contractile properties of
human motor units during voluntary isometric contractions.
J. Physiol. 1973, 228 (2), 285–306. 16. De Luca, C.J. The
use of surface electromyography in biomechanics. J. Appl.
Biomech. 1997 , 13, 135– 163. 17. Gilchrist, J.M.; Sachs,
G.M. Electrodiagnostic studies in the management and
prognosis of neuromuscular disorders. Muscle Nerve. 2004,
29 (2), 165–190. 18. Karpati, G.; Hilton-Jones, D.; Griggs,
R.C. Disorders of Voluntary Muscle, 7th Ed.; Cambridge
University Press: Cambridge, U.K., 2001. E
Electron Microscopy

9. Zhang, Y.; Zhang, M. Synthesis and characterization of


macroporous chitosan/calcium phosphate composite scaffolds
for tissue engineering. J. Biomed. Mater. Res. 2001, 55
(3), 304–312.

10. Hu, Y.; Jiang, X.; Ding, Y.; Ge, H.; Yuan, Y.; Yang, C.
Synthesis and characterization of chitosan-poly(acrylic
acid) nanoparticles. Biomaterials 2002, 23, 3193–3201.

11. Cheng, D.; Tellkamp, V.L.; Lavernia, C.J.; Lavernia,


E.J. Corrosion properties of nanocrystalline C–Cr coatings.
Ann. Biomed. Eng. 2001, 29, 803–809.

12. Roberts, S.; Flatoff, D.; Chiou, W. Low damage sample


preparation of semiconductor materials using low energry
ion milling. Microsc. Microanal. 2003, 9 (Suppl. 2),
810–811.

13. Collier, J.H.; Camp, J.P.; Hudson, T.W.; Schmidt, C.E.


Synthesis and characterization of polypyrrole-hyaluronic
acid composite biomaterials for tissue engineering
applications. J. Biomed. Mater. Res. 2000, 50 (4), 574–584.

14. Dong, Z.L.; Khor, K.A.; Quek, C.H.; White, T.J.;


Cheang, P. TEM and STEM analysis on heattreated and in
vitro plasma-sprayed hydroxyapatite/ Ti-6A1-4V composite
coatings. Biomaterials 2003, 24, 97–105.

15. Wang, X.; Li, Y.; Wei, J.; de Groot, K. Development of


biomimetic nano-hydroxyapatite/poly(hexamethylene
adipamide) composites. Biomaterials 2002, 23, 4787– 4791.

16. Zhao, F.; Yin, Y.; Lu, W.W.; Leong, J.C.; Zhang, W.;
Zhang, J.; Zhang, M.; Yao, K. Preparation and histological
evaluation of biomimetic three-dimensional
hydroxyapatite/chitosan–gelatin network composite
scaffolds. Biomaterials 2002, 23, 3227–3234.

17. Brors, D.; Aletsee, C.; Schwager, K.; Mlynski, R.;


Hansen, S.; Scha¨fers, M.; Ryan, A.F.; Dazert, S.
Interaction of spiral ganglion neuron processes with
alloplastic materials in vitro. Hear. Res. 2002, 167,
110–121.

18. Itoh, S.; Takakuda, K.; Kawabata, S.; Aso, Y.; Kasai,
K.; Itoh, H.; Shinomiya, K. Evaluation of cross-linking
procedures of collagen tubes used in peripheral nerve
repair. Biomaterials 2002, 23, 4475–4481.
19. Pieper, J.S.; van der Kraan, P.M.; Hafmans, T.; Kamp,
J.; Buma, P.; van Susante, J.L.C.; van den Berg, W.B.;
Veerkamp, J.H.; van Kuppevelt, T.H. Crosslinked type II
collagen matrices: Preparation, characterization, and
potential for cartilage engineering. Biomaterials 2002, 23,
3183–3192.

20. Yost, M.J.; Baicu, C.F.; Stonerock, C.E.; Goodwin,


R.L.; Price, R.L.; Davis, J.M.; Evans, H.; Watson, P.D.;
Gore, C.M.; Sweet, J.; Creech, L.; Zile, M.R.; Terracio, L.
A novel tubular scaffold for cardiovascular tissue. Tissue
Eng. 2004, 10, 273–284.

21. Acil, Y.; Springer, I.N.G.; Broek, V.; Terheyden, H.;


Jepsen, S. Effects of bone morphogenetic protein7
stimulation on osteoblasts cultured on different
biomaterials. J. Cell. Biochem. 2002, 86, 90–98.

22. Dong, J.; Uemura, T.; Shirasaki, Y.; Tateishi, T.


Promotion of bone formation using highly pure porous b-TCP
combined with bone marrow-derived osteoprogenitor cells.
Biomaterials 2002, 23, 4493– 4502. 23. Miura, H.; Nishibe,
T.; Yasuda, K.; Shimada, T.; Hazama, K.; Katoh, H.;
Watanabe, S.; Okuda, Y.; Kumada, T. The influence of
node-fibril morphology on healing of high-porosity expanded
polytetraflyuoroethylene grafts. Eur. Surg. Res. 2002, 34
(3), 224–231. 24. Elias, K.L.; Price, R.L.; Webster, T.L.
Enhanced functions of osteoblasts on nanometer diameter
carbon fibers. Biomaterials 2002, 23, 3279–3287. 25. Evans,
H.J.; Sweet, J.K.; Price, R.L.; Yost, M.; Goodwin, R.L.
Novel 3D culture system for study of cardiac myocyte
development. Am. J. Physiol., Heart Circ. Physiol. 2003,
285, H570–H578. 26. Dalby, M.J.; Riehle, M.O.; Johnstone,
H.; Affrossman, S.; Curtis, A.S.G. In vitro reaction of
endothelial cells to polymer demixed nanotopography.
Biomaterials 2002, 23, 2945–2954. 27. Dalby, M.J.; Yarwood,
S.J.; Riehle, M.O.; Johnstone, H.; Affrossman, S.; Curtis,
A.S.G. Increasing fibroblast response to materials using
nanotopography: Morphological and genetic measurements of
cell response to 13-nm-high polymer demixed islands. Exp.
Cell Res. 2002, 276, 1–9. 28. Deng, Y.; Zhao, K.; Zhang,
X.; Hu, P.; Chen, G. Study on the three-dimensional
proliferation of rabbit articular cartilage-derived
chondrocytes on polyhdroxyalkanoate scaffolds. Biomaterials
2002, 23, 4049–4056. 29. Perka, C.; Spitzer, R.S.;
Lindenhayn, K.; Sittinger, M.; Schultz, O. Matrix-mixed
culture: New methodology for chondrocyte culture and
preparation of cartilage transplants. J. Biomed. Mater.
Res. 2000, 49 (3), 305– 311. 30. De Aza, P.N.; Luklinska,
Z.B.; Santos, C.; Guitian, F.; De Aza, S. Mechanism of
bone-like formation on a bioactive implant in vivo.
Biomaterials 2003, 24, 1437– 1445. 31. Pina-Barba, C.;
Bosch, P.; Villarreal, E.; Martin, S.; Leo´n, B.; Palma,
B.; Torres-Villasenor, G. Bone response to 316L-SS and
zinalco implants. Microsc. Microanal. 2003, 9 (Suppl. 2),
1284–1285. 32. Germain, M.A.; Hatton, A.; Williams, S.;
Matthews, J.B.; Stone, M.H.; Fisher, J.; Ingham, E.
Comparison of cytotoxicity of clinically relevant
cobalt–chromium and alumina ceramic wear particles in
vitro. Biomaterials 2003, 24, 469–479. 33. Voggenreiter,
G.; Leiting, S.; Brauer, H.; Leiting, P.; Majetschak, M.;
Bardenheuer, M.; Obertacke, U. Immuno-inflammatory tissue
reaction to stainless-steel and titanium plates used for
internal fixation of long bones. Biomaterials 2003 , 24,
247–254. 34. Carrasquillo, K.G.; Ricker, J.A.; Rigas, I.K.;
Miller, J.W.; Gragoudas, E.S.; Adamis, A.P. Controlled
delivery of the anti-VEGF aptamer EYE001 with
poly(lactic-co-glycolic) acid microspheres. Invest.
Ophthomol. Vis. Sci. 2003, 44 (1), 290–299. 35. Sanborn,
T.J.; Messersmith, P.B.; Barron, A.E. In situ crosslinking
of a biomimetic peptide–PEG hydrogel via thermally
triggered activation of factor XIII. Biomaterials 2002, 23,
2703–2710. 36. Nsereko, S.; Amiji, M. Localized delivery of
paclitaxel in solid tumors from biodegradable chitin
microparticle formulations. Biomaterials 2002, 23,
2723–2731. 37. Kim, S.Y.; Lee, Y.M.; Baik, D.J.; Kang, J.S.
Toxic characteristics of methoxy poly(ethylene glycol)/
poly(e-caprolactone) nanospheres; in vitro and in vivo
studies in the normal mice. Biomaterials 2003, 24, 55–63.
38. Mi, F.; Lin, Y.; Wu, Y.; Shyu, S.; Tsai, Y. Chitin/
PLGA blend microspheres as a biodegradable drugdelivery
system: Phase-separation, degradation and release behavior.
Biomaterials 2002, 23, 3257–3267. E
Electrospinning

1. Gibson, P.; Schreuder-Gibson, H.; Rivin, D. Transport


properties of porous membranes based on electrospun
nanofibers. Colloids Surf., A Physicochem. Eng. Asp. 2001,
187–188, 469–481.

2. Graham, K.; Ouyang, M.; Raether, T.; Grafe, T.;


McDonald, B.; Knauf, P. In Polymeric Nanofibers in Air
Filtration Applications, The Fifteenth Annual Technical
Conference & Expo of the American Filtration & Separations
Society, Galveston, TX, April 9–12, 2002; American
Filtration & Separations Society: Falls Church, VA, 2002.

3. Bognitzki, M.; Hou, H.; Ishaque, M.; Frese, T.; Hellwig,


M.; Schwarte, C.; Schaper, A.; Wendorff, J.H.; Greiner, A.
Polymer, metal, and hybrid nanoand mesotubes by coating
degradable polymer template fibers (TUFT process). Adv.
Mater. 2000, 12, 637–640.

4. Liu, W.; Graham, M.; Evans, E.A.; Reneker, D.H.


Poly(meta-phenylene isophthalamide) nanofibers: Coating and
post processing. J. Mater. Res. 2002, 17 (12), 1–6.

5. Stizel, J.D.; Bowlin, G.L.; Mansfield, K.; Wnek, G.E.;


Simposon, G. . Int. SAMPE Tech. Conf. 2000, 32, 205.

6. Smith, D.; Reneker, D.H.; Kataphinan, W.; Dabney, S. PCT


Int. Appl. WO 2000-US27775, October 6, 2001.

7. Smith, D.; Reneker, D.H. PCT Int. Appl. WO 2000US27776,


October 6, 2001.

8. Li, W.J.; Laurencin, C.T.; Caterson, E.J.; Tuan, R.S.;


Ko, F.K. Electospun nanofibrous structure: A novel scaffold
for tissue engineering. J. Biomed. Mater. Res. 2002, 60
(4), 613–621.

9. Matthews, J.A.; Wnek, G.E.; Simpson, D.G.; Bowlin, G.L.


Electrospinning of collagen nanofibers. Biomacromolecules
2002, 3, 232–238.

10. Kenawy, E.R.; Bowlin, G.L.; Mansfield, K.; Layman, J.;


Simpson, D.G.; Sanders, E.H.; Wnek, G.E. Release of
tetracycline hydrochloride from electrospun
poly(ethylene-co-vinylacetate), poly(lactic acid), and a
blend. J. Control. Release 2002, 81, 57–64.

11. Fong, H.; Reneker, D.H. Electrospinning and Formation


of Nanofibers. In Structure Formation in Polymeric Fibers;
Salem, D.R., Sussman, M.V., Eds.; Hanser: Munich, 2001;
225–246. 12. Norris, I.D.; Shaker, M.M.; Ko, F.K.;
MacDiarmid, A.G. Electrostatic fabrication of ultrafine
conducting fibers: Polyaniline/polyethylene oxide blends.
Synth. Met. 2000, 114, 109–114. 13. Formhals, A. Process
and Apparatus for Preparing Artificial Threads. US Patent
1,975,504October 2, 1934. 14. Lord Rayleigh. On the
equilibrium of liquid conducting masses charged with
electricity. Philos. Mag. J. (Lond.) 1882, 44, 184–186. 15.
Zeleny, J. Instability of electrified liquid surfaces. Phys.
Rev. 1917, 10, 1–6. 16. Taylor, G. Disintegration of water
drops in an electric field. Proc. R. Soc., A 1964, 280,
383–397. 17. Doshi, J.; Reneker, D.H. Electrospinning
process and applications of electrospun fibers. J.
Electrost. 1995, 35, 151–160. 18. Reneker, D.H.; Chun, I.
Nanometre diameter fibres of polymer, produced by
electrospinning. Nanotechnology 1996, 7, 216–223. 19.
Reneker, D.H.; Yarin, A.L.; Fong, H.; Koombhongse, S.J.
Bending instablitity of electrically charged liquid jets of
polymer solutions in electrospinning. Appl. Phys. 2000, 87,
4531–4547. 20. Yarin, A.L.; Koombhongse, S.; Reneker, D.H.
Taylor cone and jetting from liquid droplets in
electrospinning of nanofibers. J. Appl. Phys. 2001, 90 (9),
4836–4846. 21. Spivak, A.F.; Dzenis, Y.A.; Reneker, D.H. A
model of steady state jet in the electrospinning process.
Mech. Res. Commun. 2000, 27 (1), 37–42. 22. Hohman, M.M.;
Shin, M.; Rutledge, G.; Brenner, M.P. Electrospinning and
electrically forced jets. I. Stability theory. Phys. Fluids
2001, 13 (8), 2201–2236. 23. Hohman, M.M.; Shin, M.;
Rutledge, G.; Brenner, M.P. Electrospinning and
electrically forced jets. II. Applications. Phys. Fluids
2001, 13 (8), 2201–2236. 24. Tsai, P.; Schreuder-Gibson,
H.; Gibson, P. Different electrostatic methods for making
electric filters. J. Electrost. 2002, 54, 333–341. 25. Shin,
Y.M.; Hohman, M.M.; Brenner, M.P.; Rutledge, G.C.
Experimental characterization of electrospinning: The
electrically forced jet and instabilities. Polymer 2001,
42, 9955–9967. 26. Hou, H.; Zeng, J.; Reuning, A.; Schaper,
A.; Wendorff, J.H.; Greiner, A. Poly(p-xylylene) nanotubes
by coating and removal of ultrathin polymer template fibers.
Macromolecules 2002, 35, 2429–2431. 27. Fong, H.; Reneker,
D.H. Elastomeric nanofibers of styrene–butadiene–styrene
triblock copolymer. J. Polym. Sci., B, Polym. Phys. 1999,
37, 3488–3493. 28. Hou, H.; Averdung, J.; Czado, W.;
Greiner, A.; Wendorff, J.H. Oriented Mesotubular and
Nanotubluar Oriented Nonwoven Fabrics and Their
Manufacture. German Patent 2000-10053263, October 26, 2000.
29. Hou, H.; Averdung, J.; Czado, W.; Greiner, A.;
Wendorff, J.H. Oriented Mesotubular and Nanotubluar
Oriented Nonwoven Fabrics and Their Manufacture. PCT Int.
Appl., PIXXD2 WO 0234986, May 2, 2002. 30. Yarin, A.L.;
Koombhongse, S.K.; Reneker, D.H. Bending instability in
electrospinning of nanofibers. J. Appl. Phys. 2001, 89,
3018–3029. 31. Baumgarten, P.K. Experimental
characterization of electrospinning: The electrically
forced jet and instabilities. J. Colloid Interface Sci.
1971, 36, 71. 32. Reneker, D.H.; Xu, H. In Fluid Polymer
Jets During Electrospinning of Nanofibers. Annual APS March
Meeting, Indianapolis, IN, March 18–22, 2002; American
Physical Society: College Park, MD, 2002.
www.aps.org/meet/MAR02/index.html. 33. Ita¨la¨, A.I.;
Yla¨nen, H.O.; Ekholm, C.; Karlsson, K.H.; Aro, H.T. Pore
diameter of more than 100 mm is not requisite for bone
ingrowth in rabbits. J. Biomed. Mater. Res., Appl.
Biomater. 2001, 58, 679–683. 34. Agrawal, C.M.; Ray, R.B.
Biodegradable polymeric scaffolds for musculoskeletal
tissue engineering. J. Biomed. Mater. Res. 2001, 55,
141–150. 35. Ignatious, F.; Baldoni, J.M. Electrospun
Pharmaceutical Compositions Comprising a Polymeric Carrier.
US 2000-178682, January 28, 2000. 36. Ignatious, F.;
Baldoni, J.M. Electrospun pharmaceutical compositions
comprising a polymeric carrier. PCT Int. Appl., PIXXD2 WO
0154667, August 2, 2001. 37. Lin, S.Y.; Chen, K.S.; Liang,
R.C. Design and evaluation of drug-loaded wound dressing
having thermoresponsive, adhesive, absorptive and easy
peeling properties. Biomaterials 2001, 22, 2999–3004. 38.
Winter, G.D. Formation of the scab and the rate of
epithelialization of superficial wounds in the skin of young
domestic pig. Nature 1962, 193, 257–268. 39. Bognitzki, M.;
Czado, W.; Frese, T.; Schaper, A.; Hellwig, M.; Steinhart,
M.; Greiner, A.; Wendorff, J.H. Nanostructured fibers via
electrospinning 2001, 13 (1), 70–72. 40. Matossian, J.;
Wei, R.; Vajo, J.; Hunt, G.; Gardos, M.; Chambers, G.;
Soucy, L.; Oliver, D.; Jay, L.; Taylor, C.M.; Alderson, G.;
Komanduri, R.; Perry, A. Plasmaenhanced,
magnetron-sputtered deposition (PMD) of materials. Surf.
Coat. Technol. 1998, 108–109, 496–506. 41. Caruso, R.A.;
Antonietti, M.; Giersig, M.; Hentze, H.P.; Jia, J.
Modification of TiO 2 network structures using a polymer gel
coating technique. Chem. Mater. 2001, 13 (3), 1114–1123. E
Endoscopy

6. Menze, J.; Domschke, W.; Brambs, H.J.; Frank, N.;


Hatfield, A.; Natternman, C.; Odegaard, S.; Seifert, H.;
Tamada, K.; Tio, T.L.; Foerster, E.C. Miniprobe
ultrasonography in the upper gastrointestinal tract: state
of the art 1995, and prospects. Endoscopy 1995, 28 (6),
508–513.

7. Adain, A.L.; Han-Chuan, T.; Casidy, M.; Schiano, T.D.;


Liu, J.; Miller, L.S. High-resolution endoluminal
sonography is a sensitive modality for the identification of
Barrett’s metaplasia. Gastrointest. Endosc. 1997, 46 (2),
147–151.

8. Gong, F.; Swain, P.; Mills, T. Wireless endoscopy.


Gastrointest. Endosc. 2000, 51 (6), 725–729.

9. Iddan, G.; Meron, G.; Glukhovsky, A.; Swain, P. Wireless


capsule endoscopy. Nature 2000, 405 (6785), 417.

10. Kudo, S.; Tamura, S.; Nakajima, T.; Yamano, H.; Kusaka,
H.; Watanabe, H. Diagnosis of colorectal tumorous lesions
by magnifying endoscopy. Gastrointest. Endosc. 1996, 44
(1), 8–14.

11. Siegel, L.M.; Stevens, P.D.; Lightdale, C.J.; Green,


P.H.R.; Goodman, S.; Garcia-Carrasquillo, R.J.; Rotterdam,
H. Combined magnification endoscopy with chromoendoscopy in
the evaluation of patients with suspected malabsorption.
Gastrointest. Endosc. 1997, 46 (3), 226–230.

12. Jaramillo, E.; Watanabe, M.; Befrits, R.; Ponce de


Leo´n, E.; Rubio, C.; Slezak, P. Small, flat colorectal
neoplasias in long-standing ulcerative colitis detected by
highresolution video endoscopy. Gastrointest. Endosc. 1996,
44 (1), 15–22.

13. Gono, K.; Obi, T.; Yamaguchi, M.; Ohyama, N.; Machida,
H.; Sano, Y.; Yoshida, S.; Hamamoto, Y.; Endo, T.
Appearance of enhanced tissue features in narrow-band
endoscopic imaging. J. Biomed. Opt. 2004, 9 (3), 568–577.

14. Axelrad, A.M.; Fleischer, D.E.; Geller, A.J.; Nguyen,


C.C.; Lewis, J.H.; Al-Kawas, F.H.; Avigan, M.I.;
Montgomery, E.A.; Benjamin, S.B. High-resolution
chromoendoscopy for the diagnosis of diminutive colon
polyps: implications for colon cancer screening.
Gastroenterology 1996, 110 (4), 1253–1258. 15. Kumagai, Y.;
Monma, K.; Kawada, K. Magnifying chromoendoscopy of the
esophagus: in-vivo pathological diagnosis using an
endocytoscopy system. Endoscopy 2004, 36 (7), 590–594. 16.
Kara, M.A.; Peters, F.P.; ten Kate, F.J.W.; van Deventer,
S.J.; Fockens, P.; Bergman, J.J.G.H.M. Endoscopic video
autofluorescence imaging may improve the detection of early
neoplasia in patients with barrett’s esophagus.
Gastrointest. Endosc. 2005, 61 (6), 679–685. 17. Mayinger,
B.; Neidhardt, S.; Reh, H.; Martus, P.; Hahn, E.G.
Fluorescence induced with 5-aminolevulinic acid for the
endoscopic detection and follow-up of esophageal lesions.
Gastrointest. Endosc. 2001, 54 (5), 572–578. 18. Huang, D.;
Swanson, E.A.; Lin, C.P.; Schuman, J.S.; Stinson, W.G.;
Chang, W.; Hee, M.R.; Flotte, T.; Gregory, K.; Puliafito,
C.A.; Fujimoto, J.G. Optical coherence tomography. Science
1991, 254 (5035), 1178–1181. 19. Rollins, A.M.;
Ung-arunyawee, R.; Chak, A.; Wong, R.C.K.; Kobayashi, K.;
Sivak, M.V., Jr.; Izatt, J.A. Real-time in vivo imaging of
human gastrointestinal ultrastructure by use of endoscopic
optical coherence tomography with a novel efficient
interferometer design. Opt. Lett. 1999, 24 (19), 1358–1360.
20. Corle, T.R.; Kino, G.S. Confocal Scanning Optical
Microscopy and Related Imaging Systems; Academic Press: San
Diego, California, 1996. 21. Wang, T.D.; Contag, C.H.;
Mandella, M.J.; Chan, N.Y.; Kino, G.S. Confocal fluorescence
microscope with dual-axis architecture and biaxial
postobjective scanning. J. Biomed. Opt. 2004, 9 (4),
735–742. 22. Kiesslich, R.; Burg, J.; Vieth, M.;
Gnaendiger, J.; Enders, M.; Delaney, P.; Polglase, A.;
McLaren, W.; Janell, D.; Thomas, S.; Nafe, B.; Galle, P.R.;
Neurath, M.F. Confocal laser endoscopy for diagnosing
intraepithelial neoplasias and colorectal cancer in vivo.
Gastroenterology 2004, 127, 706–713.
Ergonomics

43. Bigos, S.; Battie´, M.; Spengler, D.; Fisher, L.;


Fordyce, W.; Hansson, T. A prospective study of work
perceptions and psychosocial factors affecting the report
of back injury. Spine 1991, 16, 1–6.

44. Holmstro¨m, E.; Lindell, J.; Moritz, U. Low back and


neck/shoulder pain in construction workers: Occupational
workload and psychosocial risk factors. Part 2:
Relationship to neck and shoulder pain. Spine 1992, 17,
672–677.

45. Svensson, H.; Andersson, G. The relationship of low


back pain, work history, work environment, and stress.
Spine 1989, 14, 517–522.

46. Bigos, S.; Spengler, D.; Martin, N.; Zeh, J.; Fisher,
L.; Nachemson, A. Back injuries in industry: A
retrospective study III. Employee-related factors. Spine
1986, 11, 252–256.

47. Elo, A.; Ilmarinen, J.; Leppanen, A.; Martikainen, R.


The differences in psychological work factors and work
ability by musculoskeletal disorders among employees in the
metal industry. Adv. Occup. Ergon. Saf. I 1996, 2, 281–285.

48. Leino, P.; Ha¨nninen, V. Psychosocial factors at work


in relation to back and limb disorders. Scand. J. Work,
Environ. & Health 1995, 21, 134–142.

49. National Institute for Occupational Safety and Health.


Elements of Ergonomics Programs: A Primer Based on
Workplace Evaluations of Musculoskeletal Disorders, DHHS
(NIOSH) Publication No. 97-117; U.S. Department of Health
and Human Services: Cincinnati, OH, 1997.

50. McSweeney, K.; Craig, B.; Congleton, J.; Miller, D.


Ergonomic program effectiveness: Ergonomics and medical
intervention. Int. J. Occup. Saf. Ergon. 2002, 8, 433–449.

51. Waters, T.; Putz-Anderson, V.; Garg, A. Revised NIOSH


Lifting Equation, NIOSH Publication No. 94110; US
Department of Health and Human Services: Cincinnati, OH,
1994.

52. Snook, S.H.; Ciriello, V.M. The design of manual


handling tasks: Revised tables of maximum acceptable
weights and forces. Ergonomics 1991, 34, 1,197–1,213.
53. Snook, S.; Campanelli; Hart, J. A study of three
preventative approaches to low back injuries. J. Occup.
Med. 1978, 20, 478–481.

54. Moore, J.; Garg, A. The strain index: A proposed method


to analyze jobs for risk of distal upper extremity
disorders. Am. Ind. Hyg. Assoc. J. 1995, 56, 443–458.

55. Moore, J.S.; Rucker, N.P.; Knox, K. Validity of generic


risk factors and the strain index for predicting
nontraumatic distal upper extremity morbidity. Am. Ind.
Hyg. Assoc. J. 2001, 62, 229–235. 56. Knox, K.; Moore, J.S.
Predictive validity of the strain index in turkey
processing. J. Occup. Environ. Med. 2001, 43, 451–460. 57.
Rucker, N.P.; Moore, J.S. Predictive validity of the strain
index in manufacturing. Appl. Occup. Environ. Hyg. 2002,
17, 63–73. 58. Appendix B: Criteria for Analyzing and
Reducing WMSD Hazards for Employers Who Choose the Specific
Performance Approach, WAC 296-62-05174; Washington State
Department of Labor and Industries, May 2000. 59. Chaffin,
D.; Freivalds, A.; Evans, S. On the validity of an
isometric biomechanical model of worker strength. IIE
Trans. 1987, 19, 280–288. 60. Chaffin, D.; Erig, M.
Three-dimensional biomechanical static strength prediction
model sensitivity to postural and anthropometric
inaccuracies. IIE Trans. 1991, 23, 215–227. 61. Chaffin,
D.B. Static Biomechanical Modeling in Manual Lifting. In
The Occupational Ergonomics Handbook; Karwowski, W.,
Marras, W.S., Eds.; CRC Press: New York, 1999; 933–944. 62.
Garg, A. Occupational Biomechanics and Low-Back Pain. In
Ergonomics: Low-Back Pain, Carpal Tunnel Syndrome, and
Upper Extremity Disorders in the Workplace; Moore, J.S.,
Garg, A., Eds.; Occupational Medicine: State of the Art
Reviews, Hanley & Belfus, Inc.: Philadelphia, PA, 1992;
609–611. 63. Craig, B.; Fillyaw, J.; Joubert, B. In
Analysis of Buttock-Thigh Pressure of Five High End
Ergonomic Chairs: A Field Study, Proceedings of the
Institute of Industrial Engineers Research Conference,
Institute of Industrial Engineers: Norcross, GA, May 2002.
64. Kroemer, K.; Kroemer, H.; Kroemer-Elbert, K.
Ergonomics: How to Design for Ease and Efficiency; Prentice
Hall: Englewood Cliffs, NJ, 2000; 404–440. 65. Eastman
Kodak Company. Ergonomic Design for People at Work; Van
Nostrand Reinhold: New York, 1983; Vol. 1, 12–63. 66.
Niebel, B.; Freivalds, A. Methods, Standards, and Work
Design, 11th Ed.; McGraw-Hill: Boston, 2003; 187–195. 67.
American National Standard for Human Factors Engineering of
Visual Display Terminal Workstations (ANSI/ HFS 100-1988);
Human Factors Society, Ed.; Human Factors Society: Santa
Monica, CA, 1988. 68. Human Factors Engineering of Computer
Workstations (BSR/HFES 100-2002), Draft Standard for Trial
Use; Human Factors Society, Ed.; Human Factors Society:
Santa Monica, CA, 2002.
Excitable Tissue, Electrical Stimulation
Of

45. McCreery, D.B.; Agnew, W.F.; Bullara, L.A. The effects


of prolonged intracortical microstimulation on the
excitability of pyramidal tract neurons in the cat. Ann.
Biomed. Eng. 2002, 30 (1), 107–109.

46. Naples, G.G.; Mortimer, J.T.; Yuen, T.G.H. Overview of


peripheral nerve electrode design and implantation. In
Neural Prostheses: Fundamental Studies; Agnew, W.F.,
McCreery, D.B., Eds.; Prentice Hall: Englewood Cliffs, NJ,
1990; 107–145.

47. Nannini, N.; Horch, K. Muscle recruitment with


intrafascicular electrodes. IEEE Trans. Biomed. Eng. 1991,
38 (8), 769–776.

48. Rutten, W.L.; van Wier, H.J.; Put, J.H. Sensitivity and
selectivity of intraneural stimulation using a silicon
electrode array. IEEE Trans. Biomed. Eng. 1991, 38 (2),
192–198.

49. Naples, G.G.; Mortimer, J.T.; Scheiner, A.; Sweeney,


J.D. A spiral nerve cuff electrode for peripheral nerve
stimulation. IEEE Trans. Biomed. Eng. 1988, 35 (11),
905–916.

50. Choi, A.Q.; Cavanaugh, J.K.; Durand, D.M. Selectivity


of multiple-contact nerve cuff electrodes: a simulation
analysis. IEEE Trans. Biomed. Eng. 2001, 48 (2), 165– 172.

51. Veraart, C.; Grill, W.M.; Mortimer, J.T. Selective


control of muscle activation with a multipolar nerve cuff
electrode. IEEE Trans. Biomed. Eng. 1993, 40 (7), 640–653.

52. Tyler, D.J.; Durand, D.M. A slowly penetrating


interfascicular nerve electrode for selective activation of
peripheral nerves. IEEE Trans. Rehabil. Eng. 1997, 5,
51–61.

53. Branner, A.; Stein, R.B.; Normann, R.A. Selective


stimulation of cat sciatic nerve using an array of
varying-length microelectrodes. J. Neurophysiol. 2001, 85
(4), 1585–1594.

54. Tyler, D.J.; Durand, D.M. Functionally selective


peripheral nerve stimulation with a flat interface nerve
electrode. IEEE Trans. Neural Syst. Rehabil. Eng. 2002, 10
(4), 294–303.
55. McCreery, D.B.; Agnew, W.F.; Yuen, T.G.; Bullara, L.A.
Damage in peripheral nerve from continuous electrical
stimulation: comparison of two stimulus waveforms. Med.
Biol. Eng. Comput. 1992, 30 (1), 109–114.

56. Popovic, D.; Gordon, T.; Rafuse, V.F.; Prochazka, A.


Properties of implanted electrodes for functional
electrical stimulation. Ann. Biomed. Eng. 1991, 19 (3),
303–316. 57. Myklebust, J.B.; Cusick, J.F.; Sances, A.J.;
Larson, S.J. Neural Stimulation. RC Press, Boca Raton,
1985. 58. Caldwell, C.W.; Reswick, J.B. A percutaneous wire
electrode for chronic research use. IEEE Trans. Biomed.
Eng. 1975, 22 (5), 429–432. 59. Grandjean, P.A.; Mortimer,
J.T. Recruitment properties of monopolar and bipolar
epimysial electrodes. Ann. Biomed. Eng. 1986, 14 (1),
53–66. 60. Mortimer, J.T.; Kaufman, D.; Roessman, U.
Intramuscular electrical stimulation: tissue damage. Ann.
Biomed. Eng. 1980, 8 (3), 235–244. 61. Malmivuo, J.;
Plonsey, R. Bioelectromagnetism; 1995. 62. Basser, P.J.;
Wijesinghe, R.S.; Roth, B.J. The activating function for
magnetic stimulation derived from a threedimensional volume
conductor model. IEEE Trans. Biomed. Eng. 1992, 39 (11),
1207–1210. 63. Roth, B.J.; Basser, P.J. A model of the
stimulation of a nerve fiber by electromagnetic induction.
IEEE Trans. Biomed. Eng. 1990, 37 (6), 588–597. 64.
Nagarajan, S.S.; Durand, D.M.; Hsuing-Hsu, K. Mapping
location of excitation during magnetic stimulation: effects
of coil position. Ann. Biomed. Eng. 1997, 25 (1), 112–125.
65. Neurotech Business Report. The Market of
Neurotechnology. 2004–2008, 2004. 66. Donoghue, J.P.
Connecting cortex to machines: recent advances in brain
interfaces. Nat. Neurosci. 2002, 5 (suppl), 1085–1088. 67.
Leventhal, D.K.; Durand, D.M. Chronic measurement of the
stimulation selectivity of the flat interface nerve
electrode. IEEE Trans. Biomed. Eng. 2004, 51 (9),
1649–1658. 68. McDonnall, D.; Clark, G.A.; Normann, R.A.
Selective motor unit recruitment via intrafascicular
multielectrode stimulation. Can. J. Physiol. Pharmacol.
2004, 82 (8–9), 599–609. 69. Lawrence, S.M.; et al. Acute
peripheral nerve recording characteristics of polymer-based
longitudinal intrafascicular electrodes. IEEE Trans. Neural
Syst. Rehabil. Eng. 2004, 12 (3), 345–348. 70. Yoo, P.B.;
Durand, D.M. Selective Fascicular Recording of the
Hypoglossal Nerve Using a MultiContact Nerve Cuff
Electrode. Annals of Bioemdical Engineering 2003, 32,
511–519. 71. Lertmanorat, Z.; Durand, D.M. A novel
electrode array for diameter-dependent control of axonal
excitability: a simulation study. IEEE Trans. Biomed. Eng.
2004, 51 (7), 1242–1250.
Extracellular Matrix Scaffolds

11. Roberts, R.; Gallagher, J.; Spooncer, E.; Allen, T.D.;


Bloomfield, F.; Dexter, T.M. Heparan sulphate bound growth
factors: A mechanism for stromal cell mediated
haemopoiesis. Nature 1988, 332, 376–378.

12. Sjaastad, M.D.; Nelson, W.J. Integrin-mediated calcium


signaling and regulation of cell adhesion by intracellular
calcium. BioEssays 1997, 19, 47–55.

13. Pierschbacher, M.D.; Ruoslahti, E. Cell attachment


activity of fibronectin can be duplicated by small synthetic
fragments of the molecule. Nature 1984, 309, 30–33.

14. Yamada, K.M.; Kennedy, D.W. Dualistic nature of


adhesive protein function: Fibronectin and its biologically
active peptide fragments can autoinhibit fibronectin
function. J. Biol. Chem. 1984, 99, 29–36.

15. Humphries, M.J.; Mould, A.P.; Yamada, K.M. Matrix


Receptors in Cell Migration. In Receptors for Extracellular
Matrix; McDonald, J.A., Mecham, R.P., Eds.; Academic Press,
1991; 195–253.

16. Badylak, S.F.; Kropp, B.T.; McPherson, H.L.; Snyder,


P.W. SIS: A rapidly resorbable bioscaffold for augmentation
cystoplasty in a dog model. Tissue Eng. 1998, 4, 379–387.

17. Rickey, F.A.; Elmore, D.; Hillegonds, D.; Badylak, S.;


Record, R.; Simmons-Byrd, A. Regeneration of tissue about
an animal-based scaffold: AMS studies of the fate of the
scaffold. Nucl. Instrum. Methods Phys. Res. 2000, 172,
904–909.

18. Vanderrest, M.; Garrone, R. Collagen family of


proteins. FASEB J. 1991, 5, 2814–2823.

19. Schwarzbauer, J.E. Basement membranes: Putting up the


barriers. Curr. Biol. 1999, 9, R242–R244.

20. Timpl, R. Macromolecular organization of basement


membranes. Curr. Opin. Cell Biol. 1996, 8, 618–624.

21. Timpl, R.; Brown, J. Supramolecular assembly of


basement membranes. BioEssays 1996, 18, 123–132.

22. Ponce, M.; Nomizu, M.; Delgado, M.C.; Kuratomi, Y.;


Hoffman, M.P.; Powell, S.; Yamada, Y.; Kleinman, H.K.;
Malinda, K.M. Identification of endothelial cell binding
sites on the laminin gamma-1 chain. Circ. Res. 1999, 84,
688–694.

23. Werb, Z.; Cu, T.H.; Rinkenberger, J.L.; Coussens, L.M.


Matrix-degrading proteases and angiogenesis during
development and tumor formation. Acta Pathol. Microbiol.
Immunol. Scand. 1999, 107, 11–18.

24. Schwarzbauer, J.E. Fibronectin: From gene to protein.


Curr. Opin. Cell Biol. 1991, 3, 786–791.

25. Miyamoto, S.; Katz, B.Z.; Lafrenie, R.M.; Yamada, K.M.


Fibronectin and integrins in cell adhesion, signaling, and
morphogenesis. Ann. N.Y. Acad. Sci. 1998, 857, 119–129.

26. McPherson, T.B.; Badylak, S.F. Characterization of


fibronectin derived from porcine small intestinal submucosa.
Tissue Eng. 1998, 4, 75–83.

27. Hodde, J.; Record, R.; Tullius; Badylak, S. Fibronectin


peptides mediate HMEC adhesion to porcine-derived
extracellular matrix. Biomaterials 2002, 23, 1841–1848. 28.
Gechtman, Z.; Belleli, A.; Lechpammer, S.; Shaltiel, S. The
cluster of basic amino acids in vitronectin contributes to
its binding of plasminogen activator inhibitor-1: Evidence
from thrombin-, elastase-, and plasmin-cleaved vitronectins
and anti-peptides and antibodies. Biochem. J. 1997, 325,
339–349. 29. Varner, J.A.; Cheresh, D.A. Integrins and
cancer. Curr. Opin. Cell Biol. 1996, 8, 724–730. 30.
Hammes, H.P.; Brownlee, M.; Jonczyk, A.; Sutter, A.;
Preissner, K.T. Subcutaneous injection of a cyclic peptide
antagonist of vitronectin receptor-type integrins inhibits
retinal neovascularization. Nat. Med. 1996, 2, 529–533. 31.
Hodde, J.P.; Badylak, S.F.; Brightman, A.O.; VoytikHarbin,
S.L. Glycosaminoglycan content of small intestinal
submucosa: A bioscaffold for tissue replacement. Tissue
Eng. 1996, 2, 209–217. 32. Badylak, S.F. Modification of
Natural Polymers: Collagen. In Methods of Tissue
Engineering; Atala, A., Lanza, R.P., Eds.; Academic Press,
2002; 505–514. Chapter 42. 33. Aiken, S.W.; Badylak, S.F.;
Toombs, J.P.; Shelbourne, K.D.; Hiles, M.C.; Lantz, G.C.;
Van Sickle, D. Small intestinal submucosa as an
intra-articular ligamentous repair material: A pilot study
in dogs. Vet. Comp. Orthop. Traumatol. 1994, 7, 124–128.
34. Badylak, S.F.; Arnoczky, S.; Plouhar, P.; Haut, R.;
Mendenhall, V.; Horvath, C. Naturally-occurring ECMs as
scaffolds for musculoskeletal repair. Clin.Orthop. Relat.
Res. 1999, 367S, S333–S343. 35. Kropp, B.P.; Sawyer, B.D.;
Shannon, H.E.; Ripy, M.K.; Badylak, S.F.; Adams, M.C.;
Keating, M.A.; Rink, R.C.; Thor, K.B. Characterization of
small intestinal submucosa-regenerated canine detrusor:
Assessment of reinnervation, in vitro compliance and
contractility. J. Urol. 1996, 156, 599–607. 36. Kropp,
B.P.; Rippy, M.K.; Badylak, S.F.; Adams, M.C.; Keating,
M.A.; Rink, R.C.; Thor, K.B. Regenerative urinary bladder
augmentation using small intestinal submucosa: Urodynamic
and histopathologic assessment in long term canine bladder
augmentations. J. Urol. 1996, 155, 2098–2104. 37. Atala,
A.; Guzman, L.; Retik, A.B. A novel inert collagen matrix
for hypospadias repair. J. Urol. 1999, 162, 1148–1151. 38.
Piechota, H.J.; Dahms, S.E.; Nunes, L.S.; Dahiya, R.; Lue,
T.F.; Tanagho, E.A. In vitro functional properties of the
rat bladder regenerated by the bladder acellular matrix
graft. J. Urol. 1998, 159 , 1717–1724. 39. Piechota, H.J.;
Dahms, S.E.; Probst, M.; Gleason, C.A.; Nunes, L.S.;
Dahiya, R.; Lue, T.F.; Tanagho, E.A. Functional rat bladder
regeneration through xenotransplantation of the bladder
acellular matrix graft. Br. J. Urol. 1998, 81, 548–559. 40.
Piechota, H.J.; Gleason, C.A.; Dahms, S.E.; Dahiya, R.;
Nunes, L.S.; Lue, T.F.; Tanagho, E.A. Bladder acellular
matrix graft: In vivo functional properties of the
regenerated rat bladder. Urol. Res. 1999, 27, 206–213. 41.
Cobb, M.A.; Badylak, S.F.; Janas, W.; Boop, F.A. Histology
after dural grafting with small intestinal submucosa. Surg.
Neurol. 1996, 46, 389–394. 42. Cobb, M.A.; Badylak, S.F.;
Janas, W.; Simmons-Byrd, A.; Boop, F.A. Porcine small
intestinal submucosa as a dural substitute. Surg. Neurol.
1999, 51 (1), 99–104. 43. Sandusky, G.E.; Lantz, G.C.;
Badylak, S.F. Healing comparison of small intestine
submucosa and ePTFE grafts in the canine carotid artery. J.
Surg. Res. 1995, 58, 415–420. 44. Prevel, C.D.; Eppley,
B.L.; McCarty, M.; Harruff, R.; Brock, C.; Badylak, S.F.
Experimental evaluation of small intestine submucosa as a
microvascular graft material. J. Microsurg. 1994, 15,
588–591. 45. Badylak, S.F.; Lantz, G.; Coffey, A.; Geddes,
L.A. Small intestinal submucosa as a large diameter
vascular graft in the dog. J. Surg. Res. 1989, 47, 74–80.
46. Prevel, C.D.; Eppley, B.L.; Summerlin, D.J.; Jackson,
J.R.; McCarty, M.; Badylak, S.F. Small intestinal submucosa
(SIS): Utilization as a wound dressing in fullthickness
rodent wounds. Ann. Plast. Surg. 1995, 35, 381–388. 47.
Hodde, J.P.; Badylak, S.F.; Shelbourne, K.D. The effect of
range of motion on remodeling of small intestinal submucosa
(SIS) when used as an Achilles tendon repair material in
the rabbit. Tissue Eng. 1997, 3, 27–37. 48. Badylak, S.F.;
Park, K.; McCabe, G.; Yoder, M. Marrow-derived cells
populate scaffolds composed of xenogeneic extracellular
matrix. Exp. Hematol. 2001, 29, 1310–1318. 49. Chen, F.;
Yoo, J.J.; Atala, A. A cellular collagen matrix as a
possible ‘‘off the shelf’’ biomaterial for urethral repair.
Urology 1999, 54, 407–410. 50. Dahms, S.E.; Piechota, H.J.;
Dahiya, R.; Gleason, C.A.; Hohenfellner, M.; Tanagho, E.A.
Bladder acellular matrix graft in rats: Its
neurophysiologic properties and mRNA expression of growth
factors TGF-alpha and TGF-beta. Neurourol. Urodyn. 1998,
17, 37–54. 51. Dahms, S.E.; Piechota, H.J.; Dahiya, R.;
Lue; Tanagho, E.A. Composition and biomechanical properties
of the bladder acellular matrix graft: Comparative analysis
in rat, pig and human. Br. J. Urol. 1998, 82, 411–419. 52.
Dahms, S.E.; Piechota, H.J.; Nunes, L.; Dahiya, R.; Lue,
T.F.; Tanagho, E.A. Free ureteral replacement in rats:
Regeneration of ureteral wall components in the acellular
matrix graft. Urology 1997, 50, 818–825. 53. Probst, M.;
Piechota, H.J.; Dahiya, R.; Tanagho, E.A. Homologous
bladder augmentation in dog with the bladder acellular
matrix graft. Br. J. Urol. 2000, 85, 362–371. 54. Yoo,
J.J.; Meng, J.; Oberpenning, F.; Atala, A. Bladder
augmentation using allogenic bladder submucosa seeded with
cells. Urology 1998, 51, 221–225. E
Eye Tracking: Characteristics and Methods

17. Gilliland, A.R. Photographic methods for studying


reading. Visual Educ. 1921, 2, 21.

18. Buswell, G.T. How People Look at Pictures: A Study of


the Psychology and Perception in Art; Univ. Chicago Press:
Oxford, England, 1935; 198.

19. Totten, E. Eye movement during visual imagery. Comp.


Psychol. Monogr. 1935.

20. Perky, C.W. An experimental study of imagination. Am.


J. Psychol. 1910, 21, 422.

21. Stoy, E.G. A preliminary study of ocular attitudes in


thinking of spatial relations. J. of Gen. Psych. 1930.

22. Clark, H. Visual imagery and attention: an analytical


study. Am. J. Psychol. 1916, 27 (4), 461.

23. Goldthwait, C. Relation of eye movements to visual


imagery. Am. J. Psychol. 1933, 45.

24. Tinker, M.A. The study of eye movements in reading.


Psychol. Bull. 1946, 43.

25. Walker, R.Y. Eye-movements of good readers. Proc. Iowa


Acad. of Sci. 1933, 40.

26. Stone, L.G. Reading reactions for varied types of


subject matter: an analytical study of the eye-movements of
college freshmen. J. Exp. Educ. 1941, 10.

27. Jacobson, J.Z.; Dodwell, P.C. Saccadic eye movements


during reading. Brain Lang. 1979, 8 (3), 303.

28. Yarbus, A.L. Role of Eye Movements in the Visual


Process; Nauka: Oxford, England, 1965; 167.

29. Fender, D.H. Contact lens stability. Biomed. Sci.


Instrum. 1964, 2, 43.

30. Byford, G.H. A sensitive contact lens photoelectric eye


movement recorder. IRE Trans. Bio-Med. Electron. 1962, 9,
236.

31. Nayrac, P.; Milbled, G.; Parquet, P.H.J.; Leclerco, M.;


Dhedin, G. Un nouveau proce´de´ d’enregistrement des
mouvements oculairres. Application aux tests de tracking.
Lille Med. 1969, 14 (685–687). 32. Robinson, D. A. A method
of measuring eye movement using a scleral search coil in a
magnetic field. IEEE Trans. Bio-Med. Electron. 1963, 10,
137. 33. Young, L.R. Biomedical Engineering Systems;
Clynes, M., Milsom, J.H., Eds.; McGraw-Hill: New York,
1970. 34. Merchant, J.; Morrissette, R.; Porterfield, J.L.
Remote measurement of eye direction allowing subject motion
over one cubic foot of space. IEEE Trans. Bio-Med.
Electron. 1974, 21 (4), 309. 35. Lambert, R.H.; Monty,
R.A.; Hall, R.J. High-speed data processing and unobtrusive
monitoring of eye movements. Behav. Res. Meth. Instrum.
1974, 6 (6), 525. 36. Cornsweet, T.N.; Crane, H.D. Accurate
two-dimensional eye tracker using first and fourth Purkinje
images. J. Opt. Soc. Am. 1973, 63. 37. Ballard, D.H.;
Hayhoe, M.M.; Pelz, J.B. Memory representations in natural
tasks. J. Cogn. Neurosci. 1995, 7 (1), 66. 38. Land, M.F.;
Lee, D.N. Where do we look when we steer. Nature 1994, 369
(6483), 742. 39. Tanenhaus, M.K.; Spivey Knowlton, M.J.;
Eberhard, K.M.; Sedivy, J.C. Integration of visual and
linguistic information in spoken language comprehension.
Science 1995, 268 (5217), 1632. 40. Reder, S.M. On-line
monitoring of eye-position signals in contingent and
noncontingent paradigms. Behav. Res. Meth. Instrum. 1973,
5. 41. McConkie, G.W.; Rayner, K. The span of the effective
stimulus during a fixation in reading. Percept. Psychophys.
1975, 17 (6), 578. 42. Rayner, K. Parafoveal identification
during a fixation in reading. Acta Psychologica. 1975, 39
(4), 271. 43. McConkie, G.W.; Zola, D.; Wolverton, G.S.;
Burns, D.D. Eye movement contingent display control in
studying reading. Behav. Res. Meth. Instrum. 1978, 10 (2),
154.
Eye Tracking: Research Areas and
Applications

1. Posner, M.I. Orienting of attention. Q. J. Exp. Psychol.


1980, 32 (1), 3.

2. Treue, S. Neural correlates of attention in primate


visual cortex. Trends Neurosci. 2001, 24, 295.

3. Findlay, J.M.; Gilchrist, I.D. Eye Guidance in Reading


and Scene Perception; Underwood, G., Ed.; Elsevier Science,
Ltd.: Oxford, England, 1998; 295–312.

4. Sheliga, B.M.; Riggio, L.; Craighero, L.; Rizzolatti, G.


Spatial attention—Determined modifications in saccade
trajectories. Neuroreport: Int. J. Rapid Commun. Res.
Neurosci. 1995, 6 (3), 585.

5. Corbetta, M. Frontoparietal cortical networks for


directing attention and the eye to visual locations:
Identical, independent, or overlapping neural systems?
Proc. Natl. Acad. Sci. U. S. A. 1998, 95, 831.

6. Shepherd, M.; Findlay, J.M.; Hockey, R.J. The


relationship between eye movements and spatial attention.
Q. J. Exp. Psychol., Human Exp. Psychol. 1986, 38 (3-A),
475. 7. Sheliga, B.M.; Riggio, L.; Rizzolatti, G. Orienting
of attention and eye movements. Exp. Brain Res. 1994, 98
(507–522). 8. Hoffman, J.E.; Subramaniam, B. The role of
visual attention in saccadic eye movements. Percept.
Psychophys. 1995, 57 (6), 787. 9. Robinson, D.A.; Fuchs,
A.F. Eye movements evoked by stimulation of frontal eye
fields. J. Neurophysiol. 1969, 32. 10. Moore, T.; Armstrong,
K.M. Selective gating of visual signals by microstimulation
of frontal cortex. Nature 2003, 421 (6921), 370. 11. Moore,
T.; Fallah, M. Control of eye movements and spatial
attention. Proc. Natl. Acad. Sci. U. S. A. 2001, 98 (3),
1273. 12. Buswell, G.T. How People Look at Pictures: A
Study of the Psychology and Perception in Art; Univ.
Chicago Press: Oxford, England, 1935; 198 pp. 13. Wooding,
D.S. Eye movements of large populations: II. Deriving
regions of interest, coverage, and similarity using fixation
maps. Behav. Res. Meth. Instrum. Comput. 2002, 34 (4), 518.
14. Wooding, D.S.; Muggelstone, M.D.; Purdy, K.J.; Gale,
A.G. Eye movements of large populations: I. Implementation
and performance of an autonomous public eye tracker. Behav.
Res. Meth. Instrum. Comput. 2002, 34 (4), 509. 15.
Henderson, J.M. Human gaze control in real-world scene
perception. Trends Cogn. Sci. 2003, 7, 498. 16. Mannan,
S.K.; Ruddock, K.H.; Wooding, D.S. The relationship between
the locations of spatial features and those of fixations
made during visual examination of briefly presented images.
Spat. Vis. 1996, 10 (3), 165. 17. Mannan, S.K.; Ruddock,
K.H.; Wooding, D.S. Fixation patterns made during brief
examination of two-dimensional images. Perception 1997, 26
(8), 1059. 18. Reinagel, P.; Zador, A.M. Natural scene
statistics at the center of gaze. Network 1999, 10, 314.
19. Li, Z. A saliency map in primary visual cortex. Trends
Cogn. Sci. 2002, 6, 9. 20. Itti, L.; Koch, C. A
saliency—Based search mechanism for overt and covert shifts
of visual attention. Vision Res. 2000, 40 (10–12), 1489.
21. Yarbus, A.L. Role of Eye Movements in the Visual
Process; Nauka: Oxford, England, 1965; 167 pp. 22. Loftus,
G.R.; Mackworth, N.H. Cognitive determinants of fixation
location during picture viewing. J. Exp. Psychol. Hum.
Percept. Perform. 1978, 4 (4), 565. 23. Posner, M.I.;
Rafal, R.D.; Choate, L.S.; Vaughan, J. Inhibition of
return: Neural basis and function. Cogn. Neuropsychol.
1985, 2 (3), 211. 24. Leek, E.C.; Reppa, I.; Tipper, S.P.
Inhibition of return for objects and locations in static
displays. Percept. Psychophys. 2003, 65 (3), 388. 25.
Irwin, D.E. Integrating information across saccadic eye
movements. Curr. Dir. Psychol. Sci. 1996, 5 (3), 94. 26.
Irwin, D.E. Information integration across saccadic eye
movements. Cogn. Psychol. 1991, 23 (3), 420. 27. Peterson,
M.S.; Kramer, A.F.; Wang, R.F.; Irwin, D.E.; McCarley, J.S.
Visual search has memory. Psychol. Sci. 2001, 12 (4), 287.
28. Henderson, J.M.; Hollingworth, A. Global transsaccadic
change blindness during scene perception. Psychol. Sci.
2003, 14 (5), 493. 29. Simons, D.J.; Levin, D. Change
blindness. Trends Cogn. Sci. 1997, 1, 261. 30. Rensink,
R.A.; O’Regan, J.K.; Clark, J. Image flicker is as good as
saccades in making large scene changes invisible.
Perception 1995, 24, 26. 31. O’Regan, J.K.; Deubel, H.;
Clark, J.; Rensink, R.A. Picture changes during blinks:
Looking without seeing and seeing without looking. Vis.
Cognit. 2000, 7. 32. O’Regan, J.K.; Rensink, R.A.; Clark,
J.J. Changeblindness as a result of ‘‘mudsplashes’’. Nature
1999, 398. 33. Bridgeman, B.; Hendry, D.; Stark, L. Failure
to detect displacement of the visual world during saccadic
eye movements. Vis. Res. 1975, 15 (6), 719. 34. Henderson,
J.M.; Hollingworth, A. The role of fixation position in
detecting scene changes across saccades. Psychol. Sci.
1999, 10 (5), 438. 35. Hollingworth, A.; Schrock, G.;
Henderson, J.M. Change detection in the flicker paradigm:
The role of fixation position within the scene. Mem. Cogn.
2001, 29 (2), 296. 36. Nelson, W.W.; Loftus, G.R. The
functional visualfield during picture viewing. J. Exp.
Psychol. Hum. Learn. Mem. 1980, 6 (4), 391. 37. Diefendorf,
A.R.; Dodge, R. An experimental study of the ocular
reactions of the insane from photographic records. Brain
1908, 31, 451–492. 38. Holzman, P.S.; Proctor, L.R.;
Hughes, D.W. Eyetracking patterns in schizophrenia. Science
1973, 181. 39. Holzman, P.S.; et al. Eye-tracking
dysfunctions in schizophrenic patients and their relatives.
Arch. Gen. Psychiat. 1974, 31 (2), 143. 40. McDowell, J.E.;
Clementz, B.A.; Wixted, J.T. Timing and amplitude of
saccades during predictive saccadic tracking in
schizophrenia. Psychophysiology 1996, 33 (1), 93. 41.
Karoumi, B.; Ventre Dominey, J.; Dalery, J. Predictive
saccade behavior is enhanced in schizophrenia. Cognition
1998, 68 (3), B81. 42. Sereno, A.B.; Holzman, P.S.
Antisaccades and smooth pursuit eye movements in
schizophrenia. Biol. Psychiat. 1995, 37 (6), 394. 43.
Gooding, D.C. Antisaccade task performance in
questionnaire—Identified schizotypes. Schizophr. Res. 1999,
35 (2), 157. 44. Fukushima, J.; et al. Disturbances of
voluntary control of saccadic eye movements in
schizophrenic patients. Biol. Psychiat. 1988, 23 (7), 670.
45. Williams, L.M.; Loughland, C.M.; Green, M.J.; Harris,
A.W.F.; Gordon, E. Emotion perception in schizophrenia: An
eye movement study comparing the effectiveness of
risperidone vs. haloperidol. Psychiat. Res. 2003, 120 (1),
13. 46. Shimizu, T.; et al. Comparison of eye-movement
patterns in schizophrenic and normal adults during
examination of facial affect displays. Percept. Motor
Skills 2000, 91 (3, Pt 2), 1045. 47. Radant, A.D.;
Claypoole, K.; Wingerson, D.K.; Cowley, D.S.; Roy Byrne,
P.P. Relationships between neuropsychological and
oculomotor measures in schizophrenia patients and normal
controls. Biol. Psychiatry 1997, 42 (9), 797. 48. Ross,
R.G. Early expression of a pathophysiological feature of
schizophrenia: Saccadic intrusions into smooth-pursuit eye
movements in school-age children vulnerable to
schizophrenia. J. Am. Acad. Child Adolesc. Psychiat. 2003,
42 (4), 468. 49. Rayner, K. Eye movements in reading and
information processing: 20 Years of research. Psychol.
Bull. 1998, 124 (3), 372. 50. Rayner, K. Eye movements in
reading and information processing. Psychol. Bull. 1978, 85
(3), 618. 51. Pollatsek, A.; Rayner, K.; Collins, W.E.
Integrating pictorial information across eye movements. J.
Exp. Psychol. Gen. 1984, 113 (3), 426. 52. Morrison, R.E.;
Rayner, K. Saccade size in reading depends upon character
spaces and not visual angle. Percept. Psychophys. 1981, 30
(4), 395. 53. Carpenter, P.A.; Just, M.A. Eye Movements in
Reading: Perceptual and Language Processes; Rayner, K.,
Ed.; Academic Press: New York, 1983; 275–307. 54. Rayner,
K.; McConkie, G.W. What guides a reader’s eye movements?
Vis. Res. 1976, 16 (8), 829. 55. Kolers, P.A.; Duchnicky,
R.L.; Ferguson, D.C. Eye movement measurement of
readability of CRT displays. Hum. Factors 1981, 23 (5),
517. 56. Morrison, R.E.; Inhoff, A.W. Visual factors and
eye movements in reading. Visible Lang. 1981, 15 (2), 129.
57. Tinker, M.A. Legibility and eye movement in reading.
Psychol. Bull. 1927, 24. 58. Ferreira, F.; Clifton, C. The
independence of syntactic processing. J. Mem. Lang. 1986 ,
25, 348. 59. Rayner, K.; Carlson, M.; Frazier, L. The
interaction of syntax and semantics during sentence
processing: Eye movements in the analysis of semantically
biased sentences. J. Verbal Learn. Verbal Behav. 1983, 22
(3), 358. 60. Trueswell, J.C.; Tanenhaus, M.K.; Kello, C.
Verbspecific constraints in sentence processing: Separating
effects of lexical preference from garden-paths. J. Exp.
Psychol. Learn. Mem. Cogn. 1993, 19 (3), 528. 61. Tinker,
M.A. A photographic study of eye movements in reading
formulae. Gen. Psychol. Monogr. 1928, 3. 62. Rayner, K.;
Sereno, S.C.; Morris, R.K.; Schmauder, A.R. Eye movements
and on-line language comprehension processes. Lang. Cogn.
Processes 1989, 4 (3–4), 21. 63. Jacobson, J.Z.; Dodwell,
P.C. Saccadic eye movements during reading. Brain Lang.
1979, 8 (3), 303. 64. Levy Schoen, A. Eye Movements:
Cognition and Visual Perception; Fisher, D.F., Monty, R.A.,
Senders, J.W., Eds.; Erlbaum: Hillsdale, NJ, 1981; 299–316.
65. Morris, R.K.; Rayner, K.; Pollatsek, A. Eye movement
guidance in reading: The role of parafoveal letter and
space information. J. Exp. Psychol. Hum. Percept. Perf.
1990, 16 (2), 268. E

66. McConkie, G.W.; Rayner, K. The span of the effective


stimulus during a fixation in reading. Percept. Psychophys.
1975, 17 (6), 578.

67. McConkie, G.W.; Rayner, K. Asymmetry of the perceptual


span in reading. Bull. Psychon. Soc. 1976, 8 (5), 365.

68. Rayner, K.; McConkie, G.W.; Zola, D. Integrating


information across eye movements. Cogn. Psychol. 1980, 12
(2), 206.

69. Pollatsek, A.; Bolozky, S.; Well, A.D.; Rayner, K.


Asymmetries in the perceptual span for Israeli readers.
Brain Lang. 1981, 14 (1), 174.

70. Osaka, N.; Oda, K. Effective visual field size necessary


for vertical reading during Japanese text processing. Bull.
Psychon. Soc. 1991, 29 (4), 345.

71. Kennedy, A.; Murray, W.S. Spatial coordinates and


reading: Comments on Monk. Q. J. Exp. Psychol. 1987, 39A,
649.
72. Murray, W.S.; Kennedy, A. Spatial coding in the
processing of anaphor by good and poor readers: Evidence
from eye movement analyses. Q. J. Exp. Psychol. Hum. Exp.
Psychol. 1988, 40 (4-A), 693.

73. Just, M.A.; Carpenter, P.A. A theory of reading: From


eye fixations to comprehension. Psychol. Rev. 1980, 87 (4),
329.

74. Dixon, W.R., Jr. Studies of the eye-movements in


reading of university professors and graduate students.
Microfilm Abstr. 1948, 8.

75. Robinson, F.P. The role of eye movement habits in


determining reading efficiency. Proc. Iowa Acad. Sci. 1932,
39.

76. Eurich, A.C. Fourth-and fifth-grade standards for


photographic eye-movement records. J. Genet. Psychol. 1933,
43.

77. Eurich, A.C. The photographic eye-movement records of


successful and unsuccessful college students. J. Appl.
Psychol. 1933, 17.

78. Miles, W.R.; Bell, H.M. Eye-movement records in the


investigation of study habits. J. Exp. Psychol. 1929, 12.

79. McKane, P.F.; Maples, W.C.; Sellars, P.; McNeil, M. A


comparison of auditory/language therapy with school visual
support procedures in a public school setting. J. Optom.
Vis. Dev. 2001, 32 (2), 83.

80. The Integration of Language, Vision, and Action: Eye


movements and the Visual World; Henderson, J.M., Ferreira,
F., Eds.; Psychology Press: New York, 2004.

81. Tanenhaus, M.K.; Spivey Knowlton, M.J.; Eberhard, K.M.;


Sedivy, J.C. Integration of visual and linguistic
information in spoken language comprehension. Science 1995,
268 (5217), 1632.

82. Allopenna, P.D.; Magnuson, J.S.; Tanenhaus, M.K.


Tracking the time course of spoken word recognition using
eye movements: Evidence for continuous mapping models. J.
Mem. Lang. 1998, 38 (4), 419–439.

83. Spivey, M.J.; Tanenhaus, M.K.; Eberhard, K.M.; Sedivy,


J.C. Eye movements and spoken language comprehension:
Effects of visual context on syntactic ambiguity
resolution. Cognit. Psychol. 2002, 45 (4), 447.

84. Griffin, Z.M.; Bock, K. What the eyes say about


speaking. Psychol. Sci. 2000, 11 (4), 274. 85.
Brown-Schmidt, S.; Campana, E.; Tanenhaus, M.K.
World-Situated Language Processing: Bridging the Language
as Product and Language as Action Traditions; Trueswell,
J.C., Tanenhaus, M.K., Eds.; MIT Press: Cambridge, 2004.
86. Gibson, J.J.; Pick, A.D. Perception of another persons
looking behavior. Am. J. Psychol. 1963, 76 (3), 386. 87.
Kingstone, A.; Smilek, D.; Ristic, J.; Friesen, C.K.;
Eastwood, J.D. Attention, researchers! It is time to take a
look at the real world. Curr. Dir. Psychol. Sci. 2003, 12
(5), 176. 88. Bavelas, J.B.; Coates, L.; Johnson, T.
Listener responses as a collaborative process: The role of
gaze. J Commun. 2002, 52 (3), 566. 89. Bavelas, J.B.;
Chovil, N. Visible acts of meaning—An integrated message
model of language in face-to-face dialogue. J. Lang. Soc.
Psychol. 2000, 19 (2), 163. 90. Clark, H.H. Using Language;
Cambridge University Press: Cambridge, 1996; pp. xi, 432.
91. Irwin, D.E.; Zacks, J.L.; Brown, J.S. Visual memory and
the perception of a stable visual environment. Percept.
Psychophys. 1990, 47 (1), 35. 92. Spivey, M.J.; Geng, J.J.
Oculomotor mechanisms activated by imagery and memory: Eye
movements to absent objects. Psychol. Res./Psychol. Forsch.
2001, 65 (4), 235. 93. Richardson, D.C.; Spivey, M.J.
Representation, space and hollywood squares: Looking at
things that aren’t there anymore. Cognition 2000, 76, 269.
94. Richardson, D.C.; Kirkham, N.Z. Multi-modal events and
moving locations: Evidence for dynamic spatial indexing in
adults and six month olds. J. Exp. Psychol. Gen. 2004, 133
(1), 46–62. 95. Kosslyn, S.M.; Behrmann, M.; Jeannerod, M.
The cognitive neuroscience of mental imagery.
Neuropsychologia 1995, 33 (11), 1335. 96. Kan, I.P.;
Barsalou, L.W.; Solomon, K.O.; Minor, J.K.;
Thompson-Schill, S.L. Role of mental imagery in a property
verification task: fMRI evidence for perceptual
representations of conceptual knowledge. Cogn.
Neuropsychol. 2003, 20 (3–6), 525–540. 97. Barsalou, L.W.
Perceptual symbol systems. Behav. Brain Sci. 1999, 22 (4),
577. 98. Martin, A. Handbook of Functional NeuroImaging of
Cognition; Cabeza, R., Kingstone, A., Eds.; MIT Press:
Cambridge, 2001; 153–186. 99. Totten, E. Eye movement
during visual imagery. Comp. Psychol. Monogr. 1935, 11.
100. Perky, C.W. An experimental study of imagination. Am.
J. Psychol. 1910, 21, 422. 101. Stoy, E.G. A preliminary
study of ocular attitudes in thinking of spatial relations.
J. Gen. Psychol. 1930, 4 102. Clark, H. Visual imagery and
attention: An analytical study. Am. J. Psychol. 1916, 27
(4), 461. 103. Goldthwait, C. Relation of eye movements to
visual imagery. Am. J. Psychol. 1933, 45. 104. Goodenough,
D.R.; Shapiro, A.; Holden, M.; Steinschriber, L. A
comparison of ‘‘dreamers’’ and ‘‘nondreamers’’: Eye
movements, electroencephalograms, and the recall of dreams.
J. Abnorm. Soc. Psychol. 1959, 59. 105. Roffwarg, H.P.;
Dement, W.C.; Muzio, J.N.; Fisher, C. Dream imagery:
Relationship to rapid eye movements of sleep. Arch. Gen.
Psych. 1962, 7. 106. Antrobus, J.S.; Antrobus, J.S. Rapid
eye movements and rapid eye movement periods.
Psychophysiology 1969, 6. 107. Noton, D.; Stark, L.
Scanpaths in eye movements during pattern perception.
Science 1971, 171. 108. Brandt, S.A.; Stark, L.W.
Spontaneous eye movements during visual imagery reflect the
content of the visual scene. J. Cogn. Neurosci. 1997, 9
(1), 27. 109. Spivey, M.J.; Tyler, M.; Richardson, D.C.;
Young, E. Paper presented at the 22nd Annual Conference of
the Cognitive Science Society, Philadelphia, 2000. 110.
Bandler, R.; Grinder, J. The Structure of Magic; Science
and Behavior Books: Palo Alto, CA, 1975; pp. xv, 225. 111.
Gumm, W.B.; Walker, M.K.; Day, H.D. Neurolinguistics
programming: Method or myth? J. Couns. Psychol. 1982, 29
(3), 327. 112. Poffel, S.A.; Cross, H.J. Neurolinguistic
programming: A test of the eye-movement hypothesis.
Percept. Mot. Skills 1985, 61 (3, Pt 2), 1262. 113. Farmer,
A.; Rooney, R.; Cunningham, J.R. Hypothesized eye movements
of neurolinguistic programming: A statistical artifact.
Percept. Mot. Skills 1985, 61 (3, Pt 1), 717. 114. Elich,
M.; Thompson, R.W.; Miller, L. Mental imagery as revealed
by eye movements and spoken predicates: A test of
neurolinguistic programming. J. Counsel. Psychol. 1985, 32
(4), 622. 115. Salas, J.A.; de Groot, H.; Spanos, N.P.
Neuro-linguistic programming and hypnotic responding: An
empirical evaluation. J. Ment. Imag. 1989, 13 (1), 79. 116.
Jupp, J.J. A further empirical evaluation of
neurolinguistic primary representational systems (PRS).
Couns. Psychol. Q. 1989, 2 (4), 441. 117. Shapiro, F. Eye
Movement Desensitization and Reprocessing: Basic
Principles, Protocols, and Procedures; Guilford Press: New
York, NY, 1995; . pp. xviii, 398. . 118. Shapiro, F. EMDR
12 years after its introduction: Past and future research.
J. Clin. Psychol. 2002, 58 (1), 1. 119. Oswalt, R.;
Anderson, M.; Hagstrom, K.; Berkowitz, B. Evaluation of the
one-session eye-movement desensitization reprocessing
procedure for eliminating traumatic memories. Psychol. Rep.
1993, 73 (1), 99. 120. Acierno, R.; Hersen, M.; Van
Hasselt, V.B.; Tremont, G.; Meuser, K.T. Review of the
validation and dissemination of eye-movement
desensitization and reprocessing: A scientific and ethical
dilemma. Clin. Psychol. Rev. 1994, 14 (4), 287. 121.
Forbes, D.; Creamer, M.; Rycroft, P. Eye movement
desensitization and reprocessing in posttraumatic stress
disorder: A pilot study using assessment measures. J.
Behav. Ther. Exp. Psychiatry 1994, 25 (2), 113. 122.
Silver, S.M.; Brooks, A.; Obenchain, J. Treatment of
Vietnam War veterans with PTSD: A comparison of eye
movement desensitization and reprocessing, biofeedback, and
relaxation training. J. Trauma. Stress 1995, 8 (2), 337.
123. Wilson, S.A.; Becker, L.A.; Tinker, R.H. Eye movement
desensitization and reprocessing (EMDR) treatment for
psychologically traumatized individuals. J. Consult. Clin.
Psychol. 1995, 63 (6), 928. 124. Hyer, L.; Brandsma, J.M.
EMDR minus eye movements equals good psychotherapy. J.
Trauma. Stress 1997, 10 (3), 515. 125. Brandt, H.F. Ocular
photography as a scientific approach to the study of the
psychological aspects of seeing. Illum. Eng. Inc. Trans.
Illum. Eng. Soc. N.Y. 1944, 39. 126. Macnamara, J.J. A new
method for testing advertising effectiveness through eye
movement photography. Psychol. Rec. 1941, 4. 127. Karslake,
J.S. The Purdue eye-camera: A practical apparatus for
studying the attention value of advertisements. J. Appl.
Psychol. 1940, 24. 128. Boersema, T.; Zwaga, H.J.G. Visual
Search; Brogan, D., Ed.; Taylor & Francis: Philadelphia,
PA, 1990; 151–157. 129. Lohse, G.L. Consumer eye movement
patterns on yellow pages advertising. J. Advert. 1997, 26
(1), 61. 130. Treistman, J.; Gregg, J.P. Visual, verbal,
and sales responses to print ads. J. Advert. Res. 1979, 19
(4), 41. 131. Gerathewohl, S.J. Eye movements during radar
operations. J. Aviat. Med. 1952, 23. 132. Wallis, D.;
Samuel, J.A. Some experimental studies of radar operating.
Ergonomics 1961, 4. 133. Brues, A.M.; Damon, A. Eye
movement in sighting as related to design of turret
sighting panels; 1946. (AAF, Tech. Rep. 1943; Publ. Bd.,
No. 5183.). 134. Milton, J.L. Analysis of pilots’ eye
movements in flight. J. Aviat. Med. 1952, 23. 135. Kamyshov,
I.A.; Lazarev, V.G. Some features of changes in a pilot’s
fixations in reading instruments during actual flight. Vopr.
Psychologii. 1969, 15. 136. Steinke, T.R. An evaluation of
map design and map reading using eye movement recordings.
Dissert. Abstr. Int. 1980, 40 (8-B), 3639. 137. Haider, E.;
Luczak, H.; Rohmert, W. Ergonomics investigations of
workplaces in a police command-control centre equipped with
TV displays. Appl. Ergon. 1982, 13 (3), 163. 138. Swanston,
M.T.; Walley, C.E. Factors affecting the speed of
acquisition of tabulated information from visual displays.
Ergonomics 1984, 27 (3), 321. 139. Leermakers, M.A.;
Boschman, M.C. Eye movements, performance and visual
comfort using VDTs. IPO Ann. Prog. Rep. 1984, 19. 140.
Moray, N.; Rotenberg, I. Fault management in process
control: Eye movements and action. Ergonomics 1989 , 32
(11), 1319. 141. MacGregor, J.N.; Lee, E. Menu search:
Random or systematic? Int. J. Man–Mach. Stud. 1987, 26 (5),
627. 142. Yamamoto, S.; Kuto, Y. A method of evaluating VDT
screen layout by eye movement analysis. Ergonomics 1992, 35
(5–6), 591. 143. Deffner, G. Eye Movement Research:
Mechanisms, Processes and Applications. Studies in Visual
Information Processing, 6; Findlay, J.M., Walker, R., Eds.;
Elsevier Science: New York, NY, 1995; 479–490. E

144. Menozzi, M. Visual ergonomics of head-mounted


displays. Jpn. Psychol. Res. 2000, 42 (4), 213.

145. Ohno, R.; Hata, T.; Kondo, M. Handbook of Japan United


States Environment Behavior Research: Toward a
Transactional Approach; Wapner, S., Demick, J., Eds.;
Plenum Press: New York, NY, 1997; 163–182.

146. Martin, G. Eye movements as the cause of faulty


steering of airplanes, automobiles, and bicycles. Graefes
Arch. Ophthalmol. 1940, 142.

147. Specht, H. Fatigue and hours of service of interstate


truck drivers. V: Eye movements andrelated phenomena.
Public Health Bulletin (Wash. DC) 1941, 265.

148. Mourant, R.R.; Rockwell, T.H. Mapping eye-movement


patterns to the visual scene in driving: An exploratory
study. Hum. Factors 1970, 12.

149. Mourant, R.R.; Rockwell, T.H. Strategies of visual


search by novice and experienced drivers. Hum. Factors
1972, 14.

150. Robinson, G.H.; Erickson, D.J.; Thurston, G.L.; Clark,


R.L. Visual search by automobile drivers. Hum. Factors
1972, 14.

151. Kito, T.; Haraguchi, M.; Funatsu, T.; Sato, M.; Kondo,
M. Measurements of gaze movements while driving. Percept.
Mot. Skills. 1989, 68 (1), 19.

152. Land, M.F.; Lee, D.N. Where do we look when we steer.


Nature 1994, 369 (6483), 742.

153. Dishart, D.C.; Land, M.F. Eye Guidance in Reading and


Scene Perception; Underwood, G., Ed.; Elsevier Science,
Ltd.: Oxford, England, 1998; 419–429.

154. Underwood, G.; Chapman, P.; Brocklehurst, N.;


Underwood, J.; Crundall, D. Visual attention while driving:
Sequences of eye fixations made by experienced and novice
drivers. Ergonomics 2003, 46 (6), 629.

155. Land, M.F.; Tatler, B.W. Steering with the head: The
visual strategy of a racing driver. Curr. Biol. 2001, 11
(15), 1215.

156. Harbin, G.; Durst, L.; Harbin, D. Evaluation of


oculomotor response in relationship to sports performance.
Med. Sci. Sports Exerc. 1989, 21 (3), 258.

157. Lenoir, M.; Crevits, L.; Goethals, M.; Wildenbeest,


J.; Musch, E. Ate better eye movements an advantage in ball
games? A study of prosaccadic and antisaccadic eye
movements. Percept. Mot. Skills 2000, 91 (2), 546.

158. Trachtman, J.N. The relationship between ocular


motilities and batting average in little leaguers. Am. J.
Opt. Arch. Am. Acad. Opt. 1973, 50.

159. Vickers, J.N. Gaze control in putting. Perception


1992, 21 (1), 117.

160. Land, M.F.; McLeod, P. From eye movements to actions:


How batsmen hit the ball. Nat. Neurosci. 2000, 3 (12),
1340.

161. Land, M.F.; Hayhoe, M. In what ways do eye movements


contribute to everyday activities? Vis. Res. 2001, 41
(25–26), 3559.

162. Ballard, D.H.; Hayhoe, M.M.; Pelz, J.B. Memory


representations in natural tasks. J. Cogn. Neurosci. 1995,
7 (1), 66. 163. Ballard, D.H.; Hayhoe, M.M.; Pook, P.K.;
Rao, R.P.N. Deictic codes for the embodiment of cognition.
Behav. Brain Sci. 1997, 20, 723. 164. Hayhoe, M.M.;
Bensinger, D.G.; Ballard, D.H. Task constraints in visual
working memory. Vis. Res. 1998, 38 (1), 125. 165. Spivey,
M.J.; Richardson, D.C.; Fitneva, S. Interfacing Language,
Vision, and Action; Henderson, J.M., Ferreira, F., Eds.;
Academic Press: San Diego, 2004. 166. Kramer, A.F.;
McCarley, J.S. Oculomotor behaviour as a reflection of
attention and memory processes: Neural mechanisms and
applications to human factors. Theor. Issues Ergon. Sci.
2003, 4 (1–2), 21. 167. Duchowski, A.T. A breadth-first
survey of eye-tracking applications. Behav. Res. Meth.,
Instrum. Comput. 2002, 34 (4), 455. 168. Watson, B.;
Walker, N.; Hodges, L.F.; Worden, A. Managing level of
detail through peripheral degradation: Effects on search
performance with a headmounted display. ACM Trans.
Comput.-Hum. Interact. 1997, 4, 323. 169. Santella, A.;
DeCarlo, D. Abstracted Painterly Renderings Using
Eye-Tracking Data. In Second International Symposium on
Non-Photorealistic Animation and Rendering (NPAR); ACM
Press: New York, NY, U.S.A., 2002. 170. Ohshima, T.;
Yamamoto, H.; Tamura, H. Paper Presented at the VRAIS, Los
Alamitos, CA; 1996. 171. Stampe, D.M.; Reingold, E.M. Eye
Movement Research: Mechanisms, Processes and Applications;
Findlay, J.M., Walker, R., Kentridge, R.W., Eds.; Elsevier:
Amsterdam, 1995; 467–478. 172. Jacob, R.J.K. The use of eye
movements in humancomputer interaction techniques: What you
look at is what you get. ACM Trans. Inf. Syst. 1991, 9 (3),
152. 173. Frey, L.A.; White, K.P.; Hutchinson, T.E.
Eye-gaze word processing. IEEE Trans. Syst. Man Cybern.
1990, 20 (4), 944. 174. Majaranta, P.; Raiha, K.-J. Eye
Tracking Research & Applications: Proceedings of the
Symposium on ETRA 2002; ACM Press: New York, 2002; 15–22.
175. Gips, J.; Dimattia, P.; Curran, F.X.; Olivieri, P.
Interdisciplinary Aspects on Computers Helping People with
Special Needs; Klaus, J., Auff, E., Kremser, W., Zagler,
W., Eds.; Oldenburg: Vienna, 1996. 176. Kahn, D.A.; Heynen,
J.; Snuggs, G.L. Eye-Controlled Computing: The VisionKey
Experience. In Proceedings of the Fourteenth International
Conference on Technology and Persons with Disabilities
(CSUN’99); ACM Press: New York, NY, U.S.A., 1999. 177.
Chapman, J.E. Use of an Eye-Operated Computer System in
Locked-in Syndrome. In Proceedings of the Sixth Annual
International Conference on Technology and Persons with
Disabilities; ACM Press: New York, NY, U.S.A., 1991. 178.
Argyle, M.; Cook, M. Gaze and Mutual Gaze; Cambridge
University Press: Cambridge.
Fabrics

1. Williams, D.F.; Roaf, R. Implants in Surgery; W.B.


Saunders Company: Philadelphia, 1973.

2. Lee, H.; Neville, K. Handbook of Biomedical Plastics;


Pasadena Technology Press, 1971.

3. Nerem, R.; Sambanis, A. Tissue engineering: From biology


to biological substitutes. Tissue Eng. 1995, 1 (1).

4. Principles of Tissue Engineering; Lanza, R.P., Langer,


R., Chick, W.L., Eds.; R.G. Landes Company and Academic
Press, Inc., 1997.

5. Baer, E.; Kastelic, J.; Galeski, A.J. Connect. Tissue


Res. 1978, 6, 11.

6. Ko, F.K. Fiber Reinforced Composites in Medical


Applications. Proceedings of the Fiber Producer Conference,
Clemson University, Oct. 24, 1984; 30–33.

7. Winston, S.M. Viscoelastic Behavior of Fibers under


Physiological Conditions. M.S. Thesis; Cornell University,
1978.

8. Ozer, Z. Synthetic Absorbable Polymers for Implantation.


Textile Engineering Seminar, Philadelphia College of
Textiles and Science, October, 1982.

9. Piller, B. Bulk Yarns—Production, Processing and


Applications; SNTL: Prague, 1973.

10. Ko, F.K. Medical applications for textile structures.


Text. Asia April, 1997, 4.

11. Mechanics of Flexible Fiber Assemblies; Hearle, J.W.S.,


Thwaites, J.J., Amirbayyat, J., Eds.; NATO ASI, Ser. E
Appl. Sci., Sijthoff & Noordhoff 1980; Vol. 38.

12. Hearle, J.W.S.; Grosberg, P.; Backer, S. Structural


Mechanics of Fibers, Yarns, and Fabrics; WileyInterscience,
1969; Vol. 1.

13. Ko, F.K. Engineering Properties of Spider Silk Fibers.


In Natural Fibers, Plastics, and Composites; Wallenberger,
F.T., Weston, N., Eds.; Kluwer Academic, 2000.

14. Goswami, B.G.; Martindale, J.G.; Scardino, F.L. Textile


Yarns, Technology, Structure and Applications; John Wiley
and Sons: New York, 1977; 273–337.

15. Kaswell, E.R. Wellington-Sears Handbook of Industrial


Textiles; Wellington-Sears: New York, 1963.

16. Ko, F.K. Preform architecture for ceramic matrix


composites. Ceram. Bull. February, 1989, 68 (2).

17. Dow, N.F.; Tranfield, G. Preliminary investigations of


feasibility of weaving triaxial fabrics (Dow weave). Tex.
Res. J. 1970, 986–998. 18. Stover, E.R.; Mark, W.C.;
Marfowitz, I.; Mueller, W. Preparation of an
Omniweave-Reinforced CarbonCarbon Cylinder as a Candidate
for Evaluation in the Advanced Heat Shield Screening
Program. AFML TR-70283, Mar. 1979. 19. Herrick, J.W.
Multidimensional Advanced Composites for Improved Impact
Resistance. 10th National SAMPE Technical Conference, Oct.
17–19, 1977. 20. Pastenbaugh, J. Aerospatiale Technology.
3rd Textile Structural Composites Symposium, Philadelphia,
PA, June 1–2, 1988. 21. Bruno, P.S.; Keith, D.O.; Vicario,
A.A., Jr. Automatically woven three dimensional composite
structures. SAMPE Q. July 1986, 17 (4), 10–16. 22. O’Shea,
J. Autoweave : A Unique Automated 3-D Weaving Technology.
3rd Textile Structural Composites Symposium, Philadelphia,
PA, June 1–2, 1988. 23. Fukuta, K.; Aoki, E. 3-D Fabrics
for Structural Composites. 15th Textile Research Symposium,
1986. 24. Fukuta, K.; Aoki, E.; Onooka, R.; Magatsuka, Y.
Application of latticed structural composite materials with
three dimensional fabrics to artificial bones. Bull. Res.
Inst. Polym. Tex. 1982-2, 131, 151–159. 25. Williams, D.J.
New knitting methods offer continuous structures. Adv.
Compos. Eng. Summer 1978, 12–13. 26. Ko, F.K.; Pastore,
C.M.; Yang, J.M.; Chou, T.W. Structure and Properties of
Multidirectional Warp Knit Fabric Reinforced Composites. In
Composites ’86: Recent Advances in Japan and the United
States, Proceedings, Japan-U.S. CCM-III, Tokyo, Kawata, K.,
Umekawa, S., Kobayashi, A., Eds.; 1986; 21–28. 27. Ko,
F.K.; Pastore, C.; Head, A. Atkins and Pearce Handbook of
Industrial Braids; Drexel University, 1989. 28. Cooper,
J.A. Design, Optimization and In Vivo Evaluation of a
Tissue Engineered Anterior Crucitae Ligament Replacement.
In Ph.D. Thesis; Drexel University, 2002. 29. Lyons, J.;
Ko, F. Nanofiber Technology. In Encyclopedia of Nanoscience
and Nanotechnology; Nalwa, H.S., Ed.; American Scientific,
2004; Vol. 1–10. 30. Formhals, A. US Patent 1,975,504, Oct.
2, 1934. 31. Reneker, D.H.; Chun, I. Nanometre diameter
fibres of polymer, produced by electrospinning.
Nanotechnology 1996, 7, 216–223. 32. Norris, I.D; Shaker,
M.M.; Ko, F.K.; MacDiarmid, A.G. Electrostatic Fabrication
of Ultrafine Conducting Fibers: Polyaniline/Polyethylene
Oxide Blends. In Synthetic Metals; Elsevier, 2001. 33.
MacDiarmid, A.G.; Jones, We., Jr.; Norris, I.D.; Gao, J.;
Johnson, A.T., Jr.; Pinto, N.J.; Hone, J.; Han, B.; Ko,
F.K.; Okusaki, H.; Llanguno, M. ElectrostaticallyGenerated
Nanofibers of Electronic Polymers. In Synthetic Metals 10;
Elsevier, 2002. 34. Ko, F.; Gogotsi, Y.; Ali, A.; Naguib,
N.; Ye, H.; yang, G.; Li, C.; Willis, P. Electrospinning of
continuous carbon nanotube-filled nanofiber yarns. Adv.
Mater. July 17, 2003, 15 (14). 35. Li, W.J.; Laurencin,
C.T.; Caterson, E.J; Tuan, R.S.; Ko, F.K. Electrospun
nanofibrous structure: A novel scaffold for bioengineering.
J. Biomed. Mater. Res. March 25, 2002, 58, 613–621. Wiley
Interscience 36. Sukigara, S.; Gandhi, M.; Ayutsede, J.;
Micklus, M.; Ko, F. Regeneration of Bombyx mori silk by
electrospinningPart 1: Processing parameters and geometric
properties. Polymer 2003, 44, 5721–5727. 37. Vacanti, C.A.;
Mikos, A.G. Letter from the Editors. Tissue Eng. 1995, 1
(1). 38. In Ko, F.K.; Laurencin, C.T.; Borden, M.D.;
Reneker, D. In The Dynamics of Cell-Fiber Architecture
Interaction, Proceedings, Annual Meeting, Biomaterials
Research Society: San Diego, April 1998. 39. Fertala, A.;
Han, W.B.; Ko, F.K. Mapping critical sites in collagen II
for rational design of gene-engineered proteins for
cell-supporting materials. J. Biomed. Mater. Res. 2001, 57,
48–51. 40. Boland, E.D.; Simpson, D.G.; Wnek, G.E.; Bowlin,
G.L. Electrospinning of biopolymers for tissue engineering
scaffolds. Polym. Prepr. 2003, 44 (2), 92. F
Fiber Optic Biosensors

1. Monk, D.; Walt, D. Optical fiber-based biosensors. Anal.


Bioanal. Chem. 2004, 379 (7–8), 931–945.

2. Wolfbeis, O. Fiber-optic chemical sensors. Anal. Chem.


2004, 76 (12), 3269–3284.

3. Marazuela, M.; Moreno-Bondi, M. Fiber-optic


biosensors-an overview. Anal. Bioanal. Chem. 2002, 372
(5–6), 664–682.

4. Lee, B. Review of the present status of optical fiber


sensors. Opt. Fiber Technol. 2003, 9 (1), 57–79.

5. Paddle, B. Biosensors for chemical and biological agents


of defence interest. Biosens. Bioelectron. 1996, 11 (11),
1079–1113.

6. Mehrvar, M.; Bis, C.; Scharer, J.; Young, M.; Luong, J.


Fiber-optic biosensors—trend and advances. Anal. Sci. 2000,
16 (5), 677–692.

7. David, D.; Wilson, M.; Ruffin, D.S. Direct measurement of


ammonia in ambient air. Anal. Lett. 1976, 9 (3), 389–393.
8. Raichlin, Y.; Fel, L.; Katzir, A. Evanescent-wave
infrared spectroscopy with flattened fibers as sensing
elements. Opt. Lett. 2003, 28 (23), 2297–2299. 9. Hocde,
S.; Loreal, O.; Sire, O.; Boussard-Pledel, C.; Bureau, B.;
Turlin, B.; Keirsse, J.; Leroyer, P.; Lucas, J. Metabolic
imaging of tissues by infrared fiber-optic spectroscopy: an
efficient tool for medical diagnosis. J. Biomed. Opt. 2004,
9 (2), 404–407. 10. Jensen, J.; Hoiby, P.; Pedersen, L.H.;
Carlsen, N.L.; Bjarklev, A.; Hansen, T. Evanescent wave
sensing using a hollow-core photonic crystal fiber.
Proceedings SPIE 5317, San Jose, CA, 2004; 139–146. 11.
Ilev, I.; Waynant, R.; Byrnes, K.; Anders, J. On-off laser
delivery into a precise tissue area using smart
tissue-activated fiber probes. IEEE J. Sel. Top. Quantum
Electron. 2003, 9 (2), 331–336. 12. Ilev, I.; Waynant, R.
All-fiber-optic sensor for liquid level measurement. Rev.
Sci. Instrum. 1999, 70 (10), 2551–2555. 13. Zhang, Y.;
Shibru, H.; Cooper, K.; Wang, A. Miniature fiber-optic
multicavity Fabry–Perot interferometric biosensor. Opt.
Lett. 2005, 30 (9), 1021–1023. 14. Bhatia, V.; Campbell,
D.; Claus, R.; Vengsarkar, A. Simultaneous strain and
temperature measurement with long-period gratings. Opt.
Lett. 1997, 22 (9), 648–650. 15. Han, Y.; Lee, S.; Kim, C.;
Kang, J.; Paek, U.; Chung, Y. Simultaneous measurement of
temperature and strain using dual long-period fiber grating
with controlled temperature and strain sensitivities. Opt.
Express 2003, 11 (5), 476–481. 16. Tan, W.; Shi, Z-Y.;
Kopelman, R. Development of submicron chemical fiber optic
sensors. Anal. Chem. 1992, 64 (10), 2985–2990. 17. Tan, W.
Optical measurements on the nanometer scale. Trends Anal.
Chem. 1998, 17, 501–513. 18. Barker, S.; Thorsrud, B.;
Kopelman, R. Nitriteand chloride-selective fluorescent
nano-optodes and in vitro applications to rat conceptuses.
Anal. Chem. 1998, 70, 100–104. 19. Niedre, M.; Patterson,
M.; Wilson, B. Direct near-infrared luminescence detection
of singlet oxygen generated by photodynamic therapy in
cells in vitro and tissues in vivo. Photochem. Photobiol.
2002, 75 (4), 382–391. 20. Ilev, I.; Waynant, R.; Byrnes,
K.; Anders, J. Dualconfocal fiber-optic method for absolute
measurement of refractive index and thickness of optically
transparent media. Opt. Lett. 2002, 27, 1693–1695. 21.
Ilev, I.; Waynant, R. A simple submicron confocal
microscope with a fiber-optic output. Rev. Sci. Instrum.
2000, 71, 4161–4164. F
Fibrin

3. Doolittle, R.F.; Yang, Z.; Mochalkin, I. Crystal


structure studies on fibrinogen and fibrin. Ann. N.Y. Acad.
Sci. 2001, 936, 31–43.

4. Morawitz, P. Die chemie der blutgerinnung (The chemistry


of blood coagulation). Erg. Physiol. 1905, 4, 307–422.

5. Bergel, S. Ueber die wirkungen des fibrins (About the


effects of fibrin). Dtsch. Med. Wochenschr. 1909, 35,
663–665.

6. Seelich, T. Tissucol R (Immuno, Vienna): Biochemistry


and methods of application. J. Head Neck Pathol. 1982, 3,
65–69.

7. Bering, E.A., Jr. Chemical, clinical, and immunological


studies on the products of human plasma fractionation. XX.
The development of fibrin foam as hemostatic agent and for
use in conjunction with human thrombin. J. Clin. Invest.
1944, 23, 586–590.

8. Gerenda´s, M. Fibrin Products as Aids in Hemostasis and


Wound Healing. In Fibrinogen; Laki, K., Ed.; Marcel Dekker,
Inc.: New York, 1968; 277–316.

9. Board, P.G.; Losowsky, M.S.; Miloszewski, K.J.A. Factor


XIII: Inherited and acquired deficiency. Blood Rev. 1993, 7,
229–242.

10. Matras, H.; Dinges, H.P.; Lassmann, J.; Mamoli, B. Zur


nahtlosen interfaszikula¨ren nerventransplantation im
tierexperiment (Regarding the sutureless interfascicular
nerve transplantation in animal experiments). Wien. Med.
Wochenschr. 1972, 122, 517–523.

11. Odar, J. Die Physiologische Gewebeklebung mit


Fibrin—Prinzip, Einsatzgebiete, Klebetechniken, Kosten und
Nutzen (Physiological Gluing of Tissues with
Fibrin—Principle, Indications, Gluing Techniques, Costs and
Benefits). In Techniken und Methoden der Modernen Medizin
(Techniques and Methods of Modern Medicine); Odar, J., Ed.;
Steinkopff Verlag: Darmstadt, 1990; 75–84.

12. Jackson, M.R.; MacPhee, M.J.; Drohan, W.N.; Alving, BM.


Fibrin sealant: Current and potential clinical
applications. Blood Coagul. Fibrinolysis 1996, 7, 737–746.

13. Blomback, B. Fibrinogen and fibrin—Proteins with complex


roles in hemostasis and thrombosis. Thromb. Res. 1996, 83
(1), 1–75.

14. Mosesson, M.W.; DiOrio, J.P.; Muller, M.F.; Shainoff,


J.R.; Siebenlist, K.R.; Amrani, D.L.; Homandberg, G.A.;
Soria, J.; Soria, C.; Samama, M. Studies on the
ultrastructure of fibrin lacking fibrinopeptide B (b-FIBRIN).
Blood 1987, 69 (4), 1073–1081.

15. Ferry, J.D.; Morrison, P.R. Preparation and properties


of serum and plasma proteins VIII. The conversion of human
fibrinogen to fibrin under various conditions. J. Am. Chem.
Soc. 1947, 69, 388–400.

16. Weisel, J.W. Fibrin assembly. Lateral aggregation and


the role of the two pairs of fibrinopeptides. Biophys. J.
1986, 50, 1079–1093.

17. Amrani, D.L.; DiOrio, J.P.; Delmotte, Y. Wound healing.


Role of commercial fibrin sealants. Ann. N.Y. Acad. Sci.
2001, 936, 566–579.

18. Redl, H.; Schlag, G.; Dinges, H.P. Vergleich zweier


fibrinkleber. EinfluX ionischer zusa¨tze auf fibrinstruktur
sowie morphologie und wachstum menschlicher fibroblasten
(Comparison of two fibrin sealants: Effect of ionic
additives on fibrin structure as well as on morphology and
growth of human fibroblasts). Medwelt 1985, 36, 769–776. 19.
Schlag, G.; Redl, H.; Turnher, M.; Dinges, H.P. The
Importance of Fibrin in Wound Repair. In Fibrin Sealant in
Operative Medicine Volume 1 Otorhinolaryngology; Schlag,
G., Redl, H., Eds.; SpringerVerlag: Berlin, 1986; 3–12. 20.
Khare, A.; Woo, L.; Mclean, A.; Stewart, J.E.; DiOrio,
J.P.; Amrani, D.A.; Helgerson, S. Mechanical
characterization of fibrin gels. Blood Coagul. Fibrinolysis
1998, 9 (7), 105. 21. Roberts, W.W.; Lorand, L.; Mockros,
L.F. Viscoelastic properties of fibrin clots. Biorheology
1973, 10, 29–42. 22. Redl, H.; Schlag, G.; Dinges, H.;
Kuderna, H.; Seelich, T. Background and Methods of Fibrin
Sealing. In Biomaterials; Winter, D., Gibbons, D.F., Plenk,
H., Jr., Eds.; John Wiley and Sons: New York, 1982;
669–676. 23. Carr, M.E.; Gabriel, D.A.; McDonagh, J.
Influence of factor XIII and fibronectin on fiber size and
density in thrombin-induced fibrin gels. J. Lab. Clin. Med.
1987, 110 (6), 747–752. 24. Martin, P. Wound healing—Aiming
for perfect skin regeneration. Science 1997, 276, 75–81.
25. Clark, R.F. Fibrin and wound healing. Ann. N.Y. Acad.
Sci. 2001, 936, 355–367. 26. Martinez, J.; Ferber, A.;
Bach, T.L.; Yaen, C.H. Interaction of fibrin with
VE–Cadherin. Ann. N.Y. Acad. Sci. 2001, 936, 386–405. 27.
Stukova, S.M. Thrombin as a regulator of inflammation and
reparative processes in tissues. Biochemistry (Mosc.) 2001,
66, 14–27. 28. Danen, E.H.J.; Yamada, K.M. Fibronectin,
integrins, and growth control. J. Cell. Physiol. 2001, 189,
1–13. 29. Sahni, A.; Francis, C.W. Vascular endothelial
growth factor binds to fibrinogen and fibrin and stimulates
endothelial cell proliferation. Blood 2000, 96, 3772–3778.
30. Albes, J.M.; Klenzner, T.; Kotzerke, J.; Thiedemann,
K.U.; Schafers, H.-J.; Borst, H.-G. Improvement of tracheal
autograft revascularization by means of fibroblast growth
factor. Ann. Thorac. Surg. 1994, 57, 444–449. 31. Hasimoto,
J.; Kurosaka, M.; Yoshiya, S.; Hirohata, K. Meniscal repair
using fibrin sealant and endothelial cell growth factor. Am.
J. Sports Med. 1992, 20, 537–541. 32. MacPhee, M.J.; Singh,
M.P.; Brady, R., Jr.; Akhyani, N.; Liau, G.; Lasa, C., Jr.;
Hue, C.; Best, A.; Drohan, W. Fibrin Sealant: A Versatile
Delivery Vehicle for Drugs and Biologics. In Surgical
Adhesives and Sealants Current Technology and Applications;
Sierra, D.H., Saltz, R., Eds.; Techomic Publishing Co.:
Lancaster, PA, 1996; 109–120. 33. Schense, J.C.; Bloch, J.;
Aebischer, P.; Hubbell, J.A. Enzymatic incorporation of
bioactive peptides into fibrin matrices enhances neurite
extension. Nat. Biotechnol. 2000, 18, 415–419. 34. Andree,
C.; Voigt, M.; Wenger, A.; Erichsen, T.; Bittner, K.;
Schaefer, D.; Walgenbach, K.J.; Borges, J.; Horch, R.E.;
Eriksson, E.; Stark, G.B. Plasmid gene delivery to human
keratinocytes through a fibrinmediated transfection system.
Tissue Eng. 2001, 7, 757–766. 35. Hunyadi, J.; Farkas, B.;
Bertenyi, C.; Olah, J.; Dobozy, A. Keratinocyte grafting: A
new means of transplantation for full-thickness wounds. J.
Dermatol. Surg. Oncol. 1988, 14, 75–78. 36. Horch, R.;
Bannasch, H.; Kopp, J.; Andree, C.; Stark, G.B. Single-cell
suspension of cultured human keratinocytes in fibrin-glue
reconstitute the epidermis. Cell Transplant 1998, 7,
309–317. 37. Pellegrini, G.; Ranno, R.; Stracuzzi, G.;
Bondanza, S.; Guerra, L.; Zambruno, G.; Micali, G.; de
Luca, M. The control of epidermal stem cells (holoclones)
in the treatment of massive full-thickness burns with
autologous keratinocytes cultured on fibrin. Transplantation
1999, 68, 868–879. 38. Gorodetsky, R.; Clark, R.A.F.; An,
J.; Gailit, J.; Levdansky, L.; Vexler, A.; Berman, E.;
Marx, G. Fibrin microbeads (FMB) as biodegradable carriers
for culturing cells and for accelerating wound healing. J.
Invest. Dermatol. 1999, 112, 866–872. 39. Wechselberger,
G.; Schoeller, T.; Stenzl, A.; Ninkovic, M.; Lille, S.;
Russell, R.C. Fibrin glue as delivery vehicle for
autologous urothelial cell transplantation onto a
prefabricated pouch. J. Urol. 1998, 160, 583–586. 40. Bach,
A.D.; Bannasch, H.; Galla, T.J.; Bittner, K.M.; Stark, G.B.
Fibrin glue as matrix for cultured autologous urothelial
cells in urethral reconstruction. Tissue Eng. 2001, 7,
45–53. 41. Wechselberger, G.; Russell, RC.; Neumeister,
M.W.; Schoeller, T.; Piza-Katzer, H.; Rainer, C. Successful
transplantation of three tissue-engineered cell types using
capsule induction technique and fibrin glue as a delivery
vehicle. Plast. Reconstr. Surg. 2002, 110, 123–129. 42. van
Susante, J.; Buma, P.; Schuman, L.; Homminga, G.; van den
Berg, W.; Veth, R. Resurfacing potential of heterologous
chondrocytes suspended in fibrin glue in large
full-thickness defects of femoral articular cartilage: An
experimental study in the goat. Biomaterials 1999, 20,
1167–1175. 43. Ye, Q.; Zu¨nd, G.; Benedikt, P.;
Jockenhoevel, S.; Hoerstrup, S.P.; Sakyama, S.; Hubbell,
J.A.; Turina, M. Fibrin gel as a three-dimensional matrix
in cardiovascular tissue engineering. Eur. J.
Cardio-Thorac. Surg. 2000, 17, 587–591. 44. Tholpady, S.S.;
Sclosser, R.; Spotnitz, W.; Ogle, R.C.; Lindsey, W.H.
Repair of osseus facial critical-size defect using
augmented fibrin sealant. Laryngoscope 1999, 109, 1585–1588.
45. Daculsi, G.; Bagot d’Arc, M.; Cottreal, M. Calcium
Phosphate Ceramics (Triosite s ) in Otology: Evaluation in
Animals and Applications in Humans. In Fibrin Sealing in
Surgical and Nonsurgical Fields, Wound Healing (1); Schlag,
G., Redl, H., Eds.; Springer Verlag: Berlin, 1994; 121–126.
46. Bagot d’Arc, M.; Corlieu, P.; Daculsi, G. Mastoid
Cavity Filling for Bone Reconstruction with a Mixture of
Fibrin Glue and Ceramic Granules. In Fibrin Sealing in
Surgical and Nonsurgical Fields, Neurosurgery, Ophtalmic
Surgery, ENT (5); Schlag, G., Redl, H., Eds.; Springer
Verlag: Berlin, 1994; 174–179. 47. Hench, L.H.; Polak, J.M.
Third-generation biomedical materials. Science 2002, 295,
1014–1017. F
Fibrin Sealants

1. Morawitz, P. Die Chemie der Blutgerinnung (The chemistry


of blood coagulation). Erg. Physiol. 1905, 4, 307–422.

2. Monroe, D.M.; Hoffman, M.; Roberts, H.R. Platelets and


thrombin generation. Arterioscler. Thromb. Vasc. Biol.
2002, 22, 1381–1389.

3. Bergel, S. U¨ber Wirkungen des Fibrins. Dtsch.


Wochenschr. 1909, 35, 633–665.

4. Grey, E.G. Fibrin as a hemostatic in cerebral surgery.


Surg. Gynecol. Obstet. 1915, 21, 452–454.

5. Seddon, H.J.; Medawar, P.B. Fibrin sutures of human


nerves. Lancet 1942, ii, 87–92.

6. Cronkite, E.P.; Lozner, E.L.; Deaver, J. Use of thrombin


and fibrinogen in skin grafting. JAMA 1944, 124, 976–978.

7. Kram, H.B.; Nathan, R.C.; Mackabee, J.R.; Klein, S.R.;


Shoemaker, W.C. Clinical use of nonautologous fibrin glue.
Am. Surg. 1988, 54, 570–573.

8. Dvorak, J.F.; Harvey, V.S.; Estrella, P.; Brown, L.F.;


McDonagh, J.; Dvorak, A.M. Fibrin containing gels induce
angiogenesis. Implications for tumor stroma generation and
wound healing. Vox Sang. 1987, 57, 673–686.

9. Spotnitz, W.D.; Mintz, P.D.; Avery, N.; Bithell, T.C.;


Kaul, S.; Nolan, S.P. Fibrin glue from stored human plasma.
An inexpensive and efficient method for local blood bank
preparation. Am. Surg. 1987, 53, 460–462. 10. Martinowitz,
U.; Saltz, R. Fibrin sealant. Curr. Opin. Hematol. 1996, 3,
395–402. 11. Kjaergard, H.K.; Trumbull, H.R. Vivostat
system autologous fibrin sealant: Preliminary study in
coronary bypass grafting. Ann. Thorac. Surg. 1998, 66,
482–486. 12. Nichols, W. Adverse Antibody-Mediated
Reactions to Topical Bovine Thrombin and Fibrin Glue. In
Symposium on Fibrin Sealant: Characteristics and Clinical
Use; Uniformed Services University of the Health Sciences:
Bethesda, 1994; 5–10. 13. Kevy, S.V.; Jacobson, M.S.;
Blasetti, L. An Automated Device for the Rapid Perparation
of Autologous Fibrin/Platelet Gel, an Effective,
Convenient, Bioactive Wound Sealant. Cambridge Symposium on
Tissue Sealants: Current Practice, Future Uses, La Jolla,
1996. 14. Redl, H.; Schlag, G.; Dinges, H.P. Methods of
fibrin seal application. Thorac. Cardiovasc. Surg. 1973, 30,
223–227. 15. Holcomb, J.B.; Pusateri, A.E.; Hess, J.R.;
Hetz, S.P.; Harris, R.A.; Tock, B.B.; Drohan, W.N.;
MacPhee, M.J. Implications of new dry fibrin sealant
technology for trauma surgery. Surg. Clin. North Am. 1997,
77, 943–952. 16. Moore, M.M.; Nguyen, D.H.D.; Spotnitz,
W.D. Fibrin sealant reduces serous drainage and allows for
earlier drain removal after axillary dissection: A
randomized prospective trial. Am. Surg. 1997, 63, 97–102.
17. Vaxman, F.; Kolbe, A.; Stricher, F.; Zund, D.; Volkmar,
P.; Gros, D.; Grenier, J.F. Does fibrin glue improve
drainage after axillary lymph node dissection? Eur. Surg.
Res. 1995, 27, 346–352. 18. Suzuki, Y.; Kuroda, Y.; Morita,
A.; Fujimo, Y.; Tanoika, Y.; Kawamura, T.; Saiton, V.
Fibrin glue sealing for the prevention of pancreatic
fistulas following distal pancreatectomy. Arch. Surg. 1995,
130, 952–955. 19. D’andrea, A.A.; Costantino, V.; Sperti,
C.; Pedrazzoli, S. Humand fibrin sealant in pancreatic
surgery: A prospective randomized study. Ital. J.
Gastroenterol. 1994, 26, 283–286. 20. Rakocz, M.; Mazar,
A.; Varon, D.; Spierer, S.; Blinder, D.; Martinowitz, U.
Dental extractions inpatients with bleeding disorders: The
use of fibrin glue. Oral Surg. Oral Med. Oral Pathol. 1993,
75, 280–282. 21. Hoots, K.; McLeod, J.; E2qqers, E.; Berg,
J.; Lee, M.; Gomperts, E. Pilot study to evaluate efficacy
of fibrin sealant(human) on hemostasis in hemophiliacs
undergoing tooth extraction (abst.). Blood 1993, 82, 598.
22. Achauer, B.M.; Miller, S.R.; Lee, T.E. The hemostatic
effect of fibrin glue on graft donor sites. J. Burn Care
Rehabil. 1994, 15, 24–28. 23. Al-Yamany, M.; Del Maestro,
R.F. Prevention of subdural fluid collections following
transcortical intraventricular and/or paraventricular
procedures by using fibrin adhesive. J. Neurosurg. 2000, 92,
406–412. 24. Lee, K.-C.; Park, S.-K.; Lee, K.-S.
Neurosurgical applications of fibrin adhesive. Yonsei Med.
J. 1991, 32, 53–57. 25. Iwakawa, M.; Mizoi, K.; Tessler,
A.; Itoh, Y. Intraspinal implants of fibrin glue containing
glial cell line-derived neurotrophic factor promote dorsal
root regeneration into spinal cord. Neurorehabil. Neural
Repair 2001, 15, 173–182. 26. Martinowitz, U.; Mazar, A.L.;
Taicher, S.; Varon, D.; Gitel, S.N.; Ramot, B.; Rakocz, M.
Dental extraction for patients on oral anticoagulant
therapy. Oral Surg. Oral Med. Oral Pathol. 1990, 70,
274–277. 27. Bodner, L.; Weinstein, J.M.; Baumgarten, A.K.
Efficacy of fibrin sealant in patients on various levels of
oral anticoagulant undergoing oral surgery. Oral Surg. Oral
Med. Oral Pathol. Oral Radiol. Endo. 1998, 86, 421–424. 28.
Longstreth, G.F. Epidemiology of hospitalization for acute
upper gastrointestinal hemorrhage: A populationbased study.
Am. J. Gastroenterol. 1995, 90, 206–210. 29. Rutgeerts, P.;
Rauws, E.; Wara, P.; Swain, P.; Hoos, A.; Solleder, E.;
Halttunen, J.; Dobrilla, G.; Richter, G.; Prassler, R.
Randomised trial of single and repeated fibrin glue compared
with injection of polidocanol in treatment of bleeding
peptic ulcer. Lancet 1997, 350, 692–696. 30. Zimmer, T.;
Rucktaschel, F.; Stolzel, U.; Liehr, R.M.; Schuppan, D.;
Stallmach, A.; Zeitz, M.; Weber, E.; Riecken, E.O.
Endoscopic sclerotherapy with fibrin glue as compared with
polidocanol to prevent early esophageal variceal
rebleeding. J. Hepatol. 1998, 28, 292–297. 31. Lin, H.J.;
Hsieh, Y.H.; Tseng, G.Y.; Perng, C.L.; Chang, F.Y.; Lee,
S.D. Endoscopic injection with fibrin sealant versus
epinephrine for arrest of peptic ulcer bleeding: A
randomized, comparative trial. J. Clin. Gastroenterol.
2002, 35, 218–221. 32. Heldwein, W.; Avenhaus, W.;
Schonekas, H.; Kaess, H.; Muller-Lissner, S.; Hasford, B.;
Hasford, J. Injection of fibrin tissue adhesive versus laser
photocoagulation in the treatment of high-risk bleeding
peptic ulcers: A controlled randomized study. Endoscopy
1996, 28, 756–760. 33. Dunn, C.J.; Goa, K.L. Fibrin
sealant: A review of its use in surgery and endoscopy.
Drugs 1999, 58, 863–886. 34. Lilius, P. Fibrin adhesive:
Its use in selected skin grafting. Practical note. Scand.
J. Plast. Reconstr. Surg. Hand Surg. 1987, 21, 245–248. 35.
Luke, M.; Kvist, E.; Andersen, F.; Hjortrup, A. Reduction
of post-operative bleeding after transurethral resection of
the prostate by local instillation of fibrin adhesive
(Beriplast). Br. J. Urol. 1986, 58, 672–675. 36. McKay,
T.C.; Albala, D.M.; Gehrin, B.E.; Castelli, M. Laparoscopic
ureteral reanastomosis using fibrin glue. J. Urol. 1994,
152, 1637–1640. 37. Bach, A.D.; Bannasch, H.; Galla, T.J.;
Bittner, K.M.; Stark, G.B. Fibrin glue as matrix for
cultured autologous urothelial cells in urethral
reconstruction. Tissue Eng. 2001, 7, 45–53. 38. Muto, T.;
Ishibiki, K.; Hatakeyama, K.; et al. Efficacy and safety of
B1 91.021.3 in patients receiving anastomotic surgery of
the digestive tract: A multicenter study. Shinyaku To
Rinsho 1994, 43, 2274–2283. 39. Spotnitz, W.D.; Falstrom,
J.K.; Rodeheaver, G.T. The role of sutures and fibrin
sealant in wound healing. Surg. Clin. North Am. 1997, 77,
651–669. 40. Noun, R.; Elias, D.; Balladur, P.; Bismuth,
H.; Parc, R.; Lasser, P.; Belghiti, J. Fibrin glue
effectiveness and tolerance after elective liver resection:
A randomized trial. Hepatogastroenterology 1996, 43,
221–224. 41. Wakasugi, J.; Shimada, H. Application of fibrin
sealant in liver surgery. Biomed. Prog. 1994, 7, 33–35. 42.
Ishitani, M.B.; McGahren, E.D.; Sibley, D.A.; Spotnitz,
W.D.; Rodgers, B.M. Laparoscopically applied fibrin glue in
experimental liver trauma. J. Pediatr. Surg. 1989, 24,
867–871. 43. Thiele, H.; Berg, P.L.; Frick, B.; Kalk, J.F.
Fibrin-glue injection—a new method of haemostasis after
laparospcopic liver biopsy. Dtsch. Med. Wochenschr. 1989,
114 (31–32), 1196–1198. 44. Pessaux, P.; Tuech, J.J.;
Arnaud, J.P. Prevention of pancreatic fistulas after
surgical resection. A decade of clinical trials. Presse
Med. 2001, 30, 1359–1363. 45. Suzuki, Y.; Kuroda, Y.;
Morita, A.; Fujino, Y.; Tanioka, Y.; Kawamuro, T.; Saitoh,
Y. Fibrin glue sealing for the prevention of pancreatic
fistulas following distal pancreatectomy. Arch. Surg. 1995,
130, 952–955. 46. D’Andrea, A.A.; Constantino, V.; Sperti,
C.; Pedrazzoli, S. Human fibrin sealant in pancreatic
surgery: Is it useful in preventing fistulas? A prospective
randomized study. Ital. J. Gastroenterol. 1994, 26,
283–286. 47. Borst, H.G.; Haverich, A.; Walterbush, G.;
Maatz, W. Fibrin adhesive: An important hemsotatic adjunct
in cardiovascular operations. J. Thorac. Cardiovasc. Surg.
1982, 84, 548–553. 48. Matthew, T.L.; Spotnitz, W.D.; Kron,
I.L.; Daniel, T.M.; Tribble, C.G.; Nolan, S.P. Four years’
experience with fibrin sealant in thoracic and
cardiovascular surgery. Ann. Thorac. Surg. 1990, 50, 40–44.
49. Jackson, M.R.; Danby, C.A.; Alving, B.M. Heparinoid
anticoagulation and topical fibrin sealant in heparin
induced thrombocytopenia. Ann. Thorac. Surg. 1997, 64,
1815–1817. 50. Rousou, J.; Levitsky, S.; Gonzalez-Lavin,
L.; Cosgrove, D.; Magilligan, D.; Weldon, C.; Hiebert, C.;
Hess, P.; Joyce, L.; Bergsland, J.; et al. Randomized
clinical trial of fibrin sealant in patients undergoing
resternotomy or reoperation after cardiac operations. A
multicenter study. J. Thorac. Cardiovasc. Surg. 1989, 97,
194–203. 51. Seguin, J.R.; Frapier, J.-M.; Colson, P.;
Chaptal, P.A. Fibrin sealant improves surgical results of
type A acute aortic dissections. Ann. Thorac. Surg. 1991,
52, 745–749. 52. Seguin, J.R.; Picard, E.; Frapier, J.M.;
Chaptal, P.A. Aortic valve repair with fibrin glue for type
A acute aortic dissection. Ann. Thorac. Surg. 1994, 58,
304–307. 53. Milne, A.A.; Murphy, W.G.; Reading, S.J.;
Ruckley, C.V. Fibrin reduces suture line bleeding during
carotid endarterectomy: A randomised trial. Eur. J. Vasc.
Endovasc. Surg. 1995, 10, 91–94. 54. Ismail, S.; Combs,
M.J.; Goodman, N.C.; Teotia, S.S.; Teates, C.D.; Abbott,
R.D.; Fechner, R.E.; Nolan, S.P.; Powers, E.R.; Spotnitz,
W.D. Reduction of femoral arterial bleeding post
catheterization using percutaneous application of fibrin
sealant. Catheter. Cardiovasc. Diagn. 1995, 34, 88–95. F

55. Kipshidze, N.; Ferguson, J.J., III; Macris, M.P.;


Clubb, F.; Cloy, M.; Horn, J.; Sperl, F.; Sahota, H.;
Keelan, M.H., Jr.; Nikolaychik, V.V. Percutaneous
application of fibrin sealant to achieve hemostasis
following arterial catheterization. J. Invasive Cardiol.
1998, 10, 133–141.
56. Ney, A.L.; Kelly, P.H.; Tsukayama, D.T.; Bubrick, M.P.
Fibrin glue-antibiotic suspension in the prevention of
prosthetic graft infection. J. Trauma 1990, 30, 1000–1006.

57. Tanemoto, K.; Fujinami, H. Experimental study on


bacterial colonization of fibrin glue and its prevention.
Clin. Ther. 1994, 16, 1016–1027. 58. Mouritzen, C.; Dromer,
M.; Keinecke, H.-O. The effect of fibrin glueing to seal
bronchial and alveolar leakages after pulmonary resections:
A randomised trial. Eur. J. Cardiothorac. Surg. 1993, 7,
75–80. 59. Morikawa, T.; Katoh, H. Improved techniques of
applying fibrin glue in lung surgery. Eur. Surg. Res. 1999,
31, 180–186. 60. Kinoshita, T.; Miyoshi, S.; Katoh, M.;
Yoshimasu, T.; Juri, M.; Maebeya, S.; Naito, Y.
Intrapleural administration of a large amount of diluted
fibrin glue for intractable pneumothorax. Chest 2001, 119,
671–672.
Finite Element Analysis

1. Huebner, K.H.; Dewhirst, D.L.; Smith, D.E.; Byron, T.G.


The Finite Element Method for Engineers, 4th Ed.; John
Wiley & Sons, Inc.: New York, 2001.

2. Rao, S.S. The Finite Element Method in Engineering, 3rd


Ed.; Butterworth Heinemann: Boston, 1999.

3. Cook, R.D.; Malkus, D.S.; Plesha, M.E.; Witt, R.J.


Concepts and Applications of Finite Element Analysis, 4th
Ed.; John Wiley & Sons, Inc.: New York, 2002.

4. Finite Elements in Biomechanics; Gallagher, R.H., Simon,


B.R., Johnson, P., Gross, J., Eds.; John Wiley & Sons,
Inc.: New York, 1982.

5. Hubsch, P.F.; Middleton, J.; Knox, J. A finite element


analysis of the stress at the restoration-tooth interface,
comparing inlays and bulk fillings. Biomaterials 2000, 21
(10), 1015–1019.

6. Toparli, M.; Aykul, H.; Sasaki, S. Temperature and


thermal stress analysis of a crowned maxillary second
premolar tooth using three-dimensional finite element
method. J. Oral Rehabil. 2003, 30 (1), 99–105.

7. Shim, E.B.; Kamm, R.D.; Heldt, T.; Mark, R.G. Numerical


analysis of blood flow through a stenosed artery using a
coupled, multiscale simulation method. Comput. Cardiol.
2000, 27, 219–222.

8. Leuprecht, A.; Perktold, K.; Kozerke, S.; Boesiger, P.


Combined CFD and MRI study of blood flow in a human
ascending aorta model. Biorheology 2002, 39 (3–4), 425–429.

9. Needleman, A.; Rabinowitz, S.A.; Bogen, D.K.; McMahon,


T.A. A finite element model of the infarcted left ventricle.
J. Biomech. 1983, 16 (1), 45–58.

10. Costa, K.D.; Hunter, P.J.; Rogers, J.M.; Guccione,


J.M.; Waldman, L.K.; McCulloch, A.D. A threedimensional
finite element method for large elastic deformations of
ventricular myocardium: I–Cylindrical and spherical polar
coordinates. J. Biomech. Eng. 1996, 118 (4), 452–463. 11.
Costa, K.D.; Hunter, P.J.; Wayne, J.S.; Waldman, L.K.;
Guccione, J.M.; McCulloch, A.D. A threedimensional finite
element method for large elastic deformations of
ventricular myocardium: II–Prolate spheroidal coordinates.
J. Biomech. Eng. 1996, 118 (4), 464–472. 12. Wayne, J.S.
Load partitioning influences the mechanical response of
articular cartilage. Ann. Biomed. Eng. 1995, 23, 40–47. 13.
Dunbar, W.L., Jr.; Un, K.; Donzelli, P.S.; Spilker, R.L. An
evaluation of three-dimensional diarthrodial joint contact
using penetration data and the finite element method. J.
Biomech. Eng. 2001, 123 (4), 333–340. 14. Wayne, J.S.; Woo,
S.L.-Y.; Kwan, M.K. Application of the u-p finite element
method to the study of articular cartilage. J. Biomech.
Eng. 1991, 113, 397–403. 15. Li, L.P.; Buschmann, M.D.;
Shirazi-Adl, A. A fibril reinforced nonhomogeneous
poroelastic model for articular cartilage: Inhomogeneous
response in unconfined compression. J. Biomech. 2000, 33
(12), 1533–1541. 16. Wu, J.Z.; Herzog, W.; Hasler, E.M.
Inadequate placement of osteochondral plugs may induce
abnormal stressstrain distributionsin articular cartilage
–finite element simulations. Med. Eng. Phys. 2002, 24 (2),
85–97. 17. Hart, R.T.; Davy, D.T.; Heiple, K.G. A
computational method for stress analysis of adaptive
elastic materials with a view toward applications in
strain-induced bone remodelling. J. Biomech. Eng. 1984,
106, 342–350. 18. Huiskes, R.; Weinans, H.; Grootenboer,
H.J.; Dalstra, M.; Fudala, B.; Sloof, T.J. Adaptive bone
remodelling theory applied to prosthetic-design analysis.
J. Biomech. 1987, 20, 1135–1150. 19. Kerner, J.; Huiskes,
R.; van Lenthe, G.H.; Weinans, H.; van Rietbergen, B.;
Engh, C.A.; Amis, A.A. Correlation between pre-operative
periprosthetic bone density and post-operatire bone loss in
THA can be explained by strain-adaptive remodelling. J.
Biomech. 1999, 32, 695–703. 20. Zienkiewicz, O.C.; Taylor,
R.L. The Finite Element Method, 5th Ed.; Butterworth
Heinemann: Oxford, 2000. 21. Bathe, K.J. Finite Element
Procedures; Prentice-Hall: New Jersey, 1996. 22. Hughes,
T.J.R. The Finite Element Method: Linear Static and Dynamic
Finite Element Analysis; PrenticeHall: New Jersey, 1987. F
Flow Cytometry

23. Bass, D.A.; Parce, J.W.; DeChatelet, L.R.; Szejda, P.;


Seeds, M.C.; Thomas, M. Flow cytometric studies of
oxidative product formation by neutrophils: A graded
response to membrane stimulation. J. Immunol. 1983, 130,
1910–1917.

24. Rothe, G.; Valet, G. Flow cytometric analysis of


respiratory burst activity in phagocytes with hydroethidine
and 2u,7u-dichlorofluorescin. J. Leukoc. Biol. 1990, 47,
440–448.

25. Robinson, J.P.; Carter, W.O.; Narayanan, P.K. Oxidative


Product Formation Analysis by Flow Cytometry. In Methods in
Cell Biology: Flow Cytometry; Darzynkiewicz, Z., Robinson,
J.P., Crissman, H.A., Eds.; Academic Press: New York, 1994;
437–447.

26. Li, N.; Ragheb, K.; Lawler, G.; Sturgis, J.; Rajwa, B.;
Melendez, J.A.; Robinson, J.P. DPI induces mitochondrial
superoxide-mediated apoptosis. Free Radic. Biol. Med. 2003,
34, 465–477.

27. Crissman, H.A.; Tobey, R.A. Cell-cycle analysis in


20minutes. Science 1974, 184, 1297–1298.

28. Darzynkiewicz, Z.; Traganos, F.; Sharpless, T.K.;


Melamed, M.R. Cell cycle-related changes in nuclear
chromatin of stimulated lymphocytes as measured by flow
cytometry. Cancer Res. 1977, 37, 4635–4640.

29. Ormerod, M.G. Investigating the relationship between


the cell cycle and apoptosis using flow cytometry. J.
Immunol. Methods 2002, 265, 73–80.

30. Steen, H.B. Flow cytometry of bacteria: Glimpses from


the past with a view to the future. J. Microbiol. Methods
2000, 42, 65–74. 31. Sincock, S.A.; Robinson, J.P. Flow
cytometric analysis of microorganisms. Methods Cell Biol.
2001, 64, 511–537. 32. Galbraith, D.W.; Harkins, K.R.;
Maddox, J.M.; Ayres, N.M.; Sharma, D.P.; Firoozabady, E.
Rapid flow cytometric analysis of the cell cycle in intact
plant tissues. Science 1983, 220, 1049–1051. 33. Brown,
S.C.; Bergounioux, C. Plant Flow Cytometry. In Flow
Cytometry: Advanced Reserch and Clinical Application; Yen,
A., Ed.; CRC Press: Boca Raton, 1990; 195–212. 34. Harkins,
K.R.; Jefferson, R.A.; Kavanagh, T.A.; Bevan, M.W.;
Galbraith, D.W. Expression of photosynthesis-related gene
fusions is restricted by cell type in transgenic plants and
in transfected protoplasts. Proc. Natl. Acad. Sci. U.S.A.
1990, 87, 816–820. 35. Kuckuck, F.W.; Edwards, B.S.; Sklar,
L.A. High throughput flow cytometry. Cytometry 2001, 44,
83–90. 36. Libbus, B.L.; Perreault, S.D.; Johnson, L.A.;
Pinkel, D. Incidence of chromosome aberrations in mammalian
sperm stained with Hoechst 33342 and UV-laser irradiated
during flow sorting. Mutat. Res. 1987, 182, 265–274. 37.
Peter, A.T.; Jones, P.P.; Robinson, J.P. Fractionation of
bovine spermatozoa for sex selection: A rapid
immunomagnetic technique to remove spermatozoa that contain
the H–Y antigen. Theriogenology 1993, 40, 1177–1185. 38.
Meseguer, M.; Garrido, N.; Remohi, J.; Simon, C.; Pellicer,
A. Gender selection: Ethical, scientific, legal, and
practical issues. J. Assist. Reprod. Genet. 2002, 19,
443–446. 39. Schwartz, A.; Ferna´ndez-Repollet, E.
Development of clinical standards for flow cytometry. Ann.
N.Y. Acad. Sci. 1993, 677, 28–39.
Foreign Body Response

34. Williams, R.L.; Hunt, J.A.; Tengvall, P. Fibroblast


adhesion onto methyl–silica gradients with and without
preadsorbed protein. J. Biomed. Mater. Res. 1995, 29 (12),
1545–1555.

35. James, S.J.; Pogribna, M.; Miller, B.J.; Bolon, B.;


Muskhelishvili, L. Characterization of cellular response to
silicone implants in rats: Implications for foreign-body
carcinogenesis. Biomaterials 1997, 18 (9), 667–675.

36. Baldwin, L.; Flanagan, B.F.; McLaughlin, P.J.;


Parkinson, R.W.; Hunt, J.A.; Williams, D.F. A study of
tissue interface membranes from revision accord knee
arthroplasty: The role of T lymphocytes. Biomaterials 2002,
23 (14), 3007–3014.

37. Brodbeck, W.G.; Nakayama, Y.; Matsuda, T.; Colton, E.;


Ziats, N.P.; Anderson, J.M. Biomaterial surface chemistry
dictates adherent monocyte/macrophage cytokine expression
in vitro. Cytokine 2002, 18 (6), 311–319.

38. Harrington, P.M.; Newton, D.J.; Williams, C.M.M.; Hunt,


J.A.; Dearman, R.J.; Kimber, I.; Coleman, J.W.; Flanagan,
B.F. Eotaxin and eotaxin receptor (CCR3) expression in
Sephadex particle-induced rat lung inflammation. Int. J.
Exper. Pathol. 1999, 80 (3), 177–185.

39. Jiang, X.; Marois, Y.; Traore, A.; Tessier, D.; Dao,
L.H.; Guidoin, R.; Zhang, Z. Tissue reaction to
polypyrrole-coated polyester fabrics: An in vivo study in
rats. Tissue Eng. 2002, 8 (4), 635–647.

40. Kao, W.J.; Liu, Y.; Gundloori, R.; Li, J.; Lee, D.;
Einerson, N.; Burmania, J.; Stevens, K. Engineering
endogenous inflammatory cells as delivery vehicles. J.
Control. Release: Off. J. Control. Rel. Soc. 2002, 78
(1–3), 219–233.

41. Thomas, A.; Harding, K.G.; Moore, K. Alginates from


wound dressings activate human macrophages to secrete
tumour necrosis factor-[alpha]. Biomaterials 2000, 21 (17),
1797–1802.

42. Corry, D.C.; Delucia, A., III; Zhu, H.; Radcliffe,


R.R.; Brevetti, G.R.; El-Khatib, H.; Vance, S.J.; Moyer,
B.R.; Cotts, W.G.; Richenbacher, W.E. Time course of
cytokine release and complement activation after
implantation of the HeartMate left ventricular assist
device. ASAIO J. 1998, 44 (5), M347–M351.

43. Suska, F.; Esposito, M.; Gretzer, C.; Ka¨lltorp, M.;


Tengyall, P.; Thomsen, P. IL-1[Alpha], IL-1[Beta] and
TNF-[Alpha] secretion during in vivo/ex vivo cellular
interactions with titanium and copper. Biomaterials 2003,
24 (3), 461–468.

44. Babensee, J.E.; Stein, M.M.; Moore, L.K.


Interconnections between inflammatory and immune responses
in tissue engineering. Ann. N.Y. Acad. Sci. 2002, 961,
360–363.

45. Kirkpatrick, C.J.; Krump-Konvalinkova, V.; Unger, R.E.;


Bittinger, F.; Otto, M.; Peters, K. Tissue response and
biomaterial integration: The efficacy of in vitro methods.
Biomol. Eng. 2002, 19 (2–6), 211–217.

46. Schindler, R.; Senf, R.; Frei, U. Influencing the


inflammatory response of haemodialysis patients by cytokine
elimination using large-pore membranes. Nephrol. Dial.
Transplant. 2002, 17 (1), 17–19. 47. Ratner, B.D. Reducing
capsular thickness and enhancing angiogenesis around
implant drug release systems. J. Control. Release 2002, 78
(1–3), 211–218. 48. Yan, J.Y.J.; Cooke, F.W.; Vaskelis,
P.S.; von Recum, A.F. Titanium-coated Dacron velour. A
study of interfacial connective tissue formation. J.
Biomed. Mater. Res. 1989, 23 (2), 171–189. 49. Winkler,
P.A.; Herzog, C.; Weiler, C.; Krishnan, K.G. Foreign-body
reaction to silastic burr-hole covers with seroma
formation: Case report and review of the literature.
Pathol. Res. Pract. 2000, 196 (1), 61–66. 50. Ma, N.;
Petit, A.; Yahia, L.; Huk, O.L.; Tabrizian, M. Cytotoxic
reaction and TNF-[Alpha;] response of macrophages to
polyurethane particles. J. Biomater. Sci., Polym. Ed. 2002,
13 (3), 257–272. 51. Rhodes, N.P.; Hunt, J.A.; Williams,
D.F. Macrophage subpopulation differentiation by
stimulation with biomaterials. J. Biomed. Mater. Res. 1997,
37 (4), 481–488. 52. Ward, W.K.; Slobodzian, E.P.;
Tiekotter, K.L.; Wood, M.D. The effect of microgeometry,
implant thickness and polyurethane chemistry on the foreign
body response to subcutaneous implants. Biomaterials 2002,
23 (21), 4185–4192. 53. Lobler, M.; Sa[ss], M.; Kunze, C.;
Schmitz, K.-P.; Hopt, U.T. Biomaterial implants induce the
inflammation marker CRP at the site of implantation. J.
Biomed. Mater. Res. 2002, 61 (1), 165–167. 54. Held, M.;
Dost, P.; Erdtmann, M.; Baumann, H.; Jahnke, K.
Biocompatibility testing of new polymers in a moving
implant bed. ORL 2000, 62 (5), 247–250. 55. Bos, R.R.;
Rozema, F.R.; Boering, G.; Nijenhuis, A.J.; Pennings, A.J.;
Verwey, A.B.; Nieuwenhuis, P.; Jansen, H.W. Degradation of
and tissue reaction to biodegradable poly(L-lactide) for
use as internal fixation of fractures: A study in rats.
Biomaterials 1991, 12 (1), 32–36. 56. Bergsma, J.E.;
Rozema, F.R.; Bos, R.R.M.; Boering, G.; De Bruijn, W.C.;
Pennings, A.J. Biocompatibility study of as-polymerized
poly(L-lactide) in rats using a cage implant system. J.
Biomed. Mater. Res. 1995, 29 (2), 173–179. 57. Khabbaz, F.;
Karlsson, S.; Albertsson, A. Py-GC/MS an effective
technique to characterizing of degradation mechanism of
poly (L-lactide) in the different environment. J. Appl.
Polym. Sci. 2000, 78 (13), 2369–2378. 58. von Recum, H.A.;
Cleek, R.L.; Mikos, A.G.; Eskin, S.G. Degradation of
polydispersed poly(L-lactic acid) to modulate lactic acid
release. Biomaterials 1995, 16 (6), 441–447. 59. Van Dijk,
M.; Tunc, D.C.; Smit, T.H.; Higham, P.; Burger, E.H.;
Wuisman, P.I.J.M. In vitro and in vivo degradation of
bioabsorbable PLLA spinal fusion cages. J. Biomed. Mater.
Res. 2002, 63 (6), 752–759. 60. Iwasaki, Y.; Sawada, S.-I.;
Ishihara, K.; Khang, G.; Hai, B.L. Reduction of
surface-induced inflammatory reaction on PLGA/MPC polymer
blend. Biomaterials 2002, 23 (18), 3897–3903. 61.
Werkmeister, J.A.; Edwards, G.A.; White, J.F.; Casagranda,
F.; Hunt, J.A.; Williams, D.F.; Ramshaw, J.A.M. In vivo
evaluation of modified mandrel-grown vascular prostheses. J.
Biomed. Mater. Res. 1999, 47 (3), 316–323.
Functional MRI: Applications

21. Lee, C.C.; Ward, H.A.; Sharbrough, F.W.; Meyer, F.B.;


Marsh, W.R.; Raffel, C.; So, E.L.; Cascino, G.D.; Shin, C.;
Xu, Y.; Riederer, S.J.; Jack, C.R., Jr. Assessment of
functional MR imaging in neurosurgical planning. AJNR Am.
J. Neuroradiol. 1999, 20 (8), 1511–1519.

22. Haberg, A.; Kvistad, K.A.; Unsgard, G.; Haraldseth, O.


Preoperative blood oxygen level-dependent functional
magnetic resonance imaging in patients with primary brain
tumors: clinical application and outcome. Neurosurgery
2004, 54 (4), 902–914.

23. Roux, F.E.; Ibarrola, D.; Tremoulet, M.; Lazorthes, Y.;


Henry, P.; Sol, J.C.; Berry, I. Methodological and
technical issues for integrating functional magnetic
resonance imaging data in a neuronavigational system.
Neurosurgery 2001, 49 (5), 1145–1156.

24. Lehericy, S.; Cohen, L.; Bazin, B.; Samson, S.;


Giacomini, E.; Rougetet, R.; Hertz-Pannier, L.; Le Bihan,
D.; Marsault, C.; Baulac, M. Functional MR evaluation of
temporal and frontal language dominance compared with the
Wada test. Neurology 2000, 58 (8), 1625–1633.

25. Woermann, F.G.; Jokeit, H.; Luerding, R.; Freitag, H.;


Schulz, R.; Guertler, S.; Okujava, M.; Wolf, P.; Tuxhorn,
I.; Ebner, A. Language lateralization by Wada test and fMRI
in 100 patients with epilepsy. Neurology 2003, 61 (5),
699–701.

26. Kennedy, P.; Andreasen, D.; Ehirim, P.; King, B.;


Kirby, T.; Mao, H.; Moore, M. Using human extra-cortical
local field potentials to control a switch. J. Neural Eng.
2004, 1 (2), 72–77.

27. Elbert, T.; Rockstroh, B. Reorganization of human


cerebral cortex: the range of changes following use and
injury. Neuroscientist 2004, 10 (2), 129–141.

28. Loubinoux, I.; Carel, C.; Pariente, J.; Dechaumont, S.;


Albucher, J.F.; Marque, P.; Manelfe, C.; Chollet, F.
Correlation between cerebral reorganization and motor
recovery after subcortical infarcts. Neuroimage 2003, 20,
2166–2180. 29. Seritan, A.L.; Mendez, M.F.; Silverman,
D.H.; Hurley, R.A.; Taber, K.H. Functional imaging as a
window to dementia: corticobasal degeneration. J.
Neuropsychiatry Clin. Neurosci. 2004, 16 (4), 393–399. 30.
Leslie, R.A.; James, M.F. Pharmacological magnetic
resonance imaging: a new application for functional MRI.
Trends Pharmacol. Sci. 2000, 21 (8), 314–318. 31. Houston,
G.C.; Papadakis, N.G.; Carpenter, T.A.; Hall, L.D.;
Mukherjee, B.; James, M.F.; Huang, C.L. Mapping of brain
activation in response to pharmacological agents using fMRI
in the rat. Magn. Reson. Imaging 2001, 19 (7), 905–919. 32.
Rossini, P.M.; Dal Forno, G. Integrated technology for
evaluation of brain function and neural plasticity. Phys.
Med. Rehabil. Clin. N. Am. 2004, 15 (1), 263–306. 33.
Ramnani, N.; Behrens, T.E.; Penny, W.; Matthews, P.M. New
approaches for exploring anatomical and functional
connectivity in the human brain. Biol. Psychiatry 2004, 56,
613–619. FURTHER READING Detre, J.A.; Floyd, T.F.
Functional MRI and its applications to the clinical
neurosciences. Neuroscientist 2001, 7, 64–79. Huettel,
S.A.; Song, A.W.; McCarthy, G. Functional Magnetic
Resonance Imaging, 1st Ed.; Sinauer Associates: Sunderland,
MA, 2004. Marshall, J.C.; Fink, G.R. Cerebral localization,
then and now. Neuroimage 2003, 20 (suppl. 1), S2–S7.
Richard, B.B. Introduction to Functional Magnetic Resonance
Imaging: Principles and Techniques, 1st Ed.; Cambridge
University Press: New York, 2001. Ugurbil, K.; Toth, L.;
Kim, D.S. . How accurate is magnetic resonance imaging of
brain function? Trends Neurosci. 2003, 26 (2), 108–114.
Gait Analysis

53. Delp, S.L.; Loan, J.P.; Hoy, M.G.; Zajac, F.E.; Topp,
E.L.; Rosen, J.M. An interactive graphics-based model of
the lower extremity to study orthopaedic surgical
procedures. IEEE T. Bio-Med. Eng. 1990, 37 (8), 757–767.

54. Eames, N.W.; Baker, R.; Cosgrove, A. Defining


gastrocnemius length in ambulant children. Gait Posture
1997, 6, 9–17.

55. Eames, N.W.; Baker, R.; Hill, N.; Graham, K.; Taylor,
T.; Cosgrove, A. The effect of botulinum toxin A on
gastrocnemius length: Magnitude and duration of response.
Dev. Med. Child. Neur. 1999, 41 (4), 226–232.

56. Frigo, C.; Nielsen, J.; Crenna, P. Modelling the


triceps surae muscle-tendon complex for the estimation of
length changes during walking. J. Electromyogr. Kinesiol.
1996, 6, 191–203.

57. Vasavada, A.N.; Delp, S.L.; Maloney, W.J.; Schurman,


D.J.; Zajac, F.E. Compensating for changes in muscle length
in total hip arthroplasty. Effects on the moment generating
capacity of the muscles. Clin. Orthop. Rel. Res. 1994, 302,
121–133.

58. Arnold, A.S.; Asakawa, D.J.; Delp, S.L. Do the


hamstrings and adductors contribute to excessive internal
rotation of the hip in persons with cerebral palsy? Gait
Posture 2000, 11 (3), 181–190.

59. Arnold, A.S.; Delp, S.L. Rotational moment arms of the


medial hamstrings and adductors vary with femoral geometry
and limb position: Implications for the treatment of
internally rotated gait. J. Biomech. 2001, 34 (4), 437–447.

60. Arnold, A.S.; Salinas, S.; Asakawa, D.J.; Delp, S.L.


Accuracy of muscle moment arms estimated from MRI-based
musculoskeletal models of the lower extremity. Comput.
Aided. Surg. 2000, 5 (2), 108–119.

61. Delp, S.; Maloney, W. Effects of hip center locatoins


on the moment-generating capacity of the muscles. J.
Biomech. 1993, 26 (4/5), 485–499.

62. Delp, S.; Ringwelski, D.A.; Carroll, N.C. Transfer of


the rectus femoris: Effects of transfer site on moment arms
about the knee and hip. J. Biomech. 1994, 27 (10),
1201–1211.
63. Delp, S.; Statler, K.; Carroll, N.C. Preserving plantar
flexion strength after surgical treatment for contracture of
the triceps surae: A computer simulation study. J.
Orthopaed. Res. 1995, 1 (1), 96–104.

64. Delp, S.L.; Bleck, E.E.; Zajac, F.E.; Bollini, G.


Biomechanical analysis of the Chiari pelvic osteotomy.
Preserving hip abductor strength. Clin. Orthop. Rel. Res.
1990, 254, 189–198.

65. Delp, S.L.; Hess, W.E.; Hungerford, D.S.; Jones, L.C.


Variation of rotation moment arms with hip flexion. J.
Biomech. 1999, 32, 493–501.

66. Delp, S.L.; Wixson, R.L.; Komattu, A.V.; Kocmond, J.H.


How superior placement of the joint center in hip
arthroplasty affects the abductor muscles. Clin. Orthop.
Relat. Res. 1996, 328, 137–146.

67. Delp, S.L.; Zajac, F.E. Forceand moment-generating


capacity of lower-extremity muscles before and after tendon
lengthening. Clin. Orthop. Rel. Res. 1992, 284, 247–259.

68. Free, S.A.; Delp, S.L. Trochanteric transfer in total


hip replacement: Effects on the moment arms and
force-generating capacities of the hip abductors. J.
Orthopaed. Res. 1996, 14 (2), 245–250. 69. Duda, G.N.;
Schneider, E.; Chao, E.Y. Internal forces and moments in
the femur during walking. J. Biomech. 1997, 30 (9),
933–941. 70. Bergmann, G.; Deuretzbacher, G.; Heller, M.;
Graichen, F.; Rohlmann, A.; Strauss, J.; Duda, G.N. Hip
contact forces and gait patterns from routine activities.
J. Biomech. 2001, 34 (7), 859–871. 71. Heller, M.O.;
Bergmann, G.; Deuretzbacher, G.; Durselen, L.; Pohl, M.;
Claes, L.; Haas, N.P.; Duda, G.N. Musculo-skeletal loading
conditions at the hip during walking and stair climbing. J.
Biomech. 2001, 34 (7), 883–893. 72. O’Connor, J.; Feikes,
J.; Gill, R.; Zavatsky, A. Mobility of the Knee. In
Daniel’s Knee Injuries; D, P., J, O.C., W, A., Eds.;
Lippincott, Williams & Wilkins: Philadelphia, 2003. 73.
O’Connor, J.; Gill, R.; Zavatsky, A. Stability of the Knee.
In Daniel’s Knee Injuries; Pedowitz, D., O’Connor, J.,
Akeson, W., Eds.; Lippincott, Williams & Wilkins:
Philadelphia, 2003. 74. O’Connor, J.; Lu, T.-W.; Wilson,
D.; Feikes, J.; Leardini, A. Computer methods in
biomechanics and biomedical engineering—Kinematic pairs,
mechanisms or flexible structures? Comp. Methods Biomech.
Biomed. Eng. 1997, 1, 123–150. 75. Cavagna, G.A. Force
platforms as ergometers. J. Appl. Physiol. 1983, 39 (1),
174–179. 76. Eames, M.; Cosgrove, A.; Baker, R. Comparing
methods of estimating the total body centre of mass in
three-dimensions in normal and pathological gaits. Hum.
Mov. Sci. 1999, 18, 637–646. 77. Saini, M.; Kerrigan, D.;
Thirunarayan, M.; Duff-Raffaele, M. The vertical
displacement of the centre of mass during walking: A
comparison of four measurement methods. J. Biomech. Eng.
1998, 120, 133–139. 78. Shimba, T. An estimation of centre
of gravity from force plate data. J. Biomech. 1984, 17,
53–60. 79. Tesio, L.; Lanzi, D.; Detrembleur, C. The 3-D
motion of the centre of gravity of the human body during
level walking. 1. Normal subjects at low and intermediate
walking speeds. Clin. Biomech. 1998, 13 (2), 77–82. 80.
Tesio, L.; Lanzi, D.; Detrembleur, C. The 3-D motion of the
centre of gravity of the human body during level walking.
II lower limb amputees. Clin. Biomech. 1998, 13 (2), 83–90.
81. Whittle, M. Three-dimensional motion of the center of
gravity of the body during walking. Hum. Mov. Sci. 1997,
16, 347–355. 82. Anderson, F.C.; Pandy, M.G. Dynamic
optimization of human walking. J. Biomech. Eng. 2001, 123
(5), 381–390. 83. Anderson, F.C.; Pandy, M.G. Static and
dynamic optimization solutions for gait are practically
equivalent. J. Biomech. 2001, 34 (2), 153–161. 84.
Anderson, F.C.; Ziegler, J.M.; Pandy, M.G.; Whalen, R.T.
Application of high-performance computing to numerical
simulation of human movement. J. Biomech. Eng. 1995, 117
(1), 155–157. 85. Jonkers, I.; Spaepen, A.; Papaioannou,
G.; Stewart, C. An EMG-based, muscle driven forward
simulation of single support phase of gait. J. Biomech.
2002, 35 (5), 609–619. 86. Kerrigan, D.; R, R.; PO, R. The
modelling of adult spastic paretic stiff-legged gait swing
period based on actual kinematic data. Gait Posture 1998, 7
(2), 117–124. 87. Neptune, R.R.; Kautz, S.A.; Zajac, F.E.
Contributions of the individual ankle plantar flexors to
support, forward progression and swing initiation during
walking. J. Biomech. 2001, 34 (11), 1387–1398. 88. Pandy,
M.G.; Anderson, F.C. Dynamic simulation of human movement
using large-scale models of the body. Phonetica 2000, 57
(2–4), 219–228. 89. Piazza, S.J.; Delp, S.L. The influence
of muscles on knee flexion during the swing phase of gait.
J. Biomech. 1996, 29 (6), 723–733. 90. Riley, P.O.;
Kerrigan, D.C. Torque action of two-joint muscles in the
swing period of stiff-legged gait: A forward dynamic model
analysis. J. Biomech. 1998, 31 (9), 835–840. 91. Tashman,
S.; Zajac, F.E.; Perkash, I. Modeling and simulation of
paraplegic ambulation in a reciprocating gait orthosis. J.
Biomech. Eng. 1995, 117 (3), 300–308. 92. Veltink, P.H.;
Franken, H.M.; Van Alste, J.A.; Boom, H.B. Modelling the
optimal control of cyclical leg movements induced by
functional electrical stimulation. Int. J. Artif. Organs
1992, 15 (12), 746–755. 93. Yamaguchi, G.T.; Moran, D.W.;
Si, J. A computationally efficient method for solving the
redundant problem in biomechanics. J. Biomech. 1995, 28
(8), 999–1005. 94. Anderson, F.; Pandy, M. Individual
muscle contributions to support in normal walking. Gait
Posture. 2003, 17 (2), 159–169. 95. Kepple, T.; Siegel, K.;
Stanhope, S. Relative contributions of the lower extremity
joint moments to forward progression and support during
gait. Gait Posture 1997, 6, 1–8. 96. Schwartz, M.; Lakin,
G. The effect of tibial torsion on the dynamic function of
the soleus during gait. Gait Posture 2003, 17 (2), 113–118.
97. Siegel, K.; Kepple, T.; Stanhope, S. Joint moment
control of mechanical energy flow during normal gait. Gait
Posture 2004, 19 (1), 69–75. G
Gene-Activated Matrix

6. National Hospital Discharge Survey. In National Center


for Health Statistics, Vital Health Statistics; 13;
113–151.

7. Simons, M.; Bonow, R.; Chronos, N.; Cohen, D.; Giordano,


F.; Hammond, H.; Laham, R.; Li, W.; Pike, M.; Sellke, F.;
Stegmann, T.; Udelson, J.; Rosengart, T. Clinical trials in
coronary angiogenesis: Issues, problems, consensus: An
expert panel summary. Circulation 2000, 102 (11), E73–E86.

8. Tsurumi, Y.; Takeshita, S.; Chen, D.; Kearney, M.;


Rossow, S.; Passeri, J.; Horowitz, J.; Symes, J.; Isner, J.
Direct intramuscular gene transfer of naked DNA encoding
vascular endothelial growth factor augments collateral
development and tissue perfusion. Circulation 1996, 94
(12), 3281–3290.

9. Losordo, D.; Vale, P.; Symes, J.; Dunnington, C.;


Esakof, D.; Maysky, M.; Ashare, A.; Lathi, K.; Isner, J.
Gene therapy for myocardial angiogenesis: Initial clinical
results with direct myocardial injection of phVEGF165 as
sole therapy for myocardial ischemia. Circulation 1998, 98
(25), 2800–2804.

10. Bonadio, J. Tissue engineering via local gene delivery.


J. Mol. Med. 2000, 78 (6), 303–311.

11. Doukas, J.; Blease, K.; Craig, D.; Ma, C.; Chandler,
L.; Sosnowski, B.; Pierce, G. Delivery of FGF genes to
wound repair cells enhances arteriogenesis and myogenesis
in skeletal muscle. Molec. Ther. 2002, 5 (5 Pt. 1),
517–527.

12. Singer, A.; Clark, R. Cutaneous wound healing. N. Engl.


J. Med. 1999, 341 (10), 738–746.

13. Uherek, C.; Wels, W. DNA-carrier proteins for targeted


gene delivery. Adv. Drug Deliv. Rev. 2000, 44 (2–3),
153–166.

14. Shea, L.; Smiley, E.; Bonadio, J.; Mooney, D. DNA


delivery from polymer matrices for tissue engineering. Nat.
Biotechnol. 1999, 17 (6), 551–554.

15. Berry, M.; Gonzalez, A.; Clarke, W.; Greenlees, L.;


Barrett, L.; Tsang, W.; Seymour, L.; Bonadio, J.; Logan,
A.; Baird, A. Sustained effects of gene-activated matrices
after CNS injury. Mol. Cell. Neurosci. 2001, 17 (4),
706–716.

16. Kay, M.; Glorioso, J.; Naldini, L. Viral vectors for


gene therapy: The art of turning infectious agents into
vehicles of therapeutics. Nat. Med. 2001, 7 (1), 33–40.

17. Doukas, J.; Chandler, L.; Gonzalez, A.; Gu, D.;


Hoganson, D.; Ma, C.; Nguyen, T.; Printz, M.; Nesbit, M.;
Herlyn, M.; Crombleholme, T.; Aukerman, S.; Sosnowski, B.;
Pierce, G. Matrix immobilization enhances the tissue repair
activity of growth factor gene therapy vectors. Hum. Gene
Ther. 2001, 12 (7), 783–798.

18. Levy, R.; Song, C.; Tallapragada, S.; DeFelice, S.;


Travis Hinson, J.; Vyavahare, N.; Connoly, J.; Ryan, K.;
Li, Q. Localized adenovirus gene delivery using antiviral
IgG complexation. Gene Ther. 2001, 8, 659–667.

19. Chandler, L.; Doukas, J.; Gonzalez, A.; Hoganson, D.;


Gu, D.; Ma, C.; Nesbit, M.; Crombleholme, T.; Herlyn, M.;
Sosnowski, B.; Pierce, G. FGF2-Targeted adenovirus encoding
platelet-derived growth factor-B enhances de novo tissue
formation. Molec. Ther. 2000, 2 (2), 153–160. 20. Dvorak,
H.; Harvey, V.; Estrella, P.; Brown, L.; McDonagh, J.;
Dvorak, A. Fibrin containing gels induce angiogenesis.
Implications for tumor stroma generation and wound healing.
Lab. Invest. 1987, 57 (6), 673–686. 21. Pachence, J.; Kohn,
J. Biodegradable Polymers. In Principles of Tissue
Engineering, 2nd Ed.; Lanza, R., Langer, R., Vacanti, J.,
Eds.; Academic Press: San Diego, 2000; 263–277. 22.
Hubbell, J. Bioactive biomaterials. Curr. Opin. Biotechnol.
1999, 10 (2), 123–129. 23. Fang, J.; Zhu, Y.; Smiley, E.;
Bonadio, J.; Rouleau, J.; Goldstein, S.; McCauley, L.;
Davidson, B.; Roessler, B. Stimulation of new bone
formation by direct transfer of osteogenic plasmid genes.
Proc. Natl. Acad. Sci. U. S. A. 1996, 93 (12), 5753–5758.
24. Patil, P.V.; Graziano, G.P.; Bonadio, J.; Goldstein,
S.A. Interbody fusion augmentation using localized gene
delivery. Trans. Orthop. Res. Soc. 2000, 25, 360. 25.
Parentau, N.; Hardin-Young, J.; Ross, R. Skin. In
Principles of Tissue Engineering, 2nd Ed.; Lanza, R.,
Langer, R., Vacanti, J., Eds.; Academic Press: San Diego,
2000; 879–890. 26. Chandler, L.; Gu, D.; Ma, C.; Gonzalez,
A.; Doukas, J.; Nguyen, T.; Pierce, G.; Phillips, M.
Matrix-enabled gene transfer for cutaneous wound repair.
Wound Repair Regen. 2000, 8 (6), 473–479. 27. Tyrone, J.;
Mogford, J.; Chandler, L.; Ma, C.; Xia, Y.; Pierce, G.;
Mustoe, T. Collagen-embedded platelet-derived growth factor
DNA plasmid promotes wound healing in a dermal ulcer model.
J. Surg. Res. 2000, 93 (2), 230–236. 28. Hellstrom, M.;
Kal, N.M.; Lindahl, P.; Abramsson, A.; Betsholtz, C. Role
of PDGF-B and PDGFR-beta in recruitment of vascular smooth
muscle cells and pericytes during embryonic blood vessel
formation in the mouse. Development 1999, 126 (14),
3047–3055. 29. Horvath, K.; Doukas, J.; Lu, C.-Y.; Belkind,
N.; Greene, R.; Pierce, G.; Fullerton, D. Myocardial
functional recovery after fibroblast growth factor 2 gene
therapy as assessed by echocardiography and magnetic
resonance imaging. Ann. Thorac. Surg. 2002, 74, 481–487.
30. Bonadio, J. Local gene delivery for tissue
regeneration. E-biomed J. Regen. Med. 2000, 1, 25–29. 31.
Kyriakides, T.; Hartzel, T.; Huynh, G.; Bornstein, P.
Regulation of angiogenesis and matrix remodeling by
localized, matrix-mediated antisense gene delivery. Molec.
Ther. 2001, 3 (6), 842–849. 32. Agha-Mohammadi, S.; Lotze,
M. Regulatable systems: Applications in gene therapy and
replicating viruses. J. Clin. Invest. 2000, 105 (9),
1177–1183. G
Gene Therapy

1. Maruyama, H.; Higuchi, N.; Nishikawa, Y.; Kameda, S.;


Iino, N.; Kazama, J.J.; Takahashi, N.; Sugawa, M.; Hanawa,
H.; Tada, N.; Miyazaki, J.; Gejyo, F. High-level expression
of naked DNA delivered to rat liver via tail vein
injection. J. Gene Med. 2002, 4, 333–341.

2. Shyu, K.G.; Wang, M.T.; Wang, B.W.; Chang, C.C.; Leu,


J.G.; Kuan, P.; Chang, H. Intramyocardial injection of
naked DNA encoding HIF-1alpha/VP16 hybrid to enhance
angiogenesis in an acute myocardial infarction model in the
rat. Cardiovasc. Res. 2002, 54, 576–583.

3. De Lucca, F.L.; Sawan, F.M.; Watanabe, M.A.; de Souza,


L.R. Effect of the calcium phosphate-mediated RNA uptake on
the transfer of cellular immunity of a synthetic peptide of
HIV-1 to human lymphocytes by exogenous RNA. Mol. Cell.
Biochem. 2001, 228, 9–14.

4. Jeschke, M.G.; Richter, G.; Hofstadter, F.; Herndon,


D.N.; Perez-Polo, J.R.; Jauch, K.W. Non-viral liposomal
keratinocyte growth factor (KGF) cDNA gene transfer
improves dermal and epidermal regeneration through
stimulation of epithelial and mesenchymal factors. Gene
Ther. 2002, 9, 1065–1074.

5. Herweijer, H.; Wolff, J.A. Progress and prospects: Naked


DNA gene transfer and therapy. Gene Ther. 2003, 10,
453–458.

6. http://www-ermm.cbcu.cam.ac.uk/99000691h.htm(accessed
January 2003).

7. Glasspool-Malone, J.; Steenland, P.R.; McDonald, R.J.;


Sanchez, R.A.; Watts, T.L.; Zabner, J.; Malone, R.W. DNA
transfection of macaque and murine respiratory tissue is
greatly enhanced by use of a nuclease inhibitor. J. Gene
Med. 2002, 4, 322–323. 8. de Carvalho Bittencourt, M.;
Saas, P.; Fresnay, S.; Yerly-Motta, V.; Ferrand, C.;
Perruche, S.; Duperrier, A.; Herve, P.; Tiberghien, P.;
Chalmers, D.E. Exposure to exogenous DNA can modify the
sensitivity of the Fas apoptotic pathway. J. Gene Med.
2002, 4, 14–24. 9. Nishikawa, M.; Huang, L. Nonviral
vectors in the new millennium: Delivery barriers in gene
transfer. Hum. Gene Ther. 2001, 12, 861–870. 10. Hartikka,
J.; Sukhu, L.; Buchner, C.; Hazard, D.; Bozoukova, V.;
Margalith, M.; Nishioka, W.K.; Wheeler, C.J.; Manthorp, M.;
Sawdey, M. Electroporation-facilitated delivery of plasmid
DNA in skeletal muscle: Plasmid dependence of muscle damage
and effect of poloxamer 188. Mol. Ther. 2001, 4, 407–415.
11. Heller, L.C.; Coppola, D. Electrically mediated
delivery of vector plasmid DNA elicits an antitumor effect.
Gene Ther. 2002, 9, 1321–1325. 12. Somiari, S.;
Glasspool-Malone, J.; Drabick, J.J.; Gilbert, R.A.; Heller,
R.; Jaroszeski, M.J.; Malone, R.W. Theory and in vivo
application of electroporative gene delivery. Mol. Ther.
2000, 2, 178–187. 13. Lee, M.J.; Cho, S.S.; You, J.R.; Lee,
Y.; Kang, B.D.; Choi, J.S.; Park, J.W.; Suh, Y.L.; Kim,
J.A.; Kim, D.K.; Park, J.S. Intraperitoneal gene delivery
mediated by a novel cationic liposome in a peritoneal
disseminated ovarian cancer model. Gene Ther. 2002, 9,
859–866. 14. Baru, M.; Nahum, O.; Jaaro, H.; Sha’anani, J.;
Nur, I. Lysosome-disrupting peptide increases the efficiency
of in-vivo gene transfer by liposome-encapsulated DNA. J.
Drug Target. 1998 , 6, 191–199. 15. Tsunoo, H.; Komura, S.;
Ohishi, N.; Yajima, H.; Akiyama, S.; Kasai, Y.; Ito, K.;
Nakao, A.; Yagi, K. Effect of transfection with human
interferon-beta gene entrapped in cationic multilamellar
liposomes in combination with 5-fluorouracil on the growth
of human esophageal cancer cells in vitro. Anticancer Res.
2002, 22, 1537–1543. 16. Hailes, H.; Hurley, C.; Tabor, A.;
Barker, S.; Hart, S. Synthesis and Biological Properties of
Cationic Lipids in a Lipid/Peptide/DNA Transfection Vector.
In Molecular Therapy, American Society of Gene Therapy,
Boston, Massachusetts, June 5–9; Kellog, K., Rishizky, P.,
Wood, D., Eds.; Elsevier Science: New York, 2002; 226–275.
17. Wang, D.; Roy, K.; Hedley, M.; Barman, S. Enhanced
Intramuscular Gene Expression by DNA Delivery via Combined
Polymeric Networks/Electroporation. In Molecular Therapy,
American Society of Gene Therapy, Boston, Massachusettes,
June 5–9; Kellog, K., Rishizky, P., Wood, D., Eds.;
Elsevier Science: New York, 2002; 217–272. 18. Pun, S.;
Cheng, J.; Bellocq, N.; Jensen, G.; Davis, M. Cyclodextrin
Polymer-Based Gene Delivery Systems for Hepatocyte
Targeting. In Molecular Therapy, American Society of Gene
Therapy, Boston, Massachusetts, June 5–9; Kellog, K.,
Rishizky, P., Wood, D., Eds.; Elsevier Science: New York,
2002; 219–669. 19. Koskinen, M.; Sailynoja, E.; Ahola, M.;
Ahonen, M.; Kahari, V.; Salonen, J. Silica Xerogel Carrier
as Encapsulating Material for Controlled Release of
Recombinant Adenoviruses. In Molecular Therapy, American
Society of Gene Therapy, Boston, Massachusetts, June 5–9;
Kellog, K., Rishizky, P., Wood, D., Eds.; Elsevier Science:
New York, 2002; 224–684. 20. Ponnazhagan, S.; Mahendra, G.;
Kumar, S.; Thompson, J.A.; Castillas, M., Jr.
Conjugate-based targeting of recombinant adeno-associated
virus type 2 vectors by using avidin-linked ligands. J.
Virol. 2002, 76, 12900–12907. 21. Berns, K.I. Parvovirus
replication. Microbiol. Rev. 1990, 54, 316–329. 22.
Latchman, D.S.; Coffin, R.S. Viral vectors for gene therapy
in Parkinson’s disease. Rev. Neurosci. 2001, 12, 69–78. 23.
Fink, D.J.; Poliani, P.L.; Oligino, T.; Krisky, D.M.;
Goins, W.F.; Glorioso, J.C. Development of an HSVbased
vector for the treatment of Parkinson’s disease. Exp.
Neurol. 1997, 144, 103–121. 24. Rutledge, E.A.; Russell,
D.W. Adeno-associated virus vector integration junctions.
J. Virol. 1997, 71, 8429–8436. 25. Lesokhin, A.M.;
Delgado-Lopez, F.; Horwitz, M.S. Inhibition of chemokine
expression by adenovirus early region three (E3) genes. J.
Virol. 2002, 76, 8236–8243. 26. Snyder, R.O.; Miao, C.H.;
Patijn, G.A.; Spratt, S.K.; Danos, O.; Nagy, D.; Gown,
A.M.; Winther, B.; Meuse, L.; Cohen, L.K.; Thompson, A.R.;
Kay, M.A. Persistent and therapeutic concentrations of
human factor IX in mice after hepatic gene transfer of
recombinant AAV vectors. Nat. Genet. 1997, 16, 270–276. 27.
Chao, H.; Monahan, P.E.; Liu, Y.; Samulski, R.J.; Walsh,
C.E. Sustained and complete phenotype correction of
hemophilia B mice following intramuscular injection of AAV1
serotype vectors. Mol. Ther. 2001, 4, 217–222. 28. Gao,
G.P.; Alvira, M.R.; Wang, L.; Calcedo, R.; Johnston, J.;
Wilson, J.M. Novel adeno-associated viruses from rhesus
monkeys as vectors for human gene therapy. Proc. Natl.
Acad. Sci. U. S. A. 2002, 99, 11854–11859. 29. Brand, A.;
Griffiths, D.J.; Herve, C.; Mallon, E.; Venables, P.J. Human
retrovirus-5 in rheumatic disease. J. Autoimmun. 1999, 13,
149–154. 30. Bushman, F.D. Integration site selection by
lentiviruses: Biology and possible control. Curr. Top.
Microbiol. Immunol. 2002, 261, 165–177. 31. Ghazizadeh, S.;
Carroll, J.M.; Taichman, L.B. Repression of
retrovirus-mediated transgene expression by interferons:
Implications for gene therapy. J. Virol. 1997, 71,
9163–9169. 32. Tolba, K.A.; Bowers, W.J.; Eling, D.J.;
Casey, A.E.; Kipps, T.J.; Federoff, H.J.; Rosenblatt, J.D.
HSV amplicon-mediated delivery of LIGHT enhances the
antigen-presenting capacity of chronic lymphocytic
leukemia. Mol. Ther. 2002, 6, 455–463. 33. Delman, K.A.;
Zager, J.S.; Bennett, J.J.; Malhotra, S.; Ebright, M.I.;
McAuliffe, P.F.; Halterman, M.W.; Federoff, H.J.; Fong, Y.
Efficacy of multiagent herpes simplex virus
amplicon-mediated immunotherapy as adjuvant treatment for
experimental hepatic cancer. Ann. Surg. 2002, 236, 337–342.
Discussion 342–333. 34. Everly, D.N., Jr.; Feng, P.; Mian,
I.S.; Read, G.S. mRNA degradation by the virion host
shutoff (Vhs) protein of herpes simplex virus: Genetic and
biochemical evidence that Vhs is a nuclease. J. Virol.
2002, 76, 8560–8571. 35. Farassati, F.; Yang, A.D.; Lee,
P.W. Oncogenes in Ras signaling pathway dictate host-cell
permissiveness to herpes simplex virus 1. Nat. Cell. Biol.
2001, 3, 745–750. 36. van Dillen, I.J.; Mulder, N.H.;
Vaalburg, W.; de Vries, E.F.; Hospers, G.A. Influence of the
bystander effect on HSV-tk/GCV gene therapy. A review.
Curr. Gene Ther. 2002, 2, 307–322. 37. Federoff, H.J.;
Geschwind, M.D.; Geller, A.I.; Kessler, J.A. Expression of
nerve growth factor in vivo from a defective herpes simplex
virus 1 vector prevents effects of axotomy on sympathetic
ganglia. Proc. Natl. Acad. Sci. U. S. A. 1992, 89,
1636–1640. 38.
http://www.wiley.co.uk/wileychi/genmed/clinical (accessed
February 2004). 39. Webster, A.; Leith, I.R.; Nicholson,
J.; Hounsell, J.; Hay, R.T. Role of preterminal protein
processing in adenovirus replication. J. Virol. 1997, 71,
6381–6389. 40. McNees, A.L.; Garnett, C.T.; Gooding, L.R.
The adenovirus E3 RID complex protects some cultured human
T and B lymphocytes from Fas-induced apoptosis. J. Virol.
2002, 76, 9716–9723. 41. Reddy, P.S.; Sakhuja, K.; Ganesh,
S.; Yang, L.; Kayda, D.; Brann, T.; Pattison, S.;
Golightly, D.; Idamakanti, N.; Pinkstaff, A.; Kaloss, M.;
Barjot, C.; Chamberlain, J.S.; Kaleko, M.; Connelly, S.
Sustained human factor VIII expression in hemophilia a mice
following systemic delivery of a gutless adenoviral vector.
Mol. Ther. 2002, 5, 63–73. 42. Toietta, G.; Pastore, L.;
Cerullo, V.; Finegold, M.; Beaudet, A.L.; Lee, B.
Generation of helper-dependent adenoviral vectors by
homologous recombination. Mol. Ther. 2002, 5, 204–210. 43.
Janeway, C.; Travers, P.; Walport, M.; Capra, D.
Immunobiology: The Immune System in Health and Disease, 4th
Ed.; Elsevier Science Limited: New York, 1999. 44.
Mayer-Kuckuk, P.; Banerjee, D.; Kemeny, N.; Fong, Y.;
Bertino, J.R. Molecular therapies for colorectal cancer
metastatic to the liver. Mol. Ther. 2002, 5, 492–500. 45.
Harjai, K.J.; Chowdhury, P.; Grines, C.L. Therapeutic
angiogenesis: A fantastic new adventure. J. Interv.
Cardiol. 2002, 15, 223–229. 46. Rutanen, J.; Markkanen, J.;
Yla-Herttuala, S. Gene therapy for restenosis: Current
status. Drugs 2002, 62, 1575–1585. 47. Otsu, M.; Candotti,
F. Gene therapy in infants with severe combined
immunodeficiency. BioDrugs 2002, 16, 229–239. 48. Bragonzi,
A.; Conese, M. Non-viral approach toward gene therapy of
cystic fibrosis lung disease. Curr. Gene Ther. 2002, 2,
295–305. G

49. Thioudellet, C.; Blot, S.; Squiban, P.; Fardeau, M.;


Braun, S. Current protocol of a research phase I clinical
trial of full-length dystrophin plasmid DNA in
Duchenne/Becker muscular dystrophies. Part I: Rationale.
Neuromuscul. Disord. 2002, 12 (Suppl), S49.

50. Klink, D.T.; Glick, M.C.; Scanlin, T.F. Gene therapy of


cystic fibrosis (CF) airways: A review emphasizing targeting
with lactose. Glycoconj. J. 2001, 18, 731–740.

51. Zwiebel, J.A. Cancer gene and oncolytic virus therapy.


Semin. Oncol. 2001, 28, 336–343.

52. Morris, M.J.; Scher, H.I. Novel therapies for the


treatment of prostate cancer: Current clinical trials and
development strategies. Surg. Oncol. 2002, 11, 13–23.

53. Herman, J.R.; Adler, H.L.; Aguilar-Cordova, E.;


Rojas-Martinez, A.; Woo, S.; Timme, T.L.; Wheeler, T.M.;
Thompson, T.C.; Scardino, P.T. In situ gene therapy for
adenocarcinoma of the prostate: A phase I clinical trial.
Hum. Gene Ther. 1999, 10, 1239–1249.

54. Shalev, M.; Kadmon, D.; Teh, B.S.; Butler, E.B.;


Aguilar-Cordova, E.; Thompson, T.C.; Herman, J.R.; Adler,
H.L.; Scardino, P.T.; Miles, B.J. Suicide gene therapy
toxicity after multiple and repeat injections in patients
with localized prostate cancer. J. Urol. 2000, 163,
1747–1750.

55. Navone, N.M.; Troncoso, P.; Pisters, L.L.; Goodrow,


T.L.; Palmer, J.L.; Nichols, W.W.; von Eschenbach, A.C.;
Conti, C.J. p53 protein accumulation and gene mutation in
the progression of human prostate carcinoma. J. Natl.
Cancer Inst. 1993, 85, 1657–1669.

56. Yang, C.; Cirielli, C.; Capogrossi, M.C.; Passaniti, A.


Adenovirus-mediated wild-type p53 expression induces
apoptosis and suppresses tumorigenesis of prostatic tumor
cells. Cancer Res. 1995, 55, 4210–4213.

57. Kaufman, R.J. Advances toward gene therapy for


hemophilia at the millennium. Hum. Gene Ther. 1999, 10,
2091–2107.

58. Kay, M.A.; et al. Evidence for gene transfer and


expression of factor IX in hemophilia B patients treated
with an AAV vector. Nat. Genet. 2000, 24, 257–261.

59. High, K.A. AAV-mediated gene transfer for hemophilia.


Ann. N. Y. Acad. Sci. 2001, 953, 64–74.

60. VandenDriessche, T.; Vanslembrouck, V.; Goovaerts, I.;


Zwinnen, H.; Vanderhaeghen, M.L.; Collen, D.; Chuah, M.K.
Long-term expression of human coagulation factor VIII and
correction of hemophilia a after in vivo retroviral gene
transfer in factor VIII-deficient mice. Proc. Natl. Acad.
Sci. U. S. A. 1999, 96, 10379–10384. 61. High, K.A. Gene
therapy: A 2001 perspective. Haemophilia 2001, 7 (Suppl 1),
23–27. 62. Roth, D.A.; Tawa, N.E., Jr.; O’Brien, J.M.;
Treco, D.A.; Selden, R.F. Factor VIII Transkaryotic therapy
study group. Nonviral transfer of the gene encoding
coagulation factor VIII in patients with severe hemophilia
A. N. Engl. J. Med. 2001, 344, 1735–1742. 63. Scriver, C.R.
The Metabolic and Molecular Bases of Inherited Disease;
McGraw-Hill: New York, 2001. 64. Fischer, A.; de
Saint-Basile, G.; Disanto, J.P.; Hacein-Bey, S.; Sharara,
L.; Cavazzana-Calvo, M. [Gene therapy for hereditary
immunodeficiencies]. C. R. Seances Soc. Biol. Fil. 1996,
190, 77–93. 65. Hacein-Bey-Abina, S.; Le Deist, F.;
Carlier, F.; Bouneaud, C.; Hue, C.; De Villartay, J.P.;
Thrasher, A.J.; Wulffraat, N.; Sorensen, R.; Dupuis-Girod,
S.; Fischer, A.; Davies, E.G.; Kuis, W.; Leiva, L.;
Cavazzana-Calvo, M. Sustained correction of X-linked severe
combined immunodeficiency by ex vivo gene therapy. N. Engl.
J. Med. 2002, 346, 1185–1193. 66. Check, E. Cancer fears
cast doubts on future of gene therapy. Nature 2003, 421,
678. 67. Check, E. Second cancer case halts gene-therapy
trials. Nature 2003, 421, 305. 68. Neale, G.A.; Rehg, J.E.;
Goorha, R.M. Disruption of T-cell differentiation precedes
T-cell tumor formation in LMO-2 (rhombotin-2) transgenic
mice. Leukemia 1997, 11 (Suppl 3), 289–290. 69.
http://www.prochoiceforum.org.uk/ri6.asp (accessed January
2003). 70. http://www.ncbiotech.org/biotech101/timeline.cfm
(accessed December 2002). 71.
http://www.bentham.org/cgt1-1/bronte/brontems.htm (accessed
November 2002). 72.
http://www.niaid.nih.gov/factsheets/stdherp.htm (accessed
March 2003). 73.
http://www-micro.msb.le.ac.uk/335/peel/peel1.html (accessed
November 2002).
Glucose-Responsive Hydrogels

1. Kudela, V. Hydrogels. In Encyclopedia of Polymer Science


and Technology; Mark, H.F., Kroschwitz, J.I., Eds.;
Wiley-Interscience: New York, NY, 1985; Vol. 7, 783–807.

2. Peppas, N.A. Physiologically responsive gels. J. Bioact.


Compat. Polym. 1991, 6, 241–246.

3. Heller, J. Modulated release from drug delivery devices.


Crit. Rev. Therap. Drug Carrier Syst. 1993, 10, 253–305.

4. Bell, C.L.; Peppas, N.A. Water, solute and protein


diffusion in physiologically responsive hydrogels of poly
(methacrylic acid-g-ethylene glycol). Biomaterials 1996,
17, 1203–1218.

5. Flory, P.J. Principles of Polymer Chemistry; Cornell


University Press: Ithaca, NY, 1953; 576–590.

6. Katchalsky, A.; Lifson, S.; Eisenberg, H. Equation of


swelling for polyelectrolyte gels. J. Polym. Sci. 1951, 7,
571–574.

7. Oppermann, W. Swelling behavior and elastic properties


of ionic hydrogels. In Polyelectrolyte Gels: Properties,
Preparation, and Applications; Harland, R.S., Prud’homme,
R.K., Eds.; American Chemical Society: Washington, DC,
1992; 159–170.

8. Mandel, M. Polyelecrolytes. In Encyclopedia of Polymer


Science and Technology; Mark, H.F., Kroschwitz, J.I., Eds.;
Wiley-Interscience: New York, NY, 1985; Vol. 11, 739–829.

9. Peppas, N.A.; Khare, A.R. Preparation structure, and


diffusional behavior of hydrogels in controlled release.
Adv. Drug Del. Rev. 1993, 11, 1–35.

10. Gehrke, S.H.; Lee, P.I. Hydrogels for drug delivery


systems. In Specialized Drug Delivery Systems; Tyle, P.,
Ed.; Marcel Dekker: New York, NY, 1990; 333–392.

11. Michaeli, I.; Katchalsky, A. Potentiometric titration


of polyelectrolye gels. J. Polym. Sci. 1957, 23, 683–696.

12. Siegal, R.A.; Firestone, B.A. pH-dependent equilibrium


swelling properties of hydrophobic polyelectrolyte
copolymer gels. Macromolecules 1988, 21, 3254–3529.

13. Tanaka, T. Phase transitions in gels and a single


polymer. Polymer 1979, 20, 1404–1412.

14. Ishihara, K.; Matsui, K. Glucose responsive insulin


release from polymer capsule. J. Polym. Sci.: Polym. Lett.
Ed. 1986, 24, 413–417. 15. Ishihara, K.; Kobayashi, M.;
Ishimaru, N.; Shinohara, I. Glucose induced permeation
control of insulin through a complex membrane consisting of
immobilized glucose oxidase and a poly(amine). Polymer J.
1984, 16, 625–631. 16. Peppas, N.A.; Langer, R. New
challenges in biomaterials. Science 1994, 263, 1715–1720.
17. Okano, T.; Yoshida, R. Intelligent polymeric materials
for drug delivery. In Biomedical Applications of Polymeric
Materials; CRC Press: Boca Raton, FL, 1993; 407–428. 18.
Jacobs, I.C.; Mason, N.S. Polymer delivery systems
concepts. In Polymeric Delivery Systems: Properties and
Applications; El-Nokaly, M.A., Piatt, D.M., Charpentier,
B.A., Eds.; American Chemical Society, Washington, DC,
1993: Vol. 520, 1–5. 19. Ashcroft, F.M., Ashcroft, S.J.H.,
Eds.; Insulin: Molecular Biology to Pathology; IRL; Oxford:
NY, New York, 1992. 20. Guyton, A.C. Textbook of Medical
Physiology; Saunders: Philadelphia, PA, 1971. 21.
Decoursey, R.M. The Human Organism; McGraw Hill: New York,
NY, 1974. 22. Wallum, B.J.; Kahn, S.E.; McCulloch, D.K.;
Porte, D. Insulin secretion in the normal and diabetic
human. In International Textbook of Diabetes Mellitus;
Alberti, K.G.M.M., DeFronzo, R.A., Keen, H., Zimmet, P.,
Eds.; Wiley: New York, NY, 1992; 285–301. 23. Krall, L.P.;
Beaser, R.S. Joslin Diabetes Manual, 12th Ed.; Lea and
Febiger: Philadelphia, PA, 1989. 24. Kennedy, F.P. Recent
developments in insulin delivery techniques: current status
and future potential. Drugs 1991, 42, 213–227. 25.
Goldraich, M.; Kost, J. Glucose-sensitive polymeric
matrices for controlled drug delivery. Clinical Materials
1993, 13, 135–142. 26. Horbett, T.A.; Rattner, B.D.; Kost,
J.; Singh, M. A bioresponsive membrane for insulin
delivery. In Recent Advances in Drug Delivery Systems;
Anderson, J.M., Kim, S.W., Eds.; Plenum: New York, NY,
1983; 209–220. 27. Ito, Y.; Casolaro, M.; Kono, K.;
Imanishi, Y. An insulin-releasing system that is responsive
to glucose. J. Control. Release 1989, 13, 195–203. 28.
Heller, J.; Chang, A.C.; Rodd, G.; Grodsky, G.M. Release of
insulin from pH-sensitive poly(ortho esters). J. Control.
Release 1990, 13, 295–302. 29. Ishihara, K.; Kobayashi, M.;
Shionohara, I. Control of insulin permeation through a
polymer membrane with responsive function for glucose.
Makromol. Chem. Rapid Commun. 1983, 4, 327–331. 30. Jeong,
S.Y.; Kim, S.W. Self-regulating insulin delivery systems.
III. In vivo studies. J. Control. Release 1985, 2, 143–152.
31. Osada, Y.; Takeuchi, Y. Water and protein permeation
through polymeric membrane having mechanochemically
expanding and contracting pores. Function of chemical
valve. J. Polym. Sci.: Polym. Lett. Ed. 1981, 19, 303–308.
32. Osada, Y. Effects of polymers and their chain lengths
on contraction of poly(methacrylic acid) network. J. Polym.
Sci.: Polym. Lett. Ed. 1980, 18, 281–286. 33. Klier, J.;
Scranton, A.B.; Peppas, N.A. Self-associating networks of
poly(methacrylic acid-g-ethylene glycol). Macromolecules
1990, 23, 4944–4949. 34. Dorski, C.M.; Doyle, F.J.; Peppas,
N.A. Glucoseresponsive, complexation hydrogels. Polym.
Prepr. 1996, 37 (1), 475–476. 35. Podual, K.; Doyle, F.J.,
III; Peppas, N.A. Dynamic behavior of
glucose-oxidase-containing microparticles of poly(ethylene
glycol)-grafted cationic hydrogels in an environment of
changing pH. Biomaterials 2000, 21, 1439–1450. 36. Langer,
R.; Peppas, N.A. Advances in biomaterials, drug delivery,
and bionanotechnology. AIChE J. 2003, 49, 2990–3006. 37.
Foss, A.C.; Goto, T.; Morishita, M.; Peppas, N.A.
Development of acrylic-based copolymers for oral insulin
delivery. Europ. J. Pharm. Biopharmac. 2004, 57, 163–169.
38. Peppas, N.A. Kinetics of smart hydrogels. In Reflexive
Polymers and Hydrogels: Understanding and Designing
Fast-Responsive Polymeric Systems; Yui, N., Mrsny, R.,
Park, K., Eds.; CRC Press: Boca Rton, FL, 2004; 99–113. 39.
Morishita, M.; Goto, T.; Peppas, N.A.; Joseph, J.I.;
Torjman, M.C.; Munsick, C.; Nakamura, K.; Takayama, K.;
Lowman, A.M. Mucosal insulin delivery systems based on
complexation polymer hydrogels: effect of particle size on
insulin enteral absorption. J. Control. Release 2004, 97,
115–124. 40. Peppas, N.A. Devices based on intelligent
biopolymers for oral protein delivery. Intern. J. Pharmac.
2004, 277, 11–17. 41. Podual, K.; Doyle, F.J., III; Peppas,
N.A. Glucosesensitive cationic hydrogels: preparation
characterization and modeling of swelling properties. In
Biomaterials Carriers for Drug Delivery, and Scaffolds for
Tissue Engineering; Peppas, N.A., Mooney, D.J., Mikos,
A.G., Brannon-Peppas, L., Eds.; AIChE: New York, NY, 1997;
190–192. 42. Podual, K.; Doyle, F.J., III; Peppas, N.A.
Preparation and characterization of the dynamic response of
cationic copolymer hydrogels containing glucose oxidase.
Polymer 2000, 41, 3975–3983. 43. Podual, K.; Doyle, F.J.,
III; Peppas, N.A. Glucosesensitivity of glucose
oxidase-containing cationic copolymer hydrogels having
poly(ethylene glycol) grafts. J. Control. Release 2000, 67,
9–17. 44. Parker, R.S.; Rabinovitch, K.L.; Doyle, F.J.,
III; Peppas, N.A. Control analysis of pancreas models for
optimal insulin delivery. In Biomaterials Carriers for Drug
Delivery, and Scaffolds For Tissue Engineering; Peppas,
N.A., Mooney, D.J., Mikos, A.G., Brannon-Peppas, L., Eds.;
Aiche: New York, NY, 1997; 319–321. 45. Doyle, F.J., III;
Dorski, C.M.; Harting, J.E.; Peppas, N.A. Control and
modeling of drug delivery devices for treatment of
diabetes. Proc. Am. Cont. Conf. 1995, 1, 776–780. 46.
Podual, K.; Peppas, N.A.; Doyle, F.J., III. Insulin release
from pH-Sensitive cationic hydrogels. Proceed. Intern.
Symp. Control. Rel. Bioact. Mater. 1998, 25, 56–57. 47.
Schwarte, L.M.; Peppas, N.A. Preparation and
characterization of peg-containing pH-sensitive, cationic
hydrogels for drug delivery applications. Polym. Prepr.
1997, 38 (2), 596–597. 48. Parker, R.S.; Doyle, F.J., III;
Peppas, N.A. A modelbased algorithm for blood glucose
control in type 1 diabetic patients. Ieee Trans. Biomed.
Eng. 1999, 46, 148–157. 49. Peppas, N.A. Is there a future
in glucose-sensitive, responsive insulin delivery. J. Drug
Deliv. Sci. Techn. 2004, 14, 247–256. 50. Schwarte, L.M.;
Podual, K.; Peppas, N.A. Cationic hydrogels for controlled
release of proteins and other macromolecules. In Tailored
Polymeric Materials for Controlled Delivery Systems ACS
Symposium Series; ACS: Washington, DC, 1999; 709, 56–66.
51. Parker, R.S.; Ward, J.H.; Doyle, F.J., III.; Peppas,
N.A. Robust control in diabetes using a physiological
model. Aiche J. 2000, 46, 2537–2549. 52. Dorski, C.M.;
Doyle, F.J., III; Peppas, N.A. Preparation and
characterization of glucose-sensitive P(MAAg-EG) hydrogels.
Polym. Mater. Sci. Eng. Proceed. 1997, 76, 281–282. 53.
Hassan, C.M.; Doyle, F.J., III; Peppas, N.A. Dynamic
behavior of glucose-responsive poly(methacrylic
acidg-ethylene glycol) hydrogels. Macromolecules 1997, 30,
6166–6173. 54. Schwarte, L.M.; Peppas, N.A. Novel
poly(ethylene glycol)-grafted cationic hydrogels:
preparation, characterization and diffusive properties.
Polymer 1998, 39, 6057–6066. G
Glucose Sensors

31. Vatner, S.F.; Higgins, C.B.; White, S.; Patric, T.;


Franklin, D. The peripheral vascular response to severe
exercise in untethered dogs before and after complete heart
block. J. Clin. Invest. 1971, 50, 1950–1960. 32. Van
Citters, R.L.; Franklin, D.L. Cardiovascular performance of
Alaska sled dogs in exercise. Circ. Res. 1969, 24, 33–42.
33. Bednarik, J.A.; May, C.N. Evaluation of a transittime
system for the chronic measurement of blood flow in
conscious sheep. J. Appl. Physiol. 1995, 78, 524–530.
Glues

23. Tanaka, K.; Takamoto, S.; Ohtsuka, T.; Kotsuka, Y.;


Kawauchi, M. Application of AdvaSeal for acute aortic
dissection: Experimental study. Ann. Thorac. Surg. 1999, 68
(4), 1308–1313.

24. Nakayama, Y.; Matsuda, T. Newly designed hemostatic


technology based on photocurable gelatin. ASAIO J. 1995, 41
(3), M374–M378. 25. Auteri, J.S.; Oz, M.C.; Jeevanandam,
V.; Sanchez, J.A.; Treat, M.R.; Smith, C.R. Laser
activation of tissue sealant in hand-sewn canine esophageal
closure. J. Thorac. Cardiovasc. Surg. 1992, 103 (4),
781–783. 26. Ono, K.; Saito, Y.; Yura, H.; Ishikawa, K.;
Kurita, A.; Akaike, T.; Ishihara, M. Photocrosslinkable
chitosan as a biological adhesive. J. Biomed. Mater. Res.
2000, 49 (2), 289–295.
Glycosaminoglycans and Proteoglycans

1. Li, J.P.; Galvis, M.L.; Gong, F.; Zhang, X.; Zcharia,


E.; Metzger, S.; Vlodavsky, I.; Kisilevsky, R.; Lindahl, U.
In vivo fragmentation of heparan sulfate by heparanase
overexpression renders mice resistant to amyloid protein A
amyloidosis. Proc. Natl. Acad. Sci. USA 2005, 102 (18),
6473–6477.

2. Gama, C.I.; Hsieh-Wilson, L.C. Chemical approaches to


deciphering the glycosaminoglycan code. Curr. Opin. Chem.
Biol. 2005, 9 (6), 609–619.

3. Buckwalter, J.A.; Mankin, H.J.; Grodzinsky, A.J.


Articular cartilage and osteoarthritis. Instr. Course Lect.
2005, 54, 465–480.

4. Kiani, C.; Chen, L.; Wu, Y.J.; Yee, A.J.; Yang, B.B.
Structure and function of aggrecan. Cell Res. 2002, 12 (1),
19–32.

5. Knudson, C.B.; Knudson, W. Cartilage proteoglycans.


Semin. Cell Dev. Biol. 2001, 12 (2), 69–78.

6. Luo, W.; Guo, C.; Zheng, J.; Chen, T.L.; Wang, P.Y.;
Vertel, B.M.; Tanzer, M.L. Aggrecan from start to finish. J.
Bone Miner. Metab. 2000, 18 (2), 51–56.

7. Sztrolovics, R.; Recklies, A.D.; Roughley, P.J.; Mort,


J.S. Hyaluronate degradation as an alternative mechanism
for proteoglycan release from cartilage during
interleukin-1beta-stimulated catabolism. Biochem. J. 2002,
362 (Pt 2), 473–479.

8. Roughley, P.J. Articular cartilage and changes in


arthritis: noncollagenous proteins and proteoglycans in the
extracellular matrix of cartilage. Arthritis Res. 2001, 3
(6), 342–347.

9. Schwartz, N. Biosynthesis and regulation of expression


of proteoglycans. Front. Biosci. 2000, 5, D649–655.

10. Shi, S.; Grothe, S.; Zhang, Y.; O’Connor-McCourt, M.D.;


Poole, A.R.; Roughley, P.J.; Mort, J.S. Link protein has
greater affinity for versican than aggrecan. J. Biol. Chem.
2004, 279 (13), 12,060–12,066.

11. Sztrolovics, R.; Grover, J.; Cs-Szabo, G.; Shi, S.L.;


Zhang, Y.; Mort, J.S.; Roughley, P.J. The characterization
of versican and its message in human articular cartilage
and intervertebral disc. J. Orthop. Res. 2002, 20 (2),
257–266. 12. Roughley, P.J.; White, R.J.; Cs-Szabo, G.;
Mort, J.S. Changes with age in the structure of fibromodulin
in human articular cartilage. Osteoarth. Cartil. 1996, 4
(3), 153–161. 13. Grover, J.; Chen, X.N.; Korenberg, J.R.;
Roughley, P.J. The human lumican gene. Organization,
chromosomal location, and expression in articular
cartilage. J. Biol. Chem. 1995, 270 (37), 21,942–21,949.
14. Hildebrand, A.; Romaris, M.; Rasmussen, L.M.;
Heinegard, D.; Twardzik, D.R.; Border, W.A.; Ruoslahti, E.
Interaction of the small interstitial proteoglycans
biglycan, decorin and fibromodulin with transforming growth
factor beta. Biochem. J. 1994, 302 (Pt 2), 527–534. 15.
Tesche, F.; Miosge, N. Perlecan in late stages of
osteoarthritis of the human knee joint. Osteoarth. Cartil.
2004, 12 (11), 852–862. 16. SundarRaj, N.; Fite, D.;
Ledbetter, S.; Chakravarti, S.; Hassell, J.R. Perlecan is a
component of cartilage matrix and promotes chondrocyte
attachment. J. Cell Sci. 1995, 108 (Pt 7), 2663–2672. 17.
French, M.M.; Gomes, R.R., Jr.; Timpl, R.; Hook, M.;
Czymmek, K.; Farach-Carson, M.C.; Carson, D.D. Chondrogenic
activity of the heparan sulfate proteoglycan perlecan maps
to the N-terminal domain I. J. Bone Miner. Res. 2002, 17
(1), 48–55. 18. Jay, G.D.; Tantravahi, U.; Britt, D.E.;
Barrach, H.J.; Cha, C.J. Homology of lubricin and
superficial zone protein (SZP): products of megakaryocyte
stimulating factor (MSF) gene expression by human synovial
fibroblasts and articular chondrocytes localized to
chromosome 1q25. J. Orthop. Res. 2001, 19 (4), 677– 687.
19. Flannery, C.R.; Hughes, C.E.; Schumacher, B.L.; Tudor,
D.; Aydelotte, M.B.; Kuettner, K.E.; Caterson, B. Articular
cartilage superficial zone protein (SZP) is homologous to
megakaryocyte stimulating factor precursor and is a
multifunctional proteoglycan with potential
growth-promoting, cytoprotective, and lubricating
properties in cartilage metabolism. Biochem. Biophys. Res.
Commun. 1999, 254 (3), 535–541. 20. Spicer, A.P.;
Augustine, M.L.; McDonald, J.A. Molecular cloning and
characterization of a putative mouse hyaluronan synthase.
J. Biol. Chem. 1996, 271 (38), 23,400–23,406. 21. Spicer,
A.P.; Seldin, M.F.; Olsen, A.S.; Brown, N.; Wells, D.E.;
Doggett, N.A.; Itano, N.; Kimata, K.; Inazawa, J.;
McDonald, J.A. Chromosomal localization of the human and
mouse hyaluronan synthase genes. Genomics 1997, 41 (3),
493–497. 22. Spicer, A.P.; Olson, J.S.; McDonald, J.A.
Molecular cloning and characterization of a cDNA encoding
the third putative mammalian hyaluronan synthase. J. Biol.
Chem. 1997, 272 (14), 8957–8961. 23. Spicer, A.P.;
McDonald, J.A. Characterization and molecular evolution of
a vertebrate hyaluronan synthase gene family. J. Biol.
Chem. 1998, 273 (4), 1923– 1932. 24. Camenisch, T.D.;
Spicer, A.P.; Brehm-Gibson, T.; Biesterfeldt, J.;
Augustine, M.L.; Calabro, A., Jr.; Kubalak, S.; Klewer,
S.E.; McDonald, J.A. Disruption of hyaluronan synthase-2
abrogates normal cardiac morphogenesis and
hyaluronan-mediated transformation of epithelium to
mesenchyme. J. Clin. Invest. 2000, 106 (3), 349–360. 25.
Itano, N.; Sawai, T.; Yoshida, M.; Lenas, P.; Yamada, Y.;
Imagawa, M.; Shinomura, T.; Hamaguchi, M.; Yoshida, Y.;
Ohnuki, Y.; Miyauchi, S.; Spicer, A.P.; McDonald, J.A.;
Kimata, K. Three isoforms of mammalian hyaluronan synthases
have distinct enzymatic properties. J. Biol. Chem. 1999,
274 (35), 25,085– 25,092. 26. Recklies, A.D.; White, C.;
Melching, L.; Roughley, P.J. Differential regulation and
expression of hyaluronan synthases in human articular
chondrocytes, synovial cells and osteosarcoma cells.
Biochem. J. 2001, 354 (Pt 1), 17–24. 27. Mrosek, E.;
Erggelet, C.; McDonald, J.A.; Kurz, H. Hyaluronan synthases
in normal and regenerating joint cartilage. Cells Tissues
Organs 2003, 173 (2), 93–104. 28. Mrosek, E.H.; Lahm, A.;
Erggelet, C.; Uhl, M.; Kurz, H.; Eissner, B.; Schagemann,
J.C. Subchondral bone trauma causes cartilage matrix
degeneration: an immunohistochemical analysis in a canine
model. Osteoarth. Cartil. 2006, 14 (2), 171–178. 29.
Cs-Szabo, G.; Roughley, P.J.; Plaas, A.H.; Glant, T.T.
Large and small proteoglycans of osteoarthritic and
rheumatoid articular cartilage. Arthritis Rheum. 1995, 38
(5), 660–668. 30. Pearle, A.D.; Warren, R.F.; Rodeo, S.A.
Basic science of articular cartilage and osteoarthritis.
Clin. Sports Med. 2005, 24 (1), 1–12. 31. Kuettner, K.E.;
Cole, A.A. Cartilage degeneration in different human
joints. Osteoarth. Cartil. 2005, 13 (2), 93–103. 32. Poole,
A.R. Biochemical/immunochemical biomarkers of
osteoarthritis: utility for prediction of incident or
progressive osteoarthritis. Rheum. Dis. Clin. North Am.
2003, 29 (4), 803–818. 33. Kuettner, K.E. Biochemistry of
articular cartilage in health and disease. Clin. Biochem.
1992, 25 (3), 155–163. 34. Doege, K.J.; Coulter, S.N.;
Meek, L.M.; Maslen, K.; Wood, J.G. A human-specific
polymorphism in the coding region of the aggrecan gene.
Variable number of tandem repeats produce a range of core
protein sizes in the general population. J. Biol. Chem.
1997, 272 (21), 13,974–13,979. 35. Martin, J.A.; Brown,
T.D.; Heiner, A.D.; Buckwalter, J.A. Chondrocyte
senescence, joint loading and osteoarthritis. Clin. Orthop.
Relat. Res. 2004, 427 (Suppl), S96–103. 36. Lee, V.; Chen,
L.; Paiwand, F.; Cao, L.; Wu, Y.; Inman, R.; Adams, M.E.;
Yang, B.B. Cleavage of the carboxyl tail from the G3 domain
of aggrecan but not versican and identification of the amino
acids involved in the degradation. J. Biol. Chem. 2002, 277
(25), 22,279–22,288. 37. Cs-Szabo, G.; Melching, L.I.;
Roughley, P.J.; Glant, T.T. Changes in messenger RNA and
protein levels of proteoglycans and link protein in human
osteoarthritic cartilage samples. Arthritis Rheum. 1997, 40
(6), 1037–1045. 38. Sztrolovics, R.; White, R.J.; Poole,
A.R.; Mort, J.S.; Roughley, P.J. Resistance of small
leucine-rich repeat proteoglycans to proteolytic
degradation during interleukin-1-stimulated cartilage
catabolism. Biochem. J. 1999, 339 (Pt 3), 571–577. 39.
Whiteman, P.; Henderson, H. A method for the determination
of amniotic-fluid glycosaminoglycans and its application to
the prenatal diagnosis of Hurler and Sanfilippo diseases.
Clin. Chim. Acta. 1977, 79 (1), 99–105. 40. Danes, B.S.;
Queenan, J.T.; Gadow, E.C.; Cederqvist, L.L. Antenatal
diagnosis of mucopolysaccharidoses. Lancet 1970, 1 (7653),
946–947. 41. Kiraly, K.; Lapvetelainen, T.; Arokoski, J.;
Torronen, K.; Modis, L.; Kiviranta, I.; Helminen, H.J.
Application of selected cationic dyes for the
semiquantitative estimation of glycosaminoglycans in
histological sections of articular cartilage by
microspectrophotometry. Histochem. J. 1996, 28 (8),
577–590. 42. O’Driscoll, S.W.; Marx, R.G.; Fitzsimmons,
J.S.; Beaton, D.E. Method for automated cartilage
histomorphometry. Tissue Eng. 1999, 5 (1), 13–23. 43.
Martin, I.; Obradovic, B.; Freed, L.E.; Vunjak-Novakovic,
G. Method for quantitative analysis of glycosaminoglycan
distribution in cultured natural and engineered cartilage.
Ann. Biomed. Eng. 1999, 27 (5), 656–662. 44. Burkhardt, D.;
Hwa, S.Y.; Ghosh, P. A novel microassay for the
quantitation of the sulfated glycosaminoglycan content of
histological sections: its application to determine the
effects of Diacerhein on cartilage in an ovine model of
osteoarthritis. Osteoarth. Cartil. 2001, 9 (3), 238–247.
45. Thuy, L.P.; Nyhan, W.L. A new quantitative assay for
glycosaminoglycans. Clin. Chim. Acta. 1992, 212 (1–2),
17–26. 46. Farndale, R.W.; Sayers, C.A.; Barrett, A.J. A
direct spectrophotometric microassay for sulfated
glycosaminoglycans in cartilage cultures. Connect. Tissue
Res. 1982, 9 (4), 247–248. 47. Farndale, R.W.; Buttle,
D.J.; Barrett, A.J. Improved quantitation and
discrimination of sulphated glycosaminoglycans by use of
dimethylmethylene blue. Biochim. Biophys. Acta. 1986, 883
(2), 173–177. 48. Chandrasekhar, S.; Esterman, M.A.;
Hoffman, H.A. Microdetermination of proteoglycans and
glycosaminoglycans in the presence of guanidine
hydrochloride. Anal. Biochem. 1987, 161 (1), 103–108. 49.
Barbosa, I.; Garcia, S.; Barbier-Chassefiere, V.; Caruelle,
J.P.; Martelly, I.; Papy-Garcia, D. Improved and simple
micro assay for sulfated glycosaminoglycans quantification
in biological extracts and its use in skin and muscle
tissue studies. Glycobiology 2003, 13 (9), 647–653. 50.
Volpi, N.; Maccari, F. Detection of submicrogram quantities
of glycosaminoglycans on agarose gels by sequential
staining with toluidine blue and Stains-All.
Electrophoresis 2002, 23 (24), 4060–4066. 51. Volpi, N.;
Maccari, F.; Titze, J. Simultaneous detection of
submicrogram quantities of hyaluronic acid and dermatan
sulfate on agarose-gel by sequential staining with G
toluidine blue and Stains-All. J. Chromatogr. B Analyt.
Technol. Biomed. Life Sci. 2005, 820 (1), 131–135.

52. Horner, A.A. Electrophoresis of acidic


mucopolysaccharides in agarose gel. Can. J. Biochem. 1967,
45 (7), 1009–1013.

53. Funderburgh, J.L.; Chandler, J.W. An agarose gel


electrophoretic method for analysis of sulfated
glycosaminoglycans of cultured cells. Anal. Biochem. 1978,
91 (2), 464–472.

54. Maccari, F.; Volpi, N. Direct and specific recognition


of glycosaminoglycans by antibodies after their separation
by agarose gel electrophoresis and blotting on
cetylpyridinium chloride-treated nitrocellulose membranes.
Electrophoresis 2003, 24 (9), 1347–1352.

55. Bashir, A.; Gray, M.L.; Boutin, R.D.; Burstein, D.


Glycosaminoglycan in articular cartilage: in vivo
assessment with delayed Gd(DTPA)(2-)-enhanced MR imaging.
Radiology 1997, 205 (2), 551–558.

56. Bashir, A.; Gray, M.L.; Burstein, D. Gd-DTPA2as a


measure of cartilage degradation. Magn. Reson. Med. 1996,
36 (5), 665–673.

57. Bashir, A.; Gray, M.L.; Hartke, J.; Burstein, D.


Nondestructive imaging of human cartilage glycosaminoglycan
concentration by MRI. Magn. Reson. Med. 1999, 41 (5),
857–865.

58. Tiderius, C.J.; Olsson, L.E.; Leander, P.; Ekberg, O.;


Dahlberg, L. Delayed gadolinium-enhanced MRI of cartilage
(dGEMRIC) in early knee osteoarthritis. Magn. Reson. Med.
2003, 49 (3), 488–492.

59. Williams, A.; Gillis, A.; McKenzie, C.; Po, B.; Sharma,
L.; Micheli, L.; McKeon, B.; Burstein, D. Glycosaminoglycan
distribution in cartilage as determined by delayed
gadolinium-enhanced MRI of cartilage (dGEMRIC): potential
clinical applications. AJR. Am. J. Roentgenol. 2004, 182
(1), 167–172.

60. Gillis, A.; Bashir, A.; McKeon, B.; Scheller, A.; Gray,
M.L.; Burstein, D. Magnetic resonance imaging of relative
glycosaminoglycan distribution in patients with autologous
chondrocyte transplants. Invest. Radiol. 2001, 36 (12),
743–748.

61. Gillis, A.; Gray, M.; Burstein, D. Relaxivity and


diffusion of gadolinium agents in cartilage. Magn. Reson.
Med. 2002, 48 (6), 1068–1071.

62. Neufeld, E.F. Lysosomal storage diseases. Annu. Rev.


Biochem. 1991, 60, 257–280.

63. O’Donnell, C.M.; Kaczman-Daniel, K.; Goetinck, P.F.;


Vertel, B.M. Nanomelic chondrocytes synthesize a
glycoprotein related to chondroitin sulfate proteoglycan
core protein. J. Biol. Chem. 1988, 263 (33), 17,749–
17,754.

64. Mourao, P.A.; Kato, S.; Donnelly, P.V.


Spondyloepiphyseal dysplasia, chondroitin sulfate type: a
possible defect of PAPS—chondroitin sulfate
sulfotransferase in humans. Biochem. Biophys. Res. Commun.
1981, 98 (2), 388–396.

65. Yamaguchi, Y.; Ruoslahti, E. Expression of human


proteoglycan in Chinese hamster ovary cells inhibits cell
proliferation. Nature 1988 , 336 (6196), 244–246.

66. Rosenberg, L.C.; Choi, H.U.; Tang, L.H.; Johnson, T.L.;


Pal, S.; Webber, C.; Reiner, A.; Poole, A.R. Isolation of
dermatan sulfate proteoglycans from mature bovine articular
cartilages. J. Biol. Chem. 1985, 260 (10), 6304–6313. 67.
Quentin, E.; Gladen, A.; Roden, L.; Kresse, H. A genetic
defect in the biosynthesis of dermatan sulfate
proteoglycan: galactosyltransferase I deficiency in
fibroblasts from a patient with a progeroid syndrome. Proc.
Natl. Acad. Sci. USA 1990, 87 (4), 1342–1346. 68. Simonaro,
C.M.; D’Angelo, M.; Haskins, M.E.; Schuchman, E.H. Joint
and bone disease in mucopolysaccharidoses VI and VII:
identification of new therapeutic targets and biomarkers
using animal models. Pediatr. Res. 2005, 57 (5 Pt 1),
701–707. 69. Wraith, J.E.; Clarke, L.A.; Beck, M.; Kolodny,
E.H.; Pastores, G.M.; Muenzer, J.; Rapoport, D.M.; Berger,
K.I.; Swiedler, S.J.; Kakkis, E.D.; Braakman, T.;
Chadbourne, E.; Walton-Bowen, K.; Cox, G.F. Enzyme
replacement therapy for mucopolysaccharidosis I: a
randomized, double-blinded, placebo-controlled,
multinational study of recombinant human
alpha-L-iduronidase (laronidase). J. Pediatr. 2004, 144
(5), 581–588. 70. Felson, D.T.; McAlindon, T.E. Glucosamine
and chondroitin for osteoarthritis: to recommend or not to
recommend? Arthritis Care Res. 2000, 13 (4), 179–182. 71.
Lyman, S.; Sherman, S.; Dunn, W.R.; Marx, R.G. Advancements
in the surgical and alternative treatment of arthritis.
Curr. Opin. Rheumatol. 2005, 17 (2), 129–133. 72. Fajardo,
M.; Di Cesare, P.E. Disease-modifying therapies for
osteoarthritis: current status. Drugs Aging 2005, 22 (2),
141–161. 73. McAlindon, T.E.; LaValley, M.P.; Gulin, J.P.;
Felson, D.T. Glucosamine and chondroitin for treatment of
osteoarthritis: a systematic quality assessment and
meta-analysis. Jama 2000, 283 (11), 1469–1475. 74.
McAlindon, T.; Formica, M.; LaValley, M.; Lehmer, M.;
Kabbara, K. Effectiveness of glucosamine for symptoms of
knee osteoarthritis: results from an internet-based
randomized double-blind controlled trial. Am. J. Med. 2004,
117 (9), 643–649. 75. McAlindon, T.E.; Biggee, B.A.
Nutritional factors and osteoarthritis: recent
developments. Curr. Opin. Rheumatol. 2005, 17 (5), 647–652.
76. Persiani, S.; Roda, E.; Rovati, L.C.; Locatelli, M.;
Giacovelli, G.; Roda, A. Glucosamine oral bioavailability
and plasma pharmacokinetics after increasing doses of
crystalline glucosamine sulfate in man. Osteoarth. Cartil.
2005, 13 (12), 1041–1049. 77. Setnikar, I.; Giacchetti, C.;
Zanolo, G. Pharmacokinetics of glucosamine in the dog and
in man. Arzneimittelforschung 1986, 36 (4), 729–735. 78.
Deal, C.L.; Moskowitz, R.W. Nutraceuticals as therapeutic
agents in osteoarthritis. The role of glucosamine,
chondroitin sulfate, and collagen hydrolysate. Rheum. Dis.
Clin. North Am. 1999, 25 (2), 379–395. 79. Biggee, B.A.;
McAlindon, T. Glucosamine for osteoarthritis: part II,
biologic and metabolic controversies. Med. Health R I.
2004, 87 (6), 180–181. 80. Biggee, B.A.; Blinn, C.M.;
McAlindon, T.E.; Nuite, M.; Silbert, J.E. Low levels of
human serum glucosamine after ingestion of glucosamine
sulphate relative to capability for peripheral
effectiveness. Ann. Rheum. Dis. 2006, 65 (2), 222–226. 81.
Adebowale, A.; Du, J.; Liang, Z.; Leslie, J.L.; Eddington,
N.D. The bioavailability and pharmacokinetics of
glucosamine hydrochloride and low molecular weight
chondroitin sulfate after single and multiple doses to
beagle dogs. Biopharm. Drug Dispos. 2002, 23 (6), 217–225.
82. Biggee, B.A.; Blinn, C.M.; McAlindon, T.E. Human serum
glucosamine and sulfate levels after ingestion of
glucosamine sulfate. Arthritis Rheum. 2004, 50, 657. 83.
Conte, A.; Volpi, N.; Palmieri, L.; Bahous, I.; Ronca, G.
Biochemical and pharmacokinetic aspects of oral treatment
with chondroitin sulfate. Arzneimittelforschung 1995, 45
(8), 918–925. 84. Adebowale, A.O.; Cox, D.S.; Liang, Z.;
Eddington, N.D. Analysis of glucosamine and chondroitin
sulfate content in marketed products and the Caco-2
permeability of chondroitin sulfate raw materials. J. Am.
Nutraceutical Assoc. 2000, 3, 37–44. 85. Rindone, J.P.;
Hiller, D.; Collacott, E.; Nordhaugen, N.; Arriola, G.
Randomized, controlled trial of glucosamine for treating
osteoarthritis of the knee. West J. Med. 2000, 172 (2),
91–94. 86. Hungerford, D.S.; Jones, L.C. Glucosamine and
chondroitin sulfate are effective in the management of
osteoarthritis. J. Arthroplasty 2003, 18 (3 Suppl 1), 5–9.
87. Morelli, V.; Naquin, C.; Weaver, V. Alternative
therapies for traditional disease states: osteoarthritis.
Am. Fam. Physician. 2003, 67 (2), 339–344. 88. Hungerford,
D.; Navarro, R.; Hammad, T. Use of nutraceuticals in the
management of osteoarthritis. J. Am. Nutraceutical Assoc.
2000, 3, 23–27. 89. Richy, F.; Bruyere, O.; Ethgen, O.;
Cucherat, M.; Henrotin, Y.; Reginster, J.Y. Structural and
symptomatic efficacy of glucosamine and chondroitin in knee
osteoarthritis: a comprehensive meta-analysis. Arch.
Intern. Med. 2003, 163 (13), 1514–1522. 90. Cibere, J.;
Kopec, J.A.; Thorne, A.; Singer, J.; Canvin, J.; Robinson,
D.B.; Pope, J.; Hong, P.; Grant, E.; Esdaile, J.M.
Randomized, double-blind, placebocontrolled glucosamine
discontinuation trial in knee osteoarthritis. Arthritis
Rheum. 2004, 51 (5), 738–745. 91. Cibere, J.; Thorne, A.;
Kopec, J.A.; Singer, J.; Canvin, J.; Robinson, D.B.; Pope,
J.; Hong, P.; Grant, E.; Lobanok, T.; Ionescu, M.; Poole,
A.R.; Esdaile, J.M. Glucosamine sulfate and cartilage type
II collagen degradation in patients with knee
osteoarthritis: randomized discontinuation trial results
employing biomarkers. J. Rheumatol. 2005, 32 (5), 896–902.
92. Michel, B.A.; Stucki, G.; Frey, D.; De Vathaire, F.;
Vignon, E.; Bruehlmann, P.; Uebelhart, D. Chondroitins 4
and 6 sulfate in osteoarthritis of the knee: a randomized,
controlled trial. Arthritis Rheum. 2005, 52 (3), 779–786.
93. Altman, R.D. Status of hyaluronan supplementation
therapy in osteoarthritis. Curr. Rheumatol. Rep. 2003, 5
(1), 7–14. 94. Kirchner, M.; Marshall, D. A double-blind
randomized controlled trial comparing alternate forms of
high molecular weight hyaluronan for the treatment of
osteoarthritis of the knee. Osteoarth. Cartil. 2006, 14
(2), 154–162. 95. Fuchs, S.; Monikes, R.; Wohlmeiner, A.;
Heyse, T. Intra-articular hyaluronic acid compared with
corticoid injections for the treatment of rhizarthrosis.
Osteoarth. Cartil. 2005, 2006, 14 (1), 82–88. 96. Felson,
D.T.; Anderson, J.J. Hyaluronate sodium injections for
osteoarthritis: hope, hype, and hard truths. Arch. Intern.
Med. 2002, 162 (3), 245–247. 97. Henderson, E.B.; Smith,
E.C.; Pegley, F.; Blake, D.R. Intra-articular injections of
750 kD hyaluronan in the treatment of osteoarthritis: a
randomised single centre double-blind placebo-controlled
trial of 91 patients demonstrating lack of efficacy. Ann.
Rheum. Dis. 1994, 53 (8), 529–534. 98. Lo, G.H.; LaValley,
M.; McAlindon, T.; Felson, D.T. Intra-articular hyaluronic
acid in treatment of knee osteoarthritis: a meta-analysis.
Jama 2003, 290 (23), 3115–3121. 99. Jubb, R.W.; Piva, S.;
Beinat, L.; Dacre, J.; Gishen, P. A one-year, randomised,
placebo (saline) controlled clinical trial of 500-730 kDa
sodium hyaluronate (Hyalgan) on the radiological change in
osteoarthritis of the knee. Int. J. Clin. Pract. 2003, 57
(6), 467–474. G
Gradient Surfaces: Preparation,
Characterization, and Interactions with
Biological Species

1. http://en.wikipedia.org/wiki/Biomaterial,
www.biologyonline.org/dictionary/biomaterials;
http://www.lexiconbiology.com/biology/definition_29.html.

2. Gristina, A.G. Biomaterial-centered infection: microbial


adhesion versus tissue integration. Science 1987, 237,
1588–1595.

3. Vroman, L. Biocompatible Polymers, Metals and


Composites; Szycher, M., Ed.; Technomic: Lancaster, 1983;
81 pp.

4. Gumbiner, B.M. Cell adhesion: the molecular basis of


tissue architecture and morphogenesis. Cell 1996, 84,
345–357.

5. Schakenraad, J.M.; Busscher, H.J.; Wildevuur, C.R.H.;


Arends, J. The influence of substratum surface free energy
on growth and spreading of human fibroblasts in the presence
and absence of serum proteins. J. Biomed. Mater. Res. 1986,
20, 773–784.

6. Tamada, Y.; Ikada, Y. Effect of preadsorbed proteins on


cell adhesion to polymer surfaces. J. Colloid Interf. Sci.
1993, 155, 334–339.

7. van Wachem, P.B.; Beugeling, T.; Feijen, J.; Bantjes,


A.; Detmaers, J.P.; van Aken, W.G. Interaction of cultured
human endothelial cells with polymeric surfaces of
different wettabilities. Biomaterials 1985, 6, 403–408.

8. Lu, D.R.; Park, K. Effect of surface hydrophobicity on


the conformational changes of adsorbed fibrinogen. J.
Colloid Interf. Sci. 1991, 144, 271–281.

9. Johnson, S.D.; Anderson, J.M.; Marchant, R.E.


Biocompatibility studies on plasma polymerized interface
materials encompassing both hydrophobic and hydrophilic
surfaces. J. Biomed. Mater. Res. 1992, 26, 915–935.

10. Joschek, S.; Nies, B.; Krotz, R.G.; Opferich, A.


Chemical and physicochemical characterization of porous
hydroxyapatite ceramics made of natural bone. Biomaterials
2000, 21, 1645–1658.

11. Oh, E.; Luner, P.E. Surface free energy of


ethylcellulose films and the influence of plasticizers. Int.
J. Pharm. 1999, 188, 203–219.

12. Lee, J.H.; Park, J.W.; Lee, H.B. Cell adhesion and
growth on polymer surfaces with hydroxyl groups prepared by
water vapor plasma treatment. Biomaterials. 1991, 12,
443–448.

13. Andrade, J.D.; Smith, L.M.; Gregonis, D.E. The contact


angle and interface energetics. In Surface and Interfacial
Aspects of Biomedical Polymers. Vol. 1. Surface Chemistry
and Physics; Andrade, J.D., Ed.; Plenum Press: New York,
1985; 249–292. 14. Weiss, L. The adhesion of cells. Int.
Rev. Cytol. 1960, 9, 187–225. 15. Kaelble, D.H.; Moacanin,
J. A surface energy analysis of bioadhesion. Polymer 1977,
18, 475–482. 16. Baier, R.E.; Meyer, A.E.; Natiella, J.R.;
Natiella, R.R.; Carter, J.M. Surface properties determine
bioadhesive outcomes: methods and results. J. Biomed.
Mater. Res. 1984, 18 , 337–355. 17. van der Valk, P.; van
Pelt, A.W.J.; Busscher, H.J.; De Jong, H.P.; Wildevuur,
C.R.H.; Arends, J. Interaction of fibroblasts and polymer
surfaces: relationship between surface free energy and
fibroblast spreading. J. Biomed. Mater. Res. 1983, 17,
807–817. 18. Grinnell, F. Cellular adhesiveness and
extracellular substrates. Int. Rev. Cytol. 1978, 53,
65–144. 19. Absolom, D.R.; Hawthorn, L.A.; Chang, G.
Endothelialization of polymer surfaces. J. Biomed. Mater.
Res. 1988, 22, 271–285. 20. Tamada, Y.; Ikada, Y. Cell
attachment to various polymer surface. In Polymer in
Medicine II; Chellini, E., Giusti, P., Migliaresl, C.,
Nicolais, L., Eds.; Plenum Press: New York, 1986; 101–115.
21. Jansen, J.A.; van der Waerden, J.P.C.M.; de Groot, K.
Fibroblast and epithelial cell interactions with
surfacetreated implant materials. Biomaterials 1991, 12,
25–31. 22. Van Kooten, T.G.; Schakenraad, J.M.; van der
Mei, H.C.; Busscher, J.H. Influence of substratum
wettability on the strength of adhesion of human
fibroblasts. Biomaterials 1992, 13, 897–904. 23. Uyen,
H.M.W.; Schakenraad, J.M.; Sjollema, J.; Noordmans, J.;
Jongebloed, W.L.; Stokroos, I.; Busscher, H.J. Amount and
surface structure of albumin adsorbed to solid substrata
with different wettabilities in a parallel plate flow cell.
J. Biomed. Mater. Res. 1990, 24, 1599–1614. 24. Ruardy,
T.G.; Schakenraad, J.M.; van der Mei, H.C.; Busscher, H.J.
Preparation and characterization of chemical gradient
surfaces and their application for the study of cellular
interaction phenomena. Surf. Sci. Rep. 1997, 29, 1–30. 25.
D’Souza, S.F. Immobilization and stabilization of
biomaterials for biosensor applications. Appl. Biochem.
Biotechnol. 2001, 96, 225–238. 26. Upadhyaya, A.; van
Oudenaarden, A. Biomimetic systems for studying actin-based
motility. Curr. Biol. 2003, 13, R734–R744. 27. Fields,
G.B.; Lauer, J.L.; Dori, Y.; Forns, P.; Yu, Y.C.; Tirrell,
M. Protein-like molecular architecture: biomaterial
applications for inducing cellular receptor binding and
signal transduction. Biopolymers 1998, 47, 143–151. 28.
Walker, D.S.; Garrison, M.D.; Richert, W.M. Protein
adsorption to HEMA/EMA copolymers studied by integrated
optical techniques. J. Colloid Interface Sci. 1993, 157,
41–49. 29. Lydon, M.J.; Minett, T.W.; Tighe, B.J. Cellular
interactions with synthetic polymer surfaces in culture.
Biomaterials 1985, 6, 396–402. 30. Elwing, J.; Welin, S.;
Askendahl, A.; Nilsson, U.; Lundstrom, I. A wettability
gradient method for studies Surfaces: Preparation, of
macromolecular interactions at the liquid/solid interface.
J. Colloid Interface Sci. 1987, 119, 203–207. 31. Elwing,
J.; Askendal, A.; Lundstrom, I. Protein exchange reactions
on solid surfaces studied with a wettability gradient
method. Progr. Colloid Polym. Sci. 1987, 74, 103–107. 32.
Elwing, H.; Askendal, A.; Lundstrom, I. Competition between
adsorbed fibrinogen and high-molecularweight kininogen on
solid surfaces incubated in human plasma (the Vroman
effect): Influence of solid surface wettability. J. Biomed.
Mater. Res. 1987, 21, 1023–1028. 33. Elwing, H.; Nilsson,
B.; Svensson, K.E.; Askendahl, A.; Nilsson, U.R.;
Lundstrom, I. Conformational changes of a model protein
(complement factor 3) adsorbed on hydrophilic and
hydrophobic solid surfaces. J. Colloid Interface Sci. 1988,
125, 139–145. 34. Elwing, H.; Askendal, A.; Lundstrom, I.
Desorption of fibrinogen and g-globulin from solid surfaces
induced by a nonionic detergent. J. Colloid Interface Sci.
1989, 128, 296–300. 35. Welin-Klintstrom, S.; Wikstrom, M.;
Askendal, A.; Elwing, H.; Lundstrom, I.; Karlsson, J.O.;
Renvert, S. Proteolytic degradation of fibrinogen layers
adsorbed on hydrophilic and hydrophobic surfaces. Colloids
Surf. 1990, 44, 51–60. 36. Elwing, H.; Golander, C.G.
Protein and detergent interaction phenomena on solid
surfaces with gradients in chemical composition. Adv.
Colloid Interface Sci. 1990, 32, 317–339. 37.
Welin–Klintstrom, S.; Askendal, A.; Elwing, H. Surfactant
and protein interactions on wettability gradient surfaces.
J. Colloid Interface Sci. 1993, 158, 188–194. 38. Hlady,
V.; Golander, C.; Andrade, J.D. Hydrophobicity gradient on
silica surfaces: a study using total internal reflection
fluorescence spectroscopy. Colloids Surf. 1988, 33, 185–190.
39. Golander, C.G.; Lin, Y.S.; Hlady, V.; Andrade, J.D.
Wetting and plasma-protein adsorption studies using
surfaces with a hydrophobicity gradient. Colloids Surf.
1990, 49, 289–302. 40. Hlady, V.; Lin, J.N.; Andrade, J.D.
Spatially resolved detection of antibody-antigen reaction
on solid/liquid interface using total internal reflection
excited antigen fluorescence and charge-coupled device
detection. Biosens. Bioelect. 1990, 5, 291–301. 41. Hlady,
V. Spatially resolved adsorption kinetics of immunoglobulin
G onto the wettability gradient surface. Appl. Spectrosc.
1991, 45, 246–252. 42. Carley, J.F.; Kitze, P.T.
Corona-discharge treatment of polymeric films. II. Chemical
studies. Polym. Eng. Sci. 1980, 20, 330–338. 43. Lopez,
G.P.; Biebuyck, H.A.; Frisbie, C.D.; Whitesides, G.M.
Imaging of features on surfaces by condensation figures.
Science 1993, 260, 647–649. 44. Zhao, H.; Beysens, D. From
droplet growth to film growth on a heterogeneous surface:
condensation associated with a wettability gradient.
Langmuir 1995, 11, 627–634. 45. Lin, Y.S.; Hlady, V.;
Golander, C.G. The surface density gradient of grafted
poly(ethylene glycol): preparation, characterization and
protein adsorption. Colloids Surf. B Biointerfaces 1994, 3,
49–62. 46. Liedberg, B.; Tengvall, P. Molecular gradients
of omega-substituted alkanethiols on gold: preparation and
characterization. Langmuir 1995, 11, 3821–3827. 47. Riepl,
M.; Ostblom, M.; Lundstrom, I.S.; Svensson, C.T.; Denier
van der Gon, A.W.; Schaferling, M.; Liedberg, B. Molecular
gradients: an efficient approach for optimizing the surface
properties of biomaterials and biochips. Langumir 2005, 21,
1042–1050. 48. Pitt, W.G. Fabrication of a continuous
wettability gradient by radio frequency plasma discharge.
J. Colloid Interface Sci. 1989, 133, 223–227. 49. Golander,
C.G.; Pitt, W.G. Characterization of hydrophobicity
gradients prepared by means of radio frequency plasma
discharge. Biomaterials 1990, 11, 32– 35. 50. Lee, J.H.;
Park, J.W.; Lee, H.B. Characterization of hydroxyl group
gradient surfaces prepared by water vapor plasma treatment.
Polymer (Korea) 1990, 14, 646–652. 51. Lee, H.B.; Andrade,
J.D. Cell adhesion on gradient surfaces. Trans. 3rd World
Biomaterials Congr. 1988, 7, 43. 52. Lee, H.B. Application
of synthetic polymers in implants. In Frontiers of
Macromolecular Science; Saegusa, T., Higashimura, T., Abe,
A., Eds.; Blackwell Scientific Publications: Oxford, 1989;
579–584. 53. Bamford, C.H.; Ward, J.C. The effect of the
highfrequency discharge on the surfaces of solids. I. The
production of surface radicals on polymers. Polymer 1961,
2, 277–293. 54. Morosoff, N.; Crist, B.; Bumgarner, M.;
Hsu, T.; Yasuda, H. Free radicals resulting from plasma
polymerization and plasma treatment. J. Macromol. Sci.
Chem. 1976, A10, 451–471. 55. Mitchell, J., Jr.; Perkins,
L.R. Determination of hydroperoxide groups in oxidized
polyethylene. Appl. Polym. Symp. 1967, 4, 167–173. 56.
Clark, D.T.; Dilks, A. ESCA applied to polymers. XXII. RF
glow discharge modification of polymers in pure oxygen and
helium-oxygen mixtures. J. Polym. Sci., Polym. Chem. Ed.
1979, 17, 957–976. 57. Steinhauser, H.; Ellinghorst, G.
Corona treatment of isotactic polypropylene in nitrogen and
carbon dioxide. Angew. Makromol. Chem. 1984, 120, 177–191.
58. Iwata, H.; Kishida, A.; Suzuki, M.; Hata, Y.; Ikada, Y.
Oxidation of polyethylene surface by corona discharge and
the subsequent graft polymerization. J. Polym. Sci. Polym.
Chem. Ed. 1988, 26, 3309–3322. 59. Chan, C.M.; Ki, T.M.;
Hiraoka, H. Polymer surface modification by plasmas and
photons. Surf. Sci. Rep. 1996, 24, 1–54. 60. Lee, J.H.;
Kim, H.G.; Khang, G.; Lee, H.B.; Jhon, M.S.
Characterization of wettability gradient surfaces prepared
by corona discharge treatment. J. Colloid Interface Sci.
1992, 151, 563–570. 61. Lee, J.H.; Lee, H.B. A wettability
gradient as a tool to study protein adsorption and cell
adhesion on polymer surfaces. J. Biomater. Sci., Polymer
Ed. 1993, 4, 467–481. G with Biological

62. Kim, H.G.; Lee, J.H.; Lee, H.B.; Jhon, M.S.


Dissociation behavior of surface-grafted poly(acrylic
acid): effects of surface density and counterion size. J.
Colloid Interface Sci. 1993, 157, 82–87.

63. Lee, J.H.; Kim, H.W.; Pak, P.K.; Lee, H.B. Preparation
and characterization of functional group gradient surfaces.
J. Polym. Sci. A Polym. Chem. 1994, 32, 1569–1579.

64. Jeong, B.J.; Lee, J.H.; Lee, H.B. Preparation and


characterization of comb-like PEO gradient surfaces. J.
Colloid Interface Sci. 1996, 178, 757–763.

65. Lee, J.H.; Lee, J.W.; Khang, G.; Lee, H.B. Interaction
of cells on chargeable functional group gradient surfaces.
Biomaterials 1997, 18, 351–358.

66. Lee, J.H.; Jung, H.W.; Kang, I.J.; Lee, H.B. Cell
behavior on polymers surfaces with different functional
groups. Biomaterials 1994, 15, 705–711.

67. Lee, J.H.; Khang, G.; Lee, J.W.; Lee, H.B.; Platelet
adhesion onto chargeable functional group gradient
surfaces. J. Biomed. Mater. Res. 1998, 40, 180–186.

68. Kim, M.S.; Seo, K.S.; Khang, G.; Lee, H.B. First
preparation of biotinylated gradient polyethylene surface
to bind photoactive caged streptavidin. Langmuir 2005, 21,
4066–4070.

69. Kim, M.S.; Seo, K.S.; Khang, G.; Lee, H.B. Preparation
of a gradient biotinylated polyethylene surface to bind
streptavidin-FITC. Bioconjug. Chem. 2005, 16, 245–249.
70. Burnham, N.L. Polymers for delivering peptides and
proteins. Am. J. Hosp. Pharm. 1994, 51, 210–218.

71. Greenwald, R.B.; Choe, Y.H.; McGuire, J.; Conover, C.D.


Effective drug delivery by PEGylated drug conjugates. Adv.
Drug Deliv. Rev. 2003, 55, 217–250.

72. Lee, J.H.; Jeong, B.J.; Lee, H.B. Plasma protein


adsorption and platelet adhesion onto comb-like PEO
gradient surfaces. J. Biomed. Mater. Res. 1997, 34,
105–114.

73. Iwasaki, Y.; Ishihara, K.; Nakabayashi, N.; Khang, G.;


Jeon, J.H.; Lee, J.W.; Lee, H.B. Platelet adhesion on the
gradient surfaces grafted with phospholipid polymer. J.
Biomater. Sci. Polym. Ed. 1998, 9, 801–816.

74. Iwasaki, Y.; Sawada, S.; Nakabayashi, N.; Khang, G.;


Lee, H.B.; Ishihara, K. The effect of the chemical
structure of the phospholipid polymer on fibronectin
adsorption and fibroblast adhesion on the gradient
phospholipid surface. Biomaterials 1999, 20, 2185–2191.

75. Bain, C.D.; Whitesides, G.M. Depth sensitivity of


wetting: monolayers of omega-mercapto ethers on gold. J.
Am. Chem. Soc. 1988, 110, 5897–5898.

76. Troughton, E.B.; Bain, C.D.; Whitesides, G.M.; Nuzzo,


R.G.; Allara, D.L.; Porter, M.D. Monolayer films prepared by
the spontaneous self-assembly of symmetrical and
unsymmetrical dialkyl sulfides from solution onto gold
substrates: structure, properties, and reactivity of
constituent functional groups. Langmuir 1988, 4, 365–385.
77. Johnson, R.E.; Dettre, R.H. Surface and Colloid
Science; Matijevic, E., Ed.; Wiley Interscience: New York,
1969; Vol. 2, 85 pp. 78. Adamson, A.W. Physical Chemistry
of Surfaces, 3rd Ed.; Wiley: New York, 1976. 79. Lee, J.H.;
Khang, G.; Lee, J.W.; Lee, H.B. Interaction of different
types of cells on polymer surfaces with wettability
gradient. J. Colloid Interface Sci. 1998, 205, 323–330. 80.
Ruardy, T.G.; Moorlag, H.E.; Schakenraad, J.M.; van der
Mei, H.C.; Busscher, H.H. Growth of fibroblasts and
endothelial cells on wettability gradient surfaces. J.
Colloid Interface Sci. 1997, 188, 209–217. 81.
Ueda-Yukoshi, T.; Matsuda, T. Cellular responses on a
wettability gradient surface with continuous variations in
surface compositions of carbonate and hydroxyl groups.
Langmuir 1995, 11, 4135–4140. 82. Azzam, R.M.A.; Bashara,
N.M. Ellipsometry and Polarized Light; Elsevier: Amsterdam,
1987; 269 pp. 83. Andrade, J.D. X-ray photoelectron
spectroscopy (XPS). In Surface and Interfacial Aspects of
Biomedical Polymers Vol. 1. Surface Chemistry and Physics;
Andrade, J.D., Ed.; Plenum Press: New York, 1985; 105–195.
84. Lin, J.N.; Drake, B.; Lea, A.S.; Hansma, P.K.; Andrade,
J.D. Direct observation of immunoglobulin adsorption
dynamics using the atomic force microscope. Langmuir 1990,
6, 509–511. 85. Hlady, V.; van Waggenen, R.A.; Andrade,
J.D. Total internal reflection intrinsic fluorescence (TIRF)
spectroscopy applied to protein adsorption. In Surface and
Interfacial Aspects of Biomedical Polymers. Vol. 2. Protein
Adsorption; Andrade, J.D., Ed.; Plenum Press: New York,
1985; 81–119. 86. Roberson, S.V.; Fahey, A.J.; Sehgal, A.;
Karim, A. Multifunctional TOF-SIMS: combinatorial mapping
of gradient energy substrates. Appl. Surf. Sci. 2002, 200,
150–164. 87. Lee, J.H. Interactions of PEO-containing
polymeric surfactants with hydrophobic surfaces. Ph. D.
thesis, University of Utah, Salt Lake City, 1988. 88.
Green, N. Avidin. 1. The use of (14–C) biotin for kinetic
studies and for assay. Biochem. J. 1963, 89, 585–591. 89.
Carter, S.B. Principles of cell motility: the direction of
cell movement and cancer invasion. Nature 1965, 208,
1183–1187. 90. Lee, J.H.; Lee, S.J.; Khang, G.; Lee, H.B.
The effect of fluid shear stress on endothelial cell
adhesiveness to polymer surfaces with wettability gradient.
J. Colloid Interface Sci. 2000, 230, 84–90. Surfaces:
Preparation,
Hard Tissue: Biomaterial Interactions

10. Montanaro, L.; Arciola, C.R.; Campoccia, D.;


Cervellati, M. In vitro effects of MG63 osteoblast-like
cells following contact with two roughness-differing
fluorohydroxyapatite-coated titanium alloys. Biomaterials
2002, 23, 3651–3659.

11. Nimb, L.; Gotfredsen, K.; Steen-Jensen, J. Mechanical


failure of hydroxyapatite-coated titanium and
cobaltchromium-molybdenum alloy implants. Acta. Orthop.
Belg. 1993, 59, 333–338.

12. Rahbek, O.; Overgaard, S.; Lind, M.; Bendix, K.;


Bunger, C.; Soballe, K. Sealing effect of hydroxyapatite
coating on peri-implant migration of particles. J. Bone.
Joint Surg. 2001, 83, 441–447.

13. Coathup, M.J.; Blackburn, J.; Goodship, A.E.;


Cunningham, J.L.; Smith, T.; Blunn, G.W. Role of
hydroxyapatite coating in resisting wear particle migration
and osteolysis around acetabular components. Biomaterials
2005, 26, 4161–4169.

14. Bauer, T.W.; Taylor, S.K.; Jiang, M.; Medendorp, S.V.


An indirect comparison of third-body wear in retrieved
hydroxyapatite-coated, porous and cemented femoral
components. Clin. Orthop. Rel. Res. 1994, 298, 11–18.

15. Sul, Y.T.; Johansson, C.B.; Albrektsson, T. Oxidized


titanium screws coated with calcium ions and their
performance in rabbit bone. Int. J. Oral Maxillofac.
Implants 2002, 17, 625–634.

16. Nayab, S.N.; Jones, F.H.; Olsen, I. Effects of calcium


ion implantation on human bone cell interaction with
titanium. Biomaterials 2005, 26, 4717–4727.

17. Kornu, R.; Maloney, W.J.; Kelly, M.A.; Smith, R.L.


Osteoblast adhesion to orthopaedic implant alloys: effects
of cell adhesion molecules and diamond-like carbon coating.
J. Orthop. Res. 1996, 14, 871–877.

18. Roehlecke, C.; Witt, M.; Kasper, M.; Schulze, E.; Wolf,
C.; Hofer, A.; Funk, R.W. Synergistic effect of titanium
alloy and collagen type I on cell adhesion, proliferation
and differentiation of osteoblast-like cells. Cells Tissues
Organs 2001, 168, 178–187.

19. Goto, T.; Yoshinari, M.; Kobayashi, S.; Tanaka, T. The


initial attachment and subsequent behaviour of osteoblasts
and oral epithelial cells on titanium. Biomed. Mater. Eng.
2004, 14, 537–544.

20. Gornowicz, G.; McCarthy, M.B. Response of human


osteoblasts to implant materials: integrin-mediated
adhesion. J. Orthop. Res. 1996, 14, 878–887.

21. Zinger, O.; Anselme, K.; Denzer, A.; Habersetzer, P.;


Wieland, M.; Jeanfils, J.; Hardouin, P.; Landolt, D.
Time-dependent morphology and adhesion of osteoblastic
cells on titanium model surfaces featuring scaleresolved
topography. Biomaterials 2004, 25, 2695–2711.

22. Urban, R.M.; Jacobs, J.J.; Tomlinson, M.J.; Gavrilovic,


J.; Black, J.; Peoch, M. Dissemination of wear particles to
the liver, spleen and the abdominal lymph nodes of patients
with hip or knee replacement. J. Bone Joint Surg. 2000,
82-A, 457–477.

23. Mak, K.H.; Wong, T.K.; Poddar, N.C. Wear debris from
total hip arthroplasty presenting as an intrapelvic mass.
J. Arthroplasty 2001, 16, 674–676. 24. Doherty, A.T.;
Howell, R.T.; Ellis, L.A.; Bisbinas, I.; Learmonth, I.D.;
Newson, R.; Case, C.P. Increased chromosome translocations
and aneuploidy in peripheral blood lymphocytes of patients
having revision arthroplasty of the hip. J. Bone Joint
Surg. 2001, 83-B, 1075–1081. 25. Keel, S.B.; Jaffe, K.A.;
Petur Nielsen, G.; Rosenberg, A.E. Orthopaedic
implant-related sarcoma: a study of twelve cases. Modern
Pathol. 2001, 14, 969–977. 26. Ellingsen, J.E.;
Lyngstadaas, S.P. Bio-Implant Interface; CRC Press: Boca
Raton, U.S.A., 2003. 27. Korkusuz, P.; Korkusuz, F. Hard
tissue-biomaterial interactions. In Biomaterials in
Orthopedics; Yaszemski, M.J., Trantolo, D.J., Lewandrowski,
K.U., Hasy´rcy´, V., Altobelli, D.E., Wise, D.L., Eds.;
Marcel Dekker Inc: New York, U.S.A., 2004; 1–210. 28.
Pipino, F. The bone-prosthesis interaction. J. Orthopaed.
Traumatol. 2000, 1, 3–9. 29. Epstein, N.J.; Warme, B.A.;
Spanogle, J.; Ma, T.; Bragg, B.; Smith, R.L.; Goodman, S.B.
Interleukin1modulates periprosthetic tissue formation in an
intramedullary model of particle-induced inflammation. J.
Orthop. Res. 2005, 23, 501–510. 30. Nakashima, Y.; Sun,
D.H.; Trindade, M.C.D.; Chun, L.E.; Song, Y.; Goodman,
S.B.; Schurman, D.J.; Maloney, W.J.; Smith, R.L. Induction
of macrophage C-C chemokine expression by titanium alloy
and bone cement particles. J. Bone Join. Surg. 1999, 81-B,
155–162. 31. Sabokbar, A.; Itonaga, I.; Sun, S.G.; Kudo,
O.; Athanasou, N.A. Arthroplasty membrane-derived
fibroblasts directly induce osteoclast formation and
osteolysis in aseptic loosening. J. Orthop. Res. 2005, 23,
511–519. 32. Granchi, D.; Ciapetti, G.; Stea, S.; Savarino,
L.; Filippini, F.; Sudanse, A.; Zinghi, G.; Montanaro, L.
Cytokine release in mononuclear cells of patients with
Co-Cr hip prosthesis. Biomaterials 1999, 20, 1079–1086. 33.
Stea, S.; Visentin, M.; Granchi, D.; Cenni, E.; Ciapetti,
G.; Sudanese, A.; Toni, A. Apoptosis in peri-implant
tissue. Biomaterials 2000, 21, 1393–1398. 34. Fleming,
J.E.; Cornell, C.N.; Muschler, G.F. Bone cells and matrices
in orthopedic tissue engineering. Orthop. Clin. N. Am.
2000, 31, 357–374. 35. Korkusuz, F.; Uchida, A.; Shinto,
Y.; Araki, N.; Inoue, K.; Ono, K. Experimental
implant-related osteomyelitis treated by antibiotic-calcium
hydroxyapatite ceramic composites. J. Bone Joint Surg.
1993, 75-B, 111–114. 36. Uchida, A.; Araki, N.; Shinto, Y.;
Yoshikawa, H.; Ono, K.; Kurosaki, E. The use of
hydroxyapatite ceramic in bone tumour surgery. J. Bone
Joint Surg. 1990, 72-b, 298–302. 37. Koc, N.; Timucin, M.;
Korkusuz, F. Fabrication and characterization of porous
tricalcium phosphate ceramics. Ceramics Int. 2004, 30,
205–211. 38. Roy, M.E.; Nishimoto, S.K. Matrix Gla protein
binding to hydroxyapatite is dependent on the ionic
environment: calcium enhances binding affinity but phosphate
and magnesium decrease affinity. Bone 2002, 31, 296–302. 39.
Rees, S.G.; Hughes-Wassell, D.T.; Waddington, R.J.; Embery,
G. Interaction of bone proteoglycans and proteoglycan
components with hydroxyapatite. Biochim. Biophys. Acta
2001, 1568, 118–128. 40. Goldberg, H.A.; Warner, K.J.; Li,
M.C.; Hunter, G.K. Binding of bone sialoprotein,
osteopontin and synthetic polypeptides to hydroxyapatite.
Connect. Tissue Res. 2001, 42, 25–37. 41. Sendil-Keskin,
D.; Tezcaner, A.; Korkusuz, P.; Korkusuz, F.; Hasirci, V.
Collagen-chondroitin sulfate-based PLLA-SAIB-coated rhBMP-2
delivery system for bone repair. Biomaterials 2005, 26,
4023–4034. 42. Ko¨se, G.T.; Korkusuz, F.; Korkusuz, P.;
Hasy´rcy´, V. In vivo tissue engineering of bone using
Poly(3-hydroxybutyric acid-co-3-hydroxyvaleric acid) and
collagen scaffolds. Tissue Eng. 2004, 10, 1234–1250. 43.
Torun-Ko¨se, G.; Korkusuz, F.; O¨zkul, A.; Soysal, Y.;
O¨zdemir, T.; Yildy´z, C.; Hasirci, V. Tissue engineered
cartilage on collagen and PHBV matrices. Biomaterials 2005,
26, 5187–5197. 44. Korkusuz, F.; Korkusuz, P.; Eksiog˘lu,
F.; Gu¨rsel, I.; Hasirci, V. In vivo response to
biodegradable controlled antibiotic release systems. J.
Biomed. Mater. Res. 2001, 55, 217–228. 45. Coskun, S.;
Korkusuz, F.; Hasirci, V. Hydroxyapatite reinforced
poly(3-hydroxybutyrate) and
poly(3-hydroxybutyrate-co-3-hydroxyvalerate) based
biodegradable composite bone plate. J. Biomater. Sci.
Polym. Ed. 2005, 16, 1485–1502. 46. Idris, A.I.; van’t Hof,
R.J.; Greig, I.R.; Ridge, S.A.; Baker, D.; Ross, R.A.;
Ralston, S.H. Regulation of bone mass, bone loss and
osteoclast activity by cannabinoid receptors. Nat. Med.
2005, 11, 774–779. 47. Lynn, A.K.; Brooks, R.A.; Bonfield,
W.; Rushton, N. Repair of defects in articular joints.
Prospects for material-based solutions in tissue
engineering. J. Bone Joint Surg. 2004, 86-B, 1093–1099. 48.
Gao, J.; Dennis, J.E.; Solchaga, L.A.; Awadallah, A.S.;
Goldberg, V.M.; Caplan, A.I. Tissue-engineered fabrication
of an osteochondral composite graft using rat bone
marrow-derived mesenchymal stem cells. Tissue Eng. 2001, 7,
363–371. 49. Agung, M.; Ochi, M.; Adachi, N.; Uchio, Y.;
Takao, M.; Kawasaki, K. Osteochondritis dissecans of the
talus treated by the transplantation of tissue-engineered
cartilage. Arthroscopy 2004, 10, 1075–1080. 50. Alhadlaq,
A.; Mao, J.J. Tissue-engineered osteochondral constructs in
the shape of an articular condyle. J. Bone Joint Surg.
2005, 87-A, 936–944. H
Hard Tissue Elastic Properties

1. Glimcher, M.J. Composition, structure, and organization


of bone and other mineralized tissues and the mechanism of
calcification. Handbook of Physiology– Endocrinology VII;
Williams and Wilkins Co.: Baltimore, MD, 1976; 25–116.

2. Voet, D.; Voet, J.G. Biochemistry, 2nd Ed.; John Wiley &
Sons, Inc.: New York, 1995.

3. Le Geros, R.Z. Calcium Phosphates in Oral Biology and


Medicine; Karger: Basil, Switzerland, 1991.

4. Johnson, M.; Katz, J.L. Electromechanical effects in


bone. In Handbook of Bioengineering; Skalak, R., Chen, S.,
Eds.; McGraw-Hill Book Co.: New York, 1987; 3.1–3.11.

5. Currey, J.D. Bones: Structure and Mechanics; Princeton


University Press: Princeton, NJ, 2002.

6. Katz, J.L. Hierarchical modeling of compact haversian


bone as a fiber reinforced material. In 1976 Advances in
Bioengineering; Mates, R.E., Smith, C.R., Cowin, S.C.,
Eds.; A.S.M.E.: New York, 1976; 17–18. 7. Lakes, R.S.
Viscoelastic Solids; CRC Press: Boca Raton, FL, 1999. 8.
Wainwright, S.A.; Briggs, W.D.; Currey, J.D.; et al.
Mechanical Design in Organisms; Princeton University Press:
Princeton, NJ, 1982. 9. Eppell, S.J.; Tong, W.; Katz, J.L.;
et al. Shape and size of isolated bone mineralites using
atomic force microscopy. J. Orthop. Res. 2001, 19,
1027–1034. 10. Tong, W.; Glimcher, M.J.; Katz, J.L.; et al.
Size and shape of mineralites in young bovine bone measured
by atomic force microscopy. Calc. Tissue Int. 2003, 72 (5),
592–598. 11. Frasca, P.; Harper, R.A.; Katz, J.L. A new
method of bone preparation for collagen fiber orientation
studies by means of scanning electron microscopy. J. Dent.
Res. 1976, 55, 372–375. 12. Herring, G.M. Methods for the
study of the glycoproteins and proteoglycans of bone using
bacterial collagenase. Determination of bone sialoprotein
and chondroitin sulphate. Calcif. Tissue Res. 1977, 24,
29–36. 13. Pellegrino, E.D.; Blitz, R.M. The composition of
human bone in uremia. Medicine 1965, 44, 397–418. 14.
Vejlens, L. Glycosaminoglycans of human bone tissue: I.
Pattern of compact bone in relation to age. Calcif. Tissue
Res. 1971, 7, 175–190. 15. Knets, I.V. Mechanics of
biological tissues, a review. Polymer Mech. (Translation of
Mekhanika Polimerov) 1978, 13, 434–440. 16. Van Buskirk,
W.C.; Ashman, R.B. The elastic moduli of bone. InMechanical
Properties of Bone; Cowin, S.C., Ed.; A.S.M.E.: New York
City, 1981; AMD Vol. 41, 131–143. 17. Katz, J.L.; Lipson,
S.; Yoon, H.S.; et al. The effects of remodeling on the
elastic properties of bone. Calc. Tissue Int. 1984, 36,
S31–S36. 18. Lang, S.B. Elastic coefficients of animal bone.
Science 1969 , 165, 287–288. 19. Katz, J.L.; Ukraincik, K.
On the anisotropic elastic properties of hydroxyapatite. J.
Biomech. 1971, 4, 221–227. 20. Yoon, H.S.; Katz, J.L.
Ultrasonic wave propagation in human cortical bone. I.
Theoretical considerations for hexagonal symmetry. J.
Biomech. 1976, 9, 407–412. 21. Yoon, H.S.; Katz, J.L.
Ultrasonic wave propagation in human cortical bone. II.
Measurements of elastic properties and microhardness. J.
Biomech. 1976, 9, 459–464. 22. Lekhnitskii, S.G. Theory of
Elasticity of an Anisotropic Body; Mir Publishers: Moscow,
1981. 23. Keaveny, T.M.; Hayes, W.C. A 20-year perspective
on the mechanical properties of trabecular bone. J.
Biomech. Eng. 1993, 115, 535–542. 24. Townsend, P.R.; Rose,
R.M.; Radin, E.L. Buckling studies of single human
trabeculae. J. Biomech. 1975, 8, 199–201. 25. Ashman, R.B.;
Rho, J.Y. Elastic modulus of trabecular bone material. J.
Biomech. 1988, 21, 177–181. 26. Rho, J.Y.; Ashman, R.B.;
Turner, C.H. Young’s modulus of trabecular and cortical
bone material; ultrasonic and microtensile measurements. J.
Biomech. 1993, 26, 111–119. 27. Ryan, S.D.; Williams, J.L.
Tensile testing of rodlike trabeculae excised from bovine
femoral bone. J. Biomech. 1989, 22, 351–355. 28. Choi, K.;
Goldstein, S.A. A comparison of the fatigue behavior of
human trabecular and cortical bone tissue. J. Biomech.
1992, 25, 1371–1381. 29. Rho, J.Y.; Roy, M.E.; Tsui, T.Y.;
et al. Elastic properties of microstructural components of
human bone tissue as measured by indentation. J. Biomed.
Mat. Res. 1999, 45, 48–54. 30. Turner, C.H.; Rho, J.Y.;
Takano, Y.; et al. The elastic properties of trabecular and
cortical bone tissues are similar results from two
microscopic measurement techniques. J. Biomech. 1999, 32,
437–441. 31. Bumrerraj, S.; Katz, J.L. Scanning acoustic
microscopy study of human cortical and trabecular bone.
Ann. Biomed. Eng. 2001, 29, 1–9. 32. Chung, D.H.; Buessem,
W.R. The elastic anisotropy of crystals. In Anisotropy in
Single Crystal Refractory Compounds; Vahldiek, F.W.,
Mersol, S.A., Eds.; Plenum Press: New York, 1968; Vol. 2,
217–245. 33. Katz, J.L.; Meunier, A. The elastic anisotropy
of bone. J. Biomech. 1987, 20, 1063–1070. 34. Katz, J.L.;
Meunier, A. A generalized method for characterizing elastic
anisotropy in solid living tissues. J. Mat. Sci. Mat. Med.
1990, 1, 1–8. 35. Voigt, W. Lehrbuch der Kristallphysik;
Teubner: Berlin, Germany, 1910. 36. Reuss, A. Berechnung
der Fliessgrenze von mischkristallen auf grund der
plastizitats-bedingung fur einkristalle, z. Zeitschrift fur
Angewandte Mathematik und Mechanik 1929, 9, 49–58. 37.
Currey, J.D. Three analogies to explain the mechanical
properties of bone. Biorheol. 1964, 2, 1–10. 38. Currey,
J.D. The relationship between the stiffness and the mineral
content of bone. J. Biomech. 1969, 2, 477–480. 39. Cox,
H.L. The elasticity and strength of paper and other fibrous
materials. Br. Appl. Phys. 1952, 3, 72–79. 40. Katz, J.L.
Hard tissue as a composite material: part I. Bounds on the
elastic behavior. J. Biomech. 1971, 4, 455–473. 41.
Piekarski, K. Analysis of bone as a composite material.
Int. J. Eng. Sci. 1973 , 1, 557–565. 42. Hashin, Z.;
Shtrikman, S.J. A variational approach to the theory of
elastic behavior of multiphase materials. J. Mech. Phys.
Sol. 1963, 11, 127–140. 43. Lakes, R.S. Materials with
structural hierarchy. Nature 1993, 361, 511–515. 44. Katz,
J.L. Elastic properties of calcified tissues. Isr. J. Med.
Sci. 1971, 7, 439–441. 45. Hashin, Z.; Rosen, B.W. The
elastic moduli of fiber reinforced materials. J. Appl. Mech.
1964, 31, 223–232. 46. Katz, J.L. The structure and
biomechanics of bone. In Mechanical Properties of
Biological Materials; Currey, J.D., Vincent, J.F.V., Eds.;
Cambridge University Press: Cambridge, U.K., 1980; 137–168.
47. Bonfield, W.; Grynpas, M.D. Anisotropy of Young’s
modulus of bone. Nature 1977, 270, 453–454. 48. Katz, J.L.
On the anisotropy of Young’s modulus of bone. Nature 1980,
283, 106–107. 49. Katz, J.L. Composite material models for
bone. In Mechanical Properties of Bone; A.S.M.E.: New York,
1981; AMD-Vol. 45, 171–184. 50. Katz, J.L.; Maharidge,
R.L.; Yoon, H.S. The estimation of inter-osteonal
mechanical properties from a composite model for haversian
bone. In Biomechanics Current Interdisciplinary Research;
Perren, S.M., Scheider, E., Eds.; Martinus Injhoff:
Dordrecht, 1985; 179–184. 51. Katz, J.L.; Yoon, H.S.;
Maharidge, R.L. Calculation of inter-osteonal mechanical
properties for haversian bone based on a hierarchical
composite model. Biomechanics Symposium; A.S.M.E: New York,
1985; AMD-Vol. 68, FED-Vol. 21, 33–35. 52. Hogan, H.A.
Micromechanics modeling of haversian cortical bone
properties. J. Biomech. 1992, 25 (5), 549–556. 53. Aoubiza,
B.; Crolet, J.M.; Meunier, A. On the mechanical
characterization of compact bone structure using the
homogenization theory. J. Biomech. 1996, 29, 1539–1547. 54.
Swedlow, D.B.; Frasca, P.; Harper, R.A.; et al. Scanning
and transmission electron microscopy of calcified tissues.
Biomat. Med. Dev. Art. Org. 1975, 3, 121–153. 55. Frasca,
P.; Harper, R.A.; Katz, J.L. Isolation of single osteons
and osteon lamellae. Acta Anat. 1976, 95, 122–129. 56.
Frasca, P.; Harper, R.A.; Katz, J.L. Collagen fiber
orientation in human secondary osteons. Acta Anat. 1977,
98, 1–13. 57. Lees, S.; Davidson, C.L. The role of collagen
in the elastic properties of calcified tissues. J. Biomech.
1977, 10, 473–486. 58. Frasca, P.; Harper, R.A.; Katz, J.L.
Mineral and collagen fiber orientation studies in human
haversians. J. Dent. Res. 1978, 57, 526–533. 59. Frasca,
P.; Harper, R.A.; Katz, J.L. A new technique for studying
collagen fibers and grounds substances in bone with scanning
electron microscopy. Microsc. Acta 1978, 80, 211–214. 60.
Weiner, S.; Traub, W. Organization of hydroxyapatite
crystals within collagen fibrils. FEBS letters 1986, 206,
262–266. 61. Weiner, S.; Traub, W. Bone structure: from
angstroms to microns. FASEB 1992, 6, 879–885. 62. Wagner,
H.D.; Weiner, S. On the relationship between the
microstructure of bone and its mechanical stiffness. J.
Biomech. 1992, 25, 1311–1320. 63. Hasegawa, K.; Turner,
C.H.; Burr, D.B. Contribution of collagen and mineral to
the elastic anisotropy of bone. Calc. Tissue Int. 1994, 55,
381–386. 64. Landis, W.J.; Hodgens, K.J.; Arena, J.; et al.
Structural relations between collagen and mineral in bone
as determined by high voltage electron microscopic
tomography. Microsc. Res. Tech. 1996, 33, 192–202. 65. Rho,
J.Y.; Kuhn-Spearing, L.; Zioupos, P. Mechanical properties
and the hierarchical structure of bone. Med. Eng. Phys.
1998, 20, 92–102. 66. Rho, J.Y.; Currey, J.D.; Zioupos, P.;
et al. Microstructural elasticity and regional
heterogeneity in H human femoral bone of various ages
examined by nanoindentation. J. Biomech. 2002, 35, 189–198.

67. Lakes, R.S.; Katz, J.L. Interrelationships among the


viscoelastic functions for anisotropic solids: application
to calcified tissues and related systems. J. Biomech. 1974,
7, 259–270.

68. Sedlin, E.D. A rheological model for cortical bone.


Study of the physical properties of human femoral samples.
Acta. Orthop. Scan. 1965, 83, 1–77.

69. Smith, R.; Keiper, D. Dynamic measurement of


viscoelastic properties of bone. Am. J. Med. Elec. 1965, 4,
156–160.

70. Laird, G.W.; Kingsbury, H.B. Complex viscoelastic


moduli of bovine bone. J. Biomech. 1973, 6, 59–67.

71. Lugassy, A.A.; Korostoff, E. Viscoelastic behavior of


bovine femoral cortical bone and sperm whale dentin. In
Research in Dental and Medical Materials; Korostoff, E.,
Ed.; Plenum Press: New York, 1969; 1–17.

72. Black, J.; Korostoff, E. Dynamic mechanical properties


of viable human cortical bone. J. Biomech. 1973, 6,
435–438.
73. Knothe-Tate, M.L.; Knothe, U.; Niederer, P.
Experimental elucidation of mechanical load-induced fluid
flow and its potential role in bone metabolism and
functional adaptation. Am. J. Med. Sci. 1998, 316, 189–195.

74. Lakes, R.S.; Katz, J.L.; Sternstein, S.S. Viscoelastic


properties of wet cortical bone. I. Torsional and biaxial
studies. J. Biomech. 1979, 12, 657–678. 75. Lakes, R.S.;
Katz, J.L. Viscoelastic properties of wet cortical bone.
II. Relaxation mechanisms. J. Biomech. 1979, 12, 679–687.
76. Lakes, R.S.; Katz, J.L. Viscoelastic properties of wet
cortical bone. III. A nonlinear constitutive equation. J.
Biomech. 1979, 12, 689–698. 77. Lakes, R.S.; Katz, J.L.
Viscoelastic properties of bone. In Natural and Living
Materials III: Structure-Property Relationships in
Biomaterials; Hastings, G.W., Ducheyne, P., Eds.; CRC
Press: Boca Raton, FL, 1984; 61–87. 78. Gottesman, T.;
Hashin, Z. Analysis of viscoelastic behavior of bones on
the basis of microstructure. J. Biomech. 1979, 13, 89–96.
79. Katz, J.L.; Kinney, J.H.; Spencer, P.; et al. Elastic
anisotropy of bone and dentitional tissues. J. Mat. Sci.
Mat. Med. 2005, 16 (9). 80. Kinney, J.H.; Gladden, J.R.;
Marshall, G.W.; et al. Resonant ultrasound spectroscopy
measurements of the elastic properties of human dentin. J.
Biomech. 2004, 37 (4), 437–441. 81. Kinney, J.H.; Pople,
J.A.; Marshall, G.W.; et al. Collagen orientation and
crystallite size in human dentin: a small angle x-ray
scattering study. Calcif. Tissue. Int. 2001, 69, 31–37. 82.
Pollack, S.R. Streaming potentials in bone. In Bone
Mechanics Handbook, 2nd Ed.; Cowin, S.C., Ed.; CRC Press:
Boca Raton, FL, 2001; 24-1–24-22. 83. Singh, S.; Katz, J.L.
Electromechanical properties of bone: a review. J.
Bioelect. 1988, 7 (2), 219–238.
Healing of Bone and Connective Tissues

2. Caplan, A.I.; Bruder, S.P. Mesenchymal stem cells:


building blocks for molecular medicine in the 21st century.
Trends Mol. Med. 2001, 7, 259–264.

3. Clark, R.A.F. The Molecular and Cellular Biology of


Wound Repair, 2nd Ed., New York: Plenum, 1996; p. 611.

4. Cullinane, D.M.; Salisbury, K.T.; Alkhiary, Y.;


Eisenberg, S.; Gerstenfeld, L.; Einhorn, T.A. Effects of
the local mechanical environment on vertebrate tissue
differentiation during repair: does repair recapitulate
development? J Exp. Biol. 2003, 206, 2459–2471.

5. Daniel, D.M.; Stone, M.L.; Riehl, B. Ligament surgery,


the evaluation of results. In Knee Ligaments: Structure,
Function, Injury, and Repair; Daniel, D.M., Akeson, W.H.,
O’Connor, J.J., Eds.; Raven Press: New York, NY, 1990.

6. Diegelmann, R.F.; Evans, M.C. Wound healing: an overview


of acute, fibrotic, and delayed healing. Front. Biosci.
2004, 9, 283–289.

7. Frank, C. Medial collateral ligament healing. A


multidisciplinary assessment in rabbits. Am. J. Sports Med.
1983, 11, 379–389.

8. Gerber, H.P.; Vu, T.H.; Ryan, A.M.; Kowalski, J.; Werb,


Z.; Ferrara, N. VEGF couples hypertrophic cartilage
remodeling, ossification and angiogenesis during
endochondral bone formation. Nat. Med. 1999, 5, 623–628.

9. Hardingham, T.; Tew, S.; Murdoch, A. Tissue engineering:


chondrocytes and cartilage. Arthritis Res. 2002, 4,
S63–S68.

10. Indelicato, P.A. Non-operative treatment of complete


tears of the medial collateral ligament of the knee. J.
Bone Joint Surg. Am. 1983, 65, 323–329.

11. Kang, Q.; Sun, M.H.; Cheng, H. Characterization of the


distinct orthotopic bone-forming activity of 14 BMPs using
recombinant adenovirus-mediated gene delivery. Gene Ther.
2004, 11, 1312–1320.

12. Kim, W.J.; Gittes, G.K.; Longaker, M.T. Signal


transduction in wound pharmacology. Arch. Pharm. Res. 1998,
21, 487. 13. Kuznetsov, S.A.; Mankani, M.H.; Gronthos, S.;
Satomura, K.; Bianco, P.; Robey, P.G. Circulating skeletal
stem cells. J. Cell Biol. 2001, 153, 1133–1140. 14. Leach,
J.K.; Mooney, D.J. Bone engineering by controlled delivery
of osteoinductive molecules and cells. Expert Opin. Biol.
Ther. 2004, 4, 1015–1027. 15. Lorenz, H.P.; Longaker, M.T.
Wounds: biology, pathology, and management. In Surgery:
Basic Science and Clinical Evidence; Norton, J.A.,
Bollinger, R.R., Chang, A.E., Lowry, S.F., Mulvihill, S.J.,
Pass, H.I., Thompson, R.W., Eds.; Springer: New York, 2000;
77–88. 16. Marieb, E.N. Anatomy and Physiology; Pearson/
Benjamin Cummings: San Francisco, 2005. 17. Murphy, W.L.;
Simmons, C.A.; Kaigler, D.; Mooney, D.J. Bone regeneration
via a mineral substrate and induced angiogenesis. J. Dent.
Res. 2004, 83, 204–210. 18. O’Driscoll, S.W. The healing
and regeneration of articular cartilage. J. Bone Joint
Surg. Am. 1998, 80, 1795–1812. 19. Park, J.E.; Barbul, A.
Understanding the role of immune regulation in wound
healing. Am. J. Surg. 2004, 187, 11S–16S. 20. Redd, M.J.;
Cooper, L.; Wood, W.; Stramer, B.; Martin, P. Wound healing
and inflammation: embryos reveal the way to perfect repair.
Trans. R. Soc. Lond. 2004, 359B, 777–784. 21. Roberts,
H.R.; Tabares, A.H. Overview of the coagulaiton reactions.
In Molecular Basis of Thrombosis and Hemostasis; High,
K.A., Roberts, H.R., Eds.; Dekker: New York, 1995; 35–50.
22. Sellers, R.; Peluso, D.; Morris, E.A. The effect of
recombinant human bone morphogenic protein-2 (rhBMP-2) on
the healing of full thickness defects of articular
cartilage. J. Bone Joint Surg. Am. 1997, 79, 1452–1463.
Hearing Aids

1. Clark, G.M. Cochlear Implants: Fundamentals and


Applications; Springer-Verlag: New York, NY, 2003.

2. Nolte, J. The Human Brain: An Introduction to its


Functional Anatomy, 3rd Ed.; Mosby: St. Louis, MO, 1993.

3. Brownell, W.E.; Bader, C.R.; Bertrand, D.; de


Ribaupierre, Y. Evoked mechanical responses of isolated
cochlear outer hair cells. Science 1985, 227 (4683),
194–196.

4. Sachs, M.B.; Bruce, I.C.; Miller, R.L.; Young, E.D.


Biological basis of hearing-aid design. Ann. Biomed. Eng.
2002, 30, 157–168.

5. Robles, L.; Ruggero, M.A. Mechanics of the mammalian


cochlea. Physiol. Rev. 2001, 81, 1305–1352.

6. Bruce, I.C.; Sachs, M.B.; Young, E.D. An


auditoryperiphery model of the effects of acoustic trauma
on auditory nerve responses. J. Acoust. Soc. Am. 2003, 113
(1), 369–388.

7. Heinz, M.G.; Issa, J.B.; Young, E.D. Auditory-nerve rate


responses are inconsistent with common hypotheses for the
neural correlates of loudness recruitment. JARO 2005, 6
(2), 91–105.

8. Bondy, J.; Bruce, I.C.; Dong, R.; Becker, S.; Haykin, S.


Modeling intelligibility of hearing-aid compression
circuits. Conference Records of the Thirty-Seventh Asilomar
Conference on Signals, Systems and Computers. IEEE Press:
Piscataway, NJ, 2003; Vol. 1, 720–724.

9. Plomp, R.; Duquesnoy, A.J. A model for the


speechreception threshold in noise without and with a
hearing aid. Scand. Audiol. Suppl. 1982, 15, 95–111.

10. Dillon, H. Hearing Aids; Thieme Medical Publishers: New


York, NY, 2001.
Hearing Mechanisms

7. Wever, E.G. The Reptile Ear; Princeton University Press,


1978.

8. Lenhardt, M.L. Eardrum Saccular Coupling: Novel Form of


Hearing. In Biomedical Engineering: Recent Developments;
Vossougghi, J., Ed.; Medical and Engineering Publisher:
Washington, DC, 2002; 51–52. 9. Lenhardt, M.L.; Skellett,
R.; Wang, P.; Clarke, A.M. Human ultrasonic speech
perception. Science 1991, 5, 253 (5015), 82–85. 10.
Imaizumi, S.; Hosoi, H.; Sakaguchi, T.; Watanabe, Y.;
Sadato, N.; Nakamura, S.; Waki, A.; Yonekura, Y. Ultrasound
activates the auditory cortex of profoundly deaf subjects.
NeuroReport 2001, 5, 12 (3), 583–586. H
Heart Biomechanics

16. Humphrey, J.D.; Strumpf, R.K.; Yin, F.C.P.


Determination of a constitutive relation for passive
myocardium: I. A new functional form. J. Biomech. Eng.
1990, 112, 333–339.

17. Humphrey, J.D.; Strumpf, R.K.; Yin, F.C.P.


Determination of a constitutive relation for passive
myocardium: II. Parameter estimation. J. Biomech. Eng.
1990, 112, 340–346.

18. Guccione, J.M.; McCulloch, A.M.; Waldman, L.K. Passive


material properties of intact ventricular myocardium
determined from a cylindrical model. J. Biomech. Eng. 1991,
113, 42–55.

19. Omens, J.H.; MacKenna, D.A.; McCulloch, A.D.


Measurement of strain and analysis of stress in resting rat
left ventricular myocardium. J. Biomech. 1993, 26 (6),
665–676.

20. Holmes, J.W.; Borg, T.K.; Covell, J.W. Structure and


mechanics of healing myocardial infarcts. Ann. Rev. Biomed.
Eng. 2005, 7, 223–253.

21. Criscione, J.C.; Sacks, M.S.; Hunter, W.C.


Experimentally tractable, pseudo-elastic constitutive law
for biomembranes: I. Theory. J. Biomech. Eng. 2003, 125
(1), 94–99.

22. Criscione, J.C.; Sacks, M.S.; Hunter, W.C.


Experimentally tractable, pseudo-elastic constitutive law
for biomembranes: II. Application. J. Biomech. Eng. 2003,
125 (1), 100–105.

23. Hunter, P.J. Myocardial constitutive laws for continuum


mechanics models of the heart. Adv. Exp. Med. Biol. 1995,
382, 303–318.

24. Horowitz, A.; Lanir, Y.; Yin, F.C.P.; Perl, M.;


Sheinman, I.; Strumpf, R.K. Structural three-dimensional
constitutive law for the passive myocardium. J. Biomech.
Eng. 1988, 110, 200–207. 25. Sacks, M.S. Incorporation of
experimentally-derived fiber orientation into a structural
constitutive model for planar collagenous tissues. J.
Biomech. Eng. 2003, 125 (2), 280–287. 26. Grossman, W.;
Jones, D.; McLaurin, L.P. Wall stress and patterns of
hypertrophy in the human left ventricle. J. Clin. Invest.
1975, 56, 56–64. 27. Holmes, J.W. Candidate mechanical
stimuli for hypertrophy during volume overload. J. Appl.
Physiol. 2004, 97 (4), 1453–1460. 28. Batista, R.J.; Verde,
J.; Nery, P.; Bocchino, L.; Takeshita, N.; Bhayana, J.N.;
Bergsland, J.; Graham, S.; Houck, J.P.; Salerno, T.A.
Partial left ventriculectomy to treat end-stage heart
disease. Ann. Thorac. Surg. 1997, 64 (3), 634–638. 29.
Burkhoff, D.; Holmes, J.W.; Madigan, J.; Barbone, A.; Oz,
M.C. Left ventricular assist device-induced reverse
ventricular remodeling. Prog. Cardiovasc. Dis. 2000, 43,
19–26. 30. Barbone, A.; Oz, M.C.; Burkhoff, D.; Holmes,
J.W. Normalized diastolic properties after left ventricular
assist result from reverse remodeling of chamber geometry.
Circulation Supplement 2001, 104 (suppl I), I-229–I-232.
31. Arts, T.; Prinzen, F.W.; Snoeckx, L.H.E.H.; Rijcken,
J.M.; Reneman, R.S. Adaptation of cardiac structure by
mechanical feedback in the environment of the cell: a model
study. Biophys. J. 1994, 66, 953–961. 32. Lin, I.E.; Taber,
L.A. A model for stress-induced growth in the developing
heart. J. Biomech. Eng. 1995, 117 (3), 343–349. 33. Glass,
L.; Hunter, P.; McCulloch, A. Theory of Heart:
Biomechanics, Biophysics, and Nonlinear Dynamics of Cardiac
Function; Springer-Verlag: New York, 1991.
Heart Valve, Bioprosthetic

3. Curcio, C.; Commerford, P.; Rose, A.; Stevens, J.;


Barnard, M. Calcification of glutaraldehyde-preserved
porcine xenografts in young patients J. Thorac. Cardiovasc.
Surg. 1981, 81, 621–625.

4. Rose, A.; Forman, R.; Bowen, R. Calcification of


glutaraldehyde-fixed porcine xenograft. Thorax 1978, 33,
111–114.

5. Arom, K.V.; Emery, R.W.; Nicoloff, D.M.; Petersen, R.J.


Anticoagulant related complications in elderly patients
with St. Jude mechanical valve prostheses. J. Heart Valve
Dis. 1996, 5, 505–510.

6. Harken, D.; Soroff, H.; Taylor, W.; Lefemine, A.; Gupta,


S.; Lunzer, S. Partial and complete prostheses in aortic
insufficiency. J. Thorac. Cardiovasc. Surg. 1960, 40,
744–762.

7. Starr, A.; Edwards, M. Mitral replacement: Clinical


experiences with a ball-valve prosthesis. Ann. Surg. 1961,
154, 726.

8. Frater, R.; Berghuis, J.; Brown, A.; Ellis, F.


Autogenous pericardium for posterior mitral leaflet
replacement. Surgery 1963, 84, 260–268.

9. Frater, R.; Berghuis, J.; Brown, A.; Ellis, F. The


experimental and clinical use of autogenous pericardium for
the replacement and extension of mitral and triscuspid
valve cusps and chordae. J. Cardiovasc. Surg. 1965, 6,
214–228.

10. Ross, D. Homograft replacement of the aortic valve.


Lancet 1962, 2, 487.

11. Mohri, H.; Reichenbach, D.; Barnes, R.; Nelson, R.;


Merendino, K. Studies of antigenicity of the homologous
aortic valve. J. Thorac. Cardiovasc. Surg. 1967, 54,
564–572.

12. Davies, H.; Missen, G.; Blandford, G.; Roberts, C.;


Lessof, M.; Ross, D. Homograft replacement of the aortic
valve. A clinical and pathologic study. Am. J. Cardiol.
1968, 22, 195–217.

13. Davies, H.; Lessaf, M.; Roberts, C.; Ross, D. Homograft


replacement of the aortic valve: Follow-up studies in
twelve patients. Lancet 1965, 1, 926.

14. Ross, D. Replacement of aortic and mitral valves with a


pulmonary autograft. Lancet 1967, 2, 956–958.

15. Duran, C.; Gunning, A.; Whitehead, R. Experimental


aortic valve heterotransplantation. Thorax 1967, 22,
510–518.

16. Binet, J.; Carpentier, A.; Langlois, J.; Duran, C.;


Colvez, P. Implantation of heterogenic valves in the
treatment of aortic cardiopathies. C. R. Hebd. Seances
Acad. Sci., Ser. D, Sci. Nat. 1965, 261, 5733–5734.

17. Carpentier, A.; Lemaigre, G.; Robert, L.; Carpentier,


S.; Dubost, C. Biological factors affecting long-term
results of valvular heterografts. J. Thorac. Cardiovasc.
Surg. 1969, 58 , 467–483.

18. Rosenberg, N.; Henderson, J.; Douglas, J.; Lord, G.;


Gaughran, E. Use of enzyme-digested heterografts as
segmental arterial substitutes. III. Follow-up observations
on two-year-old aortic implants. Arch. Surg. 1958, 76, 275.

19. Lentz, D.; Pollock, E.; Olsen, D.; Andrews, E.


Prevention of intrinsic calcification in porcine and bovine
xenograft materials. Trans. Am. Soc. Artif. Intern. Organs
1982, 28, 494–497. 20. Dewanjee, M.; Solis, E.; Lanker, J.;
Mackey, S.; Lombardo, G.; Tidwell, C.; Ellefsen, R.; Kaye,
M. Effect of diphosphonate binding to collagen upon
inhibition of calcification and promotion of spontaneous
endothelial cell coverage on tissue valve prostheses. ASAIO
Trans. 1986, 32, 24–29. 21. Zilla, P.; Fullard, L.;
Trescony, P.; Meinhart, J.; Bezuidenhout, D.; Gorlitzer,
M.; Human, P.; von Oppell, U. Glutaraldehyde detoxification
of aortic wall tissue: A promising perspective for emerging
bioprosthetic valve concepts. J. Heart Valve Dis. 1997, 6,
510–520. 22. Girardot, M.; Girardot, J.; Schoen, F. Alpha
amino oleic acid, a new compound, prevents calcification of
bioprosthetic heart valves. Trans. Soc. Biomater. 1991, 14,
114(abstract) 23. Carpentier, A. Bovine Pericardium or
Porcine Valves? Pericardial Bioprostheses Mini-Symposium,
Luzern, Switzerland, 1989. 24. Perier, P. Seven Years
Clinical Follow-Up of Aortic Valve Replacement with the
Carpentier-Edwards Pericardial Valve, Pericardial
Bioprostheses MiniSymposium, Luzern, Switzerland, 1989. 25.
Gabbay, S.; Kadam, P.; Factor, S.; Cheung, T. Do heart
valve bioprostheses degenerate for metabolic or mechanical
reasons? J. Thorac. Cardiovasc. Surg. 1988, 95, 208–215.
26. Grunkemeier, G.; Bodnar, E. Comparative assessment of
bioprosthesis durability in the aortic position. J. Heart
Valve Dis. 1995, 4, 49–55. 27. Love, J.; Schoen, F.;
Breznock, E.; Shermer, S.; CS, L. Experimental evaluation
of an autologous tissue heart valve. J. Heart Valve Dis.
1992, 1, 232–241. 28. Goetz, W.; Lim, H.; Lansac, E.;
Weber, P.; Duran, C. A temporarily stented, autologous
pericardial aortic valve prosthesis. J. Heart Valve Dis.
2002, 11, 696–702. 29. Butany, J.; deSa, M.; Feindel, C.;
David, T. The Toronto SPV bioprosthesis: Review of
morphologic findings in eight valves. Semin. Thorac.
Cardiovasc. Surg. 1999, 11 (4 Suppl.), 157–162. 30.
Goldman, B.; Christakis, G.; David, T.; Rakowski, H.; Bach,
D.; Wood, J.; Goldman, S.; Pepper, J.; Yacoub, M.; Verrier,
E.; Petracek, M. Will stentless valves be durable? The
Toronto valve (TSPV) at 5 to 6 years. Semin. Thorac.
Cardiovasc. Surg. 1999, 11, 42–49. 31. Westaby, S.; Jin,
X.; Katsumata, T.; Arifi, A.; Braidley, P. Valve replacement
with a stentless bioprosthesis: Versatility of the procine
aortic root. J. Thorac. Cardiovasc. Surg. 1998, 116,
477–848. 32. Dahm, M.; Lyman, W.; Schwell, A.; Factor, S.;
Frater, R. Immunogenicity of glutaraldehyde-tanned bovine
pericardium. J. Thorac. Cardiovasc. Surg. 1990, 99,
1082–1090. 33. Cobanoglu, A.; Jamieson, W.; Miller, D.;
McKinley, C.; Grunkemeier, G.; Floten, H.; Miyagishima, R.;
Tyers, G.; Shumway, N.; Starr, A. A tri-institutional
comparison of tissue and mechanical values using a
patient-oriented definition of ‘‘treatment failure’’. Ann.
Thorac. Surg. 1987, 43, 245–253. 34. David, T.; Armstrong,
S.; Sun, Z. The Hancock II bioprosthesis at ten years. Ann.
Thorac. Surg. 1995, 60, S229–S234. 35. Gott, J.; Pan, C.;
Dorsey, L.; Jay, J.; Jett, G.; Schoen, F.; Girardot, J.;
Guyton, R. Calcification of porcine valves: A successful new
method of antimineralization. Ann. Thorac. Surg. 1992, 53,
207–215discussion 216 36. Cosgrove, D.; Frater, R.;
Scott-Page, U. Clinical communique Edwards Lifesci. LLC.
37. Maxwell, L.; Gavin, J.; Barratt-Boyes, B. Uneven host
tissue ongrowth and tissue detachment in stent mounted
heart valve allografts and xenografts. Cardiovasc. Res.
1989, 23, 709–714. 38. Schoen, F.; Hobson, C. Anatomic
analysis of removed prosthetic heart valves: Causes of
failure of 33mechanical valves and 58 bioprostheses, 1980
to 1983 Human Pathol. 1985, 16, 549–559. 39. Camilleri, J.;
Pornin, B.; Carpentier, A. Structural changes of
glutaraldehyde-treated porcine bioprosthetic valves. Arch.
Pathol. Lab. Med. 1982, 106, 490–496. 40. Zilla, P.; Zhang,
Y.; Human, P.; Koen, W.; von Oppell, U. Improved
ultrastructural preservation of bioprosthetic tissue. J.
Heart Valve Dis. 1997, 6, 492–501. 41. Human, P.;
Weissenstein, C.; Trantina, A.; Zilla, P. Fixation-related
autolysis and bioprosthetic aortic wall calcification. J.
Heart Valve Dis. 2001, 10, 656–665. 42. Simionescu, D.;
Lovekamp, J.; Vyavahare, N. Degeneration of bioprosthetic
heart valve cusp and wall tissues is initiated during
tissue preparation: An ultrastructural study. J. Heart
Valve Dis. 2003, 12, 226–234. 43. Lemasters, J.;
DiGuiseppi, J.; Nieminen, A.; Herman, B. Blebbing, free
Ca2+ and mitochondrial membrane potential preceding cell
death in hepatocytes. Nature 1987, 325, 78–81. 44. Kim, K.
Cell death and calcification of canine fibroblasts in vitro.
Cells Mater. 1994, 4, 247–261. 45. Trump, B.; Valigorsky,
J.; Dees, J.; Mergner, W.; Kim, K.; Jones, R.; Pendergrass,
R.; Garbus, J.; Cowley, R. Cellular changes in human
disease. A new method of pathological analysis. Human
Pathol. 1973, 4, 89–109. 46. Trump, B.; Berezesky, I.;
Laiho, K.; Osorino, A.; Mergner, W.; Smith, M. The role of
calcium in cell injury. A review. Scanning Electron
Microsc. 1980, II, 437–462. 47. Ferrans, V.; Spray, L.;
Billingham, M.; Roberts, W. Structural changes in
glutaraldehyde-treated porcine heterografts used as
substitute cardiac valves. Am. J. Cardiol. 1978, 41,
1159–1184. 48. Ashraf, M.; Bloor, C. Structural alterations
of the porcine heterograft after various durations of
implantation. Am. J. Cardiol. 1978, 41, 1185–1190. 49.
Anderson, H. Calcification processes. Pathol. Annu. 1980,
15, 45–75. 50. Morea, M.; De, P.R.; Galloni, M.; Gastaldi,
L.; di, S.M. Mitral valve replacement with the Biocor
stentless mitral valve: Early results. J. Heart Valve Dis.
1994, 3, 476–482. 51. Ishihara, T.; Ferrans, V.; Jones, M.;
Boyce, S.; Roberts, W. Structure of bovine parietal
pericardium and of unimplanted Ionescu-Shiley pericardial
valvular bioprostheses. J. Thorac. Cardiovasc. Surg. 1981,
81, 747–757. 52. Schoen, F.; Levy, R.; Hilbert, S.; Bianco,
R. Antimineralization treatments for bioprosthetic heart
valves. Assessment of efficacy and safety. J. Thorac.
Cardiovasc. Surg. 1992, 104, 1285–1288. 53. Chen, W.; Kim,
J.; Schoen, F.; Levy, R. Effect of 2-amino oleic acid
exposure conditions on the inhibition of calcification of
glutaraldehyde cross-linked porcine aortic valves. J.
Biomed. Mater. Res. 1994, 28, 1485–1495. 54. Zilla, P.;
Weissenstein, C.; Bracher, M.; Zhang, Y.; Koen, W.; Human,
P.; von Oppell, U. High glutaraldehyde concentrations
reduce rather than increase the calcification of aortic wall
tissue. J. Heart Valve Dis. 1997, 6, 502–509. 55. Scott,
M.; Vesely, I. Aortic valve cusp microstructure: The role
of elastin. Ann. Thorac. Surg. 1995, 60, S391–S394. 56.
Schoen, J.; Shemin, R.; Cohn, L. Pathologic evaluation of a
unileaflet pericardial bioprosthesis after implantation as
tricuspid and mitral valve replacements in sheep. Trans.
Soc. Biomater. 1986, 9, 76. 57. Sterling, H.; Saginario,
C.; Vignery, A. CD44 occupancy prevents macrophage
multinucleation. J. Cell Biol. 1998, 143, 837–847. 58.
Vignery, A.; Niven-Fairchild, T.; Ingbar, D.; Caplan, M.
Polarized distribution of Na+K+-ATPase in giant cells
elicited in vivo and in vitro. J. Histochem. Cytochem.
1989, 35, 1265–1271. 59. Valente, M.; Bortolotti, U.;
Thiene, G. Ultrastructural substrates of dystrophic
calcification in porcine bioprosthetic valve failure. Am. J.
Pathol. 1985, 119, 12–21. 60. Dahm, M.; Husmann, M.;
Eckhard, M.; Prufer, D.; Groh, E.; Oelert, H. Relevance of
immunologic reactions for tissue failure of bioprosthetic
heart valves. Ann. Thorac. Surg. 1995, 60, S348–S352. 61.
Nistal, F.; Garcia, S.E.; Artinano, E.; Duran, C.; Gallo,
I. Comparative study of primary tissue valve failure
between Ionescu-Shiley pericardial and Hancock porcine
valves in the aortic position. Am. J. Cardiol. 1986, 57,
161–164. 62. Fini, M.; Girard, M.; Matsubara, M.; Bertlett,
J. Unique regulation of the matrix metalloproteinase,
gelatinase B. Investig. Ophthalmol. Vis. Sci. 1995, 36,
622–632. 63. Galis, Z.; Sukhova, G.; Libby, P. Microscopic
localization of active proteases by in situ zymography:
Detection of matrix metalloproteinase activity in vascular
tissue. FASEB J. 1995, 9, 974–980. 64. Ellsmere, J.;
Khanna, R.; Lee, J. Mechanical loading of bovine
pericardium accelerates enzymatic degradation. Biomaterials
1999, 20, 1143–1150. 65. Trantina, Y.A.; Weissenstein, C.;
Human, P.; Zilla, P. Stentless bioprosthetic heart valve
research: Sheep versus primate model. Ann. Thorac. Surg.
2001, 71, S422–S427. 66. Stein, P.; Wang, C.; Riddle, J.;
Magilligan, D.J. Leukocytes, platelets, and surface
microstructure of spontaneously degenerated porcine
bioprosthetic valves. J. Card. Surg. 1988, 3, 253–261. H

67. Everts, V.; van, D.Z.E.; Creemers, L.; Beertsen, W.


Phagocytosis and intracellular digestion of collagen, its
role in turnover and remodelling. Histochem. J. 1996, 28,
229–245.

68. Grabenwoger, M.; Grimm, M.; Eybl, E.; Leukauf, C.;


Muller, M.; Plenck, H.J.; Bock, P. Decreased tissue
reaction to bioprosthetic heart valve material after
L-glutamic acid treatment. A morphological study. J.
Biomed. Mater. Res. 1992, 26, 1231–1240.

69. Grabenwoger, M.; Grimm, M.; Eybl, E.; Kadletz, M.;


Havel, M.; Kostler, P.; Plenk, H.; Bock, P.; Wolner, E. New
aspects of the degeneration of bioprosthetic heart valves
after long-term implantation. J. Thorac. Cardiovasc. Surg.
1992, 104, 14–21.

70. Tang, L.; Ugarova, T.; Plow, E.; Eaton, J. Molecular


determinants of acute inflammatory responses to
biomaterials. J. Clin. Invest. 1996, 97, 1329–1334.

71. Human, P.; Zilla, P. Characterization of the immune


response to valve bioprostheses and its role in primary
tissue failure. Ann. Thorac. Surg. 2001, 71, S385–S388.

72. Sherman, F.; Schoen, F.; Hawley, M.; Nichols, J.; Levy,
R. Collagen crosslinks: A critical determinant of
bioprosthetic heart valve calcification Trans. Am. Soc.
Artif. Intern. Organs 1984, 30, 577–581.

73. Human, P.; Zilla, P. Inflammatory and immune processes:


The neglected villain of bioprosthetic degeneration? J.
Long-Term Eff. Med. Implants 2001, 11.

74. Ferrans, V.; Boyce, S.; Billingham, M.; Jones, M.;


Ishihara, T.; Roberts, W. Calcific deposits in porcine
bioprostheses: Structure and pathogenesis. Am. J. Cardiol.
1980, 46, 721–734.

75. Zilla, P.; Bezuidenhout, D.; Weissenstein, C.; Van der


Walt, A.; Human, P. Diamine extension of glutaraldehyde
crosslinks mitigates bioprosthetic aortic wall calcification
in the sheep model. J. Biomed. Mater. Res. 2001, 56, 56–64.

76. Sprague, E.; Mohan, S.; Nerem, R. Shear stress


regulation of monocyte/endothelial interactions. J. Vasc.
Surg. 1999, 29, 1138–1140.

77. Ku, D. Blood flow in arteries. Annu. Rev. Fluid Mech.


1997, 29, 399–434.

78. Zilla, P.; Weissenstein, C.; Human, P.; Dower, T.; von
Oppell, U. High glutaraldehyde concentrations mitigate
bioprosthetic root calcification in the sheep model. Ann.
Thorac. Surg. 2000, 70, 2091–2095.

79. Yoganathan, A.; Ellis, J.; Healy, T.; Chatzimavroudis,


G. Fluid dynamic studies for the year 2000. J. Heart Valve
Dis. 1998, 7, 130–139.

80. Thubrikar, M.; Bosher, L.; Nolan, S. The mechanism of


opening of the aortic valve. J. Thorac. Cardiovasc. Surg.
1979, 77, 863–870.

81. Vesely, I.; Noseworthy, R. Micromechanics of the fibrosa


and the ventricularis in aortic valve leaflets. J. Biomech.
1992, 25, 101–113.
82. Kastelic, J.; Baer, E. Deformation in tendon collagen.
Symp. Soc. Exp. Biol. 1980, 34, 397–420.

83. Allen, D.; DiDio, L.; Zacharias, A.; Fentie, I.;


McGrath, A.; Puig, L.; Pomerantzeff, P.; Zerbini, E.
Microscopic study of normal parietal pericardium and
unimplanted Puig-Zerbini pericardial valvular heterografts.
J. Thorac. Cardiovasc. Surg. 1984, 87, 845–855. 84. Vesely,
I.; Mako, W. Comparison of the compressive buckling of
porcine aortic valve cusps and bovine pericardium. J. Heart
Valve Dis. 1998, 7, 34–39. 85. Sacks, M. The biomechanical
effects of fatigue on the porcine bioprosthetic heart
valve. J. Long-Term Eff. Med. Implants 2001, 11, 231–247.
86. Christie, G.; Barratt-Boyes, B. Time-Dependent Changes
to the Leaflet Elasticity of the Medtronic Intact Valve
in-vivo, World Symposium on Heart Valve Disease, London,
UK, 1999. 87. Vyavahare, N.; Ogle, M.; Schoen, F.; Zand,
R.; Gloeckner, D.; Sacks, M.; Levy, R. Mechanisms of
bioprosthetic heart valve failure: Fatigue causes collagen
denaturation and glycosaminoglycan loss. J. Biomed. Mater.
Res. 1999, 46, 44–50. 88. Simionescu, D.; Simionescu, A.;
Deac, R. Detection of remnant proteolytic activities in
unimplanted glutaraldehyde-treated bovine pericardium and
explanted cardiac bioprostheses. J. Biomed. Mater. Res.
1993, 27, 821–829. 89. Bonfield, T.; Anderson, J. Functional
versus quantitative comparison of IL-1 beta from monocytes/
macrophages on biomedical polymers. J. Biomed. Mater. Res.
1993, 27, 1195–1199. [published erratum appears in J.
Biomed. Mater. Res. 1994 Jan;28(1);137]. 90. Greisler, H.;
Dennis, J.; Endean, E.; Ellinger, J.; Friesel, R.; Burgess,
W. Macrophage/biomaterial interactions: The stimulation of
endothelialization. J. Vasc. Surg. 1989, 9, 588–593. 91.
Ciapetti, G.; Verri, E.; Granchi, D.; Cenni, E.; Gamberini,
S.; Benetti, D.; Mian, M.; Pizzoferrato, A. In vitro
assessment of phagocytosis of bovine collagen by human
monocytes/macrophages using a spectrophotometric method.
Biomaterials 1996, 17, 1703–1707. 92. Anderson, J.; Miller,
K. Biomaterial biocompatibility and the macrophage.
Biomaterials 1984, 5, 5–10. 93. Forsyth, E.; Aly, H.;
Neville, R.; Sidawy, A. Proliferation and extracellular
matrix production by human infragenicular smooth muscle
cells in response to interleukin-1 beta. J. Vasc. Surg.
1997, 26, 1002– 1007. discussion 1007–1008. 94. Clinton,
S.; Libby, P. Cytokines and growth factors in
atherogenesis. Arch. Pathol. Lab. Med. 1992, 116,
1292–1300. 95. Raines, E.; Dower, S.; Ross, R.
Interleukin-1mitogenic activity for fibroblasts and smooth
muscle cells is due to PDGF-AA. Science 1989, 243, 393–396.
96. Nathan, C. Secretory products of macrophages. J. Clin.
Invest. 1987, 79, 319–326. 97. Oppenheim, J.; Kovacs, E.;
Matasushima, K.; Durum, S. There is more than one
interleukin 1. Immunol. Today 1986, 7, 45. 98. Dinarello,
C. Interleukin 1. Rev. Infect. Dis. 1984, 6, 51. 99.
Freundlich, B.; Bomalski, J.; Neilson, E.; Jimenez, S.
Regulation of fibroblast proliferation and collagen
synthesis by cytokines. Immunol. Today 1986, 7, 303. 100.
Grinnell, F. Fibronectin and wound healing. Am. J.
Dermatopathol. 1982, 4, 185–188. 101. Oppenheim, J.; Gery,
I. Interleukin 1 is more than an interleukin. Immunol.
Today 1982, 3, 113. 102. Schmidt, J.; Mizel, S.; Cohen, D.;
Green, I. Interleukin 1, a potential regulator of fibroblast
proliferation. J. Immunol. 1982, 128, 2177–2182. 103.
Tsukamoto, Y.; Helsel, W.; Wahl, S. Macrophage production
of fibronectin, a chemoattractant for fibroblasts. J.
Immunol. 1981, 127, 673–678. 104. Wewers, M.; Marsh, C.
Role of the antibody in the pathogenesis of transplant
vascular sclerosis: A hypothesis. Transpl. Immunol. 1997,
5, 283–288. 105. Green, M.; Walsh, M.; Dare, A.; Hogan, P.;
Zhao, X.; Frazer, I.; Bansal, A.; O’Brien, M. Histologic
and immunohistochemical responses after aortic valve
allografts in the rat. Ann. Thorac. Surg. 1998, 66,
S216–S220. 106. Miller, K.; Huskey, R.; Bigby, L.;
Anderson, J. Characterization of biomedical
polymer-adherent macrophages: Interleukin 1 generation and
scanning electron microscopy studies. Biomaterials 1989,
10, 187–196. 107. Bonfield, T.; Colton, E.; Marchant, R.;
Anderson, J. Cytokine and growth factor production by
monocytes/ macrophages on protein preadsorbed polymers. J.
Biomed. Mater. Res. 1992, 26, 837–850. 108. Dower, T.;
Adler, U.; Davids, L.; Zilla, P. Increasing cross-linking
efficiency mitigates macrophage activation on bioprosthetic
tissue. Cardiovasc. Pathol. 1998, 7, 295. H
Heart Valve Failure, Bioprosthetic

1. Odell, J.; Gillmer, D.; Whitten, I.; Vythilingun, S.;


Vanker, E. Calcification of Tissue Valves in Children:
Occurrence in Porcine and Bovine Pericardial Bioprosthetic
Valves. In Biologic and Bioprosthetic Valves; Bodnar, E.,
Yacoub, M., Eds.; Yorke Medical Books: New York, 1985; 259.

2. Cheung, D.; Perelman, N.; Ko, E.; Nimni, M. Mechanism of


crosslinking of proteins by glutaraldehyde III. Reaction
with collagen in tissues. Connect. Tissue Res. 1985, 13,
109–115.

3. Hardy, P.; Nicholls, A.; Rydon, H. The nature of


crosslinking of proteins by glutaraldehyde. 1. Interaction
of glutaraldehyde with the amino-groups of 6-aminohexanoic
acid and of alpha-N-acetyl-lysine. J. Chem. Soc., Perkin
1976, 1, 958–962.

4. Woodroof, E. Use of glutaraldehyde and formaldehyde to


process tissue heart valves. J. Bioeng. 1978, 2, 1–9.

5. Johnson, T. Glutaraldehyde fixation chemistry:


Oxygen-consuming reactions. Eur. J. Cell Biol. 1987, 45,
160–169.

6. Sacks, M. The biomechanical effects of fatigue on the


porcine bioprosthetic heart valve. J. Long-Term Eff. Med.
Implants 2001, 11, 231–247.

7. Weissenstein, C.; Human, P.; Bezuidenhout, D.; Zilla, P.


Glutaraldehyde detoxification in addition to enhanced amine
cross-linking dramatically reduces bioprosthetic tissue
calcification in the rat model. J. Heart Valve Dis. 2000, 9,
230–240.

8. Zilla, P.; Fullard, L.; Trescony, P.; Meinhart, J.;


Bezuidenhout, D.; Gorlitzer, M.; Human, P.; von Oppell, U.
Glutaraldehyde detoxification of aortic wall tissue: A
promising perspective for emerging bioprosthetic valve
concepts. J. Heart Valve Dis. 1997, 6, 510–520.

9. Dewanjee, M.; Solis, E.; Lanker, J.; Mackey, S.;


Lombardo, G.; Tidwell, C.; Ellefsen, R.; Kaye, M. Effect of
diphosphonate binding to collagen upon inhibition of
calcification and promotion of spontaneous endothelial cell
coverage on tissue valve prostheses. ASAIO Trans. 1986, 32,
24–29.

10. Chen, W.; Schoen, F.; Levy, R. Mechanism of efficacy of


2-amino oleic acid for inhibition of calcification of
glutaraldehyde-pretreated porcine bioprosthetic heart
valves. Circulation 1994, 90, 323–329.

11. Grabenwoger, M.; Grimm, M.; Eybl, E.; Leukauf, C.;


Muller, M.; Plenck, H.J.; Bock, P. Decreased tissue
reaction to bioprosthetic heart valve material after
L-glutamic acid treatment. A morphological study. J.
Biomed. Mater. Res. 1992, 26, 1231–1240. 12. Golomb, G.;
Schoen, F.; Smith, M.; Linden, J.; Dixon, M.; Levy, R. The
role of glutaraldehyde-induced crosslinks in calcification
of bovine pericardium used in cardiac valve bioprostheses.
Am. J. Pathol. 1987, 127, 122–130. 13. Han, R.; Zilla, P.;
Hendricks, M.; Torrianni, M.; Human, P. Carboxyl-group
crosslinking is essential to supprress cellular
calcification in bioprosthetic tissue. J. Thorac. Cadiovasc.
Surg. 2003. (Submitted). 14. Kim, K. Pathologic
Calcification. In Pathobiology of Cell Membranes ; Trump,
B., Arstilla, A., Eds.; Academic Press: New York, 1983;
117–155. 15. Schoen, F.; Tsao, J.; Levy, R. Calcification of
bovine pericardium used in cardiac valve bioprostheses.
Implications for the mechanisms of bioprosthetic tissue
mineralization. Am. J. Pathol. 1986, 123, 134–145. 16.
Levy, R.; Schoen, F.; Flowers, W.; Staelin, S. Initiation
of mineralization in bioprosthetic heart valves: Studies of
alkaline phosphatase activity and its inhibition by AlCl3
or FeCl3 preincubations. J. Biomed. Mater. Res. 1991, 25,
905–935. 17. Hirsch, D.; Schoen, F.; Levy, R. Effects of
metallic ions and diphosphonates on inhibition of
pericardial bioprosthetic tissue calcification and
associated alkaline phosphatase activity. Biomaterials
1993, 14, 371–377. 18. Somlyo, A.P.; Somlyo, A.V. Signal
transduction and regulation in smooth muscle. Nature 1994,
372, 231–236. 19. Mintz, E.; Guillain, F. Ca2+ transport by
the sarcoplasmic reticulum ATPase. Biochim. Biophys. Acta
1997, 1318, 52–70. 20. Canet, D.; Forge, V.; Guillain, F.;
Mintz, E. Ca2+ translocation across sarcoplasmic reticulum
ATPase randomizes the two transported ions. J. Biol. Chem.
1996, 271, 20566–20572. 21. Anderson, H. Calcification
processes. Pathol. Annu. 1980, 15, 45–75. 22. Schoen, F.J.
Future directions in tissue heart valves: Impact of recent
insights from biology and pathology [see comments]. J.
Heart Valve Dis. 1999, 8, 350–358. 23. McIntosh, D.; Ross,
D.; Champeil, P.; Guillain, F. Crosslinking the active site
of sarcoplasmic reticulum Ca(2+)-ATPase completely blocks
Ca2+ release to the vesicle lumen. Proc. Natl. Acad. Sci.
U. S. A. 1991, 88, 6437–6441. 24. Ross, D.; Davidson, G.;
McIntosh, D. Mechanism of inhibition of sarcoplasmic
reticulum Ca2(+)-ATPase by active site cross-linking.
Impairment of nucleotide binding slows nucleotide-dependent
phosphoryl transfer, and loss of active site flexibility
stabilizes occluded forms and blocks E2-P formation. J.
Biol. Chem. 1991, 266, 4613–4621. 25. Coan, C.; Jakobs, P.;
Ji, J.; Murphy, A. Sarcoplasmic reticulum calcium ATPase.
Labeling of a putative Mg2+ site by reaction with a
carbodiimide and a spinlabel. FEBS Lett. 1993, 335, 33–36.
26. Cheung, W. Calmodulin. Sci. Am. 1982, 246, 62–70. 27.
Mornet, E.; Stura, E.; Lia, B.A.; Stigbrand, T.; Menez, A.;
Le, D.M. Structural evidence for a functional role of human
tissue nonspecific alkaline phosphatase in bone
mineralization. J. Biol. Chem. 2001, 276, 31171–31178. 28.
Human, P.; Zilla, P. Inflammatory and immune processes: The
neglected villain of bioprosthetic degeneration? J.
Long-Term Eff. Med. Implants 2001, 11. 29. Dahm, M.;
Husmann, M.; Eckhard, M.; Prufer, D.; Groh, E.; Oelert, H.
Relevance of immunologic reactions for tissue failure of
bioprosthetic heart valves. Ann. Thorac. Surg. 1995, 60,
S348–S352. 30. Dahm, M.; Lyman, W.; Schwell, A.; Factor,
S.; Frater, R. Immunogenicity of glutaraldehyde-tanned
bovine pericardium. J. Thorac. Cardiovasc. Surg. 1990, 99,
1082–1090. 31. Vincentelli, A.; Latremouille, C.; Zegdi,
R.; Shen, M.; Lajos, P.; Chachques, J.; Fabiani, J. Does
glutaraldehyde induce calcification of bioprosthetic
tissues? Ann. Thorac. Surg. 1998, 66, S255–S258. 32.
Carpentier, A.; Lemaigre, G.; Robert, L.; Carpentier, S.;
Dubost, C. Biological factors affecting long-term results
of valvular heterografts. J. Thorac. Cardiovasc. Surg.
1969, 58, 467–483. 33. Nimni, M. The cross-linking and
structure modification of the collagen matrix in the design
of cardiovascular prosthesis. J. Card. Surg. 1988, 3,
523–533. 34. Sheikh, K.; Tascon, M.; Nimni, M. Autoimmunity
in patients with Hancock valve implant and bypass heart
surgery. Fed. Proc. Am. Soc. Exp. Biol. 1980, 39, 472. 35.
Smith, J.; Hornick, P.; Rasmi, N.; Rose, M.; Yacoub, M.
Effect of HLA mismatching and antibody status on
‘‘homovital’’ aortic valve homograft performance. Ann.
Thorac. Surg. 1998, 66, S212–S215. 36. Wewers, M.; Marsh,
C. Role of the antibody in the pathogenesis of transplant
vascular sclerosis: A hypothesis. Transpl. Immunol. 1997,
5, 283–288. 37. Jenney, C.; Anderson, J. Adsorbed serum
proteins responsible for surface dependent human macrophage
behavior. J. Biomed. Mater. Res. 2000, 49, 435–447. 38.
Nimni, M.; Cheung, D.; Strates, B.; Kodama, M.; Sheikh, K.
Chemically modified collagen: A natural biomaterial for
tissue replacement. J. Biomed. Mater. Res. 1987, 21,
741–771. 39. Werkmeister, J.; Ramshaw, J.; Ellender, G.
Characterisation of a monoclonal antibody against native
human type I collagen. Eur. J. Biochem. 1990, 187, 439–443.
40. Werkmeister, J.; Ramshaw, J. Multiple antigenic
determinants on type III collagen. Biochem. J. 1991, 274
(Pt. 3), 895–898. 41. Gibson, M.; Hughes, J.; Fanning, J.;
Cleary, E. The major antigen of elastin-associated
microfibrils is a 31-kDa glycoprotein. J. Biol. Chem. 1986,
261, 11429– 11436. 42. Kadner, A.; Chen, R.; Mitchell, R.;
Adams, D. Lack of ABH-antigen expression on human cardiac
valves. J. Heart Valve Dis. 2000, 9, 512–516. 43. Green,
M.; Walsh, M.; Dare, A.; Hogan, P.; Zhao, X.; Frazer, I.;
Bansal, A.; O’Brien, M. Histologic and immunohistochemical
responses after aortic valve allografts in the rat. Ann.
Thorac. Surg. 1998, 66, S216–S220. 44. Melina, G.; Rubens,
M.; Birks, E.; Bizzarri, F.; Khaghani, A.; Yacoub, M. A
quantitative study of calcium deposition in the aortic wall
following Medtronic Freestyle compared with homograft
aortic root replacement. A prospective randomized trial. J.
Heart Valve Dis. 2000, 9, 97–103. 45. Dignan, R.; O’Brien,
M.; Hogan, P.; Passage, J.; Stephens, F.; Thornton, A.;
Harrocks, S. Influence of HLA matching and associated
factors on aortic valve homograft function. J. Heart Valve
Dis. 2000, 9, 504–511. 46. Zilla, P.; Weissenstein, C.;
Bracher, M.; Zhang, Y.; Koen, W.; Human, P.; von Oppell, U.
High glutaraldehyde concentrations reduce rather than
increase the calcification of aortic wall tissue. J. Heart
Valve Dis. 1997, 6, 502–509. 47. Zilla, P.; Weissenstein,
C.; Human, P.; Dower, T.; von Oppell, U. High
glutaraldehyde concentrations mitigate bioprosthetic root
calcification in the sheep model. Ann. Thorac. Surg. 2000,
70, 2091–2095. 48. Human, P.; Zilla, P. Characterization of
the immune response to valve bioprostheses and its role in
primary tissue failure. Ann. Thorac. Surg. 2001, 71,
S385–S388. 49. Human, P.; Zilla, P. Graft-specific antibody
implicated in calcification of valve bioprosthetic tissue.
J. Cardiovasc. Pathol. 2000, 9, 205. 50. Levy, R.; Schoen,
F.; Howard, S. Mechanism of calcification of porcine
bioprosthetic aortic valve cusps: Role of T-lymphocytes.
Am. J. Cardiol. 1983, 52, 629–631. 51. Levy, R.; Schoen,
F.; Levy, J.; Nelson, A.; Howard, S.; Oshry, L. Biologic
determinants of dystrophic calcification and osteocalcin
deposition in glutaraldehyde-preserved porcine aortic valve
leaflets implanted subcutaneously in rats. Am. J. Pathol.
1983, 113, 143–155. 52. Eishi, K.; Ishibashi, U.H.; Nakano,
K.; Kosakai, Y.; Sasako, Y.; Kobayashi, J.; Yutani, C.
Calcific degeneration of bioprosthetic aortic valves in
patients receiving steroid therapy. J. Heart Valve Dis.
1996, 5, 668–672. 53. Chauvaud, S.; Jebara, V.; Chachques,
J.; el Asmar, B.; Mihaileanu, S.; Perier, P.; Dreyfus, G.;
Relland, J.; Couetil, J.; Carpentier, A. Valve extension
with glutaraldehyde-preserved autologous pericardium.
Results in mitral valve repair. J. Thorac. Cardiovasc.
Surg. 1991, 102, 171–177, discussion 177–178. 54. Nimni,
M.; Myers, D.; Ertl, D.; Han, B. Factors which affect the
calcification of tissue-derived bioprostheses. J. Biomed.
Mater. Res. 1997, 35, 531–537. 55. Iyengar, A.; Sacks, M.
Dynamic Imaging of BHV Cuspal Motion Using Laser
Projection. The First Joint BMES/EMBS Conference: IEEE,
Atlanta, USA, 1999. 56. Mayne, A.; Christie, G.; Smaill,
B.; Hunter, P.; Barratt, B.B. An assessment of the
mechanical properties of leaflets from four
second-generation porcine bioprostheses with biaxial
testing techniques [see comments]. J. Thorac. Cardiovasc.
Surg. 1989, 98, 170–180. H

57. Christie, G.; Barratt-Boyes, B. Age-dependent changes


in the radial stretch of human aortic valve leaflets
determined by biaxial testing. Ann. Thorac. Surg. 1995, 60,
S156–S159.

58. Billiar, K.; Sacks, M. Biaxial mechanical properties of


the natural and glutaraldehyde treated aortic valve
cusp—Part I: Experimental results. J. Biomech. Eng. 2000,
122, 23–30.

59. Vesely, I. The influence of design on bioprosthetic


valve durability. J. Long-Term Eff. Med. Implants 2001, 11
(3&4), 137–149. 60. Walley, V.; Keon, W. Patterns of
failure in IonescuShiley bovine pericardial bioprosthetic
valves. J. Thorac. Cardiovasc. Surg. 1987, 9393, 925–933.
61. Liotta, D.; Bracco, D.; Ferrari, H.; Bertolozzi, E.;
Pisanu, A. Low profile bioprosthesis for cardiac valve
replacement: Early clinical results. Card. Dis. Bull. Tex.
Heart Inst. 1977, 4, 371–382. 62. Ius, P.; Thiene, G.;
Minarini, M.; Valente, M.; Bortolotti, U.; Talenti, E.;
Carsarotto, D. Low-profile porcine bioprosthesis (Liotta):
Pathologic findings and mode of failure in the long-term. J.
Heart Valve Dis. 1996, 5, 323–327.
Heart Valve Mechanics

33. Alam, M.; Rosenhamer, G. Atrioventricular plane


displacement and left ventricular function. J. Am. Soc.
Echocardiogr. 1992, 5, 427–433.

34. Kalmanson, D. The Mitral Valve: A Pluridisciplinary


Approach; Publishing Science Group: Acton, MA, 1976;
[Chapters 1–5], 3–45.

35. He, S.; Lemmon, J.D.; Weston, M.W.; Levine, R.A.;


Yoganathan, A.P. Mitral valve compensation for annular
dilation: In vitro study into the mechanisms of functional
mitral regurgitation with an adjustable annulus model. J.
Heart Valve Dis. 1999, 8, 294–302.

36. Silver, M.D.; Lam, J.H.C.; Raganathan, N.; Wigle, E.D.


Morphology of the human tricuspid valve. Circulation 1971,
43, 333–348.

37. Chester, A.H.; Misfeld, M.; Yacoub, M.H.


Receptormediated contraction of aortic valve leaflets. J.
Heart Valve Dis. 2000, 9, 250–255.

38. Ritchie, J.; Jimenez, J.H.; He, Z.; Sacks, M.S.;


Yoganathan, A.P. The material properties of the native
porcine mitral valve chordae tendineae: An in vitro
investigation. J. Biomech. 2006, 39 (6), 1129–1135.

39. Ritchie, J.; Warnock, J.N.; Yoganathan, A.P. Structural


characterization of the chordae tendineae in native porcine
mitral valves. Ann. Thorac. Surg. 2005, 80 (1), 189–197.

40. Kunzelman, K.S.; Cochran, R.P.; Verner, E.D.; Eberhart,


R.D. Anatomic basis for mitral valve modeling. J. Heart
Valve Dis. 1994, 3, 491–496.

41. Sedransk, K.L.; Allen, J.G.; Vesely, I. Failure


mechanics of mitral valve chordae tendineae. J. Heart Valve
Dis. 2002, 11, 644–650.

42. Liao, J.; Vesely, I. A structural basis for the


size-related mechanical properties of mitral valve chordae
tendineea. J. Biomech. 2003, 36 (8), 1125–1133.

43. Rusted, I.E.; Schiefley, C.H.; Edwards, J.E. Studies of


the mitral valve: I. Anatomical features of the normal
mitral valve and associated structures. Circulation 1952, 6
(6), 825–831.
44. Madu, E.C.; Baugh, D.S.; D’Cruz, I.A.; Johns, C. Left
ventricular papillary muscle morphology and function in
left ventricular hypertrophy and left ventricular
dysfunction. Med. Sci. Monit. 2001, 7 (6), 1212–1218.

45. Weyman, A.E. In Principles and Practices of


Echocardiography; Lea & Febiger: Philadelphia, PA, 1994.

46. Pye, M.P.; Pringle, S.D.; Cobbe, S.M. Reference values


and reproducibility of Doppler echocardiography in the
assessment of the tricuspid valve and right ventricular
diastolic function in normal subjects. Am. J. Cardiol.
1991, 67 (4), 269–273.

47. Kim, W.Y.; Walker, P.G.; Pederson, E.M.; Poulsen, J.K.;


Houlind, K.C.; Oyre, S. Left ventricular blood flow patterns
in normal subjects: A quantitative analysis of
three-dimensional magnetic resonance velocity mapping. J.
Am. Coll. Cardiol. 1995, 26 (1), 224–238.

48. Bellhouse, B.J. The fluid mechanics of a model mitral


valve and left ventricle. Cardiovasc. Res. 1972, 6,
199–210.

49. Reul, H.; Talukder, N.; Muller, E.W. Fluid mechanics of


the natural mitral valve. J. Biomech. 1981, 14 (5),
361–372. 50. Yellin, E.L.; Peskin, C.; Yoran, C.; et al.
Mechanisms of mitral valve motion during diastole. Am. J.
Physiol. 1981, 241, H389–H400. 51. Arts, T.; Meerbaum, S.;
Reneman, R.; Corday, E. Stresses in the closed mitral
valve: A model study. J. Biomech. 1983, 16, 539–547. 52.
Salgo, I.S.; Gorman, J.H., III; Gorman, R.C.; et al. Effect
of annular shape on leaflet curvature in reducing mitral
leaflet stress. Circulation 2002, 106, 711–717. 53.
Einstein, D.R.; Kunzelman, K.S.; Reinhall, P.G.; Nicosia,
M.A.; Cochran, R.P. Non-linear fluid-coupled computational
model of the mitral valve. J. Heart Valve Dis. 2005, 14
(3), 376–385. 54. Sacks, M.S.; He, Z.; Baijens, L.; et al.
Surface strains in the anterior leaflet of the functioning
mitral valve. Ann. Biomed. Eng. 2002, 30, 1281–1290. 55.
He, Z.; Sacks, M.S.; Baijens, L.; Wanant, S.; Shah, P.;
Yoganathan, A.P. Effects of papillary muscle position on
the in vitro dynamic strain on the porcine mitral valve. J.
Heart Valve Dis. 2003, 12, 488–494. 56. He, Z.; Ritchie,
J.; Grashow, J.S.; Sacks, M.S.; Yoganathan, A.P. In vitro
dynamic strain behavior of the mitral valve posterior
leaflet. J. Biomech. Eng. 2005, 127 (3), 504–511. 57.
Grashow, J.S.; Yoganathan, A.P.; Sacks, M.S. Biaxial
stress-stretch behavior of the mitral valve anterior leaflet
at physiologic strain rates. Ann. Biomed. Eng. 2006, 34
(2), 315–325. 58. Nazari, S.; Carli, F.; Bnfi, C.; et al.
Patterns of systolic stress distribution on mitral valve
anterior leaflet chordal apparatus. J. Cardiovasc. Surg.
2000, 41, 193–202. 59. He, S.; Weston, M.W.; Lemmon, J.;
Jensen, M.; Levine, R.A.; Yoganathan, A.P. Geometric
distribution of chordae tendineae: An important anatomic
feature in mitral valve function. J. Heart Valve Dis. 2000,
9, 495–501. 60. Lomholt, M.; Nielsen, S.L.; Hansen, S.B.;
Andersen, N.T.; Hasenkam, J.M. Differential tension between
secondary and primary mitral chordae in acute in-vivo
porcine model. J. Heart Valve Dis. 2002, 11, 337–345. 61.
Jimenez, J.H.; Soerensen, D.D.; He, Z.; Ritchie, J.;
Yoganathan, A.P. Effects of a papillary muscle position on
chordal force distribution: An in vitro study. J. Heart
Valve Dis. 2005, 14, 295–302. 62. Jimenez, J.H.; Sorensen,
D.D.; He, Z.; He, S.; Yoganathan, A.P. Effects of a saddle
shaped annulus on mitral valve function and papillary
muscle position. Ann. Biomed. Eng. 2003, 31, 1171–1181. 63.
Lim, K.O.; Bouchner, D.P. Mechanical properties of human
mitral valve chordae tendineae: Variation with size and
strain rate. Can. J. Physiol. Pharmacol. 1975, 53, 330–339.
64. Jensen, M.O. In Stentles Mitral Valve Fixation: Impact
on Hemodynamic Performance [Master’s Thesis, Department of
Biomedical Engineering]; Georgia Institute of Technology:
Atlanta, GA, 2000. 65. Pinto, J.G.; Fung, Y.C. Mechanical
properties of the heart muscle in the passive state. J.
Biomech. 1973, 6, 596–616. 66. Fung, Y.C. In Biomechanics:
Mechanical Properties of Living Tissues; Springer: New
York, 1981.
Heart Valves, Mechanical

2. Starr, A.; Fessler, C.L.; Grunkemeier, G.; He, G.W.


Heart valve replacement surgery: Past, present and future.
Clin. Exp. Pharmacol. Physiol. 2002, 29 (8), 735–738.

3. Ely, J.L.; Emken, M.R.; Accuntius, J.A.; Wilde, D.S.;


Haubold, A.D.; More, R.B.; Bokros, J.C. Pure pyrolytic
carbon: Preparation and properties of a new material, On-X
carbon for mechanical heart valve prostheses. J. Heart
Valve Dis. 1998, 7 (6), 626–632.

4. Bokros, J.C.; Haubold, A.D.; Akins, R.J.; Campbell,


L.A.; Griffin, C.D.; Lane, E. The Durability of Mechanical
Heart Valve Replacements: Past Experience and Current
Trends. In Replacement Cardiac Valves; Bodnar, E., Frater,
R.W.M., Eds.; Pergamon Press Inc.: New York, 1991; 21–48.

5. Hasenkam, J.M.; Pasquino, E.; Stacchino, C.; Arru, P.;


Vallana, F.; Paulsen, P.K. Wear patterns in the Sorin
Bicarbon mechanical heart valve: A clinical explant study.
J. Heart Valve Dis. 1997, 6 (2), 105–114.

6. Ritchie, R.O.; Dauskart, R.H.; Yu, W. Cyclic


fatiguecrack propagation, stress-corrosion, and
fracturetoughness in pyrolytic carbon-coated graphite for
prosthetic heart valve applications. J. Biomed. Mater. Res.
1990, 24, 189.

7. Knapp, R.J.; Daily, J.W.; Hammitt, F.G. Cavitation;


McGraw Hill Book Co.: New York, 1970; 1–2.

8. He, Z.M.; Xi, B.S.; Zhu, K.Q.; Hwang, N.H.C. Mechanism


of mechanical heart valve cavitation: Investigation using
tilting disk valve model. J. Heart Valve Dis. 2001, 10,
666–674.

9. Yoganathan, A.P.; Woo, Y.R.; Sung, H.W.; Jones, M.


Advances in prosthetic heart valves: Fluid mechanics of
aortic valve designs. J. Biomater. Appl. 1988, 2 (4),
579–614.

10. Yoganathan, A.P.; Sung, H.W.; Woo, Y.R.; Jones, M. In


vitro velocity and turbulence measurements in the vicinity
of three new mechanical aortic heart valve prostheses:
Bjork-Shiley monostrut, omni-carbon, and duromedics. J.
Thorac. Cardiovasc. Surg. 1988, 95 (5), 929–939.

11. Kini, V.; Bachmann, C.; Fontaine, A.; Deutsch, S.;


Tarbell, J.M. Integrating particle image velocimetry and
laser Doppler velocimetry measurements of the regurgitant
flow field past mechanical heart valves. Artif. Organs 2001,
25 (2), 136–145.

12. Meyer, R.S.; Deutsch, S.; Bachmann, C.B.; Tarbell, J.M.


Laser Doppler velocimetry and flow visualization studies in
the regurgitant leakage flow region of three mechanical
mitral valves. Artif. Organs 2001, 25 (4), 292–299.

13. Woo, Y.R.; Yoganathan, A.P. Pulsatile flow velocity and


shear stress measurements on the St. Jude bileaflet valve
prosthesis. Scand. J. Thorac. Cardiovasc. Surg. 1986, 20
(1), 15–28.

14. Leo, H.L.; He, Z.; Ellis, J.T.; Yoganathan, A.P.


Microflow fields in the hinge region of the CarboMedics
bileaflet mechanical heart valve design. J. Thorac.
Cardiovasc. Surg. 2002, 124 (3), 561–574. 15. Ellis, J.T.;
Travis, B.R.; Yoganathan, A.P. An in vitro study of the
hinge and near-field forward flow dynamics of the St. Jude
Medical Regent bileaflet mechanical heart valve. Ann.
Biomed. Eng. 2000 , 28 (5), 524–532. 16. Yoganathan, A.P.;
Wick, T.M.; Reul, H. Influence of Flow Characteristics of
Prosthetic Valves on Thrombus Formation. In Thrombosis,
Embolism and Bleeding; Butchart, E.G., Bodnar, E., Eds.;
ICR Publishers: United Kingdom, 1992; 123–148. 17. Hoffman,
M.; Monroe, D.M., III. A cell-based model of hemostasis.
Thromb. Haemost. 2001, 85, 958–965. 18. Ruggeri, Z.M.
Mechanisms of shear-induced platelet adhesion and
aggregation. Thromb. Haemost. 1993, 70 (1), 119–123. 19.
DeLa Cadena, R.A.; Wachtfogel, Y.T.; Colman, R.W. Contact
Activation Pathway: Inflammation and Coagulation. In
Hemostasis and Thrombosis: Basic Principles and Clinical
Practice, 3rd Ed.; Colman, R.W., Hirsh, J., Marder, V.J.,
Salzman, E.W., Eds.; J. B. Lippincott Company:
Philadelphia, 1994; 219–240. 20. Butchart, E.G.
Thrombogenicity, Thrombosis and Embolism. In Thrombosis,
Embolism and Bleeding; Butchart, E.G., Bodnar, E., Eds.;
ICR Publishers: United Kingdom, 1992; 172–205. 21. Goodman,
S.L. Sheep, pig, and human platelet–material interactions
with model cardiovascular biomaterials. J. Biomed. Mater.
Res. 1999, 45, 240–250. 22. Lamson, T.C.; Rosenberg, G.;
Geselowitz, D.B.; Deutsch, S.; Stinebring, D.R.; Frangos,
J.A.; Tarbell, J.M. Relative blood damage in the three
phases of a prosthetic heart valve flow cycle. ASAIO J.
1993, 39, M626–M633. 23. Travis, B.R. The Effects of
Bileaflet Prosthesis Pivot Geometry on Turbulence and Blood
Damage Potential. In Ph.D. Thesis; Georgia Institute of
Technology, 2001. 24. O’Brien, J.R. Shear-induced platelet
aggregation. Lancet 1990, 335, 711–713. 25. Schoephoerster,
R.T.; Oynes, F.; Nunez, G.; Kapadvanjwala, M.; Dewanjee,
M.K. Effects of local geometry and fluid dynamics on
regional platelet deposition on artificial surfaces.
Arterioscler. Thromb. 1993, 13, 1806–1813. 26. Yoganathan,
A.P.; Travis, B.R. Fluid Dynamics of Prosthetic Valves. In
The Practice of Clinical Echocardiography, 2nd Ed.; Otto,
C.M., Ed.; , 2000. 27. Chang, B.-C.; Lim, S.H.; Kim, D.K.;
Seo, J.Y.; Cho, S.Y.; Shim, W.H.; Chung, N.; Kim, S.S.;
Cho, B.K. Long-term results with St. Jude medical and
carbomedics prosthetic heart valves. J. Heart Valve Dis.
2001, 10, 185–195. 28. Grunkemeier, G.L.; Li, H.-H.;
Naftel, D.C.; Starr, A.; Rahimtoola, S.H. Long term
performance of heart valve prostheses. Curr. Probl.
Cardiol. 2000, 25 (2), 73–156. H
Hematopoietic Stem Cells and Assays

1. Quesenberry, P.; Colvin, G.A. Hematopoietic Stem Cells,


Progenitor Cells, and Cytokines. In Hematology; Beutler,
E., et al., Eds.; McGraw-Hill: New York, 2001; 153–174.

2. Madlambayan, G.J.; Rogers, I.; Casper, R.F.; Zandstra,


P.W. Controlling culture dynamics for the expansion of
hematopoietic stem cells. J. Hematother. Stem Cell Res.
2001, 10, 481–492.

3. Krause, D.S.; Fackler, M.J.; Civin, C.I.; May, W.S.


CD34: Structure, biology, and clinical utility. Blood 1996,
87, 1–13.

4. Shih, C.-C.; DiGiusto, D.; Forman, S.J. Ex vivo


expansion of transplantable human hematopoietic stem cells:
Where do we stand in the year 2000? J. Hematothe. Stem Cell
Res. 2002, 9, 621–628.

5. Sutherland, H.J.; Lansdorp, P.M.; Henkelman, D.H.;


Eaves, A.C.; Eaves, C.J. Functional characterization of
individual human hematopoietic stem cells cultured at
limiting dilution on supportive marrow stromal layers.
Proc. Natl. Acad. Sci. U.S.A. 1990, 87, 3584–3588.

6. Craig, W.; Kay, R.; Cutler, R.L.; Lansdorp, P.M.


Expression of Thy-1 on human hematopoietic progenitor
cells. J. Exp. Med. 1993, 177, 1331–1342. 7. Osawa, M.;
Hanada, K.; Hamada, H.; Nakauchi, H. Long-term
lymphohematopoietic reconstitution by CD34-low/negative
hematopoietic stem cells. Science 1996, 273, 242–245. 8.
Sato, T.; Laver, J.H.; Ogawa, M. Reversible expression of
CD34 by murine hematopoietic stem cells. Blood 1999, 94
(8), 2548–2554. 9. Matsuoka, S.; Ebihara, Y.; Xu, M.-J.;
Ishii, T.; Sugiyama, D.; Yoshino, H.; Ueda, T.; Manabe, A.;
Tanaka, R.; Ikeda, Y.; Nakahata, T.; Tsuji, K. CD34
expression on long-term repopulating hematopoietic stem
cells changes during developmental stages. Blood 2001, 97
(2), 419–425. 10. Goodell, M.; Brose, K.; Paradis, G.;
Conner, A.; Mulligan, A. Isolation and functional
properties of murine hematopoietic stem cells that are
replicating in vivo. J. Exp. Med. 1996, 183, 1797–1806. 11.
Spangrude, G.J.; Johnson, G.R. Resting and activated
subsets of mouse multipotent hematopoietic stem cells.
Proc. Natl. Acad. Sci. U.S.A. 1990, 87, 7433–7477. 12.
Zandstra, P.W.; Conneally, E.; Petzer, A.L.; Piret, J.M.;
Eaves, C.J. Cytokine manipulation of primitive human
hematopoietic cell self-renewal. Proc. Natl. Acad. Sci. U.
S. A. 1997, 94, 4698–4703. 13. Chen, C.-Z.; Li, M.; de
Graaf, D.; Monti, S.; Gottgens, B.; Sanchez, M.-J.; Lander,
E.S.; Golub, T.R.; Green, A.R.; Lodish, H.F. Identification
of endoglin as a functional marker that defines long-term
repopulating hematopoietic stem cells. Proc. Natl. Acad.
Sci. U.S.A. 2002, 99 (24), 15468–15473. 14. Fallon, P.;
Gentry, T.; Balber, A.E.; Boulware, D.; Janssen, W.E.;
Smilee, R.; Storms, R.W.; Smith, C. Mobilized peripheral
blood SSC lo ALDH br cells have phenotypic and functional
properties of primitive haematopoietic cells and their
number correlates with engraftment following autologous
transplantation. Br. J. Haematol. 2003, 122, 99–108. 15.
Williams, D.E.; Hangoc, G.; Cooper, S.; Boswell, H.S.;
Shadduck, R.K.; Gillis, S.; Waheed, A.; Urdal, D.;
Broxmeyer, H.E. The effects of purified recombinant murine
interleukin-3 and/or purified natural murine CSF-1 in vivo
on the proliferation of murine highand low-proliferative
potential colony-forming cells: Demonstration of in vivo
synergism. Blood 1987, 70, 401–403. 16. Eaves, C.J.; Eaves,
A.C. Stem cell kinetics. Bailliere’s Clin. Haematol. 1997,
10, 233–257. 17. Verfaillie, C.M. Ex Vivo Expansion of
Hematopoietic Stem Cells. In Hematopoiesis: A Developmental
Approach; Zon, L.I., Ed.; Oxford University Press: New
York, 2001; 119–129. 18. Till, J.E.; McCulloch, E.A. A
direct measurement of the radiation sensitivity of normal
mouse bone marrow cells. Radiat. Res. 1961, 14, 213–222.
19. McCune, J.M.; Namikawa, R.; Kaneshima, H.; Shultz,
L.D.; Lieberman, M.; Weissman, I.L. The SCID-hu mouse:
Murine model for the analysis of human hematolymphoid
differentiation and function. Science 1998, 24, 1632–1639.
20. Larochelle, A.; Vormoor, J.; Hanenberg, H.; Wang, J.C.;
Bhatia, M.; Lapidot, T.; Moritz, T.; Murdoch, B.; Xiao,
X.L.; Kato, I.; Williams, D.A.; Dick, J.E. Identification of
primitive human hematopoietic cells capable of repopulating
NOD/SCID mouse bone marrow: Implications for gene therapy.
Nat. Med. 1996, 2, 1329–1337. 21. Civin, C.I.;
Almeida-Porada, G.; Lee, M.J.; Olweus, J.; Terstappen,
L.W.; Zanjani, E.D. Sustained, retransplantable,
multilineage engraftment of highly purified adult human bone
marrow stem cells in vivo. Blood 1996, 88, 4102–4109. 22.
Orlic, D.; Kajstura, J.; Chimenti, S.; Jakoniuk, I.;
Anderson, S.M.; Li, B.; Pickel, J.; McKay, R.; NadalGinard,
B.; Bodine, D.M.; Leri, A.; Anversa, P. Bone marrow cells
regenerate infracted myocardium. Nature 2001, 410, 701–705.
23. Wang, X.; Ge, S.; McNamara, G.; Hao, Q.-L.; Crooks,
G.M.; Nolta, J.A. Albumin-expressing hepatocytelike cells
develop in the livers of immune-deficient mice that received
transplants of highly purified human hematopoietic stem
cells. Blood 2003, 101, 4201–4208. 24. Eglitis, M.A.;
Mezey, E. Hematopoietic cells differentiate into both
microglia in the brains of adult mice. Proc. Natl. Acad.
Sci. U.S.A. 1997, 94, 4080–4085. 25. Majka, S.M.; Jackson,
K.A.; Kienstra, K.A.; Majesky, M.W.; Goodell, M.A.;
Hirschi, K.K. Distinct progenitor populations in skeletal
muscle are bone marrow derived and exhibit different cell
fates during vascular regeneration. J. Clin. Invest. 2003,
111, 71–79. 26. Korbling, M.; Katz, R.L.; Khanna, A.;
Ruifrok, A.C.; Rondon, G.; Albitar, M.; Champlin, R.E.;
Estrov, Z. Hepatocytes and epithelial cells of donor origin
in recipients of peripheral-blood stem cells. N. Engl. J.
Med. 2002, 346 (10), 738–746. 27. Wagers, A.J.; Sherwook,
R.I.; Christensen, J.L.; Weissman, I. Little evidence for
developmental plasticity of adult hematopoietic stem cells.
Science 2002, 297, 2256–2259. 28. Terada, N.; Hamazaki, T.;
Oka, M.; Hoki, M.; Mastalerz, D.M.; Nakno, Y.; Meyer, E.M.;
Morel, L.; Petersen, B.E.; Scott, E.W. Bone marrow cells
adopt the phenotype of other cells by spontaneous cell
fusion. Nature 2002, 416, 542–545. 29. Ying, Q.-L.;
Nichols, J.; Evans, E.P.; Smith, A.G. Changing potency by
spontaneous fusion. Nature 2002, 416, 545–548. 30.
Quesenberry, P.; Colvin, G.A.; Lambert, J.-F. The
chiaroscuro stem cell: A unified stem cell theory. Blood
2002, 100 (13), 4266–4271. 31. Cairns, J. Somatic stem
cells and the kinetics of mutagenesis and carcinogenesis.
Proc. Natl. Acad. Sci. U.S.A. 2002, 99 (16), 10567–10570.
32. Corbel, S.Y.; Lee, A.; Yi, L.; Duenas, J.; Brazelton,
T.R.; Blau, H.M.; Rossi, F.M.V. Contribution of
hematopoietic stem cells to skeletal muscle. Nature Med.
2003, 9 (12), 1528–1532. 33. Collins, P.; Miller, W.M.;
Papoutsakis, E.T. Ex vivo culture systems for hematopoietic
cells. Curr. Opin. Biotechnol. 1996, 7 (2), 223–230. 34.
Wang, X.; Willenbring, H.; Akkari, Y.; Torimaru, Y.;
Foster, M.; Al-Dhalimy, M.; Lagasse, E.; Finegold, M.;
Olson, S.; Grompe, M. Cell fusion is the peripheral source
of bone-marrow-derived hepatocytes. Nature 2003, 422,
897–901. 35. Vassilopoulos, G.; Wang, P.-R.; Russel, D.W.
Transplanted bone marrow regenerates liver by cell fusion.
Nature 2003, 422, 901–904. 36. Nielsen, L.K. Bioreactors
for hematopoietic cell culture. Ann. Rev. Biomed. Eng.
1999, 1, 129–152. 37. McAdams, T.; Sandstrom, C.E.; Miller,
W.M.; Bender, J.G.; Paputsakis, E.T. Ex vivo expansion of
primitive hematopoietic cells for cellular therapies: An
overview. Cytotechnology 1995, 18, 133–146. 38. McAdams,
T.A.; Winter, J.N.; Miller, W.M.; Papoutsakis, E.T.
Hematopoietic cell culture therapies (Part II): Clinical
aspects and applications. Trends Biotech. 1996, 14 (10),
388–396. 39. Cairo, M.S.; Wagner, J.E. Placental and/or
umbilical cord blood: An alternative source of
hematopoietic stem cells for transplantation. Blood 1997,
90, 4664–4671. 40. De La Selle, V.; Gluckman, E.;
Bruley-Rosset, M. Newborn blood can engraft adult mice
without inducing graft versus host disease across non H-2
antigens. Blood 1996, 87, 3977–3984. 41. Sorg, R.V.;
Kogler, G.; Wernet, P. Identification of cord blood
dendrtitic cells an an immature CD11cpopulation. Blood
1999, 93 (7), 2302–2307. 42. Grewal, S.S.; Barker, J.N.;
Davies, S.M.; Wagner, J.E. Unrelated donor hematopoietic
cell transplantation: Marrow or umbilical cord? Blood 2003,
101 (11), 4233– 4244. 43. McNiece, I.; Kubegov, D.; Kerzic,
P.; Shpall, E.; Gross, S. Increased expansion and
differentiation of cord blood products using a two-step
expansion culture. Exp. Hematol. 2000, 28, 1181–1186. 44.
McNiece, I.; Briddell, R. Ex vivo expansion of
hematopoietic progenitor cells and mature cells. Exp.
Hematol. 2001, 29, 3–11. 45. Jaroscak, J.; Goltry, K.;
Smith, A.; Waters-Pick, B.; Martin, P.L.; Driscoll, T.A.;
Howrey, R.; Chao, N.; Douville, J.; Burhop, S.; Fu, P.;
Kurtzberg, J. Augmentation of umbilical cord (UCB)
transplantation with ex vivo-expanded UCB cells: Results of
a phase 1 trial using the AastromReplicell System. Blood
2003, 101 (12), 5061–5067. 46. Willert, K.; Brown, J.D.;
Danenberg, E.; Duncan, A.W.; Weissman, I.; Reya, T.; Yates,
J.R., III; Nusse, R. Wnt proteins are lipid-modified and can
act as stem cell growth factors. Nature 2003, 423, 448–452.
47. Mayani, H.; Lansdrop, P.M. Biology of human umbilical
cord blood-derived hematopoietic stem/ progenitor cells.
Srem. Cells 1998, 16, 153–165. 48. Kim, D.; Fujiki, Y.;
Fukushima, T.; Ema, H.; Shibuya, A.; Nakauchi, H.
Comparison of hematopoietic activities of human bone marrow
and umbilical cord blood CD34 positive and negative cells.
Stem Cells 1999, 17, 286–294. 49. Koller, M.R.; Oxender,
M.; Jensen, T.C.; Goltry, K.L.; Smith, A.K. Direct contact
between CD34 + /Lin-cells and stroma induces a soluble
activity that specifically increases primitive hematopoietic
cell production. Exp. Hematol. 1999, 27, 734–741. H

50. Bachier, C.R.; Gokmen, E.; Teale, J.; Lanzkron, S.;


Childs, C.; Franklin, W.; Shpall, E.; Douville, J.; Weber,
S.; Muller, T.; Armostrong, D.; LeMaistre, C.F. Ex-vivo
expansion of bone marrow progenitor cells for hematopoietic
reconstitution following high-dose chemotherapy for breast
cancer. Exp. Hematol. 1999, 27, 615–623.

51. Sandstrom, C.E.; Bender, J.G.; Papoutsakis, E.T.;


Miller, W.M. Effects of CD34 + cell selection and perfusion
on ex vivo expansion of peripheral blood mononuclear cells.
Blood 1995, 86, 958–970.

52. Koller, M.R.; Manchel, I.; Newsom, B.S.; Palsson, M.A.;


Palsson, B.O. Bioreactor expansion of human bone marrow:
Comparison of unprocessed, densityseparated and
CD34-enriched cells. J. Hematother. 1995, 4, 159–169.

53. Simmons, P.J.; Gronthos, S.; Zanneting, A.C.W. The


Development of Stromal CellsIn Hematopoiesis: A
Developmental Approach; Zon, L.I., Ed.; Oxford University
Press: New York, 2001; 718–726.

54. Haynesworth, S.E.; Reuben, D.; Caplan, A.I. Cell-based


tissue engineering therapies: The influence of whole body
physiology. Adv. Drug Deliv. Rev. 1997, 33, 3–14.

55. Owen, M.E. The Marrow Stromal Cell System. In Marrow


Stromal Cell in Culture; Beresford, J.N., Owen, M.E., Eds.;
Cambridge University Press: Cambridge, UK, 1988; 88–110.

56. Caplan, A.I.; Bruder, S.P. Mesenchymal stem cells:


Building blocks for molecular medicine in the 21st century.
Trends Mol. Med. 2001, 7 (6), 259–264. 57. Gutierrez-Ramos,
J.; Reyes-Maldonado, E.; Mayani, H. Characterisation of the
adherent cells developed in Dexter-type long-term cultures
from human umbilical cord blood. Stem Cells 2000, 18,
46–52. 58. Huss, R.; Lange, C.; Weissinger, E.M.; Kolb,
H.J.; Thalmeier, K. Evidence of peripheral blood-derived,
plastic-adherent CD34 �/low hematopoietic stem cell clones
with mesenchymal stem cell characteristics. Stem Cells
2000, 18, 252–260. 59. Owen, M.E. Lineage of osteogenic
cells and their relationship to the stromal system. Bone
Mineralisation Res. 1985, 3, 1–25. 60. Friedenstein, A.J.;
Gorskaja, J.F.; Kulagina, N.N. Fibroblast precursors in
normal and irradiated mouse hematopoietic organs. Exp.
Hematol. 1976, 4, 267–274. 61. Simmons, P.J.; Torok-Storb,
B. Identification of stromal cell preceursors in human bone
marrow by a novel monoclonal antibody. STRO-1. Blood 1991,
78, 55–62. 62. Gronthos, S.; Simmons, P.J. The biology and
application of human bone marrow stromal cell precursors.
J. Hematother. 1996, 5, 15–23. 63. Onyia, J.E.; Clapp,
D.W.; Long, H.; Hock, J.M. Osteoprogenitor cells as targets
for ex vivo gene transfer. J. Bone Mineral. Res. 1998, 13,
2–30. 64. Young, R.G.; Butler, D.L.; Weber, W.; Caplan,
A.I.; Gordon, S.L.; Fink, D.J. Use of mesenchymal stem
cells in collagen matrix for Achilles tendon repair. J.
Orthop. Res. 1998, 16, 406–413.
Hemocompatible Materials

1. Sefton, M.V.; Gemmel, C.H. Nonthrombogenic treatments


and strategies. In Biomaterials Science. An Introduction to
Materials in Medicine, 2nd Ed.; Ratner, B.D., Hoffman,
A.S., Schoen, F.J., Lemons, J.E., Eds.; Elsevier Academic
Press: San Diego, 2004; 456–470.

2. Kim, K.; Kim, C.; Byun, Y. Preparation of a PEGgrafted


phospholipid Langmuir-Blodgett monolayer for
blood-compatible material. J. Biomed. Mater. Res. 2000, 52,
836–840.

3. Shen, M.; Pan, Y.V.; Wagner, M.S.; Hauch, K.D.; Castner,


D.G.; Ratner, B.D.; Horbett, T.A. Inhibition of monocyte
adhesion and fibrinogen adsorption on glow discharge plasma
deposited tetraethylene grlycol dimethyl ether. J. Biomat.
Sci. Polym. Ed. 2001, 12, 961–978.

4. Lewis, A.L.; Hughes, P.D.; Kirkwood, L.C.; Leppard,


S.W.; Redman, R.P.; Tolhurst, L.A.; Stratford, P.W.
Synthesis and characterization of phosphorylcholinebased
polymers useful for coating blood filtration devices.
Biomaterials 2000, 21, 1847–1859.

5. Gu, Y.J.; Boonstra, P.W.; Rijnsburger, A.A.; Haan, J.;


van Oeveren, W. Cardiopulmonary bypass circuit treated with
surface-modifying additives: a clinical evaluation of blood
compatibility. Ann. Thorac. Surg. 1998, 65, 1342–1347.

6. Sefton, M.V.; Sawyer, A.; Gorbet, M.; Black, J.P.;


Cheng, E.; Gemmel, C.; Pottinger-Cooper, E. Does surface
chemistry affect thrombogenicity of surface modified
polymers? J. Biomed. Mater. Res. 2001, 55, 447–459.

7. Barbucci, R.; Cialdi, G.; Magnani, A. Novel HeparinLike


Sulfated Polysaccharides. European Patent–PCT WO 95/25751;
Sulfated Hyaluronic Acid Esters. U.S. Patent–6051701, April
18, 2000.

8. Park, H.D.; Lee, W.K.; Ooya, T.; Park, K.D.; Kim, Y.H.;
Yui, N. Anticoagulant activity of sulfonated polyrotaxanes
as blood-compatible materials. J. Biomed. Mater. Res. 2002,
60, 186–190. 9. Magnani, A.; Lamponi, S.; Consumi, M.;
Barbucci, R. Biological performance of two materials based
on sulphated hyaluronic acid and polyurethane. J. Mater.
Chem. 1999, 9, 2393–2398. 10. Barbucci, R.; Magnani, A.;
Rappuoli, R.; Lamponi, S.; Consumi, M. Immobilization of
sulphated hyaluronan for improved biocompatibility. J.
Inorg. Biochem. 2000, 79 (1–4), 119–125. 11. Ratner, B.D.
Surface modification of polymers: chemical, biological and
surface analytical challenges. Biosens. Bioelectron. 1995,
10, 797–804. 12. Silver, J.H.; Lin, J.C.; Lim, F.;
Tegoulia, V.A.; Chaudhury, M.K.; Cooper, S.L. Surface
properties and hemocompatibility of alkyl-siloxane
monolayers supported on silicon rubber: effect of alkyl
chain length and ionic functionality. Biomaterials 1999,
20, 1533–1543. 13. a. Larm, O.; Larsson, R.; Olsson, P. A
new nonthrombogenic surface prepared by selective covalent
binding of heparin via a modified reducing terminal residue.
Biomat. Med. Dev. Art. Org. 1983, 11, 161–173; b. Ovrum,
E.; Tangen, G.; Oystese, R.; Ringdal, M.A.L.; Istad, R.
Comparison of two heparin-coated extracorporeal circuits
with reduced systemic anticoagulation in routine coronary
artery by pass operations. J. Thorac. Cardiovasc. Surg.
2001, 121, 324–330. 14. Wendel, H.P.; Ziemer, G.
Coating-techniques to improve the hemocompatibility of
artificial devices used for extracorporeal circulation. Eur.
J. Cardiothorac. Surg. 1999 , 16, 342–350. 15. Palatianos,
G.M.; Foroulis, C.N.; Vassili, M.I.; Astras, G.;
Triantafillou, K.; Papadakis, E.; Lidoriki, A.A.;
Iliopoulou, E.; Melissari, E.N. A prospective, doubleblind
study on the efficacy of the bioline surfaceheparinized
extracorporeal perfusion circuit. Ann. Thorac. Surg. 2003,
76, 129–135. 16. Han, H.S.; Yang, S.L.; Yeh, H.Y.; Lin,
J.C.; Wu, H.L.; Shi, G.Y. Study of a novel human
thrombomodulin immobilized substrate: surface
characterization and anticoagulation activity evaluation.
J. Biomater. Sci. Polym. Ed. 2001, 12, 1075–1089. 17. Duan,
X.; Lewis, R.S. Improved haemocompatibility of
cysteine-modified polymers via endogenus nitric oxide.
Biomaterials 2002, 23, 1197–1203. 18. a. Herring, M.B.;
Baughman, S.; Glover, J.; Kesler, K.; Jesseph, J.; Dillay,
R.; Evan, A.; Gardner, A. Endothelial seeding of Dacron and
polytetrafluoroethylene grafts: the cellular events of
healing. Surgery 1984, 96, 745–754; b. Zilla, P.; Detsch,
M.; Meinhart, J.; Endothelial cell transportation. Semin.
Vasc. Surg. 1999, 12, 52–63; c. Bos, G.W.; Scharenborg,
N.M.; Poot, A.A.; Engbers, G.H.; Beugeling, T.; van Aken,
W.G.; Feijen, J. Blood compatibility of surfaces with
immobilized albumin-heparin conjugate and effect of
endothelial cell seeding on platelet adhesion. J. Biomed.
Mater. Res. 1999, 47, 279–291; d. Sipehia, R. The enhanced
attachment and growth of endothelial cells on anhydrous
ammonia gaseous plasma modified surface of polystyrene and
poly (tetrafluoroethylene). Biomater. Art. Cells Art. Org.
1990, 18, 437–446; e. Van Wachen, P.B.; Vreciks, C.N.;
Bengeling, T.; Feijen, J.; Bautjes, A.; Detmers, J.P.; van
Aken, W.G. The influence of protein adsorption on
interactions of cultured human endothelial cells with
polymers. J. Biomed. Mater. Res. 1987, 21, 701–718; f.
Massia, S.P.; Hubbel, J.A. Covalently attached GRGD on
polymer surfaces promotes biospecific adhesion of mammalian
cells. Ann. N. Y. Acad. Sci. 1990, 589, 261–270. 19.
Morice, M.C.; Serruys, P.W.; Sousa, J.E.; Fajader, J.; Ban
Hayashi, E.; Perin, M.; Colombo, A.; Schuler, G.; Barragan,
P.; Guagliumi, G.; Molnar, F.; Falotico, R. RAVEL Study
Group. Randomized study with the sirolimus-coated Bx
velocity balloon-expandable stent in the treatment of
patients with de novo native coronary artery lesion. A
randomized comparison of a sirolimuseluting stent with a
standard stent for coronary revascularization. N. Engl. J.
Med. 2002, 346, 1773–1780. 20. Vickerman, J.C. Surface
Analysis. The Principal Techniques; John Wiley & Sons Ltd:
Baffins Lane, Chichester, England, 1997. 21. Ratner, B.D.
Surface properties and surface characterization of
materials. In Biomaterials Science. An Introduction to
Materials in Medicine, 2nd Ed.; Ratner, B.D., Hoffman,
A.S., Schoen, F.J., Lemons, J.E., Eds.; Elsevier Academic
Press: San Diego, 2004; 40–59. 22. Hauch, K.D. Microscopy
for Biomaterials Science. In Biomaterials Science. An
Introduction to Materials in Medicine, 2nd Ed.; Ratner,
B.D., Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.;
Elsevier Academic Press: San Diego, 2004; 396–411. 23.
Magnani, A.; Barbucci, R. Fourier transform attenuated
total reflection infrared spectroscopy (ATR/FTIR):
applications to proteins adsorption studies. In Test
Procedures for the Blood Compatibility of Biomaterials;
Dawids, S., Ed.; Kluwer Academic Publisher: The
Netherlands, 1993; 171–184. 24. Barbucci, R.; Magnani, A.
Conformation of human plasma proteins at polymer surfaces:
the effectiveness of surface heparinization. Biomaterials
1994, 15 (12), 955–962. 25. Belu, A.M.; Graham, D.J.;
Castner, D.G. Time-offlight secondary ion mass spectrometry:
techniques and applications for the characterization of
biomaterials surfaces. Biomaterials 2003, 24, 3635–3653.
26. Norde, W.; Lyklema, J. Why proteins prefer surfaces. J.
Biomat. Sci. Polym. Ed. 1991, 2, 183–202. 27. Gray, J.J.
The interaction of protein with solid surfaces. Curr. Opin.
Struct. Biol. 2004, 14, 110–115. 28. Magnani, A.; Peluso,
G.; Margarucci, S.; Chittur, K.K. Protein adsorption and
cellular/tissue interactions. In Integrated Biomaterials
Sciences; Barbucci, R., Ed.; Kluwer Academic/Plenum
Publishers: New York, 2002; 669–689. 29. Horbett, T.A.;
Brash, J.L. Proteins at interfaces: an overview. In
Proteins at Interfaces II: Fundamentals and Applications,
ACS Symposium Series 602; Horbett, T.A., Brash, J.L., Eds.;
American Chemical Society: Washington, DC, 1995, 1–25. 30.
Slack, S.M.; Horbett, T.A. The Vroman effect: a critical
review. In Proteins at Interfaces II: Fundamentals and
Applications; Horbett T.A., Brash, J.L., Eds.; ACS
Symposium Series 602; American Chemical Society:
Washington, DC, 1995, 112–128. 31. Harris, J.M.
Introduction to biotechnical and biomedical applications of
poly(ethylene glycol). In Poly(Ethylene Glycol) Chemistry:
Biotechnical and Biomedical Applications; Harris, J.M.,
Ed.; Plenum Press: New York, 1992; 1–14. 32. Chinn, J.A.;
Posso, S.E.; Horbett, T.A.; Ratner, B.D. Post-adsorptive
transitions in fibrinogen adsorbed to polyurethanes: changes
in antibody binding and sodium dodecyl sulphate
elutability. J. Biomed. Mater. Res. 1992, 26 , 757–778. 33.
Lu, D.R.; Park, K. Protein adsorption on polymer surfaces:
calculation of adsorption energies. J. Biomat. Sci. Polym.
Ed. 1990, 1, 243–260. 34. Barbucci, R.; Lamponi, S.;
Magnani, A.; Fibrinogen conformation and platelet
reactivity in relation to material-blood interaction:
effect of stress hormones. Biomacromolecules 2003, 4 (6),
1506–1513. H
Hemodynamics, Macrocirculatory

1. Milnor, W.R. Hemodynamics; 2nd Ed.; Williams and


Wilkins: Baltimore, MA, 1989.

2. McDonald, D.A. Blood Flow in Arteries, 2nd Ed.; The


Williams and Wilkins Co.: Baltimore, MA, 1974.

3. Chandran, K.B. Cardiovascular Biomechanics; New York


University Press: New York, NY, 1992.

4. Patel, D.J.; Vaishnav, R.N. Basic Hemodynamics and Its


Role in Disease Processes; University Park Press:
Baltimore, MA, 1980.

5. Pedley, T.J. Arterial and venous fluid dynamics. In


Cardiovascular Fluid Mechanics; 1st Ed.; Pedrizzetti, G.,
Perktold, K., Eds.; Springer Wien: New York, 2004; 1–72.

6. Fung, Y.C. Biodynamics: Circulation; Springer-Verlag:


New York, NY, 1984.

7. Womersley, J.R. An Elastic Tube Theory of Pulse


Transmission and Oscillatory Flow in Mammalian Arteries;
Wright Air Development Center Technical Report TR 56-614;
U.S. Air Force: Ohio, 1957.

8. Li, J.K.-J. Dynamics of the Vascular System; Li J.K.-J.,


Series Ed.; Series on Bioengineering & Biomedical
Engineering; World Scientific Publishing Co.: Singapore,
2004; Vol. 1.

9. Lieber, B.B. Arterial macrocirculatory hemodynamics. In


The Biomedical Engineering Handbook; 2nd Ed.; Bronzino,
J.D., Ed.; CRC Press: Boca Raton, FL, 2000; 30-1–30-10.
Heparin-Binding to Improve
Biocompatibility

21. Sa´nchez, J.; Olsson, P. On the control of the plasma


contact activation system on human endothelium comparison
with heparin surface. Thromb. Res. 1999, 93, 27–34.

22. Ware, J.A.; Coller, B.S. Platetlet Morphology,


Biochemistry and Function. In Williams Hematology, 5th Ed.;
Beutler, E., Lichtman, M.A., Coller, B.S., Kipps, T.J.,
Eds.; McGraw-Hill: New York, 1995; 1161–1201.

23. Mulivor, A.W.; Lipowsky, H.H. Role of glycocalyx in


leucocyte-endothelial cell adhesion. Am. J. Physiol, Heart
Circ. Physiol. 2002, 283, H1282–H1291.

24. Larsson, R.; Rosengren, A˚.; Olsson, P. Determination


of platelet adhesion to polyethylene and heparinized
surfaces with the aid of bioluminescence. Thromb. Res.
1977, 11, 517–530.

25. Larsson, R.; Eriksson, J.C.; Lagergren, H.; Olsson, P.


Platelet and plasma coagulation compatibility of
heparinized and sulphated surfaces. Thromb. Res. 1979, 15,
157–167.

26. Weber, N.; Wendel, H.P.; Ziemer, G. Hemocompatibility


of heparin-coated surfaces and the role of selective plasma
protein adsorption. Biomaterials 2002, 23, 429–439.

27. Bickel, C.; Rupprecht, H.J.; Darius, H.; Binz, C.;


Hauro¨der, B.; Kummenauer, F.; Meyer, J. Substantial
reduction of platelet adhesion by heparin-coated stents. J.
Interv. Cardiol. 2001, 14, 407–413.

28. Larsson, R.; Sele´n, G.; Bjo¨rklund, H.; Fagerholm, P.


Intraocular PMMA lenses modified with surface-immobilized
heparin: Evaluation of biocompatibility in vitro and in
vivo. Biomaterials 1989, 10, 511–516.

29. Chronos, N.; Markou, C.; Kocsis, J.; Llanos, G.;


Hanson, S. Surface heparinization profoundly decreases
acute thrombosis on Crown and Mini Crown stents in the
baboon arteriovenous shunt model. J. Am. Coll. Cardiol.
1998, 31 (Suppl. A), 413.

30. Mollnes, T.E.; Videm, V.; Christiansen, D.; Bergseth,


G.; Riesenfelt, J.; Hovig, T. Platelet compatibility of an
artificial surface modified with functionally active heparin.
Thromb. Haemost. 1999, 82, 1132–1136.
31. Keuren, J.F.W.; Wielders, S.J.H.; Willems, G.M.; Morra,
M.; Lindhout, T. Fibrinogen adsorption, platelet adhesion
and thrombin generation at heparinized surfaces exposed to
blood. Thromb. Haemost. 2002, 87, 742–747.

32. Solberg, R.; Scholtz, T.; Videm, V.; Okkenhaug, C.;


Aasen, A.O. Heparin coating reduces cell activation and
mediator release in an in vitro venovenous bypass model for
liver transplantaton. Transpl. Int. 1998, 11, 252–258.

33. Borowiec, J.W.; Jaramillo, A.; Venge, P.; Nilsson, L.;


Thelin, S. Effects of heparin-coating of cardiopulmonary
bypass circuits on leucocytes during simulated
extracorporeal circulation. Cardiovasc. Surg. 1997, 5 (6),
568–573.

34. Barstasd, R.M.; Hamers, M.J.; Mo¨ller, A.S.;


Sakariaassen, K.S. Monocyte procoagulant activity induced
by adherence to an artificial surface is reduced by endpoint
immmobilized heparin-coating of the surface. Thromb.
Haemost. 1998, 79, 302–305. 35. Fosse, E.; Moen, O.;
Johnson, E.; Semb, G.; Brockmeier, V.; Mollnes, T.E.;
Fagerhol, M.K.; Venge, P. Reduced complement and
granulocyte activation with heparin-coated cardiopulmonary
bypass. Ann. Thorac. Surg. 1994, 58, 472–477. 36. Larsson,
R.; Olsson, P.; Lindahl, U. Inhibition of thrombin on
surfaces coated with immobilized heparin and heparin-like
substances: A crucial non-thrombogenic principle. Thromb.
Res. 1980, 19, 43–54. 37. Olsson, P.; Sa´nchez, J.;
Mollnes, T.E.; Riesenfeld, J. On the compatibility of
end-point immobilized heparin. J. Biomater. Sci., Polym.
Ed. 2000, 11, 1261–1273. 38. Sa´nchez, J.; Lundquist, P.B.;
Elgue, G.; Larsson, R.; Olsson, P. Measuring the degree of
plasma contact activation induced by artificial materials.
Thromb. Res. 2002, 105, 407–412. 39. Sa´nchez, J.; Elgue,
G.; Riesenfeld, J.; Olsson, P. Studies of adsorption,
activation and inhibition of factor XII on immobilized
heparin. Thromb. Res. 1998, 89, 41–50. 40. Sa´nchez, J.;
Elgue, G.; Riesenfeld, J.; Olsson, P. Inhibition of the
plasma contact activation system of immobilized heparin:
Relation to surface density and antithrombin binding sites.
J. Biomed. Mater. Res. 1997, 36, 37–42. 41. Hong, J.;
Larsson, A.; Elgue, G.; Nilsson Ekdahl, K.; Larsson, R.;
Nilsson, B. Contact between biomaterial and whole blood:
The sequence of events that leads to thrombin generation.
J. Clin. Lab. Med. 2000, 138, 139–145. 42. Pekna, M.;
Larsson, R.; Formgren, B.; Nilsson, U.R.; Nilsson, B.
Complement activation by polymethyl methacrylate is
minimized by end-point heparin attachment. Biomaterials
1993, 14, 189–192. 43. Gong, J.; Larsson, R.; Nilsson
Ekdahl, K.; Mollnes, T.E.; Nilsson, U.; Nilsson, B. Tubing
loops as a model for cardiopulmonary bypass circuits: Both
the biomaterial and the blood-gas phase interphases induce
complement activation in an in vitro model. J. Clin.
Immunol. 1996, 16, 223–230. 44. Videm, V.; Mollnes, T.E.;
Garred, P.; Aasen, A.; Svennevig, J.L. Biocompatibility of
extracorporeal circulation: In vitro comparison of
heparin-coated and uncoated oxygenator circuits. J. Thorac.
Cardiovasc. Surg. 1991, 101, 654–660. 45. Videm, V.;
Svennevig, J.L.; Fosse, E.; Semb, G.; O¨ sterud, A.;
Mollnes, T.E. Reduced complement activation with
herapin-coated oxygenator and tubings in coronary bypass
operations. J. Thorac. Cardiovasc. Surg. 1992, 103,
806–813. 46. Kopp, R.; Mottaghy, K.; Kirschfink, M.
Mechanism of complement activation during extracorporeal
blood– biomaterial interaction: Effects of heparin coated
and uncoated surfaces. ASAIO J. 2002, 48, 598–605. 47.
Nilsson, B.; Larsson, R.; Hong, J.; Elgue, G.; Nilsson
Ekdahl, K.; Sahu, A.; Lambris, J.D. Compstatin inhibits
complement and cellular activation in whole blood in two
models of extracorporeal circulation. Blood 1998, 92,
1661–1667. 48. Conrad, H.E. Heparin-Binding Protients;
Academic Press: New York, 1998. 49. Osmond, R.I.W.; Kett,
W.C.; Skett, S.E.; Coombe, D.R. Protein-heparin
interactions measured by BIAcore 2000 are affected by the
method of heparin immobilization. Anal. Biochem. 2002, 310,
199–207. 50. Casu, B.; Lindahl, U. Structure and biological
interactions of heparin and heparan sulfate. Adv.
Carbohydr. Chem. Biochim. 2001, 57, 159–206. 51. Underwood,
P.A.; Kirkpatrick, A.; Mitchell, S.M. New insights into
heparin binding to vitronectin: Studies with monoclonal
antibodies. Biochem. J. 2002, 365, 57–67. 52. Schwarz, I.;
Seger, D.; Shaltiel, S. Vitronectin. Int. J. Biochem. Cell
Biol. 1999, 31, 539–544. 53. Timpl, R.; Tisi, J.F.; Andac,
Z.; Sasaki, T.; Hohenester, E. Structure and function of
laminin LG modules. Matrix Biol. 2000, 19, 309–317. 54.
Lookene, A.; Chevreuil, P.; Ostergaard, P.; Olivecrona, G.
Interaction of lipoprotein lipase with heparin fragments
and with heparan sulfate: Stoichiometry, stabilization, and
kinetics. Biochemistry 1996, 35, 12155–12163. 55.
Rosenberg, R.D.; Schworak, N.W.; Liu, J.; Schwartz, J.J.;
Zhang, L. Heparan sulfate proteoglycans of the
cardiovascular system. Specific structures emerge but how is
synthesis regulated. J. Clin. Invest. 1997, 99, 2062–2070.
56. Connelly, P.W. The role of hepatic lipase in
lipoprotein metabolism. Clin. Chim. Acta 1999, 286,
243–255. 57. Calabrese, C.G.; Recondo, E.F.; Fernanadez de
Recondo, M.E. Antithrombin and first complement protein
recognize the same active heparin fraction. Thromb. Res.
2002, 15, 537–541. 58. Blom, A.M. Structural and functional
studies of complement inhibitor C4b binding. Biochem. Soc.
Trans. 2001, 30, 978–982. 59. Zipfel, P.F.; Skerka, C.;
Hellwage, J.; Jokiranta, S.T.; Meri, S.; Brade, V.;
Kraiczy, P.; Noris, M.; Remuzzi, G. Factor H family
proteins: On complement, microbes and human diseases.
Biochem. Soc. Trans. 2001, 30, 971–978. 60. Carr, J.A.;
Silverman, N. The heparin-protamine interaction. A review.
J. Cardiovasc. Surg. (Torino) 1999, 40, 659–666. 61.
Colman, R.W. Role of the light chain of high molecular
weight kininogen in adhesion, cell-associated proteolysis
and angiogenesis. Biol. Chem. 2001, 382, 65–70. 62. Kaplan,
A.P.; Joseph, K.; Silverberg, M. Pathways for bradykinin
formation and inflammatory disease. J. Allergy Clin.
Immunol. 2002, 109, 195–209. 63. Xu, X.; Takano, R.; Nagai,
Y.; Yanagida, T.; Kamei, K.; Kato, H.; Kamikubo, Y.;
Nakahara, Y.; Kumeda, K.; Hara, S. Effect of heparin chain
length on the interaction with tissue factor pathway
inhibitor (TFPI). Int. J. Biol. Macromol. 2002, 30 ,
151–160. 64. Larsson, R.; Sele´n, G.; Formgren, B.; Holst,
A.S. Longterm stability of heparin surface-modified
intraocular lenses in vivo. J. Cataract Refract. Surg.
1990, 18, 247–251. 65. Spa˚ngberg, M.; Kihlstro¨m, I.;
Bjo¨rklund, H.; Bjurstro¨m, S.; Lydahl, E.; Larsson, R.
Improved biocompatibility of intraoculoar lenses by heparin
surface modification: A 12-month implantation study in
monkeys. J. Cataract Refract. Surg. 1990, 16, 170–177. 66.
Trocme, S.D.; Li, H. Effect of heparin-surface-modified
intraocular lenses on postoperative inflammation after
phacoemulsification: A randomized trial in a United States
patient population heparin-surface-modified lens study
group. Ophthalmology 2000, 107, 1031–1037. 67. van der
Giessen, W.; van Beusekom, H.M.M.; Larsson, R.; Serruys,
P.W. Heparin-coated coronary stents. Curr. Interv. Cardiol.
Rep. 1999, 1, 234–240. 68. Kristensen, E.M.E.; Rensmo, H.;
Larsson, R.; Siegbahn, H. Characterisation of a heparin
surface using photoelectron spectroscopy and quarts crystal
microbalance. Biomaterials 2003, 24, 4153–4159. 69.
Mueller, X.M.; Tevaearai, H.T.; Jegger, D.; Augstburger,
M.; Goddar, G.; von Segesser Antithrombotic properties of
trillium coated connectors. ASAIO J. 2002, 48, 483–486. 70.
Murphy, J.A.; Savage, C.M.; Alpard, S.K.; Deyo, D.J.;
Jayroe, J.B.; Zwischenberger, J.B. Low-dose versus
high-dose heparinization during arteriovenous carbon
dioxide removal. Perfusion 2001, 16, 460–468. 71. Ereth,
M.H.; Nuttall, G.A.; Clarke, S.H.; Dearani, J.A.;
Fiechtner, B.K.; Rishavy, C.R.; Buda, D.A.; Shaw, T.A.;
Orszulak, T.A.; Oliver, W.C., Jr. Biocompatibility of
trillium biopassive surface-coated oxygenator versus
uncoated oxygenator during cardiopulmonary bypass. J.
Cardiothorac. Vasc. Anesth. 2001, 15, 545–550. 72. Palanzo,
D.A.; Zarro, D.L.; Manley, N.J.; Montesano, R.M.; Quinn,
M.; Elmore, B.A.; Gustafson, P.A.; Castagna, J.M. Effect of
Carmeda bioactive surface coating versus Trillium
biopassive surface coating of the oxygenator on circulating
platelet count drop during cardiopulmonary bypass.
Perfusion 2001, 16, 279–283. 73. Sandhu, S.; Luthra, A. New
biointeracting materials. Med. Device Technol. Oct., 2002.
74. Hsu, L.C. Heparin-coated cardiopulmonary bypass
circuits: Current status. Perfusion 2001, 16, 417–428. 75.
Cramer, R.; Moore, R.; Amplatz, K. Reduction of the
surgical complication rate by the use of a hypothrombogenic
catheter coating. Radiology 1973, 109, 585–588. 76. Hsu,
L.C.; Tong, S.D. Ionic Heparin Complex. US patent
4.871.357, 1989. 77. Bader, K.E. Biocompatibility in
extracorporeal circulation: The heparin coating method
Bioline coating. Can. Perfus. Can. 1997, 9, 9–16. 78.
Tayama, E.; Hayashida, N.; Akasu, K.; Kosuga, T.; Fukunaga,
S.; Akashi, H.; Kawara, T.; Aoyagi, S. Biocompatibility of
heparin-coated extracorporeal bypass curcuits: New heparin
bonded Bioline system. Artif. Organs 2000, 24, 618–623. 79.
Nishinaka, T.; Tatsumi, E.; Taenaka, Y.; Katagiri, N.;
Ohnishi, H.; Shioya, K.; Fukuda, T.; Oshikawa, M.; Sato,
K.; Tsukiya, T.; Homma, A.; Takewa, Y.; Takano, H.; Sato,
M.; Kashiwabara, S.; Tanaka, H.; Sakai, K.; Matsuda, T. At
least thirty-four days of animal continuous perfusion by a
newly developed extracorporeal membrane oxygenation system
without systemic anticoagulants. Artif. Organs 2002, 26,
548–551. 80. Nojiri, C.; Hagiwara, K.; Yokoyama, K.;
Kuribayashi, E.; Hidaka, K.; Ishica, N.; Horiuchi, K.;
Oshiyama, H.; Nogawa, A.; Kido, T.; Kijama, T.; Akutsu, T.
Evaluation of a new heparin bonding process in process in
prolonged extracorporeal membrane oxygenation. ASAIO J.
1995, 41, M561–M567. H

81. Usui, A.; Hiroura, M.; Kawamura, M. Heparin coating


extends the durability of oxygenators used for
cardiopulmonary support. Artif. Organs 1999, 23, 840–844.

82. Svenmarker, S.; Ha¨ggmark, S.; Jansson, E.; Lindholm,


R.; Appelblad, M.; Sandstro¨m, E.; A˚berg, T. Use of
heparin-bonded circuits in cardiopulmonary bypass improves
clinical outcome. Scand. Cardiovasc. J. 2002, 36, 241–246.

83. Ernofsson, M.; Thelin, S.; Siegbahn, A. Thrombin


generation during cardiopulmonary bypass using
heparin-coated or standard curcuits. Scand. J. Thorac.
Cardiovasc. Surg. 1995, 29, 157–165.

84. Ovrum, E.; Holen, E.A.; Tangen, G.; Brosstad, F.;


Abdelnoor, M.; Ringdal, M.; Oystese, R.; Istad, R.
Completely heparinized cardiopulmonary bypass and reduced
systemic heparin: Clinical and hemostatic effects. Ann.
Thorac. Surg. 1995, 60, 365–371.

85. Bindslev, L.; Bohm, C.; Jolin, A.; HambraeusJonzon, K.;


Olsson, P.; Ryniak, S. Extracorporeal carbon dioxide
removal performed with surface-heparinized equipment in
patients with ARDS. Acta Anaesthesiol. Scand., Suppl. 1991,
95, 125–130. Discussion 130-1.

86. Kocsis, J.; Lunn, A.; Mohammad, S. Incomplete expansion


of coronary stents: Risk of thrombosis and protection
provided by a heparin coating. J. Am. Coll. Cardiol. 1996,
27 (Suppl. A), 84A.

87. Hardhammar, P.A.; vanBeusekom, H.M.; Emanuelsson, H.U.;


Hofma, S.H.; Albertsson, P.A.; Verdouw, P.D.; Boersma, E.;
Serruys, P.W.; van der Giessen, W.J. Reduction in
thrombotic events with heparin-coated Palmaz-Schatz stents
in normal porcine coronary arteries. Circulation 1996, 93,
423–430.

88. Serruys, P.W.; Emanuelsson, H.; van der Giessen, W.;


Lunn, A.C.; Kiemeney, F.; Macaya, C.; Rutsch, W.;
Heyndrickx, W.; Suryapranata, G.; Legrand, H.; Goy, V.;
Materne, J.J.; Bonnier, P.; Morice, H.; Fajadet, M.C.;
Belardi, J.; Colombo, J.; Garcia, A.; Ruygrok, E.; de
Jaegere, P.; Morel, M.A. Heparin-coated PalmazSchatz stents
in human coronary arteries. Early outcome of the
benestent-II pilot study. Circulation 1996, 93, 412–422.

89. Serruys, P.W.; van Hout, B.; Bonnier, H.; Legrand, V.;
Garcia, E.; Macaya, C.; Sousa, E.; van der Giessen, W.;
Colombo, A.; Seabra-Gomes, R.; Kiemeneij, F.; Ruygrok, P.;
Ormiston, J.; Emanuelsson, H.; Fajadet, J.; Haude, M.;
Klugmann, S.; Morel, M.A. Randomised comparison of
implantation of heparin-coated stents with balloon
angioplasty in selected patients with coronary artery
disease. (Benestent II). Lancet 1998, 352, 673–681. 90.
Wohrle, J.; Al-Khayer, E.; Grotzinger, U.; Schindler, C.;
Kochs, M.; Hombach, V.; Hoher, M. Comparison of the heparin
coated vs. the uncoated Jostent—No influence on restenosis
or clinical outcome. Eur. Heart J. 2001, 22, 1808–1816. 91.
Shin, E.K.; Son, W.J.; Sohn, M.S.; Jin, D.K.; Park, G.S.;
Koh, K.K.; Ahn, T.H.; Choi, I.C. Efficacy of heparin-coated
stents in early setting of acute myocardial infarction.
Catheter. Cardiovasc. Interv. 2001, 52, 306–312. 92.
Kocsis, J.F.; Llanos, G.; Holmer, E. Heparin-coated stents.
J. Long-Term Eff. Med. Implants 2000, 10 , 19–45. 93.
Walpoth, B.H.; Rogulenko, R.; Tikhvinskaia, E.; Gogolewski,
S.; Schaffner, T.; Hess, O.M.; Althaus, U. Improvement of
patency rate in heparin-coated small synthetic vascular
grafts. Circulation 1998, 98 (19 Suppl.), II319–II323. 94.
Wissink, M.J.; Beernink, R.; Pieper, J.S.; Poot, A.A.;
Engbers, G.H.; Beugeling, T.; van Aken, W.G.; Feijen, J.
Binding and release of basic fibroblast growth factor from
heparinized collagen matrices. Biomaterials 2001, 22,
2291–2299. 95. Foley, P.; Barthel, C.; Brausa, H. Effect of
covalently bound heparin coating on patency and
biocompatibility of long-term indwelling catheters in the
rat jugular vein. Comp. Med. 2002, 52, 243–248. 96.
Appelgren, P.; Ransjo, U.; Bindslev, L.; Espersen, F.;
Larm, O. Surface heparinization of central venous catheters
reduces microbial colonization in vitro and in vivo:
Results from a prospective, randomized trial. Crit. Care
Med. 1996, 24, 1482–1489. 97. Nilsson, E.; Arnander, C.
Long-term monitoring of arterial pO2 in burned patients.
Clin. Physiol. 1984, 4, 13–21. 98. Nagaoka, S.; Mikami, M.;
Shimizu, Y. Antithrombogenic pO2 sensor for continuous
intravascular oxygen monitoring. Biomaterials 1990, 11,
414–418.
Hip Biomechanics

Fig. 16 Stresses occurred in the liner at three different

time intervals of the average subluxation case. The first

time step (t=0.015 sec) represents the stresses in the


instant

the femoral head first impacts with the liner. The second

time step (t=0.02 sec) represents the bouncing of the

femoral head after the first impact. The third time

(t=0.03 sec) interval represents the second impact of the

femoral head with the liner. Table 5 Average rotation angle


and vertical displacement of the liner within the
acetabular cup Load (N) Euler angle (º) Rotational
displacement (lm) Vertical displacement (lm) 250 0.0141
8.11 1.02 350 0.0151 8.69 2.35 450 0.0163 9.40 3.29 550
0.0168 9.68 4.02 650 0.0216 12.47 5.74 750 0.0233 13.41
7.31 850 0.0269 15.51 8.84 950 0.0309 17.78 10.63 1050
0.0328 18.88 12.42 1150 0.0349 20.11 14.38 1250 0.041 23.60
16.18 3. Barrack, R.L.; Folgueras, A.; Munn, B.; et al.
Pelvic lysis and polyethylene wear at 5–8 years in an
uncemented total hip. Clin. Orthop. 1997, 355, 211–217. 4.
Engh, C.A.; O’Conner, D.; Jasty, M.; et al. Quantification
of implant micromotion, strain shielding, and bone
resorption with porous-coated anatomic medullary locking
femoral prostheses. Clin. Orthop. 1992, 285, 12–29. 5.
Maquet, P.G.J. Biomechanics of the Hip; Springer-Verlag:
Berlin, Germany, 1985. 6. Alexander, E.J.; Bregler, C.;
Andriachi, T.P. Limb segment pose from range data streams
through homogeneous factorization. Proceedings of 2001 ASME
Bioengineering Conference 2001, 50. 7. Davy, D.T.; Kotzar,
G.M.; Brown, R.H. Telemetric force measurements across the
hip after total arthroplasty. J. Bone Joint Surg. Am. 1988,
70 (1), 45–50. 8. Mulroy, R.D.; Harris, W.H. The effect of
improved cementing techniques on component loosening in hip
replacement: an 11-year radiographic review. J. Bone Joint
Surg. 1990, 72B, 757–760. 9. Agins, H.J.; Alcock, N.W.;
Bansal, M.; et al. Metallic wear in failed titanium-alloy
total hip replacements. A histological and quantitative
analysis. J. Bone Joint Surg. 1988, 70 (3), 347–356. 10.
Archibeck, M.J.; Jacobs, J.J.; Black, J. Alternate bearing
surfaces in total joint arthroplasty: biologic
considerations. Clin. Orthop. 2000, 1 (379), 12–21. 11.
Arthritis Foundation, 2004 http://www.arthritis.org/). 12.
Bellemans, J. Osseointegration in porous coated knee
arthroplasty. The influence of component coating type in
sheep. Acta Orthop. Scand. Suppl. 1999, 288, 1–35. 13.
Buma, P.; van Loon, P.J.; Versleyen, H.; et al.
Histological and biomechanical analysis of bone and
interface reactions around hydroxyapatite-coated
intramedullary implants of different stiffness: a pilot
study on goat. Biomaterials 1997, 18 (18), 1251–1260. 14.
Charnley, J. Low Friction Arthroplasty of the Hip; Springer
Verlag: New York, NY, 1978. 15. Charnley, J. Low Friction
Arthroplasty, Theory and Practice; Springer Verlag: New
York, NY, 1979. 16. Charnley, J. Acrylic Cement in
Orthopaedic Surgery; E. and S. Livingstone: Edinburgh,
Scotland, 1970. 17. Cobb, T.K.; Morrey, B.F.; Ilstrup, D.M.
Effect of the elevated-rim acetabular liner on loosening
after total hip arthroplasty. J. Bone Joint Surg. Am. 1997,
79 (9), 1361–1364. 18. Davy, D.T.; Kotzar, G.M.; Brown,
R.H.; et al. Telemetric force measurements across the hip
after total arthroplasty. J. Bone Joint Surg. Am. 1998, 70
(1), 45–50. 19. Dorr, L.D.; Wan, Z.; Cohen, J. Hemispheric
titanium porous coated acetabular component without screw
fixation. Clin. Orthop. 1998, 351, 158–168. 20. El Maraghy,
A.W.; Schemitsch, E.H.; Waddell, J.P. Acetabular blood flow
during total hip arthroplasty. Can. J. Surg. 2000, 43 (3),
197–201. 21. Engh, C.A. Surgical Technique; Anatomical
Medullary Locking Total Hip System with Porocoat; DePuy
Orthopaedics: Warsaw, IN, 1992. 22. Friedman, R.J.; Black,
J.; Galante, J.O.; et al. Current concepts in orthopaedic
biomaterials and implant fixation. J. Bone Joint Surg. Am.
1993, 75 (7), 1086–1109. 23. Goldring, S.R.; Schiller,
A.L.; Roelke, M.; et al. The synovial-like membrane at the
bone-cement interface in loose total hip replacements and
its proposed role in bone lysis. J. Bone Joint Surg. Am.
1983, 65 (5), 575–584. 24. Goodman, S.B.; Song, Y.; Chun,
L.; et al. Effects of TGF beta on bone ingrowth in the
presence of polyethylene particles. J. Bone Joint Surg. Br.
1999, 81 (6), 1069–1075. 25. Jaramaz, B.; Nikou, C.;
DiGioia, A.M. Sensitivity of impingement limits to error in
cup placement. Proc. Ann. Meeting ORS 402, 1998. 26. Jasty,
M.; Burke, D.; Harris, W.H. Biomechanics of cemented and
cementless prostheses. Chir. Organi. Mov. 1992, 77 (4),
349–358. 27. Kapandji, I.A. The Physiology of the Joints;
Churchill Livingston: Edinburgh, Scotland, 1987. 28.
Kienapfel, H.; Sprey, C.; Wilke, A.; Griss, P. Implant
fixation by bone ingrowth. J. Arthroplasty 1999, 14 (3),
355–368. 29. Learmonth, I.D.; Lee, M.B.; Bitounis, V.C.
Acetabular replacement with the cementless Harris-Galante
porous cup. J. Bone Joint Surg. 2001, 83B (suppl II), 143.
30. Leunig, M.; Hertel, R. Thermal necrosis after tibial
reaming for intramedullary nail fixation. A report of three
cases. J. Bone Joint Surg. Br. 1996, 78 (4), 584–587. 31.
Macdonald, W.; Carlsson, L.V.; Charnley, G.J.; et al.
Inaccuracy of acetabular reaming under surgical conditions.
J. Arthroplasty 1999, 14 (6), 730–737. 32. Malchau, H.;
Herberts, P.; Ahnfelt, L. Prognosis of total hip
replacement in Sweden. Follow-up of 92,675 operations
performed 1978-1990. Acta Orthop. Scand. 1993, 64 (5),
497–506. 33. Manley, M.T.; Capello, W.N.; D’Antonio, J.A.;
et al. Fixation of acetabular cups without cement in total
hip arthroplasty. A comparison of three different implant
surfaces at a minimum duration of follow-up of five years.
J. Bone Joint Surg. Am. 1998, 80 (8), 1175–1185. 34.
Maquet, P.G.J. Biomechanics of the Hip; Springer-Verlag:
Berlin, Germany, 1985. 35. Morrey, B.F. Instability after
total hip arthroplasty. Orthop. Clin. N. Am. 1992, 23,
237–248. 36. McCollum, D.E.; Gray, W.J. Dislocation after
total hip arthroplasty: causes and prevention. Clin.
Orthop. 1990, 261, 159–170. 37. Woo, R.Y.G.; Morrey, B.F.
Dislocation after total hip arthroplasty. J. Bone Joint
Surg. Am. 1982, 64, 1295–1306. 38. Lombardi, A.V.; Mallory,
T.H.; Dennis, D.A.; Komistek, R.D.; Fada, R.A.; Northcut,
E.J. An in vivo determination of total hip arthroplasty
pistoning during activity. J. Arthroplasty 2000, 15,
702–709. 39. Dennis, D.A.; Komisteck, R.D.; Northcut, E.J.;
Ochoa, J.A.; Ritchie, A. In vivo determination of hip joint
separation and the forces generated due to impact loading
conditions. J. Biomech. 2001, 34, 623–629. 40. McKellop,
H.A.; Campbell, P.; Park, S.H.; et al. The origin of
submicron polyethylene wear debris in total hip
arthroplasty. Clin. Orthop. 1995, 311, 3–20. 41. Moskowitz,
R.W. Osteoarthritis, Diagnosis and Medical/ Surgical
Management, 2nd Ed. Ed.; W.B. Saunders: Philadelphia, 1992.
H

42. Mu¨ller, C.H.R.; Rahn, B.A.; Pfister, U.; et al.


Intramedullary pressure, strain on the diaphysis and
increase in cortical temperature when reaming the femoral
cavity—a comparison of blunt and sharp reamers. Injury
1993, 24 (3), S22–S30.

43. Nakata, K.; Ohzono, K.; Masuhara, K.; et al. Acetabular


osteolysis and migration in bipolar arthroplasty of the
hip: fiveto 13-year follow-up study. J. Bone Joint Surg. Br.
1997, 79 (2), 258–264.

44. Teoh, S.H.; Chan, W.H.; Thampuran, R. An elastoplastic


finite element model for polyethylene wear in total hip
arthroplasty. J. Biomech. 2002, 35, 323–330.

45. Kurtz, S.M.; Edidin, A.A.; Bartel, D.L. The role of


backside polishing, cup angle, and polyethylene thickness
on the contact stresses in metal-backed acetabular
components. J. Biomech. 1997, 30, 639–642.

46. Northcut, E.J.; Komistek, R.D.; Dennis, D.A.; et al.


‘‘In vivo’’ determination of hip joint seperation that may
lead to impulse loading conditions. J. Biomech. 2001, 34
(5), 623–629.

47. Symplastics Inc. Ramex-UHMEPE. Ultra High Molecular


Weight Polyethylene Material Properties.

48. Brand, R.A.; Pedersen, D.R.; Davy, D.T. Comparison of


hip force calculations and measurements in the same
patient. J. Arthrop. 1994, 9, 45–51.

49. Morlock, M.; Sheneider, E.; Blumh, A.; Vollmer, M.;


Bergmann, G.; Muller, V.; Honl, M. Duration and frequency
of every day activities in total hip patients. J. Biomech.
2001, 34, 873–881.

50. Bergmann, G.; Deuretzbacher, G.; Heller, M.; Graichen,


F.; Rohlmann, A.; Strauss, J.; Duda, G.N. Hip contact
forces and gait patterns from routine activities. J.
Biomech. 2001, 34, 859–871.

51. Ipavec, M.; Brand, R.A.; Pedersen, D.R.; Mavcic, B.;


Kralj-Iglic, V.; Iglic, A. Mathematical modeling of stress
in the hip during gait. J. Biomech. 1999, 32, 1229–1235.

52. Herberts; Malchau; Garellick. The Annual Report of the


Swedish National Hip Arthroplasty Register; http://
www.jru.orthop.gu.se/2003 (accessed).

53. Ong, K.; Gunsallus, K.; Ryan, M.; et al. Comparison of


the initial stability of hemispherical and elliptical
acetabular cups. Proceeding sof 2001 ASME Bioengineering
Conference 2001, 50.

54. Pellicci, P.M.; Tria, A.J.; Garvin, K.L. Orthopaedic


knowledge update. J. Am. Acad. Ortho. Surg. 1999.

55. Perona, P.G.; Lawrence, J.; Paprosky, W.G.; et al.


Acetabular micromotion as a measure of initial implant
stability in primary hip arthroplasty. J. Arthroplasty
1992, 74, 537–547.

56. Pilliar, R.M.; Lee, J.M.; Maniatopoulos, C.


Observations on the effect of movement on bone ingrowth
into porous surfaced implants. Clin. Orthop. 1986, 208,
108–113.

57. Ranawat, C.S.; Peters, L.E.; Umlas, M.E. Fixation of


the acetabular component: the case for cement. Clin.
Orthop. 1997, 344, 207–215. 58. Saleh; Thongtrangan;
Schwarz Osteolysis: medical and surgical approaches. Clin.
Orthop. 2004, 427, 138–147. 59. Schmalzried, T.P.; Kwong,
L.M.; Jasty, M.; et al. The mechanism of loosening of
cemented acetabular components in total hip arthroplasty:
Analysis of specimens retrieved at autopsy. Clin. Orthop.
1992, 274, 60–78. 60. Schmalzried, T.P.; Guttmann, D.;
Grecula, M.; Amstutz, H.C. The relationship between the
design, position, and articular wear of acetabular
components inserted without cement and the development of
pelvic osteolysis. J. Bone Joint Surg. 1994, 76 (A),
677–688. 61. Spears, I.R.; Pfleiderer, M.; Schneider, E.; et
al. Interfacial conditions between a press-fit acetabular
cup and bone during daily activities: implications for
achieving bone in-growth. J. Biomech. 2000, 33 (11),
1471–1477. 62. Van Lenthe, G.H.; Van Den Bergh, J.P.W.;
Hermus, A.R.M.M.; Huiskes, R. The prospects of estimating
trabecular bone tissue properties from the combination of
ultrasound, dual-energy x-ray absorptiometry, microcomputer
tomography, and microfinite element analysis. J. Bone Miner.
Res. 2001, 16 (3), 550–555. 63. Walde; Claus; Mohan, et al.
Advancement and separation of apex hole eliminators with
cementless duraloc 100 cups. A report of nineteen cases. J.
Bone Joint Surg. Am. 2004, 86-A (10), 2251–2256. 64.
Wessinghage, D.; Kisslinger, E. Long-term results after
cemented total hip arthroplasty in chronic polyarthritis.
Orthopade. 1998, 27 (6), 381–391. 65. Willert, H.G.
Reaction of the articular capsule to wear products of
artificial joint prostheses. J. Biomed. Mater. Res. 1977, 11
(2), 157–164. 66. Wolfe, F.; Zwillich, S.H. The long-term
outcomes of rheumatoid arthritis: a 23-year perspective,
longitudinal study of the total joint replacement and its
predictors in 1,600 patients with rheumatoid arthritis.
Arthritis Rheum. 1998, 41 (6), 1072–1082. 67. Won, C.H.;
Hearn, T.C.; Tile, M. Micromotion of cementless
hemispherical acetabular components: does press-fit need
adjunctive screw fixation? J. Bone. Joint. Surg. 1995, 77B,
484–489. 68. Finkelstein, J.A.; Anderson, G.I.; Waddell,
J.P.; et al. A study of micromotion and appositional bone
growth to a canine maderporic-surfaced femoral component.
J. Arthroplasty 1994, 9 (3), 317–324. 69. Maloney, W.J.;
Paprosky, W.; Engh, C.; et al. Surgical treatment of pelvic
osteolysis. Clin. Orthop. 2001, 393, 78–84. 70. Maloney,
W.J.; Galante, J.O.; Anderson, M.; et al. Fixation,
polyethylene wear, and pelvic osteolysis in primary total
hip replacement. Clin. Orthop. 1999, 369 , 157–164.
Hip Joint: Overuse Injuries

22. Dederich, R. Die Schnappende Hufte erweiterung des


Tractus Iliotibialis durch Z-plastik. Z. Orthop. 1983, 121,
168–170.

23. Njoo, K.H.; Van der Does, E. The occurrence and


intre-rater reliability of myofascial trigger points in the
quadratus lumborum and gluteus medius: a prospective study
in non-specific low back pain patients and controls in
general practice. Pain 1994, 58, 317–323.

24. Pec´ina, M.; Bojanic´, I.; Hasˇpl, M. The snapping hip.


Hip Int. 1994, 4, 133–136.

25. Reid, D.C. Prevention of hip and knee injuries in


ballet dancers. Sports Med. 1988, 6, 295–307.

26. Schaberg, J.E.; Harper, M.C.; Allen, W.C. The snapping


hip syndrome. Am. J. Sports Med. 1984, 12, 361–365.

27. Melamed, H.; Hutchinson, M.R. Soft tissue problems of


the hip in athletes. Sports Med. Arthrosc. Rev. 2002, 10,
168–175.

28. Zoltan, D.J.; Clancy, W.G., Jr.; Keene, J.S. A new


operative approach to snapping hip and refractory
trochanteric bursitis in athletes. Am. J. Sports Med. 1986,
14, 201–204. 29. Rask, M.R. Snapping bottom: subluxation of
the tendon of the long head of the biceps femoris muscle.
Muscle Nerve 1980, 3, 250–251. 30. Askling, C.; Tengvar,
M.; Saartok, T.; Thorstensson, A. Sports related hamstring
strains—two cases with different etiologies and injuries
sites. Scand. J. Med. Sci. Sports 2000, 10, 304–307. 31.
Orchard, J.W. Intrinsic and extrinsic risk factors for
muscle strain in Australian football. Am. J. Sports Med.
2001, 29, 300–303. 32. Puranen, J.; Orava, S. The hamstring
syndrome. Am. J. Sports Med. 1988, 16, 517–521. 33. De
Smet, A.A.; Best, T.M. MR imaging of the distribution and
location of acute hamstring injuries in athletes. Am. J.
Roentgenol. 2000, 174, 393–399. 34. Pec´ina, M.;
Krmpotic´-Nemanic´, J.; Markiewitz, A.D. Tunnel Syndromes.
Peripheral Nerve Compression Syndromes, 3rd Ed.; CRC Press:
Boca Raton, 2001. 35. Kujala, U.M.; Orava, S.; Jarvinen, M.
Hamstring injuries. Current trends in treatment and
prevention. Sports Med. 1997, 23, 397–404. 36. Orava, S.;
Kujala, U.M. Rupture of the ischial origin of the hamstring
muscles. Am. J. Sports Med. 1995, 23, 702–705.
Histogenesis

5. Wolff, J. The Law of Bone Remodeling; Springer-Verlag:


Berlin, 1986. Translated by P. Maquet.

6. Martin, R.B.; Burr, D.B.; Sharkey, N.A. Skeletal Tissue


Mechanics; Springer: New York, NY, 1998.

7. Saunders, J.W.; Cairns, J.M.; Gasseling, M.T. The role


of apical ectodermal ridge in the differentiation of the
morphological structure of and inductive specificity of limb
parts of the chick. J. Morphol. 1957, 101, 57–88.

8. Toole, B.P. Hyaluronan in Morphogenesis. Semin. Cell


Dev. Biol. 2001, 12, 79–87.

9. Ingber, D.E. Mechanochemical Switching Between Growth


and Differentiation by Extracellular Matrix. In Principles
of Tissue Engineering; Lanza, R., Langer, R., Chick, W.,
Eds.; RG Landes Company: Austin, TX, 1997; 89–100.

10. Le Mouellic, H.; Lallemand, Y.; Brulet, P. Homeosis in


the mouse induced by a null mutation in the Hox-3.1 gene.
Cell 1992, 69, 251–264.

11. Vastardis, H.; Karimbux, N.; Guthua, S.W.; Seidman,


J.G.; Seidman, C.E. A human MSX1homeodomain missense
mutation causes selective tooth agenesis. Nat. Genet. 1996,
13, 417–421.

12. Hess, A. Reaction of mammalian fetal tissue to injury.


II. Skin. Anat. Rec. 1954, 19, 435–453.

13. Shah, M.; Foreman, D.M.; Ferguson, M.W. Neutralizing


antibody to TGF-beta 1,2 reduces cutaneous scarring in
adult rodents. J. Cell. Sci. 1994, 107, 1137–1157.

14. Clark, R.A.F. Wound Repair: Lessons for Tissue


Engineering. In Principles of Tissue Engineering; Lanza,
R., Langer, R., Chick, W., Eds.; R.G. Landes Company:
Austin, TX, 1997; 737–768.

15. Marx, R.E. Platelet-Rich Plasma: A Source of Multiple


Autologous Growth Factors for Bone Grafts. In Tissue
Engineering. Application in Maxillofacial Surgery and
Periodontics; Lynch, S.E., Genco, R.J., Eds.; Quintessence
Books: Chicago, 1999; 71–82.

16. Giannobile, W. Periodontal Tissue Regeneration by


Polypeptide Growth Factors and Gene Transfer. In Tissue
Engineering. Application in Maxillofacial Surgery and
Periodontics; Lynch, S.E., Genco, R.J., Eds.; Quintessence
Books: Chicago, 1999; 231–243.

17. Eid, K.; Chen, E.; Griffith, L.; Glowacki, J. Effect of


RGD-coating on osteocompatibility of PLGA-polymer disks in
a rat tibial wound. J. Biomed. Mater. Res. 2001, 57,
224–231.

18. Glowacki, J. Tissue Response to Bone-Derived Materials.


In Bone Grafts: From Basic Science to Clinical Application;
Habal, M.B., Reddi, A.H., Eds.; W.B. Saunders:
Philadelphia, 1992; 84–92.

19. Glowacki, J.; Kaban, L.B.; Murray, J.E.; Folkman, J.;


Mulliken, J.B. Application of the biological principle of
induced osteogenesis for craniofacial defects. Lancet 1981,
(i), 959–963.

20. Upton, J.; Glowacki, J. Hand reconstruction with


allograft demineralized bone: 26 Implants in 12 patients.
J. Hand Surg. 1992, 17A, 704–713.

21. Rosenthal, R.K.; Folkman, J.; Glowacki, J.


Demineralized bone implants for non-union fractures, bone
cysts, and fibrous bone lesions. Clin. Orthop. Relat. Res.
1999, 364 , 61–69. 22. Urist, M.R. Bone: Formation by
autoinduction. Science 1956, 150, 893–899. 23. Reddi, A.H.;
Huggins, C.B. Biochemical sequences in the transformation
of normal fibroblasts in adolescent rats. Proc. Natl. Acad.
Sci. U. S. A. 1972, 69, 1601–1605. 24. Reddi, A.H. Bone
morphogenetic proteins: From basic science to clinical
applications. J. Bone Jt. Surg., Am. 2001, 83-A, S1–S6. 25.
Yates, K.E.; Glowacki, J. Altered expression of connective
tissue genes in postnatal chondroinduced human dermal
fibroblasts. Connect. Tissue Res. 2003, 44, 1–7. 26. Grande,
D.A.; Pitman, M.I.; Peterson, L.; Menche, D.; Klein, M. The
repair of experimentally produced defects in rabbit
articular cartilage by autologous chondrocyte
transplantation. J. Orthop. Res. 1989, 7, 208–218. 27.
Brittberg, M.; Lindhal, A.; Nilsson, A.; Ohlsson, C.;
Isaksson, O.; Peterson, L. Treatment of deep cartilage
defects in the knee with autologous chondrocyte
transplantation. New Engl. J. Med. 1994, 331, 889–895. 28.
Glowacki, J. In vitro engineering of cartilage. J. Rehabil.
Res. Dev. 2000, 37, 171–177. 29. Hauselmann, H.J.;
Fernandes, R.J.; Mok, S.S.; Schmid, T.M.; Block, J.A.;
Aydelotte, M.B.; Kuettner, K.E.; Thonar, E.J. Phenotypic
stability of bovine articular chondrocytes after long-term
culture in alginate beads. J. Cell Sci. 1994, 107, 17–27.
30. Mizuno, S.; Tateishi, T.; Ushida, T.; Glowacki, J.
Hydrostatic fluid pressure enhances matrix synthesis and
accumulation by bovine chondrocytes in threedimensional
culture. J. Cell. Physiol. 2002, 193, 319–327. 31. Kisiday,
J.; Jin, M.; Kurz, B.; Hung, H.; Semino, C.; Zhang, S.;
Grodzinsky, A.J. Self-assembling peptide hydrogel fosters
chondrocyte extracellular matrix production and cell
division: Implications for cartilage tissue repair. Proc.
Natl. Acad. Sci. U. S. A. 2002, 99, 9996–10001. 32.
Glowacki, J. Engineered cartilage, bone, joints, and
menisci. Cells Tissues Organs 2001, 169, 302–308. 33.
Mizuno, S.; Glowacki, J. Chondroinduction of human dermal
fibroblasts by demineralized bone in threedimensional
culture. Exp. Cell Res. 1996, 227, 89–97. 34. Mueller,
S.M.; Glowacki, J. Construction and regulation of
three-dimensional bone tissue in vitro. In Bone
Engineering; Davies, J.E., Ed.; Em squared, Inc.: Toronto,
2000; 473–487. 35. Mueller, S.M.; Mizuno, S.; Gerstenfeld,
L.C.; Glowacki, J. Medium perfusion enhances osteogenesis
by murine osteosarcoma cells in three-dimensional collagen
sponges. J. Bone Miner. Res. 1999, 14, 2118–2126. 36.
Krebsbach, P.H.; Kuznetsov, S.A.; Bianco, P.; Robey, P.G.
Bone marrow stromal cells: Characterization and clinical
application. Crit. Rev. Oral Biol. Med. 1999, 10, 165–181.
Host Reactions

18. Kirkpatrick, C.; Alves, A.; Kohler, H.; Kriegsmann, J.;


Bittinger, F.; Otto, M.; Williams, D.; Eloy, R.
Biomaterial-induced sarcoma: A novel model to study
preneoplastic change. Am. J. Pathol. 2000, 156 (4),
1455–1467.

19. Nakaoka, R.; Tsuchiya, T.; Nakamura, A. Studies on the


mechanisms of tumorigenesis induced by polyetherurethane in
rats: Production of superoxide, tumor necrosis factor, and
interleukin 1 from macrophages cultured on different
polyetherurethanes. J. Biomed. Mater. Res. 2000, 49,
99–105. 20. Nakaoka, R.; Tsuchiya, T.; Nakamura, A. Studies
on the tumor-promoting activity of polyethylene: Inhibitory
activity of metabolic cooperation of polyethylene films
containing an antioxidant. J. Long-Term Eff. Med. Implants
1995, 5 (4), 253–262. 21. Kinoshita, Y.; Kuzuhara, T.;
Kobayashi, M.; Ikada, Y. Reduction in tumor formation on
polyethylene by collagen immobilization. J. Long-Term Eff.
Med. Implants 1995, 5 (4), 275–284.
Hyaluronan

8. Toole, B.P. Hyaluronan in morphogenesis. Semin. Cell


Dev. Biol. 2001, 12 (2), 79–87.

9. Roden, L.; Campbell, P.; Fraser, J.R.; Laurent, T.C.;


Pertoft, H.; Thompson, J.N. Enzymic pathways of hyaluronan
catabolism. Ciba Found. Symp. 1989, 143, 60–76.

10. West, D.C.; Fan, T.-P.D. Hyaluronan Oligosaccharides


Promote Wound Repair. In The New Angiotherapy; Fan,
T.-P.D., Kohn, E.C., Eds.; Humana Press: Totowa, NJ, 2002;
177–188.

11. Noble, P.W. Hyaluronan and its catabolic products in


tissue injury and repair. Matrix Biol. 2002, 21 (1), 25–29.

12. Toole, B.P.; Wight, T.N.; Tammi, M.I. Hyaluronan– cell


interactions in cancer and vascular disease. J. Biol. Chem.
2002, 277 (7), 4593–4596.

13. Vercruysse, K.P.; Prestwich, G.D. Hyaluronate


derivatives in drug delivery. Crit. Rev. Ther. Drug Carr.
Syst. 1998, 15 (5), 513–555.

14. Campoccia, D.; Doherty, P.; Radice, M.; Brun, P.;


Abatangelo, G.; Williams, D.F. Semisynthetic resorbable
materials from hyaluronan esterification. Biomaterials 1998,
19 (23), 2101–2127.

15. Pouyani, T.; Prestwich, G.D. Functionalized derivatives


of hyaluronic acid oligosaccharides: Drug carriers and
novel biomaterials. Bioconjug. Chem. 1994, 5 (4), 339–347.

16. Bulpitt, P.; Aeschlimann, D. New strategy for chemical


modification of hyaluronic acid: Preparation of
functionalized derivatives and their use in the formation
of novel biocompatible hydrogels. J. Biomed. Mater. Res.
1999, 47 (2), 152–169.

17. Kuo, J.-W.; Swann, D.A.; Prestwich, G.D. Chemical


modification of hyaluronic acid by carbodiimides. Bioconjug.
Chem. 1991, 2, 232–241.

18. Barbucci, R.; Magnani, A.; Casolaro, M.; Marchettini,


N.; Rosi, C.; Bosco, M. Modification of hyaluronic acid by
insertion of sulfate groups to obtain a heparin-like
molecule. Part I. Characterization and behavior in aqueous
solution towards H+ and Cu2+ ions. Gazz. Chim. Ital. 1995,
125 (4), 169–180.
19. Magnani, A.; Albanese, A.; Lamponi, S.; Barbucci, R.
Blood-interaction performance of differently sulphated
hyaluronic acids. Thromb. Res. 1996, 81 (3), 383–395.

20. Laurent, T.C.; Hellsing, K.; Gelotte, B. Crosslinked


gels of hyaluronic acid. Acta Chem. Scand. 1964, 18 (1),
274–275.

21. Yui, N.; Okano, T.; Sakurai, Y. Inflammation responsive


degradation of crosslinked hyaluronic acid gels. J.
Control. Release 1992, 22 (2), 105–116.

22. Zhao, X.B.; Fraser, J.E.; Alexander, C.; Lockett, C.;


White, B.J. Synthesis and characterization of a novel
double crosslinked hyaluronan hydrogel. J. Mater. Sci.,
Mater. Med. 2002 , 13, 11–16.

23. Tomihata, K.; Ikada, Y. Crosslinking of hyaluronic acid


with water-soluble carbodiimide. J. Biomed. Mater. Res.
1997, 37 (2), 243–251.

24. Vercruysse, K.P.; Marecak, D.M.; Marecek, J.F.;


Prestwich, G.D. Synthesis and in vitro degradation of new
polyvalent hydrazide cross-linked hydrogels of hyaluronic
acid. Bioconjug. Chem. 1997, 8 (5), 686–694. 25. Balazs,
E.A.; Bland, P.A.; Denlinger, J.L.; Goldman, A.I.; Larsen,
N.E.; Leshchiner, E.A.; Leshchiner, A.; Morales, B. Matrix
engineering. Blood Coagul. Fibrinolysis 1991, 2 (1),
173–178. 26. Tomihata, K.; Ikada, Y. Crosslinking of
hyaluronic acid with glutaraldehyde. J. Polym. Sci., A,
Polym. Chem. 1997, 35, 3553–3559. 27. Hu, M.; Sabelman,
E.E.; Tsai, C.; Tan, J.; Hentz, V.R. Improvement of Schwann
cell attachment and proliferation on modified hyaluronic
acid strands by polylysine. Tissue Eng. 2000, 6 (6),
585–593. 28. Band, P.A. Hyaluronan Derivatives: Chemistry
and Clinical Applications. In Chemistry, Biology and
Medical Applications of Hyaluronan and Its Derivatives;
Laurent, T.C., Ed.; Portland Press: London, 1998; 33–42.
29. Anseth, K.S.; Burdick, J.A. New directions in
photopolymerizable biomaterials. Mater. Res. Soc. Bull.
2002, 27 (2), 130–136. 30. Matsuda, T.; Moghaddam, M.J.;
Miwa, H.; Sakurai, K.; Iida, F. Photoinduced prevention of
tissue adhesion. ASAIO J. 1992, 38 (3), M154–M157. 31.
Smeds, K.A.; Pfister-Serres, A.; Hatchell, D.L.; Grinstaff,
M.W. Synthesis of a novel polysaccharide hydrogel. J.
Macromol. Sci., Part A, Pure Appl. Chem. 1999, A36 (7 and
8), 981–989. 32. Leach, J.B.; Bivens, K.A.; Patrick, C.W.,
Jr.; Schmidt, C.E. Photocrosslinked hyaluronic acid
hydrogels: Natural, biodegradable tissue engineering
scaffolds. Biotechnol. Bioeng. 2003, 82, 578–589. 33.
Matsuda, T.; Magoshi, T. Preparation of vinylated
polysaccharides and photofabrication of tubular scaffolds
as potential use in tissue engineering. Biomacromolecules
2002, 3 (5), 942–950. 34. Chen, G.; Ito, Y.; Imanishi, Y.;
Magnani, A.; Lamponi, S.; Barbucci, R. Photoimmobilization
of sulfated hyaluronic acid for antithrombogenicity.
Bioconjug. Chem. 1997, 8 (5), 730–734. 35.
http://www.fda.gov/cdrh. (accessed December 2003). 36.
Pozo, M.A.; Balazs, E.A.; Belmonte, C. Reduction of sensory
responses to passive movements of inflamed knee joints by
hylan, a hyaluronan derivative. Exp. Brain Res. 1997, 116
(1), 3–9. 37. Peyron, J.G. A new approach to the treatment
of osteoarthritis: Viscosupplementation. Osteoarthr.
Cartil. 1993, 1 (2), 85–87. 38. Brun, P.; Abatangelo, G.;
Radice, M.; Zacchi, V.; Guidolin, D.; Daga Gordini, D.;
Cortivo, R. Chondrocyte aggregation and reorganization into
threedimensional scaffolds. J. Biomed. Mater. Res. 1999, 46
(3), 337–346. 39. Solchaga, L.A.; Dennis, J.E.; Goldberg,
V.M.; Caplan, A.I. Hyaluronic acid-based polymers as cell
carriers for tissue-engineering repair of bone and
cartilage. J. Orthop. Res. 1999, 17 (2), 205–213. 40.
Pavesio, A.; Renier, D.; Cassinelli, C.; Morra, M.
Antiadhesive surfaces through hyaluronan coatings. Med.
Device Technol. 1997, 8 (7), 20–21, 24–27. 41. Kito, H.;
Matsuda, T. Biocompatible coatings for luminal and outer
surfaces of small-caliber artificial grafts. J. Biomed.
Mater. Res. 1996, 30 (3), 321–330. 42. Camenisch, T.D.;
Schroeder, J.A.; Bradley, J.; Klewer, S.E.; McDonald, J.A.
Heart-valve mesenchyme formation is dependent on
hyaluronan-augmented activation of ErbB2-ErbB3 receptors.
Nat. Med. 2002, 8 (8), 850–855. 43. Kirker, K.R.; Luo, Y.;
Nielson, J.H.; Shelby, J.; Prestwich, G.D.
Glycosaminogylcan hydrogel films as biointeractive dressings
for wound healing. Biomaterials 2002, 23 (17), 3661–3671.
44. Duranti, F.; Salti, G.; Bovani, B.; Calandra, M.;
Rosati, M.L. Injectable hyaluronic acid gel for soft tissue
augmentation. A clinical and histological study. Dermatol.
Surg. 1998, 24 (12), 1317–1325. 45. Seckel, B.R.; Jones,
D.; Hekimian, K.J.; Wang, K.K.; Chakalis, D.P.; Costas,
P.D. Hyaluronic acid through a new injectable nerve guide
delivery system enhances peripheral nerve regeneration in
the rat. J. Neurosci. Res. 1995, 40 (3), 318–324. 46.
Collier, J.H.; Camp, J.P.; Hudson, T.W.; Schmidt, C.E.
Synthesis and characterization of polypyrrole-hyaluronic
acid composite biomaterials for tissue engineering
applications. J. Biomed. Mater. Res. 2000, 50 (4), 574–584.
47. Cortivo, R.; Brun, P.; Cardarelli, L.; O’Regan, M.;
Radice, M.; Abatangelo, G. Antioxidant effects of
hyaluronan and its alpha-methyl-prednisolone derivative in
chondrocyte and cartilage cultures. Semin. Arthritis Rheum.
1996, 26 (1), 492–501. 48. Moseley, R.; Leaver, M.; Walker,
M.; Waddington, R.J.; Parsons, D.; Chen, W.Y.; Embery, G.
Comparison of the antioxidant properties of HYAFF11p75,
AQUACEL and hyaluronan towards reactive oxygen species in
vitro. Biomaterials 2002, 23 (10), 2255–2264. 49. Glass,
J.R.; Dickerson, K.T.; Stecker, K.; Polarek, J.W.
Characterization of a hyaluronic acid-Arg-Gly-Asp peptide
cell attachment matrix. Biomaterials 1996, 17 (11),
1101–1108. 50. Tomer, R.; Dimitrijevic, D.; Florence, A.T.
Electrically controlled release of macromolecules from
cross-linked hyaluronic acid hydrogels. J. Control. Res.
1995, 33, 405–413. 51. Ohya, S.; Nakayama, Y.; Matsuda, T.
Thermoresponsive artificial extracellular matrix for tissue
engineering: hyaluronic acid bioconjugated with poly
(N-isopropylacrylamide) grafts. Biomacromolecules 2001, 2
(3), 856–863. 52.
http://www.glycoforum.gr.jp/science/hyaluronan/
hyaluronanE.html. (accessed December 2002). H
Hybrid Vascular Prostheses

11. Konrad, P.; Dougan, P.; Bergqvist, D. Acute


thrombogenicity of collagen coating of Dacron grafts: an
experimental study in sheep. Eur. J. Surg. 1992, 6, 67–72.

12. Guidoin, R.; Marceau, D.; Couture, J.; Rao, T.J.;


Merhi, Y.; Roy, P-E.; de la Faye, D. Collagen coatings as
biological sealants for textile arterial prostheses.
Biomaterials 1989, 10, 156–165.

13. Edwards, G.A.; Beck, C.; Charles, J.A. 2001. Evaluation


of a gelatin-sealed ePTFE vascular prosthesis in a dog
model. Proceedings 25th Anniversary Meeting of European
Society for Biomaterials, London, U.K., Sep 12–14, 2001;
106.

14. Tiwari, A.; Salacinski, H.J.; Hamilton, G.; Seifalin,


A.M. Tissue engineering of vascular bypass grafts: role of
endothelial cell extraction. Eur. J. Vasc. Endovasc. Surg.
2001, 21, 193–201.

15. Seifalian, A.M.; Tiwari, A.; Hamilton, G.; Salacinski,


H.J. Improving the clinical patency of prosthetic vascular
and coronary bypass grafts: the role of seeding and tissue
engineering. Artif. Organs 2002, 26, 307–320.

16. Deutsch, M.; Meinhart, J.; Fischlein, T.; Preiss, P.;


Zilla, P. Clinical autologous in vitro endothelialization
of infrainguinal ePTFE grafts in 100 patients: a 9-year
experience. Surgery 1999, 126, 847–855.

17. Leseche, G.; Ohan, J. Enhanced patency of venous Dacron


grafts by endothelial cell sodding. Ann. Vasc. Surg. 1995,
9, 325–326.

18. Tiwari, A.; Salacinski, H.J.; Punshon, G.; Hamilton,


G.; Seifalian, A.M. Development of a hybrid cardiovascular
graft using a tissue engineering approach. FASEB J. 2002,
16, 791–796.

19. Rashid, S.T.; Salacinski, H.J.; Button, M.J.; Fuller,


B.; Hamilton, G.; Seifalian, A.M. Cellular engineering of
conduits for coronary and lower limb bypass surgery: role
of cell attachment peptides and pre-conditioning in
optimising smooth muscle cells (SMC) adherence to compliant
poly(carbonate-urea)urethane (MyoLink) scaffolds. Eur. J.
Vasc. Endovasc. Surg. 2004, 27, 608–616.

20. Turner, N.J.; Kielty, C.M.; Walker, M.G.; Canfield, A.E.


A novel hyaluronan-based biomaterial (Hyaff-11) as a
scaffold for endothelial cells in tissue engineered
vascular grafts. Biomaterials 2004, 25, 5955–5964.

21. Swartz, D.D.; Russell, J.A.; Andreadis, S.T.


Engineering of fibrin-based functional and implantable
small-diameter blood vessels. Am. J. Physiol. Heart Circ.
Physiol. 2005, 288, H1451–H11460.

22. Birchall, I.E.; Lee, V.W.; Ketharanathan, V. Retention


of endothelium on ovine collagen biomatrix vascular
conduits under physiological shear stress. Biomaterials
2001, 22, 3139–3144.

23. Sparks, C.H. Autogenous grafts made to order. Ann.


Thorac. Surg. 1969, 8, 104–113.

24. Hoenig, M.R.; Campbell, G.R.; Rolfe, B.E.; Campbell,


J.H. Tissue-engineered blood vessels: alternative to
autologous grafts? Arterioscler. Thromb. Vasc. Biol. 2005,
25, 1128–1134. 25. Campbell, J.H.; Efendy, J.L.; Campbell,
G.R. Novel vascular graft grown within recipient’s own
peritoneal cavity. Circ. Res. 1999, 85, 1173–1178. 26.
Daly, C.D.; Campbell, G.R.; Walker, P.J.; Campbell, J.H. In
vivo engineering of blood vessels. Front. Biosci. 2004, 9,
1915–1924. 27. Ketharanathan, V.; Christie, B.A.
Glutaraldehyde-tanned ovine collagen conduits as vascular
xenografts in dogs. Arch. Surg. 1980, 115, 967–969. 28.
Edwards, G.A.; Roberts, G. Development of an ovine
collagen-based composite vascular prosthesis. Clin. Mater.
1992, 9, 211–223. 29. Werkmeister, J.A.; White, J.F.;
Ramshaw, J.A.M. Evaluation of the Omniflow collagen-polymer
vascular prosthesis. Med. Progr. Technol. 1994, 20,
231–242. 30. Koch, G.; Gutschi, S.; Pasher, O.; Fruhwirth,
J.; Glanzer, H. Analysis of 274 Omniflow vascular prostheses
implanted over an eight year period. Aust. N.Z. J. Surg.
1997, 67, 637–639. 31. Ramshaw, J.A.M.; Casagranda, F.;
White, J.F.; Edwards, G.A.; Hunt, J.A.; Williams, D.F.;
Werkmeister, J.A. Effects of mesh modification on the
structure of a mandrel grown biosynthetic vascular
prosthesis. J. Biomed. Mater. Res. 1999, 47, 309–315. 32.
Weinberg, C.B.; Bell, E. A blood vessel model constructed
from collagen and cultured vascular cells. Science 1986,
231, 397–400. 33. Hirai, J.; Matsuda, T. Venous
reconstruction using hybrid vascular tissue composed of
vascular cells and collagen: tissue regeneration process.
Cell Transplant 1997, 5, 93–105. 34. L’Heureux, N.; Paquet,
S.; Labbe, R.; Germain, L.; Auger, F.A. A completely
biological tissue-engineered human blood vessel. FASEB J.
1998, 12, 47–56. 35. Shinoka, T.; Shumtim, D.; Ma, P.X.;
Tanel, R.E.; Isogai, N.; Langer, R.; Vacanti, J.P.; Mayer,
J.E. Creation of viable pulmonary artery autografts through
tissue engineering. J. Thorac. Cardiovasc. Surg. 1998, 115,
536–545. 36. Matsuda, T. Recent progress of vascular graft
engineering in Japan. Artif. Organs 2004, 28, 64–71. 37.
L’Heureux, N.; Germain, L.; Labbe, R.; Auger, F.A. In vitro
construction of a human blood vessel from cultured vascular
cells: a morphologic study. J. Vasc. Surg. 1993, 17,
499–509. 38. Shinoka, T.; Imai, Y.; Ikeda, Y.
Transplantation of a tissue-engineered pulmonary artery.
New Engl. J. Med. 2001, 344, 532–533. 39. Shum-Tim, D.;
Stock, U.; Hrkach, J.; Shinoka, T.; Lien, J.; Moses, M.A.;
Stamp, A.; Taylor, G.; Moran, A.M.; Landis, W.; Langer, R.;
Vacanti, J.P.; Mayer, J.E., Jr. Tissue engineering of
autologous aorta using a new biodegradable polymer. Ann.
Thorac. Surg. 1999, 68, 2298–2304. 40. Berglund, J.D.;
Nerem, R.M.; Sambanis, A. Incorporation of intact elastin
scaffolds in tissue-engineered collagen-based vascular
grafts. Tissue Eng. 2004, 10, 1526–1535.
Hydrogels

12. Miyata, T.; Asami, N.; Uragami, T. A reversibly


antigen-responsive hydrogel. Nature 1999, 399, 766–769.

13. Tanaka, T.; Fillmore, D.J. Kinetics of swelling of


gels. J. Chem. Phys. 1979, 70, 1214–1218.

14. Chen, J.; Park, H.; Park, K. Synthesis of superporous


hydrogel: Hydrogels with fast swelling and superabsorbent
properties. J. Biomed. Mater. Res. 1999, 44, 53–62.

15. Kaneko, Y.; Sakai, K.; Kikuchi, A.; Yoshida, R.;


Sakurai, Y.; Okano, T. Influence of freely mobile grafted
chain length on dynamic properties of combtype grafted
poly(N-isopropyacrylamide) hydrogel. Macromolecules 1995,
28, 7717–7723.

16. Mukae, K.; Sakurai, M.; Sawamura, S.; Makino, K.; Kim,
S.W.; Ueda, I.; Shirahama, K. Swelling of poly
(N-isopropylacrylamide) gels in water-alcohol(C1–C4) mixed
solvents. J. Phys. Chem. 1993, 97, 737–741.

17. Park, K.; Shalaby, W.S.W.; Park, H. Physical Gels. In


Biodegradable Hydrogels for Drug Delivery; Technomic
Publishing Co.: Lancaster, PA, 1993; 99–140.

18. Shaheen, S.M.; Yamaura, K. Preparation of theophylline


hydrogels of atatic poly(vinyl alcohol)/NaCl/H 2 O system
for drug delivery system. J. Control. Release 2002, 81,
367–377.

19. Bezemer, J.M.; Radersma, R.; Grijpma, D.W.; Dijkstra,


P.J.; Feijen, J.; van Blitterswijk, C.A. Zero-order release
of lysozyme from poly(ethylene glycol)/poly (butylene
terephthalate) matrices. J. Control. Release 2000, 64,
179–192.

20. Molina, I.; Li, S.; Martinez, M.B.; Vert, M. Protein


release from physically crosslinked hydrogels of PMA/
PEO/PLA triblock copolymer-type. Biomaterials 2000, 22 (3),
363–369.

21. Bae, Y.H.; Vernon, B.; Han, C.K.; Kim, S.W.


Extracellular matrix for a rechargeable cell delivery
system. J. Control. Release 1998, 53, 249–258.

22. Andrianov, A.K.; Payne, L.G. Protein release from


polyphosphagene matrices. Adv. Drug Deliv. Rev. 1998, 31,
185–196. 23. Watanabe, T.; Ohtsuka, A.; Murase, N.; Barth,
P.; Gersonde, K. NMR studies on water and polymer diffusion
in dextran gels. Influence of potassium ions on
microstructure formation and gelation mechanism. Magn.
Reson. Med. 1996, 35, 697–705. 24. Holme, K.R.; Hall, L.D.
Chitosan derivatives bearing c10-alkyl glycoside branches:
A temperature-induced gelling polysaccharide.
Macromolecules 1991, 24, 3828–3833. 25. Tempel, M.;
Isenberg, G.; Sackmann, E. Temperatureinduced sol gel
transition and microgel formation in alpha-actinin
cross-linked actin networks—A rheological study. Phys. Rev.
E. 1996, 54 (2), 1802–1810. 26. Lehr, C.-M. Lectin-mediated
drug delivery: The second generation of bioadhesives. J.
Control. Release 2000, 65, 19–29. 27. Nam, K.; Watanabe,
J.; Ishihara, K. Characterization of the spontaneously
forming hydrogels composed of water-soluble phospholipid
polymers. Biomacromolecules 2002, 3 (1), 100–105. 28. Wang,
C.; Steward, R.J.; Kopecek, J. Hybrid hydrogels assembled
from synthetic polymers and coiled-coil protein domains.
Nature 1999, 397, 417–420. 29. Watanabe, J.; Eriguchi, T.;
Ishihara, K. Stereocomplex formation by enantiomeric
poly(lactic acid) graft-type phospholipid polymers for
tissue engineering. Biomacromolecules 2002, 3 (5),
1109–1114. 30. Watanabe, J.; Ooya, T.; Yui, N. Effect of
acetylation of biodegradable polyrotaxanes on its
supramolecular dissociation via terminal ester hydrolysis.
J. Biomater. Sci., Polym. Ed. 1999, 10 (12), 1275–1288. 31.
Ichi, T.; Watanabe, J.; Ooya, T.; Yui, N. Controllable
erosion time and profile in poly(ethylene glycol) hydrogels
by supramolecular structure of hydrolyzable polyrotaxane.
Biomacromolecules 2001, 2 (1), 204–210. 32. Taguchi, T.;
Kishida, A.; Akashi, M. A study on hydroxyapatite formation
on/in the hydroxyl groupsbearing nonioinic hydrogels. J.
Biomater. Sci., Polym. Ed. 1999, 10 (1), 19–32.
Hydroxyapatite

25. Barralet, J. Processing and sintering of carbonate


hydroxyapatite. Ph.D. Thesis. 1995, University of London.

26. Driessens, F.C.M.; Verbeeck, R.M.H.; Heijligers, H.J.M.


Some physical properties of Na� and CO 3 �containing
apatites synthesized at high temperatures. Inorganica.
Chemica. Acta 1983, 80, 19–23.

27. Emerson, W.H.; Fischer, E.D. The infrared absorption


spectra of carbonate in calcified tissue. Arch. Oral Biol.
1962, 7, 671–683.

28. Driessens, F.C.M.; Verbeeck, R.M.H.; Kiekens, P.


Mechanism of substitution in carbonated apatites.
Zeitschrift fur Anorganishe und allgemeine Chimie, 1983,
504, 195–200.

29. Barrelet, J.; Best, S.M.; Bonfield, W. Carbonate


substitution in precipitated hydroxyapatite: an
investigation into the effects of reaction temperature and
bicarbonate ion concentration. J. Biomed. Mater. Res. 1998,
41, 79–86.

30. Vignoles, M.; Bonel, G.; Holcomb, D.W.; Young, R.A.


Influence of preparation conditions on the composition of
type b carbonated hydroxyapatite and on the localization of
the carbonate ions. Calcif. Tissue. Int. 1998, 43, 3–40.

31. Gibson, I.R.; Bonfield, W. Novel synthesis and


charaterization of an AB-Type carbonate-substituted
hydroxyapatite. J. Biomed. Mater. Res. 2002, 59, 697–708.

32. Kim, S.R.; Lee, J.H.; Riu, D.H.; Jung, S.J.; Lee, Y.J.;
Chung, S.C.; Kim, Y.H. Synthesis of Si, Mg substituted
hydroxyapatites and their sintering behaviors. Biomaterials
2003, 24, 1389–1398.

33. Carlisle, E.M. Silicon: a possible factor in bone


calcification. Science 1970, 167, 279–280.

34. Shannon, R.D.; Prewitt, C.T. Effective ionic radii in


oxides and fluorides. Acta Cryst 1969, B25.

35. Porter, A.E.; Best, S.M.; Bonfield, W. Comparison of the


ultrastructure of hydroxyapatite and silicon-substituted
hydroxyapatite for biomedical applications. J. Biomed.
Mater. Res. 2003, 68A (1), 133–141.
36. Akao, M.; Aoki, H.; Kato, K. Mechanical properties of
sintered hydroxyapatite for prosthetic applications. J.
Mater. Sci. 1981, 28, 809.

37. Hayek, E.; Newesely, H. Pentacalcium.


Monohydroxyorthophosphate. Inorganic Syst. 1963, 7,
121–128.

38. LeGeros, R.Z.; LeGeros. Dense Hydroxyapatite. In


Introduction to Bioceramics; Hench, L.L., Wilson, J., Eds.;
World Scientific: Singapore, 1993; 139–180.

39. Monma, H.; Ueno, S.; Kanazawa, T. Properties of


hydroxyapatite prepared by the hydrolysis of tricalcium
phosphate. J. Chem. Tech. Biotechnol. 1981, 31, 15–24.

40. Rootare, H.M.; Craig, R.G. Characterization of


hydroxyapatite powders and compacts at room temperature and
after sintering at 12001C. J. Oral Rehab. 1978, 5, 293–307.

41. Fang, Y.; Agrawal, D.K.; Roy, D.M.; Roy, R. Fabrication


of porous hydroxyapatite ceramics by microwave. J. Biomed.
Mater. Res. 1981, 7 , 490–494.

42. Trombe, J.C.; Montel, G. Some features of the


incorporation of oxygen in different oxidation states in
the apatite lattice–I. On the existence of calcium and
strontium oxyapatites. Inorg. Nucl. Chem. 1978, 40, 15–21.
43. Wang, P.E.; Chaki, T.K. Sintering behaviour and
mechanical properties of hydroxyapatite and dicalcium
phosphate. J. Mater. Sci. Mater. Med. 1993, 4, 50–158. 44.
Aizawa, M.; Shinoda, H.; Uchida, H.; Itatani, K.; Okada,
I.; Matsumoto, M.; Morisue, H.; Matsumoto, H.; Toyama, Y.
Development and biological evaluation of apatite fiber
scaffolds with large pore size and high porosity for bone
regeneration. Key Eng. Mater. 2003, 240–242, 647–650. 45.
Aizawa, M.; Ueno, H.; Itatani, K.; Okada, I. Development of
scaffolds for tissue engineering using singlecrystal
apatite fibres and their biological evaluation by
osteoblastic cell. Trans. Mater. Res. Soc. Jpn. 2003, 28,
849–852. 46. Fernandez, A.; Planell, J.A.; Best, S.M.
Precipitation of carbonated apatite in the cement system
a-Ca 3 (PO 4 ) 2 Ca(H 2 PO 4 ) 2 -CaCO 3 . J. Biomed.
Mater. Res. 1999, 47, 466–471. 47. Hing, K.A.; Annaz, B.;
Saeed, S.; Revell, P.A.; Buckland, T. Microporosity
enhances bioactivity of synthetic bone graft substitutes.
J. Mater. Sci. Mater. Med. 2005, 16, 467–475. 48. Porter,
A.E.; Taak, P.; Hobbs, L.W.; Blunn, G.W.; Coathup, M.J.;
Spector, M. Bone bonding to hydroxyapatite and titanium
surfaces on human femoral stems retrieved at autopsy.
Biomaterials 2004, 25 (21), 5199–5208. 49. Bloebaum, R.D.;
Dupont, J.A. Osteolysis from a pressfit
hydroxyapatite-coated implant. A case study. J.
Arthroplasty 1993, 8 (2), 195–202. 50. van Dijk, K.;
Schaeken, H.G.; Wolke, J.C.; Maree, C.H.; Habraken, F.H.;
Verhoeven, J.; Jansen, J.A. Influence of discharge power
level on the properties of hydroxyapatite films deposited on
Ti6A14V with RF magnetron sputtering. J. Biomed. Mater.
Res. 1995, 29 (2), 269–276. 51. Thian, E.S.; Huang, J.;
Best, S.M.; Barber, Z.H.; Bonfield, W. A new way of
incorporating silicon in hydroxyapatite (Si-HA) as thin
films. J. Mater. Sci. Mater. Med. 2005, 16 (5), 411–415. . .
52. Porter, A.E.; Rea, S.M.; Galtrey, M.; Best, S.M.;
Barber, Z. Production of thin film silicon-doped
hydroxyapatite via sputter deposition. J. Mater. Sci.,
2004, 39, 1895–1898. 53. Kokubo, T.; Kushitani, H.; Sakka,
S. Solutions able to reproduce in vivo surface-structure
changes in bioactive glass-ceramic A-W. J. Biomed. Mater.
Res. 1990, 24, 721–734. . . 54. Yan, W.Q.; Nakamura, T.;
Kawanabe, K.; Nishigochi, S.; Oka, M.; Kokubo, T. Apatite
layer-coated titanium for use as bone bonding implants.
Biomaterials 1997, 18 (17), 1185–1190. 55. Lawson, A.C.;
Czernuszka, J.T. Collagen-calcium phosphate composites.
Proc. Inst. Mech. Engl. (H) 1998, 212 (6), 413–425. 56.
Song, J.; Malathong, W.; Bertozzi, C.R. Mineralization of
synthetic polymer scaffolds: a bottom-up approach for the
development of artificial bone. J. Am. Chem. Soc. 2005, 127
(10), 3366–3372. 57. Fernandez, E.; Gil, F.X.; Ginebra,
M.P.; Driessens, F.C.M.; Planell, J.A.; Best, S.M. Calcium
phosphate bone cements for clinical applications, Part I,
Solution Chemistry. J. Mater. Sci. Mater. Med. 1999, 10,
169–176. 58. Fernandez, E.; Gil, F.X.; Ginebra, M.P.;
Driessens, F.C.M.; Planell, J.A.; Best, S.M. Calcium
phosphate bone cements for clinical applications, Part II,
precipitate formation during setting reactions. J. Mater.
Sci. Mater. Med. 1999, 10, 177–184. 59. Hing, K.A.
Assessment of Porous Hydroxyapatite for Bone Replacement.
Ph.D. Thesis. University of London, 1996. 60. Hing, K.A.;
Best, S.M.; Tanner, K.E.; Bonfield, W.; Revell, P.A.
Mediation of bone ingrowth in porous hydroxyapatite bone
substitutes. J. Biomed. Mater. Res. 2004, 68, 187–200. 61.
Weng, J.; Liu, Q.; Wolke, J.G.C.; Zhang, X.; de Groot, K.
Formation and characteristics of the apatite layer on
plasma-sprayed hydroxyapatite coatings in simulated body
fluid. Biomaterials 1995, 18, 1027–1035. 62. Radin, S.R.;
Ducheyne, P. The effect of calcium phosphate ceramic
composition and structure on in vitro behavior. II.
Precipitation. J. Biomed. Mater. Res. 1993, 27, 35–45. 63.
Ducheyne, P.; Qiu, Q. Bioactive ceramics: The effect of
surface reactivity on bone formation and bone cell
function. Biomaterials 1999, 20, 2287–2303. 64. Barralet,
J.; Akao, M.; Aoki, H.; Aoki, H. Dissolution of dense
carbonate apatite subcutaneously implanted in Wistar Rats.
J. Biomed. Mater. Res. 1999, 49, 176–182. 65. Hankermeyer,
C.R.; Ohashi, K.L.; Delaney, D.C.; Ross, J.; Constantz,
B.R. Dissolution rates of carbonated hydroxyapatite in
hydrochloric acid. Biomaterials 2002, 23 (3), 743–750. 66.
Porter, A.E.; Hobbs, L.W.; Benezra Rosen, V.; Spector, M.
The ultrastructure of the plasma-sprayed
hydroxyapatite-bone interface predisposing to bone bonding.
Biomaterials 2002, 23, 725–733. 67. de Bruijn, J.D.; Flach,
J.S.; de Groot, K.; van Blitterswijk, C.A.; Davies, J.E.
Analysis of the bony interface with various types of
hydroxyapatite in vitro. Cells Mater. 1993, 3, 115–127. 68.
Layani, J.D.; Cuisnier, F.J.G.; Steuer, P.; Voegel, J.C.;
Mayer, I. High-resolution electron microscopy study of
synthetic carbonate and aluminum containing apatites. J.
Biomed. Mater. Res. 2000, 50, 199–207. 69. Webster, T.J.;
Ergun, C.; Doremus, R.H.; Siegel, R.W.; Bizios, R. Enhanced
osteoclast-like cell functions on nanophase ceramics.
Biomaterials 2001, 22, 1327–1333. 70. Klein, C.P.; de
Blieck-Hogervorst, J.M.; Wolke, J.G.; de Groot, K. Studies
of the solubility of different calcium phosphate ceramic
particles in vitro. Biomaterials 1990, 11 (7), 509–512. 71.
Gibson, I.R.; Best, S.M.; Bonfield, W. Effect of silicon
substitution on the sintering and microstructure of
hydroxyapatite. J. Am. Ceram. Soc. 2002, 85 (11), 2771–277.
72. Porter, A.E.; Patel, N.; Skepper, J.N.; Best, S.M.;
Bonfield, W. Effect of sintered silicate-substituted
hydroxyapatite on remodelling processes at the bone-implant
interface. Biomaterials 2004, 25 (16), 3303–3314. 73.
Porter, A.E.; Patel, N.; Skepper, J.N.; Best, S.M.;
Bonfield, W. Comparison of in vivo dissolution processes in
hydroxyapatite and silicon-substituted hydroxyapatite
bioceramics. Biomaterials 2003, 24, 4609–4620. H
Hydroxyapatite Coatings

1. Lemons, J.E. Hydroxyapatite coatings. Clin. Orthp. Rel.


Res. 1988, 235, 220–223. 2. Lacefield, W.R. Hydroxylapatite
coatings. In Bioceramics: Material Characteristics Versus
In vivo Behavior; Ducheyne, P., Lemons, J.E., Eds.; The New
York Academy of Science: New York, 1988; 72–80. 3.
Ducheyne, P.; Cuckler, J.M. Bioactive ceramic prosthetic
coatings. Clin. Orthop. Rel. Res. 1992, 276, 102–114. 4.
Furlong, R.J.; Osborn, J.F. Fixation of hip prostheses by
hydroxyapatite ceramic coatings. J. Bone Joint Surg. 1991,
73B, 741–745. 5. Geesink, R.G.T. Osteoconductive coating
for total joint arthroplasty. Clin. Orthop. Rel. Res. 2002,
395, 53–65. 6. Havelin, L.I.; Engesaeter, L.B.; Espehaug,
B.; Furnes, O.; Lie, S.A.; Vollset, S.E. The Norweigian
arthroplasty register, 11 years and 73,000 arthroplasties.
Acta Orthop. Scand. 2000, 71, 337–353. 7. D’Antonio, J.A.;
Capello, W.N.; Manley, M.T.; Geesink, R. Hydroxyapatite
femoral stems for total hip arthroplasty. Clin. Orthop.
Rel. Res. 2001, 393, 101–111. 8. de Groot, K.; Wolke,
J.G.C.; Jansen, J.A. State of the art: hydroxyapatite
coatings for dental implants. J. Oral Implantol. 1994, 20,
232–234. 9. Yang, Y.; Ong, J.L.; Bessho, K. Plasma-sprayed
hydroxyapatite-coated and plasma-sprayed titanium-coated
implants. In Biomaterials in Orthopedics; Yaszemski, M.J.,
Trantolo, D.J., Lewandrowski, K., Hasirci, V., Altobelli,
D.E., Wise, D.L., Eds.; Marcel Dekker, Inc.: New York,
2004; 401–423. 10. Yang, Y.; Kim, K.H.; Ong, J.L. A review
on calcium phosphate coatings produced using a sputtering
process—an alternative to plasma spraying. Biomaterials
2005, 26, 327–337. 11. Manley, M.T.; Capello, W.N.;
D’Anotnio, J.A.; Geesink, R.G.T. Fixation of acetabular
cups without cement in total hip arthroplasty. J. Bone
Joint Surg. 1998, 80A, 1175–1185. 12. Adell, R.; Lekholm,
V.; Rockler, B. A 15-year study of osseointegrated implants
in the treatment of the edentulous jaw. Int. J. Oral Surg.
1981, 10, 387–416. 13. Furlong, R. Proximal femoral bone
loss and increased rate of fracture with a proximally
hydroxyapatitecoated femoral component. J. Bone Joint Surg.
2001, 83B, 46. 14. Tonio, A.J.; Therin, M.; Doyle, C.
Hydroxyapatitecoated femoral stems. Histoloy and
histomorphometry around five components retrieved at post
mortem. J. Bone Joint Surg. 1999, 81B, 148–154. 15. Bauer,
T.W.; Stulberg, B.N.; Jiang, M.; Geesink, R.G.T. Uncemented
acetabular components: histologic analysis of retrieved
components. J. Arthoplasty 1993, 8, 167–177. 16. Adams,
C.S.; Mansfield, K.; Perlot, R.L.; Shapiro, I.M. Matrix
regulation of skeletal cell apoptosis. Role of calcium and
phosphate ions. J. Biol. Chem. 2001, 276, 20,316–20,322.
17. Lacefield, W.R. Characterization of hydroxyapatite
coatings. J. Oral Implantol. 1994, 20, 214–220. 18.
Soballe, K.; Hansen, E.S.; Brockstedt-Rasmussen, H.;
Burgess, A. Torsional stability of HA-coated and grit
blasted Ti dental implants. J. Bone Joint Surg. 1993, 75B,
270–278. 19. Driessens, F.C.M.; Boltong, M.G.; Khairoun,
I.; De Maeyer, E.A.P.; Ginebra, M.P.; Wenz, R.; Planell,
J.A.; Verbeeck, R.M.H. Applied aspects of calcium phosphate
bone cement application. In Biomaterials Engineering and
Devices. Human Applications. Volume 2; Wise, D.L.,
Trantolo, D.J., Lewandrowski, K.U., Gresser, M.V.,
Yaszemski, M.J., Eds.; Humana Press: New Jersey, Totowa,
2000; 253–260. 20. Huang, J.; Best, S.M.; Bonfield, W.;
Brooks, R.A.; Rushton, N.; Jayasinghe, S.N.; Edirisinghe,
M.J. In vitro assessment of the biological response to
nano-sized hydroxyapatite. J. Mater. Sci. Mater. Med. 2004,
15, 441–445. H
Implant, Total Hip

Fig. 5 Polarized light micrograph (190� ) of paraaortic

lymph node demonstrates abundance and morphology of

birefringent polyethylene particles within macrophages. 9.


Charnley, J. The bonding of prosthesis to bone by cement.
J. Bone Jt. Surg., Br. 1964, 46, 518. 10. Wilson, J.N.;
Scales, J.T. Loosening of total hip replacements with
cement fixation. Clinical findings and laboratory studies.
Clin. Ortop. 1970, 72, 145–160. 11. Charnley, J. Anchorage
of the femoral head prosthesis to the shaft of the femur.
J. Bone Jt. Surg., Br. 1960, 42, 28. 12. Ring, P.A.
Complete replacement arthroplasty of the hip by the ring
prosthesis. J. Bone Jt. Surg., Br. 1968, 50, 720. 13.
Black, J. Orthopaedic Biomaterials in Research and
Practice; Churchill Livingstone: New York, 1988. 14.
Jacobs, J.J.; Gilbert, J.L.; Urban, R.M. Corrosion of
Metallic Implants. In Advances in Orthopaedic Surgery Vol.
2; Stauffer, R.N., Ed.; Mosby: St. Louis, 1994. 15. Jacobs,
J.J.; Skipor, A.K.; Urban, R.M.; Black, J.; Manion, L.M.;
Starr, A.; Talbert, L.F.; Galante, J.O. Systemic
distribution of metal degradation products from titanium
alloy total hip replacements: An autopsy study. Trans.
Orthop. Res. Soc. (New Orleans) 1994, 838. 16. Jacobs,
J.J.; Silverton, C.; Hallab, N.J.; Skipor, A.K.; Patterson,
L.; Black, J.; Galante, J.O. Metal release and excretion
from cementless titanium alloy total knee replacements.
Clin. Ortop. 1999, 358, 173–180. 17. Jacobs, J.J.; Skipor,
A.K.; Patterson, L.M.; Hallab, N.J.; Paprosky, W.G.; Black,
J.; Galante, J.O. Metal release in patients who have had a
primary total hip arthroplasty. A prospective, controlled,
longitudinal study. J. Bone Jt. Surg., Am. 1998, 80,
1447–1458. 18. Urban, R.B.; Jacobs, J.; Gilbert, J.L.;
Rice, S.B.; Jasty, M.; Bragdon, C.R.; Galante, G.O.
Characterization of Solid Products of Corrosion Generated
by ModularHead Femoral Stems of Different Designs
andMaterials. In STP 1301 Modularity of Orthopedic
Implants; Marlowe, D.E., Parr, J.E., Mayor, M.B., Eds.;
ASTM: Philadelphia, 1997. 19. Urban, R.M.; Jacobs, J.J.;
Tomlinson, M.J.; Gavrilovic, J.; Black, J.; Peoc’h, M.
Dissemination of wear particles to the liver, spleen, and
abdominal lymph nodes of patients with hip or knee
replacement. J. Bone Jt. Surg., Am. 2000, 82, 457–476. 20.
Urban, R.M.; Jacobs, J.J.; Gilbert, J.L.; Galante, J.O.
Migration of corrosion products from modular hip
prostheses. Particle microanalysis and histopathological
findings. J. Bone Jt. Surg., Am. 1994, 76, 1345–1359. 21.
Merle, C.; Vigan, M.; Devred, D.; Girardin, P.; Adessi, B.;
Laurent, R. Generalized eczema from vitallium
osteosynthesis material. Contact Dermatitis 1992, 27,
257–258. I
Implanted Medical Devices, Power Sources
For

10. Mead, R.T.; Greatbatch, W.; Rudolph, F.W. Lithium–


Iodine Battery Having Coated Anode. US Patent 3,957,533,
May 18, 1976.

11. Gabano, J.-P.; Broussley, M.; Grimm, M. Lithium Solid


Cathode Batteries for Biomedical Implantable Applications.
In Batteries for Implantable Biomedical Devices; Owens,
B.B., Ed.; Plenum Press: New York, 1986; 181–212.

12. Skarstad, P.M. Lithium–Liquid Oxidant Batteries. In


Batteries for Implantable Biomedical Devices; Owens, B.B.,
Ed.; Plenum Press: New York, 1986; 215–225.

13. Holmes, C.F. The bourner lecture: Electrochemical power


sources—An important contributor to modern health care. J.
Power Sources 1997, 65, xv–xx.

14. Mirowski, M.; Mower, M.M.; Reid, P.R. The automatic


implantable defibrillator. Am. Heart J. 1980, 100,
1089–1092.

15. Horning, R.J.; Viswanathan, S. High Rate Lithium Cell


for Medical Application. In Proc. 29th Power Sources Conf.,
Atlantic City, N.J.; The Electrochemical Society:
Pennington, NJ, 1980; 64–66.

16. Liang, C.C.; Bolster, M.E.; Murphy, R.M. Metal Oxide


Composite Cathode Material for High Energy Density
Batteries. US Patent 4,310,609, January 12, 1982.

17. Liang, C.C.; Bolster, M.E.; Murphy, R.M. Metal Oxide


Composite Cathode Material for High Energy Density
Batteries. US Patent 4,391,729, July 5, 1983.

18. Schmidt, C.L.; Skarstad, P.M. Development of a


Physically-Based Model for the Lithium–Iodine Battery. In
Power Sources 13; Keily, T., Ed.; International Power
Sources Symposium Committee: Leatherhead, UK, 1991;
347–362.

19. Crespi, A.M.; Schmidt, C.L.; Norton, J.; Chen, K.;


Skarstad, P.M. Modeling and characterization of the
resistance of lithium/SVO batteries for implantable
cardioverter defibrillators. J. Electrochem. Soc. 2001, 148,
A30–A37.

20. Schmidt, C.L.; Skarstad, P.M. The future of lithium and


lithium–ion batteries in implantable medical devices. J.
Power Sources 2001, 97–98, 742–746.

21. Modern Batteries, 2nd Ed.; Vincent, C.A., Scrosati, B.,


Eds.; Arnold: London, 1997.

22. Handbook of Batteries, 3rd Ed.; Linden, D., Reddy,


T.B., Eds.; McGraw-Hill: New York, 2002.

23. Untereker, D.F.; Shepard, R.B.; Schmidt, C.L.; Crespi,


A.M.; Skarstad, P.M. Power Systems for Implantable
Pacemakers, Cardioverters and Defibrillators. In Clinical
Cardiac Pacing and Defibrillation, 2nd Ed.; Ellenbogen,
K.A., Kay, G.N., Wilkoff, B.L., Eds.; W.B. Saunders Co.:
Philadelphia, 2000; 167–193. 24. Batteries for Implantable
Biomedical Devices; Owens, B.B., Ed.; Plenum Press: New
York, 1986. 25. Greatbatch, W.; Holmes, C.F. The
lithium–iodine battery: A historical perspective. PACE
1992, 15, 2034– 2036. 26. Bard, A.J.; Faulkner, L.R.
Electrochemical Methods; John Wiley and Sons: New York,
1980; 103. 27. Schmidt, C.L.; Skarstad, P.M. Development of
an equivalent-circuit model for the lithium/iodine battery.
J. Power Sources 1997, 65, 121–128. 28. Schmidt, C.L.;
Skarstad, P.M. Development of a Physically Based Model for
the Lithium–Iodine Battery. In Power Sources 13; Keily, T.,
Baxter, B.W., Eds.; International Power Sources Symposium
Committee: Leatherhead, England, 1991; 347–361. 29.
MacLean, G.K.; Aiken, P.A.; Adams, W.A.; Mussivand, T.
Preliminary evaluation of rechargeable lithium–ion cells
for an implantable battery pack. J. Power Sources 1995, 56,
69–74. 30. Okamoto, E.; Watanabe, K.; Hashiba, K.; Inoue,
T.; Iwazawa, E.; Momoi, M.; Hashimoto, T.; Mitamura, Y.
Optimum selection of an implantable secondary battery for
an artificial heart by examination of the cycle life test.
ASAIO J. 2002, 48, 495–502. 31. Sunderland, W.C.; Rorvick,
A.W.; Merritt, D.R.; Schmidt, C.L.; Haas, D.P.
Electrochemical Cell. US Patent 5,716,729, February 10,
1998. 32. Elliot, W.B.; Paulot, W.B.; Smesko, S.A.
Electrode Edge Design. May 18, 1999. 33. Howard, W.G.;
Kelm, R.W.; Weiss, D.J.; Crespi, A.M.; Berkowitz, F.J.;
Skarstad, P.M. High Reliability Electrochemical Cell and
Electrode Assembly Therefore. US Patent 5,439,760, August
8, 1995. 34. Keister, P.P.; Mead, R.T.; Muffoletto, B.C.;
Takeuchi, E.S.; Ebel, S.J.; Zelinsky, M.A.; Greenwood, J.M.
Non-aqueous Lithium Battery. US Patent 4,830,940, May 16,
1989. 35. Takeuchi, E.S.; Quattrini, P.J.; Greatbatch, W.
Lithium/ silver vanadium oxide batteries for implantable
defibrillators. Pacing Clin Electrophysiol. 1988, 11 (11),
2035–2039. 36. Bates, J.B.; Dudney, N.J.; Neudecker, B.;
Ueda, A.; Evans, C.D. Thin-film lithium and lithium–ion
batteries. Solid State Ionics 2000, 135, 33–45. 37. Chen,
T.; Barton, S.C.; Binyamin, G.; Gao, Z.; Zhang, Y.; Kim,
H.H.; Heller, A. A miniature biofuel cell. J. Am. Chem.
Soc. 2001, 123, 8630–8631.
Infection of Medical Devices

33. Reid, G.; Habash, M.; Vachon, D.; Denstedt, J.;


Riddell, J.; Beheshti, M. Oral fluoroquinolone therapy
results in drug adsorption on ureteral stents and
prevention of biofilm formation. Int. J. Antimicrob. Agents
2001, 17 (4), 317–319. Discussion 319–320.

34. Levin, A.; Mason, A.J.; Jindal, K.K.; Fong, I.W.;


Goldstein, M.B. Prevention of hemodialysis subclavian vein
catheter infections by topical povidone-iodine. Kidney Int.
1991, 40 (5), 934–938.

35. Sesso, R.; Barbosa, D.; Leme, I.L.; Sader, H.;


Canziani, M.E.; Manfredi, S.; Draibe, S.; Pignatari, A.C.
Staphylococcus aureus prophylaxis in hemodialysis patients
using central venous catheter: Effect of mupirocin
ointment. J. Am. Soc. Nephrol. 1998, 9 (6), 1085–1092.

36. Lok, C.E.; Stanley, K.E.; Hux, J.E.; Richardson, R.;


Tobe, S.W.; Conly, J. Hemodialysis infection prevention
with polysporin ointment. J. Am. Soc. Nephrol. 2003, 14
(1), 169–179.

37. Sedor, J.; Mulholland, S.G. Hospital-acquired urinary


tract infections associated with the indwelling catheter.
Urol. Clin. North Am. 1999, 26 (4), 821–828.

38. Hedlund, H.; Hjelmas, K.; Jonsson, O.; Klarskov, P.;


Talja, M. Hydrophilic versus non-coated catheters for
intermittent catheterization. Scand J. Urol. Nephrol. 2001,
35 (1), 49–53.

39. Fader, M.; Moore, K.N.; Cottenden, A.M.; Pettersson,


L.; Brooks, R.; Malone-Lee, J. Coated catheters for
intermittent catheterization: Smooth or sticky? BJU Int.
2001, 88 (4), 373–377.

40. Johnson, J.R.; Delavari, P.; Azar, M. Activities of a


nitrofurazone-containing urinary catheter and a silver
hydrogel catheter against multidrug-resistant bacteria
characteristic of catheter-associated urinary tract
infection. Antimicrob. Agents Chemother. 1999, 43 (12),
2990–2995.

41. Darouiche, R.O.; Safar, H.; Raad, I.I. In vitro efficacy


of antimicrobial-coated bladder catheters in inhibiting
bacterial migration along catheter surface. J. Infect. Dis.
1997, 176 (4), 1109–1112.
42. Liedberg, H.; Lundeberg, T. Silver alloy coated
catheters reduce catheter-associated bacteriuria. Br. J.
Urol. 1990, 65 (4), 379–381.

43. Stickler, D.; Hewett, P. Activity of antiseptics


against biofilms of mixed bacterial species growing on
silicone surfaces. Eur. J. Clin. Microbiol. Infect. Dis.
1991, 10 (3), 157–162.

44. Nickel, J.C.; Downey, J.; Costerton, J.W. Movement of


Pseudomonas aeruginosa along catheter surfaces. A mechanism
in pathogenesis of catheter-associated infection. Urology
1992, 39 (1), 93–98.

45. Pugach, J.L.; DiTizio, V.; Mittelman, M.W.; Bruce,


A.W.; DiCosmo, F.; Khoury, A.E. Antibiotic hydrogel coated
Foley catheters for prevention of urinary tract infection
in a rabbit model. J. Urol. 1999, 162 (3 Pt 1), 883–887.

46. Niel-Weise, B.S.; Arend, S.M.; van den Broek, P.J. Is


there evidence for recommending silver-coated urinary
catheters in guidelines? J. Hosp. Infect. 2002, 52 (2),
81–87. 47. Marciante, K.D.; Veenstra, D.L.; Lipsky, B.A.;
Saint, S. Which antimicrobial impregnated central venous
catheter should we use? Modeling the costs and outcomes of
antimicrobial catheter use. Am. J. Infect. Control 2003, 31
(1), 1–8. 48. Rediske, A.M.; Roeder, B.L.; Brown, M.K.;
Nelson, J.L.; Robison, R.L.; Draper, D.O.; Schaalje, G.B.;
Robison, R.A.; Pitt, W.G. Ultrasonic enhancement of
antibiotic action on Escherichia coli biofilms: An in vivo
model. Antimicrob. Agents Chemother. 1999, 43 (5),
1211–1214. 49. Khoury, A.E.; Lam, K.; Ellis, B.; Costerton,
J.W. Prevention and control of bacterial infections
associated with medical devices. Asaio. J. 1992, 38 (3),
M174–M178. 50. Stewart, P.S.; Wattanakaroon, W.; Goodrum,
L.; Fortun, S.M.; McLeod, B.R. Electrolytic generation of
oxygen partially explains electrical enhancement of
tobramycin efficacy against Pseudomonas aeruginosa biofilm.
Antimicrob. Agents Chemother. 1999, 43 (2), 292–296. 51.
Woo, G.L.; Yang, M.L.; Yin, H.Q.; Jaffer, F.; Mittelman,
M.W.; Santerre, J.P. Biological characterization of a novel
biodegradable antimicrobial polymer synthesized with
fluoroquinolones. J. Biomed. Mater. Res. 2002, 59 (1),
35–45. 52. Hentzer, M.; Riedel, K.; Rasmussen, T.B.;
Heydorn, A.; Andersen, J.B.; Parsek, M.R.; Rice, S.A.;
Eberl, L.; Molin, S.; Hoiby, N.; Kjelleberg, S.; Givskov,
M. Inhibition of quorum sensing in Pseudomonas aeruginosa
biofilm bacteria by a halogenated furanone compound.
Microbiology 2002, 148 (Pt 1), 87–102. 53. Balaban, N.;
Goldkorn, T.; Nhan, R.T.; Dang, L.B.; Scott, S.; Ridgley,
R.M.; Rasooly, A.; Wright, S.C.; Larrick, J.W.; Rasooly,
R.; Carlson, J.R. Autoinducer of virulence as a target for
vaccine and therapy against Staphylococcus aureus. Science
1998, 280 (5362), 438–440. 54. Singh, P.K.; Parsek, M.R.;
Greenberg, E.P.; Welsh, M.J. A component of innate immunity
prevents bacterial biofilm development. Nature 2002, 417
(6888), 552–555. 55. Lyte, M.; Freestone, P.P.; Neal, C.P.;
Olson, B.A.; Haigh, R.D.; Bayston, R.; Williams, P.H.
Stimulation of Staphylococcus epidermidis growth and biofilm
formation by catecholamine inotropes. Lancet 2003, 361
(9352), 130–135. 56. Nagino, K.; Kobayashi, H. Influence of
macrolides on mucoid alginate biosynthetic enzyme from
Pseudomonas aeruginosa. Clin. Microbiol. Infect. 1997, 3
(4), 432–439. 57. van der Mei, H.C.; Free, R.H.; Elving,
G.J.; Van Weissenbruch, R.; Albers, F.W.; Busscher, H.J.
Effect of probiotic bacteria on prevalence of yeasts in
oropharyngeal biofilms on silicone rubber voice prostheses
in vitro. J. Med. Microbiol. 2000, 49 (8), 713–718. 58.
Free, R.H.; Busscher, H.J.; Elving, G.J.; van der Mei,
H.C.; van Weissenbruch, R.; Albers, F.W. Biofilm formation
on voice prostheses: In vitro influence of probiotics. Ann.
Otol. Rhinol. Laryngol. 2001, 110 (10), 946–951.
Inner Ear Implants

1. Clark, G.M. Sounds from Silence; Allen & Unwin: Sydney,


2000.

2. Clark, G.M. Cochlear Implants: Fundamentals and


Applications; Springer-Verlag: New York. in press.

3. Clark, G.M. Electrical stimulation of the auditory


nerve: The coding of frequency, the perception of pitch and
the development of cochlear implant speech processing
strategies for profoundly deaf people. Clin. Exp.
Pharmacol. Physiol. 1996, 23, 766–776.

4. Clark, G.M. Cochlear Implants. In Diseases of the Ear,


1st Ed.; Wright, A., Ludman, H., Eds.; Edward Arnold:
London, 1998; 149–163.

5. Clark, G.M. Cochlear implants: Climbing new mountains.


The Graham Fraser memorial lecture 2001. Cochlear Implants
Int. 2001, 2, 75–97.

6. Clark, G.M. Learning to Understand Speech with the


Cochlear Implant. In Perceptual Learning, 1st Ed.; Fahle,
M., Poggio, T., Eds.; MIT Press: Cambridge, MA, 2002;
147–160.

7. Surgical Procedure Manual: Nucleus 22 Channel Cochlear


Implant System, Issue 5; Cochlear Corporation: Englewood,
CO, 1987.

8. Shepherd, R.K.; Franz, B.K-H.; Clark, G.M. The


Biocompatibility and Safety of Cochlear Prostheses. In
Cochlear Prostheses, 1st Ed.; Clark, G.M., Tong, Y.C.,
Patrick, J.F., Eds.; Churchill Livingstone: London, 1990;
69–98.

9. Clark, G.M. The cochlear implant: A search for answers.


Cochlear Implants Int. 2000, 1, 1–17.

FURTHER READING

More detailed information is available in the following

texts:

Clark, G.M. The University of Melbourne—Nucleus


Multi-Electrode Cochlear Implant; Advances in
OtoRhino-Laryngology, Karger: Basel, 1987; Vol. 38.
Cochlear Implantation for Infants and Children—Advances;
Clark, G.M., Cowan, R.S.C., Dowell, R.C., Eds.; Singular
Publishing Group Inc.: San Diego, 1997.
Insulin Delivery

6. Still, J.G. Development of oral insulin: Progress and


current status. Diabetes/Metab. Res. Rev. 2002, 18 (Suppl.
1), S29–S37.

7. Lowman, A.M.; Morishita, M.; Kajita, M.; Nagai, T.;


Peppas, N.A. Oral delivery of insulin using pHresponsive
complexation gels. J. Pharm. Sci. 1999, 88, 933–937.

8. Lee, Y.-C.; Simamora, P.; Pinsuwan, S.; Yalkowsky, S.H.


Review on the systemic delivery of insulin via the ocular
route. Int. J. Pharm. 2002, 233, 1–18.

9. Modi, P.; Mihic, M.; Lewin, A. The evolving role of oral


insulin in the treatment of diabetes using a novel
RapidMistt system. Diabetes/Metab. Res. Rev. 2002, 18
(Suppl. 1), S38–S42.

10. Patton, J.S. Deep-lung delivery of therapeutic


proteins. Chemtech 1997, 27, 34–38.

11. Bindra, S.; Rosenstock, J.; Cefalu, W.T. Inhaled


insulin: A novel route for insulin delivery. Expert Opin.
Investig. Drugs 2002, 11, 687–691.

12. Klonoff, D.C. Inhaled insulin. Diabetes Technol. Ther.


1999, 1, 307–313.

13. Kost, J. Ultrasound-assisted insulin delivery and


noninvasive glucose sensing. Diabetes Technol. Ther. 2002,
4, 489–497.

14. Scheuplein, R.J.; Blank, I.H. Permeability of the skin.


Physiol. Rev. 1971, 51, 702–747.

15. Prausnitz, M.R. Overcoming skin’s barrier: The search


for effective and user-friendly drug delivery. Diabetes
Technol. Ther. 2001, 3, 233–236.

16. Dailey, G.E.; Boden, G.H.; Creech, R.H.; Johnson, D.G.;


Gleason, R.E.; Kennedy, F.P.; Weinrauch, L.A.; Weir, M.;
D’Elia, J.A. Effects of pulsatile intravenous insulin
therapy on the progression of diabetic nephropathy.
Metabolism 2000, 49, 1491–1495.

17. DCCT—The Diabetes Control and Complications Trial


Research Group. The effect of intensive treatment of
diabetes on the development and progression of long-term
complications in insulin-dependent diabetes mellitus. N.
Engl. J. Med. 1993, 329, 977–986.

18. DCCT—The Diabetes Control and Complications Trial


Research Group. The absence of a glycemic threshold for the
development of long-term complications: The perspective of
the diabetes control and complications trial. Diabetes
1996, 45, 1289–1298.

19. Pfeifer, M.A.; Halter, J.B.; Porte, D., Jr. Insulin


secretion in diabetes mellitus. Am. J. Med. 1981, 70,
579–588.

20. Gerich, J.E. Novel insulins: Expanding options in


diabetes mellitus. Am. J. Med. 2002, 113, 308–316.

21. Bolli, G.B. Physiological insulin replacement in type 1


diabetes mellitus. Exp. Clin. Endocrinol. Diabetes 2001,
109 (Suppl. 2), S317–S332.

22. Bayer Corporation Glucometer s dex diabetes care system


2003. http://wwwbayerdiag.com/products/pages
/self/glucometerdex.html.

23. Roche Diagnostics Accu-chek s test strips 2003. http://


wwwaccu-chek.com/products/main.cfm?pid=3000.

24. Potts, R.O.; Tamada, J.A.; Tierney, M.J. Glucose


monitoring by reverse iontophoresis. Diabetes/Metab. Res.
Rev. 2002, 1, S49–S53. 25. Steil, G.M.; Rebrin, K.;
Mastrototaro, J.; Bernaba, B.; Saad, M. Determination of
plasma glucose during rapid glucose excursions with a
subcutaneous glucose sensor. Diabetes Technol. Ther. 2003,
5, 27–31. 26. Eli Lilly and Company Products 2003.
http://www.lilly. com/products.html. 27. Novo Nordisk
Insulin 2003.
http://www.novonordiskus.com/view.asp?ID=1177. 28. Becton,
Dickinson, and Company BD pharmaceutical systems 2003.
http://www.bd.com/pharmaceuticals/. 29. Medtronic MiniMed
Which pump is for you? 2003.
http://www.minimed.com/patientfam/pf_ipt_paradigm_
whichpump.shtml. 30. Lacy, P. Treating diabetes with
transplanted cells. Sci. Am. 1995, 273, 50–58. 31. Weir,
G.C.; Bonner-Weir, S. Scientific and political impediments
to successful islet transplation. Diabetes 1997, 46,
1247–1256. 32. Siebers, U.; Horcher, A.; Bretzel, R.G.;
Federlin, K.; Zekorn, T. Alginate-based microcapsules for
immunoprotected islet transplantation. Ann. N.Y. Acad. Sci.
1997, 831, 304–312. 33. Reach, G. Islet transplantation: A
field on the move. Nephrol. Dial. Transplant. 2001, 16,
893–896. 34. Pericin, M.; Althage, A.; Freigang, S.;
Hengartner, H.; Rolland, E.; Dupraz, P.; Thorens, B.;
Aebischer, P.; Zinkernagel, R.M. Allogenic b-islet cells
correct diabetes and resist immune rejection. Proc. Natl.
Acad. Sci. 2002, 99, 8203–8206. 35. Duvivier-Kali, V.F.;
Omer, A.; Parent, R.J.; O’Neil, J.J.; Weir, G.C. Complete
protection of islets against allorejection and autoimmunity
by a simple barium-alginate membrane. Diabetes 2001, 50,
1698–1705. 36. Omer, A.; Duvivier-Kali, V.F.; Trivedi, N.;
Wilmot, K.; Bonner-Weir, S.; Weir, G.C. Survival and
maturation of microencapsulated porcine neonatal pancreatic
cell clusters transplanted into immunocompetent diabetic
mice. Diabetes 2003, 52, 69–75. 37. Colton, C.K.;
Avgoustiniatos, E.S. Bioengineering in development of the
hybrid artificial pancreas. J. Biomech. Eng. 1991, 113,
152–170. 38. Trivedi, N.; Keegan, M.; Steil, G.M.;
Hollister-Lock, J.; Hasenkamp, W.M.; Colton, C.K.;
Bonner-Weir, S.; Weir, G.C. Islets in alginate macrobeads
reverse diabetes despite minimal acute insulin secretory
responses. Transplantation 2001, 71, 203–211. 39. Hegre,
O.D.; Lacy, P.E.; Dionne, K.E.; Gentile, F.; Aebischer, P.;
Laurance, M.; Fiore, D.; Gardiner, A.; Hazlett, T.;
Purzycki, M. Macroencapsulation of islets of Langerhans.
Diabet. Nutr. Metab. 1992 , 5 (Suppl. 1), 159–162. 40.
Albin, G.; Horbett, T.; Ratner, B. Glucose-Sensitive
Membranes for Controlled Release of Insulin. In Pulsed and
Self-Regulated Drug Delivery; CRC Press, 1990; 160–185. 41.
Albin, G.; Horbett, T.; Ratner, B. Glucose sensitive
membranes for controlled delivery of insulin: Insulin
transport studies. J. Control. Release 1985, 2, 153–164.
42. Cartier, S.; Horbett, T.; Ratner, B. Glucose-sensitive
membrane coated porous filters for control of hydraulic
permeability and insulin delivery from a pressurized
reservior. J. Membr. Sci. 1995, 106, 17–24. 43. Klumb, L.;
Horbett, T. Design of insulin delivery devices based on
glucose sensitive membranes. J. Control. Release 1992, 18,
59–80. 44. Siegel, R.; Firestone, B. Mechanochemical
approaches to self-regulating insulin pump design. J.
Control. Release 1990, 11, 181–192. 45. Goldraich, M.;
Kost, J. Glucose-sensitive polymeric matrices for
controlled drug delivery. Clin. Mater. 1993, 13, 135–142.
46. Lowman, A.; Peppas, N. Complexation graft copolymers as
oral drug delivery systems. Polym. Prepr. 1997, 38,
566–567. 47. Hassan, C.; Doyle III, F.J.; Peppas, N.
Dynamic behavior of glucose-responsive poly(methacrylic
acid-gethylene glycol) hydrogels. Macromolecules 1997, 30,
6166–6173. 48. Kerner, W. Implantable glucose sensors:
Present status and future developments. Exp. Clin.
Endocrinol. Diabetes 2001, 109 (Suppl. 2), S341–S346. 49.
Maran, A.; Cerpaldi, C.; Tiengo, A.; Grassi, G.; Vitali,
E.; Pagaon, G.; Bistoni, S.; Calabrese, G.; Santeusanio,
F.; Leonetti, F.; Ribaudo, M.; DiMario, U.; Annuzzi, G.;
Genovese, S.; Riccardi, G.; Prefiti, M.; Cucinotta, D.;
Giorgino, F.; Bellomo, A.; Biorgino, R.; Poscia, A.;
Varalli, M. Continuous subcutaneous glucose monitoring in
diabetic patients: A multicenter analysis. Diabetes Care
2002, 25, 347–352. 50. Morari, M.; Zafiriou, E. Robust
Process Control; Prentice-Hall: Englewood Cliffs, NJ, 1989.
51. Parker, R.S.; Doyle III, F.J. Control-relevant modeling
in drug delivery. Adv. Drug Deliv. Rev. 2001, 48, 211–228.
52. Bergman, R.; Phillips, L.; Cobelli, C. Physiologic
evaluation of factors controlling glucose tolerance in man.
J. Clin. Invest. 1981, 68, 1456–1467. 53. Parker, R.S.;
Ward, J.H.; Peppas, N.A.; Doyle III, F.J. Robust H-infinity
glucose control in diabetes using a physiological model.
AIChE J. 2000, 46, 2537–2549. 54. Sorensen, J. A
Physiologic Model of Glucose Metabolism in Man and Its Use
to Design and Assess Improved Insulin Therapies for
Diabetes; Ph.D. ThesisDepartment of Chemical Engineering
MIT, 1985. 55. Parker, R.S.; Doyle III, F.J.; Peppas, N.A.
A modelbased algorithm for blood glucose control in type I
diabetic patients. IEEE Trans. Biomed. Eng. 1999, 46 (2),
148–157. 56. Yu, C.L.; Roy, R.J.; Kaufman, H.; Bequette,
B.W. Multiple-model adaptive predictive control of mean
arterial blood pressure. IEEE Trans. Biomed. Eng. 1992, 39,
765–778. 57. Linkens, D.A.; Mahfouf, M. Generalised
Predictive Control (GPC) in Clinical Anaesthesia. In
Advances in Model-Based Predictive Control; Clarke, D.,
Ed.; Oxford University Press, 1994; 429–445. 58. Parker,
R.S.; Gatzke, E.P.; Doyle III, F.J. Advanced Model
Predictive Control [MPC] for Type I Diabetic Patient Blood
Glucose Control. In Proc. American Control Conf.; , 2000;
Vol. 5, 3483–3487. 59. Tyler, M.L.; Morari, M.
Propositional logic in control and monitoring problems.
Automatica 1999, 35, 565–582. 60. Bolie, V.W. Coefficients
of normal blood glucose regulation. J. Appl. Physiol. 1961,
16, 783–788. I
Integrins

28. Ruoslahti, E. The RGD story: a personal account. Matrix


Biol. 2003, 22 (6), 459–465.

29. Krammer, A.; Craig, D.; Thomas, W.E.; Schulten, K.;


Vogel, V. A structural model for force regulated integrin
binding to fibronectin’s RGD-synergy site. Matrix Biol.
2002, 21 (2), 139–147.

30. Krammer, A.; Lu, H.; Isralewitz, B.; Schulten, K.;


Vogel, V. Forced unfolding of the fibronectin type III
module reveals a tensile molecular recognition switch.
Proc. Natl. Acad. Sci. U.S.A. 1999, 96 (4), 1351–1356.

31. Xu, J.; Rodriguez, D.; Petitclerc, E.; Kim, J.J.;


Hangai, M.; Moon, Y.S.; Davis, G.E.; Brooks, P.C.
Proteolytic exposure of a cryptic site within collagen type
IV is required for angiogenesis and tumor growth in vivo.
J. Cell Biol. 2001, 154 (5), 1069–1079.

32. Schulze, B.; Mann, K.; Poschl, E.; Yamada, Y.; Timpl,
R. Structural and functional analysis of the globular
domain IVa of the laminin alpha 1 chain and its impact on
an adjacent RGD site. Biochem. J. 1996, 314 (Pt 3),
847–851.

33. Zhong, C.; Chrzanowska-Wodnicka, M.; Brown, J.; Shaub,


A.; Belkin, A.M.; Burridge, K. Rho-mediated contractility
exposes a cryptic site in fibronectin and induces fibronectin
matrix assembly. J. Cell Biol. 1998, 141 (2), 539–551.

34. Geiger, B.; Bershadsky, A. Assembly and mechanosensory


function of focal contacts. Curr. Opin. Cell Biol. 2001, 13
(5), 584–592.

35. Wiseman, P.W.; Brown, C.M.; Webb, D.J.; Hebert, B.;


Johnson, N.L.; Squier, J.A.; Ellisman, M.H.; Horwitz, A.F.
Spatial mapping of integrin interactions and dynamics
during cell migration by image correlation microscopy. J.
Cell Sci. 2004, 117 (Pt 23), 5521– 5534.

36. Neff, N.T.; Lowrey, C.; Decker, C.; Tovar, A.; Damsky,
C.; Buck, C.; Horwitz, A.F. A monoclonal antibody detaches
embryonic skeletal muscle from extracellular matrices. J.
Cell Biol. 1982, 95, 654–666.

37. Zimerman, B.; Volberg, T.; Geiger, B. Early molecular


events in the assembly of the focal adhesion-stress fiber
complex during fibroblast spreading. Cell Motil.
Cytoskeleton 2004, 58 (3), 143–159.

38. Terracio, L.; Simpson, D.G.; Hilenski, L.; Carver, W.;


Decker, R.S.; Vinson, N.; Borg, T.K. Distribution of
vinculin in the Z-disk of striated muscle: analysis by
laser scanning confocal microscopy. J. Cell Physiol. 1990,
145 (1), 78–87.

39. Danowski, B.A.; Imanaka-Yoshida, K.; Sanger, J.M.;


Sanger, J.W. Costameres are sites of force transmission to
the substratum in adult rat cardiomyocytes. J. Cell Biol.
1992, 118 (6), 1411–1420.

40. Simpson, D.G.; Decker, M.L.; Clark, W.A.; Decker, R.S.


Contractile activity and cell-cell contact regulate
myofibrillar organization in cultured cardiac myocytes. J.
Cell Biol. 1993, 123 (2), 323–336.

41. Sharp, W.W.; Simpson, D.G.; Borg, T.K.; Samarel, A.M.;


Terracio, L. Contractile activity and external mechanical
load affect focal adhesion formation by cultured neonatal
rat cardiac myocytes. Am. J. Physiol. 1996, 273, H546–H556.
42. Zamir, E.; Katz, B.Z.; Aota, S.; Yamada, K.M.; Geiger,
B.; Kam, Z. Molecular diversity of cellmatrix adhesions. J.
Cell Sci. 1999, 112 (Pt 11), 1655–1669. 43. Matthews, B.D.;
Overby, D.R.; Alenghat, F.J.; Karavitis, J.; Numaguchi, Y.;
Allen, P.G.; Ingber, D.E. Mechanical properties of
individual focal adhesions probed with a magnetic
microneedle. Biochem. Biophys. Res. Commun. 2004, 313 (3),
758–764. 44. Hotchin, N.A.; Hall, A. The assembly of
integrin adhesion complexes requires both extracellular
matrix and intracellular rho/rac GTPases. J. Cell Biol.
1995, 131 (6 Pt 2), 1857–1865. 45. Machesky, L.M.; Hall, A.
Role of actin polymerization and adhesion to extracellular
matrix in Racand Rho-induced cytoskeletal reorganization.
J. Cell Biol. 1997, 138 (4), 913–926. 46. Nobes, C.D.;
Hall, A. Rho, rac, and cdc42 GTPases regulate the assembly
of multimolecular focal complexes associated with actin
stress fibers, lamellipodia, and filopodia. Cell 1995, 81
(1), 53–62. 47. Ridley, A.J.; Hall, A. The small
GTP-binding protein rho regulates the assembly of focal
adhesions and actin stress fibers in response to growth
factors. Cell 1992, 70 (3), 389–399. 48. Rottner, K.; Hall,
A.; Small, J.V. Interplay between Rac and Rho in the
control of substrate contact dynamics. Curr. Biol. 1999, 9
(12), 640–648. 49. Chrzanowska-Wodnicka, M.; Burridge, K.
Rhostimulated contractility drives the formation of stress
fibers and focal adhesions. J. Cell Biol. 1996, 133 (6),
1403–1415. 50. Riveline, D.; Zamir, E.; Balaban, N.Q.;
Schwarz, U.S.; Ishizaki, T.; Narumiya, S.; Kam, Z.; Geiger,
B.; Bershadsky, A.D. Focal contacts as mechanosensors:
externally applied local mechanical force induces growth of
focal contacts by an mDia1-dependent and ROCK-independent
mechanism. J. Cell Biol. 2001, 153 (6), 1175–1186. 51.
Zaidel-Bar, R.; Ballestrem, C.; Kam, Z.; Geiger, B. Early
molecular events in the assembly of matrix adhesions at the
leading edge of migrating cells. J. Cell Sci. 2003, 116
(22), 4605–4613. 52. Putnam, A.J.; Cunningham, J.J.;
Pillemer, B.B.; Mooney, D.J. External mechanical strain
regulates membrane targeting of Rho GTPases by controlling
microtubule assembly. Am. J. Cell Physiol. 2003, 284 (3),
C627–C639. 53. Kaverina, I.; Krylyshkina, O.; Small, J.V.
Microtubule targeting of substrate contacts promotes their
relaxation and dissociation. J. Cell Biol. 1999, 146,
1033–1044. 54. Kirchner, J.; Kam, Z.; Tzur, G.; Bershadsky,
A.D.; Geiger, B. Live-cell monitoring of tyrosine
phosphorylation in focal adhesions following microtubule
disruption. J. Cell Sci. 2003, 116, 975–986. 55. Ezratty,
E.J.; Partridge, M.A.; Gundersen, G.C. Microtubule-induced
focal adhesion disassembly is mediated by dynamin and focal
adhesion kinase. Nat. Cell Biol. 2005, 7, 581–590. 56.
Simpson, D.G.; Carver, W.; Borg, T.K.; Terracio, L. The
role of mechanical stimulation in the establishment and
maintenance of muscle cell differentiation. Int. Rev. Cyto.
1993, 150, 69–94. 57. Simpson, D.G.; Sharp, W.; Terracio,
L.; Price, R.L.; Borg, T.K.; Samarel, A.M. Mechanical
regulation of cardiac protein turnover and myofibrillar
structure. Am. J. Physiol. 1996, 270, C1075–C1087. 58.
Simpson, D.G.; Reaves, T.A.; Shih, D.; Burgess, W.; Borg,
T.K.; Terracio, L. Cardiac integrins: the ties that bind.
Cardiovasc. Pathol. 1997, 7, 135–143. 59. Simpson, D.G.;
Majeski, M.; Borg, T.K.; Terracio, L. Regulation of cardiac
protein turnover and myofibrillar structure in vitro by
specific directions of stretch. Circ. Res. 1999, 85,
59E–69E. 60. Civelekoglu-Scholeya, G.; Orrc, W.; Novak, I.;
Meistera, J.; Schwartze, M.A.; Mogilner, A. Model of
coupled transient changes of Rac, Rho, adhesions and stress
fibers alignment in endothelial cells responding to shear
stress. J. Theor. Biol. 2005, 232, 569–585. 61. Matthews,
J.A.; Wnek, G.E.; Simpson, D.G.; Bowlin, G.L.
Electrospinning collagen nanofibers. J. Macromol. 2002, 11,
232–238. 62. Telemeco, T.A.; Ayres, C.; Bowlin, G.L.; Wnek,
G.; Boland, E.D.; Cohen, N.M.; Baumgarten, C.M.; Mathews,
J.; Simpson, D.G. Regulation of cellular infiltration into
tissue engineering scaffolds composed of submicron diameter
fibrils produced by electrospinning. Acta Biomaterialia
2005, 1, 376–384. 63. Schlaepfer, D.D.; Jones, K.C.;
Hunter, T. Multiple Grb2-mediated integrin-stimulated
signaling pathways to ERK2/mitogen-activated protein
kinase: summation of both c-Src and focal adhesion kinase
initiated tyrosine phosphorylation events. Mol. Cell Biol.
1998, 18 (5), 2571–2585. 64. Richardson, A.; Malik, R.K.;
Hildebrand, J.D.; Parsons, J.T. Inhibition of cell
spreading by expression of the C-terminal domain of focal
adhesion kinase (FAK) is rescued by co-expression of Src or
catalytically inactive FAK: a role for paxillin tyrosine
phosphorylation. Mol. Cell Biol. 1997, 17 (12), 6906– 6914.
65. Hsia, D.A.; Mitra, S.K.; Hauck, C.R.; Streblow, D.N.;
Nelson, J.A.; Ilic, D.; Huang, S.; Li, E.; Nemerow, G.R.;
Leng, J.; Spencer, K.S.; Cheresh, D.A.; Schlaepfer, D.D.
Differential regulation of cell motility and invasion by
FAK. J. Cell Biol. 2003, 160 (5), 753–767. 66. Krylyshkina,
O.; Anderson, K.I.; Kaverina, I.; Upmann, I.; Manstein,
D.J.; Small, V.; Toomre, D.K. Nanometer targeting of
microtubules to focal adhesions. J. Cell Biol. 2003, 161,
853–859. 67. Marks, B.; Stowell, M.H.; Vallis, Y.; Mills,
I.G.; Gibson, A.; Hopkins, C.R.; McMahon, H.T. GTPase
activity of dynamin and resulting conformation change are
essential for endocytosis. Nature 2001, 410 (6825),
231–235. 68. Sorkin, A.; McClure, M.; Huang, F.; Carter, R.
Interaction of EGF receptor and grb2 in living cells
visualized by fluorescence resonance energy transfer (FRET)
microscopy. Curr. Biol. 2000 , 10 (21), 1395– 1398. 69.
Schlaepfer, D.D.; Hanks, S.K.; Hunter, T.; van der Geer, P.
Integrin-mediated signal transduction linked to Ras pathway
by GRB2 binding to focal adhesion kinase. Nature 1994, 372
(6508), 786–791. 70. Wang, Z.; Moran, M.F. Requirement for
the adapter protein GRB2 in EGF receptor endocytosis.
Science 1996, 272 (5270), 1935–1939. 71. Laukaitis, C.M.;
Webb, D.J.; Donais, K.; Horwitz, A.F. Differential dynamics
of alpha 5 integrin, paxillin, and alpha-actinin during
formation and disassembly of adhesions in migrating cells.
J. Cell Biol. 2001, 153 (7), 1427–1440. 72. Palecek, S.P.;
Schmidt, C.E.; Lauffenburger, D.A.; Horwitz, A.F. Integrin
dynamics on the tail region of migrating fibroblasts. J.
Cell Sci. 1996, 109 (Pt 5), 941–952. 73. Ding, B.; Price,
R.L.; Goldsmith, E.C.; Borg, T.K.; Yan, X.; Douglas, P.S.;
Weinberg, E.O.; Bartunek, J.; Thielen, T.; Didenko, V.V.;
Lorell, B.H. Left ventricular hypertrophy in ascending
aortic stenosis mice: anoikis and the progression to early
failure. Circulation 2000, 101 (24), 2854–2862. 74.
Goldsmith, E.C.; Carver, W.; McFadden, A.; Goldsmith, J.G.;
Price, R.L.; Sussman, M.; Lorell, B.H.; Cooper, G.; Borg,
T.K. Integrin shedding as a mechanism of cellular
adaptation during cardiac growth. Am. J. Physiol. Heart
Circ. Physiol. 2003, 284 (6), H2227–H2234. 75. Demetriou,
M.C.; Pennington, M.E.; Nagle, R.B.; Cress, A.E.
Extracellular alpha 6 integrin cleavage by urokinase-type
plasminogen activator in human prostate cancer. Exp. Cell
Res. 2004, 294 (2), 550–558. 76. Ho, A.T.; Voura, E.B.;
Soloway, P.D.; Watson, K.L.; Khokha, R. MMP inhibitors
augment fibroblast adhesion through stabilization of focal
adhesion contacts and up-regulation of cadherin function.
J. Biol. Chem. 2001, 276 (43), 40,215–40,224. 77. Dumin,
J.A.; Dickeson, S.K.; Stricker, T.P.;
Bhattacharyya-Pakrasi, M.; Roby, J.D.; Santoro, S.A.;
Parks, W.C. Pro-collagenase-1 (matrix metalloproteinase-1)
binds the alpha(2)beta(1) integrin upon release from
keratinocytes migrating on type I collagen. J. Biol. Chem.
2001, 276 (31), 29,368–29,374. 78. Richardson, A.; Parsons,
T. A mechanism for regulation of the adhesion-associated
proteintyrosine kinase pp125FAK. Nature 1996, 380 (6574),
538–540. 79. Nobes, C.D.; Lauritzen, I.; Mattei, M.G.;
Paris, S.; Hall, A.; Chardin, P. A new member of the Rho
family, Rnd1, promotes disassembly of actin filament
structures and loss of cell adhesion. J. Cell Biol. 1998,
141, 187–197. 80. Cary, L.A.; Klinghoffer, R.A.;
Sachsenmaier, C.; Cooper, J.A. SRC catalytic but not
scaffolding function is needed for integrin-regulated
tyrosine phosphorylation, cell migration, and cell
spreading. Mol. Cell Biol. 2002, 22 (8), 2427–2440. 81.
Westhoff, M.A.; Serrels, B.; Fincham, V.J.; Frame, M.C.;
Carragher, N.O. SRC-mediated phosphorylation of focal
adhesion kinase couples actin and I adhesion dynamics to
survival signaling. Mol. Cell Biol. 2004, 24 (18),
8113–8133.

82. Schlaepfer, D.D.; Hunter, T. Evidence for in vivo


phosphorylation of the Grb2 SH2-domain binding site on
focal adhesion kinase by Src-family proteintyrosine
kinases. Mol. Cell Biol. 1996, 16 (12), 7182–7184.

83. Nobes, C.D.; Hall, A. Rho GTPases control polarity,


protrusion, and adhesion during cell movement. J. Cell
Biol. 1999, 144 (6), 1235–1244.

84. Wolfsberg, T.G.; Straight, P.D.; Gerena, R.L.; Huovila,


A.P.; Primakoff, P.; Myles, D.G.; White, J.M. ADAM, a
widely distributed and developmentally regulated gene
family encoding membrane proteins with a disintegrin and
metalloprotease domain. Dev. Biol. 1995, 169 (1), 378–383.

85. Schlondorff, J.; Blobel, C.P.


Metalloproteasedisintegrins: modular proteins capable of
promoting cell-cell interactions and triggering signals by
protein-ectodomain shedding. J. Cell Sci. 1999, 112,
3603–3617.

86. White, J.M. ADAMs: modulators of cell-cell and


cellmatrix interactions. Curr. Opin. Cell Biol. 2003, 15,
598–606.

87. Higgins, J.M.; Mandlebrot, D.A.; Shaw, S.K.; Russell,


G.J.; Murphy, E.A.; Chen, Y.T.; Nelson, W.J.; Parker, C.M.;
Brenner, M.B. Direct and regulated interaction of integrin
alphaE beta7 with E-cadherin. J. Cell Biol. 1998, 140 (1),
197–210.

88. Scarborough, R.M.; Rose, J.W.; Naughton, M.A.;


Phillips, D.R.; Nannizzi, L.; Arfsten, A.; Campbell, A.M.;
Charo, I.F. Characterization of the integrin specificities
of disintegrins isolated from American pit viper venoms. J.
Biol. Chem. 1993, 268 (2), 1058– 1065.

89. Musial, J.; Niewiarowski, S.; Rucinski, B.; Stewart,


G.J.; Cook, J.J.; Williams, J.A.; Edmunds, L.H., Jr.
Inhibition of platelet adhesion to surfaces of
extracorporeal circuits by disintegrins. RGD-containing
peptides from viper venoms. Circulation 1990, 82 (1),
261–273.

90. Tilghman, R.W.; Slack-Davis, J.K.; Sergina, N.; Martin,


K.H.; Iwanicki, M.; Hershey, E.D.; Beggs, H.E.; Reichardt,
L.F.; Parsons, J.T. Focal adhesion kinase is required for
the spatial organization of the leading edge in migrating
cells. J. Cell Sci. 2005, 118 (Pt 12), 2613–23.

91. Simpson, D.G.; Terracio, L.; Terracio, M.; Price, R.L.;


Turner, D.C.; Borg, T.K. Modulation of cardiac myocyte
phenotype in vitro by the composition and orientation of
the extracellular matrix. J. Cell Physiol. 1994, 161,
89–105.

92. Maniotis, A.J.; Chen, C.S.; Ingber, D.E. Demonstration


of mechanical connections between integrins, cytoskeletal
filaments, and nucleoplasm that stabilize nuclear structure.
Proc. Natl. Acad. Sci. U.S.A. 1997, 94 (3), 849–584.

93. Chicurel, M.E.; Singer, R.H.; Meyer, C.J.; Ingber, D.E.


Integrin binding and mechanical tension induce movement of
mRNA and ribosomes to focal adhesions. Nature 1998, 392
(6677), 730–733. 94. Troussard, A.A.; Mawji, N.M.; Ong, C.;
Mui, A.; St-Arnaud, R.; Dedhar, S. Conditional knock-out of
integrin-linked kinase demonstrates an essential role in
protein kinase B/Akt activation. J Biol. Chem. 2003, 278
(25), 22,374–22,378. 95. Nho, S.; Xia, H.; Kahm, J.;
Kleidon, J.; Diebold, D.; Henke, C.A. Role of
integrin-linked kinase in regulating phosphorylation of Akt
and fibroblast survival in type I collagen matrices through
a beta 1 integrin viability signaling pathway. J. Biol.
Chem. 2005, 280 (28), 26,630–26,639. 96. Wang, X.Q.; Sun,
P.; Paller, A.S. Inhibition of integrin-linked
kinase/protein kinase B/Akt signaling: mechanism for
ganglioside-induced apoptosis. J. Biol. Chem. 2001, 276
(48), 44,504–44,511. 97. Scatena, M.; Almeida, M.;
Chaisson, M.L.; Fausto, N.; Nicosia, R.F.; Giachelli, C.M.
NF-kappaB mediates alphavbeta3 integrin-induced endothelial
cell survival. J. Cell Biol. 1998, 141 (4), 1083–1093. 98.
Gary, D.S.; Mattson, M.P. Integrin signaling via the
PI3-kinase-Akt pathway increases neuronal resistance to
glutamate-induced apoptosis. J. Neurochem. 2001, 76 (5),
1485–1496. 99. Persad, S.; Attwell, S.; Gray, V.;
Delcommenne, M.; Troussard, A.; Sanghera, J.; Dedhar, S.
Inhibition of integrin-linked kinase (ILK) suppresses
activation of protein kinase B/Akt and induces cell cycle
arrest and apoptosis of PTEN-mutant prostate cancer cells.
Proc. Natl. Acad. Sci. U.S.A. 2000, 97 (7), 3207–3212. 100.
Tremble, P.; Damsky, C.H.; Werb, Z. Components of the
nuclear signaling cascade that regulate collagenase gene
expression in response to integrin-derived signals. J. Cell
Biol. 1995, 129 (6), 1707–1720. 101. Pulai, J.I.; Chen, H.;
Im, H.J.; Kumar, S.; Hanning, C.; Hegde, P.S.; Loeser, R.F.
NF-kappa B mediates the stimulation of cytokine and
chemokine expression by human articular chondrocytes in
response to fibronectin fragments. J. Immunol. 2005, 174
(9), 5781–5788. 102. Juliano, R.L.; Reddig, P.; Alahari,
S.; Edin, M.; Howe, A.; Aplin, A. Integrin regulation of
cell signalling and motility. Biochem. Soc. Trans. 2004, 32
(Pt 3), 443–446. 103. Oktay, M.; Wary, K.K.; Dans, M.;
Birge, R.B.; Giancotti, F.G. Integrin-mediated activation
of focal adhesion kinase is required for signaling to Jun
NH2terminal kinase and progression through the G1 phase of
the cell cycle. J. Cell Biol. 1999, 145 (7), 1461–1469.
104. Brizzi, M.F.; Defilippi, P.; Rosso, A.; Venturino, M.;
Garbarino, G.; Miyajima, A.; Silengo, L.; Tarone, G.;
Pegoraro, L. Integrin-mediated adhesion of endothelial
cells induces JAK2 and STAT5A activation: role in the
control of c-fos gene expression. Mol. Biol. Cell 1999, 10
(10), 3463–3471. 105. Burridge, K.; Wennerberg, K. Rho and
Rac take center stage. Cell 2004, 116, 167–179. 106. Iyer,
V.; Pumiglia, K.; DiPersio, C.M. Alpha3 beta1 integrin
regulates MMP-9mRNA stability in immortalized
derationcytes: a novel mechanism of integrinmediated MMP
gene expression. J. Cell Sci. 2005, 118, 1185–1195. 107.
Green, K.J.; Jones, J.C. Desmosomes and hemidesmosomes:
structure and function of molecular components. FASEB J.
1996, 10 (8), 871–881. 108. Spinardi, L.; Ren, Y.L.;
Sanders, R.; Giancotti, F.G. The beta 4 subunit cytoplasmic
domain mediates the interaction of alpha6 beta4 integrin
with the cytoskeleton of hemidesmosomes. Mol. Biol. Cell
1993, 4 (9), 871–884. 109. Carver, W.; Molano, I.; Reaves,
T.A.; Borg, T.K.; Terracio, L. Role of the alpha 1 beta 1
integrin complex in collagen gel contraction in vitro by
fibroblasts. J. Cell Physiol. 1995, 165 (2), 425–437. I
Intellectual Property Management

1. Neilson vs. Minister of Public Works (NSW) (1914), 18


CLR 423.

2. Re-application of Honeywell Bull Inc (1991), 22


INTELLECTUAL PROPERTY 463.

3. IBM Corp vs. Commissioner of Patents (1991), 22


INTELLECTUAL PROPERTY 417.

4. CCOM Pty Ltd vs. Jiejing Pty Ltd (1994), 28 INTELLECTUAL


PROPERTY 481.

5. 447 US 303 (1980).

6. UNIDO. Guidelines for the Acquisition of Foreign


Technology in Developing Countries; 1973; 4–5.

7. Restatement of the Law of Torts, Art. 757.

8. Rote Taube (Red Dove) (1970), 1 IIC 136.

9. 447 US 303 (1980).

10. See 1.2.1 infra.


Interpenetrating Polymeric Networks

Fig. 5 Oscillatory swelling behavior as a function of time

and temperature at pH 7.4 and 4.0 for PMAA/PNIPAAm

IPN samples containing 70mol% of PNIPAAm.

Fig. 6 Scanning electron micrograph of a PMAA/

PNIPAAm IPN sample prepared from 40 vol% of MAA

methanolic solution (for the primary network) and 0.2 g/ml

NIPAAm methanolic solution (for the secondary network),

with 1wt% of initiator, DMPA, and 1mol% of

crosslinking agent, tetraethylene glycol dimethacrylate

(TEGDMA), for each solution. Sample was dried first in

air for two days and then in vacuum oven for five days and

was fractured after liquid nitrogen quenching. The bar

indicates 10 mm. 8. Lipatov, Y.S. Advances in


Interpenetrating Polymer Networks; Klempner, D., Frisch,
K.C., Eds.; Technomic: Lancaster, PA, 1989; 261. 9. Ma, S.;
Tang, X. Morphology of acrylic-epoxy bichain simultaneous
interpenetrating polymer networks. In Interpenetrating
Polymer Networks. Advances in Chemistry; Klempner, D.,
Sperling, L.H., Utracki, L.A., Eds.; American Chemical
Society: Washington, DC, 1994. 10. Klempner, D., Frisch,
K.C., Eds.; Advances in Interpenetrating Polymer Networks;
Technomic: Lancaster, PA, 1994. 11. Hourston, D.J.; Huson,
M.G.; McCluskey, J.A. Semiand fully interpenetrating
polymer networks based on polyurethane-polyacrylate
systems. X. Polyurethanepoly(ethyl acrylate)
interpenetrating polymer networks. J. Appl. Polym. Sci.
1986, 32, 3881. 12. Sophia, D.; Klempner, D.; Sendijarevic,
V.; Suthar, B.; Frisch, K.C. Interpenetrating polymer
networks as energy-absorbing materials. In Interpenetrating
Polymer Networks; Klempner, D., Sperling, L.H., Utracki,
L.A., Eds.; Advances in Chemistry Series, American Chemical
Society: Washington, DC, 1994; 39–75. 13. Kim, S.C.;
Sperling, L.H.; Eds. IPNs Around the World: Science and
Engineering; John Wiley & Sons: New York, NY, 1997. 14.
Klempner, D.; Frisch, H.L. Topologically interpenetrating
elastomeric networks. J. Polym. Sci. A-2, 1970, 8, 921–935.
15. Lipatov, Y.S. Equilibrium and non-equilibrium
microphase structures of interpenetrating polymer networks.
In Interpenetrating Polymer Networks; Klempner, D.,
Sperling, L.H., Utracki, L.A., Eds.; Advances in Chemistry
Series, American Chemical Society: Washington, DC, 1994;
125–139. 16. Katono, H.; Maruyama, A.; Sanui, K.; Ogata,
N.; Okano, T.; Sakurai, Y. Thermo-responsive swelling and
drug release switching of interpenetrating polymer networks
composed of poly(acrylamide-co-butyl methacrylate) and
poly(acrylic acid). J. Contr. Rel. 1991, 16, 215–228. 17.
Lim, Y.H.; Kim, D.; Lee, D.S. Drug releasing
characteristics of thermoand pHsensitive interpenetrating
polymer networks based on poly(N-isopropylacrylamide). J.
Appl. Polym. Sci. 1997, 64, 2647–2655. 18. Aoki, T.;
Kawashima, M.; Katono, H.; Sanui, K.; Ogata, N.; Okano, T.;
Sakurai, Y. Temperatureresponsive interpenetrating polymer
networks constructed with poly(acrylic acid) and
poly(N,N-dimethylacrylamide). Macromolecules 1994, 27,
947–952. 19. Buyanov, A.L.; Revelskaya, L.G.; Kuznetzov,
Y.P.; Shestakova, A.S. Cellulose-poly(acrylamide or acrylic
acid) interpenetrating polymer network membranes for the
pervaporation of water-ethanol mixture. J. Appl. Polym.
Sci. 1998, 69, 761–769. 20. Hoffman, A.S. Conventional and
environment-sensitive hydrogels for medical and industrial
uses: a review paper. Polym. Prepr. 1990, 31 (1), 220–221.
21. Kost, J.; Langer, R. Equilibrium swollen hydrogels in
controlled release applications. InHydrogels in Medicine
and Pharmacy; Peppas, N.A., Ed.; CRC Press: Boca Raton, FL,
1986; 95–107. 22. Peppas, N.A. Other biomedical
applications of hydrogels. In Hydrogels in Medicine and
Pharmacy; Peppas, N.A., Ed.; CRC Press: Boca Raton, FL,
1986; 177–194. 23. Ratner, B. Biomedical applications of
hydrogels: critical appraisal and review. In
Biocompatibility of Clinical Implant Materials; Williams,
D.F., Ed.; CRC Press: Boca Raton, FL, 1981; 145–153. 24.
Peppas, N.A.; Korsmeyer, R.W. Dynamic swollen hydrogels in
controlled release applications. In Hydrogels in Medicine
and Pharmacy; Peppas, N.A., Ed.; CRC Press: Boca Raton, FL,
1986; 109–135. 25. Langer, R.; Hsieh, D.S.T.; Rhine, W.;
Folkman, J. Control of release kinetics of macromolecules
from polymers. J. Membr. Sci. 1980, 7, 330–350. 26. Langer,
R. Polymeric delivery system for controlled drug release.
Chem. Eng. Comm. 1980, 6, 1–48. 27. Bell, C.L.; Peppas,
N.A. Poly(methacrylic acid-g-ethylene glycol) hydrogels as
pH-responsive biomedical materials. In Biomaterials for
Drug and Cell Delivery; Miikos, A.G., Murphy, R., Bemstein,
H., Peppas, N.A., Eds.; Pittsburgh, PA, 1994; 199–204. 28.
Lowman, A.M.; Peppas, N.A. Analysis of the
complexation/decomplexation phenomena in graft copolymer
networks. Macromolecules 1997, 30, 4959–4965. 29. Khare,
A.R.; Peppas, N.A. Swelling/deswelling of anionic copolymer
gels. Biomaterials 1995, 16, 559–567. 30. Brazel, C.S.;
Peppas, N.A. Synthesis and characterization of thermoand
chemomechanically responsive
poly(Nisopropylacrylamide-co-methacrylic acid) hydrogels.
Macromolecules 1995, 28, 8016–8020. 31. Vakkalanka, S.K.;
Brazel, C.S.; Peppas, N.A. Temperatureand pHsensitive
terpolymers for modulated delivery of streptokinase. J.
Biomater. Sci. Polym. Edn. 1996, 8, 119–129. 32. Seminoff,
L.; Olsen, G.; Zheng, J.; Kim, S.W. Selfregulated insulin
release. Proc. Int. Symp. Control. Rel. Bioact. Mater.
1998, 15, 160–161. 33. Shino, D.; Murata, Y.; Kubo, A.;
Kataoka, K.; Koyama, Y.; Kikuchi, A.; Yokohama, M.;
Sakurai, Y.; Okano, T. Amine containing phenylboronic acid
gel for glucose-responsive insulin release under
physiological pH. J. Contr. Rel. 1995, 37, 269–276. 34.
Gehrke, S.H. Synthesis, equilibrium swelling, kinetics,
permeability and applications of environmentally responsive
gels. Adv. Polym. Sci. 1993, 110, 81–144. 35. Bell, C.;
Peppas, N.A. Biomedical membranes from hydrogels and
interpolymer complexes. In Biopolymer II; Peppas, N.A.,
Langer, R.S., Eds.; Advances in Polymer Science;
Springer-Verlag: New York, 1995; 125–176. 36. Bae, Y.H.
Stimuli-sensitive drug delivery. In Controlled Drug
Delivery Challenges and Strategies; Park, K., Ed.; American
Chemical Society: Washington, DC, 1997; 147–162. 37.
Kaneko, Y.; Saki, K.; Kikuchi, A.; Sakurai, Y.; Okano, T.
Fast swelling/deswelling kinetics of comb-type aragted
poly(N-isopropyl acrylamide) hydrogels. Macromol. Symp.
1996, 109, 41–53. 38. Vakkalanka, S.K.; Peppas, N.A.
Swelling behavior of temperatureand pHsensitive block
terpolymers for drug delivery. Polym. Bull. 1996, 36,
221–225. I

39. Chen, G.; Hoffman, A.S. Graft copolymer that exhibit


temperature-induced phase transitions over a wide range of
pH. Nature 1995, 373, 49–52.

40. Schwarte, L.M.; Podual, K.; Peppas, N.A. Cationic


hydrogels for controlled release of proteins and other
macromolecules. In Tailored Polymeric Materials for
Controlled Delivery System; McCulloch, I., Shalaby, S.W.,
Eds.; American Chemical Society: Washington, DC, 1998;
57–66.

41. Ramkissoon-Ganokar, C.; Liu, F.; Baudys, M.; Kim, S.W.


Modulating insulin-release profile from pH/thermosensitive
polymeric beads through polymer molecular weight. J. Contr.
Rel. 1999, 59, 87–98.
42. Okano, T. Biorelated Polymers and Gels: Controlled
Release and Applications in Biomedical Engineering;
Academic Press: San Diego, CA, 1998.

43. Weiss, A.M.; Adler, K.P.; Grodzinsky, A.J.; Yarmush,


M.L. Variable permeability membranes. Network structure of
poly(methacrylic acid) and its relation to diffusive
transport. J. Membr. Sci. 1991, 58, 153–173.

44. Seno, M.; Len, M.L.; Iwamoto, K. Swelling of


poly(methacrylic acid) gels and adsorption of L-lysine and
its polymer on the gels. Colloid Polym. Sci. 1991, 269,
873–879.

45. Saito, S.; Konno, M.; Inomata, H. Volume phase


transition of N-alkylacrylamide gels. Adv. Polym. Sci.
1993, 109, 207–232.

46. Tanaka, T. Volume phase transition in


poly(N-isopropylacrylamide) gel. Phys. Rev. Lett. 1978, 40,
820.

47. Hoffman, A.S.; Afrassiabi, A.; Dong, L.C. Thermally


reversible hydrogels. II. Delivery and selective removal of
substances from aqueous solution. J. Contr. Rel. 1986, 4,
213–222.

48. Gutowska, A.; Bae, Y.H.; Kim, S.W. Heparin release from
thermosensitive hydrogels. J. Contr. Rel. 1992, 22, 95–104.

49. Bae, Y.H.; Okano, T.; Kim, S.W. ‘‘On-off’’


thermocontrol of solute transport. I. Temperature
dependence of swelling of N-isopropylacrylamide networks
modified with hydrophobic components in water. Pharm. Res.
1991, 8, 531–537.

50. Chen, J.P.; Yang, H.J.; Hoffman, A.S. Polymer-protein


conjugates. I. Effect of protein conjugation on the cloud
point of poly(N-isopropylacrylamide). Biomaterials 1990,
11, 625–630.

51. Gutowska, A.; Bae, Y.H.; Jacobs, H.; Feijen, J.; Kim,
S.W. Thermosensitive interpenetrating polymer networks:
synthesis, characterization, and macromolecular release.
Macromolecules 1994, 27, 4167–4175.

52. Mukae, K.; Bae, Y.H.; Okano, T.; Kim, S.W. A new
thermo-sensitive hydrogel: poly(ethylene oxide-dimethyl
siloxane-ethylene oxide)/poly(N-isopropylacrylamide)
interpenetrating polymer networks I. Synthesis and
characterization. Polym. J. 1990, 22, 206–217.

53. Mukae, K.; Bae, Y.H.; Okano, T.; Kim, S.W. A new
thermosensitive hydrogel: poly(ethylene oxide-dimethyl
siloxane-ethylene oxide)/poly(N-isopropylacrylamide)
interpenetrating polymer networks. II. On-off regulation of
solute release from thermo-sensitive hydrogels. Polym. J.
1902, 2, 250–265. 54. Schild, H.G.
Poly(N-isopropylacrylamide): experiment, theory and
application. Progr. Polym. Sci. 1992, 17, 163–249. 55.
Heskins, M.; Guillet, J.E. Solution properties of
poly(Nisopropylacrylamide). J. Macromol. Sci. Chem. 1968,
A2, 1441–1455. 56. Otake, K.; Inomata, H.; Konno, M.;
Saito, S. Thermal analysis of the volume phase transition
with N-isopropylacrylamide gels. Macromolecules 1990, 23,
283–289. 57. Prange, M.M.; Hooper, H.H.; Pransnitz, J.M.
Thermodynamics of aqueous systems containing hydrophilic
polymers or gels. AIChE J. 1989, 35, 803–813. 58. Hirotsu,
S.; Hirokawa, Y.; Tanaka, T. Volume-phase transitions of
ionized N-isopropylacrylamide gels. J. Chem. Phys. 1987,
87, 1392–1395. 59. Marchetti, M.; Prager, S.; Cussler, E.L.
Thermodynamic predictions of volume changes in
temperaturesensitive gels. 2. Experiments. Macromolecules
1990, 23, 3445–3450. 60. Lee, K.K.; Cussler, E.L.;
Marchetti, M.; McHugh, M.A. Pressure-dependent phase
transitions in hydrogels. Chem. Eng. Sci. 1990, 45,
766–767. 61. Otake, K.; Inomata, H.; Konno, M.; Saito, S. A
new model for the thermally induced volume phase transition
of gels. J. Chem. Phys. 1990, 91, 1345–1350. 62. Freitas,
R.F.S.; Cussler, E.L. Temperature sensitive gels as
extraction solvents. Chem. Eng. Sci. 1987, 42, 97–103. 63.
Katono, H.; Sanui, K.; Ogata, N.; Okano, T.; Sakurai, Y.
Drug release OFF behavior and deswelling kinetics of
thermo-responsive IPNs composed of poly(acrylamide-co-butyl
methacrylate) and poly(acrylic acid). Polym. J. 1991, 23,
1179–1189. 64. Kaneko, Y.; Nakamura, S.; Sakai, K.; Aoyagi,
T.; Kikuchi, A.; Sakurai, Y.; Okano, T. Rapid deswelling
response of poly(N-isopropylacrylamide) hydrogels by the
formation of water release channels using poly (ethylene
oxide) graft chains. Macromolecules 1998, 31, 6099–6105.
65. Kim, Y.-H.; Bae, Y.H.; Kim, S.W.
pH/temperaturesensitive polymers for macromolecular drug
loading and release. J. Contr. Rel. 1994, 28, 143–152. 66.
Okano, T.; Bae, Y.H.; Jacobs, H.; Kim, S.W. Thermally
on-off switching polymers for drug permeation and release.
J. Contr. Rel. 1990, 11, 255–265. 67. Dong, L.C.; Hoffman,
A.S. Synthesis and application of thermally reversible
heterogels for drug delivery. J. Contr. Rel. 1990, 13,
21–31. 68. Yoshida, R.; Sakai, K.; Okano, T.; Sakurai, Y.
Pulsatile drug delivery systems using hydrogels. Adv. Drug
Deliv. Rev. 1993 , 11, 85–108. 69. Serres, A.; Baudys, M.;
Kim, S.W. Temperature and pH-sensitive polymer for human
calcitonin delivery. Pharm. Res. 1996, 13, 196–201. 70.
Kikuchi, A.; Okano, T. Temperature-responsive polymers as
On-Off switches for intelligent biointerfaces. In
Biorelated Polymers and Gels; Okano, T., Ed.; Academic
Press: San Diego, CA, 1998; 1–28. 71. Dong, L.C.; Yan, Q.;
Hoffman, A.S. Controlled release of amylase from a thermal
and pH-sensitive, macroporous hydrogel. J. Contr. Rel.
1992, 19, 171–178. 72. Brazel, C.S.; Peppas, N.A. Pulsatile
local delivery of thrombolytic and antithrombotic agents
using poly (N-isopropylacrylamide-co-methacrylic acid)
hydrogels. J. Contr. Rel. 1996, 39 (57–64). 73. Feil, H.;
Bae, Y.H.; Feijen, J.; Kim, S.W. Mutual influence of pH and
temperature on the swelling of ionizable and
thermosensitive hydrogels. Macromolecules 1992, 25,
5528–5530. 74. Zhang, J.; Peppas, N.A. Synthesis and
characterization of pHand temperature-sensitive PMAA/PNIPAm
interpenetrating polymer networks. Macromolecules 2003, 3,
102–107. 75. Zhang, J.; Peppas, N.A. Structure and solute
size exclusion of poly(methacrylic acid)/poly(N-isopropyl a
crylamide) interpenetrating polymeric networks. In
Polymeric Drugs and Drug Delivery Systems; Ottenbrite,
R.M., Kim, S.W., Eds.; Technomic: Lancaster, PA, 2001;
157–168. 76. Zhang, J.; Peppas, N.A. Molecular interactions
in poly(methacrylic acid)/poly(N-isopropyl acrylamide)
interpenetrating polymeric networks. J. Appl. Polym. Sci.
2001, 82, 1077–1082. 77. Zhang, J.; Peppas, N.A. Morphology
of poly(methacrylic acid)/poly(N-isopropyl acrylamide)
interpenetrating polymeric networks. J. Biomater. Sci.
Polym. Ed. 2002, 13, 511–525. I This page intentionally
left blank
Intravascular Ultrasound

11. Dupis, J.; Tardif, J.C.; Cernacek, P.; Theroux, P.


Cholesterol reduction rapidly improves endothelial function
after acute coronary syndromes. The RECIFE (reduction of
cholesterol in ischemia and function of the endothelium)
trial. Circulation 1999, 99, 3227–3233.

12. Mikhailidi, D.P.; Ganotakis, E.S.; Spyropoulos, K.A.;


Jagroop, I.A.; Byrne, D.J.; Winder, A.F. Prothrombotic and
lipoproterin variables in patients attending a
cardiovascular risk management clinic: Response to
cipofibrate or lifestyle advice. Int. Angiol. 1998, 17,
225–233.

13. Grube, E.; Gerckens; Rowold, S.; Yeung, A.C.; Stertzer,


S.H. Inhibition of in-stent restenosis by a drug-eluting
polymer stent: Pilot trial with 18-month follow-up.
Circulation 2000, 102, II-554.

14. Ohnesorge, B.M.; Becker, C.R.; Flohr, T.G.; Reiser,


M.F. Multi-slice CT in Cardiac Imaging: Technical
Principles, Clinical Application and Future Developments;
Springer: New York, 2002.

15. Nishimura, D.G. Principles of Magnetic Resonance


Imaging; Nishimura: Stanford, CA, 1996.

16. Huang, D.; Swanson, E.A.; Lin, C.P.; Schuman, J.S.;


Stinson, W.G.; Chang, W.; Hee, M.R.; Flotte, T.; Gregory,
K.; Puliafito, C.A.; Fujimoto, J.G. Optical coherence
tomography. Science 1991, 254, 1178–1181.

17. Brezenski, M.E.; Fujimoto, J.G. Optical coherence


tomography: High resolution imaging in nontransparent
tissue. IEEE J. Sel. Top. Quantum Electron. 1999, 5,
1185–1192.

18. Crowe, J.R.; O’Donnell, M. Quantitative blood speed


imaging with intravascular ultrasound. IEEE Trans.
Ultrason. Ferroelectr. Freq. Control 2001, 48, 477–487.

19. Shapo, B.M.; Crowe, J.R.; Skovoroda, A.R.; Eberle,


M.J.; Cohn, N.A.; O’Donnell, M. Displacement and strain
imaging of coronary arteries with intraluminal ultrasound.
IEEE Trans. Ultrason. Ferroelectr. Freq. Control 1996, 43,
234–246.

20. O’Donnell, M.; Eberle, M.J.; Stephens, D.N.; Litzza,


J.L.; Shapo, B.M.; Crowe, J.R.; Choi, C.D.; Chen, J.J.;
Muller, D.W.M.; Kovach, J.A.; Lederman, R.J.; Ziegenbein,
R.C.; San Vicente, K.; Bleam, D. In Catheter Arrays: Can
Intravascular Ultrasound Make a Difference in Managing
Coronary Artery Disease?, Proceedings of the IEEE
Ultrasonics Symposium, Toronto, Canada, October, 1997; IEEE
UFFC, 1997; 1447–1456. 21. de Korte, C.L.; van der Steen,
A.F.W.; Cespedes, E.I.; Pasterkamp, G. Intravascular
ultrasound elastography in human arteries: Initial
experience in vivo. Ultrasound Med. Biol. 1998, 24,
401–408. 22. Cespedes, E.I.; de Korte, C.L.; van der Steen,
A.F.W. Intraluminal ultrasonic palpation: Assessment of
local and cross-sectional tissue stiffness. Ultrasound Med.
Biol. 2000, 26, 285–296. 23. Falk, E. Why do plaques
rupture? Circulation 1992, 86 (Suppl. III), III-30–III-42.
24. Wang, Y.; Stephens, D.N.; O’Donnell, M. Optimizing the
beam pattern of a forward-viewing ring-annular ultrasound
array for intravascular imaging. IEEE Trans. Ultrason.
Ferroelectr. Freq. Control 2002, 12, 1652–1664. 25. Choi,
C.D.; Skovoroda, A.R.; Emelianov, S.Y.; O’Donnell, M. An
integrated compliant balloon ultrasound catheter for
intravascular strain imaging. IEEE Trans. Ultrason.
Ferroelectr. Freq. Control 2002, 11, 1552–1560. 26. Lanza,
G.M.; Wallace, K.D.; Scott, M.J.; Catheris, W.P.; Kaufmann,
R.J.; Gaffney, P.J.; Wickline, S.A. A novel site-targeted
ultrasonic contrast agent with broad biomedical
application. Circulation 1998, 94, 3334–3340. 27. Lanza,
G.M.; Trousil, R.L.; Wallace, K.D.; Rose, J.H.; Hall, S.C.;
Scott, M.J.; Miller, J.G.; Eisenberg, P.R.; Gaffney, P.J.;
Wickline, S.A. In vitro characterization of a novel,
tissue-targeted ultrasound contrast system with acoustic
microscopy. J. Acoust. Soc. Am. 1999, 104, 3665–3672. 28.
Lawrie, A.; Brisken, A.F.; Francis, S.E.; Tayler, D.I.;
Chamberlain, J.; Crossman, D.C.; Cumberland, D.C.; Newman,
C.M. Ultrasound enhances reporter gene expression after
transfection of vascular cells in vitro. Circulation 1999,
99, 2617–2620. I
In Vivo Cardiovascular Models

1. Weinstein, P.R.; Reinert, R.L.; Brittain, F. Delayed


thrombosis of synthetic microvascular bypass grafts.
Neurosurgery 1981, 9 (3), 268–274.

2. Madras, P.N.; Johnson, W.R.; Ward, C.A. Enhancement of


thrombus formation by vascular prostheses. Can. J. Surg.
1980, 23 (6), 544–546.

3. Goldman, S.; Zadina, K.; Krasnicka, B.; Moritz, T.;


Sethi, G.; Copeland, J.; Ovitt, T.; Henderson, W.;
Department of Veterans Affairs Cooperative Study Group No.
297 Predictors of graft patency 3 years after coronary
artery bypass graft surgery. J. Am. Coll. Cardiol. 1997, 29
(7), 1563–1568.

4. Plecha, E.J.; Freischlag, J.A.; Seabrook, G.R.; Towne,


J.B. Femoropopliteal bypass revisited, an analysis of 138
cases. Cardiovasc. Surg. 1996, 4 (2), 195–199.

5. Cooper, G.J.; Underwood, M.J.; Deverall, P.B. Arterial


and venous conduits for coronary artery bypass, a current
review. Eur. J. Cardio-Thorac. Surg. 1996, 10 (2), 129–140.

6. Benedetti-Valentini, F.; Gossetti, B.; Irace, I.;


Martinelli, O.; Gattuso, R. Composite grafts for critical
ischemia. Cardiovasc. Surg. 1996, 4 (3), 372–376.

7. Donzeau-Gouge, P.; Touati, G.; Vouhe, P.R.; Roy, A.;


Farge, C.; Leca, F.; Neveux, J.Y. Coronary artery bypass
with bovine internal mammary artery graft. Arch. Mal. Coeur
Vaiss. 1990, 83 (12), 1811–1815.

8. Iaffaldano, R.A.; Lewis, B.E.; Johnson, S.A.; Piffare,


R.; McKiernan, T.L. Patency of cryopreserved saphenous vein
grafts as conduits for coronary artery bypass surgery.
Chest 1995, 108 (3), 725–729.

9. Conklin, B.S.; Richter, E.R.; Kreutziger, K.L.; Zhong,


D.S.; Chen, C. Development and evaluation of a novel
decellularized vascular xenograft. Med. Eng. Phys. 2002, 24
(3), 173–183.

10. Lee, W.K.; Park, K.D.; Han, D.K.; Suh, H.; Park, J.C.;
Kim, Y.H. Heparinized bovine pericardium as a novel
cardiovascular bioprosthesis. Biomaterials 2000, 21 (22),
2323–2330. 11. Zilla, P.; Fasol, R.; Deutch, M.; Fischlein,
T.; Minar, E.; Hammerle, A.; Krupicka, O.; Kadletz, M.
Endothelial cell seeding of polytetrafluoroethylene vascular
grafts in humans. J. Vasc. Surg. 1987, 6 (6), 535–541. 12.
Massia, S.P.; Hubbell, J.A. Tissue engineering in the
vascular graft. Cytotechnology 1992, 10 (3), 189–204. 13.
Nerem, R.M.; Seliktar, D. Vascular tissue engineering.
Annu. Rev. Biomed. Eng. 2001, 3, 225–243. 14. Dunn, P.F.;
Newman, K.D.; Jones, M.; Yamada, I.; Shayani, V.; Virmani,
R.; Dichek, D.A. Seeding of vascular grafts with
genetically modified endothelial cells. Circulation 1996, 93
(7), 1439–1446. 15. Teebken, O.E.; Haverich, A. Tissue
engineering of small diameter vascular grafts. Eur. J.
Vasc. Endovasc. Surg. 2002, 23 (6), 475–485. 16. Ratcliffe,
A. Tissue engineering of vascular grafts. Matrix Biol.
2000, 19 (4), 353–357. 17. Valente, M.; Pettenazzo, E.; Di
Filippo, L.; Laborde, F.; Rinaldi, S.; Thiene, G.
Biodegradable polymer (D,L-lactide-caprolactone) in aortic
vascular prosthesis, morphological evaluation in an animal
model. Int. J. Artif. Organs 2002, 25 (8), 777–782. 18.
Zippel, R.; Wilhelm, L.; Marusch, F.; Koch, A.; Urban, G.;
Schlosser, M. Antigenicity of polyester (Dacron) vascular
prostheses in an animal model. Eur. J. Vasc. Endovasc.
Surg. 2001, 21 (3), 202–207. 19. Macneill, B.D.;
Pomerantseva, I.; Lowe, H.C.; Oesterle, S.N.; Vacanti, J.P.
Toward a new blood vessel. Vasc. Med. 2002, 7 (3), 241–246.
20. Merhi, Y.; Bernier, J.; Marois, Y.; Guidoin, R.L. Acute
thrombogenicity of arterial prostheses exposed to reduced
blood flow in dogs, effects of heparin, aspirin and
prostacyclin. J. Cardiovasc. Pharmacol. 1995, 26 (1), 1–5.
21. Ziegler, T.; Nerem, R.M. Tissue engineering a blood
vessel, regulation of vascular biology by mechanical
stresses. J. Cell. Biochem. 1994, 56 (2), 204–209. 22.
Hess, F.; Steeghs, S.; Jerusalem, R.; Reijnders, O.;
Jerusalem, C.; Braun, B.; Grande, P. Patency and morphology
of fibrous polyurethane vascular prostheses implanted in the
femoral artery of dogs after seeding with subcultivated
endothelial cells. Eur. J. Vasc. Surg. 1993, 7 (4),
402–408. 23. Fernandez, P.; Bareille, R.; Conrad, V.; Midy,
D.; Bordenave, L. Evaluation of an in vitro endothelialized
vascular graft under pulsatile shear stress with a novel
radiolabeling procedure. Biomaterials 2001, 22 (7),
649–658. 24. Nerem, R.M. Role of mechanics in vascular
tissue engineering. Biorheology 2003, 40 (1–3), 281–287.
25. Abbott, W.M.; Callow, A.; Moore, W.; Rutherford, R.;
Veith, F.; Weinberg, S. Evaluation and performance
standarts for arterial prostheses. J. Vasc. Surg. 1993, 17
(4), 746–756. 26. Sparks, S.R.; Tripathy, U.; Broudy, A.;
Bergan, J.J.; Kumins, N.H.; Owens, E.L. Small-caliber
mesothelial cell-layered polytetraflouroethylene vascular
grafts in New Zealand white rabbits. Ann. Vasc. Surg. 2002,
16 (1), 73–76. 27. Shigematsu, K.; Yasuhara, H.;
Shigematsu, H. Topical application of antiangiogenic agent
AGM-1470 suppresses anastomotic intimal hyperplasia after
ePTFE grafting in a rabbit model. Surgery 2001, 129 (2),
220–230. 28. Yue, X.; Van der Lei, B.; Schakenraad, J.M.;
Van Oene, G.H.; Kuit, J.H.; Feijen, J. Smooth muscle cell
seeding in biodegradable grafts in rats, a new method to
enhance the process of arterial wall regeneration. Surgery
1988, 103 (2), 206–212. 29. Joshi, R.R.; Underwood, T.;
Frautschi, J.R.; Phillips, R.E.; Schoen, F.J.; Levy, R.J.
Calcification of polyurethanes implanted subdermally in rats
is enhanced by calciphylaxis. J. Biomed. Mater. Res. 1996,
31 (2), 201–207. 30. Bobryshev, Y.V.; Inder, S.J.; Cherian,
S.M.; Lord, R.S.; Ao, P.Y.; Hawthorne, W.J.; Fletcher, J.P.
Colonisation of prosthetic grafts by immunocompetent cells
in a sheep model. Cardiovasc. Surg. 2001, 9 (2), 166–176.
31. Ortenwall, P.; Bylock, A.; Kjellstrom, T.; Risberg, B.
Seeding of ePTFE carotid interposition grafts in sheep and
dogs, species-dependent results. Surgery 1988, 103 (2),
199–205. 32. Galletti, G.; Ussia, G.; Farruggia, F.;
Baccarini, E.; Biagi, G.; Gogolewski, S. Prevention of
platelet aggregation by dietary polyunsaturated fatty acids
in the biodegradable polyurethane vascular prosthesis, an
experimental model in pigs. Ital. J. Surg. Sci. 1989, 19
(2), 121–130. 33. Marois, Y.; Guidoin, R.; Wagner, E.; Roy,
R.; Douville, Y. Fresh venous allografts as arterial
substitutes in dogs, the importance of donor-recipient
tissue compatibility. J. Invest. Surg. 1994, 7 (5),
393–408. 34. Akoum, A.; Guidoin, R.; King, M.W.; Marois,
Y.; Sigot, M.; Sigot-Luizard, M.F. A new bioactive molecule
for improving vascular graft patency, exploratory trials in
dogs. Clin. Invest. Med. 1992, 15 (4), 318–330. 35. Mehran,
R.J.; Ricci, M.A.; Graham, A.M.; Carter, K.; Symes, J.F.
Porcine model for vascular graft studies. J. Invest. Surg.
1991, 4 (1), 37–44. 36. van der Bas, J.M.; Quax, P.H.; van
den Berg, A.C.; van Hinsbergh, V.W.; van Bockel, JH.
Ingrowth of aorta vascular cells into basic fibroblast
growth factor-impregnated vascular prosthesis material, a
porcine and human in vitro study on blood vessel prosthesis
healing. J. Vasc. Surg. 2002, 36 (6), 1237–1247. 37.
Tucker, O.P.; Syburra, T.; Augstburger, M.; van Melle, G.;
Gebhard, S.; Bosman, F.; von Segesser, L.K. Small intestine
without mucosa as a growing vascular conduit, a porcine
experimental study. J. Thorac. Cardiovasc. Surg. 2002, 124
(6), 1165–1175. 38. Lin, P.H.; Chen, C.; Surowiec, S.M.;
Conklin, B.; Bush, R.L.; Chaikof, E.L.; Lumsden, A.B.;
Weiss, V. A porcine model of carotid artery thrombosis for
thrombolytic therapy and angioplasty, application of PTFE
graft-induced stenosis. J. Endovasc. Ther. 2000, 7 (3),
227–235. 39. Bocan, T.M. Animal models of atherosclerosis
and interpretation of drug intervention studies. Curr.
Pharm. Des. 1998, 4 (1), 37–52. 40. Kleine, P.;
Abdel-Rahman, U.; Kleisus, A.A.; Sherer, M.; Simon, A.;
Moritz, A. Comparison of hemodynamic performance of
Medtronic Hall 21 mm versus St. Jude Medical 23 mm
prostheses in pigs. J. Heart Valve Dis. 2002, 11 (6),
857–863. 41. McLennan, G.; Trerotola, S.O.; Forney, M.;
Jellison, B.; Dreesen, R.G.; Tennery, J. Short-term patency
and safety of an expanded polytetrafluoroethylene
encapsulated endoluminal device at the venous anastomosis
of a canine arteriovenous graft model. J. Vasc. Interv.
Radiol. 2001, 12 (2), 227–234. 42. Omary, R.A.; Green,
J.D.; Schirf, B.E.; Li, Y.; Finn, J.P.; Li, D. Real-time
magnetic resonance imagingguided coronary catheterization
in swine. Circulation 2003, 107 (21), 2656–2659. 43.
Odenstendt, H.; Aneman, A.; Oi, Y.; Svensson, M.;
Stenqvist, O.; Lundin, S. Descending aortic blood flow and
cardiac output, a clinical and experimental study of
continous oesophageal echo-doppler flowmetry. Acta
Anaesthesiol. Scand. 2001, 45 (2), 180–187. 44. Dobrin,
P.B.; Mirande, R.; Kang, S.; Dong, Q.S.; Mrkvicka, R.
Mechanics of end-to-end artery-to-PTFE graft anastomoses.
Ann. Vasc. Surg. 1998, 12 (4), 317– 323. 45. Morasch, M.D.;
Dobrin, P.B.; Dong, Q.S.; Mrkvicka, R. Mechanics of
spatulated end-to-end artery-tovein anastomoses. Ann. Vasc.
Surg. 1998, 12 (1), 55–59. 46. Tozzi, P.; Hayoz, D.;
Ruchat, P.; Corno, A.; Oedman, C.; Botta, U.; von Segesser,
L.K. Animal model to compare the effects of suture
technique on cross-sectional compliance on end-to-side
anastomoses. Eur. J. Cardio-Thorac. Surg. 1999, 19 (4),
477–481. 47. Walpoth, B.H.; Ris, H.B.; Amonn, A.; Mettler,
D.; Schilt, W.; Hoflin, F.; Felix, R.; Schaffner, T.;
Althaus, U. Evaluation of recent vascular prostheses in the
arteriovenous shunt. Helv. Chir. Acta 1990, 57 (2),
347–350. 48. Walpoth, B.H.; Amonn, A.; Galdikas, J.; Ris,
H.B.; Schaffner, T.; Hoflin, F.; Schilt, W.; Mettler, D.;
Nachnbur, B.; Althaus, U. Experimental assessment of
thrombogenicity in vascular prostheses before and during
prostaglandin E1 treatment. Eur. J. Vasc. Surg. 1993, 7
(5), 493–499. 49. Walpoth, B.H.; Rogulenko, R.;
Tikhvinskaia, E.; Gogolewski, S.; Schaffner, T.; Hess,
O.M.; Althaus, U. Improvement of patency rate in
heparin-coated small synthetic vascular grafts. Circulation
1998, 98 (suppl. 19), II-319–II-324. 50. Edelman, E.R.
Vascular tissue engineering, designer arteries. Circ. Res.
1999, 85 (12), 1115–1118. 51. Hoerstrup, S.P.; Zund, G.;
Sodian, R.; Schnell, A.M.; Grunenfelder, J.; Turina, M.I.
Tissue engineering of small caliber vascular grafts. Eur.
J. Cardio-Thorac. Surg. 2001, 20 (1), 164–169. 52.
Ratcliffe, A. Tissue engineering of vascular grafts. Matrix
Biol. 2000, 19 (4), 353–357. 53. Teebken, O.E.; Bader, A.;
Steinhoff, G.; Haverich, A. Tissue engineering of vascular
grafts. Human cell seeding of decellularised porcine
matrix. Eur. J. Vasc. Endovasc. Surg. 2000, 19 (4),
381–386. I

54. Teebken, O.E.; Haverich, A. Tissue engineering of small


diameter vascular grafts. Eur. J. Vasc. Endovasc. Surg.
2002, 23 (6), 475–485.

55. Matsumura, G.; Hibino, N.; Ikada, Y.; Kurosawa, H.;


Shin’oka, T. Successful application of tissue engineered
vascular autografts, clinical experience. Biomaterials
2003, 24 (13), 2303–2308.

56. Bujan, J.; Garcia-Honduvilla, N.; Contreras, L.;


Gimeno, M.J.; Escudero, C.; Bellon, J.M.; San-Roman, J.
Coating PTFE vascular prostheses with fibroblastic matrix
improves cell retention when subjected to blood flow. J.
Biomed. Mater. Res. 1998, 39 (1), 32–39.

57. Zarge, J.I.; Gosselin, C.; Hauang, P.; Greisler, H.P.


Platelet deposition on ePTFE grafts coated with fibrin glue
with or without FGF-1 and heparin. J. Surg. Res. 1997, 67
(1), 4–8.

58. Zarge, J.I.; Husak, V.; Huang, P.; Greisler, H.P.


Fibrin glue containing fibroblast growth factor type 1 and
heparin decreases platelet deposition. Am. J. Surg. 1997,
174 (2), 188–192.

59. Chen, C.; Lumsden, A.B.; Ofenloch, J.C.; Noe, B.;


Campbell, E.J.; Stratford, P.W.; Yianni, Y.P.; Taylor,
A.S.; Hanson, S.R. Phosphorhylcholine coating of ePTFE
grafts reduces neointimal hyperplasia in canine model. Ann.
Vasc. Surg. 1997, 11 (1), 74–79.

60. Boland, E.D.; Wnek, G.E.; Simpson, D.G. Tailoring


tissue engineering scaffolds using electrostatic processing
techniques, a study of poly(glycolic acid). J. Macromol.
Sci. 2001, 38, 1231–1243.

61. Niu, S.; Kurumatani, H.; Satoh, S.; Kanda, K.; Oka, T.;
Watanabe, K. Small diameter vascular prostheses with
incorporated bio-absorbable matrices. A preliminary study.
ASAIO J. 1993, 39 (3), 750–753.

62. Izhar, U.; Schwalb, H.; Borman, J.B.; Hellener, G.R.;


Hotoveli-Salomon, A.; Marom, G.; Stern, T.; Cohn, D. Novel
synthetic selectively degradable vascular prostheses, a
preliminary implantation study. J. Surg. Res. 2001, 95 (2),
152–160.
63. Stitzel, J.D.; Pawlowski, K.J.; Wnek, G.E.; Simpson,
D.G.; Bowlin, G.L. Arterial smooth muscle cell
proliferation on a novel biomimicking biodegradable
vascular graft scaffold. J. Biomater. Appl. 2001, 16 (1),
22–33.

64. Levenberg, S.; Golub, J.S.; Amit, M.; Itskovitz-Eldor,


J.; Langer, R. Endothelial cells derived from human
embryonic stem cells. PNAS 2002, 99 (7), 4391–4396.

65. Kadner, A.; Hoerstrup, S.P.; Zund, G.; Eid, K.; Maurus,
C.; Melnitchouk, S.; Grunenfelder, J.; Turina, M.I. A new
source for cardiovascular tissue engineering, human bone
marrow stromal cells. Eur. J. Cardio-Thorac. Surg. 2002, 21
(6), 1055–1060.

66. Herring, M.B.; LeGrand, D.R. The histology of seeded


PTFE grafts in humans. Ann. Vasc. Surg. 1989, 3 (2),
96–103.

67. Sodian, R.; Lemke, T.; Fritsche, C.; Hoerstrup, S.P.;


Fu, P.; Potapov, E.V.; Hausmann, H.; Hetzer, R.
Tissue-engineering bioreactors: A new combined cellseeding
and perfusion system for vascular tissue engineering.
Tissue Eng. 2002, 8 (5), 863–870.

68. Walter, D.H.; Rittig, K.; Bahlmann, F.H.; Kirchmair,


R.; Silver, M.; Murayama, T.; Nishimura, H.; Losordo, D.W.;
Asahara, T.; Isner, J.M. Statin therapy accelerates
rendothelialization. A novel effect involving mobilization
and incorporation of bone-marrow-derived endothelial
progenitor cells. Circulation 2002, 105 (25), 3017–3024.
69. Weatherford, D.A.; Sackman, J.E.; Reddick, T.T.;
Freeman, M.B.; Stevens, S.L.; Goldman, M.H. Vascular
endothelial growth factor and heparin in a biologic glue
promotes human aortic endothelial cell proliferation with
aortic smooth muscle cell inhibition. Surgery 1996, 120
(2), 433–439. 70. Shi, Q.; Bhattacharya, V.; Hong-De Wu,
M.; Sauvage, L.R. Utilizing granulocyte colony-stimulating
factor to enhance vascular graft endothelialization from
circulating blood cells. Ann. Vasc. Surg. 2002, 16 (3),
314–320. 71. Bowlin, G.L.; Pawlowski, K.J.; Stitzel, J.D.
Electrospinning of Polymer Scaffolds for Tissue
Engineering. In Tissue Engineering and Biodegradable
Equivalents, Scientific and Clinical Applications;
Lewandrowsky, K., Ed.; Marcel Dekker: New York, 2002;
65–178. 72. Llavadot, J.; Murasawa, S.; Kureishi, Y.;
Uchida, S.; Masuda, H.; Kawamoto, A.; Walsh, K.; Isner,
J.M.; Asahara, T. HMG-CoA reductase inhibitor mobilizes
bone marrow-derived endothelial progenitor cells. J. Clin.
Invest. 2001, 108 (3), 399–405. 73. Beldi, G.; Bosshard,
A.; Hess, O.M.; Althaus, U.; Walpoth, B.H. Transit time flow
measurement, experimental validation and comparison of
three different systems. Ann. Thorac. Surg. 2000, 70 (1),
212–217. 74. Walpoth, B.H.; Beldi, G.; Bosshard, A.; Hess,
O.M.; Carrel, T. Transit Time Flow Measurements, From Bench
to Bedside. In Intraoperative Graft Patency Verification in
Cardiac and Vascular Surgery; D’Ancona, G., Karamanoukian,
H.L., Ricci, M., Salerno, T.A., Bergsland, J., Eds.; Futura
Publishing Company: New York, 2001; 191–200. 75. He, G.W.;
Yang, C.Q. Comparative study on calcium channel antagonists
in the human radial artery, clinical implications. J.
Thorac. Cardiovasc. Surg. 2000, 119 (1), 94–100. 76.
Contreras, M.A.; Quist, W.C.; Logerfo, F.W. Effect of
porosity on small-diameter vascular graft healing.
Microsurgery 2000, 20 (1), 15–21. 77. Wang, Z.G.; Zhang,
H.; Pu, L.Q.; Du, W.; Li, G.D.; Wu, J.D.; Wang, D.J.; Sha,
R.Y. Can endothelial seeding enhance patency and inhibit
neointimal hyperplasia? Experimental studies and clinical
trial of endothelial seeded venous prostheses. Int. Angiol.
2000, 19 (3), 259–269. 78. Valente, M.; Pettenazzo, E.; Di
Filippo, L.; Laborde, F.; Rinaldi, S.; Thiene, G.
Biodegradable polymer (D,L-lactide-caprolactone) in aortic
vascular prosthesis, morphological evaluation in an animal
model. Int. J. Artif. Organs 2002, 25 (8), 777–782. 79.
Gogolewski, S.; Walpoth, B.; Rheiner, P. Polyurethane
macroporous membranes as pericardial substitutes. Colloid
Polym. Sci. 1987, 265, 971–977. 80. Kambayashi, J.; Watase,
M.; Itoh, T.; Kawasaki, T.; Shiba, E.; Sakon, M.; Mori, T.
Blood compatibility of venous prosthesis made of textile or
non-textile material. Thromb. Res. 1992, 66 (4), 365–372.
81. Nojiri, C.; Okano, T.; Jacobs, H.A.; Park, K.D.;
Mohammad, S.F.; Olsen, D.B.; Kim, S.W. Blood compatibility
of PEO grafted polyurethane and HEMA/styrene block
copolymer surfaces. J. Biomed. Mater. Res. 1990, 24 (9),
1151–1171. 82. Vale, B.H. Current concepts for assessing
blood compatibility, small diameter vascular prostheses. J.
Biomater. Appl. 1987, 2 (1), 149–159. 83. Walpoth, B.H.;
Pavlicek, M.; Celik, B.; Nicolaus, B.; Schaffner, T.;
Althaus, U.; Hess, O.M.; Carrel, T.; Morris, R.E.
Prevention of neointimal proliferation by
immuno-suppression in synthetic vascular grafts. Eur. J.
Cardio-Thorac. Surg. 2001, 19 (4), 487–492. 84. Bearinger,
J.P.; Terrettaz, S.; Michel, R.; Tirelli, N.; Vogel, H.;
Textor, M.; Hubbell, J.A. Chemisorbed poly (propylene
sulphide)-based copolymers resist biomolecular
interactions. Nat. Mater. 2003, 2 (4), 259–264. 85. Chanda,
J.; Kuribayashi, R.; Abe, T. Heparin coupling in inhibition
of calcification of vascular bioprostheses. Biomaterials
1999, 20 (19), 1753–1757. 86. Shigematsu, K.; Zasuhara, H.;
Shigematsu, H. Topical application of antiangiogenic agent
AGM-1470 suppresses anastomotic intimal hyperplasia after
e-PTFE grafting in a rabbit model. Surgery 2001, 129 (2),
220–230. 87. Torem, S.; Schneider, P.A.; Paxton, L.D.;
Yasuda, H.; Hanson, S.R. Factors influencing acute thrombus
formation on carotid artery vascular grafts. ASAIO Trans.
1988, 34 (4), 916–920. 88. Helisch, A.; Schaper, W.
Arteriogenesis, the development and growth of collateral
arteries. Microcirculation 2003, 10 (1), 83–97. 89.
Hoffmann, R.; Mintz, G.S.; Dussaillant, G.R.; Popma, J.J.;
Pichard, A.D.; Satler, L.F.; Kent, K.M.; Griffin, J.; Leon,
M.B. Patterns and mechanisms of in-stent restenosis. A
serial intravascular ultrasound study. Circulation 1996, 94
(6), 1247–1254. 90. Hardhammar, P.A.; van Beusekom, H.M.;
Emanuelsson, H.U.; Hofma, S.H.; Albertsson, P.A.; Verdouw,
P.D.; Boersma, E.; Serruys, P.W.; van der Giessen, W.J.
Reduction in thrombotic events with heparin-coated
Palmaz–Schatz stents in normal porcine coronary arteries.
Circulation 1996, 93 (3), 423–430. 91. Sousa, J.E.; Costa,
M.A.; Sousa, A.G.; Abizaid, A.C.; Seixas, A.C.; Abizaid,
A.S.; Feres, F.; Mattos, L.A.; Falotico, R.; Jaeger, J.;
Popma, J.J.; Serruys, P.W. Two-year angiographic and
intravascular ultrasound follow-up after implantation of
sirolimus-eluting stents in human coronary arteries.
Circulation 2003, 107 (3), 381–383. 92. Kastrati, A.;
Schomig, A.; Dirschinger, J.; Mehilli, J.; von Welser, N.;
Pache, J.; Schuhlen, H.; Schilling, T.; Schmitt, C.;
Neumann, F.J. Increased risk of restenosis after placement
of gold-coated stents: Results of a randomized trial
comparing gold-coated with uncoated steel stents in
patients with coronary artery disease. Circulation 2000,
101 (21), 2478–2483. 93. Windecker, S.; Mayer, I.; De
Pasquale, G.; Maier, W.; Dirsch, O.; De Groot, P.; Wu,
Y.P.; Noll, G.; Leskosek, B.; Meier, B.; Hess, O.M.;
Working Group on Novel Surface Coating of Biomedical
Devices (SCOL). Stent coating with titanium-nitride-oxide
for reduction of neointimal hyperplasia. Circulation 2001,
104 (8), 928–933. 94. Grigoni, K.S.; Windecker, S.; Hubbel,
J.A. Stent coating for prevention of in-stent restenosis:
Biocompatible polymer versus inert metal. (Submitted). I
Kidneys, Artificial

Table 3 Comparison between IHD and CRRT

Intermittent hemodialysis Continuous renal replacement


therapy

Mainly diffusive Mainly convective

Low-flux membrane High-flux membrane

High dialysate flow Low dialysate flow

On-line dialysate

production Industrially manufactured substitution fluid

A few hours per day In theory, continuously

Technically demanding Technically less demanding

Less labor intensive Labor intensive

(From Ref. [117].) 10. Jebsen, R.H.; Tenckhoff, H.; Hoult,


J.C. Natural history of uremic polyneuropathy and the
effects of dialysis. N. Engl. J. Med. 1967, 277, 327–333.
11. Tenckhoff, H.; Curtis, F.K. Experience with maintenance
peritoneal dialysis in the home. Trans. Am. Soc. Artif.
Intern. Organs 1970, 16, 90–95. 12. Babb, A.L.; Popovich,
R.P.; Cristopher, T.G.; Scribner, B.H. The genesis of the
square-meter hour hypothesis. Trans. Am. Soc. Artif. Organs
1971, 17, 81–91. 13. Babb, A.L.; Ahmad, S.; Bergstrom, J.;
Scribner, B.H. The middle molecule hypothesis in
perspective. Am. J. Kidney Dis. 1981, 1, 46–50. 14. Lowrie,
E.G.; Laird, N.M.; Parker, T.F.; Sargent, J.A. Effect of
the hemodialysis prescription on patient morbidity. N.
Engl. J. Med. 1980, 305, 1176–1181. 15. Vanholder, R.; De
Smet, R.; Vogeleere, P.; Ringoir, S. Middle molecules:
toxicity and removal by hemodialysis and related
strategies. Artif. Organs 1995, 19 (11), 1120–1125. 16.
Rockel, A.; Gilge, U.; Liewald, A.; Heidland, A.
Elimination of low molecular weight proteins during
hemofiltration. Artif. Organs 1982, 6, 307–311. 17. Rockel,
A.; Abdelhamid, S.; Fiegel, P.; Walb, D. Elimination of low
molecular weight proteins with high flux membranes. Contr.
Nephrol. 1985, 46, 69–74. 18. Gejyo, F.; Yamada, T.; Odani,
S.; Nakagawa, Y.; Arakawa, M.; Kunitomo, T.; Kataoka, H.;
Suzuki, M.; Hirasawa, Y.; Shirahama, T.; Cohen, A.; Schmid,
K. A new form of amyloid protein associated with chronic
hemodialysis was identified as beta 2-microglobulin.
Biochem. Biophys. Res. Commun. 1985, 129 (3), 701–706. 19.
Gorevic, P.D.; Casey, T.T.; Stone, W.J.; Di Raimondo, C.R.;
Prelli, F.C.; Frangione, B. Beta-2 microglobulin is an
amyloidogenic protein in man. J. Clin. Invest. 1985, 76,
2425–2429. 20. Gorevic, P.D.; Munoz, P.C.; Casey, T.T.; Di
Raimondo, C.R.; Stone, W.J.; Prelli, F.C.; Rodrigues, M.M.;
Poulik, M.D.; Frangione, B. Polymerization of intact b 2
-microglobulin in tissue causes amyloidosis in patients on
chronic hemodialysis. Proc. Natl. Acad. Sci. U.S.A. 1986,
83, 7908–7912. 21. Colton, C.K. Analysis of membrane
processes for blood purification. Blood Purif. 1987, 5,
202–251. 22. Vanholder, R.; De Smet, R.; Hsu, C.;
Vogeleere, P.; Ringoir, S. Uremic toxicity: the middle
molecule hypothesis revisited. Semin. Nephrol. 1994, 14
(3), 205–218. 23. Anderstam, B.; Mamoun, A.H.; Sodersten,
P.; Bergstrom, J. Middle-sized molecule fractions isolated
from uremic ultrafiltrate and normal urine inhibit ingestive
behavior in the rat. J. Am. Soc. Nephrol. 1996, 7 (11),
2453–2460. 24. Vanholder, R.; Argiles, A.; Baurmeister, U.;
Brunet, P.; Clark, W.; Cohen, G.; De Deyn, P.P.; Deppisch,
R.; Descamps-Latscha, B.; Henle, T.; Jorresm, A.; Massy,
Z.A.; Rodriguez, M.; Stegmayr, B.; Stenvinkel, P.; Wratten,
M.L. Uremic toxicity: present state of the art. Int. J.
Artif. Organs. 2001, 24 (10), 695–725. 25. Vanholder, R.;
De Smet, R.; Glorieux, G.; Argiles, A.; Baurmeister, U.;
Brunet, P.; Clark, W.; Cohen, G.; De Deyn, P.P.; Deppisch,
R.; Descamps-Latscha, B.; Henle, T.; Jorres, A.; Lemke,
H.D.; Massy, Z.A.; Passlick-Deetjen, J.; Rodriguez, M.;
Stegmayr, B.; Stenvinkel, P.; Tetta, C.; Wanner, C.; Zidek,
W. Review on uremic toxins: classification, concentration,
and interindividual variability. Kidney Int. 2003, 63 (5),
1934–1943. 26. Vanholder, R.; De Smet, R. Pathophysiologic
effects of uremic retention solutes. J. Am. Soc. Nephrol.
1999, 10 (8), 1815–1823. 27. Graham, T. Osmotic force.
Philos. Trans. R. Soc. Lond. 1854, 144, 177–228. 28. Sakai,
K. Artificial kidney engineering–dialysis membrane and
dialyzer for blood purification. J. Chem. Eng. Jpn. 1997, 30
(4), 587–599. 29. Abel, J.J.; Rowntree, L.G.; Turner, B.B.
On the removal of diffusible substances from the
circulating blood of living animals by means of dialysis.
J. Pharmacol. Exp. Ther. 1914, 5, 275–316. 30. Haas, G.
Dialysis of the flowing blood in the patient. Klin.
Wochenschr. 1923, 70, 1888. 31. Kolff, W.J. New Ways of
Treating Uraemia: The Artificial Kidney, Peritoneal Lavage,
Intestinal Lavage; Churchill: London, 1947. 32. Kolff, W.J.
The beginning of the artificial kidney. Artif. Organs 1993,
17 (5), 293–299. 33. Kolff, W.J. The artificial kidney and
its effect on the development of other artificial organs.
Nat. Med. 2002, 8 (10), 1063–1065. 34. Kolff, W.J.;
Watschinger, B.; Further, B. Development of a coil kidney.
Disposable artificial kidney. J. Lab. Clin. Med. 1956, 47,
969–977. 35. Kiil, F. Development of a parallel-flow
artificial kidney in plastics. Acta Chir. Scand. Suppl.
1960, 253, 142–150. 36. Stewart, R.D.; Cerny, J.C.; Mahon,
H.I. The capillary kidney: preliminary report. Univ. Mich.
Med. Cent. J. 1964, 30, 116. 37. Poh, C.K.; Hardy, P.A.;
Clark, W.R.; Gao, D. Characterization of flow in
hemodialyzers using MRI. Proc. Intl. Soc. Mag. Reson. Med.
2001, 9, 1984. 38. Poh, C.K.; Hardy, P.A.; Liao, Z.; Huang,
Z.; Gao, D.; Clark, W.R. Characterization of flow
distribution in hemodialyzers using MRI (abstract). ASAIO
J. 2002, 48 (2), 179. 39. Poh, C.K.; Hardy, P.A.; Liao, Z.;
Clark, W.R.; Gao, D. Nonintrusive characterization of fluid
transport phenomena in hollow-fiber membrane modules using
MRI: an innovative experimental approach. In New Insights
into Membrane Science and Technology: Polymeric and
Biofunctional Membranes; Bhattacharyya, D., Butterfield,
D.A., Eds.; Elsevier Science B.V.: Amsterdam, The
Netherlands, 2003; 89–122. 40. Poh, C.K.; Hardy, P.A.;
Liao, Z.; Huang, Z.; Clark, W.R.; Gao, D. Effect of flow
baffles on the dialysate flow distribution of hollow-fiber
hemodialyzers: a nonintrusive experimental study using MRI.
J. Biomech. Eng. 2003, 125 (4), 481–489. K

41. Poh, C.K.; Hardy, P.A.; Liao, Z.; Huang, Z.; Clark,
W.R.; Gao, D. Effect of spacer yarns on the dialysate flow
distribution of hemodialyzers: a magnetic resonance imaging
study. ASAIO J. 2003, 49 (4), 440–448.

42. Leypoldt, J.K. Dialyzers and dialysis membranes:


research directions. ASAIO J. 1996, 42 (3), 133–135.

43. Tokars, J.I.; Alter, M.J.; Favero, M.S.; Moyer, L.A.;


Bland, L.A. National surveillance of dialysis associated
diseases in the United States, 1991. ASAIO J. 1993, 39 (4),
966–975.

44. Cheung, A.K.; Agodoa, L.Y.; Daugirdas, J.T.; Depner,


T.A.; Gotch, F.A.; Greene, T.; Levin, N.W.; Leypoldt, J.K.
Hemo Study Group. Effect of hemodialyzer reuse on
clearances of urea and b 2 -microglobulin. J. Am. Soc.
Nephrol. 1999, 10, 117–127.

45. Barth, R.H. Pros and cons of short, high efficiency, and
high flux dialysis. In Replacement of Renal Function by
Dialysis, 4th Ed.; Jacobs, C., Kjellstrand, C.M., Koch,
K.M., Winchester, J.F., Eds.; Kluwer Academic Publishers:
Boston, MA, 1996; 418–453.

46. Clark, W.R.; Hamburger, R.J.; Lysaght, M.J. Effect of


membrane composition and structure on solute removal and
biocompatibility in hemodialysis. Kidney Int. 1999, 56 (6),
2005–2015.

47. Lysaght, M.J. Hemodialysis membranes in transition.


Contrib. Nephrol. 1988, 61, 1–17.

48. Keshaviah, P.; Luehmann, D.; Ilstrup, K.; Collins, A.


Technical requirements for rapid high-efficiency therapies.
Artif. Organs 1986, 10 (3), 189–194.

49. Collins, A.J.; Keshaviah, P. High efficiency therapies


for clinical dialysis. In Clinical Dialysis, 2nd Ed.;
Nissenson, A.R., Fine, R.N., Gentile, D.E., Eds.; Appleton
& Lange: Norwalk, 1990; 687 pp.

50. Collins, A.J. High-flux, high-efficiency procedures. In


Principles and Practice of Dialysis; Henrich, W.L., Ed.;
Williams & Wilkins: Baltimore, 1994; 76 pp.

51. Held, P.J.; Levin, N.W.; Bovbjerg, R.R.; Pauly, M.V.;


Diamond, L.H. Mortality and duration of hemodialysis. JAMA
1991, 265, 871–875.

52. Pastan, S.; Bailey, J. Dialysis therapy. N. Engl. J.


Med. 1998, 338 (20), 1428–1437.

53. Akizawa, T.; Kinugasa, E.; Ideura, T. Classification of


dialysis membranes by performance. Contrib. Nephrol. 1995,
113, 25–31.

54. Craddock, P.R.; Fehr, J.; Dalmasso, A.P.; Bingham,


K.L.; Jacob, H.S. Hemodialysis leukopenia: pulmonary
vascular leukostasis resulting from complement activation
by dialyzer cellophane membranes. J. Clin. Invest. 1977,
59, 879–888.

55. Girndt, M.; Heisel, O.; Ko¨hler, H. Influence of


dialysis with polyamide vs haemophan haemodialysers on
monokines and complement activation during a 4month
long-term study. Nephrol. Dial. Transplant. 1999, 14,
676–682.

56. Deppisch, R.; Schmitt, V.; Bommer, J.; Hansch, G.M.;


Ritz, E.; Rauterberg, E.W. Fluid phase generation of
terminal complement complex as a novel index of
bioincompatibility. Kidney Int. 1990, 37 (2), 696–706.

57. Schaefer, R.M.; Rayterberg, E.W.; Deppisch, R.;


Vienken, J. Assembly of terminal SC5b-9 complement
complexes: a new index of blood-membrane interaction.
Miner. Electrolyte Metab. 1990, 16 (1), 73–76. 58. Valette,
P.; Thomas, M.; De´jardin, P. Adsorption of low molecular
weight proteins to hemodialysis membranes: experimental
results and simulations. Biomaterials 1999, 20, 1621–1634.
59. Morti, S.M.; Zydney, A.L. Protein-membrane interactions
during hemodialysis: effects on solute transport. ASAIO J.
1998, 44 (4), 319–326. 60. Morti, S.M.; Zydney, A.L.
Transport characteristics of different hemodialysis
membranes (abstract). ASAIO J. 2000, 46 (2), 220. 61.
Walton, D.; Cheung, A. Membrane biocompatibility. In
Clinical Dialysis, 3rd Ed.; Nissenson, A., Fine, R.,
Gentile, D., Eds.; Appleton & Lange: Norwalk, CT, 1995;
93–120. 62. Cheung, A.K.; Leypoldt, J.K. The hemodialysis
membranes: a historical perspective, current state and
future prospect. Semin. Nephrol. 1997, 17 (3), 196–213. 63.
U.S. Renal Data System. USRDS 2000 Annual Data Report:
Atlas of End-Stage Renal Disease in the United States;
National Institutes of Health, National Institute of
Diabetes and Digestive and Kidney Diseases: Bethesda, MD,
June 2000. 64. Clark, W.R.; Gao, D. Properties of membranes
used for hemodialysis therapy. Semin. Dial. 2002, 15 (1),
191–195. 65. Lucchi, L.; Bonucchi, D.; Acerbi, M. Improved
biocompatibility by modified cellulose membranes: the case
of hemophan. Artif. Organs 1989, 13, 417–421. 66. Bowry,
S.K.; Rintelen, T.H. Synthetically modified cellulose: a
cellulosic hemodialysis membrane with minimized complement
activation. ASAIO J. 1998, 44, M579–M583. 67. Ronco, C.;
Ballestri, M.; Cappelli, G. Dialysis membranes in
convective treatments. Nephrol. Dial. Transplant. 2000, 15
(suppl 2), 31–36. 68. Deppisch, R.; Gohl, H.; Smeby, L.
Microdomain structure of polymeric surfaces: potential for
improving blood treatment procedures. Nephrol. Dial.
Transplant. 1998, 13, 1354–1359. 69. Hakim, R.M.; Held,
P.J.; Stannard, D.C.; Wolfe, R.A.; Port, F.K.; Daugirdas,
J.T.; Agodoa, L. Effect of the dialysis membrane on
mortality of chronic hemodialysis patients. Kidney Int.
1996, 50 (2), 566–570. 70. La Greca, G.; Ronco, C. Vitamin
E-Bonded Membrane: A Further Step in Dialysis Optimization;
Karger AG: Basel, Switzerland, 1999. 71. Sasaki, M.;
Hosoya, N.; Saruhashi, M. Vitamin E modified cellulose
membrane. Artif. Organs 2000, 24 (10), 779–789. 72. Huraib,
S.; Tanimu, D.; Shaheen, F.; Hejaili, F.; Giles, C.;
Pagayon, V. Effect of vitamin-E-modified dialysers on
dialyser clotting, erythropoietin and heparin dosage: a
comparative crossover study. Am. J. Nephrol. 2000, 20 (5),
364–368. 73. Dhondt, A.; Vanholder, R.; Glorieux, G.;
Waterloos, M.A.; De Smet, R.; Lesaffer, G.; Lameire, N.
Vitamin E-bonded cellulose membrane and hemodialysis
bioincompatibility: absence of an acute benefit on
expression of leukocyte surface molecules. Am. J. Kidney
Dis. 2000, 36 (6), 1140–1146. 74. Shimazu, T.; Ominato, M.;
Toyama, K.; Yasuda, T.; Sato, T.; Maeba, T.; Owada, S.;
Ishida, M. Effects of a vitamin E-modified dialysis membrane
on neutrophil superoxide anion radical production. Kidney
Int. 2001, 59 (Suppl 78), S137–S143. 75. Mydlik, M.;
Derzsiova, K.; Racz, O.; Sipulova, A.; Lovasova, E.;
Petrovicova, J. A modified dialyzer with vitamin E and
antioxidant defense parameters. Kidney Int. 2001, 59 (Suppl
78), S144–S147. 76. MacGinley, R.; Westhuyzen, J.;
Saltissi, D.; Morgan, C.; Healy, H.; Thirlwell, G.K.;
Disney, A.P. Evaluation of a novel vitamin E coated
cellulosic membrane hollow fiber dialyzer. ASAIO J. 2001, 47
(1), 66–73. 77. Tsuruoka, S.; Kawaguchi, A.; Nishiki, K.;
Hayasaka, T.; Fukushima, C.; Sugimoto, K.; Saito, T.;
Fujimura, A. Vitamin E-bonded hemodialyzer improves
neutrophil function and oxidative stress in patients with
endstage renal failure. Am. J. Kidney Dis. 2002, 39 (1),
127–133. 78. Nakatan, T.; Takemoto, Y.; Tsuchida, A.K. The
effect of vitamin E-bonded dialyzer membrane on red blood
cell survival in hemodialyzed patients. Artif. Organs 2003,
27 (3), 214–217. 79. Kobayashi, S.; Moriya, H.; Aso, K.;
Ohtake, T. Vitamin E-bonded hemodialyzer improves
atherosclerosis associated with a rheological improvement
of circulating red blood cells. Kidney Int. 2003, 63 (5),
1881–1887. 80. Triolo, L.; Malaguti, M.; Ansali, F.;
Comunian, M.C.; Arcangeloni, O.; Coppolino, F.; Marrocco,
F.; Sicoli, R.; Biagini, M. Vitamin E-bonded cellulose
membrane, lipoperoxidation, and anemia in hemodialysis
patients. Artif. Cells Blood Substit. Immobil. Biotechnol.
2003, 31 (3), 185–191. 81. Colton, C.K.; Lowrie, E.G.
Hemodialysis: physical principles and technical
considerations. In The Kidney; Brenner, B.M., Rector, F.C.,
Eds.; W.B. Saunders: Philadelphia, PA, 1981; 2425–2489. 82.
Bird, R.B.; Stewart, W.E.; Lightfoot, E.N. Velocity
distributions in laminar flow. In Transport Phenomena, 1st
Ed.; Bird, R.B., Stewart, W.E., Lightfoot, E.N., Eds.; John
Wiley & Sons: New York, 1960; 34–70. 83. Clark, W.R.
Quantitative characterization of hemodialyzer solute and
water transport. Semin. Dial. 2001, 14 (1), 32–36. 84.
Lonnemann, G. Chronic inflammation in hemodialysis: the role
of contaminated dialysate. Blood Purif. 2000, 18 (3),
214–223. 85. Takeyama, T.; Sakai, Y.
Polymethylmethacrylate: one biomaterial for a series of
membrane. Contrib. Nephrol. 1999, 125, 9–24. 86. Kessler,
S.B.; Klein, E. Dialysis: theory. In Membrane Handbook; Ho,
W.S.W., Sirkar, K.K., Eds.; Van Nostrand Reinhold: New
York, 1992; 168 pp. 87. Welty, J.R.; Wicks, C.E.; Wilson,
R.E. Fundamental of Momentum, Heat, and Mass Transfer , 3rd
Ed.; John Wiley & Sons: New York, 1984; 494 pp. 88.
Werynski, A.; Waniewski, J. Theoretical description of mass
transport in medical membrane devices. Artif. Organs 1995,
19 (5), 420–427. 89. Jorstad, S.; Smeby, L.; Balstad, T.;
Wideroe, T. Removal, generation, and adsorption of b 2
-microglobulin during hemofiltration with five different
membranes. Blood Purif. 1988, 6 (2), 96–105. 90. Jindal,
K.K.; McDougall, J.; Woods, B.; Nowakowski, L.; Goldstein,
M.B. A study of the basic principles determining the
performance of several high-flux dialyzers. Am. J. Kidney
Dis. 1989, 14 (6), 507–511. 91. Klinke, B.; Rockel, A.;
Abdelhamid, S.; Fiegel, P.; Walb, D. Transmembranous
transport and adsorption of beta-2-microglobulin during
hemodialysis using polysulfone, polyacrylonitrile,
polymethylmethacrylate and cuprammonium rayon membranes.
Int. J. Artif. Organs 1989, 12 (11), 697–702. 92. Clark,
W.R.; Macias, W.L.; Molitoris, B.A.; Wang, N.H. Membrane
adsorption of b 2 -microglobulin: equilibrium and kinetic
characterization. Kidney Int. 1994, 46 (4), 1140–1146. 93.
Clark, W.R.; Macias, W.L.; Molitoris, B.A.; Wang, N.H.
Plasma protein adsorption to highly permeable hemodialysis
membranes. Kidney Int. 1995, 48 (2), 481–488. 94. Ronco,
C.; Heifetz, A.; Fox, K.; Curtin, C.; Brendolan, A.;
Gastaldon, F.; Crepaldi, C.; Fortunato, A.; Pietribasi, G.;
Caberlotto, A.; Brunello, A.; Milan Manani, S.; Zanella,
M.; La Greca, G. Beta 2-microglobulin removal by synthetic
dialysis membranes. Mechanisms and kinetics of the
molecule. Int. J. Artif. Organs 1997, 20 (3), 136–143. 95.
Clark, W.R.; Gao, D.; Ronco, C. Membranes for dialysis:
composition, structure and function. Contrib. Nephrol.
2002, 137, 70–77. 96. Lonnemann, G.; Koch, K.M. b 2
-microglobulin amyloidosis: effects of ultrapure dialysate
and type of dialyzer membrane. J. Am. Soc. Nephrol. 2002,
13 (Suppl 1), S72–S77. 97. Li, B.; Huang, Z.; Poh, C.;
Liao, Z.; Gao, D.; Clark, W.R. Prevention of
cytokine-inducing substance transport by a new high-flux
synthetic dialyzer (abstract). J. Am. Soc. Nephrol. 2001,
12, 269A. 98. Lim, V.S.; Flanigan, M.J.; Fangman, J. Effect
of hematocrit on solute removal during high efficiency
hemodialysis. Kidney Int. 1990, 37 (6), 1557–1562. 99.
Shinaberger, J.; Miller, J.; Gardner, P. Erythropoietin
alert: risks of high hematocrit hemodialysis. Trans. Am.
Soc. Artif. Intern. Organs 1988, 34, 179–184. 100. Gambro,
Inc. Polyflux s H Dialyzer Spec Sheet, 306150084 Rev. A;
Gambro Renal Products, Inc.: Lakewood, Colorado, U.S.A.,
2004. 101. Gambro, Inc. English Operator’s Manual POLYFLUX
R TM ; N 50 123 rev. 004; Gambro Renal Products, Inc.:
Lakewood, Colorado, U.S.A. 102. Lameire, N.; Van Biesen,
W.; Vanholder, R.; Colardijn, F. The place of intermittent
hemodialysis in the K treatment of acute renal failure in
the ICU patient. Kidney Int. 1998, 53 (Suppl 66),
S110–S119.

103. Bellomo, R.; Ronco, C. Continuous versus intermittent


renal replacement therapy in the intensive care unit.
Kidney Int. 1998, 53 (Suppl 66), S125–S128.

104. Mehta, R.; Letteri, J. National Kidney Foundation


Council on Dialysis. Current status of renal replacement
therapy for acute renal failure: a survey of US
nephrologists (abstract). J. Am. Soc. Nephrol. 1996, 7,
1457.

105. O’Reilly, P.; Tolwani, A. Renal replacement therapy


III: IHD, CRRT, SLED. Crit. Care. Clin. 2005, 21 (2),
367–378.

106. Zucchelli, P.; Santoro, A. Dialysis-induced


hypotension. A fresh look at Pathophysiology. Blood Purif.
1993, 11, 85–98.

107. Conger, J.D. Does hemodialysis delay recovery from


ARF? Semin. Dial. 1990, 3, 146–148.

108. Davenport, A.; Finn, R.; Goldsmith, H.J. Management of


patients with renal failure complicated by cerebral oedema.
Blood Purif. 1989, 7 (4), 203–209.

109. Davenport, A.; Will, E.J.; Davidson, A.M. Effect of


renal replacement therapy on patients with combined acute
renal failure and fulminant hepatic failure. Kidney Int.
1993, 43 (Suppl 41), S245–S251.

110. Schiffl, H.; Lang, S.M.; Konig, A.; Strasser, T.;


Haider, M.C.; Held, E. Biocompatible membranes in acute
renal failure: prospective case-controlled study. Lancet
1994, 344 (8922), 570–572.

111. Hakim, R.M.; Wingard, R.L.; Parker, R.A. Effect of the


dialysis membrane in the treatment of patients with acute
renal failure. N. Engl. J. Med. 1994, 331 (20), 1338–1342.

112. Kramer, P.; Wigger, W.; Rieger, J.; Matthaei, D.;


Scheler, F. Arteriovenous haemofiltration: a new and simple
method for treatment of over-hydrated patients resistant to
diuretics (in German). Klin. Wochenschr. 1977, 55 (22),
1121–1122.

113. Manns, M.; Sigler, M.H.; Teehan, B.P. Continuous renal


replacement therapies: an update. Am. J. Kidney Dis. 1998,
32 (2), 185–207.
114. Tominaga, G.T.; Ingegno, M.; Ceraldi, C.; Waxman, K.
Vascular complications of continuous arteriovenous
hemofiltration in trauma patients. Trauma 1993, 35, 285–289.

115. Lebedo, I. Predilution hemofiltration: a new technology


applied to an old therapy. Int. J. Artif. Organs. 1995, 18
(11), 735–742.

116. Sigler, M.H.; Teehan, B.O.; Daugirdas, J.T.; Ing, T.S.


Slow continuous therapies. In Handbook of Dialysis, 3rd
Ed.; Daugirdas, J.T., Blake, P.G., Ing, T.S., Eds.;
Lippincott Williams & Wilkins: Philadelphia, PA, 2001;
199–230.
Knee Joint: Overuse Injuries

1. Pecina, M.; Bojanic, I. Overuse Injuries of the


Musculoskeletal System, 2nd Ed.; CRC Press: Boca Raton,
2003; 189–254.

2. Fulkerson, J.P. Disorders of the Patellofemoral Joint,


3rd Ed.; Williams and Wilkins: Baltimore, 1997; 137–153.

3. Haspl, M.; Cicak, N.; Klobucar, H.; Pecina, M. Full


arthroscopic stabilization of the patella. Arthroscopy
2002, 18 (1), E2 (1–3).

4. Kujala, M.H.; Kvist, M.; Osterman, K. Knee injuries in


athletes, review of exertion injuries and retrospective
study of outpatient sports clinic material. Sports Med.
1986, 3, 447–460.

5. Pecina, M.; Bojanic, I. Surgical treatment of jumper’s


knee in top level athletes. In Sports, Medicine and Health;
Hermans, G.P.H., Ed.; Elsevier: Amsterdam, 1990; 299–304.
6. Fritschy, D.; Gautard, R. Jumper’s knee and
ultrasonography. Am. J. Sports Med. 1988, 16, 637–640. 7.
Fritschy, D.; Wallensten, R. Surgical treatment of patellar
tendinitis. Knee Surg. Sports Traumatol. Arthrosc. 1993, 1,
131–133. 8. Renne, J.W. The iliotibial band friction
syndrome. J. Bone Joint Surg. 1975, 57A, 1110–1111. 9.
Orava, S. Iliotibial tract friction syndrome in athletes—
an uncommon exertion syndrome on the lateral side of the
knee. Br. J. Sports Med. 1978, 12, 69–73. 10. Kennedy,
J.C.; Hawkins, R.J. Breaststroker’s knee. Phys. Sportsmed.
1974, 2, 33–38. 11. Orava, S.; Malinen, L.; Karpakka, J.;
Kvist, M; Leppilahti, J.; Rantanen, J.; Kujala, V.M.
Results of surgical treatment of unresolved
Osgood-Schlatter lesion. Ann. Chir. Gynecol. 2000, 89,
298–302. 12. Saddik, D.; McNally, E.G.; Richardson, M. MRI
of Hoffa’s fat pad. Skeletal Radiol. 2004, 33, 433–444. 13.
Dupont, J.Y. Synovial plicae of the knee. Controversies and
review. Clin. Sports Med. 1997, 16, 87–122. 14. Uson, J.;
Aguado, P.; Bernad, M.; Mayordomo, L.; Naredo, E.; Balsa,
A.; Martin-Mola, E. Pes anserinus tendino-bursitis: what
are we talking about? Scand. J. Rheumatol. 2000, 29,
184–186. 15. Faletti, C; De Stefano, N.; Giudice, G.;
Larciprete, M. Knee impingement syndromes. Eur. J. Radiol.
1998, 27, 60–69. 16. Robertson, A; Jones, S.C.; Paes, R.;
Chakrabarty, G. The fabella: a forgotten source of knee
pain? Knee. 2004, 11, 243–245.
Knee Joint Replacement

5. Andriacchi, T.P.G.J.O.; Fermier, B.S.; Illinois, C. The


influence of total knee replacement design on walking and
stair climbing. J. Bone Jt. Surg. 1982, 64-A (9),
1328–1335.

6. Buechel, F.F. Long-term followup after mobile bearing


total knee replacement. Clin. Orthop. 2002, 404, 40–50.

7. Marti, A. Cobalt-base alloys used in bone surgery.


Injury 2000, 31 (Suppl. 4), 18–21.

8. Dearnley, P.A. A review of metallic, ceramic and


surface-treated metals used for bearing surfaces in human
joint replacements. Proc. Inst. Mech. Eng., H: J. Eng. Med.
1999, 213, 107–135. (Part H).

9. Engh, C.A.O.C.D.; Jatsy, M.; McGovern, T.F.; Bobyn,


J.D.; Harris, W.H. Quantification of implant micromotion,
strain shielding, and bone resorption with porous-coated
anatomic medullary locking femoral prostheses. Clin.
Orthop. 1992, 285, 13–29.

10. Pilliar, R.M.L.J.M.; Maniatopoulos, C. Observations on


the effect of movement on bone ingrowth into poroussurfaced
implants. Clin. Orthop. 1986, 208, 108–113.

11. Volz, R.G.N.J.K.; Lee, R.W.; McMurtry, M.G. The


mechanical stability of various noncemented tibial
components. Clin. Orthop. 1988, 226, 38–42.

12. Stern, S.H.D.W.R.; Gilbert, J.L The effect of tibial


stem design on component micromotion in knee arthroplasty.
Clin. Orthop. 1997, 345, 44–52.

13. Lonner, J.H.K.M.; Levitz, C.; Lotke, P.A. Changes in


bone density after cemented total knee arthroplasty:
Influence of stem design. J. Arthroplast. 2001, 16 (1),
107–111.

14. Heim, C.S.; Postak, P.D.; Greenwald, A.S. Chapter 34:


Factors Influencing the Longevity of UHMWPE Tibial
Components. In Instructional Course Lectures; AAOS, 1996;
Vol. 45.

15. Bartel, D.; Burstein, A.; Toda, M.; Edwards, D. The


effect of conformity and plastic thickness on contact
stresses in metal-backed plastic implants. J. Biomech. Eng.
1985, 107, 193–199.
16. Blunn, G.; Walker, P.S.; Joshi, A.; Hardinge, K. The
dominance of cyclic sliding in producing wear in total knee
replacements. Clin. Orthop. Relat. Res. 1991, 273, 253–260.

17. Walker, P.S.; Blunn, G.; Joshi, A.; Sathasivam, S.


Modulation of Delamination by Surface Wear in Total Knees.
39th Annual Meeting of the ORS, San Francisco, CA, February
1993; 15–18.

18. Bartel, D.; Rawlinson, J.; Burstein, A.; Ranawat, C.;


Flynn, W. Stresses in polyethylene components of
contemporary total knee replacements. Clin. Orthop. Relat.
Res. 1995, 317, 76–82.

19. McKellop, H.; Clarke, I.; Markolf, K.; Amstutz, H.


Friction and wear properties of polymer, metal, and ceramic
prosthetic joint materials evaluated on a multichannel
screening device. J. Biomed. Mater. Res. 1981, 15, 619–653.

20. Baker, D.; Hastings, R.; Pruitt, L. Study of fatigue


resistance of chemical and radiation crosslinked medical
grade ultrhigh molecular weight polyethylene. J. Biomed.
Mater. Res. 1999, 6 (573–581), 573–581.

21. Farrar, D.F.; Brain, A.A. The microstructure of


ultra-high molecular weight polyethylene used in total
joint replacements. Biomaterials 1997, 18 (24), 1677–1685.
22. Kurtz, S.; Rimnac, C.M.; Bartel, D. Degradation rate of
ultra-high molecular weight polyethylene. J. Orthop. Res.
1997, 15, 57–61. 23. Pruitt, L.; Bailey, L. Factors
effecting near-threshold fatigue crack propagation
behaviour of orthopedic grade ultra high molecular weight
polyethylene. Polymer 1998, 39 (8–9), 1545–1553. 24.
Buechel, F.F.P.M.J. The New Jersey low-contact-stress knee
replacement system: Biomechanical rationale and review of
the first 123 cemented cases. Arch. Orthop. Trauma Surg.
1986, 105 (4), 197–204. 25. Goodfellow, J.; O’Connor, J.
The mechanics of the knee and prosthesis design. J. Bone
Jt. Surg. 1978, 60-B (3), 358–369. 26. Jordan, R.O.J.L.;
Voorhorst, P.E. Survivorship analysis of cementless
meniscal bearing total knee arthroplasty. Clin. Orthop.
1997, 338, 119–123. 27. Polyziodes, A.J.D.G.K.; Tsakonas,
H. The rotaglide total knee arthroplasty prosthesis design
and early results. J. Arthroplast. 1996, 11 (4), 453–459.
28. Psychoyois, V.C.R.W.; O’Connor, J.J.; Murray, D.W. Wear
of congruent meniscal bearings in unicompartmental knee
arthroplasty. J. Bone Jt. Surg. 1998, 80-B (6), 976–982.
29. Morgan-Jones, R.L.R.G.J.; Solis, G.; Parish, E.N.;
Cross, M.J. Meniscal bearing uncemented total knee
arthroplasty: Early clinical results at a minimum 2-year
review. J. Arthroplast. 2003, 18 (1), 41–44. 30. Murray,
D.W.G.; O’Connor, J.J. The oxford medial unicompartmental
arthroplasty. J. Bone Jt. Surg. 1998, 80-B (6), 983–989.
31. Argenson, J.N.O.C.J.J. Polyethylene wear in the
meniscal knee replacement. A one to nine-year retrieval
analysis of the oxford knee. J. Bone Jt. Surg. 1992, 74-B
(2), 228–232. 32. Matsuda, S.W.S.E.; Williams, V.G.;
McCarthy, D.S.; Whiteside, L. A Contact stress analysis in
meniscal bearing total knee arthroplasty. J. Arthroplast.
1998, 13 (6), 699–706. 33. Tsakonas, A.C.P.A.J. Reduction
of polyethylene wear in a congruent meniscal knee
prosthesis. Acta Orthop. Scand., Suppl. 1997, 275, 127–131.
34. Fu, F.H.C.D.; Vince, K.G. Total Knee Arthroplasty and
Disorders of Articular Cartilage. In Knee Surgery; Williams
and Wilkins: Baltimore, 1994; Vol. 2. 35. Palmer, S.H.;
Machan, S.; Parish, E.N.; Cross, M.J. Morphology of the
Distal Femur. 13th Trienniel Congress of the Asia Pacific
Orthopaedic Association, Adelaide, April 1–6, 2001. 36.
Anouchi, Y.S.W.L.; Kaiser, A.; Milliano, M. The effects of
axial rotation alignment of the femoral component on knee
stability and patella tracking in total knee arthroplasty
demonstrated on autopsy specimens. Clin. Orthop. 1993, 321,
168–172. 37. Whiteside, L.A.J. The anteroposterior axis for
femoral rotational alignment based on the
anterior-posterior axis in total knee arthroplasty is a
valgus knee: A technical note. J. Bone Jt. Surg. 1995,
77-A, 1331– 1334. K 38. Duffy, G.P.B.D.J.; Rand, J.A.
Cement versus cementless fixation in total knee
arthroplasty. Clin. Orthop. 1998, 356, 66–72. 39. McCaskie,
A.D.D.J.; Green, T.P.; et al. Randomised, prospective study
comparing cemented and cementless total knee replacement.
J. Bone Jt. Surg. 1998, 80-B (6), 971–975. 40. Onsten,
I.N.A.; Carlsson, A.S.; et al. Hydroxyapatite augmentation
of the porous coating improves fixation of tibial
components: A randomised RSA study in 116 patients. J. Bone
Jt. Surg. 1998, 80-B (3), 417–425. 41. Rorabeck,
C.H.B.R.B.; Nott, L. The cemented kinematic-II and the
non-cemented porous-coated anatomic prostheses for total
knee replacement. J. Bone Jt. Surg. 1988, 70-A (4),
483–490. 42. Rand, J.A.I.D.M. Survivorship analysis of
total knee arthroplasty. J. Bone Jt. Surg. 1991, 73-A, 397.
43. Ritter, M.A.H.S.A.; Keating, E.M.; et al. Long-term
survival analysis of a posterior cruciate-retaining total
condylar total knee arthroplasty. Clin. Orthop. 1994, 309,
136. 44. Collins, D.N.H.S.A.; Nelson, C.L.; Smith, P.S.
Porouscoated anatomic total knee arthroplasty. Clin.
Orthop. 1991, 267, 128–136. 45. Dodd, C.A.F.H.D.S.;
Krackow, K.A. Total knee arthroplasty fixation. Comparison
of the early results of paired cemented versus uncemented
porous coated anatomic knee prostheses. Clin. Orthop. 1990,
260, 66–70. 46. Rosenburg, A.G.B.R.; Galante, J.O. A
comparison of cemented and cementless fixation with the
millergalante total knee arthroplasty. Orthop. Clin. North
Am. 1989, 20 (1), 97–111. 47. Rand, J.A. Cement or
cementless fixation in total knee arthroplasty. Clin.
Orthop. 1991, 273, 52–62. 48. Jones, L.C.H.D.S. Cement
disease. Clin. Orthop. 1987, 225, 192–206. 49. Hoffman,
A.A.E.J.D.; Ferguson, R.P.; et al. Tento 14-year clinical
follow-up of the cementless Natural Knee system. Clin.
Orthop. 2001, 388, 85–94. 50. Parker, D.A.R.C.H.; Bourne,
R.B. Long-term followup of cementless versus hybrid fixation
for total knee arthroplasty. Clin. Orthop. 2001, 388,
68–76. 51. Nielsen, P.T.H.E.B.; Rechnagel, K. Cementless
total knee arthroplasty in unselected cases of
osteoarthritis and rheumatoid arthritis. J. Arthroplast.
1992, 7 (2), 137–143. 52. Ryuichi, G.A.S.; Takizawa, T.
Fixation of the NexGen HA-TCP-coated cementless, screwless
total knee arthroplasty. J. Arthroplast. 2002, 17 (4),
449–456. 53. Toksvig-Larsen, S.J.P.; Ryd, L.; et al.
Hydroxyapatiteenhanced tibial prosthetic fixation. Clin.
Orthop. 2000, 370, 192–200. 54. Nielsen, P.T.V.E.R.;
Rozing, P.M. The effect of hydroxyapatite on the
micromotion of total knee prosthesis: A prospective,
randomised, double-blind study. J. Bone Jt. Surg. 1998,
80-A, 1665–1672. 55. Basset, R.W. Result of 1000
performance knees: Cementless versus cemented fixation. J.
Arthroplast. 1998, 13 (4), 409–413. 56. Cross, M.J.R.G.J.;
Dimmick, R. The effect of hydroxyapatite on bone-prosthesis
interface in total knee arthroplasty. J. Bone Jt. Surg.
1998, 80-B (2S), 140. 57. Cross, M.J.D.P.; Chitnavis, J.;
Parish, E.N. Medium term clinical results of
hydroxyapatite-coated total knee replacements. Key Eng.
Mater. 2003, 240–242, 857–858. 58. Cross, M.J.R.G.J.; Holt,
M.; Bradbury, N.; Dixon, P.; Parish, E.N. Enhanced fixation
of uncemented knee replacement with hydroxyapatite. Key
Eng. Mater. 2003, 240–242, 773–776. 59. Palm, L.J.S.A.;
Ivarsson, I. Hydroxyapatite coating improves 8to 10-year
performance of the link RS cementless femoral stem. J.
Arthroplast. 2002, 17 (2), 172–175. 60. Oosterbos,
C.J.R.A.I.; Tonino, A.J. Hydroxyapatite coated hip
prosthesis followed up for 5 years. Int. Orthop. 2001, 25
(1), 17–21. 61. Insall, J.N.R.C.S.; Aglietti, P.; Shine,
J.A. A comparison of four models of total knee replacement
prosthesis. J. Bone Jt. Surg. 1976, 58-A, 754–765. 62.
Lequesne, M.G.M.C.; Samson, M.; et al. Indexes of severity
for osteoarthritis of the hip and knee. Validationvalue in
comparison with other assessment tests. Scand. J.
Rheumatol., Suppl. 1987, 65, 85–89. 63. Bellamy, N.B.W.W.;
Goldsmith, C.H.; et al. Validations of WOMAC: A health
status instrument for measuring clinically important
patient relevant outcomes to antirheumatic drug therapy in
patients with osteoarthritis of the hip or knee. J.
Rheumatol. 1988, 15 (12), 1833–1840. 64. Dawson, J.F.R.;
Murray, D.; et al. Questionnaire on the perceptions of
patients about total knee replacement. J. Bone Jt. Surg.
1998, 80-B (1), 63–69. 65. Cooper, C.M.T.; Coggon, D.;
Egger, P.; Dieppe, P. Occupational activity and
osteoarthritis of the knee. Ann. Rheum. Dis. 1994, 53 (2),
90–93. 66. Coggon, D.C.P.; Kellingray, S.; Barrett, D.;
McLaren, M.; Cooper, C. Occupational physical activities
and osteoarthritis of the knee. Arthritis Rheum. 2000, 43
(7), 1443–1449. 67. Ulbrich, J.R.A.; Alexander, N.B. Body
position used by healthy and frail older adults to rise
from the floor. J. Am. Geriatr. Soc. 2000, 48 (12),
1626–1632. 68. Schai, P.A.G.A.J.; Scott, R.D. Kneeling
ability after total knee arthroplasty: Perception and
reality. Clin. Orthop. 1999, 367, 195–200. 69. Palmer,
S.H.S.C.T.J.; Maguire, J.; Parish, E.N.; Cross, M.J.
Ability to kneel after total knee replacement. J. Bone Jt.
Surg. 2002, 84-B, 220–222. 70. Ritter, M.A.M.J.B. Total hip
arthroplasty: Can the patient play sports again?
Orthopaedics 1987, 10, 1447–1452. 71. Dubs, L.G.N.;
Munzinger, U. Sport after total hip arthroplasty. Arch.
Orthop. Trauma Surg. 1983, 101, 161–169. 72. Bradbury,
N.B.D.; Spoo, G.; Cross, M.J. Participation in sports after
total knee replacement. Am. J. Sports Med. 1998, 26 (4),
530–535.
Laboratory Animals: Ethics and
Regulations for Care and Use

7. Sideris, L.; MCCarthy, C.; Smith, D.H. Roots of concern


with nonhuman animals in biomedical ethics. In Animals in
Biomedical Ethics; ILAR, 1999; Vol. 40 (1), 3–14.

8. Animal Welfare Act and Animal Welfare Regulations,


United States Department of Agriculture, Animal and Plant
Health Inspection Service, August 2002.

9. Animals in Research, Health Research Extension Act of


1985, Public Law 99–158; November 20, 1985.

10. Public Health Service Policy on Humane Care and Use of


Laboratory Animals; OLAW, 2002.

11. National Research Council. Guide for the Care and Use
of Laboratory Animals; National Academy Press: Washington,
D.C., 1996.

12. American Veterinary Medical Association. Report of the


AVMA panel on euthanasia. JAVMA 2001, 218 (5), 669–696.

13. Guide for the Care and Use of Agricultural Animals in


Agricultural Research and Testing; Federation of Animal
Sciences, 1999.

14. Guidelines for the Care and Use of Mammals in


Neuroscience and Behavioral Research (NRC 2003); (http://
www.nap.edu/catalog/10732.html). 15. Council Directive on
the Approximation of Laws, Regulations and Administrative
Provisions of the Member States Regarding the Protection of
Animals Used for Experimental and Other Scientific Purposes.
European Union (Directive 86/609/EEC), 1986. (Adopted May
1999). 16. European Convention for the Protection of
Vertebrate Animals Used for Experimental and Other
Scientific Purposes. Council of Europe (Convention ETS 123),
1985 (Adopted May 1999); http://conventions.coe.int/
treaty/EN/cadreprincipal.htm. 17. Guide to the Care and Use
of Experimental Animals. Canadian Council on Animal Care,
1984, Vol. 2. (Adopted May 1999) Canadian Council on Animal
Care: Albert St., Ottawa, Ontario, Canada, K1R 1B1,
315–350; http://www.ccac.ca/english/gui_pol/guides/
english/TOC_V2.HTM. 18. Endangered Species Act, PL–101-707;
November, 1988. 19. Good Laboratory Practice for
Non-Clinical Studies, Title 21 Code of Federal Regulations,
Chapter 1: FDA, Department of Health and Human Services,
4-1-97 Ed., Subchapter A, Part 58.
Laser-Tissue Interaction

3. Ochsner, M. Photophysical and photobiological processes


in the photodynamic therapy of tumours. J. Photochem.
Photobiol., B 2000, 39 (1), 1–18.

4. Wachtlin, J.; Heimann, H.; Behme, T.; Foerster, M.H.


Long-term results after photodynamic therapy with
verteporfin for choroidal neovascularizations secondary to
inflammatory chorioretinal diseases Graefes. Arch. Clin.
Exp. Ophthalmol. 2003, 241 (11), 899–906.

5. Monroe, W.T.; McQuain, M.M.; Chang, M.S.; Alexander,


J.S.; Haselton, F.R. Targeting expression with light using
caged DNA. J. Biol. Chem. 1999, 274 (30), 20895–20900.

6. Thomsen, S.L. Pathologic analysis of photothermal and


photomechanical effects of laser-tissue interactions.
Photochem. Photobiol. 1991, 53 (6), 825–835.

7. Incropera & DeWitt. Introduction to Conduction; John


Wiley & Sons: New York, NY, 1990.

8. Vogel, A.; Venugopalan, V. Mechanisms of pulsed laser


ablation of biological tissues. Chem. Rev. 2003, 130 (2),
577–644.

9. Ratkay-Traub, I.; Ferincz, I.E.; Juhasz, T.; Kurtz,


R.M.; Krueger, R.R. First clinical results with the
femtosecond neodynium-glass laser in refractive surgery. J.
Refract. Surg. 2003, 19 (2), 94–103.

10. Oleinick, N.L.; Morris, R.L.; Belichenko, I. The role


of apoptosis in response to photodynamic therapy: What,
where, why and how. Photochem. Photobiol. Sci. 2003, 1 (1),
1–21. 11. Beckham, T.; Mackanos, M.A.; Crooke, C.;
Takahashi, T.; O’Conner-Rodwell, C.E.; Contag, C.H.;
Jansen, E.D. Assessment of cellular response to thermal
laser injury through bioluminescence imaging of hsp70.
Photochem. Photobiol. 2004, 79 (1), 76–85. 12. Desmettre,
T.; Maurage, C.A.; Mordon, S. Heat shock protein
hyperexpression on chorioretinal layers after
transpupillary thermotherapy. Invest. Ophthalmol. Vis. Sci.
2001, 42 (12), 2976–2980. 13. Wu, N.J.; Jansen, E.D.;
Davidson, J.M. Comparison of MMP-13 expression in
free-electron laser and scalpel incisions during wound
healing. J. Invest. Dermat. 2003, 21 (4), 926–932. 14.
Esenaliev, R.O.; Oraevsky, A.A.; Letokhov, V.S.; Karabutov,
A.A.; Malinsky, T.V. Studies of acoustical and shock waves
in the pulsed laser ablation of biotissue. Lasers Surg.
Med. 1993, 13 (4), 470–484. 15. Venugopalan, V.; Guerra,
A., III; Nahen, K.; Vogel, A. Role of laser-induced plasma
formation in pulsed cellular microsurgery and
micromanipulation. Phys. Rev. Lett. 2002, 88 (7), 078103.
16. Jacques, S.L. Laser-tissue interactions: Photochemical,
photothermal and photomechanical. Surg. Clin. N. Am. 1992,
72, 531–558. L
Laser Transmyocardial Revascularization

6. Frazier, O.H.; et al. Myocardial revascularization with


laser. Preliminary findings. Circulation 1995, 92 (Suppl.
9), II58–II65.

7. Burkhoff, D.; et al. Histologic appearance of


transmyocardial laser channels after 4 1/2 weeks. Ann.
Thorac. Surg. 1996, 61 (5), 1532–1534. discussion
1534–1535.

8. Krabatsch, T.; et al. Histological findings after


transmyocardial laser revascularization. J. Card. Surg.
1996, 11 (5), 326–331.

9. Kohmoto, T.; et al. Does blood flow through holmium: YAG


transmyocardial laser channels? Ann. Thorac. Surg. 1996, 61
(3), 861–868.

10. Jones, J.W.; et al. Holmium: YAG laser transmyocardial


revascularization relieves angina and improves functional
status. Ann. Thorac. Surg. 1999, 67 (6), 1596–1601.
discussion 1601–1602.

11. Al-Sheikh, T.; et al. Cardiac sympathetic denervation


after transmyocardial laser revascularization. Circulation
1999, 100 (2), 135–140.

12. Kwong, K.F.; et al. Transmyocardial laser treatment


denervates canine myocardium [see comments]. J. Thorac.
Cardiovasc. Surg. 1997, 114 (6), 883–889. Discussion
889–890.

13. Kwong, K.F.; et al. Nontransmural laser treatment


incompletely denervates canine myocardium. Circulation
1998, 98 (Suppl. 19), II67–II71. Discussion II71–II72.

14. Hirsch, G.M.; et al. Transmyocardial laser


revascularization does not denervate the canine heart. Ann.
Thorac. Surg. 1999, 68 (2), 460–468. Discussion 468–469.

15. Myers, J.; et al. Do transmyocardial and percutaneous


laser revascularization induce silent ischemia? An
assessment by exercise testing. Am. Heart J. 2002, 143 (6),
1052–1057.

16. Hughes, G.C.; et al. Induction of angiogenesis after


TMR: A comparison of holmium: YAG, CO2, and excimer lasers.
Ann. Thorac. Surg. 2000, 70 (2), 504– 509.
17. Kohmoto, T.; et al. Evidence of vascular growth
associated with laser treatment of normal canine myocardium
[see comments]. Ann. Thorac. Surg. 1998, 65 (5), 1360–1367.

18. Schweitzer, W.; et al. Transmyocardial laser


revascularization. Histopathology of laser channels in 10
postoperatively deceased patients 1 to 18 days after
treatment with a CO2 laser. Pathologe 1997, 18 (5),
374–384.

19. Gassler, N.; Rastar, F.; Hentz, M.W. Angiogenesis and


expression of tenascin after transmural laser
revascularization. Histol. Histopathol. 1999, 14 (1),
81–87.

20. Galli, M.; et al. Percutaneous transmyocardial


revascularization with holmium laser in patients with
refractory angina: A pilot feasibility study. G. Ital.
Cardiol. 1999, 29 (9), 1020–1026.

21. Fleischer, K.J.; et al. One-month histologic response


of transmyocardial laser channels with molecular
intervention. Ann. Thorac. Surg. 1996, 62 (4), 1051–1058.

22. Yamamoto, N.; et al. Angiogenesis is enhanced in


ischemic canine myocardium by transmyocardial laser
revascularization. J. Am. Coll. Cardiol. 1998, 31 (6),
1426–1433. 23. Ono, T.; et al. Scanning electron
microscopic studies on microvascular architecture of human
coronary vessels by corrosion casts: Normal and focal
necrosis. Scan Electron Microsc. 1986, (Pt. 1), 263–270.
24. Holmstrom, M.; et al. Wall motion and perfusion
analysis of transmyocardial laser revascularization. SCJ,
Scand. Cardiovasc. J. 2003, 37 (2), 91–97. 25. Burkhoff,
D.; et al. Factors correlating with risk of mortality after
transmyocardial revascularization. J. Am. Coll. Cardiol.
1999, 34 (1), 55–61. 26. Guleserian, K.J.; et al. Quality
of life and survival after transmyocardial laser
revascularization with the holmium: YAG laser. Ann. Thorac.
Surg. 2003, 75 (6), 1842–1847. Discussion 1847–1848. 27.
Hattler, B.G.; et al. Transmyocardial laser
revascularization in the patient with unmanageable unstable
angina. Ann. Thorac. Surg. 1999, 68 (4), 1203–1209. 28.
Jones, J.; Guleserian, K.J.; Camillo, C.J.; DAmiano, R.J.;
Moon, M.R. Is transmyocardial revascularization too
high-risk for high-risk patients. Ann. Thorac. Surg. 2003,
75, 1847–1848. Discussion. 29. Allen, K.B.; et al.
Comparison of transmyocardial revascularization with
medical therapy in patients with refractory angina [see
comments]. N. Engl. J. Med. 1999, 341 (14), 1029–1036. 30.
Frazier, O.H.; March, R.J.; Horvath, K.A. Transmyocardial
revascularization with a carbon dioxide laser in patients
with end-stage coronary artery disease [see comments]. N.
Engl. J. Med. 1999, 341 (14), 1021–1028. 31. Schofield,
P.M.; et al. Transmyocardial laser revascularisation in
patients with refractory angina: A randomised controlled
trial [see comments] [published erratum appears in Lancet
1999May 15;353(9165):1714]. Lancet 1999, 353 (9152),
519–524. 32. Burkhoff, D.; et al. Transmyocardial laser
revascularisation compared with continued medical therapy
for treatment of refractory angina pectoris: A prospective
randomised trial. ATLANTIC investigators. Angina
treatments-lasers and normal therapies in comparison.
Lancet 1999, 354 (9182), 885–890. 33. Diegeler, A.; et al.
Transmyocardial laser revascularization using the
Holium-YAG laser for treatment of end stage coronary artery
disease. Eur. J. CardioThorac. Surg. 1998, 13 (4), 392–397.
34. Milano, A.; et al. Transmyocardial revascularization
with a holmium laser: Preliminary results. G. Ital.
Cardiol. 1997, 27 (10), 1011–1018. 35. Horvath, K.A.; et
al. Transmyocardial laser revascularization: Results of a
multicenter trial with transmyocardial laser
revascularization used as sole therapy for end-stage
coronary artery disease. J. Thorac. Cardiovasc. Surg. 1997,
113 (4), 645–653. Discussion 653–654. 36. Moosdorf, R.; et
al. Transmyocardial laser revascularization in stable and
unstable angina pectoris. Herz 1997, 22 (4), 198–204. 37.
Burkhoff, D.; Jones, J.W.; Becker, L.C. Variability of
myocardial perfusion defects assessed by thallium201
scintigraphy in patients with coronary artery disease not
amenable to angioplasty or bypass surgery. J. Am. Coll.
Cardiol. 2001, 38 (4), 1033–1039. 38. Hughes, G.C.; et al.
Improved perfusion and contractile reserve after
transmyocardial laser revascularization in a model of
hibernating myocardium. Ann. Thorac. Surg. 1999, 67 (6),
1714–1720. 39. Donovan, C.L.; et al. Improvement in
inducible ischemia during dobutamine stress
echocardiography after transmyocardial laser
revascularization in patients with refractory angina
pectoris. J. Am. Coll. Cardiol. 1997, 30 (3), 607–612. L
Ligament Repair: Animal Models

14. Hunt, P.; Scheffler, S.U.; Unterhauser, F.N.; Weiler, A.


A model of soft-tissue graft anterior cruciate ligament
reconstruction in sheep. Arch. Orthop. Trauma Surg. 2005,
125 (4), 238–248.

15. Buma, P.; Kok, H.J.; Blankevoort, L.; Kuijpers, W.;


Huiskes, R.; Van Kampen, A. Augmentation in anterior
cruciate ligament reconstruction—a histological and
biomechanical study on goats. Int. Orthop. 2004, 28 (2),
91–96.

16. Clayton, M.L.; Weir, G.J., Jr. Experimental


investigations of ligamentous healing. J. Bone Joint Surg.
1969, 43A, 1167.

17. Ackermann, P.W.; Finn, A.; Ahmed, M. Sensory


neuropeptidergic pattern in tendon, ligament and joint
capsule. A study in the rat. Neuroreport 1999, 10 (10),
2055–2060. 18. Lo, I.K.Y.; Thornton, G.; Miniaci, A.;
Frank, C.B.; Rattner, J.B.; Bray, R.C. Structure and
function of diarthrodial joints. In Operative Arthroscopy,
3rd Ed.; McGinty, J.B., Ed.; Lippincot, Williams and
Wilkins: Philadelphia, 2003; 41–126. 19. McDougall, J.J.;
Bray, R.C.; Sharkey, K.A. Morphological and
immunohistochemical examination of nerves in normal and
injured collateral ligaments of rat, rabbit and human knee
joints. Anat. Record 1997, 248, 29–39. 20. Forrester, K.R.;
Tulip, J.; Leonard, C.; Stewart, C.; Bray, R.C. A laser
speckle imaging technique for measuring tissue perfusion.
IEEE Trans. Biomed. Eng. 2004, 51 (11), 1994–2005. 21.
Miller, D.; Forrester, K.; Leonard, C.; Salo, P.; Bray,
R.C. ACL deficiency and incipient osteoarthritis impairs the
vasoconstrictive efficacy of neuropeptide Y in articular
tissues: a laser speckle perfusion imaging study. J. Appl.
Physiol. 2005, 98 (1), 329–333.
Liver, Bio-Artificial

41. Catapano, G. Mass transfer limitations to the


performance of membrane bioartificial liver support devices.
Int. J. Artif. Organs 1996, 19, 18–35.

42. Flendrig, L.M.; la Soe, J.W.; Jorning, G.G.; Steenbeek,


A.; Karlsen, O.T.; Bovee, W.M.; Ladiges, N.C.; te Velde,
A.A.; Chamuleau, R.A. In vitro evaluation of a novel
bioreactor based on an integral oxygenator and a spirally
wound nonwoven polyester matrix for hepatocyte culture as
small aggregates. J. Hepatol. 1997, 26, 1379–1392. 43. Lee,
K.; Berthiaume, F.; Stephanopoulos, G.N.; Yarmush, M.L.
Metabolic flux analysis: A powerful tool for monitoring
tissue function. Tissue Eng. 1999, 5, 347–368.
Lung, Artificial: Basic Principles and
Current Applications

48. Shimono, T.; Shomura, Y.; Tahara, K.; Hioki, I.;


Tenpaku, H.; Maze, Y.; Hirano, R.; Shimpo, H.; Shionoya,
Y.; Yokoyama, A.; Morikan, T.; Yada, I. Experimental
evaluation of a newly developed ultrathin silicone layer
coated hollow fiber oxygenator. ASAIO J. 1996, 42 (5),
M451–M454.

49. Shimono, T.; Shomura, Y.; Hioki, I.; Shimamoto, A.;


Tenpaku, H.; Maze, Y.; Onoda, K.; Takao, M.; Shimpo, H.;
Yada, I. Silicone-coated polypropylene hollow-fiber
oxygenator: Experimental evaluation and preliminary
clinical use. Ann. Thorac. Surg. 1997, 63 (6), 1730–1736.

50. Watanabe, H.; Hayashi, J.; Ohzeki, H.; Moro, H.;


Sugawara, M.; Eguchi, S. Biocompatibility of a
silicone-coated polypropylene hollow fiber oxygenator in an
in vitro model. Ann. Thorac. Surg. 1999, 67 (5), 1315–1319.
51. Shimamoto, A.; Kanemitsu, S.; Fujinaga, K.; Takao, M.;
Onoda, K.; Shimono, T.; Tanaka, K.; Shimpo, H.; Yada, I.
Biocompatibility of silicone-coated oxygenator in
cardiopulmonary bypass. Ann. Thorac. Surg. 2000, 69 (1),
115–120. 52. Saito, N.; Motoyama, S.; Sawamoto, J. Effects
of new polymer-coated extracorporeal circuits on
biocompatibility during cardiopulmonary bypass. Artif.
Organs 2000, 24 (7), 547–554. 53. Peek, G.J.; Killer, H.M.;
Reeves, R.; Sosnowski, A.W.; Firmin, R.K. Early experience
with a polymethyl pentene oxygenator for adult
extracorporeal life support. ASAIO J. 2002, 48 (5),
480–482.
Lung, Artificial: Current Research and
Future Directions

Microfabrication Textile

Manufacturing

Bioengineering HYBRID ARTIFICIAL LUNG Components


Genetically Engineered Endothelial Cells Micron Scale
Blood Channels Structured 3-D Fiber Fabrics Biocompatible
Nonthrombogenic Noninflammatory Efficient Transfer 5-10μ
blood channels Short diffusion paths Compact Approach lung
A/V ∼ 100-1000 cm -1

Fig. 10 The future of artificial lungs. 6. Brunston, R.L.,


Jr.; Tao, W.; Bidani, A.; Alpard, S.K.; Traber, D.L.;
Zwischenberger, J.B. Prolonged hemodynamic stability during
arteriovenous carbon dioxide removal for severe respiratory
failure. J. Thorac. Cardiovasc. Surg. 1997, 114 (6),
1107–1114. 7. Zwischenberger, J.B.; Alpard, S.K.; Tao, W.;
Deyo, D.J.; Bidani, A. Percutaneous extracorporeal
arteriovenous carbon dioxide removal improves survival in
respiratory distress syndrome: A prospective randomized
outcomes study in adult sheep. J. Thorac. Cardiovasc. Surg.
2001, 121 (3), 542–551. 8. Zwischenberger, J.B.; Conrad,
S.A.; Alpard, S.K.; Grier, L.R.; Bidani, A. Percutaneous
extracorporeal arteriovenous CO2 removal for severe
respiratory failure. Ann. Thorac. Surg. 1999, 68 (1),
181–187. 9. Conrad, S.A.; Zwischenberger, J.B.; Grier,
L.R.; Alpard, S.K.; Bidani, A. Total extracorporeal
arteriovenous carbon dioxide removal in acute respiratory
failure: A phase I clinical study. Intensive Care Med.
2001, 27 (8), 1340–1351. 10. Luzsa. X-Ray Anatomy of the
Vascular System; J.B. Lippincott Company, 1974. 11. Tao,
W.; Zwischenberger, J.B.; Nguyen, T.T.; Tzouanakis, A.E.;
Matheis, E.J.; Traber, D.L.; Bidani, A. Performance of an
intravenous gas exchanger (IVOX) in a venovenous bypass
circuit. Ann. Thorac. Surg. 1994, 57 (6), 1484–1490. 12.
Conrad, S.A.; Bagley, A.; Bagley, B.; Schaap, R.N. Major
findings from the clinical trials of the intravascular
oxygenator. Artif. Organs 1994, 18 (11), 846–863. 13.
Conrad, S.A.; Zwischenberger, J.B.; Eggerstedt, J.M.;
Bidani, A. In vivo gas transfer performance of the
intravascular oxygenator in acute respiratory failure.
Artif. Organs 1994, 18 (11), 840–845. 14. Conrad, S.A.;
Eggerstedt, J.M.; Morris, V.F.; Romero, M.D. Prolonged
intracorporeal support of gas exchange with an
intravenacaval oxygenator. Chest 1993, 103 (1), 158–161.
15. Mortensen, J.D.; Berry, G. Conceptual and design
features of a practical, clinically effective intravenous
mechanical blood oxygen/carbon dioxide exchange device
(IVOX). Int. J. Artif. Organs 1989, 12 (6), 384–389. 16.
Mortensen, J.D. Intravascular oxygenator: A new alternative
method for augmenting blood gas transfer in patients with
acute respiratory failure. Artif. Organs 1992, 16 (1),
75–82. 17. Makarewicz, A.J.; Mockros, L.F.; Anderson, R.W.
A pumping intravascular artificial lung with active mixing.
ASAIO J. 1993, 39 (3), M466–M469. 18. Vaslef, S.N.;
Mockros, L.F.; Anderson, R.W. Intravascular Lung Assist
Device and Method. US Patent 5,037,383, 8-6-1991. 19.
Vaslef, S.N.; Mockros, L.F.; Anderson, R.W. Development of
an intravascular lung assist device. Trans. ASAIO 1989, 35,
660–664. 20. Makarewicz, A.J.; Mockros, L.F.; Anderson,
R.W. A dynamic intravascular artificial lung. ASAIO J. 1994,
40 (3), M747–M750. 21. Aldea, G.S.; Doursounian, M.;
O’Gara, P.; Treanor, P.; Shapira, O.M.; Lazar, H.L.;
Shemin, R.J. Heparinbonded circuits with a reduced
anticoagulation protocol in primary CABG: A prospective,
randomized study. Ann. Thorac. Surg. 1996, 62, 410–418. 22.
Lynch, W.R.; Montoya, J.P.; Brant, D.O.; Schreiner, R.J.;
Iannettoni, M.D.; Bartlett, R.H. Hemodynamic effect of a
low-resistance artificial lung in series with the native
lungs of sheep. Ann. Thorac. Surg. 2000, 69 (2), 351–356.
23. Lick, S.D.; Zwischenberger, J.B.; Alpard, S.K.; Witt,
S.A.; Deyo, D.M.; Merz, S.I. Development of an ambulatory
artificial lung in an ovine survival model. ASAIO J. 2001,
47 (5), 486–491. 24. Snider, M.T.; High, K.M.; Richard,
R.B.; Panol, G.; Campbell, E.A.; Service, C.V.; Stene,
J.K.; Ultman, J.S. Small intrapulmonary artery lung
prototypes: Design, construction, and in vitro water
testing. ASAIO J. 1994, 40 (3), M533–M539. 25. High, K.M.;
Nicholson, T.; Richard, R.B.; Panol, G.R.; Shelley, K.;
Snider, M.T. Effects of blood phase oscillation on gas
transfer in a microporous intravascular lung. ASAIO J.
1994, 40 (3), M735–M739. 26. Nodelman, V.; Baskaran, H.;
Ultman, J.S. Enhancement of O2 and CO2 transfer through
microporous hollow fibers by pressure cycling. Ann. Biomed.
Eng. 1998, 26 (6), 1044–1054. 27. Body, S.C.; FitzGerald,
D.; Voorhees, C.; Hansen, E.; Crowley, C.; Voorhees, M.E.;
Shernan, S.K. Effect of nitric oxide upon gas transfer and
structural integrity of a polypropylene membrane
oxygenator. ASAIO J. 1999, 45 (6), 550–554. 28. Conrad,
S.A.; Brown, E.G.; Grier, L.R.; Baier, J.; Blount, J.;
Heming, T.; Zwischenberger, J.B.; Bidani, A. Arteriovenous
extracorporeal carbon dioxide removal: A mathematical model
and experimental evaluation. ASAIO J. 1998, 44 (4),
267–277. 29. De Somer, F.; Van Belleghem, Y.; Foubert, L.;
Caes, F.; Francois, K.; Dubrulle, F.; Van Nooten, G.
Feasibility of a pumpless extracorporeal respiratory assist
device. J. Heart Lung Transplant. 1999, 18 (10), 1014–1017.
30. Gross, B.D.; Sacristan, E.; Peura, R.A.; Shahnarian,
A.; Devereaux, D.; Wang, H.L.; Fiddian-Green, R.
Supplemental systemic oxygen support using an intestinal
intraluminal membrane oxygenator. Artif. Organs 2000, 24
(11), 864–869. 31. Keszler, M.; Moront, M.G.; Cox, C.;
Milewski, M.; Visner, M.S. Oxygen delivery with a single
cannula tidal flow venovenous system for extracorporeal
membrane oxygenation. ASAIO J. 1995, 41 (4), 850–854. 32.
Fiore, G.B.; Costantino, M.L.; Fumero, R.; Montevecchi,
F.M. The pumping oxygenator: Design criteria and first in
vitro results. Artif. Organs 2000, 24 (10), 797–807. 33.
Watanabe, H.; Hayashi, J.; Ohzeki, H.; Moro, H.; Sugawara,
M.; Eguchi, S. Biocompatibility of a silicone-coated
polypropylene hollow fiber oxygenator in an in vitro model.
Ann. Thorac. Surg. 1999, 67 (5), 1315–1319. 34. Lund, L.
Measurement of Hollow Fiber Membrane Permeance in a
Gas–Liquid System Ph.D.; University of Pittsburgh, 2000. L

35. Zwischenberger, J.B.; Anderson, C.M.; Cook, K.E.; Lick,


S.D.; Mockros, L.F.; Bartlett, R.H. Development of an
implantable artificial lung: Challenges and progress. ASAIO
J. 2001, 47 (4), 316–320.

36. Boschetti, F.; Perlman, C.E.; Cook, K.E.; Mockros, L.F.


Hemodynamic effects of attachment modes and device design
of a thoracic artificial lung. ASAIO J. 2000, 46 (1), 42–48.

37. Cook, K.E.; Makarewicz, A.J.; Backer, C.L.; Mockros,


L.F.; Przybylo, H.J.; Crawford, S.E.; Hernandez, J.M.;
Leonard, R.J.; Mavroudis, C. Testing of an intrathoracic
artificial lung in a pig model. ASAIO J. 1996, 42 (5),
M604–M609.

38. Vaslef, S.N.; Mockros, L.F.; Cook, K.E.; Leonard, R.J.;


Sung, J.C.; Anderson, R.W. Computer-assisted design of an
implantable, intrathoracic artificial lung. Artif. Organs
1994, 18 (11), 813–817.

39. Vaslef, S.N.; Cook, K.E.; Leonard, R.J.; Mockros, L.F.;


Anderson, R.W. Design and evaluation of a new, low pressure
loss, implantable artificial lung. ASAIO J. 1994, 40 (3),
M522–M526.

40. Lick, S.D.; Zwischenberger, J.B.; Wang, D.; Deyo, D.J.;


Alpard, S.K.; Chambers, S.D. Improved right heart function
with a compliant inflow artificial lung in series with the
pulmonary circulation. Ann. Thorac. Surg. 2001, 72 (3),
899–904.

41. Zwischenberger, J.B.; Wang, D.; Lick, S.D.; Deyo, D.J.;


Alpard, S.K.; Chambers, S.D. The paracorporeal artificial
lung improves 5-day outcomes from lethal smoke/burn-induced
acute respiratory distress syndrome in sheep. Ann. Thorac.
Surg. 2002, 74 (4), 1011–1016. 42. Chambers, S.D.; Merz,
S.I.; Mcgillicuddy, J.W.; Bartlett, R.H. Development of the
MC3 Biolung. In IEEE, Proceedings of the Second Joint
EMBS/BMES Conference, Annual Fall Meeting of the Biomedical
Engineering Society, Houston, TX, October 2002; Omnipress,
2002; 1581–1582. 43. Gartner, M.; Litwak, P.; Borovetz, H.;
Griffith, B. Development of a Pumping Artificial Lung. In
IEEE, Proceedings of the Second Joint EMBS/BMES Conference;
Annual Fall Meeting of the Biomedical Engineering Society,
Houston, TX, October 2002; Omnipress, 2002; 1589–1590. 44.
Tatsumi, E.; Takano, H.; Taenaka, Y.; Nishimura, T.;
Kakuta, Y.; Nakata, M.; Tsukiya, T.; Nishinaka, T.
Development of an ultracompact integrated heart–lung assist
device. Artif. Organs 1999, 23 (6), 518–523. 45. Maloney,
J., Jr.; Buckberg, G. Mass and Thermal Transfer Means for
Use in Heart Lung Machines, Dialyzers, and Other
Applications. US Patent 5,900,142, 5-4-1999. 46.
http://www.cardiovention.com (accessed March 2003). 47.
Dasse, K.A.; Monzyk, B.F.; Gilbert, R.J. Development of a
Photolytic Artificial Lung (PAL). In ASAIO Journal Abstracts
New York, June 12, 2002; Lippincott Williams & Wilkins:
Baltimore, MD, 2002; 139.
Lung Surfactants

37. Hamvas, A.; Nogee, L.M.; Mallory, G.B.; Spray, T.L.;


Huddleston, C.B.; August, A.; Dehner, L.P.; deMello, D.E.;
Moxley, M.; Nelson, R.; Cole, F.S.; Colten, H.R. Lung
transplantation for treatment of infants with surfactant
protein B deficiency. J. Pediatr. 1997, 130, 231–239.

38. Nogee, L.M.; Dunbar, A.E.; Wert, S.E.; Askin, F.;


Hamvas, A.; Whitsett, J.A. A mutation in the surfactant
protein C gene associated with familial interstitial lung
disease. N. Engl. J. Med. 2001, 344, 573–579.

39. Glasser, S.W.; Burhans, M.S.; Korfhagen, T.R.; Na,


C.-L.; Sly, P.D.; Ross, G.F.; Ikegami, M.; Whitsett, J.A.
Altered stability of pulmonary surfactant in SP-C deficient
mice. Proc. Natl. Acad. Sci. 2001, 98, 6366–6371.

40. Jobe, A.H. Pulmonary surfactant therapy. N. Engl. J.


Med. 1993, 328, 861–868.

41. Anzueto, A.; Baughman, R.P.; Guntupalli, K.K.; Weg,


J.G.; Wiedemann, H.P.; Raventos, A.A.; Lemaire, F.; Long,
W.; Zaccardelli, D.S.; Pattishall, E.N.; Exosurf ARDS
Sepsis Study Group Aerosolized surfactant in adults with
sepsis-induced acute respiratory distress syndrome. N.
Engl. J. Med. 1996, 334, 1417–1421. 42. Hall, S.B.;
Venkitaraman, A.R.; Whitsett, J.A.; Holm, B.A.; Notter,
R.H. Importance of hydrophobic apoproteins as constituents
of clinical exogenous surfactants. Am. Rev. Respir. Dis.
1992, 145, 24–30. 43. Mizuno, K.; Ikegami, M.; Chen, C.-M.;
Ueda, T.; Jobe, A.H. Surfactant protein-B supplementation
improves in vivo function of a modified natural surfactant.
Pediatr. Res. 1995, 37, 271–276. 44. Seeger, W.; Grube, C.;
Gu¨nther, A.; Schmidt, R. Surfactant inhibition by plasma
proteins: Differential sensitivity of various surfactant
preparations. Eur. Respir. J. 1993, 6, 971–977. 45.
Walther, F.J.; Hernandez-Juviel, J.; Bruni, R.; Waring, A.
Spiking Survanta with synthetic surfactant peptides
improves oxygenation in surfactant-deficient rats. Am. J.
Respir. Crit. Care Med. 1997, 156, 855–861. 46. Turcotte,
J.G.; Lin, W.H.; Pivarnik, P.E.; Sacco, A.M.; Bermel, M.S.;
Lu, Z.; Notter, R.H. Chemical synthesis and surface
activity of lung surfactant phospholipid analogs. II.
Racemic N-substituted diether phosphonolipids. Biochim.
Biophys. Acta 1991, 1084, 1–12.
Magnetic and Electrophoretic Cell
Separation

1. Setchell, C.H. Magnetic separations in biotechnology. J.


Chem. Technol. Biotechnol. 1985, 35B, 175.

2. Todd, P.; Pretlow, T.G. The Separation of Living Cells.


In Cell Separation Science and Technology; Kompala, D.S.,
Todd, P., Eds.; ACS Symposium Series, American Chemical
Society: Washington, 1991; 1.

3. Rosensweig, R.E. Magnetic fluids. Sci. Am. 1982, 247, 124.

4. Patchett, R.A.; Kelly, A.F.; Kroll, R.G. The absorption


of bacteria to immobilized lectins. J. Appl. Bacteriol.
1991, 71, 271.

5. Olsvik, O.; Popovic, T.; Skjerve, E. Magnetic separation


techniques in diagnostic microbiology. Clin. Microbiol.
Rev. 1994, 7, 43.

6. Safarik, I.; Safarikova, M.; Forsythe, S.J. The


application of magnetic separation in applied microbiology.
J. Appl. Bacteriol. 1995, 78, 575.

7. Larsson, P.O.; Mosbach, K. Alcohol production by


magnetically immobilized yeast. Biotechnol. Lett. 1979, 12
(1), 501.

8. Safarik, I.; Safarikova, M. Scientific and Clinical


Applications of Magnetic Carriers. In Overview of Magnetic
Separations Used in Biochemical and Biotechnological
Applications; Hafeli, U., Schutt, W., Teller, J.,
Zborowski, M., Eds.; Plenum Press: New York, 1997; 323.

9. Todd, P. The Separation of Living Cells. In Cell


Separation Science and Technology; Kompala, D.S., Todd, P.,
Eds.; ACS Symposium Series, American Chemical Society:
Washington, 1991; 216.

10. Todd, P.; Cooper, R.P.; Doyle, J.F.; Dunn, S.;


Vellinger, J.; Deuser, M.S. Multistage magnetic particle
separator. J. Magn. Magn. Mater. 2001, 225, 294.

11. Zborowski, M. Physics of Magnetic Cell Sorting. In


Scientific and Clinical Applications of Magnetic Carriers;
Hafeli, U., Schutt, W., Teller, J., Zborowski, M., Eds.;
Plenum Press: New York, 1997; 205.

12. Zborowski, M.; Moore, L.R.; Sun, L.; Chalamers, J.J.


Continuous Flow Magnetic Cell Sorting Using Soluble
Immunomagnetic Label. In Scientific and Clinical
Applications of Magnetic Carriers; Hafeli, U., Schutt, W.,
Teller, J., Zborowski, M., Eds.; Plenum Press: New York,
1997; 247. 13. Ivory, C.F. The prospects for large-scale
electrophoresis. Sep. Sci. Technol. 1988, 23, 875. 14.
Cole, K.D.; Todd, P.; Srinivasan, K.; Dutta, B.K.
Freesolution electrophoresis of proteins in an improved
density gradient column and by capillary electrophoresis.
J. Chromatogr., A 1995, A707, 77. 15. Tulp, A.; Timmerman,
A.; Bornhorn, M.G. Electrophoresis; Stathakos, D., Ed.; W.
deGruuyter and Co.: Berlin, 1983; 317. 16. Albertsson, P.A.
Multistage Procedure. In Partition of Cells Particles and
Macromolecules, 2nd Ed.; Albertsson, P.A., Ed.; Wiley
intersciences: New York, 1986; 161. 17. Treffey, T.E.;
Sharp, P.T.; Walter, H.; Brooks, D.E. Thin Layer Counter
Current Distribution. In Partitioning in Aqueous Two Phase
Systems; Walter, H., Brooks, D.E., Fisher, D., Eds.;
Academic Press: Orlando, FL, 1985; 132. 18. Deuser, M.S.;
Vellinger, J.C.; Naumman, R.J.; Guinn, M.R.; Todd, P.
Apparatus for Aqueous Two-Phase Partitioning for
Terrestrial and Space Application, AIAA Life Sciences and
Biomedical Conference, Houston, TX, April 1995, 65. Book of
Abstracts 95-LS-42. 19. Todd, P.; Raghavarao, K.S.M.S.;
Sengupta, S.; Doyle, J.F.; Vellinger, J.; Deuser, M.S.
Multistage electrophoresis system for the separation of
cells, particles and solutes. Electrophoresis 2000, 21,
318. 20. Liberti, P.A.; Feeley, B.P. Analytical
Process-Scale Separation with Bioreceptor Ferrofluids and
High Gradient Magnetic Separation. In Cell Separation
Science and Technology; Kompala, D.S., Todd, P., Eds.; ACS
Symposium Series: Washington, 1991; 268. 21. Raghavarao,
K.S.M.S.; Srinivas, N.D.; Chethana, S.; Todd, P.
Field-assisted extraction of cells, particles and
macromolecules. Trends. Biotechnol. 2002, 20, 72. 22.
Munro, P.A.; Dunhill, P.; Lilly, M.D. Casein Hydrolysis in
stirred tank reactors using chymotrypsin immobilized on
magnetic supports. Biotechnol. Bioengg. 1981, 23, 677. 23.
Safarika, I.; Safarikova, M. Use of magnetic techniques for
the isolation of cells. J. Chromatogr. B. 1999, 722, 33.
24. Shinkai, M. Functional magnetic particles for medical
application. J. Biosci. 2002, 94, 606. 25. Weatherley,
L.R.; Campbell, I.; Slaughter, J.C.; Sutherland, K.M.
Electrically Enhanced Solvent Extraction of Biochemicals.
In Separations for Biotechnology; Verrall, M.S., Ed.; 1987;
341.
Magnetic Nanoparticles: Structure and
Bioapplications

16. Bergey, E.J.; Levy, L.; Wang, X.P.; Krebs, L.J.; Lal,
M.; Kim, K.S.; Pakatchi, S.; Liebow, C.; Prasad, P.N. DC
magnetic field induced magnetocytolysis of cancer cells
targeted by LH-RH magnetic nanoparticles in vitro. Biomed.
Microdevices 2002, 4 (4), 293–299.

17. Levy, L.; Sahoo, Y.; Kim, K.S.; Bergey, E.J.; Prasad,
P.N. Nanochemistry: synthesis and characterization of
multifunctional nanoclinics for biological applications.
Chem. Mat. 2002, 14 (9), 3715–3721.

18. Pankhurst, Q.A.; Connolly, J.; Jones, S.K.; Dobson, J.


Applications of magnetic nanoparticles in biomedicine. J.
Phys. D Appl. Phys. 2003, 36 (13), R167–R181.

19. Safarik, I.; Safarikova, M. Magnetic nanoparticles and


biosciences. Monatshefte Fur Chemie 2002, 133 (6), 737–759.

20. Safary´k, I.; Safary´kova, M. Scientific and Clinical


Applications of Magnetic Carriers; Hafeli, U., et al., Ed.;
Plenum Press: London, 1997. 21. Gijs, M.A.M. Magnetic bead
handling on-chip: new opportunities for analytical
applications. Microfluidics Nanofluidics 2004, 1, 22–40. 22.
Smistrup, K.; Hansen, O.; Bruus, H.; Hansen, M.F. Magnetic
separation in microfluidic systems using microfabricated
electromagnets-experiments and simulations. J. Magnetism
Magnetic Mat. 2005, 293 (1), 597–604. 23. Choi, J.W.; Ahn,
C.H.; Bhansali, S.; Henderson, H.T. A new magnetic
bead-based, filterless bio-separator with planar
electromagnet surfaces for integrated bio-detection
systems. Sens. Actuators B Chem. 2000, 68 (1-3), 34–39. 24.
Ahn, C.H.; Allen, M.G.; Trimmer, W.; Yun, Y.N.; Erramilli,
S. A fully integrated micromachined magnetic particle
separator. J. Microelectromech. Syst. 1996, 5 (3), 151–158.
25. Fletcher, D. Fine particle high-gradient magnetic
entrapment. IEEE Trans. Magnet. 1991, 27 (4), 3655–3677.
26. Furlani, E.P. Permanent Magnet and Electromechanical
Devices; Materials, Analysis and Applications; Academic
Press: New York, 2001.
Magnetic Resonance Imaging (MRI)

1. Bloch, F. Nuclear induction. Phys. Rev. 1946, 70,


460–461.

2. Purcell, E.M.; Torrey, H.C.; Pound, R.V. Resonance


absorption by nuclear magnetic moments in a solid. Phys.
Rev. 1946, 69, 37.

3. Lauterbur, P.C. Image formation by induced local


interactions: examples employing nuclear magnetic
resonance. Nature 1973, 242, 190.

4. Mansfield, P.; Grannell, P.K. NMR diffraction in solids.


J. Phys. C Sol. State Phys. 1973, 6 (22), L422–L426.

5. Haacke, E.M.; Brown, R.W.; Thompson, M.R.; Venkatesan,


R. Magnetic Resonance Imaging: Physical Principles and
Sequence Design; John Wiley & Sons Inc., 1999; 914 pp.

6. Hoult, D.I.; Ginsberg, N.S. The quantum origins of the


free induction decay signal and spin noise. J. Magn. Reson.
2001, 148 (1), 182–199.

7. Goodson, B.M. Nuclear magnetic resonance of


laserpolarized noble gases in molecules, materials, and
organisms. J. Magn. Reson. 2002, 155 (2), 157–216. 8.
Padhani, A., Ed.; Angiogenesis Imaging; Boetringer
Ingelheim, 2003; Vol. 76. 9. King, C.P.L. Molecular
imaging. J. Magn. Reson. Imag. 2002, 16 (4), special issue.
10. Thomas, et al. High-resolution fast spin echo imaging
of the human brain at 4.7 T: implementation and sequence
characteristics. Magn. Reson. Med. 2004, 51 (6), 1254–1264.
11. Filippi, M.; Falini, A.; Arnold, D.L.; Fazekas, F.;
Gonen, O.; Simon, J.H.; Dousset, V.; Savoiardo, M.;
Wolinsky, J.S. Magnetic resonance techniques for the in
vivo assessment of multiple sclerosis pathology: consensus
report of the white matter study group. J. Magn. Reson.
Imag. 2005, 21 (6), 669–675. 12. Petrella, J.R.
Neuroimaging and early diagnosis of Alzheimer disease: a
look to the future. Radiology 2003, 226 (2), 315–336. 13.
Wakana, S.; Jiang, H.Y.; Nagae-Poetscher, L.M.; van Zijil,
P.C.M.; Mori, S. Fiber tract-based atlas of human white
matter anatomy. Radiology 2004, 230 (1), 77–87. 14.
Jezzard, P.; Matthew, P.M.; Smith, S.M. Functional Magnetic
Resonance Imaging: An Introduction to Methods; Oxford
University Press, 2003. 15. Xavier, A.R.; Qureshi, A.I.;
Kirmani, J.F.; Yahia, A.M.; Bakshi, R. Neuroimaging of
stroke: a review. Southern Med. J. 2003, 96 (4), 367–379.
16. Schneider, G., Prince, M.R., Meaney, J.F., Vincent, B.,
Eds.; Magnetic Resonance Angiography: Techniques,
Indications and Practical Applications; Springer-Verlag
Italia Srl, 2005. 17. Woodard, P.K.; Stillman, A.E. Cardiac
MR and CT. J. Magn. Reson. Imag. 2004, 19 (6), special
issue. 18. Schulz, T.; Puccini, S.; Schneider, J.P.; Kahn,
T. Interventional and intraoperative MR: review and update
of techniques and clinical experience. Eur. Radiol. 2004,
14 (12), 2212–2227. 19. Bulte, J.W.M.; Kraitchman, D.L.
Iron oxide MR contrast agents for molecular and cellular
imaging. NMR Biomed. 2004, 17 (7), 484–499. 20. Blu¨mler,
P., Blu¨mich, P., Botto, R., Fukushima, E., Eds.; Spatially
Resolved Magnetic Resonance: Methods, Materials, Biology,
Rheology, Geology, Ecology, Hardware; Wiley-VCH: Winheim,
New York, Chichester, Brisbane, Singapore, Toronto, 1998. M
Magnetic Resonance Imaging in Temperature
Measurement

7. Farahani, K.; Black, K.L.; DeSalles, A.A.; Anzai, Y.;


Lufkin, R.B. Radiofrequency Ablation. In Interventional MR;
Joles, F.A., Young, I.R., Eds.; Martin Dunitz Ltd.: London,
1998; 153–157.

8. Hoffmann, A.L.; de la Rosette, J.J.; Wijkstra, H.


Intraprostatic temperature monitoring during transurethral
microwave thermotherapy: Status and future developments. J.
Endourol. 2000, 14 (8), 637–642.

9. Diederich, C.J.; Hynynen, K.H. Ultrasound technology for


hyperthermia. Ultrasound Med. Biol. 1999, 25 (6), 871–887.

10. Tacke, J. Thermal therapies in interventional MR


imaging. Cryotherapy. Neuroimaging Clin. N. Am. 2001, 11
(4), 759–765.

11. Butts, K.; Sinclair, J.; Daniel, B.L.; Wansapura, J.;


Pauly, J.M. Temperature quantitation and mapping of frozen
tissue. J. Magn. Reson. Imaging 2001, 13 (1), 99–104.

12. Chang, D.C.; Rorschach, H.E.; Nichols, B.L.; Hazlewood,


C.F. Implications of diffusion coefficient measurements for
the structure of cellular water. Ann. N.Y. Acad. Sci. 1973,
204, 434–443.

13. Le Bihan, D.; Delannoy, J.; Levin, R.L. Temperature


mapping with MR imaging of molecular diffusion: Application
to hyperthermia. Radiology 1989, 171 (3), 853–857.

14. Macfall, J.; Prescott, D.M.; Fullar, E.; Samulski, T.V.


Temperature dependence of canine brain tissue diffusion
coefficient measured in vivo with magnetic resonance
echo-planar imaging. Int. J. Hypertherm. 1995, 11 (1),
73–86.

15. Il’yasov, K.A.; Hennig, J. Single-shot


diffusion-weighted RARE sequence: Application for
temperature monitoring during hyperthermia session. J.
Magn. Reson. Imaging 1998, 8 (6), 1296–1305.

16. Parker, D.L.; Smith, V.; Sheldon, P.; Crooks, L.E.;


Fussell, L. Temperature distribution measurements in
two-dimensional NMR imaging. Med. Phys. 1983, 10 (3),
321–325.

17. Jolesz, F.A.; Bleier, A.R.; Jakab, P.; Ruenzel, P.W.;


Huttl, K.; Jako, G.J. MR imaging of laser–tissue
interactions. Radiology 1988, 168 (1), 249–253.

18. Graham, S.J.; Stanisz, G.J.; Kecojevic, A.; Bronskill,


M.J.; Henkelman, R.M. Analysis of changes in MR properties
of tissues after heat treatment. Magn. Reson. Med. 1999, 42
(6), 1061–1071.

19. de Jode, M.G.; Vale, J.A.; Gedroyc, W.M. MR-guided


laser thermoablation of inoperable renal tumors in an
open-configuration interventional MR scanner: Preliminary
clinical experience in three cases. J. Magn. Reson. Imaging
1999, 10 (4), 545–549.

20. Huber, P.E.; Jenne, J.W.; Rastert, R.; Simiantonakis,


I.; Sinn, H.P.; Strittmatter, H.J.; von Fournier, D.;
Wannenmacher, M.F.; Debus, J. A new noninvasive approach in
breast cancer therapy using magnetic resonance
imaging-guided focused ultrasound surgery. Cancer Res. 2001
, 61 (23), 8441–8447.

21. Hindman, J.C. Proton resonance shift of water in the


gas and liquid states. J. Chem. Phys. 1966, 44, 4582–4592.
22. Kuroda, K.; Tsutsumi, S. Temperature Imaging by Proton
Chemical Shift. In Non-invasive Thermometry; Miyakawa, M.,
Bolomey, J.C., Eds.; CRC: Boca Raton, 1994; 64–102. 23.
Ishihara, Y.; Calderon, A.; Watanabe, H.; Mori, K.;
Okamoto, K.; Suzuki, T.; Sato, K.; Kuroda, K.; Nakagawa,
N.; Tsutsumi, S. A precise and fast temperature mapping
using water proton chemical shift. Proc. SMRM 1992, 4803.
24. Chung, A.H.; Hynynen, K.H.; Cline, H.E.; Colucci, V.;
Oshio, K.; Jolesz, F.A. Optimization of spoiled
gradientecho phase imaging for in vivo localization of a
focused ultrasound beam. Magn. Reson. Med. 1996, 36 (5),
745–752. 25. De Poorter, J.; DeWagter, C.; De Deene, Y.;
Thomsen, C.; Stahlberg, F.; Achten, E. The proton resonance
frequency shift method compared with molecular diffusion
for quantitative measurements of two dimentional time
dependant temperature distributions in a phantom. J. Magn.
Reson., Ser. B 1994, 103, 234–241. 26. Kuroda, K.; Suzuki,
Y.; Ishihara, Y.; Okamoto, K.; Suzuki, Y. Temperature
mapping using water proton chemical shift obtained with
3D-MRSI: Feasibility in vivo. Magn. Reson. Med. 1996, 35,
20–29. 27. Fry, F.J. Intense focused ultrasound in
medicine. Eur. Urol. 1993, 23 (Suppl. 1), 2–7. 28. Atsumi,
H.; Matsumae, M.; Kaneda, M.; Muro, I.; Mamata, Y.; Komiya,
T.; Tsugu, A.; Tsugane, R. Novel laser system and laser
irradiation method reduced the risk of carbonization during
laser interstitial thermotherapy: Assessed by MR
temperature measurement. Lasers Surg. Med. 2001, 29 (2),
108–117. 29. Quesson, B.; Vimeux, F.; Salomir, R.; De
Zwart, J.A.; Moonen, C.T. Automatic control of hyperthermic
therapy based on real-time Fourier analysis of MR
temperature maps. Magn. Reson. Med. 2002, 47 (6),
1065–1072. 30. Vimeux, F.C.; De Zwart, J.A.; Palussiere,
J.; Fawaz, R.; Delalande, C.; Canioni, P.; Grenier, N.;
Moonen, C.T. Real-time control of focused ultrasound
heating based on rapid MR thermometry. Invest. Radiol.
1999, 34 (3), 190–193. 31. Smith, N.B.; Merrlees, N.K.;
Dahleh, M.; Hynynen, K.H. Control system for an MRI
compatible intracavitary ultrasound array for thermal
treatment of prostate disease. Int. J. Hypertherm. 2001, 17
(3), 271–282. 32. Behnia, B.; Suthar, M.; Webb, A.G.
Closed-loop feedback control of phased-array microwave
heating using thermal measurements from magnetic resonance
imaging concepts in magnetic resonance. Magn. Reson. Eng.
2002, 15 (1), 101–110. 33. Mulkern, R.V.; Panych, L.P.;
McDannold, N.J.; Jolesz, F.A.; Hynynen, K. Tissue
temperature monitoring with multiple gradient-echo imaging
sequences. J. Magn. Reson. Imaging 1998, 8 (2), 493–502.
34. Kuroda, K.; Mulkern, R.V.; Oshio, K.; Panych, L.P.;
Nakai, T.; Moriya, T.; Okuda, S.; Hynynen, K.; Jolesz,
F.A.; Joles, F.A. Temperature mapping using the water
proton chemical shift: Self-referenced method with
echo-planar spectroscopic imaging. Magn. Reson. Med. 2000 ,
43 (2), 220–225. 35. Chen, J.C.; Moriarty, J.A.;
Derbyshire, J.A.; Peters, R.D.; Trachtenberg, J.; Bell,
S.D.; Doyle, J.; Arrelano, R.; Wright, G.A.; Henkelman,
R.M.; Hinks, R.S.; Lok, S.Y.; Toi, A.; Kucharczyk, W.
Prostate cancer: MR imaging and thermometry during
microwave thermal ablation-initial experience. Radiology
2000, 214 (1), 290–297. 36. Carter, D.L.; MacFall, J.R.;
Clegg, S.T.; Wan, X.; Prescott, D.M.; Charles, H.C.;
Samulski, T.V. Magnetic resonance thermometry during
hyperthermia for human high-grade sarcoma. Int. J. Radiat.
Oncol. Biol. Phys. 1998, 40 (4), 815–822. 37. Hynynen, K.;
Pomeroy, O.; Smith, D.N.; Huber, P.E.; McDannold, N.J.;
Kettenbach, J.; Baum, J.; Singer, S.; Jolesz, F.A. MR
imaging-guided focused ultrasound surgery of fibroadenomas
in the breast: A feasibility study. Radiology 2001, 219
(1), 176–185. 38. Gilbert, J.C.; Onik, G.M.; Rubinsky, B.
MRI-Guided Tissue Ablation Using Cryosurgery. In
Interventional MR; Joles, F.A., Young, I.R., Eds.; Martin
Dunitz Ltd.: London, 1998; 159–167. 39. Smith, N.B.; Webb,
A.G.; Ellis, D.S.; Wilmes, L.J.; O’Brien, W.D., Jr.
Experimental verification of the theoretical in vivo
ultrasound heating using cobalt detected magnetic
resonance. IEEE Trans. Ultrason. Ferroelectr. Freq. Control
1995, 42 (4), 489–491. 40. Webb, A.G.; Smith, N.B.; Ellis,
D.S.; O’Brien, W.D., Jr. Non-invasive in vivo temperature
mapping of ultrasound heating using fluorine-based magnetic
resonance imaging agent 1995, 1, 1609–1612. M
Magnetic Resonance Microscopy

17. Heyn, C.; Bowen, C.V.; Rutt, B.; Foster, P.J. Detection
threshold of single SPIO-labeled cells with FIESTA. Magn.
Reson. Med. 2005, 53, 312–320.

18. Louie, A.Y.; Huber, M.M.; Ahrens, E.T.; Rothbacher, U.;


Moats, R.; Jacobs, R.E.; Fraser, S.E.; Meade, T.J. In vivo
visualization of gene expression using magnetic resonance
imaging. Nat. Biotech. 2000, 15, 321–325.

19. Kiessling, F.; Le-Huu, M.; Kunert, T.; Thorn, M.;


Vosseler, S.; Schmidt, K.; Hoffend, J.; Meinzer, H.P.;
Fusenig, N.E.; Semmler, W. Improved correlation of
histological data with DCE MRI parameter maps by 3D
reconstruction, reslicing and parameterization of the
histological images. Eur. Radiol. 2005, 15, 1079–1086. 20.
Weissleder, R. Molecular imaging in cancer. Science 2006,
312, 1168–1171. 21. Schroder, L.; Lowery, T.J.; Hilty, C.;
Wemmer, D.E.; Pines, A. Molecular imaging using a targeted
magnetic resonance hyperpolarized biosensor. Science 2006,
314, 446–449.
Mass Transfer in Tissue Function: Roles

3. Netter, F.H. Atlas of Human Anatomy, 2nd Ed.; Novartis,


1997.

4. Schmidt-Nielsen, K. Scaling; Cambridge, 1984.

5. Krebs, H.A. Body size and tissue respiration. Biochim.


Biophys. Acta 1950, 4, 249–269.

6. Calder, W.A.I. Size, Function and Life History; Dover


Edition; 1996.

7. White, C.R.; Seymour, R.S. Mammalian basal metabolic


rate is proportional to body mass to the 2/3 power. Proc.
Natl. Acad. Sci. 2003, 100 (7), 4046–4049.

8. Audesirk, T.; Audesirk, G. Biology: Life on Earth, 4th


Ed.; Prentice Hall, 1996.

9. Bassingthwaighte, J.B.; Liebovitch, L.S.; West, B.J.


Fractal Physiology; Oxford, 1994.

10. Schmidt-Nielsen, K. Animal Physiology, 5th Ed.;


Cambridge, 1997.

11. Johnson, L.R. Essential Medical Physiology;


Lippincott-Raven, 1998.

12. Kessel, R.G. Basic Medical Histology; Oxford, 1998.

13. Purves, M.K. Physiology of the Circulation; Cambridge,


1972.

14. Alberts, B.; Johnson, A.; Lewis, J.; Reff, H.; Roberts,
K.; Walter, P. Molecular Biology of the Cell, 4th Ed.;
Garland, 2002.

15. Palsson, B.O.; Bhatia, S.N. Tissue Engineering; Pearson


Prentice Hall, 2004.

16. Goodsell, D. The Machinery of Life; Springer, 1998.

17. Dudkina, N.V.; Eubel, H.; Keegstra, W. Structure of a


mitochondrial super complex formed by respirationchain
complexs I and III. Proc. Natl. Acad. Sci. 2005, 102 (9),
3225–3229.

18. Lagerholm, B.C.; Weinreb, G.E.; Jacobsen, G.E.;


Thompson, N.L. Detecting microdomains in intact cell
membranes. Annu. Rev. Phys. Chem. 2005, 56, 309–336.

19. Raman, R.; Sasikharan, V.; Sasikharan, R. Structural


insights into biological roles of protein-glycosaminoglycan
interactions. Chem. Biol. 2005, 12 (3), 267–277. 20.
Welling, P.G. Pharmacokinetics; 1997. 21. Beard, D.A.;
Bassingthwaighte, J.B. Power-law kinetcs of tracer washout
from physiological systems. Ann. Biomed. Eng. 1998, 26,
775–779. 22. Bassingthwaighte, J.B., Goresky, C.A.,
Linehan, J.H., Eds. Whole Organ Approaches to Cellular
Metabolism; Springer, 1998. 23. Kroll, K.; Wilke, N.;
Jerosch-Herold, M. Modeling regional myocardial flows from
residue functions of an intravascular indicator. Am. J.
Physiol. 1996, 271 (4), H1643–H1655. 24. Alcorn, J.;
McNamara, P.J. Pharmacokinetics in the newborn. Adv. Drug
Deliv. Rev. 2003, 55 (5), 667–686. 25. Perelson, A.S.
Modeling viral and immune system dynamics. Nature Rev.
Immunol. 2002, 2 (Jan 1), 29–35. 26. Hlastala, M.P.;
Berger, A.J. Physiology of Respiration; Oxford, 1996. 27.
Schlesinger, F.B. Comparative deposition of inhaled
aerosols in experimemtal animals and humans. J. Toxicol.
Environ. Health 1985, 13, 197–214. 28. Fung, Y.C.
Biomechanics Circulation, 2nd Ed.; Springer, 1997. 29.
Lightfoot, E.N.; Duca, K.A. The roles of mass transfer in
tissue function. In The Biomedical Engineering Handbook,
2nd Ed.; Bronzino, J.D., CRC Press: 2000; II, 115–1–115-15.
30. Okamoto, G.H.; Lightfoot, E.N. Energy cost of
intracellular organization. Ind. Drug. Chem. Res. 1992, 31
(3), 732–735. 31. Weisz, P.B. Diffusion and Chemical
Transformation Science. 1973, 179, 433–440. 32. de Duve, C.
Blueprint for a Cell; Patterson, 1991. 33. Beard, D.A.;
Bassingthwaighte, J.B. Modeling advection and diffusion of
oxygen in complex vascular networks. Ann. Biomed. Eng.
2001, 29.
Matrix Metalloproteinases

1. Zisch, A.H.; Lutolf, M.P.; Hubbell, J.A. Biopolymeric


delivery matrices for angiogenic growth factors.
Cardiovasc. Pathol. 2003, 12 (6), 295–310.

2. Woessner, J.F.; Nagase, H. Matrix Metalloproteinases and


TIMPs; Oxford University Press: New York, 2000; 1.

3. Woessner, J.F., Jr. The Matrix Metalloproteinase Family.


In Matrix Metalloproteinases; Parks, W.C., Mecham, R.P.,
Eds.; Academic Press: San Diego, 1998; 1–13.

4. Gross, J.; Lapiere, C.M. Collagenolytic activity in


amphibian tissues: A tissue culture assay. Proc. Natl.
Acad. Sci. 1962, 48, 1014–1022.

5. Werb, Z.; Chin, J.R. Extracellular matrix remodeling


during morphogenesis. Ann. N. Y. Acad Sci. 1998, 857,
110–118.

6. Cai, D.H.; Vollberg, T.M., Sr.; Hahn-Dantona, E.;


Quigley, J.P.; Brauer, P.R. MMP-2 expression during early
avian cardiac and neural crest morphogenesis. Anat. Rec.
2000, 259 (2), 168–179.

7. Itoh, Y.; Nagase, H. Matrix metalloproteinases in


cancer. Essays Biochem. 2002, 38, 21–36.

8. Uchida, M.; Shima, M.; Shimoaka, T.; Fujieda, A.; Obara,


K.; Suzuki, H.; Nagai, Y.; Ikeda, T.; Yamato, H.;
Kawaguchi, H. Regulation of matrix metalloproteinases
(MMPs) and tissue inhibitors of metalloproteinases (TIMPs)
by bone resorptive factors in osteoblastic cells. J. Cell.
Physiol. 2000, 185 (2), 207–214.

9. Henry, M.T.; McMahon, K.; Mackarel, A.J.; Prikk, K.;


Sorsa, T.; Maisi, P.; Sepper, R.; Fitzgerald, M.X.;
O’Connor, C.M. Matrix metalloproteinases and tissue
inhibitor of metalloproteinase-1 in sarcoidosis and IPF.
Eur. Respir. J. 2002, 20 (5), 1220–1227. 10. Sun, H.B.;
Yokota, H. Messenger-RNA expression of matrix
metalloproteinases, tissue inhibitors of
metalloproteinases, and transcription factors in rheumatic
synovial cells under mechanical stimuli. Bone 2001, 28 (3),
303–309. 11. Cawston, T. Matrix metalloproteinases and
TIMPs: Properties and implications for the rheumatic
diseases. Mol. Med. Today 1998, 4 (3), 130–137. 12. Ladwig,
G.P.; Robson, M.C.; Liu, R.; Kuhn, M.A.; Muir, D.F.;
Schultz, G.S. Ratios of activated matrix
metalloproteinase-9 to tissue inhibitor of matrix
metalloproteinase-1 in wound fluids are inversely correlated
with healing of pressure ulcers. Wound Repair Regen. 2002,
10 (1), 26–37. 13. Mirastschijski, U.; Impola, U.; Jahkola,
T.; Karlsmark, T.; MS, A.G.; Saarialho-Kere, U. Ectopic
localization of matrix metalloproteinase-9 in chronic
cutaneous wounds. Human Pathol. 2002, 33 (3), 355–364. 14.
Fromigue, O.; Louis, K.; Wu, E.; Belhacene, N.; Loubat, A.;
Shipp, M.; Auberger, P.; Mari, B. Active stromelysin-3
(MMP-11) increases MCF-7 survival in threedimensional
Matrigel culture via activation of p42/p44 MAP-kinase. Int.
J. Cancer 2003, 106 (3), 355–363. 15. Pan, W.; Arnone, M.;
Kendall, M.; Grafstrom, R.H.; Seitz, S.P.; Wasserman, Z.R.;
Albright, C.F. Identification of peptide substrates for
human MMP-11 (stromelysin-3) using phage display. J. Biol.
Chem. 2003 , 278 (30), 27820–27827. 16. d’Ortho, M.P.;
Will, H.; Atkinson, S.; Butler, G.; Messent, A.;
Gavrilovic, J.; Smith, B.; Timpl, R.; Zardi, L.; Murphy, G.
Membrane-type matrix metalloproteinases 1 and 2 exhibit
broad-spectrum proteolytic capacities comparable to many
matrix metalloproteinases. Eur. J. Biochem. 1997, 250 (3),
751–757. 17. Shimada, T.; Nakamura, H.; Ohuchi, E.; Fujii,
Y.; Murakami, Y.; Sato, H.; Seiki, M.; Okada, Y.
Characterization of a truncated recombinant form of human
membrane type 3 matrix metalloproteinase. Eur. J. Biochem.
1999, 262 (3), 907–914. 18. Stracke, J.O.; Fosang, A.J.;
Last, K.; Mercuri, F.A.; Pendas, A.M.; Llano, E.; Perris,
R.; Di Cesare, P.E.; Murphy, G.; Knauper, V. Matrix
metalloproteinases 19 and 20 cleave aggrecan and cartilage
oligomeric matrix protein (COMP). FEBS Lett. 2000, 478
(1–2), 52–56. 19. Bartlett, J.D.; Ryu, O.H.; Xue, J.;
Simmer, J.P.; Margolis, H.C. Enamelysin mRNA displays a
developmentally defined pattern of expression and encodes a
protein which degrades amelogenin. Connect. Tissue Res.
1998, 39 (1–3), 101–109. Discussion 141–149. 20. Armstrong,
D.G.; Jude, E.B. The role of matrix metalloproteinases in
wound healing. J. Am. Podiatr. Med. Assoc. 2002, 92 (1),
12–18. 21. Nwomeh, B.C.; Liang, H.X.; Cohen, I.K.; Yager,
D.R. MMP-8 is the predominant collagenase in healing wounds
and nonhealing ulcers. J. Surg. Res. 1999, 81 (2), 189–195.
22. Dollery, C.M.; McEwan, J.R.; Henney, A.M. Matrix
metalloproteinases and cardiovascular disease. Circ. Res.
1995, 77 (5), 863–868. 23. Robbins, J.R.; McGuire, P.G.;
Wehrle-Haller, B.; Rogers, S.L. Diminished matrix
metalloproteinase 2 (MMP-2) in ectomesenchyme-derived
tissues of the Patch mutant mouse: Regulation of MMP-2 by
PDGF and effects on mesenchymal cell migration. Dev. Biol.
1999, 212 (2), 255–263. 24. Johansson, N.; Ahonen, M.;
Kahari, V.M. Matrix metalloproteinases in tumor invasion.
Cell. Mol. Life Sci. 2000, 57 (1), 5–15. 25. Cleutjens,
J.P. The role of matrix metalloproteinases in heart
disease. Cardiovasc. Res. 1996, 32 (5), 816–821. 26.
Sternlicht, M.D.; Werb, Z. How matrix metalloproteinases
regulate cell behavior. Annu. Rev. Cell Dev. Biol. 2001,
17, 463–516. 27. Wang, X.; Pei, D. Shedding of membrane
type matrix metalloproteinase 5 by a furin-type convertase:
A potential mechanism for down-regulation. J. Biol. Chem.
2001, 276 (38), 35953–35960. 28. Kato, Y.P.; Silver, F.H.
Formation of continuous collagen fibres: Evaluation of
biocompatibility and mechanical properties. Biomaterials
1990, 11 (3), 169–175. 29. Suzuki, S.; Matsuda, K.;
Isshiki, N.; Tamada, Y.; Yoshioka, K.; Ikada, Y. Clinical
evaluation of a new bilayer ‘‘artificial skin’’ composed of
collagen sponge and silicone layer. Br. J. Plast. Surg.
1990, 43 (1), 47–54. 30. Liu, X.; Wu, H.; Byrne, M.;
Jeffrey, J.; Krane, S.; Jaenisch, R. A targeted mutation at
the known collagenase cleavage site in mouse type I
collagen impairs tissue remodeling. J. Cell Biol. 1995, 130
(1), 227–237. 31. Sorsa, T.; Konttinen, Y.T.; Lindy, O.;
Ritchlin, C.; Saari, H.; Suomalainen, K.; Eklund, K.K.;
Santavirta, S. Collagenase in synovitis of rheumatoid
arthritis. Semin. Arthritis Rheum. 1992, 22 (1), 44–53. 32.
Simpson, J.W.; Taylor, A.C. Regulation of gingival
collagenase: A possible role for a mast cell factor. Proc.
Soc. Exp. Biol. Med. 1974, 145 (1), 42–47. 33. Kruze, D.;
Wojtecka, E. Activation of leucocyte collagenase proenzyme
by rheumatoid synovial fluid. Biochim. Biophys. Acta 1972,
285 (2), 436–446. 34. Tetlow, L.C.; Adlam, D.J.; Woolley,
D.E. Matrix metalloproteinase and proinflammatory cytokine
production by chondrocytes of human osteoarthritic
cartilage: Associations with degenerative changes.
Arthritis Rheum. 2001, 44 (3), 585–594. 35. Shingleton,
W.D.; Hodges, D.J.; Brick, P.; Cawston, T.E. Collagenase: A
key enzyme in collagen turnover. Biochem. Cell. Biol. 1996,
74 (6), 759–775. 36. Hasty, K.A.; Reife, R.A.; Kang, A.H.;
Stuart, J.M. The role of stromelysin in the cartilage
destruction that accompanies inflammatory arthritis.
Arthritis Rheum. 1990, 33 (3), 388–397. 37. Reynolds, J.J.
Collagenases and tissue inhibitors of metalloproteinases: A
functional balance in tissue degradation. Oral Dis. 1996, 2
(1), 70–76. 38. Claesson, R.; Johansson, A.; Belibasakis,
G.; Hanstrom, L.; Kalfas, S. Release and activation of
matrix metalloproteinase 8 from human neutrophils triggered
by the leukotoxin of Actinobacillus actinomycetemcomitans.
J. Periodontal Res. 2002, 37 (5), 353–359. 39. Bailey, A.J.
Perspective article: The fate of collagen implants in
tissue defects. Wound Repair Regen. 2000, 8 (1), 5–12. 40.
Luttun, A.; Dewerchin, M.; Collen, D.; Carmeliet, P. The
role of proteinases in angiogenesis, heart development,
restenosis, atherosclerosis, myocardial ischemia, and
stroke: Insights from genetic studies. Curr. Atheroscler.
Rep. 2000, 2 (5), 407–416. 41. Nguyen, M.; Arkell, J.;
Jackson, C.J. Human endothelial gelatinases and
angiogenesis. Int. J. Biochem. Cell Biol. 2001, 33 (10),
960–970. 42. Murphy, G.; Nguyen, Q.; Cockett, M.I.;
Atkinson, S.J.; Allan, J.A.; Knight, C.G.; Willenbrock, F.;
Docherty, A.J. Assessment of the role of the
fibronectin-like domain of gelatinase A by analysis of a
deletion mutant. J. Biol. Chem. 1994, 269 (9), 6632–6636.
43. Shipley, J.M.; Doyle, G.A.; Fliszar, C.J.; Ye, Q.Z.;
Johnson, L.L.; Shapiro, S.D.; Welgus, H.G.; Senior, R.M.
The structural basis for the elastolytic activity of the
92-kDa and 72-kDa gelatinases. Role of the fibronectin type
II-like repeats. J. Biol. Chem. 1996, 271 (8), 4335–4341.
44. O’Farrell, T.J.; Pourmotabbed, T. The fibronectin-like
domain is required for the type V and XI collagenolytic
activity of gelatinase B. Arch. Biochem. Biophys. 1998, 354
(1), 24–30. 45. O’Farrell, T.J.; Pourmotabbed, T.
Identification of structural elements important for matrix
metalloproteinase type V collagenolytic activity as
revealed by chimeric enzymes. Role of fibronectin-like
domain and active site of gelatinase B. J. Biol. Chem.
2000, 275 (36), 27964–27972. 46. Hawkes, S.P.; Hongxia, L.;
Taniguchi, G.T. Matrix Metalloproteinase Protocols, in
Methods in Molecular Biology; Clark, I.M., Ed.; Humana
Press: Totowa, NJ, 2001. 47. Bischof, P.; Meisser, A.;
Campana, A. Control of MMP-9 expression at the
maternal-fetal interface. J. Reprod. Immunol. 2002, 55
(1–2), 3–10. 48. Lijnen, H.R. Extracellular proteolysis in
the development and progression of atherosclerosis.
Biochem. Soc. Trans. 2002, 30 (2), 163–167. 49. Galis,
Z.S.; Khatri, J.J. Matrix metalloproteinases in vascular
remodeling and atherogenesis: The good, the bad, and the
ugly. Circ. Res. 2002, 90 (3), 251–262. 50. Chang, Y.C.;
Yang, S.F.; Lai, C.C.; Liu, J.Y.; Hsieh, Y.S. Regulation of
matrix metalloproteinase production by cytokines,
pharmacological agents and periodontal pathogens in human
periodontal ligament fibroblast cultures. J. Periodontal
Res. 2002, 37 (3), 196–203. 51. Opdenakker, G.; Van Damme,
J. Chemokines and proteinases in autoimmune diseases and
cancer. Verh. K. Acad. Geneeskd. Belg. 2002 , 64 (2),
105–136. 52. Giannelli, G.; Antonaci, S. Gelatinases and
their inhibitors in tumor metastasis: From biological
research to medical applications. Histol. Histopathol.
2002, 17 (1), 339–345. 53. Strongin, A.Y.; Collier, I.;
Bannikov, G.; Marmer, B.L.; Grant, G.A.; Goldberg, G.I.
Mechanism of cell surface activation of 72-kDa type IV
collagenase. Isolation of the activated form of the
membrane metalloprotease. J. Biol. Chem. 1995, 270 (10),
5331–5338. M
54. Sato, H.; Takino, T.; Okada, Y.; Cao, J.; Shinagawa,
A.; Yamamoto, E.; Seiki, M. A matrix metalloproteinase
expressed on the surface of invasive tumour cells. Nature
1994, 370 (6484), 61–65.

55. Li, H.; Simon, H.; Bocan, T.M.; Peterson, J.T. MMP/
TIMP expression in spontaneously hypertensive heart failure
rats: The effect of ACE-and MMP-inhibition. Cardiovasc.
Res. 2000, 46 (2), 298–306.

56. Vu, T.H.; Shipley, J.M.; Bergers, G.; Berger, J.E.;


Helms, J.A.; Hanahan, D.; Shapiro, S.D.; Senior, R.M.;
Werb, Z. MMP-9/gelatinase B is a key regulator of growth
plate angiogenesis and apoptosis of hypertrophic
chondrocytes. Cell 1998, 93 (3), 411–422.

57. Ramos-DeSimone, N.; Hahn-Dantona, E.; Sipley, J.;


Nagase, H.; French, D.L.; Quigley, J.P. Activation of
matrix metalloproteinase-9 (MMP-9) via a converging
plasmin/stromelysin-1 cascade enhances tumor cell invasion.
J. Biol. Chem. 1999, 274 (19), 13066–13076.

58. Howard, E.W.; Bullen, E.C.; Banda, M.J. Preferential


inhibition of 72-and 92-kDa gelatinases by tissue inhibitor
of metalloproteinases-2. J. Biol. Chem. 1991, 266 (20),
13070–13075.

59. Ogata, Y.; Itoh, Y.; Nagase, H. Steps involved in


activation of the pro-matrix metalloproteinase 9
(progelatinase B)-tissue inhibitor of metalloproteinases-1
complex by 4-aminophenylmercuric acetate and proteinases.
J. Biol. Chem. 1995, 270 (31), 18506–18511.

60. Devy, L.; Noel, A.; Baramova, E.; Bajou, K.;


Trentesaux, C.; Jardillier, J.C.; Foidart, J.M.;
Jeannesson, P. Production and activation of matrix
metalloprotease-9 (MMP-9) by HL-60 promyelocytic leukemia
cells. Biochem. Biophys. Res. Commun. 1997, 238 (3),
842–846.

61. Irigoyen, J.P.; Munoz-Canoves, P.; Montero, L.;


Koziczak, M.; Nagamine, Y. The plasminogen activator
system: Biology and regulation. Cell. Mol. Life Sci. 1999,
56 (1–2), 104–132.

62. Woessner, J.F., Jr. Matrix metalloproteinases and their


inhibitors in connective tissue remodeling. FASEB J. 1991,
5 (8), 2145–2154. 63. Meikle, M.C.; Bord, S.; Hembry, R.M.;
Compston, J.; Croucher, P.I.; Reynolds, J.J. Human
osteoblasts in culture synthesize collagenase and other
matrix metalloproteinases in response to osteotropic
hormones and cytokines. J. Cell. Sci. 1992, 103 (Pt 4),
1093–1099. 64. Saarialho-Kere, U.K.; Pentland, A.P.;
Birkedal-Hansen, H.; Parks, W.C.; Welgus, H.G. Distinct
populations of basal keratinocytes express stromelysin-1
and stromelysin-2 in chronic wounds. J. Clin. Invest. 1994,
94 (1), 79–88. 65. Bullard, K.M.; Lund, L.; Mudgett, J.S.;
Mellin, T.N.; Hunt, T.K.; Murphy, B.; Ronan, J.; Werb, Z.;
Banda, M.J. Impaired wound contraction in
stromelysin-1-deficient mice. Ann. Surg. 1999, 230 (2),
260–265. 66. Bullard, K.M.; Mudgett, J.; Scheuenstuhl, H.;
Hunt, T.K.; Banda, M.J. Stromelysin-1-deficient fibroblasts
display impaired contraction in vitro. J. Surg. Res. 1999,
84 (1), 31–34. 67. Polette, M.; Birembaut, P. Membrane-type
metalloproteinases in tumor invasion. Int. J. Biochem. Cell
Biol. 1998, 30 (11), 1195–1202. 68. Matthews, J.A.; Wnek,
G.E.; Simpson, D.G.; Bowlin, G.L. Electrospinning of
collagen nanofibers. Biomacromolecules 2002, 3 (2), 232–238.
69. Matthews, J.A.; Boland, E.D.; Wnek, G.E.; Simpson,
D.G.; Bowlin, G.L. Electrospinning of collagen type II: A
feasibility study. J. Bioact. Compat. Polym. 2003, 18,
125–134. 70. Telemeco, T.; Bowlin, G.L.; Vaida, M.; Cohen,
N.; Simpson, D.G. Collagen as a surgical biomaterial:
Fabrication of a skeletal muscle prosthetic. Nat.
Biotechnol. 2004, in review. 71. Sawa, M.; Kiyoi, T.;
Kurokawa, K.; Kumihara, H.; Yamamoto, M.; Miyasaka, T.;
Ito, Y.; Hirayama, R.; Inoue, T.; Kirii, Y.; Nishiwaki, E.;
Ohmoto, H.; Maeda, Y.; Ishibushi, E.; Inoue, Y.; Yoshino,
K.; Kondo, H. New type of metalloproteinase inhibitor:
Design and synthesis of new phosphonamide-based hydroxamic
acids. J. Med. Chem. 2002, 45 (4), 919–929.
Mechanical Circulatory Support

1. Rose, E.A.; for the REMATCH Study Group. Long term use
of a left ventricular assist device for endstage heart
failure. NEJM 2001, 345, 1435–1443.

2. Spanier, T.B.; Chen, J.M.; Oz, M.C.; Stern, D.M.; Rose,


E.A.; Schmidt, A.M. Time-dependent cellular population of
textured-surface left ventricular assist devices
contributes to the development of a biphasic systemic
procoagulant response. J. Thorac. Cardiovasc. Surg. 1999,
118, 404–413.

3. Long, J.W. Advanced mechanical circulatory support with


the HeartMate ventricular assist device in the year 2000.
Ann. Thorac. Surg. 2001, 71, S176–S182.

4. Portner, P.M.; Oyer, P.E.; McGregor, C.G.A. First human


use of an electrically powered implantable ventricular
assist system. Artif. Organs 1985, 9, 36.

5. Robbins, R.C.; Oyer, P.E. Bridge to transplant with the


Novacor left ventricular assist system. Ann. Thorac. Surg.
1999, 68, 695–697.

6. Kasirajan, V.; McCarthy, P.M.; Hoercher, K.J.; Starling,


R.C.; Young, J.B.; Banbury, M.K.; Smedira, N.G. Clinical
experience with long term use of implantable left
ventricular assist devices: Indications, implantation, and
outcomes. Semin. Thorac. Cardiovasc. Surg. 2000, 12,
229–237.

7. Wagner, W.R.; Johnson, P.C.; Kormos, R.L.; Griffith, P.


Evaluation of bioprosthetic valve-associated thrombus in
ventricular assist device patients. Circulation 1993, 88,
2023–2029. 8. Rose, E.A.; Levin, H.; Oz, M.C.; Frazier,
O.H.; Mcmanus, Q.; Burton, N.A.; Lefrak, E.A. Artificial
circulatory support with textured interior surfaces: A
counterintuitive approach to minimizing thromboembolism.
Circulation 1994, 90, II-87–II-91. 9. Scott-Burden, T.;
Frazier, O.H. Cellular linings of ventricular assist
devices. Ann. Thorac. Surg. 1995, 60, 1561–1562. 10.
Slater, J.P.; Rose, E.A.; Levin, H.R.; Frazier, O.H.;
Roberts, J.K.; Weinberg, A.D.; Oz, M.C. Low thromboembolic
risk without anticoagulation using advanced-design left
ventricular assist devices. Ann. Thorac. Surg. 1996, 62,
1321–1327. 11. Thomas, C.E.; Jichici, D.; Petrucci, R.;
Urrutia, V.C.; Schwartzman, R.J. Neurologic complications
of the Novacor left ventricular assist device. Ann. Thorac.
Surg. 2001, 72, 1311–1315. 12. Portner, P.M.; Jansen,
P.G.M.; Oyer, P.E.; Wheeldon, D.R.; Ramasamy, N. Improved
outcomes with an implantable left ventricular assist
system: A multicenter study. Ann. Thorac. Surg. 2001, 71,
205–209. 13. Gordon, S.M.; Schmitt, S.K.; Jacobs, M.;
Smedira, N.M.; Goormastic, M.; Banbury, M.K.; Yeager, M.;
Serkey, J.; Hoercher, K.; McCarthy, P.M. Nosocomial
infections in patients with left ventricular assist
devices. Ann. Thorac. Surg. 2001, 72, 725–730. 14.
El-banayosy, A.; Arusoglu, L.; Kizner, L.; Tenderich, G.;
Minami, K.; Inoue, K.; Korfer, K. Novacor left ventricular
assist system versus heartmate vented electric left
ventricular assist system as a long term mechanical
circulatory support device in bridging patients: A
prospective study. J. Thorac. Cardiovasc. Surg. 2000 , 119.
15. Ankersmit, H.A.; Tugulea, S.; Spanier, T.; Weinberg,
A.D. Activation-induced T cell death and immune dysfunction
after implantation of left ventricular assist device.
Lancet 1999, 354, 550–555. 16. Spanier, T.; Oz, M.C.;
Levin, H.; Weinberg, A.; Stamatis, K.; Stern, D.; Rose,
E.A.; Schmidt, M. Activation of coagulation and fibrinolytic
pathways in patients with left ventricular assist devices.
J. Thorac. Cardiovasc. Surg. 1996, 112, 1090–1097. 17.
McCarthy, P.M.; Smedira, N.O.; Vargo, R.L.; Goormastic, M.;
Hobbs, R.E.; Starling, R.C.; Young, J.B. One hundred
patients with the HeartMate left ventricular assist device:
Evolving concepts and technology. J. Thorac. Cardiovasc.
Surg. 1998, 115. 18. Poirier, V.L. Inflow valve
incompetence. J. Congest. Heart Fail. Circ. Support 2001, 1
(2), 23–25.
Mechano-Regulation of Fibroblast Function

33. Bishop, J.E.; Lindahl, G. Regulation of cardiovascular


collagen synthesis by mechanical load. Cardiovasc. Res.
1999, 42, 27–44.

34. Lindahl, G.E.; Chambers, R.C.; Papakrivopoulou, J.;


Dawson, S.J.; Jacobsen, M.C.; Bishop, J.E.; Laurent, G.J.
Activation of fibroblast procollagen alpha 1(I)
transcription by mechanical strain is transforming growth
factor-beta-dependent and involves increased binding of
CCAAT-binding factor (CBF/NF-Y) at the proximal promoter.
J. Biol. Chem. 2002, 277, 6153–6161.

35. Schild, C.; Trueb, B. Mechanical stress is required for


high-level expression of connective tissue growth factor.
Exp. Cell Res. 2002, 274, 83–91.

36. Bishop, J.E.; Butt, R.; Dawes, K.; Laurent, G.


Mechanical load enhances the stimulatory effect of PDGF on
pulmonary artery fibroblast procollagen synthesis. Chest
1998, 114, 25S.

37. Parsons, M.; Kessler, E.; Laurent, G.J.; Brown, R.A.;


Bishop, J.E. Mechanical load enhances procollagen
processing in dermal fibroblasts by regulating levels of
procollagen C-proteinase. Exp. Cell Res. 1999, 252,
319–331.

38. Chiquet-Ehrismann, R.; Tannheimer, M.; Koch, M.;


Brunner, A.; Spring, J.; Martin, D.; Baumgartner, S.;
Chiquet, M. Tenascin-C expression by fibroblasts is elevated
in stressed collagen gels. J. Cell Biol. 1994, 127,
2093–2101.

39. Banes, A.J.; Horesovsky, G.; Larson, C.; Tsuzaki, M.;


Judex, S.; Archambault, J.; Zernicke, R.; Herzog, W.;
Kelley, S.; Miller, L. Mechanical load stimulates
expression of novel genes in vivo and in vitro in avian
flexor tendon cells. Osteoarthr. Cartil. 1999, 7, 141–153.

40. Skutek, M.; van Griensven, M.; Zeichen, J.; Brauer, N.;
Bosch, U. Cyclic mechanical stretching modulates secretion
pattern of growth factors in human tendon fibroblasts. Eur.
J. Appl. Physiol. 2001, 86, 48–52.

41. Wang, J.H.; Yang, G.; Li, Z. Controlling cell responses


to cyclic mechanical stretching. Ann. Biomed. Eng. 2005,
33, 337–342.
42. Yang, G.; Im, H.J.; Wang, J.H. Repetitive mechanical
stretching modulates IL-1beta induced COX-2, MMP-1
expression, and PGE(2) production in human patellar tendon
fibroblasts. Gene 2005.

43. Hsieh, A.H.; Sah, R.L.; Paul Sung, K.L. Biomechanical


regulation of type I collagen gene expression in ACLs in
organ culture. J. Orthop. Res. 2002, 20, 325–331.

44. Zhou, D.; Lee, H.S.; Villarreal, F.; Teng, A.; Lu, E.;
Reynolds, S.; Qin, C.; Smith, J.; Sung, K.L. Differential
MMP-2 activity of ligament cells under mechanical stretch
injury: an in vitro study on human ACL and MCL fibroblasts.
J. Orthop. Res. 2005, 23, 949–957.

45. Jones, B.F.; Wall, M.E.; Carroll, R.L.; Washburn, S.;


Banes, A.J. Ligament cells stretch-adapted on a
microgrooved substrate increase intercellular communication
in response to a mechanical stimulus. J. Biomech. 2005, 38,
1653–1664.

46. Von den Hoff, J.W. Effects of mechanical tension on


matrix degradation by human periodontal ligament cells
cultured in collagen gels. J. Periodontal Res. 2003, 38,
449–457. 47. Butler, D.L.; Goldstein, S.A.; Guilak, F.
Functional tissue engineering: the role of biomechanics. J.
Biomech. Eng. 2000, 122, 570–575. 48. Akhyari, P.; Fedak,
P.W.; Weisel, R.D.; Lee, T.Y.; Verma, S.; Mickle, D.A.; Li,
R.K. Mechanical stretch regimen enhances the formation of
bioengineered autologous cardiac muscle grafts. Circulation
2002, 106, I137–I142. 49. Zimmermann, W.H.;
Schneiderbanger, K.; Schubert, P.; Didie, M.; Munzel, F.;
Heubach, J.F.; Kostin, S.; Neuhuber, W.L.; Eschenhagen, T.
Tissue engineering of a differentiated cardiac muscle
construct. Circ. Res. 2002, 90, 223–230. 50. Zimmermann,
W.H.; Melnychenko, I.; Eschenhagen, T. Engineered heart
tissue for regeneration of diseased hearts. Biomaterials
2004, 25, 1639–1647. 51. Garvin, J.; Qi, J.; Maloney, M.;
Banes, A.J. Novel system for engineering bioartificial
tendons and application of mechanical load. Tissue Eng.
2003, 9, 967–979. 52. Vunjak-Novakovic, G.; Altman, G.;
Horan, R.; Kaplan, D.L. Tissue engineering of ligaments.
Annu. Rev. Biomed. Eng. 2004, 6, 131–156. 53. Ingber, D.E.;
Dike, L.; Hansen, L.; Karp, S.; Liley, H.; Maniotis, A.;
McNamee, H.; Mooney, D.; Plopper, G.; Sims, J. Cellular
tensegrity: exploring how mechanical changes in the
cytoskeleton regulate cell growth, migration, and tissue
pattern during morphogenesis. Int. Rev. Cytol. 1994, 150,
173–224. 54. Ingber, D.E. Tensegrity II. How structural
networks influence cellular information processing networks.
J. Cell Sci. 2003, 116, 1397–408. 55. Schmidt, C.E.;
Horwitz, A.F.; Lauffenburger, D.A.; Sheetz, M.P.
Integrin-cytoskeletal interactions in migrating fibroblasts
are dynamic, asymmetric, and regulated. J. Cell Biol. 1993,
123, 977–991. 56. Wang, N.; Butler, J.P.; Ingber, D.E.
Mechanotransduction across the cell surface and through the
cytoskeleton [see comment]. Science 1993, 260, 1124–1127.
57. Urbich, C.; Dernbach, E.; Reissner, A.; Vasa, M.;
Zeiher, A.M.; Dimmeler, S. Shear stress-induced endothelial
cell migration involves integrin signaling via the
fibronectin receptor subunits alpha(5) and beta(1).
Arterioscler. Thromb. Vasc. Biol. 2002, 22, 69–75. 58.
Ingber, D. Integrins as mechanochemical transducers. Curr.
Opin. Cell Biol. 1991, 3, 841–848. 59. Janmey, P.A.; Weitz,
D.A. Dealing with mechanics: mechanisms of force
transduction in cells. Trends Biochem. Sci. 2004, 29,
364–370. 60. Silver, F.H.; Siperko, L.M. Mechanosensing and
mechanochemical transduction: how is mechanical energy
sensed and converted into chemical energy in an
extracellular matrix? Crit. Rev. Biomed. Eng. 2003, 31,
255– 331. 61. Munevar, S.; Wang, Y.L.; Dembo, M. Regulation
of mechanical interactions between fibroblasts and the
substratum by stretch-activated Ca2+entry. J. Cell Sci.
2004, 117, 85–92. 62. Iqbal, J.; Zaidi, M. Molecular
regulation of mechanotransduction. Biochem. Biophys. Res.
Commun. 2005, 328, 751–755. 63. Ullrich, A.; Schlessinger,
J. Signal transduction by receptors with tyrosine kinase
activity. Cell 1990, 61, 203–212. 64. Oberhauser, A.F.;
Badilla-Fernandez, C.; CarrionVazquez, M.; Fernandez, J.M.
The mechanical hierarchies of fibronectin observed with
single-molecule AFM. J. Mol. Biol. 2002, 319, 433–447. 65.
Liu, C.; Yao, J.; Mercola, D.; Adamson, E. The
transcription factor EGR-1 directly transactivates the
fibronectin gene and enhances attachment of human
glioblastoma cell line U251. J. Biol. Chem. 2000, 275,
20,315–20,323. 66. Schwachtgen, J.L.; Houston, P.;
Campbell, C.; Sukhatme, V.; Braddock, M. Fluid shear stress
activation of egr-1 transcription in cultured human
endothelial and epithelial cells is mediated via the
extracellular signal-related kinase 1/2 mitogen-activated
protein kinase pathway. J. Clin. Invest. 1998, 101,
2540–2549. 67. Li, C.; Xu, Q. Mechanical stress-initiated
signal transductions in vascular smooth muscle cells. Cell
Signal. 2000, 12, 435–445. 68. Wilson, E.; Mai, Q.; Sudhir,
K.; Weiss, R.H.; Ives, H.E. Mechanical strain induces
growth of vascular smooth muscle cells via autocrine action
of PDGF. J. Cell Biol. 1993, 123, 741–747. 69.
Millward-Sadler, S.J.; Wright, M.O.; Lee, H.; Nishida, K.;
Caldwell, H.; Nuki, G.; Salter, D.M. Integrinregulated
secretion of interleukin 4: A novel pathway of
mechanotransduction in human articular chondrocytes. J.
Cell Biol. 1999, 145, 183–189. 70. Tsuzaki, M.; Bynum, D.;
Almekinders, L.; Yang, X.; Faber, J.; Banes, A.J. ATP
modulates load-inducible IL-1beta, COX 2, andMMP-3 gene
expression in human tendon cells. J. Cell. Biochem. 2003,
89, 556–562. 71. MacKenna, D.; Summerour, S.R.; Villarreal,
F.J. Role of mechanical factors in modulating cardiac
fibroblast function and extracellular matrix synthesis.
Cardiovasc. Res. 2000, 46, 257–263. 72. Chiquet, M.;
Renedo, A.S.; Huber, F.; Fluck, M. How do fibroblasts
translate mechanical signals into changes in extracellular
matrix production? Matrix Biol. 2003, 22, 73–80. 73.
Eastwood, M.; McGrouther, D.A.; Brown, R.A. Fibroblast
responses to mechanical forces. Proc. Inst. Mech. Eng. [H]
1998, 212, 85–92. 74. Wall, M.E.; Banes, A.J. Early
responses to mechanical load in tendon: role for calcium
signaling, gap junctions and intercellular communication.
J. Musculoskel. Neuron. Interact. 2005, 5, 70–84. 75. Gudi,
S.R.; Lee, A.A.; Clark, C.B.; Frangos, J.A. Equibiaxial
strain and strain rate stimulate early activation of G
proteins in cardiac fibroblasts. Am. J. Physiol. 1998, 274,
C1424–C1428. 76. Clark, C.B.; McKnight, N.L.; Frangos, J.A.
Strain and strain rate activation of G proteins in human
endothelial cells. Biochem. Biophys. Res. Commun. 2002, 299
, 258–262. M
Medical Imaging, 3-D

1. R + oentgen, W.K. On a new kind of rays. Nature 1896,


53, 274–276.

2. Hornak, J.P. The Basics of MRI. http://wwwcis.


rit.edu/htbooks/mri/ (accessed June 2003). 3. Lauterbur,
P.C. Image formation by induced local interactions:
Examples employing nuclear magnetic resonance. Nature 1973,
242, 190–191. 4. Kumar, A.; Welti, D.; Ernst, R. NMR
Fourier zeugmatography. J. Magn. Reson. 1975, 18, 69. 5.
Liang, Z.P.; Lauterbur, P.C. Introduction. In Principles of
Magnetic Resonance Imaging; Akay, M., Ed.; IEEE Press: New
York, 2000; 1–12. 6. Cho, Z.H.; Jones, J.P.; Singh, M.
X-ray Computerized Tomography. In Foundations of Medical
Imaging; John Wiley & Sons: New York, 1993; 148–164. 7.
Kuhl, D.E.; Edwards, R.Q. Image separation radioisotope
scanning. Radiology 1963, 80, 653–661. 8. Zaidi, H.;
Hasegawa, B. Determination of the attenuation map in
emission tomography. J. Nucl. Med. 2003, 44 (2), 291–315.
9. Cho, Z.H.; Chan, J.K.; Eriksson, L. Circular ring
transverse axial position camera for 3-D reconstruction of
radionuclide distribution. IEEE Trans. Nucl. Sci. 1976, 23,
613–622. 10. Fenster, A.; Downey, D.B.; Cardinal, H.N.
Three-dimensional ultrasound imaging. Phys. Med. Biol.
2001, 46 (5), R67–R99. 11. Zou, Y.; Guo, Z. A review of
electrical impedance techniques for breast cancer
detection. Med. Eng. Phys. 2003, 25 (2), 79–90. 12. Hagen,
G.; Wadstrom, J.; Magnusson, A. 3D rotational angiography
of transplanted kidneys. Acta Radiol. 2003, 44 (2),
193–198. 13. Handbook of Medical Imaging. Processing and
Analysis; Bankman, I.N., Ed.; Academic Press: San Diego,
2000. 14. Medical Image Processing and Analysis. In
Handbook of Medical Imaging; Sonka, M., Fitzpatrick, J.M.,
Eds.; Spie Press: Bellingham, WA, 2000; Vol. 2. 15. Medical
Image Registration; Hajnal J.V., Hill, D.L.G., Hawkes,
D.J., Eds.; Biomedical Engineering Series, CRC Press: Boca
Raton, FL, 2001. 16. Robb, R.A Biomedical Imaging,
Visualization, and Analysis; Wiley-Liss, John Wiley & Sons:
New York, 2000. 17. Bezdek, J.C.; Hall, L.O.; Clarke, L.P.
Review of MR image segmentation techniques using pattern
recognition. Med. Phys. 1993, 20 (4), 1033–1048. 18.
Clarke, L.P.; Velthuizen, R.P.; Camacho, M.A.; Heine, J.J.;
Vaidyanathan, M.; Hall, L.O.; Thatcher, R.W.; Silbiger,
M.L. MRI segmentation: Methods and applications. Magn.
Reson. Imaging 1995, 13 (3), 343–368. 19. Pal, N.R.; Pal,
S.K. A review of image segmentation techniques. Pattern
Recogn. 1993, 26 (9), 1227–1249. 20. Gonzalez, R.C.; Woods,
R.E. Digital Image Processing, 2nd Ed.; Prentice Hall:
Upper Saddle River, NJ, 2002. 21. Sahoo, P.K.; Soltani, S.;
Wond, A.K.; Chen, Y.C. A survey of thresholding techniques.
Comput. Vis. Graph. Image Process. 1988, 41, 233–260. 22.
Bramdt, M.E.; Bohan, T.P.; Kramer, L.A.; Fletcher, J.M.
Estimation of CSF, white and gray matter volumes in
hydrosephalic children using fuzzy clustering of MR
imaging. Comput. Med. Imaging Graph. 1994, 18 (1), 25–34.
23. Duda, R.O.; Hart, P.E. Pattern Recognition and Scene
Analysis; Wiley: New York, 1973. 24. Jain, A.K.; Flynn,
P.J. Image Segmentation Using Clustering. In Advances in
Image Understanding; Bowyer, K., Ahuja, N., Eds.; IEEE
Computer Society Press: Los Alamitas, CA, 1996. 25.
Haralick, R.M.; Shapiro, L.G. Survey: Image segmentation
techniques. Comput. Vis. Graph. Image Process. 1985, 29,
100–132. 26. Fletcher, L.M.; Marsotti, J.B.; Hornak, J.P. A
multispectral analysis of brain tissues. Magn. Reson. Med.
1993, 29, 623–630. 27. Reddick, W.E.; Glass, J.O.; Cook,
E.N.; Elkin, T.D.; Deaton, R.J. Automated segmentation and
classification of multispectral magnetic resonance images of
brain using artificial neural networks. IEEE Trans. Med.
Imag. 1997, 16 (6), 911–918. 28. Lucas-Quesada, F.A.;
Sinha, U.; Sinha, S. Segmentation strategies for breast
tumors from dynamic MR images. J. Magn. Reson. Imaging
1996, 6, 753–763. 29. Rogowska, J.; Preston, K., Jr.;
Hunter, G.J.; Hamberg, L.M.; Kwong, K.K.; Salonen, O.;
Wolf, G.L. Applications of similarity mapping in dynamic
MRI. IEEE Trans. Med. Imag. 1995, 14 (3), 480–486. 30.
Maintz, J.B.A.; Viergever, M.A. A survey of medical image
registration. Med. Image Anal. 1998, 2, 1–36. 31. Maurer,
C.D.; Fitzpatrick, J.M. A Review of Medical Image
Registration. In Interactive Image-Guided Neurosurgery;
Maciunas, R.J., Ed.; American Association of Neurological
Surgeons: Park Ridge, IL, 1993; 17–44. 32. Stokking, R.
Integrated Visualization of functional and Anatomical Brain
Images. PhD Thesis; Utrecht University, 1998. 33. Duncan,
J.S.; Ayache, N. Medical image analysis: Progress over two
decades and the challenges ahead. IEEE Trans. Pattern Anal.
Mach. Intell. 2000, 22 (1), 85–106. 34. Kass, M.; Witkin,
A.; Terzopoulos, D. Snakes: Active contour models. Int. J.
Comput. Vis. 1988, 1 (4), 321–331. 35. Miller, J.V.; Breen,
D.E.; Lorensen, W.E.; O’Bara, R.M.; Wozny, M.J.
Geometrically deformed models: A method for extracting
closed geometric models from volume data. Comput. Graph.
1991, 25 (4), 217–226. 36. McInerney, T.; Terzopoulos, D. A
dynamic finite element surface model for segmentation and
tracking in multidimensional medical images with
application to cardiac 4D image analysis. Comput. Med.
Imaging Graph. 1995, 19 (1), 69–83. 37. Cohen, I.; Cohen,
L.D.; Ayache, N. Using deformable surfaces to segment 3D
images and infer differential structures. CVGIP, Image
Underst. 1992, 56 (2), 242–263. 38. Zeng, X.; Staib, L.H.;
Schultz, R.T.; Duncan, J.S. Segmentation and measurement of
the cortex from 3-D MR images using coupled-surfaces
propagation. IEEE Trans. Med. Imag. 1999, 18 (10), 927–937.
39. Leventon, M.E.; Grimson, W.E.L.; Faugeras, O.
Statistical shape influence in geodesic active contours.
IEEE Conf. Comput. Vis. Pattern Recognit. 2000, 1, 316–323.
40. Studholme, C.; Hill, D.L.G.; Hawkes, D.J. An overlap
invariant entropy measure for 3d medical image alignment.
Pattern Recogn. 1999, 32 (1), 71–86. 41. Richard, F.J.P.;
Cohen, L.D. A new image registration technique with free
boundary constraints: Application to mammography. Comput.
Vis. Image Underst. 2003, 89, 166–196. 42. Bansal, R.;
Staib, J.H.; Zhe, C.; Rangarajan, A.; Knisely, J.; Nath,
R.; Duncan, J.S. Entropy-based dualportal-to-3-DCT
registration incorporating pixel correlation. IEEE Trans.
Med. Imag. 2003, 22 (1), 29–49. 43. Haili, C.; Rangarajan,
A. A Feature Registration Framework Using Mixture Models.
In IEEE Workshop on Mathematical Methods in Biomedical
Image Analysis, Proceedings; IEEE Computer Society: Los
Alamitos, CA, 2000; 190–197. 44. Pelizzari, C.A.; Chen,
G.T.Y.; Spelbring, D.R.; Weichselbaum, R.R.; Chen, C.T.
Accurate three-dimensional registration of CT, PET, and/or
MR images of brain. J. Comput. Assist. Tomogr. 1989, 13,
20–26. 45. Cuchet, E.; Knoplioch, J.; Dormont, D.;
Marsault, C. Registration in neurosurgery and
neuroradiotherapy applications. J. Image Guid. Surg. 1995,
1, 198–207. 46. Rueckert, D.; Sonoda, L.I.; Haynes, C.;
Hill, D.L.G.; Leach, M.O.; Hawkes, D.J. Non-rigid
registration using free-form deformations: Application to
breast MR images. IEEE Trans. Med. Imag. 1999, 18 (8),
712–721. 47. Johnson, H.J.; Christensen, G.E. Consistent
landmark and intensity-based image registration. IEEE
Trans. Med. Imag. 2002, 21 (5), 450–461. 48. Holden, M.;
Hill, D.L.G.; Denton, E.R.E.; Jarosz, J.M.; Cox, T.C.S.;
Rohlfing, T.; Goodey, J.; Hawkes, D.J. Voxel similarity
measures for 3D serial MR brain image registration. IEEE
Trans. Med. Imag. 2002, 19 (2), 94–102. 49. Maes, F.;
Collignon, A.; Vandermeulen, D.; Marchal, G.; Suetens, P.
Multimodality image registration by maximization of mutual
information. IEEE Trans. Med. Imag. 1997, 16 (2), 187–198.
M
Medical Robotics

28. Jack, D.; Boian, R.; Merians, A.; Tremaine, M.; Burdea,
G.; Adamovich, S.; Recce, M.; Poizner, H. Virtual
reality-enhanced stroke rehabilitation. IEEE Trans. Neural
Syst. Rehabil. Eng. 2001, 9 (3), 308–318.

29. http://www.jpl.nasa.gov/releases/2000/stepper.html
(accessed June 2003).

30. Colombo, G.; Joerg, M.; Schreier, R.; Dietz, V.


Treadmill training of paraplegic patients using a robotic
orthosis. J. Rehabil. Res. Dev. 2000, 37 (6), 693–700.

31. Reinkensmeyer, D.; Wynne, J.; Harkema, S. A Robotic


Tool for Studying Locomotor Adaptation and Rehabilitation.
Proceedings in the 2002 IEEE Engineering in Medicine and
Biology 24th Annual Conference and the 2002 Fall Meeting of
the Biomedical Engineering Society (BMES/EMBS), Houston,
TX, October 2002.

32. Van der Loos, M.; Kautz, S.; Schwandt, D.; Anderson,
J.; Chen, G.; Bevly, D. A Split-Crank, ServomotorControlled
Bicycle Ergometer Design for Studies in Human Biomechanics.
2002 IEEE/RSJ International Conference on Intelligent
Robots and Systems, Lausanne, Switzerland, September 2002.

33. Satava, R. Cybersurgery: Advanced Technologies for


Surgical Practice; John Wiley & Sons, Inc.: New York, 1997.

34. http://www.computermotion.com/, (accessed August 2003).

35. Broderick, T.; Russell, K.; Doarn, C.; Merrell, R. A


novel method forvisualizing the open surgical field. J.
Laparoendosc. Adv. Surg. Tech. 2002, 12 (4), 297–302.

36. http://www.intuitivesurgical.com/, (accessed August


2003).

37. Guthart, G.; Salisbury, K. The Intuitivet Telesurgery


System: Overview and Application. In Proceedings of 2000
IEEE International Conference on Robotics and Automation,
2000, 618–621.

38. Madhani, A.; Niemeyer, G.; Salisbury, K. The Black


Falcon: A Teleoperated Surgical Instrument for Minimally
Invasive Surgery. IEEE/RSJ Int. Conf. on Intelligent Robots
and Systems (IROS), Victoria B.C., Canada, October 1998.

39. Rosen, J.; Hannaford, B.; MacFarlane, M.; Sinanan, M.


Force controlled and teleoperated endoscopic grasper for
minimally invasive surgery—Experimental performance
evaluation. IEEE Trans. Biomed. Eng. October 1999, 46 (10),
1212–1221.

40. Marescaux, J.; Leroy, J.; Gagner, M.; Rubino, F.;


Mutter, D.; Vix, M.; Butner, S.; Smith, M.K. Transatlantic
robot-assisted telesurgery. Nat. Mag. 2001, 413, 379–380.

41. Rosen, J.; Hannaford, B.; Richards, C.; Sinanan, M.


Markov modeling of minimally invasive surgery based on
tool/tissue interaction and force/torque signatures for
evaluating surgical skills. IEEE Trans. Biomed. Eng. May
2001, 48 (5), 579–591.

42. Rosen, J.; Brown, J.; Chang, L.; Barreca, M.; Sinanan,
M.; Hannaford, B. The Blue DRAGON—A System for Measuring
the Kinematics and the Dynamics of Minimally Invasive
Surgical Tools in-vivo. Proceedings of the 2002 IEEE
International Conference on Robotics & Automation,
Washington, DC, USA, May 11–15, 2002. 43. Rosen, J.;
Solazzo, M.; Hannaford, B.; Sinanan, M. Objective
evaluation of laparoscopic skills based on haptic
information and tool/tissue interactions. Comput. Aided
Surg. July 2002, 7 (1), 49–61. 44. Rosen, J.; Chang, L.;
Brown, J.D.; Hannaford, B.; Sinanan, M.; Satava, R.
Minimally Invasive Surgery Task Decomposition—Etymology of
Endoscopic Suturing, Studies in Health Technology and
Informatics— Medicine Meets Virtual Reality; IOS Press:
January 2003; Vol. 94, 295–301. 45. Hannaford, B. Feeling
is Believing: Haptics and Telerobotics Technology. The
Robot in the Garden, Telerobotics and Telepistomology on
the Internet, Cambridge, MA, 1999; Goldberg, J.K., Ed.; MIT
Press: Cambridge, MA, 1999. 46. Green, P.; Hill, J.;
Jensen, J.; Shah, A. Telepresence surgery. IEEE Eng. Med.
Biol. 1995, 14, 324–329. 47. Taylor, R.; Lavallee, S.;
Burdea, G.; Mosges, R. Computer-Integrated Surgery; MIT
Press: Cambridge, MA, 1996. 48. Taylor, R. Medical
Robotics. In Handbook of Industrial Robotics, 2nd Ed.; Nof,
S.Y., Ed.; Wiley: New York, 1999; 1213–1230. 49. Howe, R.;
Matsuoka, Y. Robotics for surgery. Annu. Rev. Biomed. Eng.
1999, 1, 211–240. 50. Buess, G.; Schurr, M.; Fischer;
Sabine, C. Robotics and allied technologies in endoscopic
surgery. Arch. Surg. 2000, 135, 229–235. 51. Cleary, K.;
Nguyen, C. State of the art in surgical robotics: Clinical
applications and technology challenges. Comput. Aided Surg.
2001, 6, 312–328. 52. Ballantyne, G.H. The pitfalls of
laparoscopic surgery: Challenges for robotics and
telerobotic surgery, special issue on surgical robotics.
Surgical Laparoscopy, Endoscopy and Percutaneous Techniques
2002, 12 (1). 53. Li, Q.; Zamorano, L.; Pandya, A.; Perez,
R.; Gong, J.; Diaz, F. The application accuracy of the
NeuroMate robot—A quantitative comparison with frameless
and frame-based surgical localization systems. Comput.
Aided Surg. 2002, 7, 90–98. 54. http://www.immersion.com/,
(accessed August 2003). 55. http://www.sensable.com/,
(accessed August 2003). 56. Chen, E.; Marcus, B. Force
feedback for surgical simulation. Proc. I.E.E.E. March
1998, 86 (3), 524–530. 57. http://www.simcen.org/surgery/
(accessed January 2004). 58.
http://www-biocomp.stanford.edu/ (accessed January 2004).
59. Brown, J. In-vivo and Postmortem Biomechanics of
Abdominal Organs Under Compressive Loads: Experimental
Approach in a Laparoscopic Surgery Setup. Ph.D.
Dissertation; University of Washington: Seattle, WA, 2003.
60. Satava, R. Disruptive visions: The operating room of
the future. Surg. Endosc. 2003, 17 (1), 104–107. 61.
http://anesthesia.stanford.edu/VASimulator/sim.htm
(accessed June 2003). 62.
http://www.laas.fr/iarp-france/status-reports/2001/
medical.html (accessed July 2003). 63.
http://www.anthrotronix.com (accessed June 2003). 64.
Gilden, D.; Jaffe, D. Dexter—A robotic hand communication
aid for the deaf-blind. Int. J. Rehabil. Res. 1988, II (2),
198–199. 65. http://guide.stanford.edu/TTran/jrrd.html
(accessed June 2003). M
Melt Spinning

1. Nakjima, T. Advanced Fiber Spinning Technology;


Woodhead: Cambridge, UK, 1994.

2. Ziabicki, A. Fundamentals of Fiber Formation: The


Science of Fiber Spinning and Drawing; John Wiley & Sons:
London, UK, 1976. 3. Billmeyer, F.W., Jr. Textbook of
Polymer Science, 3rd Ed.; John Wiley & Sons: New York,
1984. 4. Ward, I.M. Structure and Properties of Oriented
Polymers, 2nd Ed.; Chapman & Hall: London, UK, 1997. 5.
Braun, S.; Stausberg, G. Monofilament Extrusion. In Fiber
Extrusion in Plastic Extrusion Technology, 2nd Ed.; Mayer,
M., Ed.; Hanser: Munich, Germany, 1997. 6. Chodak, I.;
Blackburn, R.S. Poly(hydroxyalkanoates) and
polycaprolactone. In Biodegradable and Sustainable Fibers;
Blackburn, R.S., Ed.; Woodhead: Cambridge, UK, 2005. 7.
Vita Vinzi Zalman, E.; Locardi, B.; Gabbi, C.; Tranquilli
Leale, P. Bioactive vitreous composition for bone implants,
filaments made therefrom and method. WO 91/12032. August 22,
1991. 8. Ella¨, V.E.; Gomes, M.E.; Reis, R.L.; To¨rma¨la¨,
P.; Kelloma¨ki, M. Studies of P(L/D)LA 96/4 non-woven
scaffolds and fibres; properties, wettability and cell
spreading before and after intrusive treatment methods.
Journal of Materials Science: Materials in Medicine 2007,
18, 1253–1261. 9. Chu, C.C. Textile-based biomaterials for
surgical applications. In Polymeric Biomaterials Second
Edition Revised & Expanded; Dumitriu, S., Ed.; Marcel
Dekker: NewYork, 2002. 10. Pazzaglia, UP.; Gabbi, C.;
Locardi, B.; Di Nucci, A.; Zatti, G.; Cherubino, P. Study
of the osteoconductive properties of bioactive glass fibers.
J Biomed Mater Res 1989, 23, 1289–1297. 11. Brink, M.
Bioactive Glasses with a Large Working Range; A˚bo Akademi
University: Turku, Finland, 1997; Doctoral Thesis. 12.
Cable, M. Classical glass technology. In Glasses and
Amorphous Materials; Zarzycki, J., Ed.; John Wiley & Sons:
Weinheim, 1991; 9. 13. Pirhonen, E. Fibers and Composites
for Potential Biomaterials Applications; Tampere University
of Technology, Finland, 2006; Doctoral Thesis. 14. Gupta,
B.S. Medical textile structures: An overview. Medical
Plastics and Biomaterials Jan. 1998, 16–31. 15. Sittinger,
M.; Reitzel, D.; Dauner, M.; Hierlemann, H.; Hammer, C.;
Kastenbauer, E.; Plack, H.; Burmester, G.R.; Bujia, J., J.
Biomed. Mater. Res. Part B: Appl. Biomater. 1996, 33,
57–63. 16. Leong, K.H.; Ramakrishna, S.; Huang, Z.M.; Bibo,
G.A. The potential of knitting for engineering composites—A
review. Composites A 2000, 31, 197–220. 17. Honkanen, P.B.;
Kelloma¨ ki, M.; Lehtima¨ki, M.Y.; To¨rma¨la¨, P.;
Ma¨kela¨, S.; Lehto, M.U.K. Bioreconstructive joint
scaffold implant arthroplasty in metacarpophalangeal
joints: Short-term results of a new treatment concept in
rheumatoid arthritis patients. Tissue Engineering 2003, 9
(5), 957–966. 18. Moimas, L.; Biosotto, M.; Di Lenarda, R.;
Olivo, A.; Schmid, C. Rabbit pilot study on the
resorbability of three-dimensional bioactive glass fibre
scaffolds. Acta Biomater. 2006, 2, 191–199.
Metallic Orthopedic Devices: Failure
Analysis

variable striation spacing; and (C) higher magnification SEM


micrograph showing striations, fluting, and terrace.
Microcirculatory Oxygen Transport

1. Krogh, A. The number and distribution of capillaries in


muscles with calculations of the oxygen pressure head
necessary for supplying the tissue. J. Physiol. 1919, 52,
409–415.

2. Duling, B.R.; Berne, R.M. Longitudinal gradients in


periarteriolar oxygen tension. A possible mechanism for the
participation of oxygen in local regulation of blood flow.
Circ. Res. 1970, 27 (5), 669–678.

3. Boron, W.F.; Boulpaep, E.L. Medical Physiology;


Saunders: Philadelphia, 2003.

4. Ellsworth, M.; Ellis, C.; Popel, A.; Pittman, R.N. Role


of microvessels in oxygen supply to tissue. News Physiol.
Sci. 1994, 9, 119–123.

5. Popel, A.S. Theory of oxygen transport to tissue. Crit.


Rev. Biomed. Eng. 1989, 17 (3), 257–321.

6. Secomb, T.W.; Hsu, R.; Dewhirst, M.W. Synergistic


effects of hyperoxic gas breathing and reduced oxygen
consumption on tumor oxygenation: a theoretical model. Int.
J. Radiat. Oncol. Biol. Phys. 2004, 59 (2), 572–578.

7. Pittman, R.N. Oxygen transport and exchange in the


microcirculation. Microcirculation 2005, 12 (1), 59–70.

8. Pittman, R.N. Influence of microvascular architecture on


oxygen exchange in skeletal muscle. Microcirculation 1995,
2 (1), 1–18.

9. Rovainen, C.M.; Wang, D.B.; Woolsey, T.A. Strobe


epiillumination of fluorescent beads indicates similar
velocities and wall shear rates in brain arterioles of
newborn and adult mice. Microvasc. Res. 1992, 43 (2),
235–239.

10. Parthasarathi, A.A.; Japee, S.A.; Pittman, R.N.


Determination of red blood cell velocity by video
shuttering and image analysis. Ann. Biomed. Eng. 1999, 27
(3), 313–325.

11. Wayland, H.; Johnson, P.C. Erythrocyte velocity


measurement in microvessels by a two-slit photometric
method. J. Appl. Physiol. 1967, 22 (2), 333–337.

12. Ellsworth, M.L.; Pittman, R.N. Evaluation of


photometric methods for quantifying convective mass
transport in microvessels. Am. J. Physiol. 1986, 251 (4 Pt
2), H869–H879.

13. Ellis, C.G.; Ellsworth, M.L.; Pittman, R.N.


Determination of red blood cell oxygenation in vivo by dual
video densitometric image analysis. Am. J. Physiol. 1990,
258 (4 Pt 2), H1216–H1223.

14. Pittman, R.N.; Duling, B.R. Measurement of percent


oxyhemoglobin in the microvasculature. J. Appl. Physiol.
1975, 38 (2), 321–327.

15. Pittman, R.N.; Duling, B.R. A new method for the


measurement of percent oxyhemoglobin. J. Appl. Physiol.
1975, 38 (2), 315–320.

16. Baker, C.; Nastuk, W. Microcirculatory Technology ;


Academic Press: Orlando, 1986. 17. Swain, D.P.; Pittman,
R.N. Oxygen exchange in the microcirculation of hamster
retractor muscle. Am. J. Physiol. 1989, 256 (1 Pt 2),
H247–H255. 18. Popel, A.S.; Pittman, R.N.; Ellsworth, M.L.
Rate of oxygen loss from arterioles is an order of
magnitude higher than expected. Am. J. Physiol. 1989, 256
(3 Pt 2), H921–H924. 19. Tsai, A.G.; Friesenecker, B.;
Mazzoni, M.C.; Kerger, H.; Buerk, D.G.; Johnson, P.C.;
Intaglietta, M. Microvascular and tissue oxygen gradients
in the rat mesentery. Proc. Natl. Acad. Sci. U.S.A. 1998,
95 (12), 6590–6595. 20. Tsai, A.G.; Johnson, P.C.;
Intaglietta, M. Oxygen gradients in the microcirculation.
Physiol. Rev. 2003, 83 (3), 933–963. 21. Vadapalli, A.;
Pittman, R.N.; Popel, A.S. Estimating oxygen transport
resistance of the microvascular wall. Am. J. Physiol. Heart
Circ. Physiol. 2000, 279 (2), H657–H671. 22. Ellsworth,
M.L.; Popel, A.S.; Pittman, R.N. Assessment and impact of
heterogeneities of convective oxygen transport parameters
in capillaries of striated muscle: experimental and
theoretical. Microvasc. Res. 1988, 35 (3), 341–362. 23.
Ellsworth, M.L.; Pittman, R.N. Arterioles supply oxygen to
capillaries by diffusion as well as by convection. Am. J.
Physiol. 1990, 258 (4 Pt 2), H1240–H1243. 24. Secomb, T.W.;
Hsu, R. Simulation of O 2 transport in skeletal muscle:
diffusive exchange between arterioles and capillaries. Am.
J. Physiol. 1994, 267 (3 Pt 2), H1214–H1221. 25. Golub,
A.S.; Pittman, R.N. Erythrocyte-associated transients in PO
2 revealed in capillaries of rat mesentery. Am. J. Physiol.
Heart Circ. Physiol. 2005, 288 (6), H2735–H2743. 26.
Federspiel, W.J.; Popel, A.S. A theoretical analysis of the
effect of the particulate nature of blood on oxygen release
in capillaries. Microvasc. Res. 1986, 32 (2), 164–189. 27.
Federspiel, W.J.; Sarelius, I.H. An examination of the
contribution of red cell spacing to the uniformity of
oxygen flux at the capillary wall. Microvasc. Res. 1984, 27
(3), 273–285. 28. Stein, J.C.; Ellis, C.G.; Ellsworth, M.L.
Relationship between capillary and systemic venous PO 2
during nonhypoxic and hypoxic ventilation. Am. J. Physiol.
1993, 265 (2 Pt 2), H537–H542. 29. Riess, J.G. Oxygen
carriers (‘‘blood substitutes’’)—raison duetre, chemistry,
and some physiology. Chem. Rev. 2001, 101 (9), 2797–2920.
30. Pittman, R.N. Oxygen supply to contracting skeletal
muscle at the microcirculatory level: diffusion vs.
convection. Acta Physiol. Scand. 2000, 168 (4), 593–602. M
Microcomputed Tomography and Its
Applications

1. Hounsfield, G.N. A method of and apparatus for


examination of a body by radiation such as X-ray or gamma
radiation. Pat. Specif. 1972, 1283915.

2. Cormack, A.M. Representation of a function by its line


integrals, with some radiological applications. J. Appl.
Phys. 1963, 34, 2722–2727.

3. Oldenhorf, W.H. Some possible applications of


computerized tomography in pathology. J. Comput. Assist.
Tomogr. 1980, 4 (2), 141–144.

4. Kujoory, M.A.; Hillman, B.J.; Barrett, H.H.


Highresolution computed tomography of the normal rat
nephrogram. Invest. Radiol. 1980, 15, 148–154.

5. Seguin, F.H.; Burstein, P.; Bjorkholm, P.J.; Homburger,


F.; Adams, R.A. X-ray computed tomography with 50-mm
resolution. Appl. Opt. 1985, 24, 4117–4123.

6. Feldkamp, L.A.; Goldstein, S.A.; Parfitt, M.A.; Jesion,


G.; Kleerekoper, M. The direct examination of
threedimensional bone architecture in vitro by computed
tomography. J. Bone Miner. Res. 1989, 4 (1), 3–11.

7. Feldkamp, L.A.; Davis, L.C. Practical cone-beam


algorithm. J. Opt. Soc. Am., A 1984, 1, 612–619.

8. Odgaard, A.; Andersen, K.; Ullerup, R.; Frich, L.H.;


Melsen, F. Three-dimensional reconstruction of entire
vertebral bodies. Bone 1994, 15 (3), 335–342.

9. Kinney, J.H.; Lane, N.E.; Haupt, D.L. In vivo,


threedimensional microscopy of trabecular bone. J. Bone
Miner. Res. 1995, 10 (2), 264–270.

10. Bonse, U.; Busch, F.; Gunnewig, O.; Beckman, F.; Pahl,
R.; Delling, G.; Graeff, HahnW. 3D computed X-ray
tomography of human cancellous bone at 8 mm spatial and 10
�4 energy resolution. Bone Miner. 1994, 25, 25–38.

11. Peyrin, F.; Salome, M.; Cloetens, P.; Laval-Jeantet,


A.M.; Ritman, E.; Ruegsegger, P. Micro-CT examinations of
trabecular bone samples at different resolutions:
14,7,2micron level. Technol. Health Care 1998, 6, 391–401.

12. Ritman, E.L.; Bolander, M.E.; Fitzpatrick, L.A.;


Turner, R.T. Micro-CT imaging of structure-to-function
relationship of bone microstructure and associated vascular
involvement. Technol. Health Care 1998, 6, 403–412.

13. Nuzzo, R.; Lafage-Proust, M.H.; Martin-Badosa, E.;


Boivin, G.; Thomas, T.; Alexandre, C.; Peyrin, F.
Synchrotron radiation microtomography allows the analysis
of three-dimensional microarchitecture and degree of
mineralization of human iliac crest biopsy specimens:
Effects of etidronate treatment. J. Bone Miner. Res. 2002,
17, 1372–1382. 14. Ruegsegger, P.; Koller, B.; Muller, R. A
microtomographic system for the nondestructive evaluation
of bone architecture. Calcif. Tissue Int. 1996, 58, 24–29.
15. Hildebrand, T.; Ru¨egsegger, P. A new method for the
model-independent assessment of thickness in
threedimensional images. J. Microsc. 1996, 185, 67–75. 16.
Hildebrand, T.; Ru¨egsegger, P. Quantification of bone
microarchitecture with the structure model index. Comput.
Methods Biomech. Biomed. Eng. 1997, 1, 15–23. 17. Salmon,
P.L.; Buelens, E.; Sasov, A. Radiation Dosimetry,
Anaesthesia and Welfare of Rodents in MicroCT In Vivo
Scanning. Skyscan, Aartselaar, Belgium, Proceedings of the
IBMS-JSBMR 2003, Osaka, Japan, June 3–7, 2003. Poster 130W.
18. David, V.; Laroche, N.; Boudignon, B.; Lafage-Proust,
M.H.; Alexandre, C.; Rueegsegger, P.; Vico, L. Noninvasive
in vivo monitoring of bone architecture alterations in
hindlimb-unloaded female rats using novel three-dimensional
microcomputed tomography. J. Bone Miner. Res. 2003, 18 (9),
1622–1631. 19. Curry, T.S.; Dowdey, J.E.; Murry, R.C.
Christensen’s Physics of Diagnostic Radiology, 4th Ed.; Lea
& Febiger: Philadelphia, PA, 1990. 20. Kak, A.C. Principles
of Computerized Tomographic Imaging; IEEE Press: New York,
NY, 1988. 21. Ritman, E.L.; Borah, B.; Dufresne, T.E.;
Phipps, R.J.; Sacha, J.P.; Josgansen S.M.R.T. 3-D
Synchrotron Micro-CT allows unique insight of changes in
bone quality. JBMR 2002, 17 (Suppl 1), S415. 22. Kak, A.C.
Aliasing Artifacts and Noise in CT Images. In Principles of
Computerized Tomographic Imaging; IEEE Press: New York, NY,
1988; 183–186. 23. Smith, B.D. Image reconstruction from
cone-beam projections: Necessary and sufficient conditions
and reconstruction methods. IEEE Trans. Med. Imag. 1985,
MI-4, 14–25. 24. Kak, A.C. Algorithms for Reconstructing
with Nondiffracting Sources. In Principles of Computerized
Tomographic Imaging; IEEE Press: New York, NY, 1988; 56–62.
25. Chmielewski, P.A; Dufresne, T.E.; Borah, B.; Combs,
K.H.; Lundy, M.W. Validation of a Micro-CT System for the
3-D Measurement of Bone. JBMR 2001, 16 (Suppl 1), p. S461.
26. Muller, R.; Van Campenhout, H.; Van Damme, B.; Van Der
Perre, G.; Dequeker, J.; Hildebrand, T.; Ruegsegger, P.
Morphometric analysis of human bone biopsies: A
quantitative structural comparison of histological sections
and micro-computed tomography. Bone 1998, 23 (1), 59–66.
27. Parfitt, A.M. The Stereologic Basis of Bone
Histomorphometry. Theory of Quantitative Microscopy and
Reconstruction of the Third Dimension. In Bone
Histomorpohometry. Techniques and Interpretations; CRC
Press: Boca Raton, FL, 1983; 53–87. 28. Ulrich, D.;
Hildebrand, T.; Van Rietbergen, B. The Quality of
Trabecular Bone Evaluated with Micro-CT, FEA and Mechanical
Testing. In Bone Research in Biomechanics; IOS Press:
Amsterdam; 1997; 97–112. 29. Consensus Development
Conference. Prophylaxis and treatment of osteoporosis. Am.
J. Med. 1991, 90 (1), 107–110. 30. Mosekilde, L. Vertebral
structure and strength in vivo and in vitro. Calcif. Tissue
Int. 1993, 53 (suppl), S121– S126. 31. Borah, B.; Dufresne,
T.E.; Chmielewski, P.A.; Gross, G.J.; Prenger, M.C.;
Phipps, R.J. Risedronate preserves trabecular architecture
and increases bone strength in vertebra of ovariectomized
minipigs as measured by 3-D micro-computed tomography. J.
Bone Miner. Res. 2002, 17, 1139–1147. 32. Ulrich, D.; van
Rietbergen, B.; Laib, A.; Ru¨egsegger, P. The ability of 3D
structural indices to reflect mechanical aspects of
trabecular bone. Bone 1999, 25 (1), 55–60. 33. Dufresne,
T.E.; Chmielewski, P.A.; Manhart, M.A.; Johnson, T.D.;
Borah, B. Risedronate Preserves Bone Architecture in Early
Postmenopausal Women in 1 Year as Measured by
Three-Dimensional Microcomputed Tomography; Calcified Tissue
International, SpringerVerlag: New York. Calc. Tissue Int.
2003, 73, 423–432. 34. Borah, B.; Dufresne, T.E.;
Chmielewski, P.A.; Prenger, M.C.; Eriksen, E.F. Risedronate
(Ris) preserves bone architecture in osteoporotic
postmenopausal women as measured by 3-D microcomputed
tomography. J. Bone Miner. Res. 2001, 16 (suppl 1), S218.
35. Borah, B.; Gross, G.J.; Dufresne, T.E.; Smith, T.S.;
Cockman, M.D.; Chmielewski, P.A.; Lundy, M.W.; Hartke,
J.R.; Sod, E.W. Three-dimensional microimaging (MrmI and
mCT), finite element modeling and rapid prototyping provide
unique insights into bone architecture in osteoporosis.
Anat. Rec. 2001, 265, 101–110. 36. Dempster, D.W.; Cosman,
F.; Kurland, E.S.; Zhou, H.; Nieves, J.; Woelfert, L.;
Shane, E.; Plavetic, K.; Muller, R.; Bilezikian, J.;
Lindsay, R. Effects of daily treatment with parathyroid
hormone on bone architecture and turnover in patients with
osteoporosis: A paired biopsy study. J. Bone Miner. Res.
2001, 16, 1846–1853. 37. Borah, B.; Dufresne, T.E.;
Chmielewski, P.A.; Pierce, S.A.; Hartke, J.R.; Lundy, M.W.
Fluprostenol, a Prostaglandin fp Agonist, Improves
3-Dimensional Trabecular Architecture in Rat Vertebrae as
Measured by Micro-computed Tomography. Proceedings of 21st
ASBMR, St. Louis, Missouri, 1999; S401. J Bone Miner Res.
38. Dufresne, T.E. Segmentation techniques for analysis of
bone by three-dimensional computed-tomographic imaging.
Technol. Health Care 1998, 6, 351–359. 39. Herzog, S.;
Sager, H.; Khmelevski, E.; Deylig, A.; Ito, W.D. Collateral
arteries grow from preexisting anastomoses in the rat
hindlimb. Am. J. Physiol, Heart Circ. Physiol. 2002, 283
(5), H2012–2020. 40. Dufresne, T.E.; Borah, B.;
Chmielewski, P.A.; Combs, K.H.; Loudy, D.E.; Lundy, M.W.;
Samuelsson, S.J. 3-D Micro-Computed Tomography in
Pharmaceutical Research. J. Scan. Microsc. 1999, 21 (2),
129–130. M
Microelectromechanical Systems (MEMS)
Manufacturing

Fig. 10 Fabrication process for the aggregated assembly

of DNA-conjugated gold nanoparticles. (From Ref. [64].) 4.


Slocum, A.H. Precision Machine Design: Macromachine Design
Philosophy and Its Applicability to the Design of
Micromachines. In Proceedings: IEEE Micro Electro
Mechanical Systems (MEMS ’92); Travemunde, Germany, 1992;
37–42. 5. Compton Compton’s Interactive Encyclopedia
(Interactive Multimedia): Computer Data and Program;
Compton’s New Media: Carlsbad, 1994. 6. Harris, T.W.
Chemical Milling; Clarendon Press: Oxford, 1976. 7. Becker,
E.W.; Ehrfeld, W.; Munchmeyer, D.; Betz, H.; Heuberger, A.;
Pongratz, S.; Glashauser, W.; Michel, H.J.; Siemens, V.R.
Production of separation nozzle systems for uranium
enrichment by a combination of x-ray lithography and
galvanoplastics. Naturwissenschaften 1982, 69, 520–523. 8.
Xia, Y.; Whitesides, G.M. Soft lithography. Angew. Chem.,
Int. Ed. Engl. 1998, 37, 550–575. 9. Szepesi, D. Sensoren
en Actuatoren in Ultraprecisie Draaibanken. In Sensoren en
Actuatoren in de Werktuigbouw/Machinebouw, Centrum voor
Micro-Electronica; The Hague, Netherlands, 1993; 99–107.
10. Smith, H.I.; Schattenburg, M.L. Why bother with x-ray
lithography? SPIE 1992, 1671, 282–298. 11. Stroscio, J.A.;
Eigler, D.M. Atomic and molecular manipulation with the
scanning tunneling microscope. Science 1991, 254,
1319–1326. 12. Vogel, S. Cats’ Paws and Catapults; W.W.
Norton: New York, 1998. 13. Ball, P. Made to Measure;
Princeton University Press: Princeton, NJ, 1997. 14.
Whitesides, G.M.; Mathias, J.P.; Seto, C.T. Molecular
self-asembly and nanochemistry: A chemical strategy for the
synthesis of nanostructures. Science 1991, 254, 1312–1318.
15. Achilles, T.; Von Kiedrowski, G. A self-replicating
system from three starting materials. Angew. Chem., Int.
Ed. Engl. 1993, 32, 1198–1201. 16. Bo¨hringer, K.F.;
Fearing, R.S.; Goldberg, K.Y. Microassembly. In Handbook of
Industrial Robotics; Nof, S., Ed.; Wiley: New York, 1998.
17. Nelson, B.; Vikramaditya, B. Visually Guided
Microassembly Using Optical Microscopes and Active Vision
Techniques. In Proceedings: IEEE Int. Conf. on Robotics and
Automation (ICRA); 1997. Albuquerque, N.M. 18. Feddema,
J.T.; Simon, R.W. CAD-Driven Microassembly and Visual
Servoing. In Proceedings: IEEE Int. Conf. on Robotics and
Automation (ICRA); 1998. Leuven, Belgium. 19. Cohn, M.B.;
Howe, R.T. Wafer-to-Wafer Transfer of Microstructures Using
Breakaway Tethers. In U.S. Patent Application; 1997. 20.
Singh, A.; Horsley, D.A.; Cohn, M.B.; Pisano, A.P.; Howe,
R.T. Batch Transfer of Microstructures Using Flip-Chip Bump
Bonding. In Digest: Int. Conf. on Solid State Sensors and
Actuators; Transducers Research Foundation: Chicago, 1997.
21. Yeh, H.-J.; Smith, J.S. Fluidic self-assembly for the
integration of GaAs light-emitting diodes on Si substrates.
IEEE Photonics Technol. Lett. 1994, 6, 706–708. 22. Yeh,
H.-J.; Smith, J.S. Fluidic Self-Assembly of Microstructures
and Its Application to the Integration of GaAs on Si. In
Proc. IEEE International Workshop on Micro Electro
Mechanical Systems (MEMS ’94); 1994; 279–284. Oiso, Japan.
23. Yeh, H.-J.; Smith, J.S. Integration of GaAs
verticalcavity surface-emitting laser on Si by substrate
removal. Appl. Phys. Lett. 1994, 64, 1466–1468. 24. Quaid,
A.E.; Hollis, R.L. Cooperative 2-Dof Robots for Precision
Assembly. In Proceedings: IEEE Int. Conf. on Robotics and
Automation (ICRA); 1996. Minneapolis, Minn. 25. Zesch, W.
Multi-Degree-of-Freedom Micropositioning Using Stepping
Principles. In Ph.D. Thesis; Swiss Federal Institute of
Technology: Zurich, Switzerland, 1997. 26. Keller, C.G.;
Howe, R.T. Hexsil Tweezers for Teleoperated Micro-Assembly.
In Proceedings: IEEE Workshop on Micro Electro Mechanical
Systems (MEMS); 1997; Nagoya, Japan. 27. Cohn, M.B.
Self-Assembly of Microfabricated Devices. U.S. Patent
5,355,577, 1992. 28. Cohn, M.B.; Howe, R.T.; Pisano, A.P.
Self-Assembly of Microsystems Using Non-Contact
Electrostatic Traps. In Proceedings of the ASME
International Congress and Exposition, Symposium on
Micromechanical Systems (IC ’95); 1995; 893–900. San
Francisco. 29. Bo¨hringer, K.-F.; Cohn, M.B.; Goldberg, K.;
Howe, R.; Pisano, A. Electrostatic Self-Assembly Aided by
Ultrasonic Vibration. AVS 44th National Symposium; 1997.
San Jose, Calif. 30. Bo¨hringer, K.-F.; Goldberg, K.; Cohn,
M.B.; Howe, R.; Pisano, A. Parallel Microassembly with
Electrostatic Force Fields. In Proceedings: IEEE Int. Conf.
on Robotics and Automation (ICRA); 1998; Leuven, Belgium.
31. Edman, C.F.; Gurtner, C.; Formosa, R.E.; Coleman, J.J.;
Heller, M.J. Electric-Field-Directed Pick-and-Place
Assembly. In HDI; October, 2000; 30–35. 32. Kim, C.-J.;
Pisano, A.P.; Muller, R.S. Silicon-Processed Overhanging
Microgripper. J. Microelectromech. Syst. 1992, 1, 31–36.
33. Pister, K.S.J.; Judy, M.W.; Burgett, S.R.; Fearing,
R.S. Microfabricated hinges. Sensors Actuators, A 1992, 33,
249–256. 34. Keller, C.; Howe, R.T. Nickel-Filled Hexsil
Thermally Actuated Tweezers. In Digest: Int. Conf. on
Solid-State Sensors and Actuators; Transducers Research
Foundation: Stockholm, Sweden, 1995. 35. Hosokawa, K.;
Shimoyama, I.; Miura, H. Dynamics of self-assembling
systems: Analogy with chemical kinetics. Artif. Life 1995,
1. 36. Ashkin, A.; Dziedzic, J.M.; Yamane, T. Optical
trapping and manipulation of single cells using infrared
laser beams. Nature 1987, 330, 769–771. 37. Brown, K.T.;
Flaming, D.G. Advanced Micropipette Techniques for Cell
Physiology. In IBRO Handbook: Methods in Neurosciences;
Chichester, Ed.; Wiley: New York, 1992. 38. Miltenyi, S.;
Mueller, W.; Weichel, W.; Radbruch, A. Cytometry 1990, 11,
231–238. M

39. Schnelle, T.; Hagedorn, R.; Fuhr, G.; Fiedler, S.;


Mu¨ller, T. . Biochim. Biophys. Acta 1993, 1157, 127–140.

40. Coackley, W.T. . TIBTECH 1997, 5, 506–511.

41. Giddings, J.C. . Science 1993, 260, 1456–1464.

42. Fu, A.; Spence, C.; Scherer, A.; Arnold, F.H.; Quake,
S.R. . Nat. Biotechnol. 1999, 17, 1109–1111.

43. Wheeler, A.R.; Morishima, K.; Arnold, D.W.; Rossi,


A.B.; Zare, R.N. Single Organelle Analysis with Integrated
Chip Electrophoresis and Optical Tweezers. In Micro Total
Analysis Systems 2000; van den Berg, A., Olthuis, W.,
Bergveld, P., Eds.; Kluwer Academic Publishers: Enschede,
The Netherlands, 2000; 25–28.

44. Ashkin, A.; Dziedzic, J.M. Observation of


radiationpressure trapping of particles by alternating
light beams. Phys. Rev. Lett. 1985, 54, 1245–1248.

45. Ashkin, A.; Dziedzic, J.M.; Bjorkholm, J.E.; Chu, S.


Observation of a single-beam gradient force optical trap
for dielectrical particles. Opt. Lett. 1986, 11, 228–290.

46. Perkins, T.T.; Smith, D.E.; Larson, R.G.; Chu, S.


Stretching of a single tethered polymer in a uniform flow.
Science 1995, 268, 83–87.

47. Ashkin, A.; Dziedzic, J.M. Optical trapping and


manipulation of viruses and bacteria. Science 1987, 235,
1517–1520.

48. Svoboda, K.; Block, S.M. Biological applications of


optical forces. Annu. Rev. Biophys. Biomol. Struct. 1994,
23, 247–285.

49. Fuhr, G.R.; Reichle, C. Living Cells in Opto-Electrical


Cages: Characterisation, Manipulation and Force
Measurements. InMicro Total Analysis System; van den Berg,
A., Olthuis, W., Bergveld, P., Eds.; Kluwer Academic
Publishers: Enschede, The Netherlands, 2000; 261–264.

50. Darling, R.B.; Suh, J.W.; Kovacs, G.T. Ciliary


microactuator array for scanning electron microscope
positioning stage. J. Vac. Sci. Technol., A 1998, 16,
1998–2002.

51. Pister, K.S.J.; Fearing, R.; Howe, R. A Planar Air


Levitated Electrostatic Actuator System. In Proceedings:
IEEE Workshop on Micro Electro Mechanical Systems (MEMS);
1990; 67–71. Napa Valley, Calif.

52. Fujita, H. Group Work of Microactuators. In


International Advanced Robot Program Workshop on
Micromachine Technologies and Systems; 1993; 24–31. Tokyo,
Japan. 53. Bo¨hringer, K.-F.; Donald, B.R.; Mihailovich,
R.; MacDonald, N.C. A Theory of Manipulation and Control
for Microfabricated Actuator Arrays. In Proceedings: IEEE
Workshop on Micro Electro Mechanical Systems (MEMS); 1994.
Oiso, Japan. 54. Storment, C.W.; Borkholder, D.A.;
Westerlind, V.; Suh, J.W.; Maluf, N.I.; Kovacs, G.T.A.
Flexible, dry-released process for aluminum electrostatic
actuators. J. Microelectromech. Syst. 1994, 3, 90–96. 55.
Suh, J.W.; Glander, S.F.; Darling, R.B.; Storment, C.W.;
Kovacs, G.T.A. Combined Organic Thermal and Electrostatic
Omnidirectional Ciliary Microactuator Array for Object
Positioning and Inspection. In Proceedings: Solid-State
Sensor and Actuator Workshop; 1996. Hilton Head, S.C. 56.
Liu, C.; Tsao, T.; Will, P.; Tai, Y.; Liu, W. A
MicroMachined Magnetic Actuator Array for MicroRobotics
Assembly Systems. In Digest: Int. Conf. on Solid-State
Sensors and Actuators; Transducers Research Foundation:
Stockholm, Sweden, 1995. 57. Liu, W.; Will, P. Parts
Manipulation on an Intelligent Motion Surface. In IEEE/RSJ
Int. Workshop on Intelligent Robots & Systems (IROS); 1995;
PIttsburgh, Penn. 58. Edman, C.F.; Swint, R.B.; Gurtner,
C.; Formosa, R.E.; Roh, S.D.; Lee, K.E.; Swanson, P.D.;
Ackley, D.E.; Coleman, J.J.; Heller, M.J. Electric field
directed assembly of an ingaas led onto silicon circuitry.
IEEE Photonics Technol. Lett. 2000, 12, 1198–1200. 59.
Seeman, N.C. J. Theor. Biol. 1982, 99, 237. 60. Seeman,
N.C. Nanotechnology 1991, 149. 61. Seeman, N.C.; Zhang, Y.;
Chen, J. J. Vac. Sci. Technol. 1993, A12, 1895. 62. Seeman,
N.C. Annu. Rev. Biophys. Biomol. Struct. 1998, 27, 225. 63.
Winfree, E.; Liu, F.; Wenzler, L.; Seeman, N.C. Nature
1998, 394, 539. 64. Mirkin, C.A.; Letsinger, R.L.; Mucic,
R.C.; Storhoff, J.J. Nature 1996, 382, 607. 65. Martin,
B.R.; Dermody, D.J.; Reiss, B.D.; Fang, M.; Lyon, L.A.;
Natan, M.J.; Mallouk, T.E. Adv. Mater. 1999, 11, 1021.
Micro-Particle Image Velocimetry in
Biomedical Applications

28. Adrian, R.J.; Yao, C.S. Pulsed laser technique


application to liquid and gaseous flows and the scattering
power of seed materials. Appl. Optics 1985, 24, 44–52.

29. Cao, J.; Wereley, S.T. Brownian Particle Distribution


in Tube Flows, Proceedings of the ASME International
Mechanical Engineering Congress and Exposition, Anaheum,
CA, Nov 13–19, 2004.

30. Fuller, C.K.; Hamilton, J.; Ackler, H.; Krulevitch, P.;


Boser, B.; Eldredge, A.; Becker, F.; Yang, J.; Gascoyne, P.
Microfabricated multi-frequency particle impedance
characterization system. Micro Total Anal. Syst. 2000, 11,
265–268.

31. Anderson, G.P.; King, K.D.; Gaffney, L.H.; Johnson,


L.H. Multi-analyte interrogation using the fiber optic
biosensor. Biosens. Bioelectronics 2000, 14, 771–777.

32. Eu, J.Y.; Wang, C.Y.; Andrade, J. Homogeneous


bioluminescence assay for Galactosuria: interference and
kinetic analysis. Anal. Biochem. 1999, 271 (2), 168–176.

33. Olsen, M.G.; Adrian, R.J. Brownian motion and


correlation in particle image velocimetry. Opt. Laser
Technol. 2000, 32, 621–627.

34. Hohreiter, V.; Wereley, S.T.; Olsen, M.G.; Chung, J.N.


Cross-correlation analysis for temperature measurement.
Meas. Sci. Technol. 2002, 13, 1072–1078. 35. Meinhart,
C.D.; Wereley, S.T.; Gray, M.H.B. Volume illumination for
two-dimensional particle image velocimetry. Meas. Sci.
Technol. 2000, 11, 809–814. 36. Olsen, M.G.; Adrian, R.J.
Out-of-focus effects on particle visibility and correlation
in microscopic particle image velocimetry. Exp. Fluids
2000, 29, 166–174. 37. Wittwer, C.T.; Fillmore, G.C.;
Garling, D.J. Minimizing the time required for DNA
amplification by efficient heat transfer to small samples.
Anal. Biochem. 1990, 186, 328–331. 38. Chiou, J.;
Matsudaira, P.; Sonin, A.; Ehrlich, D. A closed-cycle
capillary polymerase chain reaction machine. Anal. Chem.
2001, 79 (3), 2018–2021. 39. Ross, D.; Gaitan, M.;
Locascio, L.E. Temperature measurement in microfluidic
systems using a temperature-dependent fluorescent dye. Anal.
Chem. 2001, 73, 4117–4123. 40. Seger-Sauli, U.; Panayiotou,
M.; Schnydrig, S.; Jordan, M.; Renaud, P. Temperaturement
measurements in microfluidic systems: heat dissipation of
negative dielectrophoresis barriers. Electrophoresis 2005,
26 (11), 2239–2246.
Microporous Materials

1. Davies, J.E. Bone Engineering; EM 2 Incorporated:


Toronto, 2000.

2. Langer, R.; Vacanti, J.P. Tissue engineering. Science


1993, 260 (5110), 920–926.

3. Jones, J.R.; Hench, L.L. Biomedical materials for the


new millennium: A perspective on the future. J. Mater. Sci.
Technol. 2001, 17, 891–900. 4. Ohgushi, H.; Caplan, A.I.
Stem cell technology and bioceramics: From cell to gene
engineering. J. Biomed. Mater. Res. 1999, 48, 913–927. 5.
Lu, J.X.; Flautre, B.; Anselme, K.; Hardoiun, P.; Gallur,
A.; Deschamps, M.; Thierry, B. Role of interconnections in
porous bioceramics on bone recolonisation in vitro and in
vivo. J. Mater. Sci., Mater. Med. 1999, 10, 111–120. 6.
Okii, N.; Nishimura, S.; Kurisu, K.; Takeshima, Y.; Uozumi,
T. In vivo histological changes occurring in hydroxyapatite
cranial reconstruction—Case report. Neurol. Med. 2001, 41
(2), 100–104. 7. Hench, L.L. Bioceramics: From concept to
clinic. J. Am. Ceram. Soc. 1991, 74 (7), 1487–1510. 8.
Gibson, I.R.; Bonfield, W. Novel synthesis and
characterisation of an AB-type carbonate-substituted
hydroxyapatite. J. Biomed. Mater. Res. 2002, 59, 697– 708.
9. Ota, Y.; Kasuga, T.; Abe, Y. Preparation and compressive
strength behaviour of porous ceramics with bCa(PO 3 ) 2
fiber skeletons. J. Am. Ceram. Soc. 1997, 80 (1), 225–231.
10. Andrade, J.C.T.; Camilli, J.A.; Kawachi, E.Y.; Bertran,
C.A. Behaviour of dense and porous hydroxyapatite implants
and tissue response in rat femoral defects. J. Biomed.
Mater. Res. 2002, 62, 30–36. 11. Komlev, V.S.; Barimov,
S.M. Porous hydroxyapaptite ceramics of bi-modal pore size
distribution. J. Mater. Sci., Mater. Med. 2002, 13,
295–299. 12. Yin, S.; Uchida, S.; Fujishiro, Y.; Ohmori,
M.; Sato, T. Preparation of porous ceria doped tetragonal
zirconia ceramics by capsule free hot isostatic pressing.
Br. Ceram. 1999, 98 (1), 19–23. 13. Fabbri, M.; Celotti,
G.C.; Ravaglioli, A. Hydroxyapatite-based porous
aggregates: Physical-chemical nature, structure, texture
and architecture. Biomaterials 1995, 16, 225–228. 14.
Engin, N.O.; Tas, A.C. Manufacture of macroporous calcium
hydroxyapatite bioceramics. J. Eur. Ceram. Soc. 1999, 19,
2569–2572. 15. Zhou, W.; Hu, W.B.; Zhang, D. Combustion
synthesis of highly porous ceramics: The TiC–Al 2 O 3
system. J. Mater. Sci. 1999, 34, 4469–4473. 16. Fukasawa,
T.; Deng, Z.-Y.; Ando, M.; Ohji, T.; Goto, Y. Pore
structure of porous ceramics synthesised from water-based
slurry by freeze-dry process. J. Mater. Sci. 2001, 36,
2523–2527. 17. Zhang, Y.; Zhang, M. Three-dimensional
macroporous calcium phosphate bioceramics with nested
chitosan sponges for load-bearing bone implants. J. Biomed.
Mater. Res. 2002, 61 (1), 1–8. 18. Callcut, S.; Knowles,
J.C. Correlation between structure and compressive strength
in a reticulated glassreinforced hydroxyapatite foam. J.
Mater. Sci., Mater. Med. 2002, 13, 485–489. 19. Columbo,
P.; Griffoni, M.; Modesti, M. Ceramic foams from a
preceramic polymer and polyurethanes: Preparation and
morphological investigations. J. SolGel Sci. 1998, 13,
195–199. 20. Takahashi, T.; Munstedt, H.; Colombo, P.;
Modesti, M. Thermal evolution of a silicon
resin/polyurethane blend from preceramic to ceramic foam.
J. Mater. Sci. 2001, 36, 1627–1639. 21. Chu, G.T.-M.;
Orton, D.G.; Hollister, S.J.; Feinberg, S.E.; Halloran,
J.W. Mechanical and in vivo performance of hydroxyapatite
implants with controlled architectures. Biomaterials 2002,
23, 1283–1293. 22. Sepulveda, P.; Jones, J.R.; Hench, L.L.
Bioactive sol-gel foams for tissue repair. J. Biomed.
Mater. Res. 2002, 59 (2), 340–348. 23. Hench, L.L.; West,
J.K. The sol-gel process. Chem. Rev. 1990, 90, 33–72. 24.
Li, P.; Zhang, F. The electrochemistry of a glass-surface
and its application to bioactive glass in solution. J.
Non-Cryst. 1990, 119, 112–116. 25. Balas, F.; Arcos, D.;
Perez-Periente, J.; Vallet-Regi, M. Textural properties of
SiO 2 –CaO–P 2 O 5 glasses prepared by the sol-gel method.
J. Mater. Res. 2001, 16 (5), 1345–1348. 26. Nakanishi, K.
Porous gels made by phase separation: Recent progress and
future directions. J. Sol-Gel Sci. Technol. 2000, 19,
65–70. 27. Statham, M.J.; Hammett, F.; Harris, B.; Cooke,
R.G.; Jordan, R.M.; Roche, A.J. Net-shape manufacture of
low-cost ceramic shapes by freeze-gelation. J. Sol-Gel Sci.
1998, 12, 171–175. 28. Lebeau, B.; Fowler, C.E.; Mann, S.;
Farcet, C.; Charleux, B.; Sanchez, C. Synthesis of
hierarchiacally ordered dye-functionalised mesoporous
silica with macroporous architecture by dual templating. J.
Mater. Chem. 2000, 10, 2105–2108. 29. Khramov, A.N.;
Collinson, M.M. Sol-gel preparation of macroporous silica
films by templating with polystyrene microspheres. Chem.
Commun. 2001, 8, 767–768. 30. Yan, H.; Zhang, K.;
Blandford, C.F.; Francis, L.F.; Stein, A. In vitro
hydroxycarbonate apatite mineralisation of CaO–SiO 2
sol-gel glasses with a threedimensionally ordered
macroporous structure. Chem. Mater. 2001, 13, 1374–1382.
31. Sepulveda, P.; Binner, J.G.P. Processing of cellular
ceramics by foaming and in situ polymerisation of organic
monomers. J. Eur. Ceram. Soc. 1999, 19, 2059–2066. 32.
Rosen, M.J. Surfactants and Interfacial Phenomena, 2nd Ed.;
John Wiley & Sons: New York, 1989; 277–303. 33. Bikerman,
J.J. Foams; Springer Verlag: New York, 1973. 34. Sepulveda,
P.; Binner, J.G.P.; Rogero, S.O.; Higa, O.Z.; Bressiani,
J.C. Production of porous hydroxyapatite by the gel-casting
of foams and cytotoxic evaluation. J. Biomed. Mater. Res.
2000, 50, 27–34. 35. Gun, J.; Lev, O.; Regev, O.; Pevzner,
S.; Kucernak, A. Sol-gel formation of reticular
methyl-silicate materials by hydrogen peroxide
decomposition. J. Sol-Gel Sci. 1998, 13, 189–193. 36. Wu,
M.; Fujiu, T.; Messing, G.L. Synthesis of cellular
inorganic materials by foaming sol-gels. J. Non-Cryst.
1990, 121, 407–412. 37. Jones, J.R.; Hench, L.L. Effect of
porosity on the mechanical properties of bioactive foam
scaffolds. Key Eng. Mater. 2003, 240–242, 209–212. 38.
Xynos, I.D.; Edgar, A.J.; Buttery, L.D.K.; Hench, L.L.;
Polak, J.M. Ionic products of bioactive glass dissolution
increase proliferation of human osteoblasts and induce
insulin-like growth factor II mRNA expression and protein
synthesis. Biochem. Biophys. Res. Commun. 2000, 276,
461–465. 39. Lenza, R.F.S.; Jones, J.R.; Vasconcelos, W.L.;
Hench, L.L. Surface-modified 3D scaffolds for tissue
engineering. J. Mater. Sci., Mater. Med. 2002, 13, 837–842.
40. Tan, A.; Romanska, H.M.; Lenza, R.; Jones, J.; Hench,
L.L.; Polak, J.M.; Bishop, A.E. The effect of 58S bioactive
glass sol-gel derived foams on the growth of murine lung
epithelial cells. Key Eng. Mater. 2003, 240–242, 209–212.
41. Jones, J.R.; Hench, L.L. The effect of surfactant
concentration and glass composition on the structure and
properties of bioactive foam scaffolds. J. Mater. Sci.
2003, 38, 3783–3790. 42. Livingston, T.; Ducheyne, P.;
Garino, J. In vivo evaluation of a bioactive scaffold for
bone tissue engineering. J. Biomed. Mater. Res. 2002, 62,
1–13. 43. Yuan, H.; de Bruijn, J.D.; Zhang, X.; van
Blitterswijk, C.A.; de Groot, K. Bone induction by porous
glass ceramic made from Bioglass s (45S5). J. Biomed.
Mater. Res. 2001, 58 (3), 270–276. 44. Bonfield, W.;
Grynpas, M.D.; Tully, A.E.; Bowmann, J.; Abram, J.
Biomechanics of bone. Biomaterials 1981, 2 (3), 185–188.
45. Roether, J.A.; Gough, J.E.; Boccacini, A.R.; Hench,
L.L.; Maquet, V.; Jerome, R. Novel bioresorbable and
bioactive composites based on bioactive glass and
polylactide foams for bone tissue engineering. J. Mater.
Sci., Mater. Med. 2002, 13 (12), 1207–1214. 46. Day, R.;
Boccaccini, A.R.; Roether, J.A.; Surey, S.; Forbes, A.;
Hench, L.L.; Gabe, S. The effect of Bioglass s on
epithelial cell and fibroblast proliferation and
incorporation into a PGA matrix. Gastroenterology 2002, 122
(4), T875. Suppl. 1. M
Modeling, Biomedical

4. Halley, E. An estimate of the degrees of mortality of


mankind, drawn from curious tables of the births and
funerals at the city of Breslaw; With an attempt to
ascertain the price of annuities on lives. Philos. Trans.
R. Soc. Lond. 1693, 17, 596–610.

5. Einthoven, W. Die galvanometrische registrirung des


mensclichen elecktrokardiogramms, zugleich eine
beurtheilung der anwendung des capialr-elektrometers in der
physiologie. Pfluegers Arch. 1903, 99, 472–480.

6. Hodgkin, A.L.; Huxley, A.F. Currents carried by sodium


and potassium ions through the membrane of the giant axon
of Logio. J. Physiol. 1952, 116, 448–472.

7. Hodgkin, A.L.; Huxley, A.F. A quantitative description


of membrane current and its application to conduction and
excitation in nerve. J. Physiol. 1952, 117, 500–544.

8. Huxley, A.F. Muscle structure and theories of


contraction. Prog. Biophys. 1957, 7, 255–318.

9. Craig, R.; Lehman, W. Crossbridge and tropomyosin


positions observed in native, interacting thick and thin
filaments. J. Mol. Biol. 2001, 31 (311(5)), 1027–1036.

10. Moore, G.E. Cramming more components onto integrated


circuits. Electronics 1965, 38 (8), 114–117.

11. Rudy, Y. From Genome to Physiome: Integrative Models of


Cardiac Excitation. Ann. Biomed. Eng. 2000, 28, 945–950.

12. Bassingthwaighte, J. The Physiome Project: Why Now? and


How?, Proceedings of the Physiological Society of New
Zealand, Aug 2001; Vol 20. Supplement 1 (Proceedings of the
XXXIV International Congress of Physiological Sciences).

13. Mulquiney, P.J.; Smith, N.P.; Clark, K.; Hunter, P.J.


Mathematical modelling of the ischaemic heart. Nonlinear
Anal. 2001, 47, 235–244.

14. Noble, D.; Varghese, A.; Kohl, P.; Noble, P. Improved


guinea-pig ventricular cell model incorporating a diadic
space, IKr and IKs, and lengthand tensiondependent
processes. Can. J. Cardiol. 1998, 14 (1), 123–134.

15. Luo, C.; Rudy, Y. A dynamic model of the cardiac


ventricular action potential. I. Simulations of ionic
currents and concentration changes. Circ. Res. 1994, 74
(6), 1071–1096. 16. Luo, C.; Rudy, Y. A dynamic model of
the cardiac ventricular action potential. II.
Afterdepolarizations, triggered activity, and potentiation.
Circ. Res. 1994, 74 (6), 1097–1113. 17. Hunter, P.J.;
McCulloch, A.D.; ter Keurs, H.E. Modelling the mechanical
properties of cardiac muscle. Prog. Biophys. Mol. Biol.
1998, 69 (2–3), 289–331. 18. Salem, J.E.; Saiel, G.M.;
Stanley, W.C.; Cabrera, M.E. Mechanistic model of
myocardial energy metabolism under normal and ischemic
conditions. Ann. Biomed. Eng. 2002 , 30 (2), 202–216. 19.
Legrice, I.J.; Hunter, P.J.; Smaill, B.H. Laminar structure
of the heart: A mathematical model. Am. J. Physiol. 1997,
272 (5 Pt 2), H2466–H2476. 20. Hooks, D.A.; Tomlinson,
K.A.; Marsden, S.G.; LeGrice, I.J.; Smaill, B.H.; Pullan,
A.J.; Hunter, P.J. Cardiac microstructure: Implications for
electrical propagation and defibrillation in the heart.
Circ. Res. 2002, 91 (4), 331–338. 21. Smith, N.P.; Pullan,
A.J.; Hunter, P.J. Generation of an anatomically based
geometric coronary model. Ann. Biomed. Eng. 2000, 28 (1),
14–25. 22. Kassab, G.S.; Rider, C.A.; Tang, N.J.; Fung,
Y.C. Morphometry of pig coronary arterial trees. Am. J.
Physiol. 1993, 265 (1 Pt 2), H350–H365. 23. Bradley, C.P.;
Pullan, A.J.; Hunter, P.J. Effects of material properties
and geometry on electrocardiographic forward simulations.
Ann. Biomed. Eng. 2000, 28 (7), 721–741. 24. International
Human Genome Mapping Consortium. A physical map of the
human genome. Nature 2001, 409 (6822), 934–941. 25.
PricewaterhouseCoopers. An industrial revolution in R&D.
Pharma 2005, 1998. http://www.pwcglobal.com/
gx/eng/about/ind/pharma/industrial_revolution.pdf. 26.
Noble, D.; Colatsky, T.J. A return to rational drug
discovery: Computer-based models of cells, organs and
systems in drug target identification. Emerg. Ther. Targets
2000, 4, 39–49.
Nanoscale Neurosurgery

13. Dempsey, E.F.; Luse, S. Fine Strucure of the Neuropil


in Relation to the Neuroglial Cells. In Biology of
Neuroglia; Windle, W., Ed.; Charles C. Thomas: Springfield,
IL, 1958; 99.

14. Horstmann, E. Was wissen wir u¨ber den intercellula¨ren


Raum in ZNS? World Neurol. 1962, 3, 112–117.

15. Nicholson, C.; Sykova, E. Extracellular space structure


revealed by diffusion analysis. Trends Neurosci. 1998, 21,
207–215.

16. Nicholson, C. Diffusion and related transport


mechanisms in brain tissue. Rep. Prog. Phys. 2001, 64,
815–854.

17. Rech, R.H.; Domino, E.F. Observations on injections of


drugs into the brain substance. Arch. Int. Pharmacodyn.
1959, 121, 429–442.

18. Nicholson, C.; Phillips, J.M. Ion diffusion modified by


tortuosity and volume fraction in the extracellular
microenvironment of the rat cerebellum. J. Physiol. 1981,
321, 225–257.

19. Bobo, R.H.; Laske, D.W.; Akbasak, A.; Morrison, P.F.;


Dedrick, R.L.; Oldfield, E.H. Convection-enhanced delivery
of macromolecules in the brain. Proc. Natl. Acad. Sci. U.
S. A. 1994, 91, 2076–2080.

20. Nicholson, C. Diffusion from an injected volume of a


substance in brain tissue with arbitrary volume fraction
and tortuosity. Brain Res. 1985, 333, 325–329.

21. Basser, P.J. Interstitial pressure, volume, and flow


during infusion into brain tissue. Microvasc. Res. 1992,
44, 143–165.

22. Morrison, P.F.; Laske, D.W.; Bobo, R.H.; Oldfield, E.H.;


Dedrick, R.L. High-flow microinfusion: Tissue penetration
and pharmacodynamics. Am. J. Physiol. 1994, 266, R292–R305.
23. Kucharczyk, J.; Moseley, M.E. Method and Apparatus for
Use with MR Imaging. U.S. Patent 6,061,587, May 9, 2000.
24. Broaddus, W.C.; Gillies, G.T.; Kucharczyk, J. Advances
in image-guided delivery of drug and cell therapies into
the central nervous system. Neuroimaging Clin. N. Am. 2001,
11, 727–735. 25. Broaddus, W.C.; Gillies, G.T.; Kucharczyk,
J. ImageGuided Intraparenchymal Drug and Cell Delivery. In
Diagnostic and Therapeutic Imaging of the Nervous System;
Latchaw, R.E., Kucharczyk, J., Moseley, M.E., Yarborough,
B.V., Eds.; Mosby: Philadelphia, 2003. 26. Gillies, G.T.;
Allison, S.W.; Tissue, B.M. Positive pressure infusion of
fluorescent nanoparticles as a probe of the structure of
brain phantom gelatins. Nanotechnology 2002, 13, 484–486.
27. Gillies, G.T.; Wilhelm, T.D.; Humphrey, J.A.C.;
Fillmore, H.L.; Holloway, K.L.; Broaddus, W.C. Spinal cord
surrogate with nanoscale porosity for in vitro simulations
of restorative neurosurgical techniques. Nanotechnology
2002, 13, 587–591. 28. Fillmore, H.L.; Chasiotis, I.; Cho,
S.W.; Gillies, G.T. Atomic force microscopy observations of
tumour cell invadopodia: Novel cellular nanomorphologies on
collagen substrates. Nanotechnology 2003, 14, 73–76. 29.
Chasiotis, I.; Fillmore, H.L.; Gillies, G.T. Atomic force
microscopy measurement of cytostructural elements involved
in the nanodynamics of tumour cell invasion. Nanotechnology
2003, 14, 557–561. 30. Smith, D.H.; Wolf, J.A.; Meaney,
D.F. A new strategy to produce sustained grwoth of central
nervous system axons: Continuous mechanical tension. Tissue
Eng. 2001, 7, 131–139.
Nerve Guides

19. Valentini, R.F.; Sabatini, A.M.; Dario, P.; Aebischer,


P. Polymer electret guidance channels enhance peripheral
nerve regeneration in mice. Brain Res. 1989, 480 (1–2),
300–304.

20. Young, R.C.; Wiberg, M.; Terenghi, G.


Poly-3-hydroxybyturate (PHB): A resorbable conduit for long
gap repair in peripheral nerves. Br. J. Plast. Surg. 2002,
55, 235–240.

21. Yoshii, S.; Oka, M. Collagen filaments as a scaffold for


nerve regeneration. J. Biomed. Mater. Res. 2001, 56 (3),
400–405.

22. Molander, H.; Olsson, Y.; Engquist, O.; Bowald, S.;


Eriksson, I. Regeneration of peripheral nerve through a
polyglactin tube. Muscle Nerve 1982, 5, 54.

23. Gibson, K.L.; Remson, L.; Smith, A.; Satterlee, N.;


Strain, G.M.; Daniloff, J.K. Comparison of nerve
regeneration through different types of neural prostheses.
Microsurgery 1991, 12 (2), 80–85.

24. Mackinnon, S.E.; Dellon, A.L. Clinical nerve


reconstruction with a bioabsorbable polyglycolic acid tube.
Plast. Reconstr. Surg. 1990, 85 (3), 419–424.

25. Keeley, R.D.; Nguyen, K.D.; Stephanides, M.J.; Padilla,


J.; Rosen, J.M. The artificial nerve graft: A comparison of
blended elastomer-hydrogel with polyglycolic acid conduits.
J. Reconstr. Microsurg. 1991, 7 (2), 93–100.

26. den Dunnen, W.F.A.; Schakenraad, J.M.; Zondervan, G.J.;


Pennings, A.J.; van der Lei, B.; Robinson, P.H. A new
PLLA/PCL copolymer for nerve regeneration. J. Mater. Sci.,
Mater. Med. 1993, 4, 521–525.

27. Hare, G.M.; Evans, P.J.; Mackinnon, S.E.; Best, T.J.;


Bain, J.R.; Szalai, J.P.; Hunter, D.A. Walking track
analysis: A long-term assessment of peripheral nerve
recovery. Plast. Reconstr. Surg. 1992, 89 (2), 251–258.

28. Evans, G.R.D.; Brandt, K.; Widmer, M. In vivo


evaluation of poly(L-Lactic acid) porous conduits for
peripheral nerve regeneration. Biomaterials 1999, 20,
1109–1115.

29. Oudega, M.; Gautier, S.E.; Chapon, P.; Fragoso, M.;


Bates, M.L.; Parel, J.M.; Bunge, M.B. Axonal regeneration
into Schwann cell grafts within resorbable
poly(alpha-hydroxyacid) guidance channels in the adult rat
spinal cord. Biomaterials 2001, 22, 1125–1136.

30. Fabre, T.; Schappacher, M.; Bareille, R.; Dupuy, B.;


Soum, A.; Bertrand-Barat, J.; Baquey, C. Study of a
(trimethylenecarbonate-co-e-caprolactone) polymer— Part 2:
In vitro cytocompatibility analysis and in vivo ED1 cell
response of a new nerve guide. Biomaterials 2001, 22,
2951–2958.

31. den Dunnen, W.F.A.; Stokroos, I.; Blaauw, E.H.;


Holwerda, A.; Pennings, A.J.; Robinson, P.H.; Schakenraad,
J.M. Light-microscopic and electron-microscopic evaluation
of short-term nerve regeneration using a biodegradable
poly(DL-lactide-epsilon-caprolacton) nerve guide. J.
Biomed. Mater. Res. 1996 , 31, 105–115.

32. Robinson, P.H.; van der Lei, B.; Hoppen, H.J.;


Leenslag, J.W.; Pennings, A.J.; Nieuwenhuis, P. Nerve
regeneration through a two-ply biodegradable nerve guide in
the rat and the influence of ACTH4–9 nerve growth factor.
Microsurgery 1991, 12 (6), 412–419. 33. Langone, F.; Lora,
S.; Veronese, F.M.; Caliceti, P.; Parnigotto, P.P.;
Valenti, F.; Palma, G. Peripheral nerve repair using a
poly(organo)phosphazene tubular prosthesis. Biomaterials
1995, 16 (5), 347–353. 34. Bellamkonda, R.; Ranieri, J.P.;
Aebischer, P. Laminin oligopeptide derivatized agarose gels
allow three-dimensional neurite extension in vitro. J.
Neurosci. Res. 1995, 41, 501–509. 35. Schense, J.C.; Bloch,
J.; Aebischer, P.; Hubbell, J.A. Enzymatic incorporation of
bioactive peptides into fibrin matrices enhances neurite
extension. Nat. Biotechnol. 2000, 18, 415–419. 36. Woerly,
S. Microreconstruction Surgery of the Central Nervous
System by Means of Engineered Polymer Constructs; Sames,
K., Ed.; Ecomed: Landsberg-Lech, 2000; 1–14. 37. Sufan, W.;
Suzuki, Y.; Tanihara, M.; Ohnishi, K.; Suzuki, K.; Endo,
K.; Nishimura, Y. Sciatic nerve regeneration through
alginate with tubulation or nontubulation repair in cat. J.
Neurotrauma 2001, 18 (3), 329–338. 38. Toba, T.; Nakamura,
T.; Shimizu, Y.; Matsumoto, K.; Ohnishi, K.; Fukuda, S.;
Yoshitani, M.; Ueda, H.; Hori, Y.; Endo, K. Regeneration of
canine peroneal nerve with the use of a polyglycolic
acid-collagen tube filled with laminin-soaked collagen
sponge: A comparative study of collagen sponge and collagen
fibers as filling materials of nerve conduits. J. Biomed.
Mater. Res. 2001, 58, 622–630. 39. Novikov, L.N.; Novikova,
L.N.; Mosahebi, A.; Wiberg, M.; Terenghi, G.; Kellerth,
J.-O. A novel biodegradable implant for neuronal rescue and
regeneration after spinal cord injury. Biomaterials 2002,
23, 3369–3376. 40. Anton, E.S.; Weskamp, G.; Reichardt,
L.F.; Matthew, W.D. Nerve growth factor and its low-affinity
receptor promote Schwann cell migration. Proc. Natl. Acad.
Sci. U. S. A. 1994, 91 (7), 2795–2799. 41. Fu, S.Y.;
Gordon, T. The cellular and molecular basis of peripheral
nerve regeneration. Mol. Neurobiol. 1997, 14 (1–2), 67–116.
42. Barras, F.M.; Pasche, P.; Bouche, N.; Aebischer, P.
Glial cell line-derived neurotrophic factor released by
synthetic guidance channels promotes facial nerve
regeneration in the rat. J. Neurosci. Res. 2002, 70,
746–755. 43. Fine, E.G.; Decosterd, I.; Papaloizos, M.;
Zurn, A.D.; Aebischer, P. GDNF and NGF released by
synthetic guidance channels support sciatic nerve
regeneration across a long gap. Eur. J. Neurosci. 2002, 15,
589–601. 44. Bloch, J.; Fine, E.G.; Bouche, N.; Zurn, A.D.;
Aebischer, P. Nerve growth factorand
neurotrophin-3-releasing guidance channels promote
regeneration of the rat dorsal root. Exp. Neurol. 2001,
172, 425–432. 45. Nakahara, Y.; Gage, F.H.; Tuszynksi, M.H.
Grafts of fibroplasts genetically modified to secrete NGF,
BDNF, NT-3, or basic FHF elicit differential responses in
the adult spinal cord. Cell Transplant 1996, 5, 191–204.
46. Steuer, H.; Fadale, R.; Mu¨ller, E.; Mu¨ller, H.W.;
Planck, H.; Schlosshauer, B. Biohybrid nerve guide for
regeneration: Degradable polylactide fibers coated with rat
Schwann cells. Neurosci. Lett. 1999, 277, 165–168. 47.
Hobson, M.I.; Green, C.J.; Terenghi, G. VEGF enhances
intraneural angiogenesis and improves nerve regeneration
after axotomy. J. Anat. 2000, 197, 591–605. 48. Rutkowski,
J.L.; Kirk, C.J.; Lerner, M.A.; Tennekoon, G.I. Purification
and expansion of human Schwann cells in vitro. Nat. Med.
1995, 1 (1), 80–83. 49. Bamber, N.I.; Li, H.; Lu, X.;
Oudega, M.; Aebischer, P.; Xu, X.M. Neurotrophins BDNF and
NT-3 promote axonal re-entry into the distal host spinal
cord through Schwann cell-seeded mini-channels. Eur. J.
Neurosci. 2001, 13, 257–268. 50. Aebischer, P.; Guenard,
V.; Winn, S.R.; Valentini, R.F.; Galletti, P.M. Blind-ended
semipermeable guidance channels support peripheral nerve
regeneration in the absence of a distal nerve stump. Brain
Res. 1988, 454, 179–187. 51. Lundborg, G.; Dahlin, L.B.;
Danielsen, N.; Nachemson, A.K. Tissue specificity in nerve
regeneration. Scand. J. Plast. Reconstr. Surg. 1986, 20
(3), 279–283. 52. Tong, X.J.; Hirai, K.; Shimada, H.;
Mizutani, Y.; Izumi, T.; Toda, N.; Yu, P. Sciatic nerve
regeneration navigated by laminin-fibronectin double coated
biodegradable collagen grafts in rats. Brain Res. 1994, 663
(1), 155–162. 53. Aebischer, P.; Guenard, V.; Brace, S.
Peripheral nerve regeneration through blind-ended
semipermeable guidance channels: Effect of the molecular
weight cutoff. J. Neurosci. 1989, 9 (10), 3590–3595. 54.
Schlosshauer, B.; Brinker, T.; Mu¨ller, H.-W.; Meyer, J.U.
Towards micro electrode implants: In vitro guidance of rat
spinal cord neurites through polyimide sieves by Schwann
cells. Brain Res. 2001. 55. Navarro, X.; Calvet, S.;
Rodrı´guez, F.J.; Stieglitz, T.; Blau, C.; Butı´, M.;
Valderrama, E.; Meyer, J.U. Stimulation and recording from
regenerated peripheral nerves through polyimide sieve
electrodes. J. Peripher. Nerv. Syst. 1998, 3 (2), 91–101.
56. Stanec, S.; Stanec, Z. Reconstruction of
upper-extremity peripheral-nerve injuries with ePTFE
conduits. J. Reconstr. Microsurg. 1998, 14 (4), 227–232.
57. Lundborg, G.; Rosen, B.; Abrahamson, S.O.; Dahlin, L.;
Danielsen, N. Tubular repair of the median nerve in the
human forearm. Preliminary findings. J. Hand Surg., Br.
1994, 19 (3), 273–276. 58. Lundborg, G.; Rosen, B.; Dahlin,
L.; Danielsen, N.; Holmberg, J. Tubular versus conventional
repair of median and ulnar nerves in the human forearm:
Early results from a prospective, randomized, clinical
study. J. Hand Surg., Am. 1997, 22 (1), 99–106. 59.
Braga-Silva, J. The use of silicone tubing in the late
repair of the median and ulnar nerves in the forearm. J.
Hand Surg., Br. 1999, 24 (6), 703–706. 60. Weber, R.A.;
Breidenbach, W.C.; Brown, R.E.; Jabaley, M.E.; Mass, D.P. A
randomized prospective study of polyglycolic acid conduits
for digital nerve reconstruction in humans. Plast.
Reconstr. Surg. 2000, 106 (5), 1036–1045. 61. Kim, J.;
Dellon, A.L. Reconstruction of a painful posttraumatic
medial plantar neuroma with a bioresobable nerve conduit: A
case report. J. Foot Ankle Surg. 2001, 40 (5), 318–323. 62.
Dahlin, L.B.; Lundborg, G. Use of tubes in peripheral nerve
repair. Neurosurg. Clin. N. Am. 2001, 12, 341–352. 63.
Patrick, C.W.J.; Mikos, A.G.; McIntire, L.V. Prospectus in
Tissue Engineering; Pergamon: Oxford, 1998; 3–11. 64.
Langer, R.; Vacanti, J.P. Tissue engineering. Science 1993,
260, 920–926. 65. Lysaght, M.J.; Nguy, N.A.; Sullivan, K.
An economic survey of the emerging tissue engineering
industry. Tissue Eng. 1998, 4 (3), 231–238. 66. Lang, C.;
Stefan, H. Sports injuries of the nervous system. Fortschr.
Neurol. Psychiatr. 1999, 67 (8), 373–384. 67. Marino, R.J.;
Ditunno, J.F.; Donovan, W.H.; Maynard, F.J. Neurologic
recovery after traaumatic spinal cord injury: Data from the
model spinal cord injury systems. Arch. Phys. Med. Rehabil.
1999, 80 (11), 1391–1396. 68. Bonilla, I.E.; Tanabe, K.;
Strittmatter, S.M. Small proline-rich repeat protein 1A is
expressed by axotomized neurons and promotes axonal
outgrowth. J. Neurosci. 2002, 22 (4), 1303–1315. 69.
Fournier, A.E.; Takizawa, B.T.; Strittmater, S.M. Rho
kinase inhibition enhances axonal regeneration in the
injured CNS. J. Neurosci. 2003, 23 (4), 1416. N
Neuroprostheses

12. Waters, R.L.; McNeal, D.; Perry, J. Experimental


correction of foot-drop by electrical stimulation of the
peroneal nerve. J. Bone Jt. Surg., Am. 1975, 57-A,
1047–1054.

13. Strojnik, P.; Acimovic, R.; Vavken, E.; Simic, V.;


Stanic, U. Treatment of drop foot using an implantable
peroneal underknee stimulator. Scand. J. Rehab. Med. 1987,
19, 37–43.

14. Haugland, M.K.; Sinkjaar, T. Cutaneous whole nerve


recordings used for correction for footdrop in hemiplegic
man. IEEE Trans. Biomed. Eng. 1995, 3, 307–317.

15. Burridge, J.H.; Taylor, P.N.; Hagan, S.A.; Wood, D.E.;


Swain, I.D. The effects of common peroneal stimulation on
the effort and speed of walking: A randomized controlled
trial with chronic hemiplegic patients. Clin. Rehabil.
1997, 11 (3), 201–210.

16. Strojnik, P.; Kralj, A.; Ursic, I. Programmed


six-channel electrical stimulator for complex stimulation
of leg muscles during walking. IEEE Trans. Biomed. Eng.
1979, 26, 112–116.

17. Willemsen, A.T.M.; Bloemhof, F.; Boom, H.B.K. Automatic


stance-swing phase detection from accelerometer data for
peroneal nerve stimulation. IEEE Trans. Biomed. Eng. 1990,
37, 1201–1208.

18. Dai, R.; Stein, R.B.; Andrews, B.J.; James, K.B.;


Wieler, M. Application of tilt sensors in functional
electrical stimulation. IEEE Trans. Rehabil. Eng. 1996, 4
(2), 63–72.

19. Sweeney, P.C.; Lyons, G.M. Fuzzy gait event detection


in a finite state controlled FES drop foot correction
system. J. Bone Jt. Surg., Br. 1999, 81-B (Suppl. I), 93.

20. Pappas, I.P.; Popovic, M.R.; Keller, T.; Dietz, V.;


Morari, M. A reliable gait phase detection system. IEEE
Trans. Neural Syst. Rehabil. Eng. 2001, 9 (2), 113–125.

21. Kralj, A.; Bajd, T.; Turk, R. Enhancement of gait


restoration in spinal injured patients by functional
electrical stimulation. Clin. Orthop. Relat. Res. 1988,
223, 34–43.
22. Graupe, D.; Davis, R.; Kordylewski, H.; Kohn, K.H.
Ambulation by traumatic T4-12 paraplegics using functional
neuromuscular stimulation. Crit. Rev. Neurosurg. 1998, 8,
221–231.

23. Graupe, D.; Kohn, K.H. Functional neuromuscular


stimulator for short-distance ambulation by certain
thoracic-level spinal-cord-injured paraplegics. Surg.
Neurol. 1998, 50, 202–207.

24. Davis, R.; Houdayer, T.; Andrews, B.J.; Barriskill, A.


In Paraplegia: Implanted Praxis247FES System for
Multi-Functional Restoration, Proceedings of the
International Functional Electrical Stimulation Society,
Sendai, Japan, 1999.

25. Kobetic, R.; Triolo, R.; Morsolais, E. Muscle selection


and walking performance of multichannel FES systems for
ambulation in paraplegia. IEEE Trans. Rehabil. Eng. 1997 ,
5, 23–28.

26. Smith, B.; Peckham, P.H.; Keith, M.; Roscoe, D. An


externally powered, multichannel, implantable stimulator
for versatile control of paralyzed muscle. IEEE Trans.
Biomech. Eng. 1987, 34 (7), 499–508. 27. Ijzerman, M.;
Stoffers, T.; ’t Groen, F.; Klatte, M.; Snoek, G.;
Vorsteveld, J.; Nathan, R.; Hermens, H. The NESS handmaster
orthosis: Restoration of hand function in C5 and stroke
patients by means of electrical stimulation. J. Rehabil.
Sci. 1996, 9 (3), 86–89. 28. Prochazka, A.; Gauthier, M.;
Wieler, M.; Kenwell, Z. The Bionic Glove: An electrical
stimulator garment that provides controlled grasp and hand
opening in quadriplegia. Arch. Phys. Med. Rehabil. 1997,
78, 1–7. 29. Hoshimiya, N.; Handa, Y. A master-slave type
multichannel functional electrical stimulation (FES) system
for the control of the paralyzed upper extremities.
Automedica 1989, 11, 209–220. 30. Popovic, M.R.; Hajek, V.;
Takaki, J.; Bulsen, A.; Zivanovic, V. In Restoration of
Reaching and Grasping Functions in Hemiplegic Patients with
Severe Arm Paralysis, From Technology to Market: Bridging
the Gap, Proceedings of the International Functional
Electrical Stimulation Society, Maroochydore, Australia,
July 1–5, 2003. 31. Adams, M.; Takes, V.; Popovic, M.R.;
Bulsen, A.; Zivanovic, V. Restoration of Grasping Functions
in Patients with Quadriplegia. From Technology to Market:
Bridging the GapProceedings of the International Functional
Electrical Stimulation Society, Maroochydore, Australia,
July 1–5, 2003 32. Rebersek, S.; Vodovnik, L.
Proportionally controlled functional electrical stimulation
of hand. Arch. Phys. Med. Rehabil. 1973, 54, 168–172. 33.
Popovic, D.; Popovic, M.; Stojanovic, A.; Pjanovic, A.;
Radosavljevic, S.; Vulovic, D. In Clinical Evaluation of
the Belgrade Grasping System, Proceedings of the 6th Vienna
International Workshop of FES, Vienna, Austria, 1998. 34.
Smith, B.; Peckham, P.; Keith, M.; Roscoe, D. An externally
powered, multichannel, implantable stimulator for versatile
control of paralyzed muscle. IEEE Trans. Biomech. Eng.
1987, 34, 499–508. 35. Takahashi, K.; Hoshimiya, N.;
Matsuki, H.; Handa, Y. Externally powered implantable FES
system. Jpn. J. Med. Electron. Biol. Eng. 1999, 37, 43–51.
36. Merletti, R.; Acimovic´, R.; Grobelnik, S.; Cvilak, G.
Electrophysiological orthosis for the upper extremity in
hemiplegia: Feasibility study. Arch. Phys. Med. Rehabil.
1975, 56 (12), 507–513. 37. Popovic, D.; Stojanovic, A.;
Pjanovic, A.; Radosavljevic, S.; Popovic, M.; Jovic, S.;
Vulovic, D. Clinicl evaluation of the Bionic Glove. Arch.
Phys. Med. Rehabil. 1999, 80 (3), 299–304. 38. Popovic,
M.R.; Keller, T.; Pappas, I.P.I.; Dietz, V.; Morari, M.
Surface-stimulation technology for grasping and walking
neuroprostheses. IEEE Trans. Eng. Med. Biol. Mag. 2001, 20,
82–93. 39. Boyce, W.H.; Lathem, J.E.; Hunt, L.D. Research
related to the development of an artificial electrical
stimulator for the paralyzed human bladder: A review. J.
Urol. 1964, 91 , 41–51. 40. Bors, E.; Comarr, E.
Neurological Urology: Physiology of Micturition, Its
Neurological Disorders and Sequelae; Karger Verlag, 1971.
41. Brindley, G.S.; Polkey, C.E.; Rushton, D.N.; Cardozo,
L. Sacral anterior root stimulators for bladder control in
paraplegia: The first 50 cases. J. Neurol. Neurosurg.
Psychiatry 1986, 49, 1104–1114. 42. Caldwell, K.P.S. The
electrical control of sphincter incontinence (preliminary
communication). Lancet 1963, 2, 174–175. 43. Alexander, S.
Research and clinical experience in the treatment of the
neurogenic bladder by electronic implant and prosthesis.
Paraplegia 1968, 6 (3), 183–193. 44. Rijkhoff, N.J.M.;
Wijkstra, H.; van Kerrebroeck, P.E.V.; Debruyne, F.M.J.
Urinary bladder control by electrical stimulation: Review
of electrical stimulation techniques in spinal cord injury.
Neurourol. Urodyn. 1997, 16, 39–53. 45. Schurch, B.; Rodic,
B.; Jeanmonod, D. Posterior sacral rhizotomy and intradural
anterior sacral root stimulation for treatment of the
spastic bladder in spinal cord injured patients. J. Urol.
1997, 157, 610–614. 46. Creasey, G.H. Restoration of
bladder, bowel, and sexual function. Top Spinal Cord Inj.,
Rehabil. 1999, 5 (1), 21–32. 47. Vastenholt, J.M.;
Ijzerman, M.J.; Buschman, H.P.J.; Snoek, G.J.; van der Aa,
H.E. In Seven Year Follow-Up of Sacral Anterior Root
Stimulation and Sacral Posterior Root Rhizotomy for Bladder
Control in Patients with a Spinal Cord Injury,
Complications and Quality of Life, Proceedings of the
International Functional Electrical Stimulation Society,
Cleveland, Ohio, 2001. 48. Siegel, S.W. Management of
voiding dysfunction with an implantable neuroprosthesis.
Urol. Clin. North Am. 1992, 19 (1), 163–170. 49. Lenarz, T.
Cochlear implants, selection criteria and shifting borders.
Acta-Rhino-Laryngol. Belg. 1998, 52 (3), 183–199. 50.
Gstoettner, W.; Adunka, O.; Hamzavi, J.; Baumgartner, W.D.
Rehabilitation of hearing-impaired patients with cochlear
implants, a review. Wien. Klin. Wochenschr. 2000, 112 (11),
464–472. 51. Higgins, K.M.; Chen, J.M.; Nedzelski, J.M.;
Shipp, D.B.; McIlmoyl, L.D. A matched-pair comparison of
two cochlear implant systems. J. Otolaryngol. 2002, 31 (2),
97–105. 52. Popovic, M.B.; Popovic, D.B.; Sinkjaer, T.;
Stefanovic, A.; Schwirtlich, L. Restitution of reaching and
grasping promoted by functional electrical therapy. Artif.
Organs 2002, 26 (3), 271–275. 53. Thrasher, T.A.; Popovic,
M.R. In FES-Assisted Walking for Rehabilitation of
Incomplete Spinal Cord Injury, From Technology to Market:
Bridging the Gap, Proceedings of the International
Functional Electrical Stimulation Society, Maroochydore,
Australia, July 1–5, 2003. N
Neutrophil-Biomaterial Interactions

1. Edwards, S.W. Biochemistry and Physiology of the


Neutrophil; Cambridge University Press: Cambridge, 1994.

2. Videm, V. Endpoint-attached heparin blocks neutrophil


sticking and spreading. Biomaterials 2004, 25, 43–51.

3. Sundaram, S.; Lim, F.; Cooper, S.L.; Colman, R.W. Role


of leucocytes in coagulation induced by artificial surfaces:
investigation of expression of Mac-1, granulocyte elastase
release and leucocyte adhesion on modified polyurethanes.
Biomaterials 1996, 17, 1041–1047.

4. Tomczok, J.; Sliwa-Tomczok, W.; Klein, C.L.; van Kooten,


T.G.; Kirkpatrick, C.J. Biomaterial-induced alterations of
human neutrophils under fluid shear stress: Scanning
electron microscopical study in vitro. Biomaterials 1996,
18, 1359–1367.

5. Nimeri, G.; O¨hman, L.; Elwing, H.; Wettero¨, J.;


Bengtsson, T. The influence of plasma proteins and platelets
on oxygen radical production and F-actin distribution in
neutrophils adhering to polymer surfaces. Biomaterials
2002, 23, 1785–1795. 6. Eriksson, C.; Lausmaa, J.; Nygren,
H. Interactions between human whole blood and modified TiO 2
-surfaces: Influence of surface topography and oxide
thickness on leukocyte adhesion and activation.
Biomaterials 2001, 22, 1987–1996. 7. Chang, S.; Popowich,
Y.; Greco, R.S.; Haimovich, B. Neutrophil survival on
biomaterials is determined by surface topography. J. Vasc.
Surg. 2003, 37, 1082–1090. 8. Kao, W.J. Evaluation of
leukocyte adhesion on polyurethanes: The effects of shear
stress and blood proteins. Biomaterials 2000, 21,
2295–2303. 9. Shive, M.S.; Hasan, S.M.; Anderson, J.M.
Shear stress effects on bacterial adhesion, leukocyte
adhesion, and leukocyte oxidative capacity on a
polyurethane. J. Biomed. Mater. Res. 1999, 46, 511–519. 10.
Videm, V. Heparin in clinical doses ‘‘primes’’ granulocytes
to subsequent activation as measured by myeloperoxidase
release. Scand. J. Immunol. 1996, 43, 385–390. 11. Tan, J.;
Saltzman, W.M. Topographical control of human neutrophil
motility on micropatterned materials with various surface
chemistry. Biomaterials 2002, 23, 3215–3225. 12.
Borregaard, N.; Cowland, J.B. Granules of the human
neutrophilic polymorphonuclear leukocyte. Blood 1997, 89,
3503–3521. 13. A˚sberg, A.E.; Videm, V. Neutrophil
dysfunction after biomaterial contact in an in vitro model
of cardiopulmonary bypass. Eur. J. Cardiothorac. Surg.
2006, 30, 744–748. 14. De La Cruz, C.; Haimovich, B.;
Greco, R.S. Immobilized IgG and fibrinogen differentially
affect the cytoskeletal organization and bactericidal
function of adherent neutrophils. J. Surg. Res. 1998, 80,
28–34. 15. Zhou, Y.; Doerschuk, C.M.; Anderson, J.M.;
Marchant, R.E. Biomaterial surface-dependent neutrophil
mobility. J. Biomed. Mater. Res. 2004, 69A, 611–620. 16.
Kaplan, S.S; Basford, R.E.; Jeong, M.H.; Simmons, R.L.
Biomaterial–neutrophil interactions: Dysregulation of
oxidative functions of fresh neutrophils induced by prior
neutrophil–biomaterial interaction. J. Biomed. Mater. Res.
1996, 30, 67–75. 17. Chello, M.; Mastroroberto, P.;
Auirino, A.; Cuda, G.; Perticone, F.; Cirillo, F.; Covino,
E. Inhibition of neutrophil apoptosis after coronary bypass
operation with cardiopulmonary bypass. Ann. Thorac. Surg.
2002, 73, 123–130. 18. Nahar, N.; Shan, H.; Siu, J.;
Colvin, R.; Bhaskaran, M.; Ranjan, R.; Wagner, J.D.;
Singhal, P.C. Dialysis membrane-induced neutrophil
apoptosis is mediated through free radicals. Clin. Nephrol.
2001, 56, 52–59. 19. Gourlay, T.; Stefanou, D.C.;
Asimakopuolos, G.; Taylor, K.M. The effect of circuit
surface area on CD11b(mac-1) expression in a rat
recirculation model. Artif. Org. 2001, 25, 475–479. 20.
Gourlay, T.; Samartzis, I.; Taylor, K.M. The effect of
haemodilution on blood–biomaterial contact-mediated CD11b
expression on neutrophils: Ex vivo studies. Perfusion 2003,
18, 87–93.
Nickel and Nickel Alloys

28. Trepanier, C.; Tabrizian, M.; Yahia, L’H.; Bilodeau,


L.; Piron, D. Effect of modification of oxide layer on NiTi
stent corrosion resistance. J. Biomed. Mater. Res. 1998,
43, 433–440.

29. Shabalovskaya, S.A. On the nature of the


biocompatibility and on medical applications of NiTi shape
memory and superelastic alloys. Biomed. Mater. Eng. 1996,
6, 267–289.

30. Wever, D.J.; Veldhuizen, A.G.; de Vries, J.; Busscher,


H.J.; Uges, D.R.A.; van Horn, J.R. Electrochemical and
surface characterisation of nickel–titanium alloy.
Biomaterials 1998, 19, 761–769.

31. Shih, C.-C.; Lin, S.-J.; Chung, K.-h.; Chen, Y.-L.; Su,
Y.-Y. Increased corrosion resistance of stent materials by
converting current surface film of polycrystalline oxide
into amorphous oxide. J. Biomed. Mater. Res. 2000, 52,
323–332.

32. Thierry, B.; Tabrizian, M.; Savadogo, O.; Yahia, L’H.


Effects of sterilization processes on NiTi alloy: Surface
characterisation. J. Biomed. Mater. Res. 2000, 49, 88– 98.

33. Thierry, B.; Tabrizian, M.; Trepanier, C.; Savadogo,


O.; Yahia, L’H. Effect of surface treatment and
sterilization processes on the corrosion behaviour of NiTi
shape memory alloy. J. Biomed. Mater. Res. 2000, 51,
685–693.

34. Li, C.; Wu, K.H. Corrosion Behaviour of NiTi Shape


Memory Alloy in Artificial Seawater. In Proceedings of the
First International Conference on Shape Memory and
Superelastic Technologies; Pelton, A.R., Hodgson, D.,
Duerig, T., Eds.; Pacific Grove: California, 1994; 227– 232.

35. Bu¨dinger, L.; Hertl, M. Immunological mechanisms in


hypersensitivity reactions to metal ions: An overview.
Allergy 2000, 55, 108–115.

36. Shih, C.-C.; Shih, C.-M.; Chen, Y.-L.; Su, Y.-Y.; Shih,
J.-S.; Kwok, C.-F.; Lin, S.-J. Growth inhibition of
cultured smooth muscle cells by corrosion products of 316L
stainless steel wire. J. Biomed. Mater. Res. 2001, 57,
200–207.

37. Edwards, D.L.; Wataha, J.C.; Hanks, C.T. Uptake and


reversibility of uptake of nickel by human macrophages. J.
Oral Rehab. 1998, 25, 2–7. 38. Wataha, J.C.; Lockwood,
P.E.; Schedle, A.; Noda, M.; Bouillaguet, S. Ag, Cu, Hg and
Ni ions alter the metabolism of human monocytes during
extended low-dose exposures. J. Oral Rehab. 2002, 29,
133–139. 39. Wataha, J.C.; Lockwood, P.E. Release of
elements from dental casting alloys into cell-culture
medium over 10 months. Dent. Mater. 1998, 14, 158–163. 40.
Wataha, J.C.; Sun, Z.L.; Hanks, C.T.; Fang, D.N. Effect of
Ni ions on expression of intercellular adhesion molecule 1
by endothelial cell. J. Biomed. Mater. Res. 1997, 36,
145–151. 41. Takeshita, F.; Takata, H.; Ayukawa, Y.;
Suetsugu, T. Histomorphometric analysis of the response of
rat tibiae to shape memory alloy (nitinol). Biomaterials
1997, 18, 21–25. 42. Ryha¨ nen, J.; Niemi, E.; Serlo, W.;
Niemela¨, E.; Sandvik, P.; Pernu, H.; Salo, T.
Biocompatibility of nickel– titanium shape memory metal and
its corrosion behaviour in human cell cultures. J. Biomed.
Mater. Res. 1997, 35, 451–457. 43. Wever, D.J.; Veldhuizen,
A.G.; Sanders, M.M.; Schakenraad, J.M.; van Horn, J.R.
Cytotoxic, allergic and genotoxic activity of a
nickel–titanium alloy. Biomaterials 1997, 18, 1115–1120.
44. Assad, M.; Yahia, L.H.; Rivard, C.H.; Lemieux, N. In
vitro biocompatibility assessment of a nickel–titanium
alloy using electron microscopy in situ. J. Biomed. Mater.
Res. 1998, 41, 154–161. 45. Ryha¨nen, J.; Kallioinen, M.;
Tuukkanen, J.; Junila, J.; Niemela¨, E.; Sandvik, P.;
Serlo, W. In vivo biocompatibility evaluation of
nickel–titanium shape memory metal alloy: Muscle and
perineural tissue responses and encapsule membrane
thickness. J. Biomed. Mater. Res. 1998, 41, 481–488. 46.
Ryha¨nen, J.; Kallioinen, M.; Tuukkanen, J.; Lehenkari, P.;
Junila, J.; Niemela¨, E.; Sandvik, P.; Serlo, W. Bone
modeling and cell–material interface responses induced by
nickel–titanium shape memory alloy after periosteal
implantation. Biomaterials 1999, 20, 1309–1317. 47.
Trepanier, C.; Leung, T.K.; Tabrizian, M.; Yahia, L’H.;
Bienvenu, J.-G.; Tanguay, J.-F.; Piron, D.L.; Bildeau, L.
Preliminary investigation of the effects of surface
treatments on biological response to shape memory NiTi
stents. J. Biomed. Mater. Res. (Appl. Biomater.) 1999, 48,
165–171.
Nitric Oxide

40. Abman, S.H.; Griebel, J.L.; Parker, D.K.; Schmidt,


J.M.; Swanton, D.; Kinsella, J.P. Acute effects of inhaled
nitric oxide in children with severe hypoxemic respiratory
failure. J. Pediatr. 1994, 124 (6), 881–888.

41. Benzing, A.; Geiger, K. Inhaled nitric oxide lowers


pulmonary capillary pressure and changes longitudinal
distribution of pulmonary vascular resistance in patients
with acute lung injury. Acta Anaesthesiol. Scand. 1994, 38
(7), 640–645.

42. Benzing, A.; Bra¨utigam, P.; Geiger, K.; Loop, T.;


Beyer, U.; Moser, E. Inhaled nitric oxide reduces pulmonary
transvascular albumin flux in patients with acute lung
injury. Anesthesiology 1995, 83 (6), 1153–1161.

43. Day, R.W.; Guarin, M.; Lynch, J.M.; Vernon, D.D.; Dean,
J.M. Inhaled nitric oxide in children with severe lung
disease: Results of acute and prolonged therapy with two
concentrations. Crit. Care Med. 1996, 24 (2), 215–221.

44. Head, C.A.; Brugnara, C.; Martinez-Ruiz, R.; Kacmarek,


R.M.; Bridges, K.R.; Kuter, D.; Bloch, K.D.; Zapol, W.M.
Low concentrations of nitric oxide increase oxygen affinity
of sickle erythrocytes in vitro and in vivo. J. Clin.
Invest. 1997, 100 (5), 1193–1198.

45. Berger, J.I.; Gibson, R.L.; Redding, G.L.; Standaert,


T.A.; Clarke, W.R.; Truog, W.E. Effects of inhaled nitric
oxide during group B streptococcal sepsis in piglets. Am.
Rev. Respir. Dis. 1993, 147 (5), 1080–1086.

46. Dahm, P.L.; Blomquist, S.; DeRobertis, E.; Jonson, B.;


Myhre, E.; Svantesson, C.; Thorne, J. Effects of NO
inhalation on pulmonary leukocyte sequestration and blood
volume in porcine endotoxaemia. Intensive Care Med. 2000,
26 (3), 336–343.

47. Emil, S.; Berkeland, J.; Kosi, M.; Atkinson, J. Inhaled


nitric oxide prevents experimental platelet activating
factor-induced shock. Arch. Surg. 1996, 131 (8), 855–859.

48. Gessler, P.; Nebe, T.; Birle, A.; Mueller, W.; Kachel,
W. A new side effect of inhaled nitric oxide in neonates
and infants with pulmonary hypertension: Functional
impairment of neutrophil respiratory burst. Intensive Care.
Med. 1996, 22 (3), 252–258.
49. Shimpo, H.; Mitani, Y.; Tanaka, J.; Mizumoto, T.;
Onoda, K.; Tani, K.; Yuasa, H.; Yada, I.; Maruyama, K.
Inhaled low-dose nitric oxide for post-operative care in
patients with congenital heart defects. Artif. Organs 1997,
21, 10–13.

50. Belvisi, M.G.; Ward, J.K.; Mitchell, J.A.; Barnes, P.J.


Nitric oxide as a neutrotransmitter in human airways. Ach.
Int. Pharmacodyn. Ther. 1995, 329 (1), 97–110.

51. Dupuy, P.M.; Shore, S.A.; Drazen, J.M.; Frostell, C.;


Hill, W.A.; Zapol, W.M. Brochodilator effect of inhaled
nitric oxide in guinea pigs. J. Clin. Invest. 1992, 90 (2),
421–428.

52. Ho¨gman, M.; Frostell, C.G.; Hedenstro¨ m, H.;


Hedenstierna, G. Inhalation of nitric oxide modulates adult
human bronchial tone. Am. Rev. Respir. Dis. 1993, 148 (6),
1474–1478. 53. Troncy, E.; Francoeur, M.; Blaise, G.
Inhaled nitric oxide: Clinical applications, indications,
and toxicology. Can. J. Anaesth. 1997, 44 (9), 973–988. 54.
Wendel, H.P.; Ziemer, G. Coating-techniques to improve the
hemocompatibility of artificial devices used for
extracorporeal circulation. Eur. J. Cardiothorac. Surg.
1999, 16 (3), 342–350. 55. Hayashi, Y.; Sawa, Y.;
Nishimura, M.; Chang, J.C.; Amemiya, A.; Kagisaki, K.;
Taketani, S.; Yamaguchi, T.; Hirata, N.; Ohtake, S.;
Matsuda, H. Nitric oxide gas infusion to the oxygenator
enhances the biocompatibility of heparin coated
extracorporeal bypass circuits. ASAIO J. 1998, 44 (5),
M456–461. 56. Ramamurthi, A.; Lewis, R.S. Nitric oxide
inhibition of platelet deposition on biomaterials. Biomed.
Sci. Instrum. 1999, 35, 333–338. 57. Hayashi, Y.; Sawa, Y.;
Hirata, N.; Nishimura, M.; Ueda, H.; Naka, Y.; Yamaguchi,
T.; Ohtake, S.; Matsuda, H. Improvement of bypass circuit
biocompatibility: Comparison and combination of
heparin-coated circuit and nitric oxide gas infusion. J.
Card. Surg. 2002, 17 (6), 477–484. 58. Mowery, K.A.;
Schoenfisch, M.H.; Saavedra, J.E.; Keefer, L.K.; Meyerhoff,
M.E. Preparation and characterization of hydrophobic
polymeric films that are thromboresistant via nitric oxide
release. Biomaterials 2000, 21 (1), 9–21. 59. Kaul, S.;
Cercek, B.; Rengstrom, J.; Xu, X.P.; Molloy, M.D.;
Dimayuga, P.; Parikh, A.K.; Fishbein, M.C.; Nilsson, J.;
Rajavashisth, T.B.; Shah, P.K. Polymericbased perivascular
delivery of a nitric oxide donor inhibits intimal
thickening after balloon denudation arterial injury: Role
of nuclear factor-kappaB. J. Am. Coll. Cardiol. 2000, 35
(2), 493–501. 60. Buergler, J.M.; Tio, F.O.; Schulz, D.G.;
Khan, M.M.; Mazur, W.; French, B.A.; Raizner, A.E.; Ali,
N.M. Use of nitric-oxide-eluting polymer-coated coronary
stents for prevention of restenosis in pigs. Coron. Artery
Dis. 2000, 11 (4), 351–357. 61. Annich, G.M.; Meinhardt,
J.P.; Mowery, K.A.; Ashton, B.A.; Merz, S.I.; Hirschl,
R.B.; Meyerhoff, M.E.; Bartlett, R.H. Reduced platelet
activation and thrombosis in extracorporeal circuits coated
with nitric oxide release polymers. Crit. Care Med. 2000,
28 (4), 915–920. 62. Bohl, K.S.; West, J.L. Nitric
oxide-generating polymers reduce platelet adhesion and
smooth muscle cell proliferation. Biomaterials 2000, 21
(22), 2273–2278. 63. Zhang, H.; Annich, G.M.; Miskulin, J.;
Osterholzer, K.; Merz, S.I.; Bartlett, R.H.; Meyerhoff,
M.E. Nitric oxide releasing silicone rubbers with improved
blood compatibility: Preparation, characterization, and in
vivo evaluation. Biomaterials 2002, 23, 1485–1494. 64.
Frost, M.C.; Rudich, S.M.; Zhang, H.; Maraschio, M.A.;
Meyerhoff, M.E. In vivo biocompatibility and analytical
performance of intravascular amperometric oxygen sensors
prepared with improved nitric oxidereleasing silicone
rubber coating. Anal. Chem. 2002, 74 (23), 5942–5947. 65.
Lee, W.K.; Kobayashi, J.; Ooya, T.; Park, K.D.; Yui, N.
Synthesis and characterization of nitric oxide generative
polyrotaxane. J. Biomater. Sci., Polym. Ed. 2002, 13 (10),
1153–1161. 66. Scott-Burden, T.; Tock, C.L.; Schwarz, J.J.;
Casscells, S.W.; Engler, D.A. Genetically engineered smooth
muscle cells as linings to improve the biocompatibility of
cardiovascular prostheses. Circulation 1996, 94 (9 Suppl),
II235–II238. 67. Tock, C.L.; Bosley, J.P.; Parnis, S.M.;
Clubb, F.J., Jr.; Macris, M.P.; Frazier, O.H.;
Scott-Burden, T. A genetically engineered, nonthrombogenic
cellular lining for LVADs: In vitro preconditioning before
in vivo implantation. ASAIO J. 1999, 45 (3), 172–177. 68.
Dulak, J.; Jozkowicz, A.; Guevara, I.; Dembinska-Kiec, A.
Gene transfer of vascular endothelial growth factor and
endothelial nitric oxide synthase—Implications for gene
therapy in cardiovascular diseases. Pol. J. Pharmacol.
1999, 51 (3), 233–241. 69. Frey, A.; Schneider-Rasp, S.;
Marienfeld, U.; Yu, J.C.; Paul, M.; Poller, W.; Schmidt,
H.H. Biochemical and functional characterization of nitric
oxide synthase III gene transfer using a
replication-deficient adenoviral vector. Biochem. Pharmacol.
1999, 58 (7), 1155–1166. 70. Tanner, F.C.; Meier, P.;
Greutert, H.; Champion, C.; Nabel, E.G.; Luscher, T.F.
Nitric oxide modulates expression of cell cycle regulatory
proteins: A cytostatic strategy for inhibition of human
vascular smooth muscle cell proliferation. Circulation
2000, 101 (16), 1982–1989. 71. Muhs, A.; Heublein, B.;
Schletter, J.; Herrmann, A.; Rudiger, M.; Sturm, M.; Grust,
A.; Malms, J.; Schrader, J.; von der Leyen, H.E.
Preclinical evaluation of inducible nitric oxide synthase
lipoplex gene therapy for inhibition of stent—Induced
vascular neointimal lesion formation. Hum. Gene Ther. 2003,
14 (4), 375–383. 72. Ohata, T.; Sawa, Y.; Takagi, M.;
Inoue, T.; Yoshida, T.; Kogaki, S.; Matsuda, H. hybrid
artificial lung with interleukin-10 and endothelial
constitutive nitric oxide synthase gene-transfected
endothelial cells attenuates inflammatory reactions induced
by cardiopulmonary bypass. Circulation 1998, 98 (10 suppl),
II269–II274. 73. Kalra, M.; Jost, C.J.; Severson, S.R.;
Miller, V.M. Adventitial versus intimal liposome-mediated
ex vivo transfection of canine saphenous vein grafts with
endothelial nitric oxide synthase gene. J. Vasc. Surg.
2000, 32 (6), 1190–1200. 74. Campbell, A.I.; Kuliszewski,
M.A.; Stewart, D.J. Cellbased gene transfer to the
pulmonary vasculature: Endothelial nitric oxide synthase
overexpression inhibits monocrotaline-induced pulmonary
hypertension. Am. J. Respir. Cell Mol. Biol. 1999, 21 (5),
567–575. 75. Tirney, S.; Mattes, C.E.; Yoshimura, N.;
Yokayama, T.; Ozawa, H.; Tzeng, E.; Birder, L.A.; Kanai,
A.J.; Huard, J.; de Groat, W.C.; Chancellor, M.B. Nitric
oxide synthase gene therapy for erectile dysfunction:
Comparison of plasmid, adenovirus, and adenovirustransduced
myoblast vectors. Mol. Urol. 2001, 5 (1), 37–43. 76.
Chancellor, M.B.; Tirney, S.; Mattes, C.E.; Tzeng, E.;
Birder, L.A.; Kanai, A.J.; de Groat, W.C.; Huard, J.;
Yoshimura, N. Nitric oxide synthase gene transfer for
erectile dysfunction in a rat model. BJU Int. 2003, 91 (7),
691–696. 77. Bivalacqua, T.J.; Usta, M.F.; Champion, H.C.;
Adams, D.; Namara, D.B.; Abdel-Mageed, A.B.; Kadowitz,
P.J.; Hellstrom, W.J. Gene transfer of endothelial nitric
oxide synthase partially restores nitric oxide synthesis
and erectile function in streptozotocin diabetic rats. J.
Urol. 2003 , 169 (5), 1911–1917. N
Nuclear Magnetic Resonance Spectroscopy

36. Frahm, J.; Michaelis, T.; Merboldt, K.D.; Hanicke, W.;


Gyngell, M.L.; Bruhn, H. On the N-acetyl methyl resonance
in localized 1 H NMR spectra of the human brain in vivo.
NMR Biomed. 1991, 4, 201–204.

37. Birken, D.L.; Oldendorf, W.H. N-acetylL-aspartic acid:


A literature review of of a compound prominent in 1 H NMR
spectroscopic studies of brain. Neurosci. Behav. Rev. 1989,
13, 23–31.

38. Tallan, H.H. Studies on the distibution of


N-acetylLaspartic acid in brain. J. Biol. Chem. 1957, 224,
41–45.

39. Howe, F.A.; Maxwell, R.J.; Saunders, D.E.; Brown, M.M.;


Griffiths, J.R. Proton spectroscopy in vivo. Magn. Reson. Q.
1993, 9, 31–59.

40. Guimaraes, A.R.; Schwartz, P.; Prakash, M.R.; Carr,


C.A.; Berger, U.V.; Jenkins, B.G.; Coyle, J.T.; Gonzalez,
R.G. Quantitative in vivo 1 H nuclear magnetic resonance
spectroscopic imaging of neuronal loss in rat brain.
Neuroscience 1995, 69, 1095–1101.

41. Strauss, I.; Williamson, J.M.; Bertram, E.H.; Lothman,


E.W.; Fernandez, E.J. Histological and 1 H magnetic
resonance spectroscopic imaging analysis of quinolinic
acid-induced damage to the rat striatum. Magn. Reson. Med.
1997, 37, 24–33.

42. Simmons, M.L.; Frondoza, C.G.; Coyle, J.T.


Immunocytochemical localization of N-acetyl-aspartate with
monoclonal antibodies. Neuroscience 1991, 45, 37–45.

43. Alger, J.R.; Frank, J.A.; Bizzi, A.; Fulham, M.J.;


DeSouza, B.X.; Duhaney, M.O.; Inscoe, S.W.; Black, J.L.;
van Zijl, P.C.M.; Moonen, C.T.W.; Dichiro, G. Metabolism of
human gliomas: Assessment with H-1MR spectroscopy and F-18
fluorodeoxyglucose PET (see comments). Radiology 1990, 177,
633–641.

44. Arnold, D.L.; Riess, G.T.; Matthews, P.M.; Francis,


G.S.; Collins, D.L.; Wolfson, C.; Antel, J.P. Use of proton
magnetic resonance spectroscopy for monitoring disease
progression in multiple sclerosis. Ann. Neurol. 1994, 36,
76–82.

45. Davie, C.A.; Hawkins, C.P.; Barker, G.J.; Brennan, A.;


Tofts, P.S.; Miller, D.H.; McDonald, W.I. Detection of
myelin breakdown products by proton magnetic resonance
spectroscopy. Lancet 1993, 341, 630–631.

46. Barker, P.B.; Gillard, J.H.; van Zijl, P.C.; Soher,


B.J.; Hanley, D.F.; Agildere, A.M.; Oppenheimer, S.M.;
Bryan, R.N. Acute stroke: Evaluation with serial proton MR
spectroscopic imaging. Radiology 1994, 192, 723–732.

47. Petroff, O.A.; Graham, G.D.; Blamire, A.M.; al-Rayess,


M.; Rothman, D.L.; Fayad, P.B.; Brass, L.M.; Shulman, R.G.;
Prichard, J.W. Spectroscopic imaging of stroke in humans:
Histopathology correlates of spectral changes. Neurology
1992, 42, 1349–1354.

48. Cendes, F.; Andermann, F.; Preul, M.C.; Arnold, D.L.


Lateralization of temporal lobe epilepsy based on regional
metabolic abnormalities in proton magnetic resonance
spectroscopic images. Ann. Neurol. 1994, 35, 211–216.

49. Hugg, J.W.; Laxer, K.D.; Matson, G.B.; Maudsley, A.A.;


Weiner, M.W. Neuron loss localizes human temporal lobe
epilepsy by in vivo proton magnetic resonance spectroscopic
imaging. Ann. Neurol. 1993, 34, 788–794. 50. Gill, S.S.;
Small, R.K.; Thomas, D.G.; Patel, P.; Porteous, R.; Van
Bruggen, N.; Gadian, D.G.; Kauppinen, R.A.; Williams, S.R.
Brain metabolites as 1 H NMR markers of neuronal and glial
disorders. NMR Biomed. 1989, 2, 196–200. 51. Gill, S.S.;
Thomas, D.G.T.; Van Bruggen, N.; Gadian, D.G.; Peden, C.J.;
Bell, J.D.; Cox, I.J.; Menon, D.K.; Iles, R.A.; Bryant,
D.J.; Coutts, G.A. Proton MR spectroscopy of intracranial
tumours: In vivo and in vitro studies. J. Comput. Assist.
Tomogr. 1990, 14, 497–504. 52. Kreis, R.; Ross, B.D.;
Farrow, N.A.; Ackerman, Z. Metabolic disorders of the brain
in chronic hepatic encephalopathy detected with H-1MR
spectroscopy. Radiology 1992, 182, 19–27. 53. Stoll, A.L.;
Renshaw, P.F.; De Micheli, E.; Wurtman, R.; Pillay, S.S.;
Cohen, B.M. Choline ingestion increases the resonance of
choline-containing compounds in human brain: An in vivo
proton magnetic resonance study. Biol. Psychiatry 1995, 37,
170–174. 54. Urenjak, J.; Williams, S.R.; Gadian, D.G.;
Noble, M. Proton nuclear magnetic resonance spectroscopy
unambiguously identifies different neural cell types. J.
Neurosci. 1993, 13, 981–989. 55. Mathews, P.M.; Andermann,
F.; Silver, K.; Karpati, G.; Arnold, D.L. Proton MR
spectroscopic characterization of differences in regional
brain metabolic abnormalities in mitochondrial
encephalomyopathies. Neurology 1993, 43, 2484–2490. 56.
Prichard, J.; Rothman, D.; Novotny, E.; Petroff, O.;
Kuwabara, T.; Avison, M.; Howseman, A.; Hanstock, C.;
Shulman, R. Lactate rise detected by 1 H NMR in human
visual cortex during physiologic stimulation. Proc. Natl.
Acad. Sci. U. S. A. 1991, 88, 5829–5831. 57. Merboldt,
K.D.; Bruhn, H.; Hanicke, W.; Michaelis, T.; Frahm, J.
Decrease of glucose in the human visual cortex during
photic stimulation. Magn. Reson. Med. 1992, 25, 187–194.
58. Flogel, U.; Willker, W.; Leibfritz, D. Regulation of
intracellular pH in neuronal and glial tumour cells,
studied by multinuclear NMR spectroscopy. NMR Biomed. 1994,
7, 157–166. 59. Ross, B.D.; Jacobson, S.; Villamil, F.;
Korula, J.; Kreis, R.; Ernst, T.; Shonk, T.; Moats, R.A.
Subclinical hepatic encephalopathy: Proton MR spectroscopic
abnormalities. Radiology 1994, 193, 457–463. 60. Shonk,
T.K.; Moats, R.A.; Gifford, P.; Michaelis, T.; Mandigo,
J.C.; Izumi, J.; Ross, B.D. Probable Alzheimer disease:
Diagnosis with proton MR spectroscopy. Radiology 1995, 195,
65–72. 61. Kruse, B.; Hanefeld, F.; Christen, H.J.; Bruhn,
H.; Michaelis, T.; Hanicke, W.; Frahm, J. Alterations of
brain metabolites in metachromatic leukodystrophy as
detected by localized proton magnetic resonance
spectroscopy in vivo. J. Neurol. 1993, 241, 68–74. 62.
Rothman, D.L.; Petroff, O.A.; Behar, K.L.; Mattson, R.H.
Localized 1 H NMR measurements of gamma-aminobutyric acid
in human brain in vivo. Proc. Natl. Acad. Sci. U. S. A.
1993, 90, 5662–5666. 63. Gruetter, R.; Garwood, M.;
Ugurbil, K.; Seaquist, E.R. Observation of resolved glucose
signals in 1 H NMR spectra of the human brain at 4 Tesla.
Magn. Reson. Med. 1996, 36, 1–6. 64. Michaelis, T.; Helms,
G.; Merboldt, K.D.; Hanicke, W.; Bruhn, H.; Frahm, J.
Identification of Scyllo-inositol in proton NMR spectra of
human brain in vivo. NMR Biomed. 1993, 6, 105–109. 65. van
Zijl, P.C.; Moonen, C.T. In situ changes in purine
nucleotide and N-acetyl concentrations upon inducing global
ischemia in cat brain. Magn. Reson. Med. 1993, 29, 381–385.
66. Hwang, J.H.; Graham, G.D.; Behar, K.L.; Alger, J.R.;
Prichard, J.W.; Rothman, D.L. Short echo time proton
magnetic resonance spectroscopic imaging of macromolecule
and metabolite signal intensities in the human brain. Magn.
Reson. Med. 1996, 35, 633–639. 67. Kvistad, K.A.; Bakken,
I.J.; Gribbestad, I.S.; Ehrnholm, B.; Lundgren, S.; Fjosne,
H.E.; Haraldseth, O. Characterization of neoplastic and
normal breast tissues with 1 H MR spectroscopy. JMRI 1999,
10, 159–164. 68. Bhakoo, K.K.; Pearce, D. In vitro
expression of Nacetyl aspartate by oligodendrocytes:
Implications for proton magnetic resonance spectroscopy
signal in vivo. J. Neurochem. 2000, 74 (1), 254–262. 69.
Barker, P.B. N-acetyl aspartate—A neuronal marker? Ann.
Neurol. 2001, 49, 423–424. 70. Jacobs, M.A.; Horska, A.;
van Zijl, P.C.M.; Barker, P.B. Quantitative proton MR
spectroscopic imaging of normal human cerebellum and brain
stem. Magn. Reson. Med. 2001, 46, 699–705. N
Ocular Implants

7. Werblin, T.P.; Peiffer, R.L.; Binder, P.S.; McCarey,


B.E.; Patel, A.S. Eight years experience with permalens
intracorneal lenses in nonhuman primates. Refract. Corneal
Surg. 1992, 8, 12–22.

8. Lane, S.S.; Lindstrom, R.L. Polysulfone intracorneal


lenses. Int. Ophthalmol. Clin. 1991, 31, 38–46.

9. Hoffman, F.; Kruse, H.; Schuler, A. Mechanical methods


in refractive corneal surgery. Curr. Opin. Ophthalmol.
1993, 4, 84–90.

10. Lehrer, M.S.; Sun, T.T.; Lavker, R.M. Strategies of


epithelial repair: Modulation of stem cell and transit
amplifying cell proliferation. J. Cell. Sci. 1998, 111,
2867–2875.

11. Catroveijo, R.; Cardona, H.; DeVoe, A.G. Present state


of prosthokeratoplasty. Am. J. Ophthalmol. 1969, 68,
613–625.

12. Cardona, H. Mushroom transcorneal keratoprosthesis. Am.


J. Ophthalmol. 1969, 68, 604–612.

13. Girard, L.J. Keratoprosthesis. Cornea 1983, 2, 207–224.

14. Doane, M.G.; Dohlman, C.H.; Bearse, G. Fabrication of a


keratoprosthesis. Cornea 1996, 15, 179–184.

15. Barber, J.C.; Feaster, F.T.; Prior, D.J. The acceptance


of a vitreous carbon alloplastic material, Proplast, into
the rabbit eye. Investig. Ophthalmol. Vis. Sci. 1980, 19,
182–190.

16. Legeais, J.M.; Rossi, C.; Renard, G.; Savoldelli, M.;


D’hermies, F.; Pouliquen, Y. A new fluorocarbon for
keratoprosthesis. Cornea 1992, 11, 538–545.

17. Chirila, T.V. Artificial cornea with a porous polymeric


skirt. Trends Polym. Sci. 1998, 5, 346–348.

18. Chirila, T.V. An overview of the development of


artificial corneas with porous skirts and the use of PHEMA
for such an application. Biomaterials 2001, 22, 3311– 3317.

19. Strampelli, B. Osteo-odonto-keratoprosthesis. Ann.


Ottalmol. Clin. Ocul. 1963, 89, 1039–1044.
20. Schepens, C.L.; Acosta, F. Scleral implants: An
historical perspective. Surv. Ophthalmol. 1991, 35,
447–453.

21. Hahn, Y.S.; Lincoff, A.; Lincoff, H.; Kreissig, I.


Infection after sponge implantation for scleral bucking.
Am. J. Ophthalmol. 1979, 87, 180–185.

22. Molteno, A.C.B. New implant for drainage in glaucoma.


Animal trial. Br. J. Ophthalmol. 1969, 53, 161–168.

23. Lim, K.S.; Allan, B.D.S.; Lloyd, A.W.; Muir, A.; Khaw,
P.T. Glaucoma filtration implants: Past, present and future.
Br. J. Ophthalmol. 1998, 82, 1083–1089. 24. Lim, K.S.;
Allan, B.; Khaw, P.T.; Willis, S.; Lloyd, A.W.; Muir, A.;
Gard, P.; Faragher, R.G.A.; Olliff, C.J.; Hanlon, G.W.;
Wong, L.; Reed, S.; Denyer, S. Experimental flow studies in
glaucoma drainage device development. Br. J. Ophthalmol.
2001, 85, 1231–1236. 25. Obstbaum, A.S. Biologic
relationship between poly(methyl methacrylate) intraocular
lenses and uveal tissue. J. Cataract Refract. Surg. 1992,
18, 219–231. 26. Lloyd, A.W.; Dropcova, S.; Faragher,
R.G.A.; Gard, P.R.; Hanlon, G.W.; Mikhalovsky, S.V.;
Olliff, C.J.; Denyer, S.P. The development of in vitro
biocompatibility tests for the evaluation of intraocular
biomaterials. J. Mater. Sci., Mater. Med. 1999, 10,
621–627. 27. Lloyd, A.W.; Bowers, R.W.J.; Dropcova, S.;
Denyer, S.P.; Faragher, R.G.A.; Gard, P.R.; Hall, B.;
Hanlon, G.W.; Jones, S.A.; Muir, A.; Olliff, C.J.; Rosen,
P.H.; Riding, M. In vitro evaluation of novel biomimetic
intraocular lens materials. Investig. Ophthalmol. Vis. Sci.
1997, 38, S178. 28. Findl, O.; Kiss, B.; Petternel, V.;
Menapace, R.; Georgopoulos, M.; Rainer, G.; Drexler, W.
Intraocular lens movement caused by ciliary muscle
contraction. J. Cataract Refract. Surg. 2003, 29, 669–676.
29. Legeais, J.M.; Werner, L.; Werner, L.; Abenhaim, A.;
Renard, G. Pseudoaccommodation: BioComFold versus a
foldable silicone intraocular lens. J. Cataract Refract.
Surg. 1999, 25, 262–267. 30. Rosen, E.S. Intraocular
lenses. Curr. Opin. Ophthalmol. 1993, 4, 44–53. 31. Ravi,
N.; Chuck, R.S. Development of an injectable intraocular
lens for preservation of accommodation. Investig.
Ophthalmol. Vis. Sci. 1996, 37, 3539. 32. Capone, A., Jr.;
Aaberg, T.M. Silicone oil in vitreoretinal surgery. Curr.
Opin. Ophthalmol. 1995, 6, 33–37. 33. Arroyo, M.H.; Refojo,
M.F.; Ariaz, J.J.; Tolentino, F.I.; Cajita, V.N.; Elner,
V.M. Silicone oil as a delivery vehicle for BCNU in rabbit
proliferative vitreoretinopathy. Retina 1993, 13, 245–250.
34. Liesegang, T.J. Viscoelastic substances in
ophthalmology. Surv. Ophthalmol. 1990, 34, 268–293. 35.
Goa, K.L.; Benfield, P. Hyaluronic acid—A review of its
pharmacology and use as a surgical aid in ophthalmology,
and its therapeutic potential in joint disease and
wound-healing. Drugs 1994, 47, 536–566. 36. Ding, S. Recent
developments in ophthalmic drug delivery. Pharm. Sci.
Technol. Today 1998 , 8, 328–335.
Ocular Implants for Drug Delivery

38. Hashizoe, M.; Ogura, Y.; Takanashi, T.; Kunou, N.;


Honda, Y.; Ikada, Y. Biodegradable polymeric device for
sustained intravitreal release of ganciclovir in rabbits.
Curr. Eye Res. 1997, 16 (7), 633–639.

39. Chang, D.F.; Wong, V. Two clinical trials of an


intraocular steroid delivery system for cataract surgery.
Trans. Am. Ophthalmol. Soc. 1999, 97, 261–274. discussion
274–269.

40. Tan, D.T.; Chee, S.P.; Lim, L.; Lim, A.S. Randomized
clinical trial of a new dexamethasone delivery system
(Surodex) for treatment of post-cataract surgery
inflammation. Ophthalmology 1999, 106 (2), 223–231.

41. Tan, D.T.; Chee, S.P.; Lim, L.; Theng, J.; Van Ede, M.
Randomized clinical trial of Surodex steroid drug delivery
system for cataract surgery: Anterior versus posterior
placement of two Surodex in the eye. Ophthalmology 2001,
108 (12), 2172–2181.

42. Morita, Y.; Saino, H.; Tojo, K. Polymer blend implant


for ocular delivery of fluorometholone. Biol. Pharm. Bull.
1998, 21 (1), 72–75.

43. Rubsamen, P.E.; Davis, P.A.; Hernandez, E.; O’Grady,


G.E.; Cousins, S.W. Prevention of experimental
proliferative vitreoretinopathy with a biodegradable
intravitreal implant for the sustained release of
fluorouracil. Arch. Ophthalmol. 1994, 112 (3), 407–413.

44. Borhani, H.; Peyman, G.A.; Rahimy, M.H.; Thompson, H.


Suppression of experimental proliferative vitreoretinopathy
by sustained intraocular delivery of 5-FU. Int. Ophthalmol.
1995, 19 (1), 43–49.

45. Morales, J.; Kelleher, P.J.; Campbell, D.; Crosson,


C.E. Effects of daunomycin implants on filtering surgery
outcomes in rabbits. Curr. Eye Res. 1998, 17 (8), 844–850.

46. Uppal, P.; Jampel, H.D.; Quigley, H.A.; Leong, K.W.


Pharmacokinetics of etoposide delivery by a bioerodible
drug carrier implanted at glaucoma surgery. J. Ocul.
Pharmacol. 1994, 10 (2), 471–479.

47. Hardten, D.R.; Samuelson, T.W. Ocular toxicity of


mitomycin-C. Int. Ophthalmol. Clin. 1999, 39 (2), 79–90.
48. Rahimy, M.H.; Peyman, G.A.; Fernandes, M.L.; el-Sayed,
S.H.; Luo, Q.; Borhani, H. Effects of an intravitreal
daunomycin implant on experimental proliferative
vitreoretinopathy: Simultaneous pharmacokinetic and
pharmacodynamic evaluations. J. Ocul. Pharmacol. 1994, 10
(3), 561–570.

49. Rahimy, M.H.; Peyman, G.A.; Chin, S.Y.; Golshani, R.;


Aras, C.; Borhani, H.; Thompson, H. Polysulfone capillary
fiber for intraocular drug delivery: In vitro and in vivo
evaluations. J. Drug Target. 1994, 2 (4), 289–298.

50. Apel, A.; Oh, C.; Chiu, R.; Saville, B.; Cheng, Y.L.;
Rootman, D. A subconjunctival degradable implant for
cyclosporine delivery in corneal transplant therapy. Curr.
Eye Res. 1995, 14 (8), 659–667.

51. Hillman, J.S.; Marsters, J.B.; Broad, A. Pilocarpine


delivery by hydrophilic lens in the management of acute
glaucoma. Trans. Ophthalmol. Soc. U. K. 1975, 95 (1),
79–84.

52. Yamakawa, I.; Ishida, M.; Kato, T.; Ando, H.; Asakawa,
N. Release behavior of poly(lactic acid-coglycolic acid)
implants containing phosphorothioate oligodeoxynucleotide.
Biol. Pharm. Bull. 1997, 20 (4), 455–459. 53. Simpson,
A.E.; Gilbert, J.A.; Rudnick, D.E.; Geroski, D.H.; Aaberg,
T.M.; Edelhauser, H.F., Jr. Transscleral diffusion of
carboplatin: An in vitro and in vivo study. Arch.
Ophthalmol. 2002, 120 (8), 1069–1074. 54. Einmahl, S.;
Zignani, M.; Varesio, E.; Heller, J.; Veuthey, J.L.;
Tabatabay, C.; Gurny, R. Concomitant and controlled release
of dexamethasone and 5-fluorouracil from poly(ortho ester).
Int. J. Pharm. 1999, 185 (2), 189–198. 55. Quigley, H.A.;
Pollack, I.P.; Harbin, T.S., Jr. Pilocarpine ocuserts.
Long-term clinical trials and selected pharmacodynamics.
Arch. Ophthalmol. 1975, 93 (9), 771–775. 56. Hitchings,
R.A.; Smith, R.J. Experience with pilocarpine Ocuserts.
Trans. Ophthalmol. Soc. U. K. 1977, 97 (1), 202–205. 57.
Ashton, P.; Blandford, D.L.; Pearson, P.A.; Jaffe, G.J.;
Martin, D.F.; Nussenblatt, R.B. Review: Implants. J. Ocul.
Pharmacol. 1994, 10 (4), 691–701. 58. Peters, B.S.; Beck,
E.J.; Anderson, S.; Coleman, D.; Coker, R.; Main, J.;
Migdal, C.; Harris, J.R.; Pinching, A.J. Cytomegalovirus
infection in AIDS. Patterns of disease, response to therapy
and trends in survival. J. Infect. 1991, 23 (2), 129–137.
59. McGavin, J.K.; Goa, K.L. Ganciclovir: An update of its
use in the prevention of cytomegalovirus infection and
disease in transplant recipients. Drugs 2001, 61 (8),
1153–1183. 60. Anand, R.; Nightingale, S.D.; Fish, R.H.;
Smith, T.J.; Ashton, P. Control of cytomegalovirus
retinitis using sustained release of intraocular
ganciclovir. Arch. Ophthalmol. 1993, 111 (2), 223–227. 61.
Ashton, P.; Brown, J.D.; Pearson, P.A.; Blandford, D.L.;
Smith, T.J.; Anand, R.; Nightingale, S.D.; Sanborn, G.E.
Intravitreal ganciclovir pharmacokinetics in rabbits and
man. J. Ocul. Pharmacol. 1992, 8 (4), 343–347. 62. Smith,
T.J.; Pearson, P.A.; Blandford, D.L.; Brown, J.D.; Goins,
K.A.; Hollins, J.L.; Schmeisser, E.T.; Glavinos, P.;
Baldwin, L.B.; Ashton, P. Intravitreal sustained-release
ganciclovir. Arch. Ophthalmol. 1992, 110 (2), 255–258. 63.
Martin, D.F.; Parks, D.J.; Mellow, S.D.; Ferris, F.L.;
Walton, R.C.; Remaley, N.A.; Chew, E.Y.; Ashton, P.; Davis,
M.D.; Nussenblatt, R.B. Treatment of cytomegalovirus
retinitis with an intraocular sustained-release ganciclovir
implant. A randomized controlled clinical trial. Arch.
Ophthalmol. 1994, 112 (12), 1531–1539. 64. Martin, D.F.;
Ferris, F.L.; Parks, D.J.; Walton, R.C.; Mellow, S.D.;
Gibbs, D.; Remaley, N.A.; Ashton, P.; Davis, M.D.; Chan,
C.C.; Nussenblatt, R.B. Ganciclovir implant exchange.
Timing, surgical procedure, and complications. Arch.
Ophthalmol. 1997, 115 (11), 1389–1394. 65. Charles, N.C.;
Freisberg, L. Endophthalmitis associated with extrusion of
a ganciclovir implant. Am. J. Ophthalmol. 2002, 133 (2),
273–275. 66. Guembel, H.O.; Krieglsteiner, S.; Rosenkranz,
C.; Hattenbach, L.O.; Koch, F.H.; Ohrloff, C. Complications
after implantation of intraocular devices in patients with
cytomegalovirus retinitis. Graefe Arch. Clin. Exp.
Ophthalmol. 1999, 237 (10), 824–829. 67. Berger, A.S.;
Cheng, C.K.; Pearson, P.A.; Ashton, P.; Crooks, P.A.;
Cynkowski, T.; Cynkowska, G.; Jaffe, G.J. Intravitreal
sustained release corticosteroid-5-fluoruracil conjugate in
the treatment of experimental proliferative
vitreoretinopathy. Invest. Ophthalmol. Vis. Sci. 1996, 37
(11), 2318–2325. 68. Hainsworth, D.P.; Pearson, P.A.;
Conklin, J.D.; Ashton, P. Sustained release intravitreal
dexamethasone. J. Ocular Pharmacol. Ther. 1996, 12 (1),
57–63. 69. Jaffe, G.J.; Yang, C.H.; Guo, H.; Denny, J.P.;
Lima, C.; Ashton, P. Safety and pharmacokinetics of an
intraocular fluocinolone acetonide sustained delivery
device. Invest. Ophthalmol. Vis. Sci. 2000, 41 (11),
3569–3575. 70. Masuda, K.; Nakajima, A.; Urayama, A.;
Nakae, K.; Kogure, M.; Inaba, G. Double-masked trial of
cyclosporin versus colchicine and long-term open study of
cyclosporin in Behcet’s disease. Lancet 1989, 1 (8647),
1093–1096. 71. Gilger, B.C.; Wilkie, D.A.; Davidson, M.G.;
Allen, J.B. Use of an intravitreal sustained-release
cyclosporine delivery device for treatment of equine
recurrent uveitis. Am. J. Vet. Res. 2001, 62 (12),
1892–1896. 72. Velez, G.; Whitcup, S.M. New developments in
sustained release drug delivery for the treatment of
intraocular disease. Br. J. Ophthalmol. 1999, 83 (11),
1225–1229. 73. Yang, C.S.; Khawly, J.A.; Hainsworth, D.P.;
Chen, S.N.; Ashton, P.; Guo, H.; Jaffe, G.J. An
intravitreal sustained-release triamcinolone and
5-fluorouracil codrug in the treatment of experimental
proliferative vitreoretinopathy. Arch. Ophthalmol. 1998,
116 (1), 69–77. 74. Enyedi, L.B.; Pearson, P.A.; Ashton,
P.; Jaffe, G.J. An intravitreal device providing sustained
release of cyclosporine and dexamethasone. Curr. Eye Res.
1996, 15 (5), 549–557. 75. Zhou, T.; Lewis, H.; Foster,
R.E.; Schwendeman, S.P. Development of a multiple-drug
delivery implant for intraocular management of
proliferative vitreoretinopathy. J. Control Release. 1998,
55 (2–3), 281–295. 76. Tao, W.; Wen, R.; Goddard, M.B.;
Sherman, S.D.; O’Rourke, P.J.; Stabila, P.F.; Bell, W.J.;
Dean, B.J.; Kauper, K.A.; Budz, V.A.; Tsiaras, W.G.;
Acland, G.M.; Pearce-Kelling, S.; Laties, A.M.; Aguirre,
G.D. Encapsulated cell-based delivery of CNTF reduces
photoreceptor degeneration in animal models of retinitis
pigmentosa. Invest. Ophthalmol. Vis. Sci. 2002, 43 (10),
3292–3298. 77. Zaffaroni, A. Overview and evolution of
therapeutic systems. Ann. N. Y. Acad. Sci. 1991, 618,
405–421. 78. Michelson, J.B.; Nozik, R.A. Experimental
endophthalmitis treated with an implantable osmotic
minipump. Arch. Ophthalmol. 1979, 97 (7), 1345–1346. O
Optical Coherence Tomography

19. Jang, I.K.; MacNeill, B.D.; Yabushita, H.; DeJoseph,


D.; Kauffman, C.R.; Tearney, G.J.; Bouma, B.E. In-vivo
characterization of coronary plaques in patients with ST
elevation acute myocardial infarction using optical
coherence tomography (OCT). Circulation 2002, 106 (19),
698.

20. Saleh, B.E.A.; Teich, M.C. Foundamentals of Photonics;


John Wiley & Sons, Inc.: New York, 1991.

21. Matsumoto, H.; Hirai, A. A white-light interferometer


using a lamp source and heterodyne detection with
acousto-optic modulators. Opt. Commun. 1999, 170 (4–6),
217–220.

22. Vabre, L.; Dubois, A.; Boccara, A.C. Thermal-light


full-field optical coherence tomography. Opt. Lett. 2002, 27
(7), 530–532.

23. Morgner, U.; Kartner, F.X.; Cho, S.H.; Chen, Y.; Haus,
H.A.; Fujimoto, J.G.; Ippen, E.P.; Scheuer, V.; Angelow,
G.; Tschudi, T. Sub-two-cycle pulses from a Kerr-lens
mode-locked Ti: Sapphire laser. Opt. Lett. 1999, 24 (6),
411–413.

24. Chudoba, C.; Fujimoto, J.G.; Ippen, E.P.; Haus, H.A.;


Morgner, U.; Kartner, F.X.; Scheuer, V.; Angelow, G.;
Tschudi, T. All-solid-state Cr: Forsterite laser generating
14-fs pulses at 1.3mu m. Opt. Lett. 2001, 26 (5), 292–294.

25. Birks, T.A.; Wadsworth, W.J.; Russell, P.S.


Supercontinuum generation in tapered fibers. Opt. Lett.
2000, 25 (19), 1415–1417.

26. Ranka, J.K.; Windeler, R.S.; Stentz, A.J. Visible


continuum generation in air–silica microstructure optical
fibers with anomalous dispersion at 800 nm. Opt. Lett. 2000,
25 (1), 25–27.

27. Unterhuber, A.; Povazay, B.; Hermann, B.; Sattmann, H.;


Drexler, W.; Yakovlev, V.; Tempea, G.; Schubert, C.; Anger,
E.M.; Ahnelt, P.K.; Stur, M.; Morgan, J.E.; Cowey, A.;
Jung, G.; Le, T.; Stingl, A. Compact, lowcost Ti: Al 2 O 3
laser for in vivo ultrahigh-resolution optical coherence
tomography. Opt. Lett. 2003, 28 (11), 905–907.

28. Drexler, W.; Morgner, U.; Kartner, F.X.; Pitris, C.;


Boppart, S.A.; Li, X.D.; Ippen, E.P.; Fujimoto, J.G. In
vivo ultrahigh-resolution optical coherence tomography.
Opt. Lett. 1999, 24 (17), 1221–1223.

29. Bouma, B.E.; Tearney, G.J.; Bilinsky, I.P.; Golubovic,


B.; Fujimoto, J.G. Self-phase-modulated Kerr-lens
mode-locked Cr: Forsterite laser source for optical
coherence tomography. Opt. Lett. 1996, 21 (22), 1839–1841.

30. Kowalevicz, A.M.; Schibli, T.R.; Kartner, F.X.;


Fujimoto, J.G. Ultralow-threshold Kerr-lens mode-locked Ti:
Al2O3 laser. Opt. Lett. 2002, 27 (22), 2037–2039.

31. Hartl, I.; Li, X.D.; Chudoba, C.; Ghanta, R.K.; Ko,
T.H.; Fujimoto, J.G.; Ranka, J.K.; Windeler, R.S.
Ultrahigh-resolution optical coherence tomography using
continuum generation in an air–silica microstructure
optical fiber. Opt. Lett. 2001, 26 (9), 608–610. 32. Wang,
Y.M.; Zhao, Y.H.; Nelson, J.S.; Chen, Z.P.; Windeler, R.S.
Ultrahigh-resolution optical coherence tomography by
broadband continuum generation from a photonic crystal
fiber. Opt. Lett. 2003, 28 (3), 182–184. 33. Hitzenberger,
C.K.; Baumgartner, A.; Drexler, W.; Fercher, A.F.
Dispersion effects in partial coherence interferometry:
Implications for intraocular ranging. J. Biomed. Opt. 1999,
4 (1), 144–151. 34. Niblack, W.K.; Schenk, J.O.; Liu, B.;
Brezinski, M.E. Dispersion in a grating-based optical delay
line for optical coherence tomography. Appl. Opt. 2003, 42
(19), 4115–4118. 35. Fercher, A.F.; Hitzenberger, C.K.;
Kamp, G.; Elzaiat, S.Y. Measurement of intraocular
distances by backscattering spectral interferometry. Opt.
Commun. 1995, 117 (1–2), 43–48. 36. Hausler, G.; Lindner,
M.W. Coherent radar and spectral radar—New tools for
dermatological diagnosis. J. Biomed. Opt. 1998, 3 (1),
21–31. 37. Wojtkowski, M.; Leitgeb, R.; Kowalczyk, A.;
Bajraszewski, T.; Fercher, A.F. In vivo human retinal
imaging by Fourier domain optical coherence tomography. J.
Biomed. Opt. 2002, 7 (3), 457–463. 38. Yun, S.H.; Tearney,
G.J.; de Boer, J.F.; Iftimia, N.; Bouma, B.E. High-speed
optical frequency-domain imaging. Opt. Express 2003, 11
(22), 2953–2963. 39. Nassif, N.; Cense, B.; Park, B.H.;
Yun, S.H.; Chen, T.C.; Bouma, B.E.; Tearney, G.J.; de Boer,
J.F. In vivo human retinal imaging by ultrahigh-speed
spectral domain optical coherence tomography. Opt. Lett.
2004, 29 (5), 480–482. 40. Kwong, K.F.; Yankelevich, D.;
Chu, K.C.; Heritage, J.P.; Dienes, A. 400-Hz Mechanical
scanning optical delay-line. Opt. Lett. 1993, 18 (7),
558–560. 41. Tearney, G.J.; Bouma, B.E.; Fujimoto, J.G.
High-speed phaseand group-delay scanning with a
gratingbased phase control delay line. Opt. Lett. 1997, 22
(23), 1811–1813. 42. Rollins, A.M.; Kulkarni, M.D.;
Yazdanfar, S.; Ungarunyawee, R.; Izatt, J.A. In vivo video
rate optical coherence tomography. Opt. Express 1998, 3
(6), 219–229. 43. Treacy, E.B. Optical pulse compression
with diffraction gratings. IEEE J. Quantum Electron. 1969,
QE-5 (9), 454–458. 44. Heritage, J.P.; Weiner, A.M.;
Thurston, R.N. Picosecond pulse shape manipulation in a
grating pulse— Compressor. J. Opt. Soc. Am., A, Opt. Image
Sci. Vision 1985, 2 (13), P43. 45. Oppenheim, A.V.;
Willsky, A.S.; Nawab, S.H. Signals & Systems; Prentice
Hall: Upper Saddle River, NJ, 1997. 46. Su, C.B. Achieving
variation of the optical path length by a few millimeters
at millisecond rates for imaging of turbid media and
optical interferometry: A new technique. Opt. Lett. 1997,
22 (10), 665–667. 47. Szydlo, J.; Delachenal, N.;
Giannotti, R.; Walti, R.; Bleuler, H.; Salathe, R.P.
Air-turbine driven optical low-coherence reflectometry at
28.6-kHz scan repetition rate. Opt. Commun. 1998, 154
(1–3), 1–4. 48. Liu, X.M.; Cobb, M.J.; Li, X.D. Rapid
scanning allreflective optical delay line for real-time
optical coherence tomography. Opt. Lett. 2004, 29 (1),
80–82. 49. Chen, N.G.; Zhu, Q. Rotary mirror array for high
speed optical coherence tomography. Opt. Lett. 2002, 27
(8), 607–609. 50. Hsiung, P.L.; Li, X.D.; Chudoba, C.;
Hartl, I.; Ko, T.H.; Fujimoto, J.G. High-speed path-length
scanning with a multiple-pass cavity delay line. Appl. Opt.
2003, 42 (4), 640–648. 51. Swanson, E.A.; Izatt, J.A.; Hee,
M.R.; Huang, D.; Lin, C.P.; Schuman, J.S.; Puliafito, C.A.;
Fujimoto, J.G. In-vivo retinal imaging by optical coherence
tomography. Opt. Lett. 1993, 18 (21), 1864–1866. 52.
Schmitt, J.M.; Yadlowsky, M.J.; Bonner, R.F. Subsurface
imaging of living skin with optical coherence microscopy.
Dermatology 1995, 191 (2), 93–98. 53. Pan, Y.; Farkas, D.L.
Noninvasive imaging of living human skin with
dual-wavelength optical coherence tomography in two and
three dimensions. J. Biomed. Opt. 1998, 3 (4), 446–455. 54.
Boppart, S.A.; Bouma, B.E.; Pitris, C.; Tearney, G.J.;
Fujimoto, J.G.; Brezinski, M.E. Forward-imaging instruments
for optical coherence tomography. Opt. Lett. 1997, 22 (21),
1618–1620. 55. Li, X.D.; Drexler, W.; Pitris, C.; Ghanta,
R.; Jesser, C.; Hermann, J.; Stamper, D.; Golden, D.;
Martin, S.; Fujimoto, J.G.; Brezinski, M.E. Imaging of
Osteoarthritic Cartilage With Optical Coherence Tomography:
Micronstructure and Polarization Sensitivity. In Conference
on Lasers and Electro-Optics; Optical Society of America
and IEEE: Baltimore, MD, 1999; 339. 56. Tearney, G.J.;
Boppart, S.A.; Bouma, B.E.; Brezinski, M.E.; Weissman,
N.J.; Southern, J.F.; Fujimoto, J.G. Scanning single-mode
fiber optic catheter-endoscope for optical coherence
tomography. Opt. Lett. 1996, 21 (7), 543–545. 57. Sergeev,
A.M.; Gelikonov, V.M.; Gelikonov, G.V.; Feldchtein, F.I.;
Kuranov, R.V.; Gladkova, N.D.; Shakhova, N.M.; Suopova,
L.B.; Shakhov, A.V.; Kuznetzova, I.A.; Denisenko, A.N.;
Pochinko, V.V.; Chumakov Yu, P.; Streltzova, O.S. In vivo
endoscopic OCT imaging of precancer and cancer states of
human mucosa. Opt. Express 1997, 1 (13) 58. Rollins, A.M.;
Ung-arunyawee, R.; Chak, A.; Wong, R.C.K.; Kobayashi, K.;
Sivak, M.V.; Izatt, J.A. Realtime in vivo imaging of human
gastrointestinal ultrastructure by use of endoscopic
optical coherence tomography with a novel efficient
interferometer design. Opt. Lett. 1999, 24 (19), 1358–1360.
59. Pan, Y.T.; Xie, H.K.; Fedder, G.K. Endoscopic optical
coherence tomography based on a microelectromechanical
mirror. Opt. Lett. 2001, 26 (24), 1966–1968. 60. Li, X.D.;
Chudoba, C.; Ko, T.; Pitris, C.; Fujimoto, J.G. Imaging
needle for optical coherence tomography. Opt. Lett. 2000,
25 (20), 1520–1522. 61. Li, X.D.; Ko, T.H.; Fujimoto, J.G.
Intraluminal fiberoptic Doppler imaging catheter for
structural and functional coherence tomography. Opt. Lett.
2001, 26 (23), 1906–1908. 62. Tearney, G.J.; Brezinski,
M.E.; Bouma, B.E.; Boppart, S.A.; Pitris, C.; Southern,
J.F.; Fujimoto, J.G. In vivo endoscopic optical biopsy with
optical coherence tomography. Science 1997, 276 (5321),
2037–2039. 63. Bouma, B.E.; Tearney, G.J.; Compton, C.C.;
Nishioka, N.S. High-resolution imaging of the human
esophagus and stomach in vivo using optical coherence
tomography. Gastrointest. Endosc. 2000, 51 (4 Pt 1),
467–474. 64. Sivak, M.V., Jr.; Kobayashi, K.; Izatt, J.A.;
Rollins, A.M.; Ung-Runyawee, R.; Chak, A.; Wong, R.C.;
Isenberg, G.A.; Willis, J. High-resolution endoscopic
imaging of the GI tract using optical coherence tomography.
Gastrointest. Endosc. 2000, 51 (4 Pt 1), 474–479. 65. Li,
X.D.; Boppart, S.A.; Van Dam, J.; Mashimo, H.; Mutinga, M.;
Drexler, W.; Klein, M.; Pitris, C.; Krinsky, M.L.;
Brezinski, M.E.; Fujimoto, J.G. Optical coherence
tomography: Advanced technology for the endoscopic imaging
of Barrett’s esophagus. Endoscopy 2000, 32 (12), 921–930.
66. Feldchtein, F.I.; Gelikonov, G.V.; Gelikonov, V.M.;
Kuranov, R.V.; Sergeev, A.M.; Gladkova, N.D.; Shakhov,
A.V.; Shakhova, N.M.; Snopova, L.B.; Terent’eva, A.B.;
Zagainova, E.V.; Chumakov Yu, P.; Kuznetzova, I.A.
Endoscopic applications of optical coherence tomography.
Opt. Express 1998, 3 (6) 67. Fujimoto, J.G.; Boppart, S.A.;
Tearney, G.J.; Bouma, B.E.; Pitris, C.; Brezinski, M.E.
High resolution in vivo intra-arterial imaging with optical
coherence tomography. Heart 1999, 82 (2), 128–133. 68.
Feldchtein, F.I.; Gelikonov, V.M.; Gelikonov, G.V. Handbook
of Optical Coherence Tomography; Bouma, B.E.; Tearney,
G.J., Eds.; Marcel Dekker, Inc.: New York, 2001; 125–142.
69. Liu, X.M.; Cobb, M.J.; Chen, Y.C.; Li, X.D. Miniature
Lateral Priority Scanning Endoscope for Real-Time
Forward-Imaging Optical Coherence Tomography. Opt. Lett. in
press. 70. Kulkarni, M.D.; Izatt, J.A. Spectroscopic
Optical Coherence Tomography. In Conference on Lasers and
Electro Optics (CLEO’96); Optical Society of America:
Anaheim, CA, 1996; 59–60. 71. Morgner, U.; Drexler, W.;
Kaertner, F.X.; Li, X.D.; Pitris, C.; Ippen, E.P.;
Fujimoto, J.G. Spectroscopic optical coherence tomography.
Opt. Lett. 2000, 25 (2), 111–113. 72. Tanaka, T.; Riva, C.;
Ben-Sira, B. Blood velocity measurements in human retinal
vessels. Science 1974, 186 (4166), 830–831. 73. Stern, M.D.
In vivo evaluation of microcirculation by coherent light
scattering. Nature 1975, 254 (5495), 56–58. 74. Bonner,
R.F.; Nossal, R. Laser Doppler Blood Flowmetry; Shepherd,
A.P., Odberg, P.A., Eds.; Kluwer Academic: Boston, 1990;
17–45. 75. Zhao, Y.H.; Chen, Z.P.; Saxer, C.; Xiang, S.H.;
de Boer, J.F.; Nelson, J.S. Phase-resolved optical
coherence tomography and optical Doppler tomography for
imaging blood flow in human skin with fast scanning speed
and high velocity sensitivity. Opt. Lett. 2000, 25 (2),
114–116. O

76. Yang, V.X.D.; Gordon, M.L.; Mok, A.; Zhao, Y.H.; Chen,
Z.P.; Cobbold, R.S.C.; Wilson, B.C.; Vitkin, I.A. Improved
phase-resolved optical Doppler tomography using the Kasai
velocity estimator and histogram segmentation. Optics
Commun. 2002, 208 (4–6), 209–214.

77. Major, A.; Kimel, S.; Mee, S.; Milner, T.E.; Smithies,
D.J.; Srinivas, S.M.; Chen, Z.P.; Nelson, J.S.
Microvascular photodynamic effects determined in vivo using
optical Doppler tomography. IEEE J. Sel. Top. Quant.
Electron. 1999, 5 (4), 1168–1175.

78. Wong, R.C.K.; Yazdanfar, S.; Izatt, J.A.; Kulkarni,


M.D.; Barton, J.K.; Welch, A.J.; Willis, J.; Sivak, M.V.
Visualization of subsurface blood vessels by color Doppler
optical coherence tomography in rats: Before and after
hemostatic therapy. Gastrointest. Endosc. 2002, 55 (1),
88–95.

79. Hee, M.R.; Huang, D.; Swanson, E.A.; Fujimoto, J.G.


Polarization-sensitive low-coherence reflectometer for
birefringence characterization and ranging. J. Opt. Soc.
Am., B 1992, 9, 903–908.

80. de Boer, J.F.; Srinivas, S.M.; Malekafzali, A.; Chen,


Z.P.; Nelson, J.S. Imaging thermally damaged tissue by
polarization sensitive optical coherence tomography. Opt.
Express 1998, 3 (6), 212–218.
81. Jiao, S.L.; Wang, L.H.V. Two-dimensional depth-resolved
Mueller matrix of biological tissue measured with
double-beam polarization-sensitive optical coherence
tomography. Opt. Lett. 2002, 27 (2), 101–103.

82. Humphrey-Website. (Information on commercial ophthalmic


optical coherence tomography is available on
http://www.meditec.zeiss.com/).

83. Jackle, S.; Gladkova, N.; Feldchtein, F.; Terentieva,


A.; Brand, B.; Gelikonov, G.; Gelikonov, V.; Sergeev, A.;
Fritscher-Ravens, A.; Freund, J.; Seitz, U.; Schroder, S.;
Soehendra, N. In vivo endoscopic optical coherence
tomography of the human gastrointestinal tract—Toward
optical biopsy. Endoscopy 2000, 32 (10), 743–749.

84. Pfau, P.R.; Sivak, M.V.; Chak, A.; Kinnard, M.; Wong,
R.C.K.; Isenberg, G.A.; Izatt, J.A.; Rollins, A.; Westphal,
V. Criteria for the diagnosis of dysplasia by endoscopic
optical coherence tomography. Gastrointest. Endosc. 2003,
58 (2), 196–202.

85. Wax, A.; Yang, C.H.; Dasari, R.R.; Feld, M.S.


Measurement of angular distributions by use of
low-coherence interferometry for light-scattering
spectroscopy. Opt. Lett. 2001, 26 (6), 322–324.

86. Wax, A.; Yang, C.H.; Backman, V.; Badizadegan, K.;


Boone, C.W.; Dasari, R.R.; Feld, M.S. Cellular organization
and substructure measured using angle— Resolved
low-coherence interferometry. Biophys. J. 2002, 82 (4),
2256–2264. 87. Colston, B.W.; Sathyam, U.S.; DaSilva, L.B.;
Everett, M.J.; Stroeve, P.; Otis, L.L. Dental OCT. Opt.
Express 1998, 3 (6), 230–238. 88. Otis, L.L.; Everett,
M.J.; Sathyam, U.S.; Colston, B.W., Jr. Optical coherence
tomography: A new imaging technology for dentistry. J. Am.
Dent. Assoc. 2000, 131 (4), 511–514. 89. Tearney, G.J.;
Yabushita, H.; Houser, S.L.; Aretz, H.T.; Jang, I.K.;
Schlendorf, K.H.; Kauffman, C.R.; Shishkov, M.; Halpern,
E.F.; Bouma, B.E. Quantification of macrophage content in
atherosclerotic plaques by optical coherence tomography.
Circulation 2003, 107 (1), 113–119. 90. Virmani, R.;
Kolodgie, F.D.; Burke, A.P.; Farb, A.; Schwartz, S.M.
Lessons from sudden coronary death: A comprehensive
morphological classification scheme for atherosclerotic
lesions. Arterioscler. Thromb. Vasc. Biol. 2000, 20 (5),
1262–1275. 91. Patwari, P.; Weissman, N.J.; Boppart, S.A.;
Jesser, C.; Stamper, D.; Fujimoto, J.G.; Brezinski, M.E.
Assessment of coronary plaque with optical coherence
tomography and high-frequency ultrasound. Am. J. Cardiol.
2000, 85 (5), 641–644. 92. Boppart, S.A.; Brezinski, M.E.;
Bouma, B.E.; Tearney, G.J.; Fujimoto, J.G. Investigation of
developing embryonic morphology using optical coherence
tomography. Dev. Biol. 1996, 177 (1), 54–63. 93. Boppart,
S.A.; Tearney, G.J.; Bouma, B.E.; Southern, J.F.;
Brezinski, M.E.; Fujimoto, J.G. Noninvasive assessment of
the developing xenopus cardiovascular system using optical
coherence tomography. Proc. Natl. Acad. Sci. U. S. A. 1997,
94 (9), 4256–4261. 94. Yelbuz, T.M.; Choma, M.A.; Thrane,
L.; Kirby, M.L.; Izatt, J.A. Optical coherence tomography—A
new highresolution imaging technology to study cardiac
development in chick embryos. Circulation 2002, 106 (22),
2771–2774. 95. Cobb, M.J.; Chen, Y.C.; Underwood, R.A.;
Thariani, R.A.; Usui, M.L.; Olerud, J.E.; Li, X.D.
Assessment of Cutaneous Wound Healing Using Optical
Coherence Tomography. In OSA Biomedical Optics Topical
Meetings; Optical Society of America: Miami Beach, FL,
2004; FH22. 96. Patterson, J.; Li, X.D.; Herring, S.;
Stayton, P.S. Regenerative Matrices for Oriented
Craniofacial Bone Growth. In The 2003 Annual BMES Fall
Meeting; Session 2.6: Tissue Engineering of Bone and
Cartilage; Biomedical Engineering Society: Nashville, TN,
2003. 97. Dunkers, J.P.; Cicerone, M.T.; Washburn, N.R.
Collinear optical coherence and confocal fluorescence
microscopies for tissue engineering. Opt. Express 2003, 11
(23), 3074–3079.
Optical Detection of Cancers

1. ACS. Cancer Facts & Figures 2005. American Cancer


Society, 2005.

2. Lodish, H.F.; Berk, A.; Matsudaira, P.; Kaiser, C.A.;


Krieger, M.; Scott, M.P.; Zipursky, S.L.; Darnell, J.
Molecular Cell Biology; 5th Ed.; W.H. Freeman and Company:
New York, 2003.

3. Cotran, R.S.; Kumar, V.; Collins, T.; Robbins, S.L.


Robbins’ Pathologic Basis of Disease; 6th Ed.; Saunders:
Philadelphia, 1999.

4. Lu, J.Q.; Yang, P.; Hu, X.H. Simulations of light


scattering from a biconcave red blood cell using the FDTD
method. J. Biomed. Opt. 2005, 10 (2) 024022.

5. Chandrasekhar, S. Radiative Transfer; Dover


Publications: New York, 1960.

6. van de Hulst, H.C. Light Scattering by Small Particles;


Wiley: New York, 1957.

7. Ma, X.; Lu, J.Q.; Brock, R.S.; Jacobs, K.M.; Yang, P.;
Hu, X.H. Determination of complex refractive index of
polystyrene microspheres from 370 to 1610 nm. Phys. Med.
Biol. 2003, 48, 4165–4172.

8. Wilson, B.C.; Adam, G. A Monte Carlo model for the


absorption and flux distributions of light in tissue. Med.
Phys. 1983, 10 (6), 824–830.

9. Li, K.; Lu, J.Q.; Brock, R.S.; Yang, B.; Hu, X.H.
Quantitative modeling of skin images using parallel Monte
Carlo methods. SPIE Proc. 2005, 5693, 82–87.

10. Kak, A.C.; Slaney, M. Principles of Computerized


Tomographic Imaging; IEEE Press: New York, 1988.

11. Cutler, M. Transillumination as an aid in the diagnosis


of breast lesions. Surg. Gynecol. Obstet. 1929, 48 (6),
721–729.

12. D’Orsi, C.J.; Bartrum, R.J.; Moskowitz, M.M. Light


scanning of the breast. In Mammography, Thermography, and
Ultrasound in Breast Cancer Detection; 2nd Ed.; Gold, R.H.,
Ed.; Grune & Stratton: New York, 1987; 169–177.

13. Profio, A.E.; Navarro, G.A.; Sartorius, O.W. Scientific


basis of breast diaphanography. Med. Phys. 1989, 16 (1),
60–65.

14. Moncrieff, M.; Cotton, S.; Claridge, E.; Hall, P.


Spectrophotometric intracutaneous analysis: a new technique
for imaging pigmented skin lesions. Br. J. Dermatol. 2002,
146 (3), 448–457.

15. Elbaum, M.; Kopf, A.W.; Rabinovitz, H.S.; Langley,


R.G.; Kamino, H.; Mihm, M.C., Jr.; Sober, A.J.; Peck, G.L.;
Bogdan, A.; Gutkowicz-Krusin, D.; Greenebaum, M.; Keem, S.;
Oliviero, M.; Wang, S. Automatic differentiation of
melanoma from melanocytic nevi with multispectral digital
dermoscopy: a feasibility study. J. Am. Acad. Dermatol.
2001, 44 (2), 207–218.

16. Farina, B.; Bartoli, C.; Bono, A.; Colombo, A.; Lualdi,
M.; Tragni, G.; Marchesini, R. Multispectral imaging
approach in the diagnosis of cutaneous melanoma:
potentiality and limits. Phys. Med. 2000, 45 (5),
1243–1254.

17. Kienle, A.; Lilge, L.; Patterson, M.S.; Hibst, R.;


Steiner, R.; Wilson, B.C. Spatially resolved absolute
diffuse reflectance measurements for noninvasive
determination of the optical scattering and absorption
coefficients of biological tissue. Appl. Opt. 1996, 35,
2304–2314. 18. Pfefer, T.J.; Matchette, L.S.; Bennett,
C.L.; Gall, J.A.; Wilke, J.N.; Durkin, A.J.; Ediger, M.N.
Reflectancebased determination of optical properties in
highly attenuating tissue. J. Biomed. Opt. 2003, 8 (2),
206– 215. 19. Nordstrom, R.J.; Burke, L.; Niloff, J.M.;
Myrtle, J.F. Identification of cervical intraepithelial
neoplasia (CIN) using UV-excited fluorescence and
diffuse-reflectance tissue spectroscopy. Lasers Surg. Med.
2001, 29 (2), 118–127. 20. Mirabal, Y.N.; Chang, S.K.;
Atkinson, E.N.; Malpica, A.; Follen, M.; Richards-Kortum,
R. Reflectance spectroscopy for in vivo detection of
cervical precancer. J. Biomed. Opt. 2002, 7 (4), 587–594.
21. Bigio, I.J.; Bown, S.G.; Briggs, G.; Kelley, C.;
Lakhani, S.; Pickard, D.; Ripley, P.M.; Rose, I.G.;
Saunders, C. Diagnosis of breast cancer using
elastic-scattering spectroscopy: preliminary clinical
results. J. Biomed. Opt. 2000, 5 (2), 221–228. 22.
Utzinger, U.; Brewer, M.; Silva, E.; Gershenson, D.; Blast,
R.C., Jr.; Follen, M.; Richards-Kortum, R. Reflectance
spectroscopy for in vivo characterization of ovarian
tissue. Lasers Surg. Med. 2001, 28 (1), 56–66. 23.
Garcia-Uribe, A.; Kehtarnavaz, N.; Marquez, G.; Prieto, V.;
Duvic, M.; Wang, L.V. Skin cancer detection by
spectroscopic oblique-incidence reflectometry: classification
and physiological origins. Appl. Opt. 2004, 43, 2643–2650.
24. Zonios, G.; Perelman, L.T.; Backman, V.; Manoharan, R.;
Fitzmaurice, M.; Van Dam, J.; Feld, M.S. Diffuse reflectance
spectroscopy of human adenomatous colon polyps in vivo.
Appl. Opt. 1999, 38 (31), 6628–6637. 25. Johnson, K.S.;
Chicken, D.W.; Pickard, D.C.; Lee, A.C.; Briggs, G.;
Falzon, M.; Bigio, I.J.; Keshtgar, M.R.; Bown, S.G. Elastic
scattering spectroscopy for intraoperative determination of
sentinel lymph node status in the breast. J. Biomed. Opt.
2004, 9 (6), 1122– 1128. 26. Canpolat, M.; Mourant, J.R.
Particle size analysis of turbid media with a single
optical fiber in contact with the medium to deliver and
detect white light. Appl. Opt. 2001, 40 (22), 3792–3799.
27. Georgakoudi, I.; Jacobson, B.C.; Van Dam, J.; Backman,
V.; Wallace, M.B.; Muller, M.G.; Zhang, Q.; Badizadegan,
K.; Sun, D.; Thomas, G.A.; Perelman, L.T.; Feld, M.S.
Fluorescence, reflectance, and lightscattering spectroscopy
for evaluating dysplasia in patients with Barrett’s
esophagus. Gastroenterology 2001, 120 (7), 1620–1629. 28.
Madsen, S.J.; Anderson, E.R.; Haskell, R.C.; Tromberg, B.
Portable, high-bandwidth frequency-domain photon migration
instrument for tissue spectroscopy. Opt. Lett. 1994, 19
(23), 1934–1936. 29. Fantini, S.; Walker, S.A.;
Franceschini, M.A.; Kaschke, M.; Schlag, P.M.; Moesta, K.T.
Assessment of the size, position, and optical properties of
breast tumors in vivo by noninvasive optical methods. Appl.
Opt. 1998, 37 (10), 1982–1989. 30. Fishkin, J.B.; Coquoz,
O.; Anderson, E.R.; Brenner, M.; Tromberg, B.J.
Frequency-domain photon migrationmeasurements of normal and
malignant tissue optical properties in a human subject.
Appl. Opt. 1997, 36 (31), 10–20. 31. Hornung, R.; Pham,
T.H.; Keefe, K.A.; Berns, M.W.; Tadir, Y.; Tromberg, B.J.
Quantitative near-infrared spectroscopy of cervical
dysplasia in vivo. Hum. Reprod. 1999, 14 (11), 2908–2916.
32. Heffer, E.; Pera, V.; Schutz, O.; Siebold, H.; Fantini,
S. Near-infrared imaging of the human breast: complementing
hemoglobin concentration maps with oxygenation images. J.
Biomed. Opt. 2004, 9 (6), 1152–1160. 33. Das, B.B.; Liu,
F.; Alfano, R.R. Time-resolved fluorescence and photon
migration studies in biomedical and model random media.
Rep. Prog. Phys. 1997, 60, 227– 292. 34. Patterson, M.S.;
Chance, B.; Wilson, B.C. Time resolved reflectance and
transmittance for the noninvasive measurement of tissue
optical properties. Appl. Opt. 1989, 28 (12), 2331–2336.
35. Kienle, A.; Patterson, M.S. Determination of the
optical properties of turbid media from a single Monte
Carlo simulation. Phys. Med. Biol. 1996, 41, 2221– 2227.
36. Zacharakis, G.; Zolindaki, A.; Sakkalis, V.;
Filippidis, G.; Papazoglou, T.G.; Tsiftsis, D.D.;
Koumantakis, E. In vitro optical characterization and
discrimination of female breast tissue during near infrared
femtosecond laser pulses propagation. J. Biomed. Opt. 2001,
6 (4), 446–449. 37. Torricelli, A.; Spinelli, L.; Pifferi,
A.; Taroni, P.; Cubeddu, R. Use of a nonlinear perturbation
approach for in vivo breast lesion characterization by
multiwavelength time-resolved optical mammography. Opt.
Exp. 2003, 11 (8), 853–867. 38. Grosenick, D.; Wabnitz, H.;
Moesta, K.T.; Mucke, J.; Moller, M.; Stroszczynski, C.;
Stossel, J.; Wassermann, B.; Schlag, P.M.; Rinneberg, H.
Concentration and oxygen saturation of haemoglobin of 50
breast tumours determined by time-domain optical
mammography. Phys. Med. Biol. 2004, 49 (7), 1165–1181. 39.
Arridge, S.R. Optical tomography in medical imaging.
Inverse Problems 1999, 15, R41–R93. 40. Arridge, S.R.;
Hebden, J.C. Optical imaging in medicine: II. Modelling and
reconstruction. Phys. Med. Biol. 1997, 42 (5), 841–853. 41.
Li, A.; Miller, E.L.; Kilmer, M.E.; Brukilacchio, T.J.;
Chaves, T.; Stott, J.; Zhang, Q.; Wu, T.; Chorlton, M.;
Moore, R.H.; Kopans, D.B.; Boas, D.A. Tomographic optical
breast imaging guided by three-dimensional mammography.
Appl. Opt. 2003, 42 (25), 5181–5190. 42. Dehghani, H.;
Pogue, B.W.; Poplack, S.P.; Paulsen, K.D. Multiwavelength
three-dimensional near-infrared tomography of the breast:
initial simulation, phantom, and clinical results. Appl.
Opt. 2003, 42 (1), 135–145. 43. Huang, D.; Swanson, E.A.;
Lin, C.P.; Schuman, J.S.; Stinson, W.G.; Chang, W.; Hee,
M.R.; Flotte, T.; Gregory, K.; Puliafito, C.A., et al.
Optical coherence tomography. Science 1991 , 254 (5035),
1178–1181. 44. Shields, C.L.; Mashayekhi, A.; Luo, C.K.;
Materin, M.A.; Shields, J.A. Optical coherence tomography
in children: analysis of 44 eyes with intraocular tumors
and simulating conditions. J. Pediatr. Ophthalmol.
Strabismus 2004, 41 (6), 338–344. 45. Knuttel, A.;
Boehlau-Godau, M. Spatially confined and temporally resolved
refractive index and scattering evaluation in human skin
performed with optical coherence tomography. J. Biomed.
Opt. 2000, 5 (1), 83–92. 46. Strasswimmer, J.; Pierce,
M.C.; Park, B.H.; Neel, V.; de Boer, J.F.
Polarization-sensitive optical coherence tomography of
invasive basal cell carcinoma. J. Biomed. Opt. 2004, 9 (2),
292–298. 47. Wilder-Smith, P.; Jung, W.G.; Brenner, M.;
Osann, K.; Beydoun, H.; Messadi, D.; Chen, Z. In vivo
optical coherence tomography for the diagnosis of oral
malignancy. Lasers Surg. Med. 2004, 35 (4), 269–275. 48.
Sergeev, A.M.; Gelikonov, V.M.; Gelikonov, G.V.;
Feldchtein, F.I.; Kuranov, R.V.; Gladkova, N.D.; Shakhova,
N.M.; Snopova, L.B.; Shakhov, A.V.; Kuznetzova, I.A.;
Denisenko, A.N.; Pochinko, V.V.; Chumakov, Y.P.;
Streltzova, O.S. In vivo endoscopic OCT imaging of
precancer and cancer states of human mucosa. Opt. Exp.
1997, 1 (13), 432–440. 49. Poneros, J.M.; Nishioka, N.S.
Diagnosis of Barrett’s esophagus using optical coherence
tomography. Gastrointest. Endosc. Clin. N. Am. 2003, 13
(2), 309–323. 50. Bouma, B.E.; Tearney, G.J.; Compton,
C.C.; Nishioka, N.S. High-resolution imaging of the human
esophagus and stomach in vivo using optical coherence
tomography. Gastrointest. Endosc. 2000, 51 (4 pt 1),
467–474. 51. Jackle, S.; Gladkova, N.; Feldchtein, F.;
Terentieva, A.; Brand, B.; Gelikonov, G.; Gelikonov, V.;
Sergeev, A.; Fritscher-Ravens, A.; Freund, J.; Seitz, U.;
Schroder, S.; Soehendra, N. In vivo endoscopic optical
coherence tomography of esophagitis, Barrett’s esophagus,
and adenocarcinoma of the esophagus. Endoscopy 2000, 32
(10), 750–755. 52. Alfano, R.R.; Tata, D.B.; Cordero, J;
Tomashefsky, P.; Longo, F.W.; Alfano, M.A. Laser induced
fluorescence spectroscopy from native cancerous and normal
tissue. IEEE J. Quantum Electron. 1984, 20 (12), 1507–1511.
53. Alfano, R.R.; Tang, G.C.; Pradhan, A.; Lam, W.; Choy,
D.S.J.; Opher, E. Fluorescence spectra from cancerous and
normal human breast and lung tissues. IEEE J. Quantum
Electron. 1987, 23 (10), 1806–1811. 54. Gupta, P.K.;
Majumder, S.K.; Uppal, A. Breast cancer diagnosis using N2
laser excited autofluorescence spectroscopy. Lasers Surg.
Med. 1997, 21 (5), 417–422. 55. Hage, R.; de la Riviere,
A.B.; Seldenrijk, C.A.; van den Bosch, J.M. Update in
pulmonary carcinoid tumors: a review article. Ann. Surg.
Oncol. 2003, 10 (6), 697–704. 56. Zellweger, M.; Goujon,
D.; Conde, R.; Forrer, M.; van den Bergh, H.; Wagnieres, G.
Absolute autofluorescence spectra of human healthy,
metaplastic, and early cancerous bronchial tissue in vivo.
Appl. Opt. 2001, 40 (22), 3784–3791. 57. Qu, J.Y.; Wing,
P.; Huang, Z.; Kwong, D.; Sham, J.; Lee, S.L.; Ho, W.K.;
Wei, W.I. Preliminary study of in vivo autofluorescence of
nasopharyngeal carcinoma O and normal tissue. Lasers Surg.
Med. 2000, 26 (5), 432–440.

58. Ramanujam, N.; Mitchell, M.F.; Mahadevan, A.; Thomsen,


S.; Malpica, A.; Wright, T.; Atkinson, N.; Richards-Kortum,
R. Development of a multivariate statistical algorithm to
analyze human cervical tissue fluorescence spectra acquired
in vivo. Lasers Surg. Med. 1996, 19 (1), 46–62.

59. Lin, W.; Yuan, X.; Yuen, P.; Wei, W.I.; Sham, J.; Shi,
P.; Qu, J. Classification of in vivo autofluorescence spectra
using support vector machines. J. Biomed. Opt. 2004, 9 (1),
180–186.

60. Zangaro, R.A.; Silveira, L.J.; Manoharan, R.; Zonios,


G.; Itzkan, I.; Dasari, R.R.; Van Dam, J.; Feld, M.S. Rapid
multiexcitation fluorescence spectroscopy system for in vivo
tissue diagnosis. Appl. Opt. 1996, 35 (25), 5211–5219.

61. Ramanujam, N.; Mitchell, M.F.; Mahadevan, A.; Warren,


S.; Thomsen, S.; Silva, E.; Richards-Kortum, R. In vivo
diagnosis of cervical intraepithelial neoplasia using
337-nm-excited laser-induced fluorescence. Proc. Natl. Acad.
Sci. USA 1994, 91 (21), 10193–10197.

62. Nath, A.; Rivoire, K.; Chang, S.; West, L.; Cantor,
S.B.; Basen-Engquist, K.; Adler-Storthz, K.; Cox, D.D.;
Atkinson, E.N.; Staerkel, G.; MacAulay, C.;
Richards-Kortum, R.; Follen, M. A pilot study for a
screening trial of cervical fluorescence spectroscopy. Int.
J. Gynecol. Cancer 2004, 14 (6), 1097–1107.

63. Brewer, M.; Utzinger, U.; Silva, E.; Gershenson, D.;


Bast, R.C., Jr.; Follen, M; Richards-Kortum, R.
Fluorescence spectroscopy for in vivo characterization of
ovarian tissue. Lasers Surg. Med. 2001, 29 (2), 128–135.
64. Tadrous, P.J.; Siegel, J.; French, P.M.; Shousha, S.;
Lalani, el N.; Stamp, G.W. Fluorescence lifetime imaging of
unstained tissues: early results in human breast cancer. J.
Pathol. 2003, 199 (3), 309–317. 65. Neerken, S.; Lucassen,
G.W.; Bisschop, M.A.; Lenderink, E.; Nuijs, T.A.
Characterization of age-related effects in human skin: a
comparative study that applies confocal laser scanning
microscopy and optical coherence tomography. J. Biomed.
Opt. 2004, 9 (2), 274– 281. 66. Rajadhyaksha, M.; Gonzalez,
S.; Zavislan, J.M.; Anderson, R.R.; Webb, R.H. In vivo
confocal scanning laser microscopy of human skin II:
advances in instrumentation and comparison with histology.
J. Invest. Dermatol. 1999, 113 (3), 293–303. 67. Hanlon,
E.B.; Manoharan, R.; Koo, T.W.; Shafer, K.E.; Motz, J.T.;
Fitzmaurice, M.; Kramer, J.R.; Itzkan, I.; Dasari, R.R.;
Feld, M.S. Prospects for in vivo Raman spectroscopy. Phys.
Med. Biol. 2000, 45 (2), R1–59. 68. Skala, M.C.; Squirrell,
J.M.; Vrotsos, K.M.; Eickhoff, J.C.; Gendron-Fitzpatrick,
A.; Eliceiri, K.W.; Ramanujam, N. Multiphoton microscopy of
endogenous fluorescence differentiates normal, precancerous,
and cancerous squamous epithelial tissues. Cancer Res.
2005, 65 (4), 1180–1186. 69. Yasui, T.; Tohno, Y.; Araki,
T. Characterization of collagen orientation in human dermis
by two-dimensional second-harmonic-generation polarimetry.
J. Biomed. Opt. 2004, 9 (2), 259–264.
Optical Mapping

24. Fast, V.G.; Rohr, S.; Gillis, A.M.; Kleber, A.G.


Activation of cardiac tissue by extracellular electrical
shocks: Formation of ‘secondary sources’ at intercellular
clefts in monolayers of cultured myocytes. Circ. Res. 1998,
82 (3), 375–385.

25. Bub, G.; Glass, L.; Publicover, N.G.; Shrier, A.


Bursting calcium rotors in cultured cardiac myocyte
monolayers. Proc. Natl. Acad. Sci. U. S. A. 1998, 95 (17),
10283–10287.

26. Entcheva, E.; Lu, S.N.; Troppman, R.H.; Sharma, V.;


Tung, L. Contact fluorescence imaging of reentry in
monolayers of cultured neonatal rat ventricular myocytes.
J. Cardiovasc. Electrophysiol. 2000, 11 (6), 665–676. 27.
Arutunyan, A.; Swift, L.M.; Sarvazyan, N. Initiation and
propagation of ectopic waves: Insights from an in vitro
model of ischemia-reperfusion injury. Am. J. Physiol, Heart
Circ. Physiol. 2002, 283 (2), H741–H749. 28. Cohen, L.B.;
Lesher, S. Optical Monitoring of Membrane Potential:
Methods of Multisite Optical Measurement. In Optical
Methods in Cell Physiology; De Weer, P., Salzberg, B.M.,
Eds.; Wiley-Interscience, 1986. 29. Montana, V.; Farkas,
D.L.; Loew, L.M. Dual-wavelength ratiometric fluorescence
measurements of membrane potential. Biochemistry 1989, 28
(11), 4536–4539. 30. Efimov, I.R.; Rendt, J.M.; Salama, G.
Optical maps of intracellular [Ca2+]i transients and action
potentials from the surface of perfused guinea pig hearts.
Circulation 1994, 90 (II), 632.
Optics, Biomedical

2. O’Leary, M.A.; Boas, D.A.; Chance, B.; Yodh, A.G.


Refraction of diffuse photon density waves. Phys. Rev.
Lett. 1992, 69, 2658–2661.

3. Chance, B.; Kang, K.; He, L.; Weng, J.; Sevick, E.


Highly sensitive object location in tissue models with
linear in-phase and anti-phase multi-element optical arrays
in one and two dimensions. Proc. Natl. Acad. Sci. U.S.A.
1993, 90, 3423–3427.

4. Boas, D.A.; O’Leary, M.A.; Chance, B.; Yodh, A.G.


Scattering and wavelength transduction of diffuse photon
density waves. Phys. Rev., E 1993, 47, R2999– R3002.

5. Hetzel, F.; Patterson, M.; Preuss, L.; Wilson, B.


Recommended Nomenclature for Physical Quantities in Medical
Applications of Light; AAPM Report No. 57, Ed.; American
Institute of Physics: Woodbury, NY, 1996; 1–6.

6. Hale, G.M.; Querry, M.R. Optical constants of water in


the 200 nm to 200m wavelength region. Appl. Opt. 1973, 12,
555–563.

7. S. Prahl (of the Oregon Medical Laser Center, Portland,


OR) has tabulated the molar extinction coefficients for
oxy-hemoglobin and deoxy-hemoglobin using data from W.B.
Gratzer and N. Kollias. These tabulated data are available
at http://omlc.ogi.edu/spectra/ hemoglobin/summary.html.

8. Jacques, S.L.; Wang, L. Monte Carlo modeling of light


transport in tissues. In Optical-Thermal Response of
Laser-Irradiated Tissue; Welch, A.J., van Gemert, M.J.C.,
Eds.; Plenum Press: New York, NY, 1995; 73–100.

9. Niemz, M.H. Laser-Tissue Interactions: Fundamentals and


Applications; Springer-Verlag: Berlin, Germany, 1996.

10. Alfano, R.R.; Tata, D.B.; Cordero, J.J.; Tomashefsky,


P.; Longo, F.W.; Alfano, M.A. Laser induced fluorescence
spectroscopy from native cancerous and normal tissues. IEEE
J. Quantum Electron. 1984, QE-20, 1507–1511.

11. Prasad, P.N. Introduction to Biophotonics; John Wiley


and Sons: Hoboken, NJ, 2003.

12. Duderstadt, J.J.; Hamilton, L.J. Nuclear Reactor


Analysis; Wiley: New York, NY, 1976; 113.
13. Arridge, S.R.; Cope, M.; Delpy, D.T. The theoretical
basis for the determination of optical pathlengths in
tissue: Temporal and frequency analysis. Phys. Med. Biol.
1992, 37, 1531–1560.

14. Perelman, L.T.; Backman, V.; Wallace, M.; Zonios, G.;


Manohaan, R.; Nusrat, A.; Shields, S.; Seiler, M.; Lima,
C.; Hamamo, T.; Itzkan, I.; Van Dam, J.; Crawford, J.M.;
Feld, M.S. Observation of periodic fine structure in
reflectance from biological tissue: A new technique for
measuring nuclear size distribution. Phys. Rev. Lett. 1998,
80, 627–630.

15. Maier, J.S.; Walker, S.A.; Fantini, S.; Franceschini,


M.A.; Gratton, E. Possible correlation between blood
glucose concentration and the reduced scattering coefficient
of tissues in the near infrared. Opt. Lett. 1994, 19,
2062–2064. 16. Bruulsema, J.T.; Hayward, J.E.; Farrel,
T.J.; Patterson, M.S.; Heinemann, L.; Berger, M.;
Koschinsky, T.; Sandahl-Christiansen, J.; Orskov, H.;
Essenpreis, M.; Schmelzeisen-Redeker, G.; Bo¨cker, D.
Correlation between blood glucose concentration in
diabetics and noninvasively measured tissue optical
scattering coefficient. Opt. Lett. 1997, 22, 190–192. 17.
MacVicar, B.A.; Hochman, D. Imaging of synaptically evoked
intrinsic optical signals in hippocampal slices. J.
Neurosci. 1991, 11, 1458–1469. 18. Stepnoski, R.A.;
LaPorta, A.; Raccuia-Behling, F.; Blonder, G.E.; Slusher,
R.E.; Kleinfeld, D.J. Noninvasive detection of changes in
membrane potential in cultured neurons by light scattering.
Proc. Natl. Acad. Sci. U. S. A. 1991, 88, 9382–9386. 19.
Mahadevan-Jansen, A.; Richards-Kortum, R. Raman
spectroscopy for the detection of cancers and precancers.
J. Biomed. Opt. 1996, 1, 31–70. 20. Wilson, T.; Sheppard,
C. Theory and Practice of Scanning Optical Microscopy;
Academic Press: New York, NY, 1984. 21. Denk, W.;
Strickler, J.H.; Webb, W.W. Two-photon laser scanning
fluorescence microscopy. Science 1990, 248, 73–76. 22.
Tadrous, P.J. Methods for imaging the structure and
function of living tissues and cells: 2. Fluorescence
lifetime imaging. J. Pathol. 2000, 191, 229–234. 23.
Schmitt, J.M. Optical coherence tomography (OCT): A review.
IEEE J. Sel. Top. Quantum Electron. 1999, 5, 1205–1215. 24.
Fercher, A.F. Optical coherence tomography. J. Biomed. Opt.
1996, 1, 157–173. 25. Marchesini, R.; Bono, A.; Bartoli,
C.; Lualdi, M.; Tomatis, S.; Cascinelli, N. Optical imaging
and automated melanoma detection: Questions and answers.
Melanoma Res. 2002, 12, 279–286. 26. Frostig, R.D.; Lieke,
E.E.; Ts’o, D.Y.; Grinvald, A. Cortical functional
architecture and local coupling between neuronal activity
and the microcirculation revealed by in vivo
high-resolution imaging of intrinsic signals. Proc. Natl.
Acad. Sci. U. S. A. 1990, 87, 6082–6086. 27. Farkas, D.L.;
Becker, D. Applications of spectral imaging: Detection and
analysis of human melanoma and its precursors. Pigment Cell
Res. 2001, 14, 2–8. 28. Hebden, J.C.; Arridge, S.R.; Delpy,
D.T. Optical imaging in medicine: I. Experimental
techniques. Phys. Med. Biol. 1997, 42, 825–840. 29.
Arridge, S.R.; Hebden, J.C. Optical imaging in medicine:
II. Modelling and reconstruction. Phys. Med. Biol. 1997,
42, 841–853.
Organic and Inorganic Matrices

8. Boivin, G.; Meunier, P.J. The degree of mineralization


of bone tissue measured by computerized quantitative
contact microradiography. Calcif. Tissue Int. 2002, 70 (6),
503–511.

9. Cummings, S.R.; Bates, D.; Black, D.M. Clinical use of


bone densitometry: scientific review. JAMA 2002, 288 (15),
1889–1897.

10. Ding, M.; Day, J.S.; Burr, D.B.; Mashiba, T.; Hirano,
T.; Weinans, H.; Sumner, D.R.; Hvid, I. Canine cancellous
bone microarchitecture after one year of high-dose
bisphosphonates. Calcif. Tissue Int. 2003, 72 (6), 737–44.

11. JCPDS. Joint Committee on Powder Diffraction Standards.


Annual Book of ASTM Standards.

12. Boskey, A.L. Mineral Analysis provides insights into


the mechanism of biomineralization. Calcif. Tissue Int.
2003, 72 (5), 533–536.

13. Boskey, A.; Spevak, L.; Tan, M.; Doty, S.B.; Butler,
W.T. Dentin sialoprotein (DSP) has limited effects on in
vitro apatite formation and growth. Calcif. Tissue Int.
2000, 67 (6), 472–478.

14. Frank, R.M.; Klewansky, P.; Hemmerle, J.; Tenenbaum, H.


Ultrastructural demonstration of the importance of crystal
size of bioceramic powders implanted into human periodontal
lesions. J. Clin. Periodontol. 1991, 18 (9), 669–680.

15. Tong, W.; Glimcher, M.J.; Katz, J.L.; Kuhn, L.; Eppell,
S.J. Size and shape of mineralites in young bovine bone
measured by atomic force microscopy. Calcif Tissue Int.
2003, 72 (5), 592–598.

16. Zhang, Y.; Cheng, F.; Li, D.; Wang, Y.; Zhang, G.;
Liao, W.; Tang, T.; Huang, Y.; He, W. Investigation of
elemental content distribution in femoral head slice with
osteoporosis by SRXRF microprobe. Biol. Trace. Elem. Res.
2005, 103 (2), 177–185.

17. Boskey, A.L.; Mendelsohn, R. Infrared analysis of bone


in health and disease. J. Biomed. Optics 2005, 10(031102-1
-9).

18. Tarnowski, C.P.; Ignelzi, M.A., Jr.; Morris, M.D.


Mineralization of developing mouse calvaria as revealed by
Raman microspectroscopy. J. Bone Miner. Res. 2002, 17 (6),
1118–1126.

19. Faibish, D.; Gomes, A.; Boivin, G.; Binderman, I.;


Boskey, A. Infrared imaging of calcified tissue in bone
biopsies from adults with osteomalacia. Bone. 2005, 36 (1),
6–12.

20. Paschalis, E.P.; Betts, F.; DiCarlo, E.; Mendelsohn,


R.; Boskey, A.L. FTIR microspectroscopic analysis of normal
human cortical and trabecular bone. Calcif. Tissue. Int.
1997, 61 (6), 480–486.

21. Gadeleta, S.J.; Boskey, A.L.; Paschalis, E.; Carlson,


C.; Menschik, F.; Baldini, T.; Peterson, M.; Rimnac, C.M. A
physical, chemical, and mechanical study of lumbar
vertebrae from normal, ovariectomized, and nandrolone
decanoate-treated cynomolgus monkeys (Macaca fascicularis)
Bone 2000, 27 (4), 541–550. 22. Paschalis, E.P.; Betts, F.;
DiCarlo, E.; Mendelsohn, R.; Boskey, A.L. FTIR
microspectroscopic analysis of human iliac crest biopsies
from untreated osteoporotic bone. Calcif. Tissue Int. 1997,
61 (6), 487–492. 23. Paschalis, E.P.; Boskey, A.L.; Kassem,
M.; Eriksen, E.F. Effect of hormone replacement therapy on
bone quality in early postmenopausal women. J. Bone Miner.
Res. 2003, 18 (6), 955–959. 24. Boskey, A.L.; DiCarlo, E.;
Paschalis, E.; West, P.; Mendelsohn, R. Comparison of
mineral quality and quantity in Iliac crest biopsies from
high-turnover and low-turnover osteoporosis: an FT-IR
Microspectroscopic Invest. Osteopor. Int. 2005, Aug 9, ePub
ahead of print. 25. Yamauchi, M. Collagen biochemistry: an
overview. Advances in tissue banking 2002, 6, 445–500. 26.
Eyre, D.R. Collagens and cartilage matrix homeostasis.
Clin. Orthop. Relat. Res. 2004, 427, Suppl S118–S122. 27.
Dalgleish, R. The human type I collagen mutation database.
Nucleic Acids Res. 1997, 25 (1), 181–187. 28. Byers, P.H.
Molecular genetics of chondrodysplasias, including clues to
development, structure, and function. Curr. Opin.
Rheumatol. 1994, 6 (3), 345–350. 29. Prockop, D.J.;
Kuivaniemi, H.; Tromp, G. Molecular basis of osteogenesis
imperfecta and related disorders of bone. Clin. Plast.
Surg. 1994, 21 (3), 407–413. 30. Niyibizi, C.; Wang, S.;
Mi, Z.; Robbins, P.D. Gene therapy approaches for
osteogenesis imperfecta. Gene Ther. 2004, 11 (4), 408–416.
31. Germain, D. Ehlers-Danlos syndromes. Clinical, genetic
and molecular aspects. Ann. Dermatol. Venereol. 1995, 122
(4), 187–204. 32. Bailey, A.J.; Wotton, S.F.; Sims, T.J.;
Thompson, P.W. Post-translational modifications in the
collagen of human osteoporotic femoral head. Biochem.
Biophys. Res. Commun. 1992, 185 (3), 801–805. 33. Krane,
S.M. Elucidation of the potential roles of matrix
metalloproteinases in skeletal biology. Arthritis Res.
Ther. 2003, 5 (1), 2–4. 34. Fisher, L.W.; Termine, J.D.
Noncollagenous proteins influencing the local mechanisms of
calcification. Clin. Orthop. Relat. Res. 1985, 200, 362–385.
35. Gokhale, J.; Robey, P.G.; Boskey, A.L. The biochemistry
of bone. In Osteoporosis, 2nd Ed.; San DiegoRobert Marcus,
David Feldman, Jennifer Kelsey, Eds.; Academic Press, 2001;
Vol. 1, 107–189. 36. Bonucci, E.; Nanci, A. Alkaline
phosphatase and tartrate-resistant acid phosphatase in
osteoblasts of normal and pathologic bone. Ital. J. Anat.
Embryol. 2001, 106 (2 Suppl. 1), 129–33. 37. Ho, A.M.;
Johnson, M.D.; Kingsley, D.M. Role of the mouse ank gene in
control of tissue calcification and arthritis. Science.
2000, 289 (5477), 265–270. 38. Johnson, K.; Terkeltaub, R.
Upregulated ank expression in osteoarthritis can promote
both chondrocytes MMP-13 expression and calcification via
chondrocyte extracellular PPi excess. Osteoarthritis
Cartil. 2004, 12 (4), 321–335. 39. Veis, A.; Sfeir, C.; Wu,
CB. Phosphorylation of the proteins of the extracellular
matrix of mineralized tissues by casein kinase-like
activity. Crit. Rev. Oral. Biol. Med. 1997, 8 (4), 360–379.
40. Mott, J.D.; Werb, Z. Regulation of matrix biology by
matrix metalloproteinases. Curr. Opin. Cell Biol. 2004, 16
(5), 558–564. 41. Drake, F.H.; Dodds, R.A.; James, I.E.;
Connor, J.R.; Debouck, C.; Richardson, S.; Lee-Rykaczewski,
E.; Coleman, L.; Rieman, D.; Barthlow, R.; Hastings, G.;
Gowen, M. Cathepsin K, but not cathepsins B, L, or S, is
abundantly expressed in human osteoclasts. J. Biol. Chem.
1996, 271 (21), 12,511–12,516. 42. Frenkel, S.R.; Bradica,
G.; Brekke, J.H.; Goldman, S.M.; Ieska, K.; Issack, P.;
Bong, M.R.; Tian, H.; Gokhale, J.; Coutts, R.D.;
Kronengold, R.T. Regeneration of articular
cartilage—evaluation of osteochondral defect repair in the
rabbit using multiphasic implants. Osteoarthritis Cartil.
2005, 13 (9), 798–807. 43. Blair, H.C.; Athanasou, N.A
Recent advances in osteoclast biology and pathological bone
resorption. Histol. Histopathol. 2004, 19 (1), 189–99. 44.
Goldberg, M.; Boskey, A.L. Lipids and biomineralizations.
Prog. Histochem. Cytochem. 1996, 31 (2), 1–187. 45. van der
Meulen, M.C.; Jepsen, K.J.; Mikic, B. Understanding bone
strength: size isn’t everything. Bone 2001, 29 (2),
101–104. 46. Ballock, R.T.; OuKeefe, R.J. Physiology and
pathophysiology of the growth plate. Birth Defects Res. C
Embryo Today 2003, 69 (2), 123–143. 47. Tuan, R.S. Biology
of developmental and regenerative skeletogenesis. Clin.
Orthop. Relat. Res. 2004, 427, Suppl. S105–S17. 48. Colnot,
C.I.; Helms, J.A. A molecular analysis of matrix remodeling
and angiogenesis during long bone development. Mech. Dev.
2001, 100 (2), 245–50. 49. Zelzer, E.; Olsen, B.R. Multiple
roles of vascular endothelial growth factor (VEGF) in
skeletal development, growth, and repair. Curr. Top. Dev.
Biol. 2005, 65, 169–87. 50. Xue, Y.; Karaplis, A.C.; Hendy,
G.N.; Goltzman, D.; Miao, D. Genetic models show that
parathyroid hormone and 1,25-dihydroxyvitamin D3 play
distinct and synergistic roles in postnatal mineral ion
homeostasis and skeletal development. Hum Mol Genet. 2005,
14 (11), 1515–1528. 51. Ferguson, C.M.; Miclau, T.; Hu, D.;
Alpern, E.; Helms, J.A. Common molecular pathways in
skeletal morphogenesis and repair. Ann. N Y Acad. Sci.
1998, 23 (857), 33–42. 52. Crane, G.M.; Ishaug, S.L.;
Mikos, A.G. Bone tissue engineering. Nat. Med. 1995, 1
(12), 1322–1324. 53. Ni, S.; Chang, J.; Chou, L. A novel
bioactive porous CaSiO(3) scaffold for bone tissue
engineering. J. Biomed. Mater. Res. A. 2005, Epub ahead of
print. 54. Causa, F.; Netti, P.A.; Ambrosio, L.; Ciapetti,
G.; Baldini, N.; Pagani, S.; Martini, D.; Giunti, A.
Poly-epsilon-caprolactone/hydroxyapatite composites for
bone regeneration: in vitro characterization and human
osteoblast response. J. Biomed. Mater. Res. A. 2005, Epub
ahead of print. 55. Gonzalez-McQuire, R.; Green, D.; Walsh,
D.; Hall, S.; Chane-Ching, J.Y.; Oreffo, R.O.; Mann, S.
Fabrication of hydroxyapatite sponges by dextran
sulphate/amino acid templating. Biomaterials 2005, 26 (33),
6652–6656. 56. Du, C.; Schneider, G.B.; Zaharias, R.;
Abbott, C.; Seabold, D.; Stanford, C.; Moradian-Oldak, J.
Apatite/ Amelogenin coating on titanium promotes osteogenic
gene expression. J. Dent. Res. 2005, 84 (11), 1070–1074.
57. Michenfelder, M.; Fu, G.; Lawrence, C.; Weaver, J.C.;
Wustman, B.A.; Taranto, L.; Evans, J.S.; Morse, D.E.
Characterization of two molluscan crystal-modulating
biomineralization proteins and identification of putative
mineral binding domains. Biopolymers 2003, 70 (4), 522–533.
58. Swan, E.E.; Popat, K.C.; Grimes, C.A.; Desai, T.A.
Fabrication and evaluation of nanoporous alumina membrane
for osteoblast culture. J. Biomed. Mater. Res. A 2005, 72
(3), 288–295. 59. Ber, S.; Torun Kose, G.; Hasirci, V. Bone
tissue engineering on patterned collagen films: an in vitro
study. Biomaterials 2005, 26 (14), 1977–1986. 60. He, G.;
George, A. Dentin matrix protein 1 immobilized on type I
collagen fibrils facilitates apatite deposition in vitro. J.
Biol. Chem. 2004, 279 (12), 11,649–11,656. 61. Song, J.;
Malathong, V.; Bertozzi, C.R. Mineralization of synthetic
polymer scaffolds: a bottom-up approach for the development
of artificial bone. J. Am. Chem. Soc. 2005, 127 (10),
3366–3372. 62. Kumarasuriyar, A.; Jackson, R.A.; Grondahl,
L.; Trau, M.; Nurcombe, V.; Cool, S.M.
Poly(beta-hydroxybutyrate-co-beta-hydroxyvalerate) supports
in vitro osteogenesis. Tissue Eng. 2005, 11 (7–8),
1281–1295. 63. Jansen, E.J.; Sladek, R.E.; Bahar, H.;
Yaffe, A.; Gijbels, M.J.; Kuijer, R.; Bulstra, S.K.;
Guldemond, N.A.; Binderman, I.; Koole, L.H. Hydrophobicity
as a design criterion for polymer scaffolds in bone tissue
engineering. Biomaterials 2005, 26 (21), 4423–4431. 64.
Hargerink, J.D.; Beniash, E.; Stupp, S.I. Self-assembly and
mineralization of peptide-amphiphile nanofibers. Science
2001, 294 (5547), 1684–1688. 65. Vogt, S.; Berger, S.;
Wilke, I.; Larcher, Y.; Weisser, J.; Schnabelrauch, M.
Design of oligolactone-based scaffolds for bone tissue
engineering. Bio-Medical Materials and Engineering 2005, 15
(1/2), 73–85. 66. Lahiri, J.; Xu, G.; Dabbs, D.M.; Yao, N.;
Aksay, I.A.; Groves, J.T. Prophyrin amphiphiles as
templates for the nucleation of calcium carbonate. J. Am.
Chem. Soc. 1997, 119, 5449–5450. 67. Liang, P.; Shen, Q.;
Zhao, Y.; Zhou, Y.; Wei, H.; Lieberwirth, I.; Huang, Y.;
Wang, D.; Xu, D. Petuniashaped superstructures of CaCO3
aggregates modulated by modified chitosan. Langmuir. 2004 ,
20 (24), 10,444–10,448. 68. Mukherjee, D.P.; Tunkle, A.S.;
Roberts, R.A.; Clavenna, A.; Rogers, S.; Smith, D. An
animal evaluation of a paste of chitosan glutamate and
hydroxyapatite as a synthetic bone graft material. J Biomed
Mater Res B Appl Biomater. 2003, 67 (1), 603–609. 69. Zhao,
F.; Grayson, W.L.; Ma, T.; Bunnell, B.; Lu, W.W. Effects of
hydroxyapatite in 3-D chitosan-gelatin O polymer network on
human mesenchymal stem cell construct development.
Biomaterials 2005, epub ahead of print.

70. Bigi, A.; Torricelli, P.; Fini, M.; Bracci, B.;


Panzavolta, S.; Sturba, L.; Giardino, R. A biomimetic
gelatincalcium phosphate bone cement. Int. J. Artif. Organs
2004, 27 (8), 664–673.

71. Fan, Y.; Duan, K.; Wang, R. A composite coating by


electrolysis-induced collagen self-assembly and calcium
phosphate mineralization. Biomaterials. 2005, 26 (14),
1623–1632.

72. Hempel, U.; Reinstorf, A.; Poppe, M.; Fischer, U.;


Gelinsky, M.; Pompe, W.; Wenzel, K.W. Proliferation and
differentiation of osteoblasts on Biocement D modified with
collagen type I and citric acid. J. Biomed. Mater. Res. B.
Appl. Biomater. 2004, 71 (1), 130–143.

73. Yamauchi, K.; Goda, T.; Takeuchi, N.; Einaga, H.;


Tanabe, T. Preparation of collagen/calcium phosphate
multilayer sheet using enzymatic mineralization.
Biomaterials. 2004, 25 (24), 5481–5489.

74. Mistry, A.S.; Mikos, A.G. Tissue engineering strategies


for bone regeneration. Adv. Biochem. Eng. Biotechnol. 2005,
94, 1–22.

75. Schek, R.M.; Taboas, J.M.; Segvich, S.J.; Hollister,


S.J.; Kresbach, P.H. Engineered osteochondral grafts using
biphasic composite solid free-form fabricated scaffolds.
Tissue Eng. 2004, 10 (9–10), 1376–1385.

76. Holland, T.A.; Bodde, E.W.; Baggett, L.S.; Tabata, Y.;


Mikos, A.G.; Jansen, J.A. Osteochondral repair in the
rabbit model utilizing bilayered, degradable oligo(poly
(ethylene glycol) fumarate) hydrogel scaffolds. J. Biomed.
Mater. Res. A. 2005, 75 (1), 156–167.

77. Logeart-Avramoglou, D.; Anagnostou, F.; Bizios, R.;


Petite, H. Engineering bone: challenges and obstacles. J.
Cell. Mol. Med. 2005, 9 (1), 72–84.

78. Hwang, J.J.; Iyer, S.N.; Li, L.-S.; Claussen, R.;


Harrington, D.A.; Stupp, S.I. Self-assembling biomaterials:
liquid crystal phases of cholesteryl oligo( L-lactic acid)
and their interactions with cells. PNAS 2002, 99 (15),
9662–9667.

79. Stendahl, J.C.; Li, L.; Claussen, R.C.; Stupp, S.I.


Modification of fibrous poly( L-lactic acid) scaffolds with
self-assembling triblock molecules. Biomaterials 2004, 25
(27), 5847–5856.

80. Yu, G.; Ji, J.; Shen, J. Cholesterol tethered poly(


DL-lactic acid) for promoting osteoblast attachment and
growth. J. Bioact. Compat. Polym. 2005, 20 (6), 527–540.

81. Stevens, M.M.; Marini, R.P.; Schaefer, D.; Aronson, J.;


Langer, R.; Shastri, V.P. In vivo engineering of organs:
the bone bioreactor. PNAS USA. 2005, 102 (32), 11450–11455.

82. Cheng, H.; Jiang, W.; Phillips, F.M.; Haydon, R.C.;


Peng, Y.; Zhou, L.; Luu, H.H.; An, N.; Breyer, B.;
Vanichakarn, P.; Szatkowski, J.P.; Park, J.Y.; He, T.C.
Osteogenic activity of the fourteen types of human bone
morphogenetic proteins (BMPs). J. Bone Joint Surg. Am.
2003, 85-A (8), 1544–1552.

83. Butler, W.T.; Brunn, J.C.; Qin, C. Dentin extracellular


matrix (ECM) proteins: comparison to bone ECM and
contribution to dynamics of dentinogenesis. Connect. Tissue
Res. 2003, 44 (Suppl 1), 171–178. 84. Butler, W.T.;
Ritchie, H. The nature and functional significance of dentin
extracellular matrix proteins. Int. J. Dev. Biol. 1995, 39
(1), 169–179. 85. Butler, W.T.; Brunn, J.C.; Qin, C.;
McKee, M.D. Extracellular matrix proteins and the dynamics
of dentin formation. Connect. Tissue Res. 2002, 43 (2–3),
301–317. 86. Sreenath, T.; Thyagarajan, T.; Hall, B.;
Longenecker, G.; DuSouza, R.; Hong, S.; Wright, J.T.;
MacDougall, M.; Sauk, J.; Kulkarni, A.B. Dentin
sialophosphoprotein knockout mouse teeth display widened
predentin zone and develop defective dentin mineralization
similar to human dentinogenesis imperfecta type III. J.
Biol. Chem. 2003, 278 (27), 24,874–24,880. 87. Sharpe,
P.T.; Young, C.S. Test-tube teeth. Scientific American 2005,
293 (2), 34–41. 88. Verdelis, K.; Crenshaw, M.A.;
Paschalis, E.P.; Doty, S.; Atti, E.; Boskey, A.L.
Spectroscopic imaging of mineral maturation in bovine
dentin. J. Dent. Res. 2003, 82, 697–702. 89. Iejima, D.;
Saito, T.; Uemura, T. A collagen-phosphophoryn sponge as a
scaffold for bone tissue engineering. J. Biomater. Sci.
Polym. Ed. 2003, 14 (10), 1097–1103. 90. He, G.; Dahl, T.;
Veis, A.; George, A. Nucleation of apatite crystals in
vitro by self-assembled dentin matrix protein 1. Nat.
Mater. 2003, 2 (8), 552–558. 91. Young, C.S.; Kim, S.W.;
Qin, C.; Baba, O.; Butler, W.T.; Taylor, R.R.; Bartlett,
J.D.; Vacanti, J.P.; Yelick, P.C. Developmental analysis
and computer modelling of bioengineered teeth. Arch. Oral.
Biol. 2005, 50 (2), 259–265. 92. Deng, M.; Shi, J.; Smith,
A.J.; Jin, Y. Effects of transforming growth factor beta1
(TGFbeta-1) and dentin non-collagenous proteins (DNCP) on
human embryonic ectomesenchymal cells in a
three-dimensional culture system. Arch. Oral. Biol. 2005,
50 (11), 937–945. 93. Giannobile, W.V.; Somerman, M.J.
Growth and amelogenin-like factors in periodontal wound
healing. A systematic review. Ann. Periodontol. 2003, 8
(1), 193–204. 94. Zhao, M.; Jin, Q.; Berry, J.E.; Nociti,
F.H., Jr.; Giannobile, W.V.; Somerman, M.J. Cementoblast
delivery for periodontal tissue engineering. J.
Periodontol. 2004, 75 (1), 154–61. 95. Bosshardt, D.D. Are
cementoblasts a subpopulation of osteoblasts or a unique
phenotype?. J. Dent. Res. 2005, 84 (5), 390–406. 96.
Weatherell, J.A.; Robinson, C.; Hallsworth, A.S. Variations
in the chemical composition of human enamel. J. Dent. Res.
1974, 53 (2), 180–192. 97. Aoba, T.; Komatsu, H.; Shimazu,
Y.; Yagishita, H.; Taya, Y. Enamel mineralization and an
initial crystalline phase. Connect Tissue Res. 1998, 38
(1–4), 129–137. 98. Deutsch, D.; Dafni, L.; Palmon, A.;
Hekmati, M.; Young, M.F.; Fisher, L.W. Tuftelin: enamel
mineralization and amelogenesis imperfecta. Ciba Found
Symp. 1997, 205, 135–47. 99. Kirkham, J.; Brookes, S.J.;
Shore, R.C.; Bonass, W.A.; Smith, D.A.; Wallwork, M.L.;
Robinson, C. Atomic force microscopy studies of crystal
surface topology during enamel development. Connect. Tissue
Res. 1998, 38 (1–4), 91–100. 100. Robinson, C.; Kirkham,
J.; Brookes, S.J.; Bonass, W.A.; Shore, R.C. The chemistry
of enamel development. Int. J. Dev. Biol. 1995, 39 (1),
145–52. 101. Du, C.; Falini, G.; Fermani, S.; Abbott, C.;
MoradianOldak, J. Supramolecular assembly of amelogenin
nanospheres into birefringent microribbons. Science 2005,
307 (5714), 1450–1454. 102. Sire, J.Y.; Delgado, S.;
Fromentin, D.; Girondot, M. Amelogenin: lessons from
evolution. Arch. Oral. Biol. 2005, 50 (2), 205–212. 103.
Beniash, E.; Simmer, J.P.; Margolis, H.C. The effect of
recombinant mouse amelogenins on the formation and
organization of hydroxyapatite crystals in vitro. J.
Struct. Biol. 2005, 149 (2), 182–190. 104. Devecioglu, D.;
Tozum, T.F.; Sengun, D.; Nohutcu, R.M. Biomaterials in
periodontal regenerative surgery: effects of cryopreserved
bone, commercially available coral, demineralized
freeze-dried dentin, and cementum on periodontal ligament
fibroblasts and osteoblasts. J. Biomater. Appl. 2004, 19
(2), 107–120. 105. Chen, H.; Clarkson, B.H.; Sun, K.;
Mansfield, J.F. Self-assembly of synthetic hydroxyapatite
nanorods into an enamel prism-like structure. J. Colloid
Interface Sci. 2005, 288 (1), 97–103. 106. Wilt, F.H.
Developmental biology meets materials science:
morphogenesis of biomineralized structures. Dev. Biol.
2005, 280 (1), 15–25. 107. Krampitz, G.; Witt, W.
Biochemical aspects of biomineralization. Top. Curr. Chem.
1979, 78, 57–144. 108. Albeck, S.; Addadi, I.; Weiner, S.
Regulation of calcite crystal morphology by
intracrystalline acidic proteins and glycoproteins.
Connect. Tissue Res. 1996, 35 (1–4), 365–370. 109. Falini,
G.; Fermani, S. Chitin mineralization. Tissue Eng. 2004, 10
(1–2), 1–6. 110. Wright, G.M.; Keeley, F.W.; Robson, P. The
unusual cartilaginous tissues of jawless craniates,
cephalochordates and invertebrates. Cell Tissue Res. 2001,
04 (2), 165–174. 111. Wustman, B.A.; Morse, D.E.; Evans,
J.S. Structural characterization of the N-terminal mineral
modification domains from the molluscan crystal-modulating
biomineralization proteins, AP7 and AP24. Biopolymers 2004,
74 (5), 363–376. 112. Wustman, B.A.; Weaver, J.C.; Morse,
D.E.; Evans, J.S. Structure-function studies of the lustrin
A polyelectrolyte domains, RKSY and D4. Connect Tissue Res.
2003, 44 (Suppl. 1), 10–15. 113. Thompson, J.B.; Paloczi,
G.T.; Kindt, J.H.; Michenfelder, M.; Smith, B.L.; Stucky,
G.; Morse, D.E.; Hansma, P.K. Direct observation of the
transition from calcite to aragonite growth as induced by
abalone shell proteins. Biophys. J. 2000, 79 (6), 3307–
3312. 114. Walters, D.A.; Smith, B.L.; Belcher, A.M.;
Paloczi, G.T.; Stucky, G.D.; Morse, D.E.; Hansma, P.K.
Modification of calcite crystal growth by abalone shell
proteins: an atomic force microscope study. Biophys. J.
1997, 72 (3), 1425–1433. 115. Weiner, S.; Hood, L. Soluble
protein of the organic matrix of mollusk shells: a
potential template for shell formation. Science 1975,
(4218), 987–989. 116. Chandy, T.; Sharma, C.P. Chitosan–as
a biomaterial. Biomater. Artif. Cells. Artif. Organs. 1990,
18 (1), 1–24. 117. Aimin, C.; Chunlin, H.; Juliang, B.;
Tinyin, Z.; Zhichao, D. Antibiotic loaded chitosan bar. An
in vitro, in vivo study of a possible treatment for
osteomyelitis. Clin. Orthop. Relat. Res. 1999, 366,
239–247. 118. Chen, F.; Chen, S.; Tao, K.; Feng, X.; Liu,
Y.; Lei, D.; Mao, T. Marrow-derived osteoblasts seeded into
porous natural coral to prefabricate a vascularised bone
graft in the shape of a human mandibular ramus:
experimental study in rabbits. Br. J. Oral Maxillofac.
Surg. 2004, 42 (6), 532–537. 119. Martina, M.; Subramanyam,
G.; Weaver, J.C.; Hutmacher, D.W.; Morse, D.E.;
Valiyaveettil, S. Developing macroporous bicontinuous
materials as scaffolds for tissue engineering.
Biomaterials. 2005, 26 (28), 5609–5616. 120. Green, D.;
Walsh, D.; Mann, S.; Oreffo, R.O. The potential of
biomimesis in bone tissue engineering: lessons from the
design and synthesis of invertebrate skeletons. Bone 2002,
30 (6), 810–815. O
Orthodontic Wires

10. Flinn, R.A.; Trojan, P.K. Engineering Materials and


Their Applications, 3rd Ed.; Houghton Mifflin: Boston, 1986;
530–531.

11. Budinski, K.G. Engineering Materials: Properties and


Selection, 5th Ed.; Prentice Hall: Englewood Cliffs, NJ,
1996; 418–420.

12. Backofen, W.A.; Gales, G.F. Heat treating stainless


steel wire for orthodontics. Am. J. Orthod. 1952, 38,
755–765.

13. Goldberg, A.J.; Burstone, C.J. An evaluation of beta


titanium alloys for use in orthodontic appliances. J. Dent.
Res. 1979, 58, 593–600.

14. Fillmore, G.M.; Tomlinson, J.L. Heat treatment of


Cobalt-Chromium wire. Angle Orthod. 1976, 46, 187–195.

15. Fillmore, G.M.; Tomlinson, J.L. Heat treatment of


Cobalt-Chromium alloys of various tempers. Angle Orthod.
1979, 49, 126–130.

16. Nelson, K.R.; Burstone, C.J.; Goldberg, A.J. Optimal


welding of b titanium orthodontic wires. Am. J. Orthod.
Dentofac. Orthop. 1987, 92, 213–219.

17. Kusy, R.P.; Whitley, J.Q. Effects of sliding velocity


on the coefficients of friction in a model orthodontic
system. Dent. Mater. 1989, 5, 235–240.

18. Bradley, T.G.; Mitchell, J.C. Brantley, Surface


Composition and Microtopography of nickel-titanium
Orthodontic Wires. In Special Issue— J Dent Res, 74th
General Session and Exhibition of The International
Association for Dental Research, San Francisco, CA,
March13–17, Herzberg, M.C., Ed.; American Association for
Dental Research: Alexandria, VA, 1996; 61. 19. Kula, K.;
Phillips, C.; Gibilaro, A.; Proffit, W.R. Effect of ion
implantation of TMA archwires on the rate of orthodontic
sliding space closure. Am. J. Orthod. Dentofac. Orthop.
1998, 114, 577–580. 20. Buehler, W.J.; Wiley, R.C.
TiNi-ductile intermetallic compound. Trans. Aktueller
Softw.-Markt 1962, 55, 269–276. 21. Andreasen, G.F.;
Hilleman, T.B. An evaluation of 55 cobalt substituted
nitinol wire for use in orthodontics. J. Am. Dent. Assoc.
1971, 82, 1373–1375. 22. Goldberg, A.J.; Morton, J.;
Burstone, C.J. The flexure modulus of elasticity of
orthodontic wires. J. Dent. Res. 1983, 62, 856–858. 23.
Andreasen, G.F.; Morrow, R.E. Laboratory and clinical
analyses of nitinol wire. Am. J. Orthod. 1978, 73, 142–151.
24. Burstone, C.J.; Qin, B.; Morton, J.Y. Chinese NiTi
wire: A new orthodontic alloy. Am. J. Orthod. 1985 , 87,
445–452. 25. Miura, F.; Mogi, M.; Ohura, Y.; Hamanaka, H.
The super-elastic property of Japanese NiTi alloy wire for
use in orthodontics. Am. J. Orthod. Dentofac. Orthop. 1986,
90, 1–10. 26. Meling, T.R.; Ødegaard, J. The effect of
short-term temperature changes on superelastic
nickel-titanium archwires activated in orthodontic bending.
Am. J. Orthod. Dentofac. Orthop. 2001, 119, 263–273. 27.
Copper Ni-Ti Wire to Wire Consistency; Ormco: Orange, CA,
1998. Promotional literature.
Orthopedic Biomaterials

10. Mayer, M.H.; Hollinger, J.O. Biodegradable Bone


Fixation Devices. In Biomedical Applications of Synthetic
Biodegradable Polymers; Hollinger, J., Ed.; CRC Press: Boca
Raton, 1995; 173–195.

11. Self-Reinforced Bioabsorbable Polymeric Composites in


Surgery; Rokkamen, P., To¨rma¨la¨, P., Eds. Tampereen,
Pikakapio: Tampere, Finland, 1995.

12. Incardona, S.D.; Fambri, L.; Migliaresi, C.; University


of Trento, Italy. Poly-L-Lactic acid braided fibres produced
by melt spinning: Characterization and in vitro
degradation. J. Mater. Sci., Mater. Med. 1996, 7 (7),
387–391.

13. Athanasiou, K.A.; Niederauer, G.G.; Agrawal, C.M.


Sterilization, toxicity, biocompatibility and clinical
applications of polylactic acid/polyglycolic acid
copolymers. Biomaterials 1996, 17, 93–102.

14. Bronzino, J.D.; Joseph, D. The Biomedical Engineering


Handbook, 2nd Ed.; CRC Press: Boca Raton, FL, 2000; Vol. 1,
39.

15. Bostman, O.M. Osteolytic changes accompanying


degradation of absorbable fracture fixation implants. J.
Bone Jt. Surg., Br. 1991, 73-B (4), 679–682. David L.
Helfet— Advances in Orthopedic Surgery, 16 (5), 335–337,
March/April 1993.

16. Pietrzak, W.S.; Eppley, B.L. Resorbable polymer fixation


for craniomaxillofacial surgery: Development and
engineering paradigms. J. Craniofac. Surg. 2000, 11,
575–585.

17. Ratner, B.D.; et al. Biomaterials Science an


Introduction to Materials in Medicine; Academic Press;
1996.

18. den Dunnen, W.F.A.; et al. Biological performance of a


degradable poly(lactic acid-e-caprolactone) nerve guide:
Influence of tube dimensions. J. Biomed. Mater. Res. 1995,
29 (6), 757–776.

19. Nakamura, T.; et al. Biodegradation and tumorigenicity


of implanted plates made from a copolymer of e-caprolactone
and L-lactide. J. Biomed. Mater. Res. 1998, 42 (4),
475–484.
20. Barrows, T.H. Synthetic Bioabsorbable Polymers. In High
Performance Biomaterials; Szycher, M., Ed.; Technomic
Publishing Co.: Lancaster, 1991; 243–257.

21. Wenger, K.H.; Univ. Utah School of Med. Changes in


Mechanical Properties of Three Bioabsorbable Fracture
Fixation Pin Types Due to Canine Implantation, 40th Annual
Meeting, Orthopedic Research Society, New Orleans, Feb.
21–24, 1994; 538.

22. Daniels, A.U.; Andriano, K.P.; Smutz, W.P.; Chang,


M.K.O.; Heller, J. Evaluation of absorbable poly(ortho
Esters) for use in surgical implants. J. Appl. Biomater.
1994, 5 (1), 51–64.

23. Rawlings, C.E., III. Modem bone substitutes with


emphasis on calcium phosphate ceramics and osteoinductors.
Neurosurgery 1993, 33 (5), 935–938.

24. Rawlings, C.E., III. Modem bone substitutes with


emphasis on calcium phosphate ceramics and osteoinductors.
J. Neurosurg. 1992, 76, 275–279.

25. Behravesh, E.; et al. Synthetic biodegradable polymers


for orthopedic applications. Clin. Orthop. Relat. Res.
1999, 367s, s118–s125.

26. Attawia, M.A.; Uhrich, K.E.; Botchwey, E.; Fan, M.;


Langer, R.; Laurencin, C.T. Cytotoxicity testing of
poly(anhydride-co-imides) for orthopedic applications. J.
Biomed. Mater. Res. 1995, 29 (10), 1222–1240. 27. Gurav,
N.; Downes, S. A qualitative in vitro evaluation of the
degradable materials poly(caprolactone), poly
(hydroxybutyrate) and a
poly(hydroxybutyrate)-(hydroxyvalerate) copolymer. J.
Mater. Sci., Mater. Med. 1994, 5 (11), 784–787. 28. Lee,
K.Y.; Ha, W.S.; Park, W.H. Blood compatibility and
biodegradability of partially N-acylated chitosan
derivatives. Biomaterials 1995, 16 (16), 1210–1216. 29.
Radder, A.M.; Leenders, H.; van Blitterswijk, C.A.
Interface reactions to PEO/PBT copolymers (Polyactive reg )
after implantation in cortical bone. J. Biomed. Mater. Res.
1994, 28 (2), 141–151. (Also similar paper pp. 269–277).
30. Laurencin, C.T.; Norman, M.E.; Elgendy, H.M.; El-Amin,
S.F.; Allcock, H.R.; Rucher, S.R.; Ambriosio, A.A. Use of
polyphosphazenes for skeletal tissue regeneration. J.
Biomed. Mater. Res. 1993, 27 (7), 963–973. 31. Kettumen,
J.; Makela, A.; Miettinen, H.; Nevalainen, T.; Heikkila,
M.; Tormaua, P.; Rokkanen, P. The effect of an
intramedullary carbon-fiber-reinforced liquid crystalline
polymer impant on bone: An experimental study on rabbits.
J. Biomed. Mater. Res. 1998, 42 (3), 407–411. [Finland].
32. Tulles, G.L.; Cassidy, P.E.; St. Clair, A.K. Polymers
derived from hexafluoroacetone: 12F-poly(ether ketone).
Macromolecules 1991, 24, 6059–6064. 33. Nguyen, H.X.;
Ishida, H. Poly(aryl-ether-ether-ketone) and its advanced
composites: A review. Polym. Compos. 1987, 8 (2), 57–73.
34. Johnson, R.N.; Farnham, A.G.; Clendinning, R.A.; Hale,
W.F.; Merriam, C.N. . J. Polym. Sci., Polym. Chem. Ed.
1967, 15, 2375–2398. 35. Klawitter, J.J.; Hulbert, S.F.
Tissue reaction to three ceramics of porous and nonporous
structures. J. Biomed. Res. 1972, 6 (5), 347–374. 36.
Haher, T. New and Transfer Technology, Materials; Spinal
Instrumentation from Harrington to the Future, Continuing
Education Course by AAOS and SRS, Kansas City, Kansas,
September 23, 1992. 37. Dhert, W.J.A.; Klein, C.P.A.T.;
Jansen, J.A.; van der Velde, E.A.; Vriesde, R.C.; Rozing,
P.M.; de Groot, K. A histological and histomorphometrical
investigation of fluorapatite, magnesium whitlockite, and
hydroxylapatite plasma-sprayed coatings in goats. J.
Biomed. Mater. Res. 1993, 27 (1), 127–138. 38. Radin, S.R.;
Ducheyne, P. The effect of varying the calcium phosphate
composition and structure on dissolution and precipitation
reactions in simulated interstitial fluid. Trans. Soc.
Biomater. May 1–5, 1991, XIV, 193. 39. Denissen, H.W.; et
al. Stable loaded HA coatings in comparison with
experimental plasma sprayed alternatives. Trans. Soc.
Biomater.May 1–5, 1991, XIV, 189. 40. Wiltfang, J.; et al.
Degradation characteristics of a and b
tri-calcium-phosphate (TCP) in minipigs. J. Biomed. Mater.
Res. (Appl. Biomater.) 2002, 63 (2), 115–121. (Germany).
41. Daculsi, G.; LeGerros, R.Z.; Nery, E.; Lynch, K.;
Kerebel, B. Transformation of biphasic calcium phosphate
ceramics in vivo: Ultrastructural and physiochemical
characterization. J. Biomed. Mater. Res. 1989, 23 (8),
883–894. 42. Nicholas, R.W.; Lange, T.A.; St. Paul-Ramsey
Medical Center, St. Paul, MN. Granular tricalcium phosphate
grafting of nonstructural cavitary bone lesions in humans.
Trans. Soc. Biomater.May 1–5, 1991, XIV, 8. 43. Passuti,
N.; Daculsi, G.; Martin, S.; Deudon, C. Macroporous
Polycrystalline Calcium Phosphate Implant for Spinal Fusion
in Man and Dogs. In Handbook of Bioactive Ceramics, Volume
II—Calcium Phosphate and Hydroxylapatite Ceramics;
Yamamuro, T., Hench, L., Wilson, J., Eds.; CRC Press: Boca
Raton, 1990; 345–354. 44. Chuckler, J.M.; Ducheyne, P.;
Nangle, A.E. The Effect of Osteoporosis on Skeletal
Fixation of Porous and Calcium Phosphate Coated Implants,
Transactions of Fourth World Biomaterials Congress, Berlin,
Germany, April 24–28, 1992; 97. 45. Guigui, P.; Plais,
P.Y.; Flautre, B.; Viguier, E.; Blary, M.C.; Chopin, D.;
Lavaste, F.; Hardouin, P. Experimental model of
posterolateral spinal arthrodesis in sheep: Part 2.
Application of the model: Evaluation of vertebral fusion
obtained with coral (porites) or with a biphasic ceramic
(triosite). Spine 1994, 19 (24), 2798–2803. (France). 46.
Berger, G.; Gildehaar, R.; Ploska, U. Rapid resorbable
glassy crystalline materials on the basis of calcium alkali
orthophosphates. Biomaterials 1995, 16 (16), 1241–1248. 47.
Oonishi, H. Bioceramics in Orthopedic Surgery—Our Clinical
Experiences. In Bioceramics; Hulbert, S., Ed.; Rose-Hulman
Institute of Technology, 1992; Vol. 3, 31–42. 48. Ishihara,
K.; Arai, H.; Nakabayashi, N. Adhesive bone cement
containing hydroxyapatite particle as bone compatible
filler. J. Biomed. Mater. Res. 1992, 26, 937–945. 49. Dhert,
W.J.A.; de Groot, K.; et al. Biomaterials Research Group,
Departments of Biomaterials and Orthopaedics, School of
Medicine, University of Leiden, The Netherlands The
response of trabecular bone to fluorapatite, magnesium
whitlockite, and hydroxylapatite plasma-spray coated
implants, an experimental study in goats. Trans. Soc.
Biomater. May 1–5, 1991, XIV, 3. 50. Kangasniemi, I.;
Verheyen, C.C.P.M.; Wolke, J.G.C.; de Groot, K. Tensile
Test of the Bone Bonding Strength to Fluorand
Hydroxylapatite and Titanium, Transactions of the Fourth
World Biomaterials Congress, Berlin, Germany, April 24–28,
1992; 99. 51. Hotter, D.S. Band-aids for broken bones.
Mach. Des. April 4, 1996, 39–44. 52. Neo, M.; Nakamura, T.;
Ohtsuki, C.; Kokubo, T.; Yamamuro, T. Apatite formation on
three kinds of bioactive material at an early stage in
vivo: A comparative study by transmission electron
microscopy. J. Biomed. Mater. Res. 1993, 27 (8), 999–1006.
53. David, A.; Eitenmuller, J.; Hangst, K.; Bar, H.F.;
Muhr, G. Bone-Defect Substitution in Load Bearing Skeletal
Segments by Titanium and HydroxyapatiteCoated
Titanium-Implants, Transactions of the Fourth World
Biomaterials Congress, Berlin, Germany, April 24–28, 1992;
93. 54. Sanden, B.; Olerud, C.; Johansson, C.; Larsson, S.
Improved extraction torque of hydroxyapatite-coated pedicle
screws. E. Spine J. 2000, 9. pp. 6, 534–537. 55. Graziano,
G.P.; More, D.C.; Maitra, R.S.; Farjo, L.A.; Goldstein,
S.A. Calcium Phosphate Ceramic vs. PMMA for the
Augmentation of Transpedicular Screws, Scoliosis Research
Society, 28th Annual Meeting, Dublin, Ireland, September
18–23, 1993, 92. Exhibit #43. 56. Frankenburg, E.P.;
Goldstein, S.A.; Harris, S.A.; Toomajian, L.R.; Pellizzon,
G.G.; Balker, D.A.; Bauer, T.W.; Orthop. Res. Lab. U. Mich.
and CCF, Cleveland Evaluation of an in situ-setting calcium
phosphate grout in an in vivo canine metaphyseal defect
model. Orthop. Trans. 1994, 18 (2), 413. 57. Caddes, D.E.;
Norian Corporation. Novel bone repair biomaterials. Orthop.
Trans. 1994, 18 (2), 3C–22C. 58. Orthop. Today 1995, 15
(5), 10. 59. Yoshikawa, T.; et al. Biochemical Sequence of
Bone Formation in Porous Ceramics. In Bioceramics; Hulbert,
S., Ed.; Rose-Hulman Institute of Technology, 1992; Vol. 3;
157–169. 60. Katz, J.L. On the anisotropy of Young’s
modulus of bone. Nature 1980, 283, 106–107. 61. Damien,
C.J.; Parsons, J.R. Bone graft and bone graft substitutes:
A review of current technology and applications. J. Appl.
Biomater. 1991, 2 (3), 187–208. 62. Nathan, R.; et al.
Osteogenesis in a Rat Femoral Defect with a
Collagen-Ceramic Composite Containing Bone Marrow,
Transactions of the 37th Annual Meeting, Orthopedic
Research Society, Anaheim, CA, March 4–7, 1991; 406. 63.
St. John, K.R. Histological and electron microscopic
analysis of tissue response to synthetic composite bone
graft in the canine. J. Appl. Biomater. 1995, 6 (2), 89–97.
64. Muschler, G.F.; Huber, B.; Ullman, T.; Barth, R.;
Easley, K.; Otis, J.O.; Lane, J.M. Evaluation of
bonegrafting materials in a new canine segmental spinal
fusion model. J. Orthop. Res. 1993, 11 (4), 514–524. 65.
Stupp, S.I. Organoapatites: Materials for artificial bone.
II. Hardening reactions and properties. J. Biomed. Mater.
Res. 1993, 27 (3), 289–299. 66. Smith & nephew licenses
composite from BTG. Clinica April 3, 1995, 648, 14.
Additional reference on page 17. 67. Witschger, P. New bone
joint stronger than natural bone. Jpn. Chem. Week Oct. 3,
1996, 3. 68. Witschger, P. New bone joint stronger than
natural bone. Comline Biotechnol. Med. Oct. 9, 1996. 69.
Neve, A.D.; Piddock, V.; Combe, E.C. Development of novel
dental cements. I & II. Clin. Mater. 1992, 9, 7–20. 70.
Jones, D.W.; Rizkalla, A.S.; Sutow, E.J.; Hall, G.C.
Influence of Composition on Mechanical Properties of
Polyalkenoate Cements, Transactions of the Fourth World
Biomaterials Congress, Berlin, Germany, April 24–28, 1992;
91. 71. Rokkanen, P.; Bostman, O.; Vainionpaa, S.; et al.
Biodegradable implants in fracture fixation: Early results
of treatment of fractures of the ankle. Lancet 1985, 1,
1422–1424. O

72. Rokkanen, P.U. Bioabsorbable fixation devices in


orthopaedics and traumatology. Ann. Chir. Gynaecol. 1998,
87, 13–20.

73. Bostman, O.; Vainionpaa, S.; Hirvensalo, E.; et al.


Biodegradable internal fixation for malleolar fractures. A
prospective randomized trial. J. Bone Jt. Surg., Br. 1987,
69, 516–619.

74. Peltoniemi, H.H.; Tulamo, R.M.; Toivonen, T.; et al.


Biodegradable semirigid plate and miniscrew fixation
compared with rigid titanium fixation in experimental
calvarial osteotomy. J. Neurosurg. 1999, 90, 910–917.

75. Yamamuro, T.; Matsusue, Y.; Uchida, A.; et al.


Bioabsorbable osteosynthetic implants of ultra high
strength poly-L-lactide. A clinical study. Int. Orthop.
1994, 18, 332–340.

76. Maruyama, M.; Terayama, K.; Itou, M.; Yamagishi, T.


Experimental Development of a Chitosan-Bonded
Hydroxyapatite Self-Hardening Paste, Transactions of the
Fourth World Biomaterials Congress, Berlin, Germany, April
24–28, 1992; 93.

77. Ito, M.; Miyazaki, A.; Yamagishi, T.; Yagasaki, H.;


Matsumoto Dental College, Nagano Japan Experimental
development of a chitosan-bonded b-tricalcium phosphate
bone filling paste. Bio-Med. Mater. Eng. 1994, 4 (6),
439–449.

78. Daniel, A.U.; Chang, M.K.O.; Andriano, K.P. Mechanical


properties of biodegradable polymers and composites
proposed for internal fixation of bone. J. Appl. Biomater.
1990, 1 (1), 57–78.

79. Gogolewski, S.; Jovanovic, M.; Perren, S.M.; Dillon,


J.G.; Hughes, M.K. Tissue response and in vivo degradation
of selected polyhydroxyacids: Polylactides (PLA),
poly(3-hydroxybutyrate) (PHB), and poly
(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHB/VA). J.
Biomed. Mater. Res. 1993, 27 (9), 1135–1148.

80. Barrows, T.H. Semiabsorbable Bone Plate Spacer. US


Patent 5,013,315, filed 12 January 1985, assigned to
Minnesota Mining and Manufacturing Company, issued 7 May
1991.

81. Yoda, R.; Komatsuzaki, S.; Hayashi, T. Surface


properties and biocmpatibility of A-B-A type block
copolymer membranes consisting of
poly(g-benzyl-L-glutamate) as the A Component and
Polyisoprene as the B Component. Biomaterials 1995, 16
(16), 1203–1210.

82. Ertel, S.I.; Joachim, K.Z.; Zimmerman, M.C.; Parsons,


J.R. Evaluation of poly(DTH carbonate), a tyrosinederived
degradable polymer, for orthopedic applications. J. Biomed.
Mater. Res. 1995, 29 (11), 1337–1348.
83. Storey, R.F.; Wiggins, J.S. Design and fabrication of
polyester-fiber and matrix composites for totally absorbable
biomaterials. Med. Plast. Biomater. 1996, 3 (2), 41–44.

84. Kadiyala, S.; Lo, H.; Ponticiello, M.S.; Leong, K.W.


Poly(phosphoesters): Synthesis, Physio-chemical
Characterization and Biological Response. In Biomedical
Applications of Synthetic Biodegradable Polymers;
Hollinger, J.O., Ed.; CRC Press, 1995; 33–57. Chapter 3.
85. Chvapil, M.; et al. Collagen fibers as a temporary
scaffold for replacement of ACL in goats. J. Biomed. Mater.
Res. 1993, 27 (3), 313–325. 86. Bulstra, S.; et al.
Analysis of the Osteoconductive Properties of Polyactive in
the Rabbit Femur by Dual Energy X-Ray Absorptiometric
Analysis and Quantitative Histological Analysis, 40th
Annual Meeting, Orthopedic Research Society, New Orleans,
Feb. 21–24, 1994; 515. 87. Whitecloud, T.S.; Cook, S.D.;
Dalton, J.E.; Prewett, A. Reduction in Perineural Scar
Formation Following Laminectomy with Polyactive Membrane
Sheets, 40th Annual Meeting, Orthopedic Research Society,
New Orleans, Feb. 21–24, 1994; 711. 88. Cook, S.D.;
Prewett, A.B.; Dalton, J.E.; Whitecloud, T.S., III.
Reduction in perineural scar formation after laminectomy
with polyactive membrane sheets. Spine 1994, 19 (16),
1815–1825. 89. Conversation of Tom Sander of Polymedics
(partnership of U.S. Surgical and Biomet), Warsaw, IN {219
267-6639x1663} with CRM on 6 September 1994. 90. Damien,
C.J. A Comparative Analysis of Various Bone Graft
Substitutes. Ph.D. Thesis; Rutgers The State University of
New Jersey: New Brunswick, 1990. U.M.D.N.J. 91. Cifkova,
I.; Lopour, P.; Vondracek, P.; Jelinek, F. Silicone
rubber-hydrogel composites as polymeric biomaterials. I.
Biological properties of the silicone rubber-p(HEMA)
composite. Biomaterials 1990, 11 (6), 393–396. 92. Lopour,
P.; Vondracek, P.; Janatova, V.; Sulc, J.; Vacik, J.
Silicone rubber-hydrogel composites as polymeric
biomaterials. II. Hydrophilicity and permeability to
water-soluble low-molecular-weight compounds. Biomaterials
1990, 11 (6), 397–402. 93. Reissis, N.; Downes, S.;
Bentley, G.; IRC Biomedical Materials, Middlesex UK.
Cartilage Repair Using a New Synthetic Polymer, 40th Annual
Meeting, Orthopedic Research Society, New Orleans, Feb.
21–24, 1994; 482. 94. Hydrophilic-hydrophobic binary
systems of poly(2hydroxyethyl methacrylate) and
polycaprolactone. Part I: Synthesis and characterization.
J. Bioact. Compat. Polym. January 1994, 9. Also ‘‘Part II:
Degradation’’ in April 1994. 95. Zachariades, A.E. A New
Class of UHMWPE Orthopedic Prosthetic Devices with Enhanced
Mechanical Properties, Transactions of Fourth World
Biomaterials Congress, Berlin, April 24–28, 1992; 623. 96.
Kerns, L. Clinical trials to begin for injectable
fracturehealing bone mineral substitute. Orthop. Today
1995, 15 (2), 23–24.
Orthopedic Fixation Devices

37. Fleming, B.; Paley, D.; Kristiansen, T.; Pope, M. A


biomechanical analysis of the Ilizarov external fixator.
Clin. Orthop. 1989, 241, 95–105.

38. Behrens, F. General theory and principles of external


fixation. Clin. Orthop. 1989, 241, 15–23.

39. Behrens, F. A primer of fixator devices and


configurations. Clin. Orthop. 1989, 241, 5–14.

40. Duda, G.N.; Sollmann, M.; Sporrer, S.; Hoffman, J.E.;


Kassi, J.-P.; Khodadadyan, C.; Raschke, M. Interfragmentary
motion in tibial osteotomies stabilized with ring fixators.
Clin. Orthop. 2002, 396, 163–172.

41. Paley, D.; Catagni, M.A.; Argnani, F.; Villa, A.;


Benedetti, G.B.; Cattaneo, R. Ilizarov treatment of tibial
nonunions with bone loss. Clin. Orthop. 1989, 241, 146–165.
42. Schwartsman, V.; Choi, S.H.; Schwartsman, R. Tibial
nonunions: Treatment tactics with the Ilizarov method.
Orthop. Clin. 1990, 21 (4), 639–653. 43. Dagher, F.;
Roukoz, S. Compound tibial fractures with bone loss treated
by the Ilizarov technique. J. Bone Joint Surg., Br 1991,
73-B, 316–321. 44. Shtarker, H.; Volpin, G.; Stolero, J.;
Kaushansky, A.; Samchukov, M. Correction of combined
angular and rotational deformities by the Ilizarov method.
Clin. Orthop. 2002, 402, 184–195. 45. Weber, B.G. On the
Biomechanics of External Fixation. In The External Fixator;
Weber, B.G., Magerl, F., Eds.; Springer-Verlag: Berlin,
1985; 27–53.
Osteoclastic Bone Resorption

1. Lakkakorpi, P.T.; Vaananen, H.K. Cytoskeletal changes in


osteoclasts during the resorption cycle. Microsc. Res.
Tech. 1996, 33, 171–181. 2. Nesbitt, S.; Nesbit, A.;
Helfrich, M.; Horton, M.; et al. Biochemical
characterization of human osteoclast integrins. Osteoclasts
express alpha v beta 3, alpha 2 beta 1, and alpha v beta 1
integrins. J. Biol. Chem. 1993, 268, 16737–16745. 3. Ross,
F.P.; Chappel, J.; Alvarez, J.I.; et al. Interactions
between the bone matrix proteins osteopontin and bone
sialoprotein and the osteoclast integrin alpha v beta 3
potentiate bone resorption. J. Biol. Chem. 1993, 268,
9901–9907. 4. Helfrich, M.H.; Nesbitt, S.A.; Lakkakorpi,
P.T.; et al. Beta 1 integrins and osteoclast function:
involvement in collagen recognition and bone resorption.
Bone 1996, 19, 317–328. 5. Fisher, J.E.; Caulfield, M.P.;
Sato, M.; et al. Inhibition of osteoclastic bone resorption
in vivo by echistatin, an ‘‘arginyl-glycyl-aspartyl’’
(RGD)-containing protein. Endocrinology 1993, 132,
1411–1413. 6. Horton, M.A.; Taylor, M.L.; Arnett, T.R.;
Helfrich, M.H. Arg-Gly-Asp (RGD) peptides and the
antivitronectin receptor antibody 23C6 inhibit dentine
resorption and cell spreading by osteoclasts. Exp. Cell.
Res. 1991, 195, 368–375. 7. Felding-Habermann, B.; Cheresh,
D.A. Vitronectin and its receptors. Curr. Opin. Cell Biol.
1993, 5, 864–868. 8. Helfrich, M.H.; Nesbitt, S.A.; Dorey,
E.L.; Horton, M.A. Rat osteoclasts adhere to a wide range
of RGD (Arg-Gly-Asp) peptide-containing proteins, including
the bone sialoproteins and fibronectin, via a beta 3
integrin. J. Bone Miner. Res. 1992, 7, 335–343. 9.
Lakkakorpi, P.T.; Horton, MA; Helfrich, M.H.; Karhukorpi,
E.-K; Va¨a¨na¨nen, H.K. Vitronectin receptor has a role in
bone resorption but does not mediate tight sealing zone
attachment of osteoclasts to the bone surface. J. Cell
Biol. 1991, 115, 1179–1186. 10. Meredith, J.E., Jr; Winitz,
S.; Lewis, J.M.; et al. The regulation of growth and
intracellular signaling by integrins. Endocr. Rev. 1996,
17, 207–220. 11. Duong, L.T.; Lakkakorpi, P.; Nakamura, I.;
Rodan, G.A. Integrins and signaling in osteoclast function.
Matrix Biol. 2000, 19, 97–105. 12. Blair, H.C.; Teitelbaum,
S.L.; Ghiselli, R.; Gluck, S. Osteoclastic bone resorption
by a polarized vacuolar proton pump. Science 1989, 245,
855–857. 13. Vaananen, H.K.; Karhukorpi, E.K.; Sundquist,
K.; et al. Evidence for the presence of a proton pump of
the vacuolar H(+)-ATPase type in the ruffled borders of
osteoclasts. J. Cell Biol. 1990, 111, 1305–1311. 14. Salo,
J.; Metsikko, K.; Palokangas, H.; Lehenkari, P.; Vaananen,
H. Bone-resorbing osteoclasts reveal a dynamic division of
basal plasma membrane into two different domains. J. Cell
Sci. 1996, 109, 301–307. 15. Vaananen, H.K.; Zhao, H.;
Mulari, M.; Halleen, J.M. The cell biology of osteoclast
function. J. Cell Sci. 2000, 113, 377–381. 16. Gay, C.V.;
Mueller, W.J. Carbonic anhydrase and osteoclasts:
localization by labeled inhibitor autoradiography. Science
1974, 183, 432–434. 17. Palokangas, H.; Mulari, M.;
Vaananen, H.K. Endocytic pathway from the basal plasma
membrane to the ruffled border membrane in bone-resorbing
osteoclasts. J. Cell Sci. 1997, 110, 1767–1780. 18. Goto,
T.; Yamaza, T.; Tanaka, T. Cathepsins in the osteoclast. J.
Electron Microsc. (Tokyo) 2003, 52, 551–558. 19.
Littlewood-Evans, A.; Kokubo, T.; Ishibashi, O.; et al.
Localization of cathepsin K in human osteoclasts by in situ
hybridization and immunohistochemistry. Bone 1997, 20,
81–86. 20. Kafienah, W.; Bromme, D.; Buttle, D.J.; Croucher,
L.J.; Hollander, A.P. Human cathepsin K cleaves native type
I and II collagens at the N-terminal end of the triple
helix. Biochem. J. 1998, 331, 727–732. 21. Bromme, D.;
Okamoto, K.; Wang, B.B.; Biroc, S. Human cathepsin O2, a
matrix protein-degrading cysteine protease expressed in
osteoclasts. Functional expression of human cathepsin O 2
in Spodoptera frugiperda and characterization of the
enzyme. J. Biol. Chem. 1996, 271, 2126–2132. 22. Bossard,
M.J.; Tomaszek, T.A.; Thompson, S.K.; et al. Proteolytic
activity of human osteoclast cathepsin K. Expression,
purification, activation, and substrate identification. J.
Biol. Chem. 1996, 271, 12517–12524. 23. Ljusberg, J.; Wang,
Y.; Lang, P.; et al. Proteolytic excision of a repressive
loop domain in tartrate-resistant acid phosphatase by
cathepsin K in osteoclasts. J. Biol. Chem. 2005, 280 (31),
28370–28381. 24. Saftig, P.; Hunziker, E.; Wehmeyer, O.; et
al. Impaired osteoclastic bone resorption leads to
osteopetrosis in cathepsin-K-deficient mice. Proc. Natl
Acad. Sci. USA. 1998, 95, 13453–13458. 25. Yamaza, T.;
Tsuji, Y.; Goto, T.; et al. Comparison in localization
between cystatin C and cathepsin K in osteoclasts and other
cells in mouse tibia epiphysis by immunolight and
immunoelectron microscopy. Bone 2001, 29, 42–53. 26.
Lerner, U.H.; Johansson, L.; Ranjso, M.; Rosenquist, J.B.;
Reinholt, F.P.; Grubb, A. Cystatin C, and inhibitor of bone
resorption produced by osteoblasts. Acta Physiol. Scand.
1997, 161, 81–92. 27. Motyckova, G.; Fisher, D.E.
Pycnodysostosis: role and regulation of cathepsin K in
osteoclast function and human disease. Curr. Mol. Med.
2002, 2, 407–421. 28. Delaisse, J.M.; Andersen, T.L.;
Engsig, M.T.; Henriksen, K.; Troen, T.; Blavier, L. Matrix
metalloproteinases (MMP) and cathepsin K contribute
differently to osteoclastic activities. Microsc. Res. Tech.
2003, 61, 504–513. 29. Teronen, O.; Heikkila, P.;
Konttinen, Y.T.; et al. MMP inhibition and downregulation
by bisphosphonates. Ann. N.Y. Acad. Sci. 1999, 878,
453–565. 30. Wu, X.; McKenna, M.; Feng, X.; Nagy, T.R.;
McDonald, J.M. Osteoclast apoptosis: the role of Fas in
vivo and in vitro. Endocrinology 2003, 144, 5545–5555. 31.
Chang, S.S.; Suratwala, S.J.; Jung, K.M.; et al.
Bisphosphonates may reduce recurrence in giant cell tumor
by inducing apoptosis. Clin. Orthop. Relat. Res. 2004,
103–109. 32. Lacey, D.L.; Timms, E.; Tan, H.-L.; et al.
Osteoprotegerin ligand is a cytokine that regulates
osteoclast differentiation and activation. Cell 1998, 93,
165–176. 33. Wang, R.; Zhang, L.Y.; Zhang, X.R.; et al.
Regulation of activation-induced receptor activator of
NF-kappaB ligand (RANKL) expression in T cells. Eur. J.
Immunol. 2002, 32, 1090–1098. 34. van’t Hof, R.J.;
Tuinenburg-Bol Raap, A.C.; Nijweide, P.J. Induction of
osteoclast characteristics in cultured avian blood
monocytes; modulation by osteoblasts and 1,25-(OH) 2
vitamin D 3 . Int. J. Exp. Pathol. 1995, 76, 205–214. 35.
Rivollier, A.; Mazzorana, M.; Tebib, J.; et al. Immature
dendritic cell transdifferentiation into osteoclasts: a
novel pathway sustained by the rheumatoid arthritis
microenvironment. Blood 2004, 104, 4029–4037. 36. Darnay,
B.G.; Haridas, V.; Ni, J.; Moore, P.A.; Aggarwal, B.B.
Characterization of the intracellular domain of receptor
activator of NF-kappaB (RANK). Interaction with tumor
necrosis factor receptor-associated factors and activation
of NF-kappab and c-Jun N-terminal kinase. J. Biol. Chem.
1998, 273, 20551– 20555. 37. Khosla, S. Minireview: the
OPG/RANKL/RANK system. Endocrinology 2001, 142, 5050–5055.
38. Hsu, H.; Lacey, D.L.; Dunstan, C.R.; et al. Tumor
necrosis factor receptor family member RANK mediates
osteoclast differentiation and activation induced by
osteoprotegerin ligand. Proc. Natl Acad. Sci. USA. 1999,
96, 3540–3545. 39. Myers, D.E.; Collier, F.M.; Minkin, C.;
et al. Expression of functional RANK on mature rat and
human osteoclasts. FEBS Lett. 1999, 463, 295–300. 40.
Yasuda, H.; Shima, N.; Nakagawa, N.; et al. Osteoclast
differentiation factor is a ligand for osteoprotegerin/
osteoclastogenesis-inhibitory factor and is identical to
TRANCE/RANKL. Proc. Natl Acad. Sci. USA. 1998, 95,
3597–3602. 41. Skoumal, M.; Kolarz, G.; Haberhauer, G.;
Woloszczuk, W.; Hawa, G.; Klingler, A. Osteoprotegerin and
the receptor activator of NF-kappa B ligand in the serum
and synovial fluid. A comparison of patients with
longstanding rheumatoid arthritis and osteoarthritis.
Rheumatol. Int. 2005, 25, 373–378. 42. Grimaud, E.;
Soubigou, L.; Couillaud, S; et al. Receptor activator of
nuclear factor kappaB ligand (RANKL)/osteoprotegerin (OPG)
ratio is increased in severe osteolysis. Am. J. Pathol.
2003, 163, 2021–2031. 43. Goranov, S.E.; Goranova-Marinova,
V.S. Bone lesions in multiple myeloma–the OPG/RANK-ligand
system. Folia Med. (Plovdiv) 2004, 46, 5–11. 44. Kodama,
H.; Yamasaki, A.; Nose, M.; et al. Congenital osteoclast
deficiency in osteopetrotic (op/op) mice is cured by
injections of macrophage colony-stimulating factor. J. Exp.
Med. 1991, 173, 269–272. 45. Tanaka, S.; Takahashi, N.;
Udagawa, N.; et al. Macrophage colony-stimulating factor is
indispensable for both proliferation and differentiation of
osteoclast progenitors. J. Clin. Invest. 1993, 91, 257–263.
46. Dinarello, C.A. Interleukin-1. Cytokine Growth Factor
Rev. 1997, 8, 253–265. 47. Wei, S.; Kitaura, H.; Zhou, P.;
Ross, F.P.; Teitelbaum, S.L. IL-1mediates TNF-induced
osteoclastogenesis. J. Clin. Invest. 2005, 115, 282–290. O

48. Ma, T.; Miyanishi, K.; Suen, A.; Epstein, N.J.; Tomita,
T.; Smith, R.L. Human interleukin-1-induced murine
osteoclastogenesis is dependent on RANKL, but independent
of TNF-alpha. Cytokine 2004, 26, 138–144.

49. Li, H.; Cuartas, E.; Cui, W.; et al. IL-1


receptor-associated kinase M is a central regulator of
osteoclast differentiation and activation. J. Exp. Med.
2005, 201, 1169–1177.

50. Kamolmatyakul, S.; Chen, W.; Yang, S.; et al. IL-1alpha


stimulates cathepsin K expression in osteoclasts via the
tyrosine kinase-NF-kappaB pathway. J. Dent. Res. 2004, 83,
791–796.

51. Kobayashi, K.; Takahashi, N.; Jimi, E.; et al. Tumor


necrosis factor alpha stimulates osteoclast differentiation
by a mechanism independent of the ODF/ RANKL–RANK
interaction. J. Exp. Med. 2000, 191, 275–286.

52. Zou, W.; Hakim, I.; Tschoep, K.; Endres, S.; BarShavit,
Z. Tumor necrosis factor-alpha mediates RANK ligand
stimulation of osteoclast differentiation by an autocrine
mechanism. J. Cell Biochem. 2001, 83, 70–83.

53. Holt, I.; Davie, M.W.; Marshall, M.J. Osteoclasts are


not the major source of interleukin-6 in mouse parietal
bones. Bone 1996, 18, 221–226.

54. Kurihara, N.; Bertolini, D.; Suda, T.; Akiyama, Y.;


Roodman, G.D. IL-6 stimulates osteoclast-like
multinucleated cell formation in long term human marrow
cultures by inducing IL-1 release. J. Immunol. 1990, 144,
4226–4230.

55. O’Brien, C.A.; Lin, S.C.; Bellido, T.; Manolagas, S.C.


Expression levels of gp130 in bone marrow stromal cells
determine the magnitude of osteoclastogenic signals
generated by IL-6-type cytokines. J. Cell Biochem. 2000,
79, 532–541.

56. Naito, A.; Azuma, S.; Tanaka, S.; et al. Severe


osteopetrosis, defective interleukin-1 signalling and lymph
node organogenesis in TRAF6-deficient mice. Genes Cells
1999, 4, 353–362.

57. Clohisy, J.C.; Frazier, E.; Hirayama, T.; Abu-Amer, Y.


RANKL is an essential cytokine mediator of
polymethylmethacrylate particle-induced osteoclastogenesis.
J. Orthop. Res. 2003, 21, 202–212. 58. Lubberts, E.;
Schwarzenberger, P.; Huang, W.; et al. IL-17 promotes bone
erosion in murine collagen-induced arthritis through loss
of the receptor activator of NF-kappa B
ligand/osteoprotegerin balance. J. Immunol. 2003, 170,
2655–2662. 59. Menaa, C.; Reddy, S.V.; Kurihara, N.; et al.
Enhanced RANK ligand expression and responsivity of bone
marrow cells in Paget’s disease of bone. J. Clin. Investig.
2000, 105, 1833–1838. 60. Collin-Osdoby, P. Regulation of
vascular calcification by osteoclast regulatory factors
RANKL and osteoprotegerin. Circ. Res. 2004, 95, 1046–1057.
61. Good, C.R.; O’Keefe, R.J.; Puzas, J.E.; Schwarz, E.M.;
Rosier, R.N. Immunohistochemical study of receptor
activator of nuclear factor kappa-B ligand (RANK-L) in
human osteolytic bone tumors. J. Surg. Oncol. 2002, 79 ,
174–179. 62. Kitazawa, S.; Kitazawa, R. RANK ligand is a
prerequisite for cancer-associated osteolytic lesions. J.
Pathol. 2002, 198, 228–236. 63. Teitelbaum, S.L. Bone
resorption by osteoclasts. Science 2000, 289, 1504–1508.
64. Quinn, J.M.; Neale, S.; Fujikawa, Y.; McGee, J.O.;
Athanasou, N.A. Human osteoclast formation from blood
monocytes, peritoneal macrophages, and bone marrow cells.
Calcif. Tissue Int. 1998, 62, 527–531. 65. Reddy, S.V.;
Roodman, G.D. Control of osteoclast differentiation. Crit.
Rev. Eukaryot. Gene Expr. 1998, 8, 1–17. 66. Teitelbaum,
S.L. Bone resorption by osteoclasts. Science 2000, 289,
1504–1508. 67. Takahashi, N.; Akatsu, T.; Udagawa, N.; et
al. Osteoblastic cells are involved in osteoclast
formation. Endocrinology 1988, 123, 2600–2602. 68.
Tondravi, M.M.; McKercher, S.R.; Anderson, K.; et al.
Osteopetrosis in mice lacking haematopoietic transcription
factor PU.1. Nature 1997, 386, 81–84.
Osteogenic Progenitor Cells of Bone

9. Bianco, P.; Robey, P.G. Stem cells in tissue


engineering. Nature 2001, 414 (6859), 118–121.

10. Bianco, P.; Riminucci, M.; Gronthos, S.; Robey, P.G.


Bone marrow stromal stem cells: Nature, biology, and
potential applications. Stem Cells 2001, 19 (3), 180–192.

11. Jiang, Y.; Jahagirdar, B.N.; Reinhardt, R.L.; Schwartz,


R.E.; Keene, C.D.; Ortiz-Gonzalez, X.R.; Reyes, M.; Lenvik,
T.; Lund, T.; Blackstad, M.; Du, J.; Aldrich, S.; Lisberg,
A.; Low, W.C.; Largaespada, D.A.; Verfaillie, C.M.
Pluripotency of mesenchymal stem cells derived from adult
marrow. Nature 2002, 418 (6893), 41–49.

12. Triffitt, J.T.; Oreffo, R.O.C. Osteoblast Lineage, 1st


Ed.; JAI Press, Inc., 1998; 475–498.

13. Nishida, S.; Endo, N.; Yamagiwa, H.; Tanizawa, T.;


Takahashi, H.E. Number of osteoprogenitor cells in human
bone marrow markedly decreases after skeletal maturation.
J. Bone Miner. Metab. 1999, 17 (3), 171–177.

14. D’Ippolito, G.; Schiller, P.C.; Ricordi, C.; Roos,


B.A.; Howard, G.A. Age-related osteogenic potential of
mesenchymal stromal stem cells from human vertebral bone
marrow. J. Bone Miner. Res. 1999, 14 (7), 1115– 1122.

15. Majors, A.K.; Boehm, C.A.; Nitto, H.; Midura, R.J.;


Muschler, G.F. Characterization of human bone marrow
stromal cells with respect to osteoblastic differentiation.
J. Orthop. Res. 1997, 15 (4), 546–557.

16. Egrise, D.; Martin, D.; Vienne, A.; Neve, P.;


Schoutens, A. The number of fibroblastic colonies formed
from bone marrow is decreased and the in vitro
proliferation rate of trabecular bone cells increased in
aged rats. Bone 1992, 13 (5), 355–361.

17. Bergman, R.J.; Gazit, D.; Kahn, A.J.; Gruber, H.;


McDougall, S.; Hahn, T.J. Age-related changes in osteogenic
stem cells in mice. J. Bone Miner. Res. 1996, 11 (5),
568–577.

18. Oreffo, R.O.C.; Bennett, A.; Carr, A.J.; Triffitt, J.T.


Patients with primary osteoarthritis show no change with
ageing in the number of osteogenic precursors. Scand. J.
Rheumatol. 1998, 27 (6), 415–424.
19. Oreffo, R.O.C.; Bord, S.; Triffitt, J.T. Skeletal
progenitor cells and ageing human populations. Clin. Sci.
(Colch.) 1998, 94 (5), 549–555.

20. Stenderup, K.; Justesen, J.; Eriksen, E.F.; Rattan,


S.I.; Kassem, M. Number and proliferative capacity of
osteogenic stem cells are maintained during aging and in
patients with osteoporosis. J. Bone Miner. Res. 2001, 16
(6), 1120–1129.

21. Pfeilschifter, J.; Diel, I.; Pilz, U.; Brunotte, K.;


Naumann, A.; Ziegler, R. Mitogenic responsiveness of human
bone cells in vitro to hormones and growth factors
decreases with age. J. Bone Miner. Res. 1993, 8 (6),
707–717.

22. Erdmann, J.; Kogler, C.; Diel, I.; Ziegler, R.;


Pfeilschifter, J. Age-associated changes in the stimulatory
effect of transforming growth factor beta on human
osteogenic colony formation. Mech. Ageing Dev. 1999, 110
(1–2), 73–85.

23. Verfaillie, C.M. Adult stem cells: Assessing the case


for pluripotency. Trends Cell Biol. 2002 , 12 (11),
502–508. 24. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.;
Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.;
Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage
potential of adult human mesenchymal stem cells. Science
1999, 284 (5411), 143–147. 25. Bruder, S.P.; Horowitz,
M.C.; Mosca, J.D.; Haynesworth, S.E. Monoclonal antibodies
reactive with human osteogenic cell surface antigens. Bone
1997, 21 (3), 225–235. 26. Simmons, P.J.; Torok-Storb, B.
CD34 expression by stromal precursors in normal human adult
bone marrow. Blood 1991, 78 (11), 2848–2853. 27.
Haynesworth, S.E.; Baber, M.A.; Caplan, A.I. Cell surface
antigens on human marrow-derived mesenchymal cells are
detected by monoclonal antibodies. Bone 1992, 13 (1),
69–80. 28. Joyner, C.J.; Bennett, A.; Triffitt, J.T.
Identification and enrichment of human osteoprogenitor cells
by using differentiation stage-specific monoclonal
antibodies. Bone 1997, 21 (1), 1–6. 29. Gronthos, S.;
Graves, S.E.; Ohta, S.; Simmons, P.J. The STRO-1+ fraction
of adult human bone marrow contains the osteogenic
precursors. Blood 1994, 84 (12), 4164–4173. 30. Stewart,
K.; Walsh, S.; Screen, J.; Jefferiss, C.M.; Chainey, J.;
Jordan, G.R.; Beresford, J.N. Further characterization of
cells expressing STRO-1 in cultures of adult human bone
marrow stromal cells. J. Bone Miner. Res. 1999, 14 (8),
1345–1356. 31. Howard, D.; Partridge, K.; Yang, X.; Clarke,
N.M.; Okubo, Y.; Bessho, K.; Howdle, S.M.; Shakesheff,
K.M.; Oreffo, R.O.C. Immunoselection and adenoviral genetic
modulation of human osteoprogenitors: In vivo bone
formation on PLA scaffold. Biochem. Biophys. Res. Commun.
2002, 299 (2), 208–215. 32. Bruder, S.P.; Ricalton, N.S.;
Boynton, R.E.; Connolly, T.J.; Jaiswal, N.; Zaia, J.;
Barry, F.P. Mesenchymal stem cell surface antigen SB-10
corresponds to activated leukocyte cell adhesion molecule
and is involved in osteogenic differentiation. J. Bone
Miner. Res. 1998, 13 (4), 655–663. 33. Barry, F.P.;
Boynton, R.E.; Haynesworth, S.; Murphy, J.M.; Zaia, J. The
monoclonal antibody SH-2, raised against human mesenchymal
stem cells, recognizes an epitope on endoglin (CD105).
Biochem. Biophys. Res. Commun. 1999, 265 (1), 134–139. 34.
Triffitt, J.T.; Oreffo, R.O.C.; Virdi, A.S.; Xia, Z.
Osteogenic stem-cell characterization and development:
Potentials for cytotherapy. Cytotherapy 2001, 3 (5),
413–416. 35. Park, S.R.; Oreffo, R.O.C.; Triffitt, J.T.
Interconversion potential of cloned human marrow adipocytes
in vitro. Bone 1999, 24 (6), 549–554. 36. Kuznetsov, S.A.;
Mankani, M.H.; Gronthos, S.; Satomura, K.; Bianco, P.;
Robey, P.G. Circulating skeletal stem cells. J. Cell Biol.
2001, 153 (5), 1133–1140. 37. Friedenstein, A.J.;
Deriglasova, U.F.; Kulagina, N.N.; Panasuk, A.F.; Rudakowa,
S.F.; Luria, E.A.; Ruadkow, I.A. Precursors for fibroblasts
in different populations of hematopoietic cells as detected
by the in vitro colony assay method. Exp. Hematol. 1974, 2
(2), 83–92. 38. Nuttall, M.E.; Gimble, J.M. Is there a
therapeutic opportunity to either prevent or treat
osteopenic disorders by inhibiting marrow adipogenesis?
Bone 2000, 27 (2), 177–184. 39. Nuttall, M.E.; Patton,
A.J.; Olivera, D.L.; Nadeau, D.P.; Gowen, M. Human
trabecular bone cells are able to express both osteoblastic
and adipocytic phenotype: Implications for osteopenic
disorders. J. Bone Miner. Res. 1998, 13 (3), 371–382. 40.
Wozney, J.M.; Rosen, V. Bone morphogenetic protein and bone
morphogenetic protein gene family in bone formation and
repair. Clin. Orthop. 1998, 346, 26–37. 41. Chen, D.; Ji,
X.; Harris, M.A.; Feng, J.Q.; Karsenty, G.; Celeste, A.J.;
Rosen, V.; Mundy, G.R.; Harris, S.E. Differential roles for
bone morphogenetic protein (BMP) receptor type IB and IA in
differentiation and specification of mesenchymal precursor
cells to osteoblast and adipocyte lineages. J. Cell Biol.
1998, 142 (1), 295–305. 42. Pfeilschifter, J.; Wolf, O.;
Naumann, A.; Minne, H.W.; Mundy, G.R.; Ziegler, R.
Chemotactic response of osteoblastlike cells to
transforming growth factor beta. J. Bone Miner. Res. 1990,
5 (8), 825–830. 43. Locklin, R.M.; Oreffo, R.O.C.; Triffitt,
J.T. Effects of TGFbeta and bFGF on the differentiation of
human bone marrow stromal fibroblasts. Cell Biol. Int. 1999,
23 (3), 185–194. 44. Walsh, S.; Jefferiss, C.; Stewart, K.;
Beresford, J.N. TGFbeta1 limits the expansion of the
osteoprogenitor fraction in cultures of human bone marrow
stromal cells. Cell Tissue Res. 2003, 311 (2), 187–198. 45.
Wang, E.A.; Israel, D.I.; Kelly, S.; Luxenberg, D.P. Bone
morphogenetic protein-2 causes commitment and
differentiation in C3H10T1/2 and 3T3 cells. Growth Factors
1993, 9 (1), 57–71. 46. Rickard, D.J.; Sullivan, T.A.;
Shenker, B.J.; Leboy, P.S.; Kazhdan, I. Induction of rapid
osteoblast differentiation in rat bone marrow stromal cell
cultures by dexamethasone and BMP-2. Dev. Biol. 1994, 161
(1), 218–228. 47. Gronthos, S.; Simmons, P.J. The growth
factor requirements of STRO-1–positive human bone marrow
stromal precursors under serum-deprived conditions in
vitro. Blood 1995, 85 (4), 929–940. 48. Kuznetsov, S.A.;
Friedenstein, A.J.; Robey, P.G. Factors required for bone
marrow stromal fibroblast colony formation in vitro. Br. J.
Haematol. 1997, 97 (3), 561–570. 49. Oreffo, R.O.C.;
Romberg, S.; Virdi, A.S.; Joyner, C.J.; Berven, S.;
Triffitt, J.T. Effects of interferon alpha on human
osteoprogenitor cell growth and differentiation in vitro.
J. Cell. Biochem. 1999, 74 (3), 372–385. 50. Walsh, S.;
Jefferiss, C.; Stewart, K.; Jordan, G.R.; Screen, J.;
Beresford, J.N. Expression of the developmental markers
STRO-1 and alkaline phosphatase in cultures of human marrow
stromal cells: Regulation by fibroblast growth factor
(FGF)-2 and relationship to the expression of FGF receptors
1–4. Bone 2000, 27 (2), 185–195. 51. Martin, I.; Muraglia,
A.; Campanile, G.; Cancedda, R.; Quarto, R. Fibroblast
growth factor-2 supports ex vivo expansion and maintenance
of osteogenic precursors from human bone marrow.
Endocrinology 1997, 138 (10), 4456–4462. 52. Locklin, R.M.;
Williamson, M.C.; Beresford, J.N.; Triffitt, J.T.; Owen,
M.E. In vitro effects of growth factors and dexamethasone
on rat marrow stromal cells. Clin. Orthop. 1995, 313,
27–35. 53. Oreffo, R.O.C.; Virdi, A.S.; Triffitt, J.T.
Modulation of osteogenesis and adipogenesis by human serum
in human bone marrow cultures. Eur. J. Cell Biol. 1997, 74
(3), 251–261. 54. Cheng, S.L.; Yang, J.W.; Rifas, L.;
Zhang, S.F.; Avioli, L.V. Differentiation of human bone
marrow osteogenic stromal cells in vitro: Induction of the
osteoblast phenotype by dexamethasone. Endocrinology 1994,
134 (1), 277–286. 55. Kimoto, S.; Cheng, S.L.; Zhang, S.F.;
Avioli, L.V. The effect of glucocorticoid on the synthesis
of biglycan and decorin in human osteoblasts and bone
marrow stromal cells. Endocrinology 1994, 135 (6),
2423–2431. 56. Walsh, S.; Jordan, G.R.; Jefferiss, C.;
Stewart, K.; Beresford, J.N. High concentrations of
dexamethasone suppress the proliferation but not the
differentiation or further maturation of human osteoblast
precursors in vitro: Relevance to glucocorticoid-induced
osteoporosis. Rheumatol. (Oxf.) 2001, 40 (1), 74–83. 57.
Beresford, J.N.; Joyner, C.J.; Devlin, C.; Triffitt, J.T.
The effects of dexamethasone and 1,25-dihydroxyvitamin D3
on osteogenic differentiation of human marrow stromal cells
in vitro. Arch. Oral Biol. 1994, 39 (11), 941–947. 58.
Scutt, A.; Bertram, P. Bone marrow cells are targets for
the anabolic actions of prostaglandin E2 on bone: Induction
of a transition from nonadherent to adherent osteoblast
precursors. J. Bone Miner. Res. 1995, 10 (3), 474–487. 59.
Still, K.; Scutt, A. Stimulation of CFU—F formation by
prostaglandin E2 is mediated in part by its degradation
product, prostaglandin A2. Prostaglandins Other Lipid
Mediat. 2001, 65 (1), 21–31. 60. Kasugai, S.; Oida, S.;
Iimura, T.; Arai, N.; Takeda, K.; Ohya, K.; Sasaki, S.
Expression of prostaglandin E receptor subtypes in bone:
Expression of EP2 in bone development. Bone 1995, 17 (1),
1–4. 61. Oreffo, R.O.C.; Wells, N.; Johnstone, D. The
presence of prostaglandin receptors on osteoblasts and
osteoclasts. J. Bone Miner. Res. 1991, 6 (Suppl. 1), S208.
62. Yang, X.; Tare, R.S.; Partridge, K.A.; Roach, H.I.;
Clarke, N.M.; Howdle, S.M.; Shakesheff, K.M.; Oreffo,
R.O.C. Induction of human osteoprogenitor chemotaxis,
proliferation, differentiation, and bone formation by
osteoblast stimulating factor-1/pleiotrophin:
Osteoconductive biomimetic scaffolds for tissue
engineering. J. Bone Miner. Res. 2003, 18 (1), 47–57. 63.
Tare, R.S.; Oreffo, R.O.C.; Clarke, N.M.; Roach, H.I.
Pleiotrophin/osteoblast-stimulating factor 1: Dissecting
its diverse functions in bone formation. J. Bone Miner.
Res. 2002, 17 (11), 2009–2020. 64. Jilka, R.L. Cytokines,
bone remodeling, and estrogen deficiency: A 1998 update.
Bone 1998 , 23 (2), 75–81. 65. Manolagas, S.C.; Jilka, R.L.
Bone marrow, cytokines, and bone remodeling. Emerging
insights into the O pathophysiology of osteoporosis. N.
Engl. J. Med. 1995, 332 (5), 305–311.

66. Boyce, B.F.; Aufdemorte, T.B.; Garrett, I.R.; Yates,


A.J.; Mundy, G.R. Effects of interleukin-1 on bone turnover
in normal mice. Endocrinology 1989, 125 (3), 1142–1150.

67. Chapekar, M.S. Tissue engineering: Challenges and


opportunities. J. Biomed. Mater. Res. 2000, 53 (6),
617–620.

68. Jordan, K.M.; Cooper, C. Epidemiology of osteoporosis.


Best. Pract. Res. Clin. Rheumatol. 2002, 16 (5), 795–806.

69. Spector, M. Biomaterial failure. Orthop. Clin. North


Am. 1992, 23 (2), 211–217.
70. Rose, F.R.; Oreffo, R.O.C. Bone tissue engineering:
Hope vs. hype. Biochem. Biophys. Res. Commun. 2002, 292
(1), 1–7.

71. Langer, R.; Vacanti, J.P. Tissue engineering. Science


1993, 260 (5110), 920–926.

72. Kadiyala, S.; Young, R.G.; Thiede, M.A.; Bruder, S.P.


Culture expanded canine mesenchymal stem cells possess
osteochondrogenic potential in vivo and in vitro. Cell
Transplant. 1997, 6 (2), 125–134.

73. Gundle, R.; Joyner, C.J.; Triffitt, J.T. Human bone


tissue formation in diffusion chamber culture in vivo by
bone-derived cells and marrow stromal fibroblastic cells.
Bone 1995, 16 (6), 597–601.

74. Nakahara, H.; Bruder, S.P.; Haynesworth, S.E.; Holecek,


J.J.; Baber, M.A.; Goldberg, V.M.; Caplan, A.I. Bone and
cartilage formation in diffusion chambers by subcultured
cells derived from the periosteum. Bone 1990, 11 (3),
181–188.

75. Ohgushi, H.; Goldberg, V.M.; Caplan, A.I. Heterotopic


osteogenesis in porous ceramics induced by marrow cells. J.
Orthop. Res. 1989, 7 (4), 568–578.

76. Simmons, P.J.; Storb, B. Torok-identification of stromal


cell precursors in human bone marrow by a novel monoclonal
antibody, STRO-1. Blood 1991, 78 (1), 55– 62.

77. Stewart, K.; Walsh, S.; Screen, J.; Jefferiss, C.M.;


Chainey, J.; Jordan, G.R.; Beresford, J.N. Further
characterization of cells expressing STRO-1 in cultures of
adult human bone marrow stromal cells. J. Bone Miner. Res.
1999, 14 (8), 1345–1356.

78. Bruder, S.P.; Jaiswal, N.; Haynesworth, S.E. Growth


kinetics, self-renewal, and the osteogenic potential of
purified human mesenchymal stem cells during extensive
subcultivation and following cryopreservation. J. Cell.
Biochem. 1997, 64 (2), 278–294.

79. Jaiswal, N.; Haynesworth, S.E.; Caplan, A.I.; Bruder,


S.P. Osteogenic differentiation of purified,
culture-expanded human mesenchymal stem cells in vitro. J.
Cell. Biochem. 1997, 64 (2), 295–312.

80. Kadiyala, S.; Young, R.G.; Thiede, M.A.; Bruder, S.P.


Culture expanded canine mesenchymal stem cells possess
osteochondrogenic potential in vivo and in vitro. Cell
Transplant 1997, 6 (2), 125–134.

81. Horwitz, E.M.; Prockop, D.J.; Fitzpatrick, L.A.; Koo,


W.W.; Gordon, P.L.; Neel, M.; Sussman, M.; Orchard, P.;
Marx, J.C.; Pyeritz, R.E.; Brenner, M.K. Transplantability
and therapeutic effects of bone marrowderived mesenchymal
cells in children with osteogenesis imperfecta. Nat. Med.
1999, 5 (3), 309–313. 82. Horwitz, E.M.; Prockop, D.J.;
Gordon, P.L.; Koo, W.W.; Fitzpatrick, L.A.; Neel, M.D.;
McCarville, M.E.; Orchard, P.J.; Pyeritz, R.E.; Brenner,
M.K. Clinical responses to bone marrow transplantation in
children with severe osteogenesis imperfecta. Blood 2001,
97 (5), 1227–1231. 83. Dominici, M.; Hofmann, T.J.;
Horwitz, E.M. Bone marrow mesenchymal cells: Biological
properties and clinical applications. J. Biol. Regul.
Homeost. Agents 2001, 15 (1), 28–37. 84. Connolly, J.F.
Injectable bone marrow preparations to stimulate osteogenic
repair. Clin. Orthop. 1995, 313, 8–18. 85. Quarto, R.;
Mastrogiacomo, M.; Cancedda, R.; Kutepov, S.M.; Mukhachev,
V.; Lavroukov, A.; Kon, E.; Marcacci, M. Repair of large
bone defects with the use of autologous bone marrow stromal
cells. N. Engl. J. Med. 2001, 344 (5), 385–386. 86. Perry,
C.R. Bone repair techniques, bone graft, and bone graft
substitutes. Clin. Orthop. 1999, 360, 71–86. 87. Boden,
S.D. Bioactive factors for bone tissue engineering. Clin.
Orthop. 1999, 367 (Suppl. 1), 84–94. 88. Oreffo, R.O.C.;
Triffitt, J.T. Future potentials for using osteogenic stem
cells and biomaterials in orthopedics. Bone 1999, 25 (2
Suppl.), 5S–9S. 89. Lieberman, J.R.; Daluiski, A.;
Stevenson, S.; Wu, L.; McAllister, P.; Lee, Y.P.; Kabo,
J.M.; Finerman, G.A.; Berk, A.J.; Witte, O.N. The effect of
regional gene therapy with bone morphogenetic
protein-2-producing bone-marrow cells on the repair of
segmental femoral defects in rats. J. Bone Jt. Surg. Am.
1999, 81 (7), 905– 917. 90. Breitbart, A.S.; Grande, D.A.;
Mason, J.M.; Barcia, M.; James, T.; Grant, R.T.
Gene-enhanced tissue engineering: Applications for bone
healing using cultured periosteal cells transduced
retrovirally with the BMP-7 gene. Ann. Plast. Surg. 1999,
42 (5), 488–495. 91. Olmsted, E.A.; Blum, J.S.; Rill, D.;
Yotnda, P.; Gugala, Z.; Lindsey, R.W.; Davis, A.R.
Adenovirus-mediated BMP2 expression in human bone marrow
stromal cells. J. Cell. Biochem. 2001, 82 (1), 11–21. 92.
Turgeman, G.; Pittman, D.D.; Muller, R.; Kurkalli, B.G.;
Zhou, S.; Pelled, G.; Peyser, A.; Zilberman, Y.;
Moutsatsos, I.K.; Gazit, D. Engineered human mesenchymal
stem cells: A novel platform for skeletal cell mediated
gene therapy. J. Gene Med. 2001, 3 (3), 240–251. 93.
Partridge, K.; Yang, X.; Clarke, N.M.; Okubo, Y.; Bessho,
K.; Sebald, W.; Howdle, S.M.; Shakesheff, K.M.; Oreffo,
R.O.C. Adenoviral BMP-2 gene transfer in mesenchymal stem
cells: In vitro and in vivo bone formation on biodegradable
polymer scaffolds. Biochem. Biophys. Res. Commun. 2002, 292
(1), 144–152. 94. Musgrave, D.S.; Bosch, P.; Ghivizzani,
S.; Robbins, P.D.; Evans, C.H.; Huard, J.
Adenovirus-mediated direct gene therapy with bone
morphogenetic protein-2 produces bone. Bone 1999, 24 (6),
541–547. 95. Patel, N.; Padera, R.; Sanders, G.H.;
Cannizzaro, S.M.; Davies, M.C.; Langer, R.; Roberts, C.J.;
Tendler, S.J.; Williams, P.M.; Shakesheff, K.M. Spatially
controlled cell engineering on biodegradable polymer
surfaces. FASEB J. 1998, 12 (14), 1447–1454. 96. Pearson,
R.G.; Bhandari, R.; Quirk, R.A.; Shakesheff, K.M. Recent
advances in tissue engineering: An invited review. J.
Long-Term Eff. Med. Implants 2002, 12 (1), 1–33. 97.
Rezania, A.; Healy, K.E. Integrin subunits responsible for
adhesion of human osteoblast-like cells to biomimetic
peptide surfaces. J. Orthop. Res. 1999, 17 (4), 615–623.
98. Dee, K.C.; Anderson, T.T.; Bizios, R. Osteoblast
population migration characteristics on substrates modified
with immobilized adhesive peptides. Biomaterials 1999, 20
(3), 221–227. 99. Yang, X.B.; Roach, H.I.; Clarke, N.M.;
Howdle, S.M.; Quirk, R.; Shakesheff, K.M.; Oreffo, R.O.C.
Human osteoprogenitor growth and differentiation on
synthetic biodegradable structures after surface
modification. Bone 2001, 29 (6), 523–531. O
Osteoinductive Substances and Materials

31. Yabe, T.; Nemoto, A.; Uemura, T. Recognition of


osteopontin by rat bone marrow derived osteoblastic primary
cells. Biosci. Biotechnol. Biochem. 1997, 61, 754–756.

32. Uemura, T.; Liu, Y.-K.; Feng, Y.; Nemoto, A.; Yabe, T.;
Ushida, T.; Miyamoto, H.; Tateishi, T. The role of
sialoprotein in recognition of bone surface by osteoblasts
via integrin. Mater. Sci. Eng. 1997, C4, 303–309.

33. Nemoto, A.; Uemura, T. Integrin signal transduction via


FAK in osteoblasts. Seikagaku 1999, 71, 140–145.

34. Uemura, T.; Nemoto, A.; Deng, H.-Y. Differentiation and


function of bone cells induced by bone matrix proteins.
Tanpakushitsu Kakusan Koso 1999, 44, 143–148.

35. Liu, Y.-K.; Uemura, T.; Nemoto, A.; Yebe, T.; Fujii,
N.; Ushida, T.; Tateishi, T. Osteopontin involvement in
integrin-mediated cell signaling and regulation of
expression of alkaline phosphatase during early
differentiation of UMR cells. FEBS Lett. 1997, 420,
112–116.

36. Uemura, T.; Nemoto, A.; Liu, Y.-K.; Kojima, H.; Dong,
J.; Yabe, T.; Yoshikawa, T.; Ohgushi, H.; Ushida, T.;
Tateishi, T. Osteopontin involvement in bone remodeling and
its effects on in vivo osteogenic potential of bone
marrow-derived osteoblasts/porous hydroxyapatite
constructs. Mater. Sci. Eng. 2001, C17, 33–36.

37. Uemura, T.; Nemoto, A.; An, J.; Liu, Y.-K.; Yoshikawa,
T.; Ohgushi, H.; Kuboki, Y.; Ushida, T.; Tateishi, T.
Osteopontin involvement in osteoblast differentiation and
its effect on in vivo osteogenic potential of bone marrow
derived osteoblasts. Bioceramics 1999, 6, 127–129.

38. Buttler, W.T.; Ridall, A.L.; McKee, M.D. Osteopontin.


In Principles of Bone Biology; Bilezikian, J.P., Raisz,
L.G., Rodan, G.A., Eds.; Academic Press, 1996; 167– 181.

39. Butler, W.T. Dentin matrix proteins and dentinogenesis.


Connect. Tissue Res. 1995, 33, 59–65.

40. Boskey, A.L. Mineral-matrix interactions in bone and


cartilage. Clin. Orthop. 1992, 281, 244–274.

41. Boskey, A.L.; Maresca, M.; Doty, S.; Sabsay, B.; Veis,
A. Concentration-dependent effects of dentin phosphophoryn
in the regulation of in vitro hydroxyapatite formation and
growth. Bone Miner. 1990, 11, 55–65.

42. Campbell, A.A.; Ebrahimpour, A.; Perez, L.; Smesko,


S.A.; Nancollas, G.H. The dual role of polyelectrolytes and
proteins as mineralization promoters and inhibitors of
calcium oxalate monohydrate. Calcif. Tissue Int. 1989, 45,
122–128.

43. Weinstock, M.; Leblond, C.P. Radioautographic


visualization of the deposition of a phosphoprotein at the
mineralization front in the dentin of rat incisor. J. Cell
Biol. 1973, 56, 838–845.

44. Kuboki, Y.; Fujisawa, R.; Aoyama, K.; Sasaki, S.


Calcium-specific precipitation of dentin phosphoprotein: A
new method of purification and the significance for the
mechanism of calcification. J. Dent. Res. 1979 , 58,
1926–1932. 45. Marsh, M.E. Self-association of calcium and
magnesium complexes of dentin phosphophoryn. Biochemistry
1989, 28, 339–345. 46. Butler, W.T. Dentin Collagen:
Chemical Structure and Role in Mineralization. In Dentin
and Dentinogenesis; Linde, A., Ed.; CRC Press: Boca Raton,
1984; 37–54. 47. Stetler-Stevenson, W.G.; Veis, A. Type I
collagen shows a specific binding affinity for bovine dentin
phosphophoryn. Calcif. Tissue Int. 1986, 38, 135–141. 48.
Traub, W.; Jodaikin, A.; Arad, T.; Veis, A.; Sabsay, B.
Dentin phosphophoryn binding to collagen fibrils. Matrix
1992, 12, 197–201. 49. Fujisawa, R.; Kuboki, Y. Affinity of
bone sialoprotein and several other bone and dentin acidic
proteins to collagen fibrils. Calcif. Tissue Int. 1992, 51,
438–442. 50. Saito, T.; Arsenault, A.L.; Yamauchi, M.;
Kuboki, Y.; Crenshaw, M.A. Mineral induction by immobilized
phosphoproteins. Bone 1997, 21, 305–311. 51. Saito, T.;
Yamauchi, M.; Crenshaw, M.A. Apatite induction by insoluble
dentin collagen. J. Bone Miner. Res. 1998, 13, 265–270. 52.
Saito, T.; Yamauchi, M.; Abiko, Y.; Matsuda, K.; Crenshaw,
M.A. In vitro apatite induction by phosphophoryn
immobilized to collagen fibrils. J. Bone Miner. Res. 2000,
15, 1615–1619. 53. Maniatopoulos, C.; Sodek, J.; Melcher,
A.H. Bone formation in vitro by stromal cells obtained from
marrow of young adult rats. Cell Tissue Res. 1988, 254,
317–330. 54. Uemura, T.; Dong, J.; Wang, Y.C.; Kojima, H.;
Saito, T.; Iejima, D.; Kikuchi, M.; Tanaka, J.; Tateishi,
T. Transplantation of cultured bone cells using
combinations of scaffolds and culture techniques.
Biomaterials 2003, 24, 2277–2286. 55. Hench, L.L.;
Splinter, R.J.; Allen, W.C.; Greenlee, T.K. Bonding
mechanisms at the interface of ceramic prosthetic
materials. J. Biomed. Mater. Res. Symp. 1971, 2, 117–141.
56. Wilson, J.; Yli-Urpo, A.; Risto-Pekka, H. Bioactive
Glasses: Clinical Applications. In An Introduction to
Bioceramics; Hench, L.L., Wilson, J., Eds.; World
Scientific: Singapore, 1993; 63–74. 57. Kokubo, T.; Kim,
H.M.; Kawashita, M. Novel bioactive materials with
different mechanical properties. Biomaterials 2003, 24
(13), 2161–2175. 58. Glowacki, J.; Altobelli, D.; Mulliken,
J.B. Fate of mineralized and demineralized osseous implants
in cranial defects. Calcif. Tissue Int. 1981, 33 (1),
71–76. 59. Ray, R.D.; Holloway, J.A. Bone implants.
Preliminary results of an experimental study. J. Bone Jt.
Surg. 1957, 39A , 1119–1128. 60. Mulliken, J.B.; Glowacki,
J. Induced osteogenesis for repair and construction in the
craniofacial region. Plast. Reconstr. Surg. 1980, 65,
553–559. 61. Reddi, A.H.; Huggins, C. Biochemical sequences
in the transformation of normal fibroblasts in adolescent
rats. Proc. Natl. Acad. Sci. U. S. A. Jun. 1972, 69 (6),
1601–1605. 62. Gepstein, R.; Weiss, R.E.; Hallel, T.
Bridging large defects in bone by demineralized bone matrix
in the O form of a powder. A radiographic, histological,
and radioisotope-uptake study in rats. J. Bone Jt. Surg.,
Am. 1987, 69 (7), 984–992. 63. Glowacki, J.; Kaban, L.B.;
Murray, J.E.; Folkman, J.; Mulliken, J.B. Application of
the biological principle of induced osteogenesis for
craniofacial defects. Lancet 1981, 1 (8227), 959–962. 2.
64. Kaban, L.B.; Mulliken, J.B.; Glowacki, J. Treatment of
jaw defects with demineralized bone implants. J. Oral
Maxillofac. Surg. 1982, 40 (10), 623–626. 65. Sonis, S.T.;
Kaban, L.B.; Glowacki, J. Clinical trial of demineralized
bone powder in the treatment of periodontal defects. J.
Oral Med. 1983, 38 (3), 117–122. 66. Upton, J.; Boyajian,
M.; Mulliken, J.B.; Glowacki, J. The use of demineralized
xenogeneic bone implants to correct phalangeal defects: A
case report. J. Hand Surg., Am. 1984, 9 (3), 388–391. 67.
Upton, J.; Glowacki, J. Hand reconstruction with allograft
demineralized bone: Twenty-six implants in twelve patients.
J. Hand Surg., Am. 1992, 17 (4), 704– 713. 68. Neo, M.;
Kotani, S.; Fujita, Y.; Nakamura, T.; Yamamuro, T.; Bando,
Y.; Ohtsuki, C.; Kokubo, T. Differences in ceramic-bone
interface between surfaceactive ceramics and resorbable
ceramics: A study by scanning and transmission electron
microscopy. J. Biomed. Mater. Res. 1992, 26, 255–267. 69.
Kitsugi, T.; Yamamuro, T.; Nakamura, T.; Kotani, S.;
Kokubo, T.; Takeuchi, H. Four calcium phosphate ceramics as
bone substitutes for non-weight-bearing. Biomaterials 1993,
14, 216–224. 70. Yang, Z.; Yuan, H.; Tong, W.; Zou, P.;
Chen, W.; Zhang, X. Osteogenesis in extraskeletally
implanted porous calcium phosphate ceramics: Variability
among different kinds of animals. Biomaterials 1996, 17,
2131– 2137. 71. Ohura, K.; Bohner, M.; Hardouin, P.;
Lemaitre, J.; Pasquier, G.; Flautre, B. Resorption of, and
bone formation from, new b-tricalcium phosphate-monocalcium
phosphate cements: An in vivo study. J. Biomed. Mater. Res.
1996, 30, 193–200. 72. Ayers, R.A.; Simske, S.J.; Nunes,
C.R.; Wolford, L.M. Long-term bone ingrowth and residual
microhardness of porous block hydroxyapatite implants in
humans. J. Oral Maxillofac. Surg. 1998, 56, 1297–1301. 73.
Tancred, D.C.; McCormack, B.A.; Carr, A.J. A synthetic bone
implant macroscopically identical to cancellous bone.
Biomaterials 1998, 19, 2303–2311. 74. Furusawa, T.;
Mizunuma, K.; Yamashita, S.; Takahashi, T. Investigation of
early bone formation using resorbable bioactive glass in
the rat mandible. Int. J. Oral Maxillofac. Implants 1998,
13, 672–676. 75. Ohsawa, K.; Neo, M.; Matsuoka, H.;
Akiyama, H.; Ito, H.; Kohno, H.; Nakamura, T. The
expression of bone matrix protein mRNAs around beta-TCP
particles implanted into bone. J. Biomed. Mater. Res. 2000
, 52, 460–466. 76. Merten, H.A.; Wiltfang, J.; Grohmann,
U.; Hoenig, J.F. Intraindividual comparative animal study
of alphaand b-tricalcium phosphate degradation in
conjunction with simultaneous insertion of dental implants.
J. Craniofac. Surg. 2001, 12, 59–68. 77. Kurashina, K.;
Kurita, H.; Wu, Q.; Ohtsuka, A.; Kobayashi, H. Ectopic
osteogenesis with biphasic ceramics of hydroxyapatite and
tricalcium phosphate in rabbits. Biomaterial 2002, 2 (23),
407–412. 78. Grenga, T.E.; Zins, J.E.; Bauer, T.W. The rate
of vascularization of coralline hydroxyapatite. Plast.
Reconstr. Surg. 1989, 84, 245–249. 79. Schliephake, H.;
Neukam, F.W.; Klosa, D. Influence of pore dimensions on bone
ingrowth into porous hydroxyapatite blocks used as bone
graft substitutes. An isometric study. Int. J. Oral
Maxillofac. Surg. 1991, 20, 53–58. 80. Tancred, D.C.;
McCormack, B.A.; Carr, A.J. A synthetic bone implant
macroscopically identical to cancellous bone. Biomaterials
1998, 19, 2303–2311. 81. Saito, M.; Shimizu, H.; Beppu, M.;
Takagi, M. The role of b-tricalcium phosphate in
vascularized periosteum. J. Orthop. Sci. 2000, 5, 275–282.
82. Schliephake, H.; Kage, T. Enhancement of bone
regeneration using resorbable ceramics and a polymerceramic
composite material. J. Biomed. Mater. Res. 2001, 56,
128–136. 83. Nicholas, R.W.; Lange, T.A. Granular
tricalcium phosphate grafting of cavitary lesions in human
bone. Clin. Orthop. 1994, 306, 197–203. 84. Nery, E.B.;
LeGeros, R.Z.; Lynch, K.L.; Lee, K. Tissue response to
biphasic calcium phosphate ceramic with different ratios of
HA/b TCP in periodontal osseous defects. J. Periodontol.
1992, 63, 729–735. 85. Johnson, K.D.; Frierson, K.E.;
Keller, T.S.; Cook, C.; Scheinberg, R.; Zerwekh, J.;
Meyers, L.; Sciadini, M.F. Porous ceramics as bone graft
substitutes in long bone defects: A biomechanical,
histological, and radiographic analysis. J. Orthop. Res.
1996, 14, 351–369. 86. Krebsbach, P.H.; Kuznetsov, S.A.;
Satomura, K.; Emmons, R.V.; Rowe, D.W.; Robey, P.G. Bone
formation in vivo: Comparison of osteogenesis by
transplanted mouse and human marrow stromal fibroblasts.
Transplantation 1997, 63, 1059–1069. 87. Wang, J.; Chen,
W.; Li, Y.; Fan, S.; Weng, J.; Zhang, X. Biological
evaluation of biphasic calcium phosphate ceramic vertebral
laminae. Biomaterials 1998, 19, 1387–1392. 88. Alam, I.;
Asahina, I.; Ohmamiuda, K.; Enomoto, S. Comparative study
of biphasic calcium phosphate ceramics impregnated with
rhBMP-2 as bone substitutes. J. Biomed. Mater. Res. 2001 ,
54, 129–138. 89. Yokoyama, N.; Minabe, M.; Sugaya, A.;
Hori, T.; Otaki, K.; Hasegawa, A.; Irie, H.; Hakamatsuka,
Y. Application of b-tricalcium phosphate to periodontal
therapy. Part 1. Fundamental studies and biological tests
of b-TCP prepared by mechanochemical method. Nippon
Shishubyo Gakkai Kaishi 1989, 31, 213–223. 90. Dong, J.;
Uemura, T.; Shirasaki, Y.; Tateishi, T. Promotion of bone
formation using highly pure porous beta-TCP combined with
bone marrow-derived osteoprogenitor cells. Biomaterials
2002, 23 (23), 4493–4502.
Particulate Matter and Host Reactions

1. Wright, T.M.; Goodman, S.B. Implant wear in total joint


replacement. Rosemount, Ill: J. Am. Acad. Orthop. Surg.
2001, 224.

2. Bauer, T. Particles and periimplant bone resorption.


Clin. Orthop. Rel. Res. 2002, 405, 138–143.

3. Dorr, L.D.; Wan, Z.; Shahrdar, C.; Sirianni, L.;


Boutary, M.; Yun, A. Clinical performance of a Durasul
highly cross-linked polyethylene acetabular liner for total
hip arthroplasty at five years. J. Bone Joint Surg. 2005,
87A, 1816–1821.

4. Ingram, J.; Stone, M.; Fisher, J.; Ingham, E. The


influence of molecular weight, crosslinking and counterface
roughness on TNF-a production by macrophages in response to
ultra high molecular weight polyethylene particles.
Biomaterials 2004, 25, 3511–3522.

5. Greenfield, E.M.; Bi, Y.; Ragab, A.A.; Goldberg, V.M.;


Van De Motter, R.R. The role of osteoclast differentiation
in aseptic loosening. J. Orthop. Res. 2002, 20, 1–8.

6. Greenfield, E.M.; Bi, Y.; Ragab, A.A.; Goldberg, V.M.;


Nalepka, J.L.; Seabold, J.M. Does endotoxin contribute to
aseptic loosening of orthopaedic implants? J. Biomed.
Mater. Res. B. Appl. Biomater. 2005, 72B, 179–185.

7. Kobayashi, S.; Takaoka, K.; Saito, N.; Hisa, K. Factors


affecting aseptic failure of fixation after primary Charnley
total hip arthroplasty. J. Bone Joint Surg. 1997, 79A,
1618–1627.

8. Wilkinson, J.M.; Hamer, A.J.; Stockley, I.; Eastell, R.


Polyethylene wear rate and osteolysis: critical threshold
versus continuous dose-response relationship. J. Orthop.
Res. 2005, 23, 520–525.

9. Haynes, D.R.; Boyle, S.J.; Rogers, S.D.; Howie, D.W.;


Vernon-Roberts, B. Variation in cytokines induced by
particles from different prosthetic materials. Clin.
Orthop. Rel. Res. 1998, 352, 223–230.

10. Taki, N.; Tatro, J.M.; Nalepka, J.L.; Togawa, D.;


Goldberg, V.M.; Rimnac, C.M.; Greenfield, E.M. Polyethylene
and titanium particles induce osteolysis by similar,
lymphocyte-independent, mechanisms. J. Orthop. Res. 2005,
23, 376–383.
11. Beidelschies, M.; Islam, A.; Smith, M.; Goldberg, V.;
Chen, X.; Greenfield, E. ERK1/2 activation by orthopaedic
wear particles is strongly stimulated by adherent endotoxin
and is required for stimulation of TNFa production. 2005,
submitted.

12. Wilkinson, J.M.; Wilson, A.; Stockley, I.; Scott, I.;


Macdonald, D.; Hamer, A.; Duff, G.; Eastell, R. Variation
in the TNF gene promoter and risk of osteolysis after total
hip arthroplasty. J. Bone Miner. Res. 2003, 18, 1995–2001.

13. Gordon, A.; Wilkinson, J.M.; Wilson, A.G.; Stockley,


I.; MacDonald, D.; Eastell, R. Polymorphisms in the
interleukin-one gene cluster and the risk of aseptic
loosening after total hip arthroplasty. J. Bone Miner. Res.
2003, 18 (S2), 326.

14. Smith, M.; Innocenti, M.; Lee, M.; Rohrer, J.;


Sobieraj, M.; Goldberg, V.; Greenfield, E. The PI3K/Akt
pathway mediates the biological activity of titanium
particles. Trans. Orthop. Res. Soc. 2005, 30, 1619. 15.
Wei, X.; Zhang, X.; Zuscik, M.; Drissi, M.; Schwarz, E.;
O’keefe, R. Fibroblasts express RANKL and support
osteoclastogenesis in a COX-2-dependent manner after
stimulation with titanium particles. J. Bone Miner. Res.
2005, 20, 1136–1148. 16. Jiranek, W.A.; Machado, M.; Jasty,
M.; Jevsevar, D.; Wofe, H.J.; Goldring, S.R.; Goldberg,
M.J.; Harris, W.H. Production of cytokines around loosened
cemented acetabular components. J. Bone Joint Surg. 1994,
75A, 863–879. 17. Hallab, N.; Merritt, K.; Jacobs, J. Metal
sensitivity in patients with orthopaedic implants. J. Bone
Joint Surg. 2001, 83A, 428–436. 18. Davies, A.; Willert,
H.; Cambell, P.; Learmouth, I.; Case, C. An unusual
lymphocytic perivascular infiltration in tissue around
contemporary metal-on-metal joiunt replacements. J. Bone
Joint Surg. 2005, 87A, 18–27. 19. Park, Y.; Moon, Y.; Lim,
S.; Yang, J.; Ahn, G.; Choi, Y. Early osteolysis following
second-generation metalon-metal hip replacement. J. Bone
Joint Surg. 2005, 87A, 1515–1521. 20. Willert, H.;
Buchhorn, G.; Fayyazi, A.; Flury, R.; Windler, M.; Koster,
G.; Lohmann, C. Metal-on-metal bearings and
hypersensitivity in patients with artificial joints. J. Bone
Joint Surg. 2005, 87A, 28–36. 21. Nanes, M.; Pacifici, R.
Inflammatory cytokines. In Bone Resorption; Bronner, F.,
Farach-Carson, M., Rubin, J., Eds.; London: Springer, 2005;
67–90. 22. Skripitz, R.; Bohling, S.; Ruther, W.;
Aspenberg, P. Stimulation of implant fixation by parathyroid
hormone (1-34)-A histomorphometric comparison of PMMA
cement and stainless steel. J. Orthop. Res. 2005, 23,
1266–1270. 23. Fleischmann, R.; Iqbal, I.; Stern, R.
Considerations with the use of biological therapy in the
treatment of rheumatoid arthritis. Expert Opin. Drug Saf.
2004, 3, 391–403. 24. Kurth, A.; Eberhardt, C.; Muller, S.;
Steinacker, M.; Schwarz, M.; Bauss, F. The bisphosphonate
ibandronate improves implant integration in osteopenic
ovariectomized rats. Bone 2005, 37, 204–210. 25. Peter, B.;
Pioletti, D.; Laib, S.; Bujoli, B.; Pilet, P.; Janvier, P.;
Guicheux, J.; Zambelli, P.; Bouler, J.; Gauthier, O.
Calcium phosphate drug delivery system: influence of local
zoledronate release on bone implant osteointegration. Bone
2005, 36, 52–60. 26. Bhandari, M.; Bajammal, S.; Guyatt,
G.; Griffith, L.; Busse, J.; Scunemann, H.; Einhorn, T.
Effect of bisphosphonates on periprosthetic bone mineral
density after total joint arthroplasty. A meta-analysis. J.
Bone Joint Surg. 2005, 87A, 293–301. 27. Wilkinson, J.;
Eagleton, A.; Stockley, I.; Peel, N.; Hamer, A.; Eastell,
R. Effect of pamidronate on bone turnover and implant
migration after total hip arthroplasty: a randomized trial.
J. Orthop. Res. 2005, 23, 1–8. 28. Albagha, O.; Ralston, S.
Genetic determinants of susceptibility to osteoporosis.
Endocrinol. Metab. Clin. North Am. 2003, 32, 65–81. 29.
Engesaeter, L.B.; Lie, S.A.; Espehaug, B.; Furnes, O.;
Vollset, S.E.; Havelin, L.I. Antibiotic prophylaxis in
total hip arthroplasty. Acta. Orthop. Scand. 2003, 74,
644–651. 30. Parvizi, J.; Wickstrom, E.; Zeiger, A.; Adams,
C.; Shapiro, I.; Purtill, J.; Sharkey, P.; Hozack, W.;
Rothman, R.; Hickok, N. Titanium surface with biologic
activity against infection. Clin. Orthop. Rel. Res. 2004,
429, 33–38. 31. Bhumbra, R.; Walker, P.; Berman, A.;
Emmanual, J.; Barrett, D.; Blunn, G. Prevention of
loosening in total hip replacements using guided bone
regeneration. Clin. Orthop. Rel. Res. 2000, 372, 192–204. P
Peripheral Nerve Repair and Regeneration:
Historical Perspective

45. Theriault, M.; Dort, J.; Sutherland, G.; Zochodne, D.W.


A prospective quantitative study of sensory deficits after
whole sural nerve biopsies in diabetic and nondiabetic
patients. Surgical approach and the role of collateral
sprouting. Neurol. February 1998, 50 (2), 480–484.

46. LaStayo, P.C.; Wheeler, D.L. Reliability of passive


wrist flexion and extension goniometric measurements: A
multicenter study. Physical Therapy 1994, 74 (2), 162–176.

47. Daniels, L.; Worthingham, C. Muscle Testing; Techniques


of Manual Examination, 5th Ed.; W.B. Saunders:
Philadelphia, 1986.

48. Jebsen, R.H.; Taylor, N.; Trieschmann, R.B.; Trotter,


M.J.; Howard, L.A. An objective and standardized test of
hand function. Arch. Phys. Med. Rehabil. 1969, 56, 311–319.

49. Lin, F.M.; Pan, Y.C.; Hom, C.; Sabbahi, M.; Shenaq, S.
Ankle stance angle: a functional index for the evaluation
of sciatic nerve recovery after complete transection. J.
Reconstr. Microsurg. April 1996, 12 (3), 173–177.

50. Jones, E.; Hanly, J.G.; Mooney, R.; Rand, L.L.;


Spurway, P.M.; Eastwood, B.J.; Jones, J.V. Strength and
function in the normal and rheumatoid hand. J. Rheumatol.
1991, 18, 1313–1318.

51. Greenleaf EVALtm system. http://www.steeval.com/.

52. Doyle, J.R. Factors Affecting Clinical Results of Nerve


Suture. In Nerve Repair and Regeneration, Its Clinical and
Experimental Basis; Jewett, D.L., McCarroll, H.R., Jr.
Eds.; C.V. Mosby: St. Louis, 1980; 263–266.

53. Houle, J.D.; Ye, J.H. Changes occur in the ability to


promote axonal regeneration as the post injury period
increases. Neuro. Report 1997, 8, 751–755.

54. Dellon, A.L. Wound healing in nerve. Clinics in Plastic


Surgery 1990, 17, 545–570.

55. Cairns, H.; Young, J.Z. Treatment of gunshot wounds of


peripheral nerves. Lancet. 3 August 1940, 123–130.

56. Millesi, H. The interfascicular nerve grafting of the


median and ulnar nerves. J. Bone Joint Surg. Am. 1972, 54,
727–748.

57. Waller, A.V. Experiments on the glossopharyngeal and


hypoglossal nerves of the frog and observations produced
thereby in the structure of their primative fibres. Phil.
Trans. R. Soc. Lond. 1850, 140, 423.

58. Mitchell, S.W.; Morehouse, G.R.; Keen, W.W. Gunshot


Wounds and Other Injuries of Nerves; J.B. Lippincott:
Philadelphia, 1864.

59. Langley, J.N.; Hashimoto, M. On the suture of separate


nerve bundles in a nerve trunk and on internal nerve
plexus. J. Phys. (Lond.) 1917, 51.

60. Smith, J.W. Microsurgery of peripheral nerves. Plast.


Reconstr. Surg. 1964, 33, 317.

61. Woodhall, B.; Beebe, W.G. In Peripheral Nerve


Regeneration: A Follow-up Study of 3,656 World War II
Injuries, VA Medical Monograph; US Government Printing
Office: Washington, DC, 1956.

62. Weiss, P.A. From Cell Research to Nerve Repair, 2nd


Ed.; Futura Publishing: Mt. Kisco, New York, 1976; 316. 63.
Weiss, P.; Taylor, A.C. Further experimental evidence
against ‘‘neurotropism’’ in nerve regeneration. J. Exp.
Zool. 1944, 95, 289–300. 64. Seckel, B.R.; Ryan, S.E.;
Gagne, R.G.; Chiu, T.H.; Watkins, E., Jr. Target-specific
nerve regeneration through a nerve guide in the rat. Plast.
Reconstr. Surg. 1986, 78 (6), 793–800. 65. Brushart, T.M.;
Seiler, W.A. Selective reinervation of distal motor stumps
by peripheral motor axons. Exp. Neurol. 1987, 97, 289–300.
66. Lundborg, G. Nerve Injury and Repair; Longman Group UK,
Ltd.: New York, 1988; 64–70, 158–178, 196–199. 67. Kallio,
P.K.; Vastamaki, M.; Kolonen, K.A. The results of secondary
microsurgical repair of radial nerve in 33 patients. J.
Hand Surg. 1993, 18B, 320–322. 68. Capek, L.; Clarke, H.M.;
Zuker, R.M. Endoscopic sural nerve harvest in the pediatric
patient. Plast. Reconstr. Surg. October 1996, 98 (5),
884–888. 69. Jaroszynski, G.; Johnston, G.H. Harvesting of
the sural nerve with a tendon stripper. Microsurg. 1996, 17
(4), 217–220. 70. Terzis, J.K. Principles and Techniques of
Peripheral Nerve Surgery. In Reconstructive Microsurgery;
Daniel, R.K., Terzis, J.K., Eds.; Little Brown: Boston,
1977; 387–390. 71. Hentz, V.R.; Rosen, J.M.; Xiao, S.;
McGill, K.C.; Abraham, G.A. The nerve gap dilemma. A study
comparing nerves repaired end-to-end under tension to
tension-free nerve graft techniques in a clinically
relevant primate model. J. Hand Surg. 1993, 18, 417– 425.
72. Staniforth, P.; Fisher, T.R. The effects of sural nerve
excision in autogenous nerve grafting. The Hand 1978, 10,
187–190. 73. Stancic, M.F.; Micovic, V.; Bobinac, D.;
Starcevic, G.; Fuzinac, A.; Tomljanovic, Z.
Electromyographic evaluation of experimental nerve grafts
suggests better recovery with microscope assistance.
Pflugers Arch. 1996, 431 (6 Suppl. 2), R285–R286. 74. Chase,
R.A. Atlas of Hand Surgery, Vol. 2; W.B. Saunders:
Philadelphia, 1984; 78–91. 75. Shaw-Wilgis, E.F. Nerve
Repair and Grafting. In Operative Hand Surgery, 2nd Ed.,
Vol. 2; Green, D.P., Ed.; Churchill Livingstone: New York,
1988; 1373–1403. 76. Zhao, S.; Beuerman, R.W.; Kline, D.G.
Neurotization of motor nerves innervating the lower
extremity by utilizing the lower intercostal nerves. J.
Reconstr. Microsurg. January 1997, 13 (1), 39–45. 77.
Mucci, S.; Dellon, A.L. Restoration of lower lip sensation:
Neurotization of the mental nerve with the supraclavicular
nerve. J. Reconstr. Microsurg. 1997, 13, 151–155. 78.
London, P.S. Neuroma and neurosis in the hand.
Handchirurgie 1972, 4, 103–104. 79. Zehr, E.P.; Stein,
R.B.; Komiyama, T. Function of sural nerve reflexes during
human walking. J. Physiol. (Lond.) February 1998, 507 (Pt.
1), 305–314. 80. Allieu, Y.; Chammas, M.; Picot, M.C.
Paralysis of the brachial plexus caused by supraclavicular
injuries in Peripheral the adult. Long-term comparative
results of nerve grafts and transfers. Rev. Chir. Orthop.
Reparatrice Appar. Mot. 1997, 83 (1), 51–59. 81. Madison,
R.D.; Archibald, S.J.; Krarup, C. Peripheral Nerve Injury.
In Wound Healing. Biochemical and Clinical Aspects; Cohen,
I.K., Diegelmann, R.F., Lindblad, W.J., Eds.; W.B.
Saunders: Philadelphia, 1992; 450–487. 82. Rosen, J.M.
Concepts of peripheral nerve repair. Ann. Plast. Surg.
1981, 7, 165. 83. Sunderland, S. Nerves and Nerve Injuries,
2nd Ed.; Churchill Livingston: New York, 1978; 423–428. 84.
Fields, R.D.; Le Beau, J.M.; Longo, F.M.; Ellisman, M.H.
Nerve regeneration through artificial tubular implants.
Prog. Neurobiol. 1989, 33, 87. 85. Katsube, K.; Doi, K.;
Fukumoto, T.; Fujikura, Y.; Shigetomi, M.; Kawai, S. Nerve
regeneration and origin of Schwann cells in peripheral
nerve allografts in immunologically pretreated rats.
Transplantation 15 December 1996, 62 (11), 1643–1649. 86.
Midha, R.; Munro, C.A.; Mackinnon, S.E.; Ang, L.C. Motor
and sensory specificity of host nerve axons influence nerve
allograft rejection. J. Neuropathol. Exp. Neurol. April
1997, 56 (4), 421–434. 87. Dumont, C.E.; Hentz, V.R.
Enhancement of axon growth by detergent-extracted nerve
grafts. Transplantation 15 May 1997, 63 (9), 1210–1215. 88.
Sondell, M.; Lundborg, G.; Kanje, M. Regeneration of the
rat sciatic nerve into allografts made acellular through
chemical extraction. Brain Res. June 1998, 795 (1–2),
44–54. 89. Chiu, D.T.W.; Janecka, I.; Krizek, T.J.; Wolff,
M.; Lovelace, R.E. Autogenous vein graft as a conduit for
nerve regeneration. Surg. 1982, 91, 226–233. 90. Strauch,
B.; Ferder, M.; Lovelle-Allen, S.; Moore, K.; Kim, D.J.;
Llena, J. Determining the maximal length of a vein conduit
used as an interposition graft for nerve regeneration. J.
Reconstr. Microsurg. November 1996, 12 (8), 521–527. 91.
Lundborg, G.; Dahlin, L.B.; Danielsen, N.P.; et al.
Reorganization and orientation of regenerating nerve fibres,
perineurium, and epineurium in preformed mesothelial tubes.
An experimental study on the sciatic nerve of rats. J.
Neurosci. Res. 1981, 6, 265. 92. Herndon, J.H. Neuromas. In
Operative Hand Surgery, 2nd Ed.; Green, D.P., Ed.;
Churchill Livingstone: New York, 1988; 1405–1422. 93.
Lundborg, G.; Kanje, M. Bioartificial nerve grafts. A
prototype. Scand. J. Plast. Reconstr. Surg. Hand Surg. June
1996, 30 (2), 105–110. 94. Strauch, B. Use of nerve
conduits in peripheral nerve repair. Hand Clin. February
2000, 16 (1), 123–130. 95. Ellis, D.L.; Yannas, I.V. Recent
advances in tissue synthesis in vivo by use of
collagen-glycosaminoglycan copolymers. Biomaterials
February 1996, 17 (3), 291–299. 96. Integra.
http://www.integra-ls.com/products/?product= 88. 97.
Mackinnon, S.E.; Dellon, A.L. A study of nerve regeneration
across synthetic (Maxon) and biological (collagen) nerve
conduits for nerve gaps up to 5 cm in the primate. J.
Reconstr. Microsurg. 1990, 6, 117–121. 98. Woerly, S.;
Plant, G.W.; Harvey, A.R. Neural tissue engineering: From
polymer to biohybrid organs. Biomaterials February 1996, 17
(3), 301–310. 99. Jenq, C.-B.; Coggeshall, R.E. Nerve
regeneration through holey silicone tubes. Brain Res. 1985,
361, 233–241. 100. Rich, K.M.; Alexander, T.D.; Pryor,
J.C.; Hollowell, J.P. Nerve growth factor enhances
regeneration through silicone chambers. Exper. Neurol.
1989, 105, 162–170. 101. Borkenhagen, B.; Stoll, R.C.;
Neuenschwander, P.; Suter, U.W.; Aebischer, P. In vivo
performance of a new biodegradable polyesterurethane system
used as a nerve guidance channel. Biomaterials 1998, 19
(23), 2154–2166. 102. Merle, M.; Dellon, A.; Campbell, J.;
Chang, P. Complications from silicon-polymer intubulation
of nerves. Microsurg. 1989, 10, 130–133. 103. Archibald,
S.J.; Krarup, C.; Shefner, J.; Li, S.-T.; Madison, R.D. A
collagen-based nerve guide conduit for peripheral nerve
repair: An electrophysiological study of nerve regeneration
in rodents and nonhuman primates. J. Compar. Neurol. 1991,
306, 685–696. 104. Kline, D.G.; Hayes, G.J. The use of a
resorbable wrapper for peripheral nerve repair. J.
Neurosurg. 1964, 21, 737. 105. Crawley, W.A.; Dellon, A.L.
Inferior alveolar nerve reconstruction with a polyglycolic
acid bioabsorbable nerve conduit. Plast. Reconstr. Surg.
1992, 90, 300–302. 106. Rosen, J.M.; Padilla, J.A.; Nguyen,
K.D.; Siedman, J.; Pham, H.N. Artificial nerve graft using
glycolide trimethylene carbonate as a nerve conduit filled
with collagen compared to sutured autograft in a rat model.
J. Rehabil. Res. Devel. 1992, 29 (2), 1–12. 107. den
Dunnen, W.F.A.; van der Lei, B.; Robinson, P.H.; Holwerda,
A.; Pennings, A.J.; Schakenraad, J.M. Biological
performance of a degradable poly(lactic
acid-e-caprolactone) nerve guide: Influence of tube
dimensions. J. Biomed. Mater. Res. 1995, 29, 757–766. 108.
Satou, T.; Nishida, S.; Hiruma, S.; Tanji, K.; Takahashi,
M.; Fujita, S.; Mizuhara, Y.; Akai, F.; Hashimoto, S. A
morphological study on the effects of collagen gel matrix
on regeneration of severed rat sciatic nerve in silicone
tubes. Acta Pathol. Jpn. 1986, 36 (2), 199–208. 109.
Miller, E.J.; Gay, E.S. The Collagens: an overview and
update. Methods in Enzymol. 1987, 144, 4–40. 110. Seyer,
J.M.; Kang, A.H.; Whitaker, J.N. The characterization of
type I and type III of collagens from human peripheral
nerve. BioChim. Biophys. Acta 1977, 492, 415–425. 111.
Junqueira, L.C.U.; Montes, G.S.; Bezerra, M.S.F. Do Schwann
cells produce collagen type III? Experientia 1978, 35, 114.
112. Sabelman, E.E. Biology, Biotechnology and
Biocompatibility of Collagen. In Biocompatibility of Tissue
Analogs, Vol. 1; Williams, D.F, Ed.; CRC Press: Boca Raton,
Florida, 1985; 27–66. P

113. Reiser, K.; McCormick, R.J.; Rucker, R.B. Enzymatic


and nonenzymatic cross-linking of collagen and elastin.
FASEB J. 1992, 6 (7), 2439–2449.

114. Madison, R.; Da Silva, C.; Dikkes, P. Entubulation


repair with protein additives increases the maximum nerve
gap distance successfully bridged with tubular prostheses.
Brain Res. 1988, 447, 325–334.

115. Rosen, J.M.; Padilla, J.A.; Nguyen, K.D.; Padilla,


M.A.; Sabelman, E.E.; Pham, H.N. Artificial Nerve graft
using collagen as an extracellular matrix for nerve repair
compared with sutured autograft in a rat model. Annals of
Plast. Surg. 1990, 25 (5), 375–387.

116. Satou, T.; Nishida, S.; Hiruma, S.; Tanji, K.;


Takahashi, M.; Fujita, S.; Mizuhara, Y.; Akai, F.;
Hashimoto, S. A morphological study on the effects of
collagen gel matrix on regeneration of severed rat sciatic
nerve in silicone tubes. Acta Pathol. Jpn. 1986, 36 (2),
199–208.

117. Colin, W.; Donoff, R.B. Nerve regeneration through


collagen tubes. J. Dental Res. 1984, 63, 987.

118. Takahashi, M.; Satou, T.; Hashimoto, S. Experimental


in vivo regeneration of peripheral nerve axons and
perineurium guided by resorbable collagen film. Acta Pathol.
Jpn. 1988, 38, 489–502.

119. Liu, S.; Bodjarian, N.; Langlois, O.; Bonnard, A.S.;


Boisset, N.; Peulve`, P.; Saoˆd, G.; Tadie`, M. Axonal
regrowth through a collagen guidance channel bridging
spinal cord to the avulsed C6 roots: functional recovery in
primates with brachial plexus injury. J. Neurosci. Res.
March 1998, 51 (6), 723–734.

120. Aguayo, A.J.; Bray, G.M.; Perkins, C.S.; Duncan, I.D.


Axon-sheath cell interactions in peripheral and central
nervous system transplants. Soc. Neurosci. Symp. 1979, 4,
361–383.

121. Griffiths, R.; Horch, K.; Stensaas, L. A collagen and


fibrin tube for nerve repair. Rest. Neuro. and Neurosci.
1990, 1, 339–346.

122. Eccleston, P.A.; Mirsky, R.; Jessen, K.R. Type I


collagen preparations inhibit DNA synthesis in glial cells
of the peripheral nervous system. Exp. Cell Res. 1989, 182,
173–185.

123. Duby, N.; Letourneau, P.C.; Tranquillo, R.T. Enhanced


neurite and Schwann cell invasion into magnetically-aligned
collagen gel rods for peripheral nerve entubulation repair.
Trans. Soc. for Biomaterials— Symposium: Engineering Neural
Tissue, San Diego, California, April 22–25, 1998.

124. Rizvi, A.H.; Wasserman, A.J.; Zazanis, G.; Silver,


F.H. Evaluation of peripheral nerve regeneration in the
presence of longitudinally aligned collagen fibers. Cells &
Materials 1991, 1, 279–289.

125. Hudson, R.; Evans, G.R.D.; Schmidt, C. Engineering


strategies for peripheral nerve repair. In Clinics Plastic
Surgery, Vol. 31, No. 3; Edington, H., Ed.; W. B. Saunders:
Philadelphia, 2000; 485–497. Reprinted in Orthopaedic
Clinics of North America.

126. Ansselin, A.D.; Fink, T.; Davey, D.F. Peripheral nerve


regeneration through nerve guides seeded with adult Schwann
cells. Neuropathol. Appl. Neurobiol. October 1997, 23 (5),
387–398. 127. Sittinger, M.; Bujia, J.; Rotter, N.;
Reitzel, D.; Minuth, W.W.; Burmester, G.R. Tissue
engineering and autologous transplant formation: practical
approaches with resorbable biomaterials and new cell
culture techniques. Biomaterials February 1996, 17 (3),
237– 242. 128. Levi, A.D.O.; Gue´nard, V.; Aebischer, P.;
Bunge, R. The functional characteristics of Schwann cells
cultured from the human peripheral nerve after
transplantation into a gap within the rat sciatic nerve. J.
Neurosci. 1994, 14, 1309–1319. 129. Sørensen, J.; Haase,
G.; Krarup, C.; Gilgenkrantz, H.; Kahn, A.; Schmalbruch, H.
Gene transfer to Schwann cells after peripheral nerve
injury: a delivery system for therapeutic agents. Ann.
Neurol. February 1998, 43 (2), 205–211. 130. Casella, G.T.;
Bunge, R.P.; Wood, P.M. Improved method for harvesting
human Schwann cells from mature peripheral nerve and
expansion in vitro. Glia 1996, 17, 327. 131. Moretto, G.;
Kim, S.U.; Shin, D.H.; Pleasure, D.E.; Rizzuro, N.
Long-term cultures of human adult Schwann cells isolated
from autopsy materials. Acta Neuropathol. (Berlin) 1984,
64, 15–21. 132. Livesey, F.J.; O’Brien, J.A.; Li, M.;
Smith, A.G.; Murphy, L.J.; Hunt, S.P. A Schwann cell
mitogen accompanying regeneration of motor neurons. Nature
December 1997, 390, 614–618. 133. Sondell, M.;
Fex-Svenningsen, A.; Kanje, M. The insulin-like growth
factors I and II stimulate proliferation of different types
of Schwann cells. Neuroreport September 1997, 8 (13),
2871–2876. 134. Brockes, J.P. Studies of cultured rat
Schwann cells. Establishment of purified populations from
cultures of peripheral nerve. Brain Res. 1979, 165,
105–118. 135. Askanas, V.; Engel, W.K.; Dalakas, M.C.;
Lawrence, J.V.; Carter, L.S. Human Schwann cells in tissue
culture. Histochemical and ultrastructural studies. Arch.
Neurol. 1980, 37, 329–337. 136. Hardman, P.; Spooner, B.S.;
Klement, B.J. A technique for promoting Schwann cell growth
from fresh and frozen biopsy nerve utilizing D-valine
medium (letter). In vitro Cell. & Dev. Biol. 1990, 12 (26),
1116– 1117. 137. Rosen, J.M.; Kaplan, E.N.; Jewett, D.L.
Suture and Sutureless Methods of Repairing Experimental
Nerve Injuries. In Nerve Repair and Regeneration, Its
Clinical and Experimental Basis; Jewett, D.L., McCarroll,
H.R., Jr. Eds.; C.V. Mosby: St.Louis, 1980; 235–242. 138.
Rosen, J.M.; Pham, H.N.; Hentz, V.R. Fascicular
tubulization: A comparison of experimental nerve repair
techniques in the cat. Ann. Plas. Surg. 1989, 22 (6),
467–468. 139. Bain, J.R.; Mackinnon, S.E.; Hunter, D.A.
Functional evaluation of complete sciatic peroneal and
posterior tibial nerve lesions in the rat. Plast. and
Reconstr. Surg. 1989 , 3 (1), 129–136. 140. Keeley, R.D.;
Sabelman, E.; Kadlcik, P.; Agras, J.; Eng, L.; Wiedman,
T-W.; Nguyen, K.; Sudekum, A.; Rosen, J. Synthetic nerve
graft containing collagen and syngeneic Schwann cells
improves functional, Peripheral electrophysiological and
histological parameters of peripheral nerve regeneration.
Restorative Neurol. and Neurosci. 1993, 5, 353–366. 141.
Eilert, J.B.; Binder, P.; McKinney, P.W.; et al.
Polyglycolic acid synthetic absorbable sutures. Am. J.
Surg. 1971, 121, 561. 142. Jeffre, J. Collagen Degradation.
In Wound Healing: Biochemical and Clinical Aspects; Cohen,
I.K., Diegelmann, R.F., Lindblad, W.J., Eds.; W. B.
Saunders: Philadelphia, 1992; 177–194. 143. Roe, S.C.;
Milthorpe, B.K.; Schindhelm, K. Collagen cross-linking and
resorption: effect of glutaraldehyde concentration.
Artificial Organs 1990, 14, 443–448. 144. Chiu, D.T.W.;
Lovelace, R.E.; Yu, L.I.; Wolff, M.; Stengel, S.;
Middleton, L.; Janecka, I.P.; Krizek, T.J. Comparative
electrophysiologic evaluation of nerve grafts and vein
grafts as nerve conduits. Surg. Forum 1983, 34, 601. 145.
Lundborg, G. A 25-year perspective of peripheral nerve
surgery: evolving neuroscientific concepts and clinical
significance. J. Hand Surg. 2000, 25A, 391–414. 146. Ohara,
K.; Doi, K.; Sakai, K.; Kuwata, N.; Kawai, S. Restoration
of sensibility in the hand after complete brachial plexus
injury. J. Hand Surg. (Am.)May 1996, 21 (3), 381–386. 147.
Bertelli, J.A.; Taleb, M.; Mira, J.C.; dos Santos, A.R.;
Calixto, J.B.; Kassar, L. Selective restoration of
sensation by peripheral nerve grafts directly implanted
into the contralateral C7 dorsal root ganglion: an
experimental study in rat brachial plexus. Neurosurg.
January 1998, 42 (1), 125–129. 148. Jacobs, J.M.; Laing,
J.H.; Harrison, D.H. Regeneration through a long nerve
graft used in the correction of facial palsy. A qualitative
and quantitative study. Brain February 1996, 119 (Pt. 1),
271–279. 149. Kozek, J.; Voska, P.; Tiche´, M. Contemporary
state of surgical treatment of facial nerve paresis.
Preliminary experience with new procedures. Acta Chir.
Plast. 1997, 39 (4), 125–131. 150. Moissonnier, P.;
Reviron, T.; Ye, J.H.; Horvat, J.C. Motoneurons of the
injured spinal cord of the adult dog can grow lengthy axons
into an autologous peripheral nerve graft. A retrograde
axonal tracing study. Spinal Cord 1996, 34 (6), 320–325.
151. Gage, F.H. Method of Grafting Genetically Modified
Cells to Treat Defects, Disease or Damage of the Central
Nervous System. US Patent 5,650,148, July 22, 1997. 152.
Stichel, C.C.; Lips, K.; Wunderlich, G.; Mu¨ller, H.W.
Reconstruction of transected postcommissural fornix in
adult rat by Schwann cell suspension grafts. Exp. Neurol.
July 1996, 140 (1), 21–36. 153. Date, I.; Imaoka, T.;
Miyoshi, Y.; Ono, T.; Asari, S.; Ohmoto, T. Chromaffin cell
survival and host dopaminergic fiber recovery in a patient
with Parkinson’s disease treated by cografts of adrenal
medulla and pretransected peripheral nerve. Case report. J.
Neurosurg. April 1996, 84 (4), 685–689. 154. Wirth, E.D.;
Fessler, R.G.; Reier, P.J.; Thompson, F.J.; Uthman, B.;
Behrman, A.; Remson, E.J.; Anderson, D.K. Neural tissue
transplantation in syringomyelia patients: feasibility and
safety. Amer. Soc. for Neural Transplantation, 5th Annual
Conference, 1998. 155. Romano, V.M.; Blair, S.J.; Wurster,
R.D. Rerouting peripheral nerves for spinal cord lesions.
J. Orthop. Res. 1991, 9, 54–60. 156. Eberhard, D.; Millesi,
H. Split nerve grafting. J. Reconstr. Microsurg. February
1996, 12 (2), 71–76. P
Phosphorylcholine (PC) Technology

28. Ishihara, K.; Nomura, H.; Mihara, T.; Kurita, K.;


Iwasaki, Y.; Nakabayashi, N. Why do phospholipid polymers
reduce protein adsorption? J. Biomed. Mater. Res. 1998, 39,
323–330.

29. Campbell, E.J.; Hall, B.; Parmer, A.R.; Sullivan, A.M.


Improved haemocompatibility of artificial surfaces by
treatment using phosphorylcholine derivatives. Thromb.
Haemost. 1993, 69, 935.

30. Murphy, E.F.; Lu, J.R.; Brewer, J.; Russell, J.C.;


Penfold, J. The reduced adsorption of proteins at the
phosphoryl choline incorporated polymer-water interface.
Langmuir 1999, 15, 1313–1322.

31. Murphy, E.F.; Keddie, J.L.; Lu, J.R.; Brewer, J.;


Russell, J.C. The reduced adsorption of lysozyme at the
phosphorylcholine incorporated polymer/aqueous solution
interface studied by spectroscopic ellipsometry.
Biomaterials 1999, 20 (16), 1501–1511.

32. Clarke, S.; Davies, M.C.; Lewis, A.L.; O’Byrne, V.;


Roberts, C.J.; Tendler, S.J.B.; Williams, P.M. Novel
biomimetic polymers incorporating phosphorylcholine. J.
Pharm. Pharmacol. 2000, 52 (Suppl.), 114.

33. Clarke, S.; Davies, M.C.; Roberts, C.J.; Tendler,


S.J.B.; Williams, P.M.; Lewis, A.L.; O’Byrne, V. Atomic
force microscope and surface plasmon resonance
investigation of polymer blends of
poly([2-(methacryloyloxy)ethyl] phosphorylcholine-co-lauryl
methacrylate) and poly (lauryl methacrylate).
Macromolecules 2001, 34, 4166– 4172.

34. Murphy, E.F.; Lu, J.R.; Lewis, A.L.; Brewer, J.;


Russell, J.C.; Stratford, P.W. Characterisation of protein
adsorption at the phosphorylcholine incorporated
polymer-water interface. Macromolecules 2000, 33 (12),
4545–4554.

35. Faragher, R.G.A.; Bowers, R.W.J.; Denyer, S.P.;


Dropcova, S.; Gard, P.R.; Hall, B.; Hanlon, G.W.; Jones,
S.A.; Lloyd, A.W.; Mikhalovsky, S.V.; Olliff, C.J.; Riding,
M.; Rosen, P.H. In-vitro evaluation of biomimetic
hydrogels: Potential new base materials for intraocular
lenses. J. Pharm. Pharmocol. 1995, 47 (Suppl.), 1068.

36. Lloyd, A.W.; Dropcova, S.; Mikhalovsky, S.V.; Denyer,


S.P.; Gard, P.R.; Hanlon, G.W.; Olliff, C.J.; Faragher,
R.G.A.; Bowers, R.W.J.; Hall, B.; Muir, A.; Jones, S.A.;
Rosen, P.; Riding, M. Biocompatible hydrogels for IOLs.
Vis. Res. 1996, 36, S113.

37. Andrews, C.S.; Lloyd, A.W.; Sharma, D.P.; Hall, B.;


Hanlon, G.W.; Denyer, S.P. In-vitro evaluation of
biomimetic hydrogels. Vis. Res. 1996, 36, S200.

38. Young, G.; Bowers, R.W.J.; Hall, B.; Port, M. Clinical


comparison of omafilcon A with four control materials. CLAO
J. 1997, 23 (4), 249.

39. Bowers, R.W.J.; Port, M.; Young, G. Optician 1995, 210,


23.
Photopolymerization

16. Peppas, N.A.; Merrill, E.W. Polyvinyl-alcohol


hydrogels—Reinforcement of radiation—Crosslinked networks
by crystallization. J. Polym. Sci. Polym. Chem. 1976, 14
(2), 441–457.

17. Elbert, D.L.; Hubbell, J.A. Conjugate addition


reactions combined with free-radical cross-linking for the
design of materials for tissue engineering.
Biomacromolecules 2001, 2 (2), 430–441.

18. Timmer, M.D.; Ambrose, C.G.; Mikos, A.G. In vitro


degradation of polymeric network of poly(propylene
fumarate) and the crosslinking macromer poly(propylene
fumarate)-dicacrylate. Biomaterials 2003, 24 (4), 571–577.

19. Poshusta, A.K.; Anseth, K.S. Photopolymerized


biomaterials for application in the temporomandibular
joint. Cells Tissue Organs 2001, 169, 272–278.

20. Nakayama, Y.; Kim, J.Y.; Nishi, S.; Ueno, H.; Matsuda,
T. Development of high-performance stent: Gelations
photogel-coated stent that permits drug delivery and gene
transfer. J. Biomed. Mater. Res. 2001, 57 (4), 559–566.

21. Halstenberg, S.; Panitch, A.; Rizzi, S.; Hall, H.;


Hubbell, J.A. Biologically engineered
protein-graftpoly(ethylene glycol) hydrogels: A cell
adhesive and plasmin-degradable biosynthetic material for
tissue repair. Biomacromolecules 2002, 3 (4), 710–723.

22. Elisseeff, J.H.; Anseth, K.; Langer, R.; Hrkach, J.S.


Synthesis and characterization of photo-cross-linked
polymers based on poly( L-lactic acid-co-aspartic acid).
Macromolecules 1997, 30 (7), 2182–2184.

23. Li, Q.; Wang, J.; Sun, D.D.N.; Shahani, S.; Sharma, B.;
Elisseeff, J.H.; Leong, K.W. Biodegradable and
photocrosslinkable polyphosphoester hydrogel. Biomaterials
2004. Submitted.

24. Wang, D.A.; Williams, C.G.; Cher, N.; Lee, S.;


Elisseeff, J.H. Bioresponsive phosphoester hydrogels for
bone tissue. Adv. Funct. Mater. 2003. Submitted.

25. Sefton, M.V.; May, M.H.; Lahooti, S.; Babensee, J.E.


Making mcroencapsulation work: Conformal coating,
immobilization gels and in vivo performance. J. Control.
Release 2000, 65 (1–2), 173–186.
26. Lum, L.Y.B.; Cher, N.L.C.; Williams, C.G.; Elisseeff,
J.H. An extracellular matrix extract for tissue-engineered
cartilage. IEEE Eng. Med. Biol. 2003, 22 (5), 71–76.

27. Mann, B.K.; Schmedlen, R.H.; West, J.L. TetheredTGF-b


increases extracellular matrix production of vascular
smooth muscle cells. Biomaterials 2001, 22 (5), 439–444.
28. Burdick, J.A.; Anseth, K.S. Photoencapsulation of
osteoblasts in injectable RGD-modified PEG hydrogels for
bone tissue engineering. Biomaterials 2002, 23 (22),
4315–4323. 29. Luo, N.; Hutchison, J.B.; Anseth, K.S.;
Bowman, C.N. Surface-initiated photopolymerization of
poly(ethylene glycol) methyl ether methacrylate on a
diethyldithiocarbamate-mediated polymer substrate.
Macromolecules 2002, 35 (7), 2487–2493. 30. Guan, J.J.;
Gao, C.Y.; Feng, L.X.; Sheng, J.C. Surface photo-grafting
of polyurethane with 2-hydroxyethyl acrylate for promotion
of human endothelial cell adhesion and growth. J. Biomater.
Sci. Polym. Ed. 2000, 11 (5), 523–536. 31. Cao, Y.L.;
Vacanti, J.P.; Paige, K.T.; Upton, J.; Vacanti, C.A.
Transplantation of chondrocytes utilizing a polymer–cell
construct to produce tissue-engineered cartilage in the
shape of a human ear. Plast. Reconstr. Struct. 1997, 100
(2), 297–302. 32. Liu, V.A.; Bhatia, S.N. Three-dimensional
photopatterning of hydrogels containing living cells.
Biomed. Microdevices 2002, 4 (4), 257–266. 33. Griffith, M.;
Hakim, M.; Shimmura, S.; Watsky, M.A.; Li, F.; Carlsson,
D.; Doillon, C.J.; Nakamura, M.; Suuronen, E.; Shinozaki,
N.; Nakata, K.; Sheardown, H. Artificial human corneas
scaffolds for transplantation and host regeneration. Cornea
2002, 21 (Suppl. 2), S54–S61. 34. Williams, C.G.; Anseth,
K.; Gurkan, I.; Sharma, B.; Parks, B.; Wang, D.A.; Meyers,
J.D.; Wenz, J.; Elisseeff, J.H. Cartilage repair using a
photocurablemesenchymal stem cell hydrogel in large animal
model. ORS’50, The (50th Annual Meeting of the Orthopedic
Research Society, San Francisco, CA, U.S.A., March 7–10,
2004. 35. Elisseeff, J.; Anseth, K.; Sims, D.; McIntosh,
W.; Randolph, M.; Langer, R. Transdermal
photopolymerization for minimally invasive implantation.
Proc. Natl. Acad. Sci. U. S. A. 1999, 96 (6), 3104–3107.
36. Williams, C.G.; Kim, T.K.; Taboas, A.; Malik, A.;
Manson, P.; Elisseeff, J.H. In vitro chondrogenesis of bone
marrow-derived mesenchymal stem cells in a
photopolymerizing hydrogel. Tissue Eng. 2003, 9 (4),
679–688. 37. Kim, T.K.; Sharma, B.; Williams, C.G.;
Ruffner, M.A.; Malik, A.; McFarland, B.G.; Elisseeff, J.H.
Experimental model for cartilage tissue engineering to
regenerate the zonal organization of articular cartilage.
Osteoarthr Cartilage 2003, 11 (9), 653–664.
Piezoelectric Materials

1. Curie, J.; Curie, P. De´veloppement, par pression, de


l’e´lectricite´ polaire dans les cristaux he´mie`dres a`
faces incline´es. C. R. Acad. Sci. 1880, 91, 294–295.

2. Poynting, J.H.; Thomson, J.J. Textbook of Physics,


Electricity and Magnetism; Griffen: London, 1914; 148–163.

3. Voigt, V.W. Lehrbuch der Kristallphysik; B. G. Teubner:


Leipzig-Berlin, 1910.

4. Mason, W.P. Piezoelectricity, its history and


applications. J. Acoust. Soc. Am. 1981, 70, 1561–1566.

5. Busch, G.; Scherrer, P. A new seignettoelectric


substance. Naturwissenschaften 1935, 23, 737.

6. Fukada, E. The piezoelectric effect in fibrous proteins.


Rep. Prog. Polym. Phys. Jpn. 1959, 2, 168–170.

7. Kawai, H. The piezoelectricity of poly(vinylidene


fluoride). Jpn. J. Appl. Phys. 1969, 8, 975.

8. Trainer, M. Kelvin and piezoelectricity. Eur. J. Phys.


2003, 24, 535–542.

9. Cady, W.G. Piezoelectricity; An Introduction to the


Theory and Applications of Electromechanical Phenomena in
Crystals; Dover Publications: New York, 1964.

10. Jaffe, B.; Cook, W.R., Jr.; Jaffe, H. Piezoelectric


Ceramics; Academic Press: London, 1971.

11. Ballato, A. Piezoelectricity: History and New Thrusts,


IEEE Ultrasonics Symposium, 1996; 575–583.

12. Mossi, K.; Selby, G.; Bryant, R. Thin-layer composite


unimorph ferroelectric driver and sensor properties. Mater.
Lett. 1998, 35, 39–49.

13. Furman, E.; Li, G.; Haertling, G. An investigation of


the resonance properties of RAINBOW devices. Ferroelectrics
1994, 160, 357–369.

14. Davis, G.T. Piezoelectric and Pyroelectric Polymers. In


Polymers for Electronic and Photonic Applications; Academic
Press, Inc.: Boston, MA, 1993; 435. 15. Omote, K.;
Ohigashi, H.; Koga, K. Temperature dependence of elastic,
dielectric and piezoelectric properties of ‘single
crystalline’ films of vinylidene fluoride trifluoroethylene
copolymer. J. Appl. Phys. 1997, 81 (6), 2760. 16. Kawai,
H.; Henji, I. Electrostriction and piezoelectricity of
elongated polymer films. Oyo Butsuri 1970, 39, 413. 17.
Newman, B.A.; Chen, P.; Pae, K.D.; Scheinbeim, J.I.
Piezoelectricity in nylon-11. J. Appl. Phys. 1980, 51 (10),
5161–5164. 18. Mei, B.Z.; Schienbeim, J.I.; Newman, B.A.
The ferroelectric behavior of odd-numbered nylons.
Ferroelectrics 1993, 144, 51–60. 19. Newman, B.A.; Kim,
K.G.; Scheinbeim, J.I. The effects of water content on the
piezoelectric properties of nylon11 and nylon-7. J. Mater.
Sci. 1990, 25, 1779–1783. 20. Harrison, J.; Ounaies, Z.
Piezoelectric Polymers. In Encyclopedia of Smart Materials;
John Wiley and Sons: New York, 2002. 21. Hilczer, B.;
Malecki, J. Electrets: Studies in Electrical and Electronic
Engineering; Elsevier: New York, NY, 1986; Vol. 14; 19. 22.
Comstock, R.J.; Stupp, S.I.; Carr, S.H. Thermally
stimulated discharge currents from polyacrylonitrile. J.
Macromol. Sci., Phys. 1977, B13 (1), 101–115. 23. Ueda, H.;
Carr, S.H. Piezoelectricity in polyacrylonitrile. Polym. J.
1984, 16, 661–667. 24. von Berlepsch, H.; Kunstler, W.;
Wedel, A.; Danz, R.; Geib, D. Piezoelectric activity in a
copolymer of acrylonitrile and methylacrylate. IEEE Trans.
Electr. Insul. 1989, 24, 357–362. 25. Miyata, S.;
Yoshikawa, M.; Tasaka, S.; Ko, M. Piezoelectricity revealed
in the copolymer of vinylidenecyanide and vinylacetate.
Polym. J. 1980, 12, 857–860. 26. Furukawa, T.; Date, M.;
Nakajima, K.; Kosaka, T.; Seo, I. Large dielectric
relaxations in an alternate copolymer of vinylidene cyanide
and vinyl acetate. Jpn. J. Appl. Phys. 1986, 25, 1178–1182.
27. Seo, I. Piezoelectricity of vinylidene cyanide
copolymers and their applications. Ferroelectrics 1995,
171, 45–55. 28. Sakurai, M.; Ohta, Y.; Inoue, Y.; Chujo, R.
An important factor generating piezoelectric activity of
vinylidene cyanide copolymers. Polym. Commun. 1991, 32,
397–399. 29. Tasaka, S.; Toyama, T.; Inagaki, N. Ferroand
pyroelectricity in amorphous polyphenylethernitrile. Jpn.
J. Appl. Phys. 1994, 33, 5838–5844. 30. Takahashi, T.;
Kato, H.; Ma, S.P.; Sasaki, T.; Sakurai, K. Morphology of a
wholly aromatic thermoplastic, poly(ether nitrile). Polymer
1995, 36, 3803–3808. 31. Hall, H.K.; Chan, R.J.H.; Oku, J.;
Hughes, O.R.; Scheinbeim, J.; Newman, B. Piezoelectric
activity in films of poly(1-bicyclobutanecarbonitrile).
Polym. Bull. 1987, 17, 135–136. 32. Furukawa, T.
Piezoelectricity in polymers. IEEE Trans. Electr. Insul.
1989, 24, 375–393. 33. Bharti, V.; Nath, R. Quantitative
analysis of piezoelectricity in simultaneously stretched
and corona poled polyvinyl chloride films. J. Appl. Phys.
1997, 82, 3488–3492. 34. Ounaies, Z.; Young, J.A.;
Harrison, J.S. An Overview of the Piezoelectric Phenomenon
in Amorphous Polymers; Amer. Chem. Society: Washington, DC,
1999. 35. Martin, A.J.P. Tribo-electricity in wool and
hair. Proc. Phys. Soc. 1941, 53, 183. 36. Fukada, E.
Piezoelectricity and Pyroelectricity of Biopolymers. In
Ferroelectric Polymers, Chemistry, Physics and Application;
Marcel Dekker, Inc.: New York, NY, 1995; 393. 37. Maeda,
H.; Fukada, E. Effect of water on piezoelectric dielectric
and elastic properties of bone. Biopolymers 1982, 21, 2055.
38. Date, M.; Takashita, S.; Fukada, E. Temperature
variation of piezoelectric moduli in oriented
poly(g-methylL-glutamate). J. Polym. Sci. 1970, A-2 (8),
61. 39. Furukawa, T.; Fukada, E. Piezoelectric relaxation
in poly(g-methyl-L-glutamate). J. Polym. Sci., Polym. Phys.
1976, 14, 1979. 40. Ando, Y.; Fukada, E. Piezoelectric
properties of oriented deoxyribonucleate films. J. Polym.
Sci., Polym. Phys. 1976, 14, 63. 41. Fukada, E.
Piezoelectric properties of poly-L-lactic acid. Rep. Prog.
Polym. Phys. Jpn. 1991, 34, 269. 42. Ando, Y.; Fukada, E.;
Glimcher, M.J. Piezoelectricity of chitin in lobster shell
and apodeme. Biorheology 1977, 14, 175. 43. Fukada, E.;
Yasuda, L. On the piezoelectric effect of bone. J. Phys.
Soc. Jpn. 1957, 12, 1158. 44. Bassett, C.A.L.; Becker, R.O.
Generation of electric potentials by bone in response to
mechanical stress. Science 1962, 137, 1063. 45. Shamos,
M.H.; Lavine, L.S.; Shamos, M.I. Piezoelectric effect in
bone. Nature 1963, 1978–1981. 46. Fukada, E.
Piezoelectricity of Bone and Osteogenesis of Piezoelectric
Films. In Mechanisms of Growth Control; Charles C. Thomas
ed; Springfield, IL, 1981; 192. 47. Bar-Cohen, Y.
Electroactive Polymer (EAP) Actuators as Artificial Muscles:
Reality, Potential and Challenges; SPIE Press: Bellingham,
WA, 2001; 615. 48. DeRossi, D.; Dario, P. Medical
Applications of Piezoelectric Polymers; Gordon and Breach
Science Publishers, 1988; 83. 49. Garner, G.M. The
Applications of Ferroelectric Polymers; Blackie: London,
UK, 1988; 190. 50. Meeker, T.R. The Applications of
Ferroelectric Polymers; Blackie: London, UK, 1988; 305. 51.
Yamaka, E. The Applications of Ferroelectric Polymers;
Blackie: London, UK, 1988; 329. P
Plasma Modification of Materials

1. Grill, A. Cold Plasma in Materials Fabrication: From


Fundamentals to Applications; IEEE Press: New York, 1994.

2. Lieberman, M.A.; Lichtenberg, A.J. Principles of Plasma


Discharges and Materials Processing; John Wiley & Sons: New
York, 1994.

3. Ohring, M. The Materials Science of Thin Films; Academic


Press: Boston, 1992. 4. Klee, D.; Hocker, H. Polymers for
biomedical applications: improvement of the interface
compatibility. Adv. Polym. Sci. 2000, 149, 1–57. 5.
Flemming, R.G.; Murphy, C.J.; Abrams, G.A.; Goodman, S.L.;
Nealey, P.F. Effects of synthetic microand nano-structured
surfaces on cell behavior. Biomaterials 1999, 20 (6),
573–588. 6. Chen, C.S.; Mrksich, M.; Huang, S.; Whitesides,
G.M.; Ingber, D.E. Micropatterned surfaces for control of
cell shape, position, and function. Biotechnol. Prog. 1998,
14 (3), 356–363. 7. Schroder, K.; Meyer-Plath, A.; Keller,
D.; Ohl, A. On the applicability of plasma assisted
chemical micropatterning to different polymeric
biomaterials. Plasmas Polym. 2002, 7 (2), 103–125. 8. Den
Braber, E.T.; Jansen, H.V.; De Boer, M.J.; Croes, H.J.E.;
Elwenspoek, M.; Ginsel, L.A.; Jansen, J.A. Scanning
electron microscopic, transmission electron microscopic,
and confocal laser scanning microscopic observation of
fibroblasts cultured on microgrooved surfaces of bulk
titanium substrate. J. Biomed. Mater. Res. 1998, 40 (3),
425–433. 9. Ha, S.W.; Hauert, R.; Ernst, K.H.;
Wintermantel, E. Surface analysis of chemically-etched and
plasmatreated polyetheretherketone (PEEK) for biomedical
applications. Surf. Coat. Technol. 1997, 96 (2–3), 293–299.
10. Damdorenea, J.D.; Conde, A.; Palacio, C.; Rodriguez, R.
Modification of corrosion properties of titanium by
N-implantation. Surf. Coat. Technol. 1997, 91 (1–2), 1–6.
11. Ueda, M.; Silva, M.M.; Otani, C.; Reuther, H.;
Yatsuzuka, M.; Lepienski, C.M.; Berni, L.A. Improvement of
tribological properties of Ti6Al4V by nitrogen plasma
immersion ion implantation. Surf. Coat. Technol. 2003, 169,
408–410. 12. Poon, R.W.Y.; Ho, J.P.Y.; Liu, X.Y.; Chung,
C.Y.; Chu, P.K.; Yeung, K.W.K.; Lu, W.W.; Cheung, K.M.C.
Anti-corrosion performance of oxidized and oxygen
plasma-implanted NiTi alloys. Mater. Sci. Eng. A 2005, 390
(1–2), 444–451. 13. Tan, L.; Dodd, R.A.; Crone, W.C.
Corrosion and wearcorrosion behavior of NiTi modified by
plasma source ion implantation. Biomaterials 2003, 24 (22),
3931–3939. 14. Hanawa, T. In vivo metallic biomaterials and
surface modification. Mater. Sci. Eng. A. 1999, 267 (2),
260– 266. 15. Liu, X.; Poon, R.W.Y.; Kwok, S.C.H.; Chu,
P.K.; Ding, C. Structure and properties of
Ca-plasma-implanted titanium. Surf. Coat. Technol. 2005,
191 (1), 43–48. 16. Yoshinari, M.; Oda, Y.; Ueki, H.;
Yokose, S. Immobilization of bisphosphonates on surface
modified titanium. Biomaterials 2001, 22 (7), 709–715. 17.
Krupa, D.; Baszkiewicz, J.; Kozubowski, J.A.; Barcz, A.;
Sobczak, J.W.; Bilinski, A.; LewandowskaSzumiel, M.;
Rajchel, B. Effect of phosphorus-ion implantation on the
corrosion resistance and biocompatibility of titanium.
Biomaterials 2002, 23 (16), 3329–3340. 18. Pham, M.T.;
Maitz, M.F.; Matz, W.; Reuther, H.; Richter, E.; Steiner,
G. Promoted hydroxyapatite nucleation on titanium
ion-implanted with sodium. Thin Solid Films 2000, 379
(1–2), 50–56. 19. Li, D.J.; Niu, L.F. Cell attachment of
polypropylene surface-modified by COOH + ion implantation.
Nucl. Instrum. Methods B. 2002, 192 (4), 393–401. 20.
Nakao, A.; Kaibara, M.; Iwaki, M.; Suzuki, Y.; Suzuki, Y.;
Kusakabe, M. XPS and SERS studies of cell
adhesion-controlled polymer modified by ion implantation.
Surf. Interface Anal. 1996, 24, 252–256. 21. Bacakova, L.;
Mares, V.; Lisa, V.; Svorcik, V. Molecular mechanisms of
improved adhesion and growth of an endothelial cell line
cultured on polystyrene implanted with fluorine ions.
Biomaterials 2000, 21, 1173–1179. 22. Li, D.J.; Cui, F.Z.;
Gu, H.Q. F + ion implantation induced cell attachment on
intraocular lens. Biomaterials 1999, 20, 1889–1896. 23.
Colwell, J.M.; Wentrup-Byrne, E.; Bell, J.M.; Wielunski,
L.S. A study of the chemical and physical effects of ion
implantation of micro-porous and nonporous PTFE. Surf.
Coat. Technol. 2003, 168 (2–3), 216–222. 24. Suzuki, Y.;
Iwaki, M.; Tani, S.; Oohashi, G.; Kamio, M. Ion
implantation into ePTFE for application of a dural
substitute. Nucl. Instrum. Methods B 2003, 206, 538–542.
25. Yang, P.; Kwok, S.C.H.; Fu, R.K.Y.; Leng, Y.X.; Wang,
J.; Wan, G.J.; Huang, N.; Leng, Y.; Chu, P.K. Structure and
properties of annealed amorphous hydrogenated carbon
(a-C:H) films for biomedical applications. Surf. Coat.
Technol. 2004, 177–178, 747–751. 26. Wang, J.; Huang, N.;
Pan, C.J.; Kwok, S.C.H.; Yang, P.; Leng, Y.X.; Chen, J.Y.;
Sun, H.; Wan, G.J.; Liu, Z.Y.; Chu, P.K. Bacterial
repellence from polyethylene terephthalate surface modified
by acetylene plasma immersion ion implantation–deposition.
Surf. Coat. Technol. 2004, 186 (1–2), 299–304. 27. Huang,
N.; Yang, P.; Leng, Y.X.; Chen, J.Y.; Sun, H.; Wang, J.;
Wang, G.J.; Ding, P.D.; Xi, T.F.; Leng, Y.
Hemocompatibility of titanium oxide films. Biomaterials
2003, 24 (13), 2177–2187. 28. Chen, J.Y.; Wan, G.J.; Leng,
Y.X.; Yang, P.; Sun, H.; Wang, J.; Huang, N. Behavior of
cultured human umbilical vein endothelial cells on titanium
oxide films fabricated by plasma immersion ion implantation
and deposition. Surf. Coat. Technol. 2004, 186 (1–2),
270–276. 29. Chen, J.Y.; Leng, Y.X.; Tian, X.B.; Wang,
L.P.; Huang, N.; Yang, P.; Chu, P.K. Antithrombogenic
investigation of surface energy and optical bandgap and
hemocompatibility mechanism of Ti(Ta +5 )O 2 thin films.
Biomaterials 2002, 23 (12), 2545–2552. 30. Tsyganov, I.;
Maitz, M.F.; Wieser, E. Blood compatibility of
titanium-based coatings prepared by metal plasma immersion
ion implantation and deposition. Appl. Surf. Sci. 2004, 235
(1–2), 156–163. 31. Klee, D.; Ademovic, Z.; Bosserhoff, A.;
Hoecker, H.; Maziolis, G.; Erli, H.J. Surface modification
of poly(vinylidenefluoride) to improve the osteoblast
adhesion. Biomaterials 2003, 24 (21), 3663–3670. 32. Kang,
I.K.; Seo, E.J.; Huh, M.W.; Kim, K.H. Interaction of blood
components with heparin-immobilized polyurethanes prepared
by plasma glow discharge. J. Biomater. Sci. Polym. Ed.
2001, 12 (10), 1091–1108. 33. Dong, Z.L.; Khor, K.A.; Quek,
C.H.; White, T.J.; Cheang, P. TEM and STEM analysis on
heat-treated and in vitro plasma-sprayed
hydroxyapatite/Ti-6Al-4V composite coatings. Biomaterials
2003, 24 (1), 97–105. 34. Liu, X.Y.; Zhao, X.B.; Fu,
R.K.Y.; Ho, J.P.Y.; Ding, C.X.; Chu, P.K. Plasma-treated
nanostructued TiO 2 surface supporting biomimetic growth of
apatite. Biomaterials 2005, 26 (31), 6143–6150. 35. Liu,
X.; Tao, S.; Ding, C. Bioactivity of plasma sprayed
dicalcium silicate coatings. Biomaterials 2002, 23 (3),
963–968. 36. Sodhi, R.N.S. Application of surface
analytical and modification techniques to biomaterials
research. J. Electron. Spectrosc. Phenom. 1996, 81 (3),
269–284. 37. Liu, X.; Chu, P.K.; Ding, C. Surface
modification of titanium, titanium alloys, and related
materials for biomedical application. Mater. Sci. Eng. R.
2004, 47, 49–121. 38. Chu, P.K.; Chen, J.Y.; Wang, L.P.;
Huang, N. Plasmasurface modification of biomaterials. Mater.
Sci. Eng. R. 2002, 36, 143–206. 39. Ratner, B.D.; Hoffman,
A.S.; Schoen, F.J.; Lemons, J.E. Biomaterials Science: An
Introduction to Materials in Medicine; Academic Press: New
York, 1996. 40. Park, J.B.; Bronzino, J.D. Biomaterials:
Principles and Applications; CRC Press: New York, 2003. P
Polyamides (Synthetic and Natural)

42. http://www.ewg.org/reports/skindeep2/report.php?type=
INGREDIENT&id=2065 (accessed February 2007).

43. Maarek, J.M.; Holschneider, D.P.; Harimoto, J.


Fluorescence of indocyanine green in blood: intensity
dependence on concentration and stabilization with sodium
polyaspartate. J. Photochem. Photobiol. B 2001, 65 (2–3),
157–164.

44. Whilton, N.T.; Vickers, P.J.; Mann, S. Bioinorganic


clays: Synthesis and characterization of aminoand polyamino
acid intercalated layered double hydroxides. J. Mater.
Chem. 1997, 7 (8), 1623–1629.

45. Trauger, J.W.; Baird, E.E.; Dervan, P.B. Recognition of


DNA by designed ligands at subnanomolar concentrations.
Nature 1996, 382, 559–561.

46. White, S.; Szewczyk, J.W.; Turner, J.M.; Baird, E.E.;


Dervan, P.B. Recognition of the four Watson-Crick base
pairs in the DNA minor groove by synthetic ligands. Nature
1998, 391, 468–471.

47. Murty, M.S.R.C.; Sugiyama, H. Biology of


N-methylpyrrole-N-methylimidazole hairpin polyamide. Biol.
Pharm. Bull. 2004, 27 (4), 468–474.

48. Wang, Y.-D.; Dziegielewski, J.; Wurtz, N.R.;


Dziegielewska, B.; Dervan, P.B.; Beerman, T.A. DNA
crosslinking and biological activity of a hairpin
polyamide–chlorambucil conjugate. Nucleic Acids Res. 2003,
31 (4), 1208–1215. 49. Wurtz, N.R.; Dervan, P.B. Sequence
specific alkylation of DNA by hairpin pyrrole–imidazole
polyamide conjugates. Chem. Biol. 2000, 7 (3), 153–161. 50.
Zheng, Z.; Feldman, D. Synthetic fibre-reinforced concrete.
Prog. Polym. Sci. 1995, 20, 185–210. 51.
http://www.technica.net/NT/NT3/composites.htm (accessed
February 2007). 52.
http://www.horizonpress.com/gateway/pna.html (accessed
February 2007). 53. Ray, A.; Norden, B. Peptide nucleic
acid (PNA): Its medical and biotechnical applications and
promise for the future. FASEB J. 2000, 14, 1041–1060. 54.
Pooga, M.; Land, T.; Bartfai, T.; Langel, U. PNA oligomers
as tools for specific modulation of gene expression. Biomol.
Eng. 2001, 17, 183–192. 55. Paulasova, P.; Pellestor, F.
The peptide nucleic acids (PNAs): A new generation of
probes for genetic and cytogenetic analyses. Ann. Genet.
2004, 47 (4), 349–358. 56.
http://daecr1.harvard.edu/pdf/smnr_2000-2001_Peterson_
Gretchen.pdf (accessed March 2007). 57.
http://www.medfarm.uu.se/forskning/program.php?ve
tenskapsid=2&forskomr=43&id=216&lang=en (accessed March
2007). 58. Walensky, L.D.; Kung, A.L.; Escher, I.; Malia,
T.J.; Barbuto, S.; Wright, R.D.; Wagner, G.; Verdine, G.L.;
Korsmeyer, S.J. Activation of apoptosis in vivo by a
hydrocarbon-stapled BH3 helix. Science. 2004 , 305 (5689),
1466–1470.
Polycaprolactone

47. Lemarchand, C.; Couvreur, P.; Besnard, M.; Costantini,


D.; Gref, R. Novel polyester-polysaccharide nanoparticles.
Pharm. Res. 2003, 20 (8), 1284–1292.

48. Sinha, V.R.; Bansal, K.; Kaushik, R.; Kumria, R.;


Trehan, A. Poly-e-caprolactone microspheres and
nanospheres: an overview. Int. J. Pharm. 2004, 278 (1),
1–23.

49. Attivi, D.; Wehrle, P.; Ubrich, N.; Damge, C.; Hoffman,
M.; Maincent, P. Formulation of insulin-loaded polymeric
nanoparticles using response surface methodology. Drug Dev.
Ind. Pharm. 2005, 31 (2), 179–189.

50. Shenoy, D.B.; D’Souza, R.J.; Tiwari, S.B.; Udupa, N.


Potential applications of polymeric microsphere suspension
as subcutaneous depot for insulin. Drug Dev. Ind. Pharm.
2003, 29 (5), 555–563.

51. Park, E.K.; Lee, S.B.; Lee, Y.M. Preparation and


characterization of methoxy poly(ethylene glycol)/poly
(e-caprolactone) amphiphilic block copolymeric nanospheres
for tumor-specific folate-mediated targeting of anticancer
drugs. Biomaterials 2005, 26 (9), 1053–1061.

52. Shuai, X.T.; Ai, H.; Nasongkla, N.; Kim, S.; Gao, J.M.
Micellar carriers based on block copolymers of poly
(e-caprolactone) and poly(ethylene glycol) for doxorubicin
delivery. J. Control. Release 2004, 98 (3), 415–426.

53. Chawla, J.S.; Amiji, M.M. Cellular uptake and


concentrations of tamoxifen upon administration in poly
(e-caprolactone) nanoparticles. AAPS. Pharmsci. 2003, 5
(1), 1–7.

54. Ameller, T.; Marsaud, W.; Legrand, P.; Gref, R.;


Renoir, J.M. Pure antiestrogen RU 58668-loaded nanospheres:
morphology, cell activity and toxicity studies. Eur. J.
Pharm. Sci. 2004, 21 (2–3), 361–370.

55. Sastre, R.L.; Blanco, M.D.; Teijon, C.; Olmo, R.;


Teijon, J.M. Preparation and characterization of
5-fluorouracilloaded poly(e-caprolactone) microspheres for
drug administration. Drug Dev. Res. 2004, 63 (2), 41–53.

56. Zhang, X.Z.; Lewis, P.J.; Chu, C.C. Fabrication and


characterization of a smart drug delivery system:
microsphere in hydrogel. Biomaterials 2005, 26 (16),
3299–3309.

57. Dhanaraju, M.D.; Jayakumar, R.; Vamsadhara, C. Influence


of manufacturing parameters on development of contraceptive
steroid loaded injectable microspheres. Chem. Pharm. Bull.
2004, 52 (8), 976–979.

58. Williamson, M.R.; Chang, H.I.; Coombes, A.G.A. Gravity


spun polycaprolactone fibres: controlling release of a
hydrophilic macromolecule (ovalbumin) and a lipophilic drug
(progesterone). Biomaterials 2004, 25 (20), 5053–5060.

59. Barbault-Foucher, S.; Gref, R.; Russo, P.; Guechot, J.;


Bochot, A. Design of poly-e caprolactone nanospheres coated
with bioadhesive hyaluronic acid for ocular delivery. J.
Control. Release 2002, 83 (3), 365–375.

60. Lboutounne, H.; Chaulet, J.F.; Ploton, C.; Falson, F.;


Pirot, F. Sustained ex vivo skin antiseptic activity of
chlorhexidine in poly(e-caprolactone) nanocapsule
encapsulated form and as a digluconate. J. Control. Release
2002, 82 (2–3), 319–334.

61. Dai, N.T.; Williamson, M.R.; Khammo, N.; Adams, E.F.;


Coombes, A.G.A. Composite cell support membranes based on
collagen and polycaprolactone for tissue engineering of
skin. Biomaterials 2004, 25 (18), 4263–4271. 62. Ng, K.W.;
Khor, H.L.; Hutmacher, D.W. In vitro characterization of
natural and synthetic dermal matrices cultured with human
dermal fibroblasts. Biomaterials 2004, 25 (14), 2807–2818.
63. Khor, H.L.; Ng, K.W.; Schantz, J.T.; Phan, T.T.; Lim,
T.C.; Teoh, S.H.; Hutmacher, D.W. Poly (e-caprolactone)
films as a potential substrate for tissue engineering an
epidermal equivalent. Mater. Sci. Eng. C-Biomimetic
Supramol. Syst. 2002, 20 (1–2), 71–75. 64. Woei, K.;
Hutmacher, D.W.; Schantz, J.T.; Seng, C.; Too, H.P.; Chye,
T.; Phan, T.T.; Teoh, S.H. Evaluation of ultra-thin
poly(e-caprolactone) films for tissueengineered skin. Tissue
Eng. 2001, 7 (4), 441–455. 65. Meek, M.F.; Jansen, K.;
Steendam, R.; van Oeveren; van Wachem, P.B.; van Luyn,
M.J.A. In vitro degradation and biocompatibility of poly (
DL-lactide-e-caprolactone) nerve guides. J. Biomed. Mater.
Res. A 2004, 68A (1), 43–51. 66. Galla, T.J.; Vedecnik,
S.V.; Halbgewachs, J.; Steinmann, S.; Friedrich, C.; Stark,
G.B. Fibrin/Schwann cell matrix in poly-e-caprolactone
conduits enhances guided nerve regeneration. Int. J. Artif.
Organs 2004, 27 (2), 127–136. 67. Heijkants, R.G.J.C.; van
Calck, R.V.; De Groot, J.H.; Pennings, A.J.; Schouten,
A.J.; van Tienen, T.G.; Ramrattan, N.; Buma, P.; Veth,
R.P.H. Design, synthesis and properties of a degradable
polyurethane scaffold for meniscus regeneration. J. Mater.
Sci. Mater. Med 2004, 15 (4), 423–427. 68. Li, W.J.;
Danielson, K.G.; Alexander, P.G.; Tuan, R.S. Biological
response of chondrocytes cultured in threedimensional
nanofibrous poly(e-caprolactone) scaffolds. J. Biomed.
Mater. Res. A 2003, 67A (4), 1105–1114. 69. Zhao, J.; Yuan,
X.Y.; Cui, Y.L.; Ge, Q.B.; Yao, K.D. Preparation and
characterization of poly ( L-lactide)/ poly(e-caprolactone)
fibrous scaffolds for cartilage tissue engineering. J. Appl.
Polym. Sci. 2004, 91 (3), 1676–1684. 70. Huang, Q.;
Hutmacher, D.W.; Lee, E.H. In vivo mesenchymal cell
recruitment by a scaffold loaded with transforming growth
factor b1 and the potential for in situ chondrogenesis.
Tissue Eng. 2002, 8 (3), 469–482. 71. Aho, A.J.; Tirri, T.;
Kukkonen, J.; Strandberg, N.; Rich, J.; Seppala, J.;
Yli-Urpo, A. Injectable bioactive glass/biodegradable
polymer composite for bone and cartilage reconstruction:
concept and experimental outcome with thermoplastic
composites of poly (e-caprolactone-coD,L-lactide) and
bioactive glass S53P4. J. Mater. Sci.-Mater. Med. 2004, 15
(10), 1165–1173. 72. Cao, T.; Ho, K.H.; Teoh, S.H. Scaffold
design and in vitro study of osteochondral coculture in a
threedimensional porous polycaprolactone scaffold
fabricated by fused deposition modelling. Tissue Eng. 2003,
9 (Suppl. 1), S103–S112. 73. Aydin, H.M.; Piskin, E.;
Calimli, A. Microporous scaffolds from
poly(lactide-co-e-caprolactone) composites with
hydroxyapatite and tricalcium phosphates using
supercritical CO 2 for bone tissue engineering. J. Bioact.
Compat. Polym. 2004, 19 (5), 383–394. 74. Endres, M.;
Hutmacher, D.W.; Salgado, A.J.; Kaps, C.; Ringe, J.; Reis,
R.L.; Sittinger, M.; Brandwood, A.; Schantz, J.T.
Osteogenic induction of human bone marrow-derived
mesenchymal progenitor cells in novel synthetic
polymer-hydrogel matrices. Tissue Eng. 2003, 9 (4),
689–702. 75. Looss, P.; Le Ray, A.M.; Grimandi, G.;
Daculsi, G.; Merle, C. A new injectable bone substitute
combining poly(e-caprolactone) microparticles with biphasic
calcium phosphate granules. Biomaterials 2001, 22 (20),
2785–2794. 76. Kim, H.W.; Lee, E.J.; Kim, H.E.; Salih, V.;
Knowles, J.C. Effect of fluoridation of hydroxyapatite in
hydroxyapatite-polycaprolactone composites on osteoblast
activity. Biomaterials 2005, 26 (21), 4395–4404. 77. Rai,
B.; Teoh, S.H.; Ho, K.H.; Hutmacher, D.W.; Cao, T.; Chen,
F.; Yacob, K. The effect of rhBMP-2 on canine osteoblasts
seeded onto 3D bioactive polycaprolactone scaffolds.
Biomaterials 2004, 25 (24), 5499–5506. 78. Rhee, S.H.; Lee,
Y.K.; Lim, B.S. Evaluation of a novel
poly(epsilon-caprolactone)-organosiloxane hybrid material
for the potential application as a bioactive and degradable
bone substitute. Biomacromolecules 2004, 5 (4), 1575–1579.
79. Rhee, S.H. Bone-like apatite-forming ability and
mechanical properties of poly(e-aprolactone)/silica hybrid
as a function of poly(e-caprolactone) content. Biomaterials
2004, 25 (7–8), 1167–1175. 80. Im, S.Y.; Cho, S.H.; Hwang,
J.H.; Lee, S.J. Growth factor releasing porous
poly(e-caprolactone)-chitosan matrices for enhanced bone
regenerative therapy. Arch. Pharm. Res. 2003, 26 (1),
76–82. 81. Christian, P.; Jones, I.A. Polymerisation and
stabilisation of polycaprolactone using a borontrifluoride–
glycerol catalyst system. Polymer 2001, 42 (9), 3989–3994.
82. Carlisle, E.S.; Mariappan, M.R.; Nelson, K.D.; Thomes,
B.E.; Timmons, R.B.; Constantinescu, A.; Eberhart, R.C.;
Bankey, P.E. Enhancing hepatocyte adhesion by pulsed plasma
deposition and polyethylene glycol coupling. Tissue Eng.
2000, 6 (1), 45–52. 83. Chua, K.N.; Lim, W.S.; Zhang, P.C.;
Lu, H.F.; Wen, J.; Ramakrishna, S.; Leong, K.W.; Mao, H.Q.
Stable immobilization of rat hepatocyte spheroids on
galactosylated nanofiber scaffold. Biomaterials 2005, 26
(15), 2537–2547. 84. Barralet, J.E.; Wallace, L.L.; Strain,
A.J. Tissue engineering of human biliary epithelial cells
on polyglycolic acid/polycaprolactone scaffolds maintains
long-term phenotypic stability. Tissue Eng. 2003, 9 (5),
1037–1045. 85. Shin, M.; Ishii, O.; Sueda, T.; Vacanti,
J.P. Contractile cardiac grafts using a novel nanofibrous
mesh. Biomaterials 2004, 25 (17), 3717–3723. 86. Huard, J.;
Fu, F.H. Eds. Gene Therapy and Tissue Engineering in
Orthopedic and Sports Medicine; Birkhauser publishers:
Boston, 2000. 87. Shuai, X.T.; Merdan, T.; Unger, F.;
Wittmar, M.; Kissel, T. Novel biodegradable ternary
copolymers hy-PEI-g-PCL-b-PEG: synthesis, characterization,
and potential as efficient nonviral gene delivery vectors.
Macromolecules 2003, 36 (15), 5751–5759. 88. Chen, L.G.;
Wang, Z.R.; Wan, C.M.; Chao, L.; Chao, J.; Xing, H.Y.
Encapsulated transgene cells attenuate hypertension,
cardiac hypertrophy and enhance renal function in Goldblatt
hypertensive rats. J. Gene. Med. 2004, 6 (7), 786–797. 89.
Ciapetti, G.; Ambrosio, L.; Savarino, L.; Granchi, D.;
Cenni, E.; Guizzardi, N.S.; Causa, F.; Giunti, A.
Osteoblast growth and function in porous poly
e-caprolactone matrices for bone repair: a preliminary
study. Biomaterials 2003, 24, 3815–3824. 90. Kwon, I.K.;
Kidoaki, S.; Matsuda, T. Electrospun nanoto microfiber
fabrics made of biodegradable copolyesters: structural
characteristics, mechanical properties and cell adhesion
potential. Biomaterials 2005, 26 (18), 3929–3939. 91. Mo,
X.M.; Xu, C.Y.; Kotaki, M.; Ramakrishna, S. Electrospun
P(LLA-CL) nanofiber: a biomimetic extracellular matrix for
smooth muscle cell and endothelial cell proliferation.
Biomaterials 2004, 25 (10), 1883–1890. 92. Hsu, C.M.;
Shivkumar, S. Nano-sized beads and porous fiber constructs
of poly(e-caprolactone) produced by electrospinning. J.
Mater. Sci. 2004, 39 (9), 3003–3013. 93. Yoshimoto, H.;
Shin, Y.M.; Terai, H.; Vacanti, J.P. A biodegradable
nanofiber scaffold by electrospinning and its potential for
bone tissue engineering. Biomaterials 2003, 24 (12),
2077–2082. 94. Busby, A.J.; Zhang, J.X.; Roberts, C.J.;
Lester, E.; Howdle, S.M. Novel nanostructured polymeric
composites of polycaprolactone and
ultra-high-molecularweight polyethylene via a
supercritical-fluid route. Adv. Mater. 2005, 17 (3), 364.
95. Stassin, F.; Jerome, R. Contribution of supercritical
CO 2 to the preparation of aliphatic polyesters and
materials thereof. Macromol Symposia 2004, 217, 135–146.
96. Wang, F.; Shor, L.; Darling, A.; Khalil, S.; Sun, W.;
Guceri, S.; Lau, A. Precision extruding deposition and
characterization of cellular poly-e-caprolactone tissue
scaffolds. Rapid Prototyping J. 2004, 10 (1), 42–49. P
Poly(carbonate)urethanes

Table 4 ChronoFlex C Biodurable poly(carbonate)urethane


elastomers

CardioTech International, Inc. Technical data sheet

Properties ASTM procedure Typical values

Hardness (Shore durometer) ASTM D-2240 80A 55D 75D

Appearance Visual Slightly cloudy Slightly cloudy Slightly


cloudy

Ultimate tensile strength (psi) ASTM D-638 a W5000 W6000


W7000

Ultimate tensile strain (%) ASTM D-638 a 400–490 365–440


255–320

100% Secant modulus (psi) ASTM D-638 a 770–1250 1850–2200


5300–5700

300% Secant modulus (psi) ASTM D-638 a 700–1400 1700–2000


2700–3200

Flexural strength (psi) ASTM D-790 350 550 10,000

Flexural modulus (psi) ASTM D-790 5500 9300 300,000

Melt index (g/10 min) 2101C;

2.17kg ASTM D-1238 8 5 3

Vicat softening point (1F/1C) ASTM D-1525 160/70 180/80 –

Water absorption ASTM D-5170 1.2 1.0 0.8

Dielectric strength (volts/mil) ASTM D-149 360 520 420

Specific gravity ASTM D-792 1.2 1.2 1.2

Coefficient of friction (kinetic) ASTM D-1894 1.5 0.8 0.64

Abrasion resistance (% loss at

1000 cycles) ASTM D-1044 0.008 0.035 0.053

Melt-process. temp. (1F/1C) 375–430/190–220


Recommended sterilization

method Gamma; E-beam; ethylene oxide

Class VI biocompatibility test U.S.P. XXII Pass Pass Pass

a Specimens conditioned for 7 days at room temperature and


50% relative humidity. Metal Catalyzed Oxidation in
Implanted Polymers. In Polyurethanes in Biomedical
Engineering II; Planck, H., et al., Eds.; Elsevier:
Amsterdam, The Netherlands, 1987; 109–127. 5. Szycher, M.
Biostability of polyurethanes: A critical review. J.
Biomater. Appl. 1988, 3 (2), 297–402. 6. Szycher, M.
Biostable Polyurethane Products. US Patent 5,254,662.
FURTHER READING Bernacca, G.M.; O’Connor, B.; Williams,
D.F.; Wheatley, D.J. Hydrodynamic function of polyurethane
prosthetic heart valves: Influences of Young’s modulus and
leaflet thickness. Biomaterials Jan 2002, 23 (1), 45–50.
Chen, C.T.; et al. Synthesis, characterization, and
permeation properties of polyether-based polyurethanes. J.
Appl. Polym. Sci. 1972, 16, 2105–2114. Farrar, D.J. The
thoratec ventricular assist device: A paracorporeal pump
for treating acute and chronic heart failure. Semin.
Thorac. Cardiovasc. Surg. Jul 2000, 12 (3), 243–250.
Farrar, D.J. Development of a prosthetic coronary artery
bypass graft. Heart Surg. Forum 2000, 3 (1), 36–40. Farrar,
D.J.; et al. In vivo evaluations of a new thromboresistant
polyurethane for artificial heart blood pumps. J. Thorac.
Cardiovasc. Surg. Feb 1988, 95 (2), 191–200. Fields, C.;
Cassano, A.; Allen, C.; Meyer, A.; Pawlowski, K.J.; Bowlin,
G.L.; Rittgers, S.E.; Szycher, M. Endothelial cell seeding
of a 4 mm ID polyurethane vascular graft. J. Biomater.
Appl. Jul 2002, 17 (1), 45–70. Grasel, T.G.; et al. Surface
properties and blood compatibility of polyurethaneureas.
Biomaterials Sep 1986, 7, 315–328. Hergenrother, R.W.; et
al. Blood-contacting properties of
4-4u-polydimethylsiloxane polyurea-urethanes. Biomaterials
1994, 15 (8), 635–640. Hutchinson, D.T.; Savory, K.M.;
Bachus, K.N. Crack-growth properties of various elastomers
with potential application in small joint prosthesis. J.
Biomed. Mater. Res. Oct 1997, 37 (1), 94–99. Jahangir,
A.R.; McClung, W.G.; Cornelius, R.M.; McCloskey, C.B.;
Brash, J.L.; Santerre, J.P. Fluorinated surface-modifying
macromolecules: Modulating adhesive protein and platelet
interactions on a polyetherurethane. J. Biomed. Mater. Res.
Apr 2002, 60 (1), 135–147. Lim, F.; et al. Synthesis,
characterization and ex vivo evaluation of
polydimethylsiloxane polyurea-urethanes. Biomaterials 1994,
15 (6), 408–416. Nyilas, E.; et al. Development of
Blood-Compatible Elastomers. V. Surface Structure and Blood
Compatibility of Avcothane Elastomers. In Biological
Interactions at Polymer Surfaces, J. Biomed. Mater. Res.,
Biomed. Mater. Symp., John Wiley & Sons, Inc.: New York,
1977; Vol. 8, 69–84. Okkema, A.Z.; et al. Bulk, surface,
and blood-contacting properties of polyetherurethanes
modified with polyethylene oxide. J. Biomater. Sci., Polym.
Ed. 1989, 1 (1), 43–62. Park, J.H.; Lee, K.B.; Kwon, I.C.;
Bae, Y.H. PDMS-based polyurethanes with MPEG grafts:
Mechanical properties, bacterial repellency, and release
behavior of rifampicin. J. Biomater. Sci. Polym. Ed. 2001 ,
12 (6), 629–645. Park, J.C.; Song, M.J.; Hwang, Y.S.; Suh
Yonsei, H. Calcification comparison of polymers for vascular
graft. Med. J. Jun 2001, 42 (3), 304–310. Paynter, R.W.; et
al. The hydrolytic stability of mitrathane (a polyurethane
urea)—An X-ray photoelectron spectroscopy study. J. Biomed.
Mater. Res. Jul 1988, 22 (7), 687–698. Sakurai, S.; et al.
Changes in structure and properties due to mechanical
fatigue for polyurethanes containing
poly(dimethylsiloxane). Polymer 1994, 35 (3), 532–539.
Speckhard, T.A.; et al. Ultimate tensile properties of
segmented polyurethane elastomers: Factors leading to
reduced properties for polyurethanes based on nonpolar soft
segments. Rubber Chem. Technol. 1986, 59, 405–429. Perena,
M.F.; Castillo, J.; Medrano, J.; De Gregorio, M.A.; Loras,
E.; Cristobal, J.A. Nasolacrimal polyurethane stent
placement: Preliminary results. Eur. J. Ophthalmol. Jan–Mar
2001, 11 (1), 25–30. Pulat, M.; Akdogan, A.; Ozkan, S.
Microbiological interaction and diffusion properties of
hydrophilic and hydrophobic PU membranes. J. Biomater.
Appl. Apr 2002, 16 (4), 293–303. Salacinski, H.J.; Tai,
N.R.; Punshon, G.; Giudiceandrea, A.; Hamilton, G.;
Seifalian, A.M. Optimal endothelialisation of a new
compliant poly(carbonate-urea)urethane vascular graft with
effect of physiological shear stress. Eur. J. Vasc.
Endovasc. Surg. Oct 2000, 20 (4), 342–352. Salacinski,
H.J.; Punshon, G.; Krijgsman, B.; Hamilton, G.; Seifalian,
A.M. A hybrid compliant vascular graft seeded with
microvascular endothelial cells extracted from human
omentum. Artif. Organs Dec 2001, 25 (12), 974–982.
Salacinski, H.J.; Tai, N.R.; Carson, R.J.; Edwards, A.;
Hamilton, G.; Seifalian, A.M. In vitro stability of a novel
compliant poly(carbonate-urea)urethane to oxidative and
hydrolytic stress. J. Biomed. Mater. Res. Feb 2002, 59 (2),
207–218. Seifalian, A.M.; Salacinski, H.J.; Punshon, G.;
Krijgsman, B.; Hamilton, G. A new technique for measuring
the cell growth and metabolism of endothelial cells seeded
on vascular prostheses. J. Biomed. Mater. Res. Jun 15,
2001, 55 (4), 637–644. Soldani, G.; Varelli, G.; Minnocci,
A.; Dario, P. Manufacturing and microscopical
characterization of polyurethane nerve guidance channel
featuring a highly smooth internal surface. J. Biomater.
Nov 1998, 19 (21), 1919–1924. Sugimoto, K.; Hirota, S.;
Imanaka, K.; Ohnishi, H.; Tomioka, H.; Tada, K.; Aoki, M.;
Sugimura, K. Covered Gianturco stent for tracheal
stricture: Application of polychlorovinylidene and
polyurethane as covering materials. Eur. Radiol. 2001, 11
(10), 1933–1938. Szycher, M.; Reed, A.M. In vivo testing of
a biostable polyurethane. J. Biomater. Appl. Oct 1991 , 6
(2), 110–130. P

Szycher, M.; et al. Blood compatible polyurethane


elastomers. J. Biomater. Appl. Oct 1987, 2, 290–313.

Tai, N.R.; Salacinski, H.J.; Edwards, A.; Hamilton, G.;


Seifalian, A.M. Compliance properties of conduits used in
vascular reconstruction. Br. J. Surg. Nov 2000, 87 (11),
1516–1524.

Takahara, A.; et al. Surface molecular mobility and


platelet reactivity of segmented poly(etherurethaneuraneas)
with hydrophilic and hydrophobic soft segment components.
J. Biomater. Sci., Polym. Ed. 1989, 1 (1), 17–29.

Takahara, A.; et al. Effect of soft segment chemistry on


the biostability of segmented polyurethanes. II. In vitro
hydrolytic degradation and lipid sorption. J. Biomed.
Mater. Res. 1992, 26, 801–818.

Tanzi, M.C.; Fare, S.; Petrini, P. In vitro stability of


polyether and polycarbonate urethanes. J. Biomater. Appl.
Apr 2000, 14 (4), 325–348.

Thomas, V.; Jayabalan, M.; Sandhya, S. Studies on


polyurethane potting compound based on isocyanurate of
aliphatic diisocyanate for fabrication of a haemodialyser.
J. Biomater. Appl. Oct 2000, 15 (2), 86–105.

Tiwari, A.; Salacinski, H.; Seifalian, A.M.; Hamilton, G.


New prostheses for use in bypass grafts with special
emphasis on polyurethanes. Cardiovasc. Surg. Jun 2002, 10
(3), 191–197.

Van Den Hazel, S.J.; Mulder, C.J.; Den Hartog, G.; Thies,
J.E.; Westhof, W. A randomized trial of polyurethane and
silicone percutaneous endoscopic gastrostomy catheters.
Aliment. Pharmacol. Ther. Oct 2000, 14 (10), 1273–1277.

Visai, L.; Rindi, S.; Speziale, P.; Petrini, P.; Fare, S.;
Tanzi, M.C. In vitro interactions of biomedical
polyurethanes with macrophages and bacterial cells. J.
Biomater. Appl. Jan 2002, 16 (3), 191–214.

Ward, R.S. Surface modification prior to surface formation:


Control of polymer surface properties via bulk composition,
medical plastics and biomaterials. Mater. Forum 1995,
34–41. Ward, R.S.; et al. Production of Biomedical I.
Polymers, Silicone/Urethane Synergy in Avcothane
Elastomers. In Organometallic Polymers; Carraher, E.C.,
Jr., Ed.; Academic Press: New York, 1978; 219–229. Ward,
R.S.; et al. BPS-215M: A Polyurethaneurea for Biomedical
Devices: Development and in vivo Testing in the
Pierce–Donachy VAD, The 13th Annual Meeting of the Society
for Biomaterials, New York, NY, USA, Jun. 2–6, 1987; 259.
Ward, R.S.; et al. Development of a hybrid artificial
pancreas with a dense polyurethane membrane. ASAIO J. 1993,
39 (3), M261–M267. Ward, R.S.; et al. Improved Polymer
Biostability via Oligomeric End Groups Incorporated During
Synthesis. In Polymeric Materials Science and Engineering,
Fall Meeting, Boston, MA, Aug. 23–27, 1998; 526–527
Wheatley, D.J.; Raco, L.; Bernacca, G.M.; Sim, I.; Belcher,
P.R.; Boyd, J.S. Polyurethane: Material for the next
generation of heart valve prostheses? Eur. J. Cardiothorac.
Surg. Apr 2000, 17 (4), 440–448. White, K.A.; et al.
Surface Modification of Segmented Polyurethaneureas via
Oligomeric End Groups Incorporated During Synthesis. In
Surface Modification of Polymeric Biomaterials; Rainer,
B.D., et al., Eds.; Plenum Press: New York, NY, 1996;
27–33. Wiggins, M.J.; Wilkoff, B.; Anderson, J.M.; Hiltner,
A. Biodegradation of polyether polyurethane inner
insulation in bipolar pacemaker leads. J. Biomed. Mater.
Res. May 1, 2001, 58 (3), 302–307. Yang, M.; Zhang, Z.;
Hahn, C.; King, M.W.; Guidoin, R. Assessing the resistance
to calcification of polyurethane membranes used in the
manufacture of ventricles for a totally implantable
artificial heart. J. Biomed. Mater. Res. 1999, 48 (5),
648–659.
Polyelectrolyte Multilayers

37. Muller, M.; Rieser, T.; Lunkwitz, K.; Meier-Haack, J.


Polyelectrolyte complex layers: a promising concept for
antifouling coatings verified by in-situ ATR-FTIR
spectroscopy. Macromol. Rapid Commun. 1999, 20, 607–611.

38. Watanabe, S.; Regen, S.L. Dendrimers as buildingblocks


for multilayer construction. J. Am. Chem. Soc. 1994, 116,
8855–8856.

39. Keller, S.W.; Kim, H.-N.; Mallouk, T.E. Layer-bylayer


assembly of intercalation compounds and heterostructures on
surfaces—toward molecular beaker epitaxy. J. Am. Chem. Soc.
1994, 116, 8817–8818.

40. Hakkarainen, S; Gilbert, S.L.; Kontturi, A.K.;


Kontturi, K. Amperometric method for determining the degree
of complexation of polyelectrolytes with cationic
surfactants. J. Colloid Interface Sci. 2004, 272, 404–410.

41. Panambur, G.; Zhang, Y.B.; Yesayan, A.; Galstian, T.;


Bazuin, C.G.; Ritcey, A.M. Preparation and characterization
of polyion-complexed LangmuirBlodgett films containing an
NLO chromophore. Langmuir 2004, 20, 3606–3615.

42. Huang, H.X.; Qian, D.J.; Nakamura, N.; Nakamura, C.;


Wakayama, T.; Miyake, J. Quartz crystal microbalance and
electrochemical studies on the electrode modified by
layer-by-layer multilayers of viologen polyelectrolytes.
Electrochim. Acta 2004, 49, 1491– 1498.

43. Hoffmannova, H.; Fermin, D.; Krtil, P. Growth and


electrochemical activity of the
polyL-lysine9poly-L-glutamic acid thin layer films: an EQCM
and electrochemical study. J. Electroanal. Chem. 2004, 562,
261– 265.

44. Serpe, M.J.; Lyon, L.A. Optical and acoustic studies of


pH-dependent swelling in microgel thin films. Chem. Mater.
2004, 16, 4373–4380.

45. Zhu, H.G.; Ji, J.; Shen, J.C. Biomacromolecules


electrostatic self-assembly on 3-dimensional tissue
engineering scaffold. Biomacromolecules 2004, 5, 1933–1939.

46. Haynie, D.T.; Balkundi, S.; Palath, N.; Chakravarthula,


K.; Dave, K. Polypeptide multilayer films: role of molecular
structure and charge. Langmuir 2004, 20, 4540–4547.
47. Welsh, E.R.; Schauer, C.L.; Santos, J.P.; Price, R.R.
In situ cross-linking of alternating polyelectrolyte
multilayer films. Langmuir 2004, 20, 1807–1811.

48. Caruso, F.; Niikura, K.; Furlong, D.N.; Okahata, Y.


Ultrathin multilayer polyelectrolyte films on gold:
construction and thickness determination. 1. Langmuir 1997,
13, 3422–3426.

49. Muller, M.; Meier-Haack, J.; Schwarz, S.; Buchhammer,


H.M.; Eichhorn, E.J.; Janke, A.; Kessler, B.; Nagel, J.;
Oelmann, M.; Reihs, T.; Lunkwitz, K. Polyelectrolyte
multilayers and their interactions. J. Adhesion 2004, 80,
521–547.

50. Schoeler, B.; Poptoschev, E.; Caruso, F. Growth of


multilayer films of fixed and variable charge density
polyelectrolytes: effect of mutual charge and secondary
interactions. Macromolecules 2003, 36, 5258–5264.

51. Schwarz, S.; Nagel, J.; Jaeger, W. Comparison of


polyelectrolyte multilayers built up with
polydiallyldimethylammonium chloride and
poly(ethyleneimine) from salt-free solutions by in-situ
surface plasmon resonance measurements. Macromol. Symp.
2004, 211, 201–216. 52. Dante, S.; Advincula, R.; Frank,
C.W.; Stroeve, P. Photoisomerization of polyionic
layer-by-layer films containing azobenzene. Langmuir 1999,
15, 193–201. 53. Keller, S.W.; Johnson, S.A.; Brigham,
E.S.; Yonemoto, E.H.; Mallouk, T.E. Photoinduced charge
separation in multilayer thin films grown by sequential
adsorption of polyelectrolytes. J. Am. Chem. Soc. 1995,
117, 12879–12880. 54. Cheng, L.; Niu, L.; Gong, J.; Dong,
S.J. Electrochemical growth and characterization of
polyoxometalate-containing monolayers and multilayers on
alkanethiol monolayers self-assembled on gold electrodes.
Chem. Mater. 1999, 11, 1465–1475. 55. Dermody, D.L.; Peez,
R.F.; Bergbreiter, D.E.; Crooks, R.M. Chemically grafted
polymeric filters for chemical sensors: hyperbranched
poly(acrylic acid) films incorporating beta-cyclodextrin
receptors and aminefunctionalized filter layers. Langmuir
1999, 15 , 885–890. 56. Delcorte, A.; Bertrand, P.;
Wischerhoff, E.; Laschewsky, A. Adsorption of
polyelectrolyte multilayers on polymer surfaces. Langmuir
1997, 13, 5125–5136. 57. Laschewsky, A.; Wischerhoff, E.;
Bertrand, P.; Delcorte, A. Polyelectrolyte multilayers
containing photoreactive groups. Macromol. Chem. Phys.
1997, 198, 3239–3253. 58. Buscher, K.; Graf, K.; Ahrens,
H.; Helm, C.A. Influence of adsorption conditions on the
structure of polyelectrolyte multilayers. Langmuir 2002,
18, 3585– 3591. 59. Serpe, M.J.; Lyon, L.A. Optical and
acoustic studies of pH-dependent swelling in microgel thin
films. Chem. Mater. 2004, 16, 4373–4380. 60. Zhu, H.G.; Ji,
J.; Shen, J.C. Biomacromolecules electrostatic
self-assembly on 3-dimensional tissue engineering scaffold.
Biomacromolecules 2004, 5, 1933– 1939. 61. Haynie, D.T.;
Balkundi, S.; Palath, N.; Chakravarthula, K.; Dave, K.
Polypeptide multilayer films: role of molecular structure
and charge. Langmuir 2004, 20, 4540–4547. 62. Welsh, E.R.;
Schauer, C.L.; Santos, J.P.; Price, R.R. In situ
cross-linking of alternating polyelectrolyte multilayer
films. Langmuir 2004, 20, 1807–1811. 63. Caruso, F.;
Niikura, K.; Furlong, D.N.; Okahata, Y. Ultrathin
multilayer polyelectrolyte films on gold: construction and
thickness determination. 1. Langmuir 1997, 13, 3422–3426.
64. Schoeler, B.; Poptoschev, E.; Caruso, F. Growth of
multilayer films of fixed and variable charge density
polyelectrolytes: effect of mutual charge and secondary
interactions. Macromolecules 2003, 36, 5258– 5264. 65.
Schwarz, S.; Nagel, J.; Jaeger, W. Comparison of
polyelectrolyte multilayers built up with
polydiallyldimethylammonium chloride and poly
(ethyleneimine) from salt-free solutions by in-situ surface
plasmon resonance measurements. Macromol. Symp. 2004, 211,
201–216. 66. Picart, C.; Ladam, G.; Senger, B.; Voegel,
J.C.; Schaaf, P.; Cuisinier, F.J.G.; Gergely, C.
Determination of structural parameters characterizing thin
films by optical methods: a comparison between scanning
angle reflectometry and optical waveguide lightmode
spectroscopy. J. Chem. Phys. 2001, 115, 1086–1094. 67.
McAloney, R.A.; Sinyor, M.; Dudnik, V.; Goh, M.C. Atomic
force microscopy studies of salt effects on polyelectrolyte
multilayer film morphology. Langmuir 2001, 17, 6655–6663.
68. McAloney, R.A.; Dudnik, V.; Goh, M.C. Kinetics of
salts-induced annealing of a polyelectrolyte multilayer film
morphology. Langmuir 2003, 19, 3947–3952. 69. Jaber, J.A.;
Schlenoff, J.B. Polyelectrolyte multilayers with reversible
thermal responsivity. Macromolecules 2005, 38, 1300–1306.
70. Sullivan, D.M.; Bruening, M.L. Ultrathin, crosslinked
polyimide pervaporation membranes prepared from
polyelectrolyte multilayers. J. Membr. Sci. 2005, 248,
161–170. 71. Sukhishvili, S.A.; Granick, S. Layered,
erasable polymer multilayers formed by hydrogen-bonded
sequential self-assembly. Macromolecules 2002, 35, 301–310.
72. Schwinte, P.; Voegel, J.C.; Picart, C.; Haikel, Y.;
Schaaf, P.; Szalontai, B. Stabilizing effects of various
polyelectrolyte multilayer films on the structure of
adsorbed/embedded fibrinogen molecules: an ATRFTIR study. J.
Phys. Chem. B. 2001, 105, 11906– 11916. 73. Muller, M.;
Rieser, T.; Lunkwitz, K.; Berwald, S.; Meier-Haack, J.;
Jehnichen, D. An in-situ ATR-FTIR study on polyelectrolyte
multilayer assemblies on solid surfaces and their
susceptibility to fouling. Macromol. Rapid Commun. 1998,
19, 333–336. 74. Heuberger, R.; Sukhorukov, G.; Voros, J.;
Textor, M.; Mohwald, H. Biofunctional polyelectrolyte
multilayers and microcapsules: control of non-specific and
bio-specific protein adsorption. Adv. Funct. Mater. 2005,
15, 357–366. 75. Picart, C.; Gergely, C.; Arntz, Y.;
Voegel, J.C.; Schaaf, P.; Cuisinier, F.J.G.; Senger, B.
Measurement of film thickness up to several hundreds of
nanometers using optical waveguide lightmode spectroscopy.
Biosensors Bioelectron. 2004, 20, 553–561. 76. Vodouhe, C.;
Schmittbuhl, M.; Boulmedais, F.; Bagnard, D.; Vautier, D.;
Schaaf, P.; Egles, C.; Voegel, J.C.; Ogier, J. Effect of
functionalization of multilayered polyelectrolyte films on
motoneuron growth. Biomaterials 2005, 26, 545–554. 77.
Green, R.J.; Frazier, R.A.; Shakesheff, K.M.; Davies, M.C.;
Roberts, C.J.; Tendler, S.J.B. Surface plasmon resonance
analysis of dynamic biological interactions with
biomaterials. Biomaterials 2000, 21, 1823–1835. 78. Nelson,
B.P.; Frutos, A.G.; Brockman, J.M.; Corn, R.M.
Near-infrared surface plasmon resonance measurements of
ultrathin films. 1. Angle shift and SPR imaging experiments.
Anal. Chem. 1999, 71, 3928–3934. 79. Furst, E.M.; Pagac,
E.S.; Tilton, R.D. Coadsorption of polylysine and the
cationic surfactant cetyltrimethylammonium bromide on
silica. Ind. Eng. Chem. Res. 1996, 35, 1566–1574. 80.
Koper, G.J.M. Optical properties of colloidal films.
Colloids Surf. A Physicochem. Eng. Asp. 2000, 165, 39–57.
81. Voros, J.; Ramsden, J.J.; Csucs, G.; Szendro, I.; De
Paula, S.M.; Textora, M.; Spencer, N.D. Optical grating
coupler biosensors. Biomaterials 2002, 23, 3699–3710. 82.
Katz, H.E.; Lovinger, A.J.; Johnson, J.; Kloc, C.;
Siegrist, T.; Li, W.; Lin, Y.-Y.; Dodabalapur, A. A soluble
and air-stable organic semiconductor with high electron
mobility. Nature 2000, 404, 478–480. 83. Burroughes, J.H.;
Bradley, D.D.C.; Brown, A.R.; Marks, R.N.; Mackay, K.;
Friend, R.H.; Burns, P.L.; Homes, A.B. Light-emitting
diodes based on conjugated polymers. Nature 1990, 347,
539–541. 84. Sirringhaus, H.; Kawase, T.; Friend, R.H.;
Shimoda, T.; Inbasekaran, M.; Wu, W.; Woo, E.P.
Highresolution inkjet printing of all-polymer transistor
circuits. Science 2000, 290, 2123–2126. 85. Bharathan, J.;
Yang, Y. Polymer electroluminescent devices processed by
inkjet printing: I. Polymer lightemitting logo. Appl. Phys.
Lett. 1998, 72, 2660–2662. 86. Fou, A.C.; Onitsuka, O.;
Ferreira, M.; Rubner, M.F. Fabrication and properties of
light-emitting diodes based on self-assembled multilayers
of poly(phenylene vinylene). J. Appl. Phys. 1996, 79,
7501–7509. 87. Jiang, X.; Hammond, P.T. Selective
deposition in layer-by-layer assembly: functional graft
copolymers as molecular templates. Langmuir 2000, 16, 8501–
8509. 88. Stroeve, P.; Vasquez, V.; Coelho, M.A.N.; Rabolt,
J.F. Gas transfer in supported films made by molecular
self-assembly of ionic polymers. Thin Solid Films 1996,
284–285, 708–712. 89. Levasalmi, J.-M.; McCarthy, T.J.
Poly(4-methylLpentene)-supported polyelectrolyte multilayer
films: preparation and gas permeability. Macromolecules
1997, 30, 1752–1757. 90. Dai, J.; Jensen, A.W.; Mohanty,
D.K.; Erndt, J.; Bruening, M.L. Controlling the
permeability of multilayered polyelectrolyte films through
derivatization, cross-linking, and hydrolysis. Langmuir
2001, 17, 931–937. 91. Katayama, H.; Ishihama, Y.; Asakawa,
N. Stable capillary coating with successive multiple ionic
polymer layers. Anal. Chem. 1998, 70, 2254–2260. 92.
Katayama, H.; Ishihama, Y.; Asakawa, N. Stable cationic
capillary coating with successive multiple ionic polymer
layers for capillary electrophoresis. Anal. Chem. 1998, 70,
5272–5277. 93. Graul, T.W.; Schlenoff, J.B. Capillaries
modified by polyelectrolyte multilayers for electrophoretic
separations. Anal. Chem. 1999, 71, 4007–4013. 94. Rmaile,
H.H.; Schlenoff, J.B. Optically active polyelectrolyte
multilayers as membranes for chiral separations. J. Am.
Chem. Soc. 2003, 125, 6602–6603. 95. Alberts, B.; Johnson,
A.; Lewis, J.; Raff, M.; Roberts, K.; Walters, P. Cell
junctions, cell adhesion, and the extracellular matrix. In
Molecular Biology of the Cell; 4th. Alberts, B., Johnson,
A., Lewis, J., Raff, M., Roberts, K., Walters, P., Eds.;
Garland Science: New York, 2002; 1065–1125. P

96. Yavin, E.; Yavin, Z.J. Attachment and culture of


dissociated cells from rat embryo cerebral hemispheres on
polylysine-coated surface. J. Cell Biol. 1974, 62, 540–546.

97. Letourneau, P.C. Possible roles for cell-to-substratum


adhesion in neuronal morphogenesis. Dev. Biol. 1975, 44,
77–91.

98. McKeehan, W.L.; Ham, R.G. Stimulation of clonal growth


of normal fibroblasts with substrata coated with basic
polymers. J. Cell Biol. 1976, 71, 727–734.

99. Freshney, R.I. Culture of Animal Cells: A Manual of


Basic Techniques, 4th Ed.; Wiley-Liss: New York, 2000.

100. Brandley, B.K.; Schnaar, R.L. Covalent attachment of


an Arg-Gly-Asp sequence peptide to derivatizable
polyacrylamide surfaces—support of fibroblast adhesion and
long-term growth. Anal. Biochem. 1988, 172, 270–278.
101. Tryoen-Toth, P.; Vautier, D.; Haikel, Y.; Voegel,
J.C.; Schaaf, P.; Chluba, J.; Ogier, J. Viability,
adhesion, and bone phenotype of osteoblast-like cells on
polyelectrolyte multilayer films. J. Biomed. Mater. Res.
2002, 60, 657–667.

102. Vautier, D.; Hemmerle, J.; Vodouhe, C.; Koenig, G.;


Richert, L.; Picart, C.; Voegel, J.C.; Debry, C.; Chluba,
J.; Ogier, J. 3-D surface charges modulate protrusive and
contractile contacts of chondrosarcoma cells. Cell Motil.
Cytoskeleton 2003, 56, 147–158.

103. Barker, S.L.R.; Ross, D.; Tarlov, M.J.; Gaitan, M.;


Locascio, L.E. Control of flow direction in microfluidic
devices with polyelectrolyte multilayers. Anal. Chem. 2000,
72, 5925–5929.

104. Cui, L.; Tang, J.; Li, W.J.; Wang, Z.C.; Sun, C.Q.;
Zhao, M.Y. Preparation of catalytically active enzyme thin
film by alternate deposition of horseradish peroxidase and
bipolar quaternary ammonium on solid surface. Mater. Chem.
Phys. 2001, 71, 23–27. 105. Wang, M.; Wang, L.; Yuan, H.;
Ji, X.; Sun, C.; Ma, L.; Bai, Y.; Li, T.; Li, J.
Immunosensors based on layer-by-layer self-assembled Au
colloidal electrode for the electrochemical detection of
antigen. Electroanalysis 2004, 16, 757–764. 106. Donath,
E.; Sukhorukov, G.B.; Caruso, F.; Davis, S.A.; Mohwald, H.
Novel hollow polymer shells by colloid-templated assembly
of polyelectrolytes. Angew. Chem. 1998, 37, 2201–2205. 107.
Tiourina, O.P.; Antipov, A.A.; Sukorukov, G.B.; Larionova,
N.I.; Lvov, Y.; Mohwald, H. Entrapment of -chymotrypsin
into hollow polyelectrolyte microcapsules. Macromol.
Biosci. 2001, 1, 209–214. 108. Balabushevitch, N.G.;
Sukhorukov, G.B.; Moroz, N.A.; Volodkin, D.V.; Larionova,
N.I.; Donath, E.; Mohwald, H. Encapsulation of proteins by
layer-bylayer adsorption of polyelectrolytes onto protein
aggregates: factors regulating the protein release.
Biotechnol. Bioeng. 2001, 76, 207–213. 109. Barker, S.L.R.;
Tarlov, M.J.; Canavan, H.; Hickman, J.J.; Locascio, L.E.
Plastic microfluidic devices modified with polyelectrolyte
multilayers. Anal. Chem. 2000, 72, 4899–4903. 110.
Bertrand, P.; Jonas, A.; Laschewsky, A.; Legras, R.
Ultrathin polymer coatings by complexation of
polyelectrolytes at interfaces: suitable materials,
structure and properties. Macromol. Rapid Commun. 2000, 21,
319–348.
Polyepoxy Compound Fixation

45. Wang, E.Y.; Giclas, P.C.; Tu, R.H.; Hata, C.; Quijano,
R.C. A comparative study of complement activation by
Denaflex, Bioflow, and BioPolyMeric vascular grafts. ASAIO J.
1993, 39, 691–694.

46. Tu, R.; Wang, E.; Hata, C.; Shen, S.H.; Lin, D.;
Quijano, R.C. A compliant biological vascular prosthesis.
Int. J. Artif. Organs 1993, 16, 141–145.

47. Nojiri, C.; Noishiki, Y.; Koyanagi, H. Aorta-coronary


bypass grafting with heparinized vascular grafts in dogs. A
preliminary study. J. Thorac. Cardiovasc. Surg. 1987, 93,
867–877.

48. Noishiki, Y.; Yamane, Y.; Miyata, T. Long-term


evaluation of a growable graft. ASAIO Trans. 1989, 35,
267–270.

49. Noishiki, Y.; Yamane, Y.; Furuse, M.; Miyata, T.


Development of a growable vascular graft. ASAIO Trans.
1988, 34, 308–313. 50. Stenzel, K.H.; Miyata, T.; Rubin,
A.L. Collagen as a biomaterial. Annu. Rev. Biophys. Bioeng.
1974, 3, 231–253. 51. Kuru, S. Collagen Fiber Hemostatic
Material and Method of Producing the Same. European Patent,
0,463,887 B1, July 1, 1991. 52. Tomizawa, Y.; Noishiki, Y.;
Endo, M.; Hashimoto, A.; Koyanagi, H. Evaluation of blood
absorption, hemostatic ability and purity of a polyepoxy
compound crosslinked cotton type collagen hemostat. Kyobu
Geka Feb. 1996, 49, 126–129. 53. Miyata, T.; Fususe, M.;
Yamane, Y.; Noishiki, Y. A biodegradable antiadhesion
collagen membrane with slow release heparin. ASAIO Trans.
1988, 34, 687–691. 54. Noishiki, Y.; Miyata, T.
Antiadhesive collagen membrane with heparin slow release.
J. Bioact. Compat. Polym. 1987, 2, 325–333.
Poly(Glycolic Acid)

4. Belenkaya, B.; Sakharova, V.; Sinevich, E.; Belousov,


S.; Kuptsov, A. Synthesis and biodegradation of modified
polyglycolide. Polymer Prepr. 1999, 39 (2), 164.

5. Chatani, Y.; Suehiro, K.; Okita, Y.; Tadokoro, H.;


Chujo, K. Structural studies of polyesters I. Crystal
structure of polyglycolide. Makromol. Chem. 1968, 113,
215–229.

6. Hurrell, S.; Cameron, R. Polyglycolide: Degradation and


drug release. Part I: Changes in morphology during
degradation. J. Mater. Sci. Mater. Med. 2001, 12, 811–816.

7. Chu, C. The in vitro degradation of poly(glycolic acid)


sutures—Effect of pH. J. Biomed. Mater. Res. 1981, 15 (6),
795–804.

8. Chu, C.; Campbell, N. Scanning electron microscopic


study of the hydrolytic degradation of poly(glycolic acid)
suture. J. Biomed. Mater. Res. 1982, 16 (4), 417–430.

9. Tomihata, K.; Suzuki, M.; Ikada, Y. The pH dependence of


monofilament sutures on hydrolytic degradation. J. Biomed.
Mater. Res. 2001, 58 (5), 511–518.

10. Williams, D.; Mort, E. Enzyme-accelerated hydrolysis of


poly(glycolic acid). J. Bioeng. 1977, 1 (3), 231–238.

11. Barrows, T. Synthetic Bioabsorbable Polymers. In High


Performance Biomaterials: A Comprehensive Guide to Medical
and Pharmaceutical Applications; Szycher, M., Ed.;
Technomic: Lancaster, PA, 1991; 243–257.

12. Hurrell, S.; Cameron, R. Polyglycolide: Degradation and


drug release. Part II: Drug release. J. Mater. Sci. Mater.
Med. 2001, 12, 817–820.

13. Eilbert, J.; McKinney, P.; Conn, J.; Binder, P.; Beal,
J. Poly(glycolic acid) synthetic absorbable sutures. Am. J.
Surg. 1971, 121 (5), 561–565.

14. Ibnabddjalil, M.; Loh, M.; Chu, C.; Blumenthal, N.


Effect of surface plasma treatment on the chemical,
physical, morphological, and mechanical properties of
totally absorbable bone internal fixation devices. J.
Biomed. Mater. Res. 1994, 28 (3), 289–301.

15. Vasenius, J.; Vainionpa¨a¨, S.; Vihtonen, K.; Ma¨kela¨,


A.; Rokkanen, P. Comparison of in vitro hydrolysis,
subcutaneous and intramedullary implantation to evaluate
the strength retention of absorbable osteosynthesis
implants. Biomaterials 1990, 11 (7), 501–504. 16. Browning,
A.; Chu, C.C. The effects of annealing treatments on the
tensile properties and hydrolytic degradative properties of
polyglycolic acid sutures. J. Biomed. Mater. Res. 1986, 20
(5), 613–632. 17. Deitzel, J.; Kleinmeyer, M.; Harris, J.;
Beck, D.; Tan, N. The effect of processing variables on the
morphology of electrospun nanofibers and textiles. Polymer
2001, 42, 261–272. 18. Boland, E.D.; Wnek, G.E.; Simpson,
D.G.; Pawlowski, K.J.; Bowlin, G.L. Tailoring tissue
engineering scaffolds using electrostatic processing
techniques: A study of poly(glycolic acid) electrospinning.
J. Macromol. Sci. 2001, 38 (12), 1231–1243. 19. Kitchens,
W.H. Legal Issues Involved in Tissue Engineering: FDA
Regulation of Tissue Engineering. In Synthetic
Biodegradable Polymer Scaffolds; Atala, A., Mooney, D.,
Vacanti, J., Langer, R., Eds.; Birhauser: Boston, 1997;
33–49. 20. http://wtec.org/loyola/te/final/te_final.pdf.
(accessed Oct. 2002). 21. Babanalbandi, D.; Hill, D.;
O’Donnell, J.; Pomery, P. An electron spin resonance
analysis on g-irradiated poly (glycolic acid) and its
copolymers with lactic acid. Polym. Degrad. Stab. 1996, 52,
59–66. 22. Santavirta, S.; Konttinen, Y.; Saito, T.;
Gro¨nblad, M.; Partio, E.; Kemppinen, P.; Rokkanen, P.
Immune response to poly(glycolic acid) implants. J. Bone
Joint. Surg. Br. 1990, 72 (4), 597–600. 23. Holder, W.;
Gruber, H.; Moore, A.; Culberson, C.; Anderson, W.; Burg,
K.; Mooney, D. Cellular ingrowth and thickness changes in
polyL-lactide and polyglycolide matrices implanted
subcutaneously in the rat. J. Biomed. Mater. Res. 1998, 41,
412–421. 24. Goa, J.; Niklason, L.; Langer, R. Surface
hydrolysis of poly(glycolic acid) meshes increases the
seeding density of vascular smooth muscle cells. J. Biomed.
Mater. Res. 1998, 42 (3), 417–424. 25. Mooney, D.J.;
Langer, R.S. Engineering Biomaterials for Tissue
Engineering: The 10–100 Micron Size Scale. In The
Biomedical Engineering Handbook; Bronzino, J.D., Ed.; CRC
Press: Boca Raton, 1995; 1609–1618.
Poly(lactic acid)s

11. Vert, M.; Chabot, F.; Leray, J.; Chrsitel, P.


Bioresorbable polyesters for bone surgery. Makromol. Chem.,
Suppl. 1981, 5, 30–41.

12. Li, S.M.; Vert, M. Biodegradation of Aliphatic


Polyesters. In Degradable Polymers. Principles and
Applications; Scott, G., Ed.; Kluwer Academic Publ.:
Dordrecht, The Netherlands, 2002; 71–132.

13. Lillie, E.; Schulz, R.C. 1 H and 13 C-{1H}-NMR spectra


of stereocopolymers of lactide. Makromol. Chem. 1975, 176,
1901–1906.

14. Chabot, F.; Vert, M.; Chapelle, S.; Granger, P.


Configurational structures of lactic acid stereocopolymers
as determined by 13 C1 H-NMR. Polymer 1993, 24, 53–59.

15. Ajioka, M.; Enamoto, E.; Suzuki, K.; Yamaguchi, A.


Basic properties of polylactic acid produced by the direct
condensation polymerization of lactic acid. Bull. Chem.
Soc. Jpn. 1995, 68, 2125–2131.

16. Moon, S.I.; Lee, C.W.; Miyamoto, M.; Kimura, Y. Melt


polycondensation of L-lactic acid with Sn(II) catalysts
activated by various proton acids: A direct manufacturing
route to high molacular weight poly ( L-lactic acid). J.
Polym. Sci., A, Polym. Chem. 2000, 38, 1673–1679.

17. Ikada, Y.; Jamshidi, K.; Tsuji, H.; Hyon, S.H.


Stereocomplex formation between enantiomeric
poly(lactides). Macromolecules 1987, 20, 904–906.

18. Kricheldorf, H.R.; Kreiser, I. Polylactones. 13.


Transesterification of poly( L-lactide with poly(glycolide),
poly(b-propiolactone) and poly(e-caprolactone). J.
Macromol. Sci., Chem. 1987, A24, 1345–1358.

19. Garlotta, D. A literature review of poly(lactic acid).


J. Polym. Environ. 2001, 9, 63–84.

20. Albertsson, A.-C.; Varma, I.K. Aliphatic Polyesters. In


Biopolymers: Biology, Chemistry, Biotechnology,
Applications, Polyesters III; Doi, Y., Steinbu¨chel, A.,
Eds.; Wiley-VCH, Verlag GmbH: Weinhiem, Germany, 2002;
25–52.

21. Tsuji, H. Polylactides. In Biopolymers: Biology,


Chemistry, Biotechnology, Applications, Polyesters III;
Doi, Y., Steinbu¨chel, A., Eds.; Wiley-VCH, Verlag GmbH:
Weinheim, Germany, 2002; 129–178.

22. Vert, M. Polyglycolide and Copolyesters with Lactides.


In Biopolymers: Biology, Chemistry, Biotechnology,
Applications, Polyesters III; Doi, Y., Steinbu¨chel, A.,
Eds.; Wiley-VCH, Verlag GmbH: Weinhiem, Germany, 2002;
179–202.

23. Kleine, J.; Kleine, H. Unber hochmolekulare,


insbesondere optische aktive polyester des milchsa¨ure, ein
beitrag zur stereochemie makromolekularer verbindungen.
Makromol. Chem. 1959, 30, 23–38.

24. Kricheldorf, H.R.; Kreiser-Saunders, I.; Stricker, A.


Polylactones 48. Sn-Oct2-initiated polymerizations of
lactide: A mechanistic study. Macromolecules 2000, 33,
702–709.

25. Dubois, P.; Jacobs, C.; Je´roˆme, R.; Teyssie´, P.


Macromolecular engineering of polylactones and
polylactides. IV. Mechanism and kinetics of lactide
homopolymerization by aluminum isopropoxide. Macromolecules
1991, 24, 2266–2270.

26. Kowalski, A.; Duda, A.; Penczek, S. Kinetics and


mechanism of cyclic esters polymerization initiated with
tin(II) octoate. 3. Polymerization of L-L-dilactide.
Macromolecules 2000, 33, 7359–7370. 27. Schwach, G.;
Coudane, J.; Engel, R.; Vert, M. Ring opening
polymerization of DL-lactide in the presence of zinc–metal
and zinc–lactate. Polym. Int. 1998, 46, 177–182. 28.
Zhiyan-Zhong; Dijkstra, P.J.; Birg, C.; Westerhausen, M.;
Feijen, J. A novel and versatile calcium-based initiator
system for the ring-opening polymerization of cyclic
esters. Macromolecules 2001, 34, 3863–3868. 29. Sarasua,
J.R.; Prud’homme, R.E.; Wisniewski, M.; Le-Borgne, A.;
Spassky, N. Crystallization and melting behavior of
polylactides. Macromolecules 1998, 31, 3895–3905. 30.
Leray, J.; Vert, M.; Blanquaert, D. Nouveau mate´riau de
prothe`se osseuse et son application. FR7628629, September
20, 1976. 31. Vert, M.; Schwach, G.; Engel, R.; Coudane, J.
Something new in the field of PLA/GA bioresorbable polymers.
J. Control. Release 1998, 53, 85–92. 32. Schwach, G.;
Coudane, J.; Engel, R.; Vert, M. Zn lactate as initiator of
DL-lactide ring opening polymerization and comparison with
Sn octoate. Polym. Bull. 1996, 37, 771–776. 33. Deasy,
P.B.; Finan, M.P.; Meegan, M.J. Preparation and
characterization of lactic/glycolic acid polymers and
copolymers. J. Microencapsul 1989, 6, 369–378. 34. He, B.;
Bei, J.; Wang, S. Synthesis and characterization of a
functionalized biodegradable copolymer: Poly (
L-lactide-co-RS-b-malic acid). Polymer 2003, 44, 989–994.
35. Barakat, I.; Dubois, Ph.; Grandsfils, Ch.; Jerome, R.
Poly(e-caprolactone-b-glycolide) and poly(
D,L-lactideb-glycolide) diblock copolyesters: Controlled
synthesis, characterization, and colloidal dispersions. J.
Polym. Sci., A, Polym. Chem. 2001, 39, 294–306. 36. Dong,
C.M.; Qiu, K.Y.; Gu, Z.W.; Feng, X.D. Living polymerization
of D,L-3-methylglycolide initiated with bimetallic (Al/Zn)
m-oxo alkoxide and copolymers thereof. J. Polym. Sci., A,
Polym. Chem. 2001, 39, 357– 367. 37. Inata, H.; Matsumura,
S. Chain extenders for polyester. II: Reactivities of
carboxyl-addition-type chain extenders; bis
cyclic-imino-ethers. J. Appl. Polym. Sci. 1986, 32,
5193–5202. 38. Younes, H.; Cohn, D. Morphological study of
biodegradable PEO/PLA block copolymers. J. Biomed. Mater.
Res. 1987, 21, 1301–1316. 39. Kurcock, P.; Penczek, H.J.;
Franek, J.; Jedlinski, Z. Anionic polymerization of
lactones. XIV: Anionic block copolymerization of
d-valerolactone and L-lactide initiated with potassium
methoxide. Macromolecules 1992, 25, 2285–2289. 40. Dubois,
P.; Dege´e, P.; Ropson, N.; Jerome, R. Macromolecular
Engineering of Polylactones and Polylactides by Ring
Opening Polymerization. In Macromolecular Design of
Polymeric Materials; Hatada, K., Kitayama, T., Vogel, O.,
Eds.; Marcel Dekker Inc.: New York, 1997; 247–272. 41.
Zell, M.T.; Padden, B.E.; Paterick, A.J.; Thakur, K.A.M.;
Kean, R.T.; Hillmyer, M.A.; Munson, E.J. Unambiguous
determination of the 13 C and 1 H NMR stereosequence
assignments of polylactide using high resolution solution
NMR spectroscopy. Macromolecules 2002, 35, 7700–7707. 42.
Urayama, H.; Kanamori, T.; Kimura, Y. Microstructure and
thermomechanical properties of glassy polylactides with
different optical purity of the lactate units. Macromol.
Mater. Eng. (Print) 2001, 286, 705–713. 43. Kister, G.;
Cassanas, G.; Vert, M. Effects of morphology, conformation
and configuration on the IR and Raman spectra of various
poly(lactic acid)s. Polymer 1998, 39, 267–273. 44.
Libiszowski, J.; Kowalski, A.; Duda, A.; Penczek, S.
Kinetics and mechanism of cyclic esters polymerization
initiated with covalent metal carboxylates, 5. End-group
studies in the model e-caprolactone and L,L-dilactide/
tin(II) and zinc octoate/butyl alcohol systems. Macromol.
Chem. Phys. (Print) 2002, 203, 1694–1701. 45. Braud, C.;
Devarieux, R.; Garreau, H.; Vert, M. Capillary
electrophoresis to analyze water-soluble oligo (hydroxy
acids) issued from degraded or biodegraded aliphatic
polyesters. J. Environ. Polym. Degrad. 1996, 4, 135–148.
46. Vert, M.; Se´le´gny, E. The optical properties of
chiral centres as tools for studying irreversible or
reversible reactions of synthetic polymers in solution. J.
Polym. Sci., C, Polym. Symp. 1973, 42, 1239–1248. 47. Pitt,
C.G.; Gratzel, M.M.; Kimmel, G.L.; Surles, J.; Schindler,
A. Aliphatic polyesters. 2. The degradation of poly(
DL-lactide), poly (e-caprolactone) and their complexes in
vivo. Biomaterials 1981, 2, 215–220. 48. Li, S.; Garreau,
H.; Vert, M. Structure–property relationships in the case
of degradation of solid aliphatic poly (a-hydroxy acids) in
aqueous media: 3. Amorphous and semi-crystalline PLA100. J.
Mater. Sci., Mater. Med. 1990, 1, 198–206. 49. Li, S.;
Garreau, H.; Vert, M. Structure–property relationships in
the case of degradation of solid aliphatic poly (a-hydroxy
acids) in aqueous media: 2. PLA37.5GA25 and PLA75GA25
copolymers. J. Mater. Sci., Mater. Med. 1990, 1, 131–139.
50. Schmitt, E.A.; Flanagan, D.R.; Lindhart, R.J.
Importance of distinct water environments in the hydrolysis
of poly( DL-lactide-co-glycolide). Macromolecules 1994, 27,
743–748. 51. Vert, M. Bioresorbable Synthetic Polymers and
Their Operation Field. In Biomaterials in Surgery;
Walenkamp, G., Ed.; Georg Thieme Verlag: Stuttgart, 1998;
97–101. 52. Vert, M.; Li, S.; Garreau, H. Attempts to map
structure and degradation characteristics of aliphatic
polyesters derived from lactic and glycolic acids. J.
Biomater. Sci., Polym. Ed. 1994, 6, 639–649. 53. Vert, M.
Degradation of Polymeric Biomaterials with Respect to
Temporary Applications. In Degradable Materials; Baremberg,
S.A., Brash, J.L., Narayan, R., Redpath, A.E., Eds.; CRC
Press: Boca Raton, 1990; 11–37. 54. Mauduit, J.; Perouse,
E.; Vert, M. Hydrolytic degradation of films prepared from
blends of high and low molecular weight poly( DL-lactic
acid)s. J. Biomed. Mater. Res. 1996, 30, 201–207. 55.
Cordeweiner, F.W.; Rozema, F.R.; Bos, R.R.; Boering, G.
Material properties and tissue reaction during degradation
of poly(96L/4D-lactide)—A study in vitro and in rats. J.
Mater. Sci., Mater. Med. 1995, 6, 211–218. 56. Li, S.M.;
Vert, M. Hydrolytic degradation of coral/ poly( DL-lactic
acid) bioresorbable materials. J. Biomater. Sci., Polym.
Ed. 1996, 7, 817–827. 57. Li, S.M.; Girod-Holland, S.;
Vert, M. Hydrolytic degradation of poly( DL-lactic acid) in
the presence of caffein base. J. Control. Release 1996, 40,
41–53. 58. Mauduit, J.; Bukh, N.; Vert, M. Gentamycin/poly
(lactic acid) blends aimed at sustained release local
antibiotic therapy administered per-operatively. III. The
case of gentamycin sulfate in films prepared from high and
low molecular weight poly( DL-lactic acids). J. Control.
Release 1993, 25, 43–49. 59. Li, S.; Vert, M. Biodegradable
Polymers: Polyesters. In The Encyclopaedia of Controlled
Drug Delivery; Mathiowitz, Ed.; J. Wiley & Sons: New York,
1999; 71–93. 60. Brannon-Peppas, L.; Vert, M. Polylactic
and Polyglycolic Acids as Drug Delivery Carriers. In
Handbook of Pharmaceutical Controlled Release Technology;
Wise, L.R., Klibanov, D.L., Mikos, A., Brannon-Peppas, L.,
Peppas, N.A., Trantalo, D.J., Wnek, G.E., Michael, J.,
Yaszemski, M.J., Eds.; Marcel Dekker: New York, 2000;
90–130. 61. Grizzi, I.; Garreau, H.; Li, S.; Vert, M.
Biodegradation of devices based on poly( DL-lactic acid):
Size dependence. Biomaterials 1995, 16, 305–311. 62.
Schwach, G.; Engel, R.; Coudane, J.; Vert, M. Stannous
octoate versus zinc-initiated polymerization of racemic
lactide: Effect of configurational structures. Polym. Bull.
1994, 32, 617–623. 63. Zhang, X.; MacDonald, D.A.; Goosen,
M.F.A.; Mcauley, K.B. Mechanism of lactide polymerization
in the presence of stannous octoate: The effect of hydroxy
and carboxylic acid substances. J. Polym. Sci., A, Polym.
Chem. 1994, 32, 2965–2970. 64. Schwach, G.; Coudane, J.;
Engel, R.; Vert, M. More about the initiation mechanism of
lactide polymerization in the presence of stannous octoate.
J. Polym. Sci., A, Polym. Chem. 1997, 35, 3431–3440. 65.
Cutright, D.E.; Perez, B.; Beasley, J.D.; Larson, W.J.;
Rosey, W.R. Degradation rates of polymers and copolymer of
polylactic and polyglycolic acids. Oral Surg. 1974, 37,
142–152. 66. Miller, R.A.; Brady, J.M.; Cutright, D.E.
Degradation rates of oral resorbable polyesters for
orthopaedic surgery. J. Biomed. Mater. Res. 1977, 11,
711–719. 67. Guilding, D.K.; Reed, A.M. Biodegradable
polymers for use in surgery—Polyglycolic/poly(lactic acid)
homoand copolymers: 1. Polymer 1979, 20, 1459–1464. 68.
Sedel, L.; Chabot, F.; Christel, P.; de Charantenay, F.X.;
Leray, J.; Vert, M. Les implants biode´gradables en
chirurgie orthope´dique. Rev. Chir. Orthop. 1978, 64,
92–101. 69. Reed, A.M.; Guilding, D.K. Biodegradable
polymers for use in surgery—Poly(glycolic)/poly(lactic
acid) homo and copolymers. Polymer 1981, 22, 494–498. 70.
Vert, M.; Li, S.M.; Spenlehauer, G.; Guerin, P.
Bioresorbability and biocompatibility of aliphatic
polyesters. J. Mater. Sci., Mater. Med. 1992, 3, 432–446. P

71. Ignatius, A.A.; Claes, L.E. In vitro biocompatibility


of bioresorbable polymers: Poly( L,DL-lactide) and poly (
L-lactide-co-glycolide). Biomaterials 1996, 17, 831–839.

72. Hollinger, J.O. Preliminary report on the osteogenic


potential of a biodegradable copolymer of polylactide (PLA)
and polyglycolide (PGA). J. Biomed. Mater. Res. 1983, 17,
71–82.

73. Sharma, C.P.; Williams, D.F. The effects of lipids on


the mechanical properties of polyglycolic acid structures.
Eng. Med. 1981, 10, 8–10.
74. Lee, K.H.; Chu, C.C. The role of superoxide ions in the
degradation of synthetic absorbable sutures. J. Biomed.
Mater. Res. 2000, 49, 25–35.

75. Kenley, R.A.; Ott Lee, M.; Mahoney, T.R.; Sanders, L.


Poly(lactide-co-glycolide) decomposition kinetics in vivo
and in vitro. Macromolecules 1987, 20, 2398–2403.

76. The´rin, M.; Christel, P.; Li, S.; Garreau, H.; Vert,
M. Degradation of massive poly(a-hydroxy acids) in aqueous
living medium: In vivo validation of in vitro findings.
Biomaterials 1992, 13, 594–600.

77. Gogolewski, S.; Jovanovic, M.; Perren, S.M.; Dillon,


J.G.; Hughes, M.K. Tissue response and in vivo degradation
of selected polyhydroxyacids: Polylactides (PLA),
poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHB/VA). J.
Biomed. Mater. Res. 1993, 27, 1135–1148.

78. Mainil-Varlet, P.; Curtis, R.; Gogolewski, S. Effect of


in vivo and in vitro degradation on molecular and
mechanical properties of various low-molecular weight
polylactides. J. Biomed. Mater. Res. 1997, 36, 360–380.

79. Tabata, Y.; Ikada, Y. Macrophage phagocytosis of


biodegradable microspheres composed of L-lactic acid/
glycolic acid homo and copolymers. J. Biomed. Mater. Res.
1988, 22, 837–858.

80. Serizawa, T.; Arikawa, Y.; Hamada, K.; Yamashita, H.;


Fujikawa, T.; Kimura, Y.; Akashi, M. Alkaline hydrolysis of
enantiomeric poly(lactide)s stereocomplexes deposited on
solid substrates. Macromolecules 2003, 36, 1762–1765.

81. Dos Santos, I.; Morgat, J.L.; Vert, M. Hydrogen isotope


exchange as a means of labeling lactides. J. Labelled Compd
Radiopharm. 1998, 41, 1005–1012.

82. Dos Santos, I.; Morgat, J.L.; Vert, M. Glycolide


deuteration by hydrogen isotope exchange using the HSCIE
method. J. Labelled Compd Radiopharm. 1999, 42, 1093–1101.

83. Middleton, J.C.; Tipton, A.J. Synthetic biodegradable


polymers as orthopeadic devices. Biomaterials 2000, 21,
2325–2346.

84. Vert, M.; Chabot, F.; Leray, J.; Christel, P. Nouvelles


pie`ces d’oste´osynthe`se, leur pre´paration et leur
application. FR782987, October 20, 1978.
85. To¨rma¨la¨, P.; Vasenius, J.; Vainionpa¨a¨, S.; Laiho,
J.; Pohjonen, T.; Rokkanen, P. Ultra high strength
absorbable self-reinforced polyglycolide (SR-PGA) composite
rods for internal fixation of bone fractures: In vitro and
in vivo study. J. Biomed. Mater. Res. 1991, 25, 22.85.

86. Joukainen, A.; Pihlajamaki, H.; Makela, E.A.;


Ashammakhi, N.; Viljanen, J.; Patiala, H.; Kellomaki, M.;
Tormala, P.; Rokkanen, P. Strength retention of
self-reinforced drawn polyL/DL-lactide 70/30 (SR-PLA70)
rods and fixation properties of distal femoral osteotomies
with these rods. An experimental study on rats. J.
Biomater. Sci., Polym. Ed. 2000, 11, 1411–1428. 87.
Chanavaz, M.; Chabot, F.; Donazzan, M.; Vert, M. Further
Clinical Applications of Bioresorbable PLA 37.5 GA 25 and
PLA 50 Polymers for Limited Bone Augmentation and Bone
Replacement in Human. In Biological and Biomechanical
Performance of Biomaterials; Christel, P., Meunier, A.,
Lee, A.J.C., Eds.; Elsevier, 1986; 233–238. 88. Oudega, M.;
Gautier, S.E.; Chapon, P.; Fragoso, M.; Bates, M.L.; Parel,
J.-M.; Bunge, M.B. Axonal regeneration into Schwann cell
grafts within resorbable poly(alpha-hydroxyacid) guidance
channels in the adult rat spinal cord. Biomaterials 2001,
22, 1125–1136. 89. Anderson, J.M.; Shive, M.S.
Biodegradation and biocompatability of PLA and PLGA
microspheres. Adv. Drug Deliv. Rev. 1997, 28, 5–24. 90.
Brannon-Peppas, L. Recent advances on the use of
biodegradable microparticles and nanoparticles in
controlled drug delivery. Int. J. Pharm. 1995, 116, 1–9.
91. Agrawal, C.M.; Niedersuer, C.C.; Micallef, D.M.;
Athanasiou, K.A. The Use of PLA–PGA Polymers in
Orthopaedics. In Encyclopedic and Book of Biomaterials and
Bioengineering; Marcel Dekker: New York, 1995; 2081–2115.
Chap. 30. 92. Athanasiou, K.A.; Schmitz, J.P.; Agrawal,
C.M. The effects of porosity on in vitro degradation of
polylactic acid–polyglycolic acid implants used in repair
of articular cartilage. Tissue Eng. 1998, 4, 53–63. 93.
Robert, P.M.; Frank, R.M. Periodontal guided tissue
regeneration with a new bioresorbable polylactic acid
membrane. J. Periodontol. 1994, 65, 414–422. 94. Ishaug,
S.L.; Yashemski, M.J.; Bizios, R.; Mikos, A.G. Osteoblast
function on synthetic biodegradable polymers. J. Biomed.
Mater. Res. 1994, 28, 1445–1453. 95. Holder, W.D., Jr.;
Gruber, H.E.; Moore, A.L.; Culberson, C.R.; Anderson, W.;
Burg, K.J.L.; Mooney, D.J. Cellular ingrowth and thickness
changes in polyL-lactide and polyglycolide matrices
implanted subcutaneously in the rat. J. Biomed. Mater. Res.
1998, 41, 412–421. 96. Ikada, Y.; Tsuji, H. Biodegradable
polyesters for medical and environmental applications.
Rapid Commun. 2000, 21, 117–132. 97. Gruber, P.; O’Brien,
M. Polylactides Nature Workst PLA. In Biopolymers: Biology,
Chemistry, Biotechnology, Applications, Polyesters III;
Doi, Y., Steinbu¨chel, A., Eds.; Wiley-VCH, Verlag GmbH:
Weinhiem, Germany, 2002; 235–250. 98. Kawashima, N.; Ogawa,
S.; Obuchi, S.; Matsuo, M.; Yagi, T. Polylactic Acid
)Lacea*. In Biopolymers: Biology, Chemistry, Biotechnology,
Applications, Polyesters III; Doi, Y., Steinbu¨chel, A.,
Eds.; Wiley-VCH, Verlag GmbH: Weinhiem, Germany, 2002;
251–274.
Polylactic-Co-Glycolic Acid (PLGA)

1. Wise, D.L.; Fellman, T.D.; Sanderson, J.E.; Wentworth,


R.L. Lactic/glycolic acid polymers. In Drug Carriers in
Biology and Medicine; Gregoriadis, G., Ed.; Academic Press:
New York, 1979; 237–270.

2. Ajioka, M.; Suizu, H.; Higuchi, C.; Kashima, T.


Aliphatic polyesters and their copolymers synthesized
through direct condensation polymerization. Polym. Degrad.
Stab. 1998, 39, 137–143.

3. Takahashi, K.; Taniguchi, I.; Miyamoto, M.; Kimura, Y.


Melt/solid polycondensation to obtain high-molecular-weight
poly(glycolic acid). Polymer 2000, 41, 137–143.

4. Chabot, F.; Vert, M.; Chapelle, S.; Granger, P.


Configurational structures of lactic acid stereocopolymers
as determined by 13 C1 {H} n.m.r.. Polymer 1983, 24 (2),
53–59.

5. Gilding, D.K.; Reed, A.M. Biodegradable polymers for use


in surgery-polyglycolic/poly(lactic acid) homoand
copolymers: 1. Polymer 1979, 20 (12), 137–143.

6. Dittrich, W.; Schulz, R.C. Kinetic und Mechanismus der


ringoffnenden polymerization von L(�)�Lactid. Angew.
Macromol. Chem. 1971, 15, 109–126.

7. Kowalski, A.; Duda, A.; Penczek, S. Kinetics and


mechanism of cyclic esters polymerization initiated with
tin(II) octoate 3. Polymerization of L,L-dilactide.
Macromolecules 2000, 33, 7359–7370.

8. Kricheldorf, H.R.; Kreiser-Saunders, I.; Stricker, A.


Polylactones 48. SnOct2-initited polymerizations of
lactide. Macromolecules 2000, 33, 702–709.

9. Avgoustakis, K.; Nixon, J.R. Biodegradable controlled


release tablets 1: preparative variables affecting the
properties of poly(lactide-co-glycolide) copolymers as
matrix forming material. Int. J. Pharm. 1991, 70, 77–85.

10. Hyon, S.H.; Jamshidi, K.; Ikada, Y. Synthesis of


polylactides with different molecular weights. Biomaterials
1997, 18 (22), 1503–1508.

11. Perrin, D.E.; English, J.P. Polyglycolide and


polylactide. In Handbook of Biodegradable Polymers; Domb,
A.J., Kost, J., Wiseman, D.M., Eds.; In Drug Targeting and
Delivery; Florence, A.T., Gregoriadis, G., Series Eds.;
Harwood Academic Publishers: Amsterdam, 1997; 3–27.

12. Vert, M.; Chabot, F.; Leray, J.; Christel, P.


Stereoregular bioresorbable polyesters for orthopaedic
surgery. Macromol. Chem. Suppl. 1981, 5, 30–41.

13. Daniels, A.U.; Chang, M.K.; Andriano, K.P. Mechanical


properties of biodegradable polymers and composites
proposed for internal fixation of bone. J. Appl. Biomater.
1990, 1 (1), 57–78.

14. Pitt, C.G.; Gu, Z. Modification of the rates of chain


cleavage of poly(e-caprolactone) and related polyesters in
the solid state. J. Controlled Release 1987, 4, 283–292.
15. Tracy, M.A.; Ward, K.L.; Firouzabadian, L.; Wang, Y.;
Dong, N.; Qian, R.; Zhang, Y. Factors affecting the
degradation rate of poly(lactide-co-glycolide) microspheres
in vivo and in vitro. Biomaterials 1999, 20, 1057–1062. 16.
Grizzi, I.; Garreau, H.; Li, S.; Vert, M. Hydrolytic
degradation of devices based on poly( DL-lactic acid)
size-dependence. Biomaterials 1995, 16 (4), 305–311. 17.
Avgoustakis, K. In Synthesis and Evaluation of Some
Poly(Lactide-co-Glycolides) for Use in Sustained Release
Tablets. Ph.D. thesis, University of London, 1992. 18.
Williams, D.F. Biodegradation of surgical polymers. J.
Mater. Sci. 1982, 17, 1233–1246. 19. Vert, M. Polyglycolide
and copolyesters with lactides. In Biopolymers: Biology,
Chemistry, Biotechnology, Applications, Vol. 4 Polyesters
III; Doi, Y., Steinbuchel, A., Eds.; Wiley-VCH Verlag GmbH:
Weinheim, Germany, 2002; 179–202. 20. Anderson, J.M.;
Shive, S.M. Biodegradation and biocompatibility of PLA and
PLGA microspheres. Adv. Drug Deliv. Rev. 1997, 28, 5–24.
21. Drury, D.R.; Wick, A.N. Chemistry and metabolism of
L(+) and D(�) lactic acids. Am. J. Physiol. 1956, 184,
1061–1069. 22. Alexis, F. Factors affecting the degradation
and drug-release mechanism of poly(lactic acid) and
poly[(lactic acid)-co-(glycolic acid)]. Polym. Int. 2005,
54, 36–46. 23. Vert, M. Degradation of polymeric systems
aimed at temporary therapeutic applications:
structure-related complications. e-Polymers 2005, 8, 1–10.
24. Hyon, S.H.; Jamshidi, K.; Ikada, Y. Effects of residual
monomer on the degradation of DL-lactide polymer. Polym.
Int. 1998, 46 (3), 196–202. 25. Miller, R.A.; Brady, J.M.;
Cutright, D.E. Degradation rates of oral resorbable
implants (polylactates and polyglycolates): rate
modification with changes in PLA/ PGA copolymer ratios. J.
Biomed. Mater. Res. 1977, 11, 711–719. 26. Dunn, R.L.;
English, J.P.; Strobel, J.D.; Cowsar, D.R.; Tice, T.R.
Preparation and evaluation of lactide/glycolide copolymers
for drug delivery. In Polymers in Medicine III; Migliaresi,
C., Nikolais, L., Giusti, P., Chiellini, E., Eds.;
Elsevier: Amsterdam, 1988; 149–160. 27. Kamei, S.; Inoue,
Y.; Okada, H.; Yamada, M.; Ogawa, Y.; Toguchi, H. New
method for analysis of biodegradable polyesters by
high-performance liquid chromatography after alkali
hydrolysis. Biomaterials 1992, 13, 953–958. 28. Schwach,
G.; Goudane, J.; Engel, R.; Vert, M. More about the
initiation mechanism of lactide polymerization in the
presence of stannous octoate. J. Polym. Sci. A. Polym.
Chem. 1997, 35, 3431–3440. 29. Loo, S.C.J.; Ooi, C.P.;
Boey, Y.C.F. Radiation effects on
poly(lactide-co-glycolide) (PLGA) and poly(lactide) (PLLA).
Polym. Degrad. Stab. 2004, 83, 259–265. 30. Gogolewski, S.;
Mainil-Varlet, P.; Dillon, J.G. Sterility, mechanical
properties, and molecular stability of polylactide
internal-fixation devices treated with lowtemperature
plasmas. J. Biomed. Mater. Res. 1996, 32, 227–235. 31.
Vert, M.; Christel, P.; Chabot, F.; Leray, J. Bioresorbable
plastic materials for bone surgery. In Macromolecular
Biomaterials; Hastings, G.W., Ducheyne, P., Eds.; CRC
Press: Boca Raton, FL, 1984; 119–142. 32. Visscher, G.E.;
Robison, R.L.; Maulding, H.V.; Fong, J.W.; Pearson, J.E.;
Argentieri, G.J. Biodegradation of and tissue reaction to
50:50 poly( DL-lactide-co-glycolide) microcapsules. J.
Biomed. Mater. Res. 1985, 19, 349–365. 33. Tiainen, J.;
Soini, Y.; Tormala, P.; Waris, T.; Ashammakhi, N.
Self-reinforced polylactide/polyglycolide 80/20 s crews
take more than 11/2 years to resorb in rabbit cranial bone.
J. Biomed. Mater. Res. B Appl. Biomater. 2004, 70, 49–55.
34. Racey, G.L.; Wallace, W.R.; Cavalaris, J.; Marguard,
J.V. Comparison of a polyglycolic–polylactic acid suture to
black silk and plain catgut in human oral tissues. J. Oral
Surg. 1978, 36, 766–770. 35. Rokkanen, P.; Vainionpaa, S.;
Tormala, P.; Kilpikari, J.; Bostman, O; Vihtonen, K.; Laiho
J. Tamminmaki, M. Biodegradable implants in fracture
fixation: early results of treatment of fracture of the
ankle. Lancet 1985, 1, 1422–1424. 36. Ashammakhi, N.;
Renier, D.; Arnaud, E.; Marshac, D.; Ninkovich, M.;
Donaway, D.; Jones, B.; Serlo, W.; Laurikainen, K.;
Tormala, P.; Waris, T. Successful use of Biosorb
osteofixation devices in 165 cranial and maxillofacial
cases: a multicenter report. J. Craniofac. Surg. 2004, 15
(4), 692–701. 37. Athanasiou, K.A.; Niederauer, G.G.;
Agrawal, C.M. Sterilization, toxicity, biocompatibility,
and clinical applications of polylactic acid/poly-glycolic
acid copolymers. Biomaterials 1996, 17, 93–102. 38.
Rokkanen, P.; Bostman, O.; Hirvensalo, E.; Partio, E.K.;
Makela, E.A.; Patiala, H.; Vihtonen, K. Bioabsorbable
implants in orthopaedics. Curr. Orthop. 1999, 13, 223–228.
39. Middleton, J.C.; Tipton, A.J. Synthetic biodegradable
polymers as orthopedic devices. Biomaterials 2000, 21,
2335–2346. 40. Jain, R.; Shah, N.H.; Malick, A.W.; Rhodes,
C.T. Controlled drug delivery by biodegradable poly-(ester)
devices: different preparative approaches. Drug Dev. Ind.
Pharm. 1998, 24 (8), 703–727. 41. Panyam, J.; Labhasetwar,
V. Biodegradable nanoparticles for drug and gene delivery
to cells and tissue. Adv. Drug Deliv. Rev. 2003, 55 (3),
329–347. 42. Johansen, P.; Men, Y.; Merckle, H.P.; Gander,
B. Revisiting PLA/PLGA microspheres: an analysis of their
potential in parenteral vaccination. Eur. J. Pharm.
Biopharm. 2000, 50, 129–146. 43. Mooney, D.J.; Baldwin,
D.F.; Suh, N.P.; Vacanti, J.P.; Langer, R. Novel approach
to fabricate porous sponges of poly( D,L-lactic-co-glycolic
acid) without the use of organic solvents. Biomaterials
1996, 17, 1417–1422. 44. Griffith, L.G.; Naughton, G. Tissue
engineering-current challenges and expanding opportunities.
Science 2002, 295, 1009–1014. 45. Avgoustakis, K. Pegylated
poly(lactide) and poly(lactide-co-glycolide) nanoparticles:
preparation, properties, and possible applications in drug
delivery. Curr. Drug Deliv. 2004, 1 (4), 321–333. FURTHER
READING Tsuji, H. Polylactides. In Biopolymers: Biology,
Chemistry, Biotechnology, Applications, Vol. 4 Polyesters
III; Doi, Y., Steinbuchel, A., Eds.; Wiley-VCH, Verlag
GmbH: Weinheim, Germany, 2002; 129–178. P
Polymer Foams

1. Gibson, L.J.; Ashby, M.J. Cellular Solids: Structure and


Properties, 2nd Ed.; Pergamon: Oxford, United Kingdom,
1997.

2. Suh, K.W.; Skochdopole, R.E. Encyclopedia of Chemical


Technology, 3rd Ed.; Kirk-Othmer, Ed.; John Wiley & Sons,
1980; Vol 2, 82.

3. Mooney, D.J.; Baldwin, D.F.; Suh, N.P.; Vacanti, J.P.;


Langer, R. Novel approach to fabricate porous sponges of
poly( DL-lactic-co-glycolic acid) without the use of
organic solvents. Biomaterials 1996, 17 (14), 1417– 1422.

4. Mikos, A.G.; Thorsen, A.J.; Czerwonka, L.A.; Bao, Y.;


Langer, R. Preparation and characterization of poly (
L-lactic acid) foams. Polymer 1994, 35, 1068–1077.

5. Chen, G.; Ushida, T.; Tateishi, T. Preparation of poly(


L-lactic acid) and poly(DL-lactic-co-glycolic acid) foams
by use of ice microparticles. Biomaterials 2001, 22 (18),
2563–2567. 6. Shastri, V.P.; Martin, I.; Langer, R.
Macroporous polymeric foams by hydrocarbon templating.
Proc. Natl. Acad. Sci. U.S.A. 2000, 97 (5), 1970–1975. 7.
Schaefer, D.; Martin, I.; Shastri, V.P.; Padera, R.F.;
Langer, R.; Freed, L. E.; Vunjak-Novakovic, G. In vitro
engineering of bone-cartilage composite tissues.
Biomaterials 2000, 21, 2599–2606. 8. Martin, I.; Shastri,
V.P.; Padera, R.F.; Langer, R.; Yang, J.; MacKay, A.;
Vunjak-Novakovic, G.; Freed, L.E. Selective in vitro
differentiation of mammalian mesenchymal progenitor cells
into three dimensional skeletal tissues. J. Biomed. Mat.
Res. 2001, 55 (2001), 229–235. 9. Ma, P.X.; Choi, J.W.
Biodegradable polymer scaffolds with well-defined
interconnected spherical pore network. Tissue Eng. 2001, 7
(1), 23–33. 10. Zhang, R.; Ma, P.X. Poly(alpha-hydroxyl
acids)/ hydroxyapatite porous composites for bone-tissue
engineering I. Preparation and morphology. J. Biomed. Mat.
Res. 1999, 44 (4), 446–455. 11. Lo, H.; Kadiyala, S.;
Guggino, S.F.; Leong, K.W. Poly( L-lactic acid) foams with
cell-seeding and controlled release capacity. J. Biomed.
Mat. Res. 1996, 30 (4), 475–484. 12. Hile, D.; Amirpour,
M.L.; Akgerman, A.; Pishko, M.V. Active growth factor
delievry from poly( DL-lactideco-glycolide) foams prepared
in super critical CO 2 . J. Control. Release 2000, 66
(2–3), 177–185. 13. Schugens, C.; Maquet, V.; Grandfils, C.;
Jerome, R.; Teyssie, P. Polylactide macroporous
biodegradable implants for cell transplantation. II.
Preparation of polylactide foams by liquid-liquid phase
separation. J. Biomed. Mater. Res. 1996, 30 (4), 449–461.
14. Teng, Y.D.; Lavik, E.B.; Qu, X.; Park, K.I.; Ourednik,
J.; Zurakowski, D.; Langer, R.; Snyder, E.Y. Functional
recovery following traumatic spinal cord injury mediated by
a unique polymer scaffold seeded with neural stem cells.
Proc. Natl. Acad. Sci. U.S.A. 2002, 99 (5), 3024–3029. 15.
Thomson, R.C.; Yaszemski, M.J.; Powers, J.M.; Mikos, A.G.
Fabrication of biodegradable polymer scaffolds to engineer
trabecular bone. J. Biomater. Sci., Polym. Ed. 1995, 7 (1),
23–38. 16. Freed, L.E.; Vunjak-Novakovic, G.; Biron, R.J.;
Eagles, D.B.; Lesnoy, D.C.; Barlow, S.K.; Langer, R.
Biodegradable polymer scaffolds for tissue engineering.
Biotechnology (NY) 1994, 2 (7), 689–693. 17. Freed, L.E.;
Marquis, J.C.; Nohria, A.; Emmanual, J.; Mikos, A.G.;
Langer, R. Neocartilage formation in vitro and in vivo
using cells cultured on synthetic biodegradable polymers.
J. Biomed. Mat. Res. 1993, 27 (1), 11–23. 18. Mikos, A.G.;
Bao, Y.; Cima, L.G.; Ingber, D.E.; Vacanti, J.P.; Langer,
R. Preparation of poly(glycolic acid) bonded fiber
structures for cell attachment and transplantation. J.
Biomed. Mat. Res. 1993, 27 (2), 183–189.
Polymers

1. Morawetz, H. Polymers: The Origin and Growth of a


Science; Wiley-Interscience: New York, 1985.

2. Allcock, H.R.; Lampe, F.; Mark, J. Contemporary Polymer


Chemistry, 3rd Ed.; Prentice Hall: Englewood Cliffs, NJ,
2003.

3. Qui, J.; Charleux, B.; Matyjaszewski, K. . Prog. Polym.


Sci. 2001, 26, 2083.

4. Organometallic Catalysts and Olefin Polymerization; Blom,


R., Follestad, A., Rytter, E., Tilset, M., Ystenes, M.,
Eds.; Springer-Verlag: Berlin, 2001.

5. Roovers, J.; Comanita, B. . Adv. Polym. Sci. 1999, 142,


180.

6. Materials Research to Meet 21st-Century Defense Needs;


National Academies Press: Washington, DC, 2003. P
Polymers Used in Tissue Engineering

13. Mooney, D.; Hansen, L.; Vacanti, J.; Langer, R.;


Farmer, S.; Ingber, D. Switching from differentiation to
growth in hepatocytes:control by extracellular matrix. J.
Cell. Physiol. 1992, 151, 497–505.

14. Ruoslahti, E.; Pierschbacher, M.D. New perspectives in


cell adhesion: RGD and integrins. Science 1987, 238 (4826),
491–497.

15. Griffith, L.G. Polymeric biomaterials. Acta Mater. 2000,


48, 263–277.

16. Rouslahti, E. RGD and other recognition sequences for


integrins. Annu. Rev. Cell Dev. Biol. 1996, 12, 697–715.

17. Clark, E.A.; Brugge, J.S. Integrins and signal


transduction pathways: the road taken. Science 1995, 268
(5208), 233–239.

18. Badylak, S.F. The extracellular matrix as a scaffold


for tissue reconstruction. Semin. Cell Dev. Biol. 2002, 13,
377–383.

19. Van Wachem, P.B.; Brouwer, L.A.; Zeeman, R.; Dijkstra,


P.J.; Feijen, J.; Hendriks, M.; Cahalan, P.T.; van Luyn,
M.J.A. Tissue reactions to epoxy-crosslinked porcine heart
valves post-treated with detergents or a dicarboxylic acid.
J. Biomed. Mater. Res. 2001, 55 (3), 415–423.

20. Chen, G.; Ushida, T.; Tateishi, T. Scaffold design for


tissue engineering. Macromol. Biosci. 2002, 2 (2), 67–77.

21. Yannas, I.V.; Burke, J.F. Design of an artificial skin:


basic design principles. J. Biomed. Mater. Res. 1980, 14,
65–81.

22. Bonassar, L.J.; Vacanti, C.A. Tissue engineering: the


first decade and beyond. J. Cell. Biochem. Suppl. 1998,
30/31, 297–303.

23. Katz, J. Developments in Medical Polymers for


Biomaterials Applications. Medical Device & Diagnostic
Industry Magazine, January 2001.

24. Middleton, J.C.; Tipton, A.J. Synthetic Biodegradable


Polymers as Medical Devices. Medical Plastics and
Biomaterials Magazine (Materials), March 1998.
25. Pachence, J.M.; Kohn, J. Biodegradable polymers for
tissue engineering. In Principles of Tissue Engineering;
Lanza, R.P., Langer, R., Chick, W.L., Eds.; Texas, USA,
1997; 273.

26. Agrawal, C.M.; Ray, R.B. Biodegradable polymeric


scaffolds for musculoskeletal tissue engineering. J.
Biomed. Mater. Res. 2001, 55, 141–150.

27. Gilding, D.K.; Reed, A.M. Biodegradable polymers for


use in surgery-poly(glycolic)/poly(lactic acid) homo and
copolymers. Polymer 1979, 20, 1459–1464.

28. Reed, A.M.; Gilding, D.K. Biodegradable polymers for


use in surgerypoly(glycolic)/poly(lactic acid) homo and
copolymers: 2. In vitro degradation. Polymer 1981, 22,
494–498.

29. Gilding, D.K. Biocompatibility of clinical implant


materials. In Williams, D.F., Ed.; Biodegradable Polymers;
CRC Press: Boca Raton, FL, 1981; Vol. II, 209–232. 30.
Bostman, O.M.; Pihlajamaki, H.K. Adverse tissue reactions
to bioabsorbable fixation devices. Clin. Orthop. Relat. Res.
2000, 371, 216–217. 31. Niklason, L.F.; Langer, R.S.
Advances in tissue engineering of blood vessels and other
tissues. Transpl. Immunol. 1997, 5, 303–306. 32. Shi, G.;
Cai, Q.; Wang, C.; Lu, N.; Wang, S.; Bei, J. Fabrication
and biocompatibility of cell sacffolds of poly ( L-lactic
acid) and poly (L-lacticco-glycolic acid). Polym. Adv.
Tech. 2002, 13, 227–232. 33. Hasirci, V.; Berthiaume, F.;
Bondre, S.P.; Gresser, J.D.; Trantolo, D.J.; Toner, M.;
Wise, D.L. Expression of liver-specific functions by rat
hepatocytes seeded in treated poly(lactic-co-glycolic) acid
biodegradable foams. Tissue Eng. 2001, 7 (4), 385–394. 34.
Woodward, S.C.; Brewer, P.S.; Moatamed, F.; Schindler, A.;
Pitt, C.G. The intracellular degradation of
poly(epsilon-caprolactone). J. Biomed. Mater. Res. 1985,
19, 437–444. 35. Williams, J.M.; Adewunmi, A.; Schek, R.M.;
Flanagan, C.L.; Krebsbach, P.H.; Feinberg, S.E.; Hollister,
S.J.; Das, S. Bone tissue engineering using
polycaprolactone scaffolds fabricated via selective laser
sintering. Biomaterials 2005, 26, 4817–4827. 36. Cao, T.;
Ho, K.-H.; Teoh, S.-H. Scaffold design and in vitro study
of osteochondral coculture in a threedimensional porous
polycaprolactone scaffold fabricated by fused deposition
modeling. Tissue Eng. 2003, 9 (1), 103–112. 37. Katti,
D.S.; Lakshmi, S.; Langer, R.; Laurencin, C.T. Toxicity,
biodegradation and elimination of polyanhydrides. Adv. Drug
Deliv. Rev. 2002, 54, 933–961. 38. Ibim, S.E.; Uhrich,
K.E.; Attawia, M.; Shastri, V.R.; El-Amin, S.F.; Bronson,
R.; Langer, R.; Laurencin, C.T. Preliminary in vivo report
on the osteocompatibility of poly(anhydride-co-imides)
evaluated in a tibial model. J. Biomed. Mater. Res. 1998,
43, 374–379. 39. Ibim, S.M.; Uhrich, K.E.; Bronson, R.;
El-Amin, S.F.; Langer, R.S.; Laurencin, C.T.
Poly(anhydride-coimides): In vivo biocompatibility in a rat
model. Biomaterials 1998, 19, 941–951. 40. Peter, S.J.;
Miller, S.T.; Zhu, G.; Yasko, G.; Mikos, A.G. In vivo
degradation of a poly(propylene fumarate)/b-tricalcium
phosphate injectable scaffold. J. Biomed. Mater. Res. 1998,
41, 1–7. 41. Peter, S.J.; Lu, L.; Kim, D.J.; Stamatas,
G.N.; Miller, M.J.; Yaszemski, M.J.; Mikos, A.G. Effects of
transforming growth factor b1 released from biodegradable
polymer microparticles on marrow stromal osteoblasts
cultured on poly(propylene fumarate) substrates. J. Biomed.
Mater. Res. 2000, 50, 452–462. 42. Payne, R.G.; McGonigle,
J.S.; Yaszemski, M.J.; Yasko, A.W.; Mikos, A.G. Development
of an injectable, in situ crosslinkable, degradable
polymeric carrier for osteogenic cell populations. Part 3.
Proliferation and differentiation of encapsulated marrow
stromal osteoblasts cultured on crosslinking poly
(propylene fumarate). Biomaterials 2002, 23 (22),
4381–4387. 43. Muggli, D.S.; Burkoth, A.K.; Keyser, S.A.;
Lee, H.R.; Anseth, K.S. Reaction behaviour of
biodegradable, photocross-linkable polyanhydrides.
Macromolecules 1998, 31, 4120–4125. 44. Betre, H.; Setton,
L.A.; Meyer, D.E.; Chilkoti, A. Characterization of a
genetically engineered elastinlike polypeptide for
cartilaginous tissue repair. Biomacromolecules 2002, 3 (5),
910–916. 45. Jeschke, B.; Meyer, J.; Jonczyk, A.; Kessler,
H.; Adamietz, P.; Meenen, N.M.; Kantlehner, M.; Goepfert,
C.; Nies, B. RGD-peptides for tissue engineering of
articular cartilage. Biomaterials 2002, 23 (16), 3455–3463.
46. Holmes, T.C.; Lacalle, S.; Su, X.; Liu, G.; Rich, A.;
Zhang, S. Extensive neurite outgrowth and active synapse
formation on self-assembling peptide scaffolds. PNAS 2000,
97 (12), 6728–6733. 47. Cho, N.; Heller, J. Drug delivery
devices manufactured from polyorthoesters and
polyorthocarbonates. US Patent 4093709, 1978. 48. Heller,
J.; Penhale, D.; Helwing, R. Preparation of poly(ortho
esters) by the reaction of kiketene acetals and polyols. J.
Polym. Sci. 1980, 18, 619–624. 49. Guo, Q.; Slavov, S.;
Halley, P.J. Phase behavior, crystallization, and
morphology in thermosetting blends of a biodegradable
poly(ethylene glycol)-type epoxy resin and poly(e
caprolactone). J. Polym. Sci. Part B: Polym. Phys. 2004,
42, 2833–2843. 50. Deschamps, A. Transactions of the Sixth
World Biomaterials Congress, Society for Biomaterials:
Minneapolis, 2000; 364 51. Drury, J.L.; Mooney, D.J.
Hydrogels for tissue engineering:scaffold design variables
and applications. Biomaterials 2003, 24, 4337–4351. 52.
Jeong, B.; Bae, Y.H.; Lee, D.S.; Kim, S.W. Biodegradable
block copolymers as injectable drug-delivery systems.
Nature 1997, 388, 860–862. 53. Huh, K.M.; Bae, Y.H.
Synthesis and characterization of poly(ethylene
glycol)/poly( L-lactic acid) alternating multiblock
copolymers. Polymer 1999, 40, 6147–6155. 54. Metters, A.T.;
Anseth, K.S.; Bowman, C.N. Fundamental studies of a novel,
biodegradable PEG-b-PLA hydrogel. Polymer 2000, 41,
3993–4004. 55. Suggs, L.J.; Mikos, A.G. Development of poly
(propylene fumarate-co-ethylene glycol) as an injectable
carrier for endothelial cells. Cell Trans. 1999, 8,
345–350. 56. Rosso, F.; Marino, G.; Giordano, A.;
Barbarisi, M.; Parmeggiani, D.; Barbarisi, A. Smart
materials as scaffolds for tissue engineering. J. Cell.
Physiol. 2005, 203, 465–470. 57. Park, S-N.; Park, J-C.;
Kim, H.O.; Song, M.J.; Suh, H. Characterization of porous
collagen/hyaluronic acid scaffold modified by
1-ethyl-3-(3-dimethylaminopropyl) carbodiimide
cross-linking. Biomaterials 2002, 23, 1205–1212. 58. Tan,
W.; Krishnaraj, R.; Desai, T.A. Evaluation of
nanostructured composite collagen–chitosan matricies for
tissue engineering. Tissue Eng. 2001, 7, 203–210. 59.
Huang, L.; Nagapudi, K.; Apkarian, R.P.; Chaikof, E.L.
Engineered collagen–PEO nanofibers and fabrics. J. Biomater.
Sci.: Polym. Ed. 2001, 12, 979–993. 60. Wakitani, S.; Goto,
T.; Young, R.; Mansour, J.; Goldberg, V.; Caplan, A. Repair
of large full-thickness articular cartilage defects with
allograft articular chondrocytes embedded in a collagen
gel. Tissue Eng. 1998, 4, 429–444. 61. Orwin, E.; Hubel, A.
In vitro culture characteristics of corneal epithelial,
endothelial, and keratocyte cells in a native collagen
matrix. Tissue Eng. 2000, 6, 307–319. 62. Ber, S.; Torun
Kose, G.; Hasirci, V. Bone tissue engineering on patterned
collagen films:an in vitro study. Biomaterials 2005, 26,
1977–1986. 63. Valluzzi, R.; Winkler, S.; Wilson, D.;
Kaplan, D.L. Silk: Molecular organization and control of
assembly. Phil. Trans. R. Soc. Lond. B 2002, 357, 165–167.
64. Altman, G.H.; Diaz, F.; Jakuba, C.; Calabro, T.; Horan,
R.L.; Chen, J.; Lu, H.; Richmond, J.; Kaplan, D.L.
Silk-based biomaterials. Biomaterials 2003, 24, 401–416.
65. Minoura, N.; Aiba, S.; Gotoh, Y.; Tsukada, M.; Imai, Y.
Attachment and growth of cultured fibroblast cells on silk
protein matrices. J. Biomed. Mater. Res. 1995, 29,
1215–1221. 66. Sofia, S.; McCarthy, M.B.; Gronowicz, G.;
Kaplan, D.L. Functionalized silk-based biomaterials for
bone formation. J. Biomed. Mater. Res. 2001, 54, 139–148.
67. Unger, R.E.; Wolf, M.; Peters, K.; Motta, A.;
Migliaresi, C.; Kirkpatrick, C.J. Growth of human cells on
a non-woven silk fibroin net: a potential for use in tissue
engineering. Biomaterials 2004, 25, 1069–1075. 68. Walton,
A.G.; Blackwell, J. Fibrous proteins and biopolymer models.
In Biopolymers; Academic Press Inc.: New York, USA. 1973;
p. 456. 69. Ye, Q.; Zund, G.; Benedikt, P.; Jockenhoevel,
S.; Hoerstrup, S.P.; Sakyama, S.; Hubbell, J.A.; Turina, M.
Fibrin gel as a three dimensional matrix in cardiovascular
tissue engineering. Eur. J. Cardiothorac. Surg. 2000, 17,
587–591. 70. Pellegrini, G.; Ranno, R.; Stracuzzi, G.;
Bondanza, S.; Guerra, L.; Zambruno, G.; Micali, G.; De
Luca, M. The control of epidermal stem cells (holoclones)
in the treatment of massive full-thickness burns with
autologous keratinocytes cultured on fibrin. Transplantation
1999, 68 (6), 868–879. 71. Suh, J.K.F.; Matthew, H.W.T.
Application of chitosan-based polysaccharide biomaterials
in cartilage tissue engineering: a review. Biomaterials
2000, 21, 2589–2598. 72. Madihally, S.V.; Matthew, H.W.
Porous chitosan scaffolds for tissue engineering.
Biomaterials 1999, 20 (12), 1133–1142. 73. Tuzlakoglu, K.;
Catarina, M.; Alves, C.M.; Mano, J.F.; Reis, R.L.
Production and characterization of chitosan fibers and 3-D
fiber mesh scaffolds for tissue engineering applications.
Macromol. Biosci. 2004, 4, 811–819. 74. Zhao, F.; Yin, Y.;
Lu, W.W.; Leong, J.C.; Zhang, W.; Zhang, J.; Zhang, M.;
Yao, K. Preparation and histological evaluation of
biomimetic threeP dimensional
hydroxyapatite/chitosan-gelatin network composite
scaffolds. Biomaterials 2002, 23, 3227– 3234.

75. Zhu, H.; Ji, J.; Lin, R.; Gao, C.; Feng, L.; Shen, J.
Surface engineering of poly( D,L-lactic acid) by entrapment
of chitosan-based derivatives for the promotion of
chondrogenesis. J. Biomed. Mater. Res. 2002, 62, 532–539.

76. Zavan, B.; Brun, P.; Vindigni, V.; Amadori, A.;


Habeler, W.; Pontisso, P.; Montemurro, D.; Abatangelo, G.;
Cortivo, R. Extracellular matrixenriched polymeric
scaffolds as a substrate for hepatocyte cultures: in vitro
and in vivo studies. Biomaterials 2005, 26 (34), 7038–7045.

77. Aigner, J.; Tegeler, J.; Hutzler, P.; Campoccia, D.;


Pavesio, A.; Hammer, C.; Kastenbauer, E.; Naumann, A.
Cartilage tissue engineering with novel nonwoven structured
biomaterial based on hyaluronic acid benzyl ester. J.
Biomed. Mater. Res. 1998, 42, 172–181.

78. Kolybaba, M.; Tabil, L.G.; Panigrahi, S.; Crerar, W.J.;


Powell, T.; Wang, B. Biodegradable polymers: past, present,
and future ASAE/CSAE North Central Intersectional Meeting,
Fargo, North Dakota, October 3–4, 2003 ASAE Paper No.
RRV03-0007.
79. Jopski, T. Biodegradable plastics. Starch 1993, 83
(10), 17–20.

80. Salgado, A.J.; Gomes, M.E.; Chou, A.; Coutinho, O.P.;


Reis, R.L.; Hutmacher, D.W. Preliminary study on the
adhesion and proliferation of human osteoblasts on
starch-based scaffolds. Mater. Sci. Eng. C 2002, 20, 27–33.

81. Li, Z.; Ramay, H.R.; Hauch, K.D.; Xiao, D.; Zhang, M.
Chitosan–alginate hybrid scaffolds for bone tissue
engineering. Biomaterials 2005, 26, 3919–3928.

82. Williams, S.F.; Martin, D.P.; Horowitz, D.M.; People,


O.P. PHA applications: Addressing the price performance
issue I. Tissue engineering. Int. J. Biol. Macromol. 1999,
25, 111–121.

83. Zinn, M.; Witholt, B.; Egli, T. Occurrence, synthesis


and medical application of bacterial polyhydroxyalkanoate.
Adv. Drug Deliv. Rev. 2001, 53, 5–21.

84. Adams, J.H.; Irving, G.; Koeslag, J.H.; Lochner, J.D.;


Sandell, R.C.; Wilkinson, C. Beta-adrenergic blockade
restores glucose’s antiketogenic activity after exercise in
carbohydrate-depleted athletes. J. Physiol. Lond. 1987,
386, 439–454.

85. Torun Kose, G.; Kenar, H.; Hasirci, N.; Hasirci, V.


Macroporous poly(3-hydroxybutyrate-co-3-hydroxyvalerate)
matrices for bone tissue engineering. Biomaterials 2003, 24
(11), 1949–1958.

86. Torun Kose, G.; Korkusuz, F.; Korkusuz, P.; Purali, N.;
Ozkul, A.; Hasırcı, V. Bone generation on PHBV matrices: An
in vitro study. Biomaterials 2003, 24, 4999–5007.

87. Torun Kose, G.; Korkusuz, F.; Ozkul, A.; Soysal, Y.;
Ozdemir, T.; Yildiz, C.; Hasirci, V. Tissue engineered
cartilage on collagen and PHBV matrices. Biomaterials 2005,
26, 5187–5197.

88. Sun, J.; Wu, J.; Li, H.; Chang, J. Macroporous poly
(3-hydroxybutyrate-co-3-hydroxyvalerate) matrices for
cartilage tissue engineering. Eur. Poly. J. 2005, 41 (10),
2443–2449. 89. Cho, C.S.; Seo, S.J.; Park, I.K.; Kim, S.H.;
Kim, T.H.; Hoshiba, T.; Harada, I.; Akaike, T.
Galactose-carrying polymers as extracellular matrices for
liver tissue engineering. Biomaterials 2006, 27 (4),
576–585. 90. Grzesiak, J.J.; Pierschbacher, M.D.; Amodeo,
M.F.; Malaney, T.I.; Glass, J.R. Enhancement of cell
interactions with collagen/glycosaminoglycan matrices by
RGD derivatization. Biomaterials 1997, 18, 1625–1632. 91.
Santiago, L.Y.; Nowak, R.W.; Peter Rubin, J.; Marra, K.G.
Peptide-surface modification of poly(caprolactone) with
laminin-derived sequences for adiposederived stem cell
applications. Biomaterials 2006, 27 (15), 2962–2969. 92.
He, W.; Ma, Z.; Yong, T.; Teo, W.E.; Ramakrishna, S.
Fabrication of collagen-coated biodegradable polymer
nanofiber mesh and its potential for endothelial cells
growth. Biomaterials 2005, 26 (36), 7606–7615. 93.
Tezcaner, A.; Bugra, K.; Hasirci, V. Retinal pigment
epithelium cell culture on surface modified poly
(hydroxybutyrate-co-hydroxyvalerate) thin films.
Biomaterials 2003, 24, 4573–4583. 94. Roy, I.; Gupta, M.N.
Smart polymeric materials: emerging biochemical
applications. Chem. Biol. 2003, 10, 1161–1171. 95. Gupta,
M.N.; Roy, I. Applied biocatalysis: An overview. Indian J.
Biochem. Biophys. 2002, 39, 220–228. 96. Ganorkar, C.R.;
Liu, F.; Baudy, M.; Kim, S.W. Modulating insulin-release
profile from pH/thermosensitive polymeric beads through
polymer molecular weight. J. Control. Release 1999, 59,
287–298. 97. Shimizu, T.; Yamato, M.; Kikuchi, A.; Okano,
T. Two-dimensional manipulation of cardiac myocyte sheets
utilizing temperature-responsive culture dishes augments
the pulsatile amplitude. Tissue Eng. 2001, 7, 141–151. 98.
Jeong, B.; Kim, S.W.; Bae, Y.H. Thermosensitive sol– gel
reversible hydrogels. Adv. Drug Deliv. Rev. 2002, 54,
37–51. 99. Yoshida, R.; Sakai, Y.; Okano, T.; Sakurai, Y.
Modulating the phase transition temperature and
thermosensitivity in N-isopropylacrylamide copolymer gels.
J. Biomater. Sci. Polym. Ed. 1994, 6, 585–598. 100.
Yoshida, R.; Uchida, K.; Kaneko, Y.; Sakal, K.; Kiruchi,
A.; Sakurai, Y.; Okano, T. Comb-type grafted hydrogels with
rapid deswelling response to temperature changes. Nature
1995, 374, 240–242. 101. Kaneko, Y. Rapid deswelling
response of poly(N-isopropylacrylamide) hydrogels by the
formation of water release channels using poly(ethylene
oxide) graft chains. Macromolecules 1998, 31, 6099–6105.
102. Li, C.; Tang, Y.; Armes, S.P. Synthesis and
characterization of biocompatible thermo-responsive
gelators based on ABA triblock copolymers.
Biomacromolecules 2005, 6, 994–999. 103. Au, A.; Ha, J.;
Polotsky, A.; Krzyminski, K.; Gutowska, A.; Hungerford,
D.S.; Frondoza, C.G. Thermally reversible polymer gel for
chondrocyte culture. J. Biomed. Mater. Res. 2003, 67A,
1310–1319. 104. Cho, J.H.; Kim, S.-H.; Park, K.D.; Jung,
M.C.; Yang, W.I.; Han, S.W.; Noh, J.Y.; Lee, J.W.
Chondrogenic differentiation of human mesenchymal stem
cells using a thermosensitive
poly(N-isopropylacrylamide)and water-soluble chitosan
copolymer. Biomaterials 2004, 25, 5743–5751. 105. Wang,
D.A.; Williams, C.G.; Yang, F.; Cher, N.; Lee, H.;
Elisseeff, J.H. Bioresponsive phosphoester hydrogels for
bone tissue engineering. Tissue Eng. 2005, 11 (1–2),
201–213. 106. Jung, Y.; Kim, S.-S.; Kim, Y.H.; Kim, S.H.;
Kim, B.-S.; Kim, S.; Choi, C.Y.; Kim, S.H. A poly(lactic
acid)/calcium metaphosphate composite for bone tissue
engineering. Biomaterials 2005, 26, 6314–6322. 107. Ma,
P.X.; Zhang, R.; Xiao, G.; Franceschi, R. Engineering new
bone tissue in vitro on highly porous poly(-hydroxyl
acids)/hydroxyapatite composite scaffolds. J. Biomed.
Mater. Res. 2000, 54 (2), 284–293. 108. Alsberg, E.;
Anderson, K.W.; Albeiruti, A.; Franceschi, R.T.; Mooney,
D.J. Cell-interactive alginate hydrogels for bone tissue
engineering. J. Dent. Res. 2001, 80, 2025–2029. 109. Torun
Kose, G.; Korkusuz, F.; Korkusuz, P.; Hasirci, V. In vivo
tissue engineering of bone using poly (3-hydroxybutyric
acid-co-3-hydroxyvaleric acid) and collagen scaffolds.
Tissue Eng. 2004, 10 (7), 1234–1250. 110. Gomes, M.E.;
Sikavitsas, V.I.; Behravesh, E.; Reis, R.L.; Mikos, A.G.
Effect of flow perfusion on the osteogenic differentiation
of bone marrow stromal cells cultured on starch-based
three-dimensional scaffolds. J. Biomed. Mater. Res. 2003,
67A, 87–95. 111. Li, W.J.; Tuli, R.; Okafor, C.; Derfoul,
A.; Danielson, K.G.; Hall, D.J.; Tuan, R.S. A
three-dimensional nano fibrous scaffold for cartilage tissue
engineering using human mesenchymal stem cells.
Biomaterials 2005, 26, 599–609. 112. Chen, G.; Sato, T.;
Ohgushi, H.; Ushida, T.; Tateishi, T.; Tanaka, J. Culturing
of skin fibroblasts in a thin PLGA–collagen hybrid mesh.
Biomaterials 2005, 26, 2559–2566. 113. Besseau, L.;
Coulomb, B.; Lebreton-Decoster, C.; Giraud-Guille, M.M.
Production of ordered collagen matrices for
three-dimensional cell culture. Biomaterials 2002, 23,
27–36. 114. Wen, S.J.; Zhao, L.M.; Li, P.; Li, J.X.; Liu,
Y.; Liu, J.L.; Chen, Y.C. Blood vessel tissue engineering:
Seeding vascular smooth muscle cells and endothelial cells
sequentially on biodegradable scaffold in vitro. Zhonghua
Yi Xue Za Zhi. 2005, 85 (12), 816–818. 115. Dvir-Ginzberg,
M.; Gamlieli-Bonshtein, I.; Agbaria, R.; Cohen, S. Liver
tissue engineering within alginate scaffolds: Effects of
cell-seeding density on hepatocyte viability, morphology,
and function. Tissue Eng. 2003, 9 (4), 757–766. P
Polytetrafluoroethylene

1. http://www.dupont.com/teflon/newsroom/history.html.

2. Allcock, H.R.; Lampe, F.W. Contemporary Polymer


Chemistry, 3rd Ed.; Prentice Hall: Englewood Cliffs, NJ,
1990; 284.

3. http://www.polymerprocessing.com/polymers/PTFE. html.

4. http://www.matweb.com/

5. Bahadur, P.; Sastry, N.V. Principles of Polymer Science;


CRC Press: Boca Raton, FL, 2000.

6. Altman, R.; Chubb, S.; Dorrenstein, G.M.; Quesenberry,


K. Avian Medicine and Surgery; W.B. Saunders, Inc.:
Philadelphia, PA, 1997; 609.

7. Boucher, M.; Ehmler, T.J.; Bermudez, A.J.


Polytetrafluoroethylene gas intoxication in broiler
chickens. Avian Dis. 2000, 44 (2), 449–453. 8. Seidel,
W.C.; Scherer, K.V., Jr; Cline, D., Jr; et al. Chemical,
physical, and toxicological characterization of fumes
produced by heating tetrafluoroethene homopolymer and its
copolymers with hexafluoropropene and perfluoro (propyl vinyl
ether). Chem. Res. Toxicol. 1991, 4 (2), 229–236. 9.
Waritz, R.S. An industrial approach to evaluation of
pyrolysis and combustion hazards. Environ. Health Perspect.
1975, 11, 197–202. 10. Johnston, C.J.; Finkelstein, J.N.;
Mercer, P.; Corson, N.; Gelein, R.; Oberdorster, G.
Pulmonary effects induced by ultrafine PTFE particles.
Toxicol. Appl. Pharmacol. 2000, 168 (3), 208–215. 11.
Nuttall, J.B.; Kelly, R.J.; Smith, B.S.; Whiteside, C.K.
Aerospace Med. 1964, July. 12. Zanen, A.L.; Rietveld, A.P.
Inhalation trauma due to overheating in a microwave oven.
Thorax 1993, 48 (3), 300–302. 13. Allcock, H.R.; Lampe,
F.W. Contemporary Polymer Chemistry, 3rd Ed.; Prentice
Hall: Englewood Cliffs, NJ, 1990; 173. 14.
http://www.epa.gov/opptintr/pfoa/ P
Polyurethane Biomaterials

9. Grapski, J.; Cooper, S.L. Synthesis and characterization


of non-leaching biocidal polyurethanes. Biomaterials 2001,
22, 2239–2246.

10. Jayabalan, M.; Lizymol, P.P.; Thomas, V. Synthesis of


hydrolytically stable low elastic modulus polyurethaneurea
for biomedical applications. Polym. Int. 2000, 49 (1),
88–92.

11. Labow, R.S.; Erfle, D.; Santerre, J.P.


Neutrophilmediated degradation of segmented polyurethanes.
Biomaterials 1995, 16, 51–59.

12. Ratner, B.D.; Gladhill, K.W.; Horbett, T.A. Analysis of


in vitro enzymatic and oxidative degradation of
polyurethanes. J. Biomed. Mat. Res. 1988, 22, 509–527.

13. Tang, Y.; Labow, R.S.; Santerre, J.P. Enzyme-induced


biodegradation of polycarbonate polyurethanes: dependence
of hard-segment concentration. J. Biomed. Mat. Res. 2001,
56, 516–528.

14. Gorna, K.; Polowinski, S.; Gogolewski, S. Synthesis and


characterization of biodegradable poly(caprolactone
urethane)s. I. Effect of the polyol molecular weight,
catalyst and chain extender on the molecular and physical
characteristics. J. Polym. Sci. A Polym. Chem. 2001, 40,
156–170.

15. Guan, J.J.; Fujimoto, K.L.; Sacks, M.S.; Wagner, W.R.


Preparation and characterization of highly porous,
biodegradable polyurethane scaffolds for soft tissue
applications. Biomaterials 2005, 26 (18), 3961–3971.

16. Guan, J.J.; Sacks, M.S.; Beckman, E.J.; Wagner, W.R.


Biodegradable poly(ether ester urethane)urea elastomers
based on poly(ether ester) triblock copolymers and
putrescine: synthesis, characterization and
cytocompatibility. Biomaterials 2004, 25 (1), 85–96.

17. Fromstein, J.D.; Woodhouse, K.A. Elastomeric


biodegradable polyurethane blends for soft tissue
applications. J. Biomat. Sci. Polym. Ed. 2002, 13 (4),
391–406.

18. Elliott, S.L.; Fromstein, J.D.; Santerre, J.P.;


Woodhouse, K.A. Identification of biodegradation products
formed by L-phenylalanine based segmented
polyurethaneureas. J. Biomat. Sci. Polym. Ed. 2002, 13 (6),
691–711.

19. Zhang, J.Y.; Doll, B.A.; Beckman, E.J.; Hollinger, J.O.


A biodegradable polyurethane-ascorbic acid scaffold for
bone tissue engineering. J. Biomed. Mat. Res. A 2003, 67A
(2), 389–400.

20. Covolan, V.L.; Di Ponzio, R.; Chiellini, F.; Fernandes,


E.G.; Solaro, R.; Chiellini, E. Polyurethane based
materials for the production of biomedical materials.
Macromol. Symp. 2004, 218, 273–282.

21. Petrini, P.; Fare, S.; Piva, A.; Tanzi, M.C. Design,
synthesis and properties of polyurethane hydrogels for
tissue engineering. J. Mat. Sci. Mat. Med. 2003, 14 (8),
683–686.

22. Kidoaki, S.; Kwon, I.K.; Matsuda, T. Mesoscopic spatial


designs of nanoand microfiber meshes for tissue-engineering
matrix and scaffold based on newly devised multilayering
and mixing electrospinning techniques. Biomaterials 2005,
26, 37–46. 23. Sanders, J.E.; Sanders, J.E.; Lamont, S.E.;
Karchin, A.; Golledge, S.L.; Ratner, B.D. Fibro-porous
meshes made from polyurethane micro-fibers: effects of
surface charge on tissue response. Biomaterials 2005, 26
(7), 813–818. 24. Stankus, J.J.; Guan, J.J.; Wagner, W.R.
Fabrication of biodegradable elastomeric scaffolds with
sub-micron morphologies. J. Biomed. Mat. Res. A 2004, 70A
(4), 603–614. 25. Takahashi, A.; Kita, R.; Kaibara, M.
Effects of thermal annealing of segmented polyurethanes on
surface properties, structure and antithrombogenicity. J.
Mat. Sci. Mat. Med. 2002, 13, 259–264. 26. Woo, G.;
Mittleman, M.; Santerre, J.P. Synthesis and
characterization of a novel biodegradable antimicrobial
polymer. Biomaterials 2000, 21, 1235–1246. 27. Jun, H.-K.;
West, J.L. Modification of polyurethaneurea with PEG and
YIGSR peptide to enhance endothelialization without
platelet adhesion. J. Biomed. Mat. Res. B Appl. Biomat.
2005, 72B, 131–139. 28. Ho, J.Y.; Matsuura, T.; Santerre,
J.P. The effect of fluorinated surface modifying
macromolecules on the surface morphology of
polyethersulfone membranes. J. Biomat. Sci. Polym. Ed.
2000, 11 (10), 1085–1104. 29. Jahangir, A.R.; McClung,
W.G.; Cornelius, R.M.; McCloskey, C.B.; Brash, J.L.;
Santerre, J.P. Fluorinated surface-modifying
macromolecules: modulating adhesive protein and platelet
interactions on a polyether-urethane. J. Biomed. Mat. Res.
2002, 60 (1), 135–147. 30. Jahangir, R.; McCloskey, C.B.;
McClung, W.G.; Labow, R.S.; Brash, J.L.; Santerre, J.P. The
influence of protein adsorption and surface modifying
macromolecules on the hydrolytic degradation of a poly
(ether-urethane) by cholesterol esterase. Biomaterials
2003, 24 (1), 121–130. 31. Ward, R.S.; Tian, Y.; White,
K.A. Improved polymer biostability via oligomeric end
groups incorporated during synthesis. Abstr. Pap. Am. Chem.
Soc. 1998, 216, U907–U907. 32. Zhu, Y.B.; Gao, C.Y.; He,
T.; Shen, J.C. Endothelium regeneration on luminal surface
of polyurethane vascular scaffold modified with diamine and
covalently grafted with gelatin. Biomaterials 2004, 25 (3),
423–430. 33. Wang, D.A.; Feng, L.X.; Ji, J.; Sun, Y.H.;
Zheng, X.X.; Elisseeff, J.H. Novel human endothelial
cell-engineered polyurethane biomaterials for
cardiovascular biomedical applications. J. Biomed. Mat.
Res. A 2003, 65A (4), 498–510. 34. van Wachem, P.B.;
Hendricks, M.; Blaauw, E.H.; Dijk, F.; Verhoeven, M.;
Cahalan, P.T.; van Luyn, M.J.A. Electron microscopic
observations on tissue integration of collagen-immobilized
polyurethane. Biomaterials 2002, 23 (6), 1401–1409. 35.
Gorman, S.P.; Tunney, M.M.; Keane, P.F.; VanBladel, K.;
Bley, B. Characterization and assessment of a novel
poly(ethylene oxide)/polyurethane composite hydrogel
(Aquavene s ) as a ureteral stent biomaterial. J. Biomed.
Mat. Res. 1998, 39, 642–649.
Positron Emission Tomography (PET)

1. Sweet, W.H. The use of nuclear disintegration in


diagnosis and treatment of brain tumors. N. Engl. J. Med.
1951, 245, 875–878.

2. Brownell, G.L.; Sweet, W.H. Localization of brain tumors


with positron emitters. Nucleonics 1953, 11, 40–45.

3. Chesler, D.D. Three-dimensional activity distribution


from multiple positron scintigraphs. J. Nucl. Med. 1971,
12, 347–348.

4. Chesler, D.A. Positron Emission Tomography and


Three-Dimensional Reconstruction Technique. In Tomographic
Imaging in Nuclear Medicine; Freedman, G.S., Ed.; The
Society of Nuclear Medicine: New York, 1973; 176–183.

5. Ambrose, J. Computerized transverse axial scanning


(tomography): Part II. Clinical application. Br. J. Radiol.
1973, 46, 1023–1047.

6. Hounsfield, G.N. Computerized transverse axial scanning


(tomography). Part 1: Description of system. Part II:
Clinical applications. Br. J. Radiol. 1973, 46, 1016–1022.

7. Cormack, A.M. Representation of a function by its line


integrals, with some radiological applications. J. Appl.
Phys. 1973, 34, 2722–2727.

8. Cormack, A.M. Reconstruction densities from their


projections, with applications in radiological physics.
Phys. Med. Biol. 1973, 18, 195–207.

9. Cherry, S.R.; Shao, Y.; Silverman, R.W.; Chatziioannou,


A.; Meadows, K.; Siegel, S.; Farguhar, T.; Young, J.;
Jones, W.F.; Newport, D.; Moyers, C.; Andreaco, M.; Paulus,
M.; Binkley, D.; Nutt, R.; Phelps, M.E. MicroPET: A high
resolution PET scanner for imaging small animals. IEEE
Trans. Nucl. Sci. 1997, 44, 1161–1166.

10. Correia, J.; Burnham, C.A.; Kaufman, D.; Fischman, A.


Development of a small animal PET imaging device with
resolution approaching 1mm. IEEE Trans. Nucl. Sci. 1999, 46
(3), 631–635.

11. Melcher, C.L. Lutetium Orthosilicate Single Crystal


Scintillator Detector. US Patent 4,958,080, 1990. 12.
www.itnonline.net. ‘‘Product Comparison Chart’’ or
www.reillycomm.com/cgi-bin/proj/compare2.cgi. 13. Burnham,
C.A.; Bradshaw, J.; Kaufman, D.; Chesler, D.A.; Brownell,
G.L. Positron source position sensing detector and
electronics. US Patent 4,531,058, July 23, 1985. 14. Casey,
M.; Nutt, R. A multislice two-dimensional BGO detector
system for PET. IEEE Trans. Nucl. Sci. 1986, 33, 760–763.
15. Brownell, G.L.; Burnham, C.A.; Chesler, D.A. High
Resolution Tomograph Using Analog Coding. In The Metabolism
of the Human Brain Studies with Positron Emission
Tomography; Greitz, T., Ingvar, D.H., Widen, L., Eds.;
Raven Press: New York, 1985; 13–19. 16. Schmand, M.;
Dahlbom, M.; Eriksson, L.; Casey, M.E.; Andreaco, M.S.;
Vagneur, K.; Phelps, M.E.; Nutt, R. Performance of a
LSO/NaI(Tl) phoswitch detector for a combined PET/SPECT
imaging system. J. Nucl. Med. 1998, 39 (5), 9. 17.
Brownell, G.L. US Atomic Energy Commission 1971.
(Inspection document). 18. Brownell, G.L.; Burnham, C.A.
MGH Positron Camera. In Tomographic Imaging in Nuclear
Medicine; Freedman, G.S., Ed.; The Society of Nuclear
Medicine: New York, 1973; 154–164. 19. Chesler, D.A.; Hoop,
B., Jr.; Brownell, G.L. Transverse section imaging of
myocardium with 13NH4. J. Nucl. Med. 1973, 14, 623. 20.
Robertson, J.S.; Marr, R.B.; Rosenblum, M.; Radeka, V.;
Yamamoto, Y.L. 32-Crystal Positron Transverse Section
Detector. In Tomographic Imaging in Nuclear Medicine;
Freedman, G.S., Ed.; The Society of Nuclear Medicine: New
York, 1973. 21. Cho, Z.H.; Chan, J.K.; Eriksson, L.
Circular ring transverse axial positron camera for
3-dimensional reconstruction of radionuclide distribution.
IEEE Trans. Nucl. Sci. 1976, 23, 613–623. 22. Derenzo,
S.E.; Budinger, T.F.; Cahoon, J.L.; Greenberg, W.L.;
Huesman, R.; Vuletich, T. The Donner 280-crystal high
resolution positron tomograph. IEEE Trans. Nucl. Sci. 1979,
26, 2790–2793. 23. Ter-Pogossian, M.M.; Phelps, M.E.;
Hoffman, E.J. A positron emission transaxial tomograph for
nuclear medicine imaging (PETT). Radiology 1975, 114,
89–98. 24. Brownell, G.; Burnham, CA.; Stearns, CW.;
Chesler, D.A.; Brownell, A.-L.; Palmer, M. Development in
highresolution positron emission tomography at MGH. Int. J.
Imaging Syst. Technol. 1989, 1, 207–217. 25. NEMA.
Performance Measurements of Positron Emission Tomographs;
National Electrical Manufacturers Association: Rosslyn, VA,
2001. 26. Hoffman, E.; Huang, S.C.; Phelps, M.E.
Quantitation in positron emission tomography. 1—Effect of
object size. J. Comput. Assist. Tomogr. 1979, 3, 299–308.
27. Ficke, D.C.; Beecher, D.E.; Bergmann, S.R.; Hoffman,
G.R.; Hood, J.T.; Ter-Pogossian, M.M. In Performance
Characterization of a Whole Body PET System Designed for
Dynamic Cardiac Imaging, IEEE Nuclear Science Symposium and
Medical Imaging Conference, Santa Fe, NM, 1991. 28. Bohm,
C.; Erikssson, L.; Bergstrom, M.; Litton, J.; Sundman, R.;
Singh, M. A computer assisted ringdetector positron camera
system for reconstruction tomography of the brain. IEEE
Trans. Nucl. Sci. 1978, 25, 624–637. 29. Bendriem, B.;
Soussaline, F.; Campagnolo, R.; Verrey, B.; Wajnberg, P.;
Syrota, A. A technique for correction of scattered
radiation in a PET system using timeof-flight information.
J. Comput. Assist. Tomogr. 1986, 10, 287–295. 30.
Bergstrom, M.; Eriksson, L.; Bohm, C.; Blomqvist, G.;
Litton, J. Correction for scattered radiation in a ring
detector positron camera by integral transformation of the
projections. J. Comput. Assist. Tomogr. 1983, 7, 42–50. 31.
Oda, K.; Toyama, H.; Uemura, K.; Ikoma, Y.; Kimura, Y.;
Senda, M. Comparison of parametric FBP and OS-EM
reconstruction algorithm images for PET dynamic study. Ann.
Nucl. Med. 2001, 15, 417–423. 32. Subramanyam, R.; Alpert,
N.M.; Hoop, B., Jr.; Brownell, G.L.; Taveras, J.M. A model
for regional cerebral oxygen distribution during continuous
inhalation of 15O2, C15O, and C15O2. J. Nucl. Med. 1978,
19, 43–53. 33. Phelps, M.E.; Huang, S.C.; Hoffman, E.J.;
Selin, C.; Sokoloff, L.; Kuhl, D. Tomographic measurement
of local cerebral glucose metabolic rate in humans with
(F-18-2-fluoro-2-deoxyD-glucose: Validation of method. Ann.
Neurol. 1979, 6, 371–388. 34. Farde, L.; Hall, H.; Ehrin,
E.; Sedvall, G. Quantitative analysis of D2 dopamine
receptor binding in the living human brain by PET. Science
1986, 231, 258–260. 35. Delforges, J.; Loc’h, C.; Hantraye,
P.; Stulzaft, O.; Khalili-Varasteh, M.; Maziere, M.;
Syrota, A.; Maziere, B. Kinetic analysis of central
(76Br)bromolisuride binding to dopamine D2-receptor studies
by PET. J. Cereb. Blood Flow Metab. 1991, 11, 914–925. 36.
Gjedde, A.; Wong, D.F. Modeling Neuroreceptor Binding of
Radioligands in vivo. In Quantitative Imaging:
Neuroreceptors Neurotransmitters and Enzymes; Frost, J.,
Wagner, H.W., Jr., Eds.; Raven Press: New York, 1990;
51–79. 37. Jones, T.; Chesler, DA.; Ter-Pogossian, M.M. The
continuous inhalation of oxygen-15 for assessing regional
oxygen extraction fraction in the brain of man. Br. J.
Radiol. 1976, 49, 339–343. 38. Lenzi, G.I.; Frackowiak,
R.S.J.; Jones, T. Cerebral oxygen metabolism and blood flow
in human cerebral ischemic infarction. J. Cereb. Blood Flow
Metab. 1982, 2, 321–335. 39. Rubin, G.; Firlik, A.D.; Levy,
E.I.; Pindzola, R.R.; Yonas, H. Relationship between
cerebral blood flow and clinical outcome in acute stroke.
Cerebovasc. Disord. 2000, 10 (4), 298–306. 40. Brooks, D.J.
Cerebral blood flow activation studies in Parkinson’s
disease. Parkinson’s Dis. Adv. Neurol. 2001, 86, 225–235.
41. Akutsu, Y.; Harumi, K.; Michihata, T.; Watanabe, T.;
Yamanaka, H.; Okazaki, O.; Kashida, M.; Hasegawa, M.;
Katagiri, T. Correlations between resting regional wall
motion and regional myocardial blood flow (at rest and
during exercise) in infarct-related myocardium—A study with
[13N]ammonia positron emission tomography. Jpn. Circ. J.
1997, 61, 665–672. 42. Di Chiro, G.; DeLaPaz, R.L.; Brooks,
R.A.; Sokoloff, L.; Kornblith, P.L.; Smith, B.H.; Patronas,
N.J.; Kufta, C.V.; Kessler, R.M.; Johnston, G.S.; Manning,
R.G.; Wolf, A.P. Glucose utilization of cerebral gliomas
measured by [18F]fluorodeoxyglucose and positron emission
tomography. Neurology 1982, 32 (12), 1323–1329. 43. Di
Chiro, G. Which PET radiopharmaceutical for brain tumors?
J. Nucl. Med. 1991, 32 (7), 1346–1348. 44. Frackowiak, R.;
Lenzi, G.L.; Jones, T.; Heather, J.D. Quantitative
measurement of regional cerebral blood flow and oxygen
metabolism in man using 15O and positron emission
tomography: Theory, procedure and normal values. J. Comput.
Assist. Tomogr. 1980, 4, 727–736. 45. Ericson, K.; Lilja,
A.; Bergstrom, M.; Collins, V.P.; Eriksson, L.; Ehrin, E.;
von Holst, H.; Lundqvist, H.; Langstrom, B.; Mosskin, M.
Positron emission tomography with 11C-methyl-L-methionine,
11CD-glucose and 68Ga-EDTA in supratentorial tumors. J.
Comput. Assist. Tomogr. 1985, 9, 683–689. 46. Geltman, E.M.
Assessment of myocardial fatty acid metabolism with
1-11C-palmitate. J. Nucl. Cardiol. 1994, 1 (Supp1), 15–22.
47. Taylor, M.; Wallhaus, T.R.; Degrado, T.R.; Russell,
D.C.; Stanko, P.; Nickles, R.J.; Stone, C.K. An evaluation
of myocardial fatty acid and glucose uptake using PET with
18F-fluoro-6-thia-heptadecanoic acid. J. Nucl. Med. 2001,
42, 55–62. 48. Frost, J.J.; Rosier, A.J.; Reich, S.G.;
Smith, J.S.; Ehlers, M.D.; Snyder, S.H.; Ravert, H.T.;
Dannals, R.F. Positron emission tomographic imaging of the
dopamine transporter with 11C-WIN 35,428 reveals marked
decline in mild Parkinson’s disease. Ann. Neurol. 1993, 34,
423–431. 49. Hantraye, P.; Brownell, A.-L.; Elmaleh, D.R.;
Spealman, R.D.; Wullner, U.; Brownell, G.L.; Isacson, O.
Dopamine fiber detection by [11C]-CFT and PET in a primate
model of parkinsonism. NeuroReport 1992, 3, 265–268. 50.
Weeks, R.A.; Piccini, P.; Harding, A.E.; Brooks, D.J.
Striatal D1 and D2 dopamine receptor loss in asymptomatic
mutation carriers of Huntington’s disease. Ann. Neurol.
1996, 40 (1), 49–54. 51. Juhasz, C.; Nagy, F.; Watson, C.;
da Silva, E.A.; Muzik, O.; Chugani, D.C.; Shah, J.;
Chugani, HT. Glucose and [11C]flumazenil positron emission
tomography abnormalities of thalamic nuclei in temporal
lobe epilepsy. Neurology 1999, 53, 2037–2045. 52. Martinez,
D.; Hwang, D.; Mawlawi, O.; Slifstein, M.; Kent, J.;
Simpson, N.; Parsey, R.V.; Hashimoto, T.; Huang, Y.; Shinn,
A.; Van Heertum, R.; Abi-Dargham, A.; Caltabiano, S.;
Malizia, A.; Cowley, H.; Mann, J.J.; Laruelle, M.
Differential occupancy of somatodendritic and postsynaptic
5HT(1A) receptors by pindolol: A dose-occupancy study with
[11C]WAY 100635 and positron emission tomography in humans.
Neuropsychopharmacology 2001, 24 (3), 209–229. 53. Jones,
A.; Kitchen, N.D.; Watabe, H.; Cunningham, V.J.; Jones, T.;
Luthra, S.K.; Thomas, D.G. Measurement of P changes in
opioid receptor binding in vivo during trigeminal neuralgic
pain using [11C] diprenorphine and positron emission
tomography. J. Cereb. Blood Flow Metab. 1999, 19, 803–808.

54. Saji, H.; Tsutsumi, D.; Magata, Y.; Iida, Y.; Konishi,
J.; Yokoyama, A. Preparation and biodistribution in mice of
[11C]carfentanil: A radiopharmaceutical for studying brain
mu-opioid receptors by positron emission tomography. Ann.
Nucl. Med. 1992, 6, 63–67.

55. Mortimer, J.E.; Dehdashti, F.; Siegel, B.A.;


Katzenellenbogen, J.A.; Fracasso, P.; Welch, M.J. Positron
emission tomography with 2-[18F]fluoro-2-deoxyD-glucose and
16alpha-[18F]fluoro-17beta-estradiol in breast cancer:
Correlation with estrogen receptor status and response to
systemic therapy. Clin. Cancer Res. 1996, 2, 933–939.

56. Shields, A.F.; Grierson, J.R.; Dohmen, B.M.; Machulla,


H.J.; Stayanoff, J.C.; Lawhorn-Crews, J.M.; Obradovich,
J.E.; Muzik, O.; Mangner, T.J. Imaging proliferation in
vivo with [F-18]FLT and positron emission tomography. Nat.
Med. 1998, 4 (11), 1334–1336.

57. Wells, P.; Gunn, R.N.; Alison, M.; Steel, C.; Golding,
M.; Ranicar, A.S.; Brady, F.; Osman, S.; Jones, T.; Price,
P. Assessment of proliferation in vivo using
2-[(11)C]thymidine positron emission tomography in advanced
intra-abdominal malignancies. Cancer Res. 2002, 62 (20),
5698–5702.

58. Tjuvajev, J.; Chen, S.H.; Joshi, A.; Joshi, R.; Guo,
Z.S.; Balatoni, J.; Ballon, D.; Koutcher, J.; Finn, R.;
Woo, S.L.; Blasberg, R.G. Imaging adenoviral-mediated
herpes virus thymidine kinase gene transfer and expression
in vivo. Cancer Res. 1999, 59, 5186–5193.

59. Gambhir, S.; Barrio, J.R.; Herschman, H.R.; Phelps,


M.E. Assays for noninvasive imaging of reporter gene
expression. J. Nucl. Med. 1999, 26, 481–490.

60. Gambhir, S.; Barrio, J.R.; Wu, L.; Iyer, M.; Namavari,
M.; Satyamurthy, N.; Bauer, E.; Parrish, C.; MacLaren,
D.C.; Borghei, A.R.; Green, L.A.; Sharfstein, S.; Berk,
A.J.; Cherry, S.R.; Phelps, M.E.; Herschman, H. Imaging of
adenoviral-directed herpes simplex virus type 1 thymidine
kinase reporter gene expression in mice with radiolabeled
ganciclovir. J. Nucl. Med. 1998, 39, 2003–2011.
61. Schellinger, P.D.; Jansen, O.; Fiebach, J.B.; Pohlers,
O.; Ryssel, H.; Heiland, S.; Steiner, T.; Hacke, W.;
Sartor, K. Feasibility and practicality of MR imaging of
stroke in the management of hyperacute cerebral ischemia.
Am. J. Neuroradiol. 2000, 21 (7), 1184–1189.

62. Mintun, M.A.; Lundstrom, B.N.; Snyder, A.Z.; Vlassenko,


A.G.; Shulman, G.l.; Raichle, M.E. Blood flow and oxygen
delivery to human brain during functional activity:
Theoretical modeling and experimental data. Proc. Natl.
Acad. Sci. U. S. A. 2001, 98, 6859–6864.

63. Ito, H.; Takahashi, K.; Hatazawa, J.; Kim, S.G.; Kanno,
I. Changes in human regional cerebral blood flow and
cerebral blood volume during visual stimulation measured by
positron emission tomography. J. Cereb. Blood Flow Metab.
2001, 21, 608–612.

64. Fukuda, M.; Mentis, M.; Ghilardi, M.F.; Dhawan, V.;


Antonini, A.; Hammerstad, J.; Lozano, A.M.; Lang, A.;
Lyons, K.; Koller, W.; Ghez, C.; Eidelberg, D. Functional
correlates of pallidal stimulation for Parkinson’s disease.
Ann. Neurol. 2001, 49, 155–164. 65. Marshall, R.C.;
Tillisch, J.H.; Phelps, M.E.; Huang, S.C.; Carson, R.;
Henze, E.; Schelbert, H.R. Identification and
differentiation of resting myocardial ischemia and
infarction in man with positron computed tomography,
18F-labeled fluorodeoxyglucose and N-13 ammonia. Circulation
1983, 67 (4), 766–778. 66. Kitsiou, A.N.; Bacharach, S.L.;
Bartlett, M.L.; Srinivasan, G.; Summers, R.M.; Quyyumi,
A.A.; Dilsizian, V. 13N-ammonia myocardial blood flow and
uptake: Relation to functional outcome of asynergic regions
after revascularization. J. Am. Coll. Cardiol. 1999, 33,
678–686. 67. Anderson, H.; Yap, J.; Price, P.M. Measurement
of tumour and normal tissue (NT) perfusion by positron
emission tomography (PET) in the evaluation of antivascular
therapy: Results in the phase I study of Combretastatin A4
Phosphate (CA4P). Proc. Ann. Meet. Am. Soc. Clin Oncol.
2000, 9, 179a. 68. Herbst, R.S.; Mullani, N.A.; Davis,
D.W.; Hess, K.R.; McConkey, D.J.; Charnsangavej, C.;
O’Reilly, M.S.; Kim, H.W.; Baker, C.; Roach, J.; Ellis,
L.M.; Rashid, A.; Pluda, J.; Bucana, C.; Madden, T.L.;
Tran, H.T.; Abbruzzese, J.L. Development of biologic
markers of response and assessment of antiangiogenic
activity in a clinical trial of human recombinant
endostatin. J. Clin. Oncol. 2002, 20, 3804–3814. 69.
Sokoloff, L.; Reivich, M.; Kennedy, C.; Des Rosiers, M.H.;
Patlak, C.S.; Pettigrew, K.D.; Sakurada, O. The (C-14)
deoxy glucose method for the measurement of local cerebral
glucose utilization: Theory, procedure, the normal values
in the conscious and anesthetized albino rat.
Neurochemistry 1977, 28, 897–916. 70. Ratib, O.; Phelps,
M.E.; Huang, S.C.; Henze, E.; Selin, C.E.; Schelbert, H.R.
Positron tomography with deoxyglucose for estimating local
myocardial glucose metabolism. J. Nucl. Med. 1982, 23,
577–586. 71. Kapoor, R.; Spence, A.M.; Muzi, M.; Graham,
M.M.; Abbott, G.L.; Krohn, K. Determination of the
deoxyglucose and glucose phosphorylation and the lumped
constant in rat brain and a transplantable rat glioma. J.
Neurochem. 1989, 53, 37–44. 72. Bokenmeyer, C.;
Kollmannsberger, C.; Oechsle, K.; Dohmen, B.M.;
Pfannenberg, A.; Claussen, C.D.; Bares, R.; Kanz, L. Early
prediction of treatment response to high-dose salvage
chemotherapy in patients with relapsed germ cell cancer
using [18F]FDG-PET. Br. J. Cancer 2002, 86, 506–511. 73.
Brock, C.S.; Young, H.; O’Reilly, S.M.; Matthews, J.;
Osman, S.; Evans, H.; Newlands, E.S.; Price, P.M. Early
evaluation of tumour metabolic response using
[18F]flurodeoxyglucose and positron emission tomography: A
pilot study following the phase II chemotherapy schedule
for temozolomide in recurrent high-grade gliomas. Br. J.
Cancer 2000, 82 (3), 608–615. 74. Benabid, A.L.; Pollak,
P.; Gao, D.; Hoffman, D.; Limousin, P.; Gay, E. Chronic
electrical stimulation of the ventralis intermedius nucleus
of the thalamus as a treatment of movement disorders. J.
Neurosurg. 1996, 84, 203–214. 75. Lindvall, O.; Sawle, G.;
Widner, H.; Rothwell, J.C.; Bjorklund, A.; Brooks, D.;
Brundin, P.; Frackowiak, R.S.; Marsden, C.D.; Odin, P.;
Rehncrona, S. Evidence of long-term survival and function
of dopaminergic grafts in progressive Parkinson’s disease.
Ann. Neurol. 1994, 32, 172–180. 76. Frankle, W.G.;
Laruelle, M. Neuroreceptor imaging in psychiatric
disorders. Ann. Nucl. Med. 2002, 16, 437–446. 77. Volkow,
N.D.; Fowler, J.S.; Wang, G.J. Role of dopamine in drug
reinforcement and addiction in humans: Results from imaging
studies. Behav. Pharmacol. 2002, 13, 355–366. 78.
Dougherty, D.D.; Shin, L.M.; Alpert, N.M.; Pitman, R.K.;
Orr, S.P.; Lasko, M.; Macklin, M.L.; Fischman, A.J.; Rauch,
S.L. Anger in healthy men: A PET study using script-driven
imagery. Biol. Psychiatry 1999, 46, 466–472. 79. Volkow,
N.D.; Fowler, J.S.; Wang, G.; Ding, Y.; Gatley, S.J.
Mechanism of action of methylphenidate: Insights from PET
imaging studies. J. Atten. Disord. 2002, 6 (Suppl. 1),
31–43. 80. Dehdashti, F.; Mortimer, J.E.; Siegel, B.A.;
Griffeth, L.K.; Bonasera, T.J.; Fusselman, M.J.; Detert,
D.D.; Cutler, P.D.; Katzenellenbogen, J.A.; Welch, M.J.
Positron tomographic assessment of estrogen receptors in
breast cancer: Comparison with FDG-PET and in vitro
receptor assays. J. Nucl. Med. 1995, 36, 1766–1774. 81.
Dehdashti, F.; Flanagan, F.L.; Mortimer’, J.E.;
Katzenelenbogen, J.A.; Welch, M.J.; Siegel, B.A. Positron
emission tomographic assessment of ‘‘metabolic flare’’ to
predict response of metastatic breast cancer to
antiestrogen therapy. Eur. J. Nucl. Med. 1999, 26, 51–56.
82. Young, H.; Baum, R.; Cremerius, U.; Herholz, K.;
Hoekstra, O.; Lammertsma, A.A.; Pruim, J.; Price, P.
Measurement of clinical and subclinical tumour response
using [18F]-fluorodeoxyglucose and positron emission
tomography: Review and 1999 EORTC recommendations. European
Organization for Research and Treatment of Cancer (EORTC)
PET Study Group. Eur. J. Cancer 1999, 35, 1773–1782. 83.
Mintun, M.A.; Welch, M.J.; Siegel, B.A.; Mathias, C.J.;
Brodack, J.W.; McGuire, A.H.; Katzenellenbogen, J.A. Breast
cancer: PET imaging of estrogen receptors. Radiology 1988,
169, 45–48. 84. Gupta, N.; Price, P.M.; Aboagye, E.O. PET
for in vivo pharmacokinetic and pharmacodynamic
measurements. Eur. J. Cancer 2002, 38, 2094–2107. P
Protein Adsorption Modeling

1. Branden, C.; Tooze, J. Introduction To Protein


Structure, 1st Ed.; Garland: New York, 1991; 1–410.

2. Cleland, J.L. Introduction to Protein Engineering. In


Protein Engineering: Principles and Practice, 1st Ed.;
Cleland, J.L., Craik, C.S., Eds.; Wiley-Liss: New York,
1996; 1–32.

3. Horbett, T.A.; Ratner, B.D.; Schakenraad, J.M.; Schoen,


F.J. Biomaterials Science: An Introduction to Materials in
Medicine, 1st Ed.; Ratner, B.D., Hoffman, A.S., Schoen,
F.J., Lemons, J.E., Eds.; Academic Press: New York, 1996;
133–141. 4. Duan, Y.; Kollman, P.A. Pathways to a protein
folding intermediate observed in a 1-microsecond simulation
in aqueous solution. Science 1998, 282, 740–744. 5. Yoon,
B.J.; Lenhoff, A.M. Computation of the electrostatic
interaction energy between a protein and a charged surface.
J. Phys. Chem. 1992, 96, 3130–3134. 6. Waldman-Meyer, H.;
Knippel, E. A surface charge density model for structure
and orientation of polymer-bound proteins. J. Colloid
Interface Sci. 1992, 148, 508–516. 7. Lu, D.R.; Park, K.
Protein adsorption on polymer surfaces: Calculation of
adsorption energies. J. Biomater. Sci., Polym. Ed. 1990, 1,
243–260. 8. Noinville, V.; Vidal-Madjar, C.; Sebille, B.
Modeling of protein adsorption on polymer surfaces:
Computation potential. J. Phys. Chem. 1995, 99, 1516–1522.
9. Song, D.; Forciniti, D. Monte Carlo simulations of
peptide adsorption on solid surfaces. J. Chem. Phys. 2001,
115, 8089–8900. 10. Daura, X.; Mark, A.E.; van Gunsteren,
W.F. Peptide folding simulations: No solvent required?
Comput. Phys. Commun. 1999, 123, 97–102. 11. Andricioaei,
I.; Karplus, M. On the calculation of entropy from
covariance matrices of the atomic fluctuations. J. Chem.
Phys. 2001, 115, 6289–6292. 12. Bujnowski, A.M.; Pitt, W.G.
Water structure around enkephalin near a PE surface: A
molecular dynamics study. J. Colloid Interface Sci. 1998,
203, 47–58. 13. Bockris, J.O’M.; Reddy, A.K.N.;
Gamboa-Aldeco, M. Modern Electrochemistry:Ionics, 2nd Ed.;
Plenum Pub. Corp.: New York, 2001; 1–2053. 14. Ben-Tal, N.;
Honig, B.; Peitzsch, R.M.; Denisov, G.; McLaughlin, S.
Binding of small basic peptides to membranes containing
acidic lipids: Theoretical models and experimental results.
Biophys. J. 1996, 71, 561–575. 15. West, J.K.; Latour, R.,
Jr.; Hench, L.L. Molecular modeling study of adsorption of
polyL-lysine onto silica glass. J. Biomed. Mater. Res.
1997, 37, 585–591. 16. Latour, R.A., Jr. Molecular modeling
of biomaterial surfaces. Curr. Opin. Solid State Mater.
Sci. 1999, 4, 413–417. 17. Daggett, V.; Levitt, M.A. Model
of the molten globule state from molecular dynamics
simulations. Proc. Natl. Acad. Sci. U. S. A. 1992, 89,
5142–5146. 18. Dauber-Osguthorpe, P.; Roberts, V.A.;
Osguthorpe, D.J.; Wolff, J.; Genest, M.; Hagler, A.T.
Structure and energetics of ligand binding to proteins: E.
coli dihydrofolate reductase-trimethoprim, a drug-receptor
system. Proteins 1988, 4, 31–37. 19. Lewis, G.V.; Catlow,
C.R.A. Potential models for ionic oxides. J. Phys. C. Solid
State Phys. 1985, 18, 1149. 20. Kittel, C. Introduction to
Solid State Physics, 7th Ed.; John Wiley and Sons, Inc.:
New York, 1996; 56–65. 21. Kiselev, A.V.; Lopatkin, A.A.;
Shulga, A.A. Molecular statistical calculation of gas
adsorption by silicalite. Zeolites 1985, 5, 261. 22.
Catlow, C.R.A.; Freeman, C.M.; Vessal, B.; Tomlinson, S.M.;
Leslie, M. Molecular dynamics studies of hydrocarbon
diffusion in zeolites. J. Chem. Soc., Faraday Trans. 1991,
87, 1947. 23. Sastre, B.G.; Catlow, C.R.A.; Chica, A.;
Corma, A. Molecular dynamics of C7 hydrocarbon diffusion in
ITQ-2. The benefits of zeolite structures containing
accessible pockets. J. Phys. Chem., B 2000, 104, 416. 24.
Langel, W.; Parrinello, M.J. Ab Initio molecular dynamics
of H 2 O adsorbed on solid MgO. Chem. Phys. 1995, 103,
3240. 25. McCarthy, M.I.; Schenter, G.K.; Scamehorn, C.A.;
Nicholas, J.B. Structure and dynamics of the water/ MgO
interface. J. Phys. Chem. 1996, 100, 16989. 26. De Leeuw,
N.H.; Watson, G.W.; Parker, S.C. Atomistic simulation of
the effect of dissociative adsorption of water on the
surface structure and stability of calcium and magnesium
oxide. J. Phys. Chem. 1995, 99, 17219. 27. Berendsen,
H.J.C.; Postma, J.P.M.; von Gunsteren, W.F.; Hermans, J.
Intermolecular Forces; Pullman, B., Ed.; Reidel: Dordrecht,
Holland, 1981; 331. 28. Westwood, A.D.; Youngman, R.A.;
Mccartney, M.R.; Cormack, A.N.; Notis, M.R. Oxygen
incorporation in AlN via extended defects: I refinement of a
structural model for the planar inversion domain boundary
(IDB). J. Mater. Res. 1995, 10, 1270. 29. Cormack, A.N.;
Lewis, R.J.; Goldstein, A.H. In preparation. P This page
intentionally left blank
Protein and Cell Signaling with
Biomaterials: Influence of Surfactants

2. Khubchandani, K.R.; Snyder, J.M. Surfactant protein A


(SP-A): the alveolus and beyond. FASEB J. 2001, 15, 59–69.

3. Hills, B.A. Boundary lubrication in vivo. Proc. Inst.


Mech. Eng. H 2000, 214 (1), 83–94.

4. Mills, P.C.; Hills, Y.; Hills, B.A. Surface-active


phospholipid (surfactant) in equine tendon and tendon
sheath fluid. N. Z. Vet. J. 2005, 53 (2), 154–156.

5. Anderson, E.H.; Ruegsegger, M.A.; Murugesan, G.;


Kottke-Marchant, K.; Marchant, R.E. Extracellular
matrix-like surfactant polymers containing
arginineglycine-aspartic acid (RGD) peptides. Macromol.
Biosci. 2004, 4 (8), 766–775.

6. Wang, Y.C.; Ho, C.C. Micropatterning of proteins and


mammalian cells on biomaterials. FASEB J. 2004, 18 (3),
525–527.

7. Maier, R.M. Biosurfactants: evolution and diversity in


bacteria. Adv. Appl. Microbiol. 2003, 52, 101–121.

8. Veldhuizen, R.; Possmayer, F. Phospholipid metabolism in


lung surfactant. Subcell. Biochem. 2004, 37, 359–388.

9. Bakewell, W.E.; Smith, G.J.; Miller, B.E.; Viviano,


C.J.; Hook, G.E. Confocal laser scanning immunofluorescence
microscopy of the pulmonary surfactant system. Association
of surfactant protein A with the nucleus of the alveolar
type II cell. Lab. Invest. 1993, 68 (5), 566–576.

10. Allen, L.V., Jr.; Popovich, N.G.; Ansel, H.C. Ansel’s


Pharmaceutical Dosage Forms and Drug Delivery Systems, 8th
Ed.; Lippincott Williams & Wilkins: Philadelphia, 2005.

11. Chattaraj, S.C.; Das, S.K. Physicochemical


characterization of influenza viral vaccine loaded
surfactant vesicles. Drug Deliv. 2003, 10 (2), 73–77.

12. Abrol, S.; Trehan, A.; Katare, O.P. Formulation,


characterization, and in vitro evaluation of
silymarinloaded lipid microspheres. Drug Deliv. 2004, 11
(3), 185–191.

13. Chekhonin, V.P.; Zhirkov, Y.A.; Gurina, O.L.;


Ryabukhin, I.A.; Lebedev, S.V.; Kashparov, I.A.;
Dmitriyeva, T.B. PEGylated immunoliposomes directed against
brain astrocytes. Drug Deliv. 2005, 12 (1), 1–6.

14. Sato, H. Enzymatic procedure for site-specific


PEGylation of proteins. Adv. Drug Deliv. Rev. 2002, 54 (4),
487–504.

15. Krishna, S.H.; Srinivas, N.D.; Raghavarao, K.S.;


Karanth, N.G. Reverse micellar extraction for downstream
processing of proteins/enzymes. Adv. Biochem. Eng.
Biotechnol. 2002, 75, 119–183.

16. Orlich, B.; Schomacker, R. Enzyme catalysis in reverse


micelles. Adv. Biochem. Eng. Biotechnol. 2002, 75, 185–208.

17. Boontheekul, T.; Mooney, D.J. Protein-based signaling


systems in tissue engineering. Curr. Opin. Biotechnol.
2003, 14 (5), 559–565. 18. Siebers, M.C.; ter Brugge, P.J.;
Walboomers, X.F.; Janeen, J.A. Integrins as linker proteins
between osteoblasts and bone replacing materials. A
critical review. Biomaterials 2005, 26 (2), 137–146. 19.
Mahmood, T.A.; de Jong, R.; Riesle, J.; Langer, R.; van
Blitterswijk, C.A. Adhesion-mediated signal transduction in
human articular chondrocytes: the influence of biomaterial
chemistry and tenascin-C. Exp. Cell Res. 2004, 301,
179–188. 20. Whitsett, J.A.; Weaver, T.E. Hydrophobic
surfactant proteins in lung function and disease. N. Engl.
J. Med. 2002, 347 (26), 2141–2148. 21. Leung, S.W.; Lai,
J.C.K. Differential effects of anionic surfactants on
activities of GDH, LDH, and MDH. Biochem. Eng. J. 2005, 25
(1), 79–88. 22. Sanford, K.; Meyer, D.; Mathison, M.;
Figueras, J. Selective inactivation of lactate
dehydrogenase isoenzymes with ionic surfactants.
Biochemistry 1981, 20, 3207–3214. 23. Lehnert, T.; Berlet,
H.H. Selective inactivation of lactate dehydrogenase of rat
tissues by sodium deoxycholate. Biochem. J. 1979, 117,
813–818. 24. Vincenzini, M.; Favilli, F.; Treves, C.;
Vanni, P. Specific interaction among some enzymes and sodium
dodecyl sulfate. Life Sci. 1982, 31, 463–470. 25. Chander,
A.; Sen, N.; Spitzer, A. Synexin and GTP increase
surfactant secretion in permeabilized alveolar Type II
cells. Am. J. Physiol. (Lung Cellular and Molecular
Physiology) 2001, 280, 991–998. 26. Castro, G.R.;
Knubovets, T. Homogeneous biocatalysis in organic solvents
and water-organic mixtures. Crit. Rev. Biotechnol. 2003,
23, 195–231. 27. Kirk, O.; Borchert, T.V.; Fuglsang, C.C.
Industrial enzyme applications. Curr. Opin. Biotechnol.
2002, 13, 345–351. 28. Cartmell, J.S.; Dunn, M.G. Effect of
chemical treatments on tendon cellularity and mechanical
properties. J. Biomed. Mat. Res. 2000, 49, 134–140. 29.
Kellam, B.; De Bank, P.A.; Shakesheff, K.M. Chemical
modification of mammalian cell surfaces. Chem. Soc. Rev.
2003, 32, 327–337. 30. Levis, S.R.; Deasy, P.B.
Pharmaceutical applications of size reduced grades of
surfactant co-processed microcrystalline cellulose. Int. J.
Pharm. 2001, 230, 25–33. 31. Varshosaz, J.; Pardakhty, A.;
Hajhashemi, V.I.; Najafabadi, A.R. Development and physical
characterization of sorbitan monoester niosomes for insulin
oral delivery. Drug Deliv. 2003, 10, 251–262. 32. Leung,
S.W.; Rivera, G.; Lai, J.C.K. Effects of surfactants on
membrane separation of proteins at trace levels for
drinking water, Proceedings of the 3rd LeadingEdge
Conference on Water and Wastewater Treatment Technologies,
June 6–8, 2005; Sapporo, Japan. 33. Chu, W.; Jafvert, C.T.
Photodechlorination of polychlorobenzene congeners in
surfactant micelle solution. Environ. Sci. Technol. 1994,
28, 2415–2422. P Surfactants
Protein and Cell Signaling with
Biomaterials: Interfacial Transport

8. Ratner, B.D.; Bryant, S.J. Biomaterials: where we have


been and where we are going. Annu. Rev. Biomed. Eng. 2004,
6, 41–75.

9. Lu, D.R.; Kadambi, B.N.; Lai, J.C.K. Protein and cell


interactions with biomaterials—protein conformational
considerations. In Encyclopedic Handbook of Biomaterials
and Bioengineering, Part A: Materials, 1st Ed.; Wise, D.L.,
Trantolo, D.J., Altobelli, D.E., Yaszemski, M.J., Gresser,
J.D., Schwartz, E.R., Eds.; Marcel Dekker, Inc.: New York,
1995; Vol. 1, 305–328.

10. C & EN. In Chemical and Engineering News, 2000, Mar 6,


60–61.

11. Hwang, S.-T.; Kammermeyer, K. Membranes in Separations;


Wiley-Interscience Publications: New York, 1975.

12. Tamagawa, H.; Sakurai, M.; Inoue, Y. Theoretical study


of intermolecular interactions at the lipid water
interface. 2. Analysis based on the Poisson–Boltzmann
equation. J. Phys. Chem. B. 1997, 101 (24), 4817–4825.

13. Tohda, K.; Umezawa, Y.; Yoshiyagawa, S. Cation


permselectivity at the phase boundary of
ionophoreincorporated solvent polymer membranes as studied
by optical second harmonic generation. Anal. Chem. 1995,
67, 570–577.

14. Bailey, A.L.; Cullis, P.R. Membrane fusion with


cationic liposomes: effects of target membrane lipid
composition. Biochemistry 1997, 36, 1628–1634.

15. Morf, W.E. The Principles of Ion-Selective Electrodes


and of Membrane Transport; Elsevier: New York, 1981.

16. Coombs, H.; Miller, A.J.; Sanders, D. Disruptive


effects of protein on performance of liquid membrane-based
microelectrodes. Am. J. Physiol. 1994, 267 (pt 1), C1027–
C1035.

17. Ibanez, J.G.; Batina, N.; Gutierrez-Granados, S.;


Fernandez, L. Conducting polymers for environmental
remediation. Proc. ACS Environ. Chem. 1998, 38 (suppl. 1),
42–43.

18. Martin, C.R.; Mitchell, D.T. Nanomaterials in


analytical chemistry. Anal. Chem. 1998, 70 (9), 322A– 327A.

19. Nishizawa, M.; Menon, V.P.; Martin, C.R. Metal


nanotubule membranes with electrochemically switchable
ion-transport selectivity. Science 1995, 268, 700– 702.

20. Jirage, K.B.; Hulteen, J.C.; Martin, C.R.


Nanotubulebased molecular-filtration membranes. Science
1997, 278 (24), 655–658.

21. Yamada, K.; Gasparac, R.; Martin, C.R. Electrochemical


and transport properties of templated gold/
polypyrrole-composite microtube membranes. J. Electrochem.
Soc. 2004, 151 (1), 14–19. 22. Leung, S.W.; Lai, J.C.K.
Macromolecular diffusion through artificial membranes.
Proceedings of WTA International Conference on Nanoand
Microparticles in Water and Waste Water Treatment, Zurich,
Switzerland, Sep 21–24, 2003, 363–370. 23. Bar-Cohen, Y.
Electroactive Polymer (EAP) Actuators as Artificial Muscles,
Reality Potential, and Challenges; SPIE Press, 2001. 24.
Shahinpoor, M.; Kim, K.J. The effect of surface-electrode
resistance on the performance of ionic polymermetal
composite (IPMC) artificial muscles. Smart Mater. Struct. J.
2000, 9 (4), 543–551. 25. Nemat-Nasser, S. Micromechanics
of actuation of ionic polymer-metal composites. J. Appl.
Phys. 2002, 92 (5), 2899–2915. 26. Baughman, R.H. Muscles
made from metal. Science 2003, 300 (Apr 11), 268–269. 27.
Metcalf and Eddy Inc. In Wastewater Engineering, Treatment,
and Reuse; McGraw Hill: New York, 2003. 28. DeGroot, S.R.
Thermodynamics of Irreversible Processes; North Holland:
Amsterdam, 1961, 186. 29. Skilland, A.H.P. Diffusional Mass
Transfer; Wiley and Sons: New York, 1974. 30. White, J.S.;
White, D.C. Source Book of Enzymes; CRC Press: New York,
1997. 31. Vos, K.D.; Burris, F.O.; Riley, R.L. Kinetic
study of the hydrolysis of cellulose acetate in the pH
range of 2–10. J. Appl. Polym. Sci. 1966, 10, 825–832. 32.
Weber, W. Physicochemical Processes for Water Quality
Control; Wiley and Sons: New York, 1972. 33. Snoeyink,
V.L.; Jenkins, D. Water Chemistry; Wiley and Sons: New
York, 1980. 34. Kesting, R.E. Synthetic Polymeric
Membranes, a Structure Perspective; Wiley and Sons: New
York, 1985. 35. Lonsdale, H.; Meerten, U.; Riley, R.L.
Transport properties of cellulose acetate osmotic
membranes. J. Appl. Polym. Sci. 1965, 9, 1341–1362. FURTHER
READING Leung, S.W.; Lai, J.C.K. Protein and cell signaling
with biomaterials: influence of surfactants. In Encyclopedia
of Biomaterials and Biomedical Engineering; Wnek, G.E.,
Bowlin, G.L., Eds.; Marcel Dekker, Inc.: New York, 2005.
Protein and
Protein Delivery Systems

28. Brannon-Peppas, L. Polymers in controlled drug


delivery. Medical Plastics and Biomaterials 1997, 4, 34–44.

29. Sahlin, J.J.; Peppas, N.A. Hydrogels as mucoand


bioadhesive materials: a review. Biomaterials 1996, 17,
1553–1561.

30. Peppas, N.A. Hydrogels and drug delivery. Curr. Opinion


Coll. Inter. Sci. 1997, 2, 531–537.

31. Kim, S.W.; Bae, Y.H.; Okano, T. Hydrogels: swelling,


drug loading, and release. Pharm. Res. 1992, 9, 283–290.

32. Lowman, A.M.; Peppas, N.A. Hydrogels. In Encyclopedia


of Controlled Release; Mathiowitz, E., Ed.; Wiley: New
York, NY; Vol. 1, 397–418.

33. Bell, C.L.; Peppas, N.A. Biomedical membranes from


hydrogels and interpolymer networks. Adv. Polym. Sci. 1995,
22, 126–175.

34. Khare, A.R.; Peppas, N.A. Swelling/deswelling of


anionic copolymer gels. Biomaterials 1995, 16, 559–567.

35. Peppas, N.A. Hydrogels in Medicine and Pharmacy, Vol. 1


Fundamentals; Peppas, N.A., Ed.; CRC Press: Boca Raton, FL,
1986.

36. Peppas, N.A.; Bures, P. Molecular dynamics of


pHsensitive hydrogels based on poly(acrylic acid). Polym.
Prepr. 1999, 40 (2), 506–507.

37. Peppas, N.A.; Klier, J. Controlled release by using


poly(methacrylic acid-g-ethylene glycol) hydrogels. J.
Control Release 1991, 16, 203–214.

38. Bell, C.L.; Peppas, N.A. Water, solute and protein


diffusion in physiologically-responsive hydrogels of
poly(methacrylic acid-g-ethylene glycol). Biomaterials
1996, 17, 1203–1218.

39. Lowman, A.M.; Peppas, N.A. Solute transport analysis in


pH-responsive, complexing hydrogels of poly(methacrylic
acid-g-ethylene glycol). J. Biomat. Sci. Polym. Edn. 1999,
10, 999–1009.

40. Peppas, N.A.; Lowman, A.M. Pulsatile drug delivery


based on a complexation-decomplexation mechanism. In
Intelligent Materials for Controlled Release; Dinh, S.M.,
DeNuzzio, J.D., Comfort, A.R., Eds.; ACS Symposium Series:
Washington, DC, 1999; Vol. 728, 30–42.

41. Bell, C.L.; Peppas, N.A. Equilibrium and dynamic


swelling of polyacrylates. Polym. Eng. Sci. 1996, 36,
1856–1861.

42. Drummond, R.K.; Klier, J.; Alameda, J.A.; Peppas, N.A.


Preparation of poly(methacrylic acid-g-ethylene oxide)
microspheres. Macromolecules 1989, 22, 3816–3818.

43. Nakamura, K.; Maitani, Y.; Lowman, A.M.; Takayama, K.;


Peppas, N.A.; Nagai, T. Uptake and release of budesonide
from mucoadhesive, pH-sensitive copolymers and their
application to nasal delivery. J. Control Release 1999, 61,
329–335.

44. Lowman, A.M.; Morishita, M.; Kajita, M.; Nagai, T.;


Peppas, N.A. Oral delivery of insulin using pH-responsive
complexation gels. J. Pharm. Sci. 1999, 88, 933–937.

45. Lowman, A.M.; Peppas, N.A.; Morishita, M.; Nagai, T.


Novel bioadhesive complexation network for oral protein
drug delivery. In Materials for Controlled Release
Applications; McCullough, I., Shalby, S., Eds.; ACS
Symposium Series: Washington, DC, 1999. 46. Torres-Lugo,
M.; Peppas, N.A. Molecular design and in vitro studies of
novel pH sensitive hydrogels for oral delivery of
calcitonin. Macromolecules 1999, 32, 6646– 6651. 47.
Constantino, H.R.; Liauw, S.; Mitragotri, S.; Langer, R.;
Klibanov, A.M.; Sluzky, V. The pharmaceutical development
of insulin: historical perspectives and future directions.
In Therapeutic Protein and Peptide Formulation and
Delivery; Shahrokh, Z., Sluzky, V., Cleland, J.L., Shire,
S.J., Randolph, T.W., Eds.; American Chemical Society:
Washington, DC, 1997; 29–66. 48. Saffran, M.; Sudesh Kumar,
G.; Savariar, C.; Burnhan, J.C.; Willims, F.; Neckers, D.C.
A new approach to the oral administration of insulin and
other peptide drugs. Science 1986, 233, 1081–1084. 49.
Saffran, M. Oral colon-specific drug delivery with emphasis
on insulin. In Oral Colon-Specific Drug Delivery; Friend,
D.R., Ed.; CRC Press: Boca Raton, FL, 1992; 115–142. 50.
Morishita, I.; Morishita, M.; Takayama, K.; Machida, Y.;
Nagai, T. Hypoglycemic effect of novel oral microspheres of
insulin with protease inhibitor in normal and diabetic
rats. Int. J. Pharm. 1992, 78, 9–16. 51. Carino, G.P.;
Jacob, J.S.; Mathiowitz, E. Nanospheres based oral insulin
delivery. J. Control Release 2000, 65, 261–269. 52.
Ashcroft, F.; Ashcroft, S. Insulin: Molecolar Biology to
Pathology; Oxford University Press: New York, 1992. 53.
Goodman Gilman, A. Insulin, oral hypoglycemic agents, and
the pharmacology of the endocrine pancreas. In The
Pharmacological Basis of Therapeutics; McGrawnHill, Ed.;
2001. 54. Harrison’s Principles of Internal Medicine,
Genetics of Diabetes Mellitus; McGrawn-Hill Ed., 2001,
http:// books,mcgraw-hill.com/medical/harrisons/index.html.
55. Trehan, A.; Ali, A. Recent approaches in insulin
delivery. Drug Dev. Ind. Pharm. 1998, 26, 589–597. 56.
Rohde, T.D.; Buchwald, H. Implantable pumps: recent
progress and anticipated future advances. ASAIO Journal,
1992, 38, 772–778. 57. Charles, M.A.; Selam, J.L. Devices
for insulin administration. Diabetes Care 1990 , 13,
955–979. 58. Ziv, E.; Hoffman, A. Pharmacokinetic
consideration of new insulin formulations and routes of
administrations. Clin. Pharmacokinet. 1997, 33, 285–301.
59. Mitragotri, S.; Blankschtein, D.; Langer, R.
Ultrasoundmediated transdermal protein delivery. Science
1995, 269, 850–853. 60. Nagai, T.; Nishimoto, Y.; Nambu,
N.; Suzuki, Y.; Sekine, K. Powder dosage form of insulin
for nasal administration. J. Control Release 1984, 1,
15–22. 61. Longenecker, J.P. Nazlin s transnasal systemic
delivery of insulin. In Delivery Systems for Peptide Drugs;
Davis, S.S., Illum, L., Tomlinson, E., Eds.; Plemun: New
York, 1986; 211–220. 62. Moses, A.C.; Gordon, G.S.; Carey,
M.C.; Flier, J.S. Insulin administered intranasally as an
insulin-bile salt aerosol-effectiveness and reproducibility
in normal and diabetic subjects. Diabetes 1983, 32, 1040–
1047. 63. Fernandez_Urrusuno, R.; Calvo, P.; Reuman-Lopez,
C.; Vila-Jato, J.L.; Alonso, M.J. Enhancement of nasal
absorption of insulin using chitosan nanoparticles. Pharm.
Res. 1993, 16, 1576–1581. 64. Danforth, E.; Ur, R.; Moore,
O. Intestinal absorption of insulin in the rat.
Endocrinology 1959, 65, 118. 65. Laskowski, M.; Haessier,
H.A.; Mieon, R.R.; Peanasky, R.J.; Laskowski, M. Effect of
trypsin inhibitor on passage of insulin across the
intestinal barrier. Science 1958, 127, 1115. 66. Patel,
H.M.; Ryman, B.E. Oral administration of insulin by
encapsulation within liposomes. FEBS Lett. 1976, 62, 60–74.
67. Mehnert, W.; Mader, K. Solid lipid nanospheres.
Production, characterization and applications. Adv. Drug
Del. Rev. 2001, 47, 165–196. 68. De Jaeghere, F.; Doelker,
E.; Gurny, R. Nanospheres. In Encyclopedia of Controlled
Drug Delivery; Mathiowitz, E., Ed.; John Wiley & Sons,
Inc.: New York, 1999; Vol. 2, 641–664. 69. Mittal, K.L.;
Kumar, T.M. Handbook of Microemulsion Science and
Technology; Marcel Dekker Eds.: New York, NY, 1999. 70.
Wisendanger, R. Scanning Probe Microscopy and Spectroscopy.
Methods and Applications; Cambridge University Press:
Cambridge, 1994. 71. Sudol, E.D. Dispersion polymerization.
In Polymeric Dispersions: Principles and Applications;
Asua, J.M., Ed.; Kluwer Academic Publishers: Boston, 1997;
141–154. 72. Finsy, R. Particle sizing by quasi-elastic
light scattering. Adv. Coll. Inter. Sci. 1999, 52, 79–143.
73. Flory, P.J. Principles of Polymer Chemistry; Cornell
Univ. Press: Ithaca, New York, 1953. 74. Suzuki, M. Gels
Handbook, The Fundamentals; Osada, Y., Kajiwara, K., Eds.;
Academic Press: New York, 2001; 228–234. 75. Odian, G.
Principle of Polymerization; John Wiley & Sons, Inc.: New
York, 1990. 76. Arshady, R. Suspension, emulsion and
dispersion polymerization: a methodological survey. Colloid
Polym. Sci. 1990. 77. Goldstein, J.I.; Newbury, D.E.;
Echlin, P.; Joy, D.C.; Romig, A.D.; Lyman, C.E.; Fiori, C.;
Lifshin, E. Scanning Electron Microscopy and X-Ray
Microanalysis; Plenum Press, 1992. 78. Kreuter, J.
Nanospheres. In Colloidal Drug Delivery Systems; Kreuter,
J., Ed.; Marcel Dekker: New York, NY, 1994; 219–342. 79.
Weiner, B.B. Modern Methods of Particle Size Analysis;
Barth, H.G., Ed.; John Wiley & Sons: New York, 1984;
93–116. 80. Madsen, F.; Peppas, N.A. Complexation graft
copolymer networks: swelling properties, calcium binding,
and proteolytic enzyme inhibition. Biomaterials 1999 , 20,
1701–1708. P
Pyrolytic Carbon

1. LaGrange, L.D.; Gott, V.L.; Bokros, J.C.; Ramos, M.D.


Compatibility of Carbon and Blood, Chapter 5. Artificial
Heart Program Conference, National Heart Institute
Artificial Heart Program, Washington, DC, June 9–13, 1969;
Johnson Hegyeli, R., Ed.; 47–58. 2. Bokros, J.C.
Deposition, Structure and Properties of Pyrolytic Carbon.
In Chemistry and Physics of Carbon; Walker, P.L., Ed.;
Marcel Dekker, Inc.: New York, 1969; Vol. 4, 1–118. 3.
Akins, R.J.; Bokros, J.C. The deposition of pure and
alloyed isotropic carbons and steady state fluidized beds.
Carbon 1974, 12, 439–452. 4. Brewer, L.A. Prosthetic Heart
Valves; Charles C Thomas Pub: Springfield, IL, 1969. 5.
Bokros, J.C.; Haubold, A.D.; Akins, R.J.; Campbell, A.C.;
Griffin, C.D.; Lane, E. The Durability of Mechanical Heart
Valve Replacements: Past Experience and Current Trends. In
Replacement Cardiac Valves; Bodnar, E., Frater, R.W.M.,
Eds.; Pergamon Press: New York, 1991; 21–48. 6. Sadeghi, H.
Dysfonctions des prostheses valvulaires cardaques et leur
traitement chirurgical. Schwiez. Med. Wschr. 1987, 117,
1665–1670. 7. Black, M.M.; Ccochrane, T.; Lawford, P.V.;
Reul, H.; Yoganathan, A. Design and Flow Characteristics.
In Replacement Cardiac Valves; Bodnar, E., Frater, R.W.M.,
Eds.; Pergamon Press: New York, 1991; 1–20. 8. Milano, A.;
Bortolotti, U.; Mazzucco, A.; Mossuto, E.; Testolin, L.;
Thiene, G.; Gallucci, U. Heart valve replacement with the
Sorin tilting-disc prosthesis. J. Thorac. Cardiovasc. Surg.
1992, 103, 267–275. 9. Vallana, F.; Rinaldi, S.; Galletti,
P.M.; Nguyen, A.; Piwnica, A. Pivot design in bileaflet
valves. ASAIO Trans. 1992, 38, M600–M606. 10. Arru, P.;
Santi, M.; Vallana, F.; Majini, G.; Ottaviani, G.;
Paccagnella, A. A New PyC Film for Biomedical Applications.
In High Tech Ceramics; Vincenzini, P., Ed.; Elsevier
Science Pub. B.V.: Amsterdam, 1987; 117–126. 11. Bokros,
J.C.; Atkins, R.J. Process for Depositing Pyrolytic Carbon
Coatings. US patent 3,977,896, August 31, 1976. 12. Ely,
J.L.; Emken, M.R.; Accuntius, J.A.; Wilde, D.S.; Haubold,
A.D.; More, R.B.; Bokros, J.C. Pure pyrolytic carbon:
Preparation and properties of a new material, On-X carbon,
for mechanical heart valve prostheses. J. Heart Valve Dis.
1998, 7 (6), 626–632.

Table 4 Carbomedics valve damage-tolerant lifetimes

Component and location Proof stress (MPa) Initial flaw size


(mm) Design stress (MPa) Final flaw size (mm) Lifetime
(years)

Leaflet B-datum 167 21.5 47 250 1.8� 10 30


Leaflet ear shoulder 64 143 33 431 4.6� 10 16

Leaflet tip 157 24.3 40 325 5.2� 10 32

Leaflet ear 530 2.67 310 7.8 7.8� 10 11

Orifice pivot 530 2.67 310 7.8 7.8� 10 11

Orifice groove 67 163 30 565 1.1� 10 20

Orifice rim 78 98 34 414 3.0� 10 20 13. Bokros, J. Carbon


biomedical devices. Carbon 1977, 15, 355–371. 14. Tian,
C.L.; Hetherington, V.J.; Reed, S. A review of pyrolytic
carbon: Application in bone and joint surgery. J. Foot
Ankle Surg. 1993, 32 (5), 490–498. 15. Cook, S.D.; Thomas,
K.A.; Kester, M.A. Wear characteristics of the canine
acetabulum against different femoral prostheses. J. Bone
Jt. Surg. 1989, 71B, 189–197. 16. Cook, S.D.; Beckenbaugh,
R.D.; Redondo, J.; Popich, L.S.; Klawitter, J.J.;
Linscheid, R.L. Long term followup of pyrolytic carbon
metacarpophalangeal implants. J. Bone Jt. Surg. 1999, 81A
(5), 635–648. 17. More, R.B.; Kepner, J.L.; Strzepa, P.
Hertzian Fracture in Pyrolite Carbon. In Bioceramics Volume
6; Ducheyne, P., Christiansen, D., Eds.;
Butterworth–Heinemann: Oxford, 1993; 225–228. 18. Chwirut,
D.J.; Regnault, W.F. Fracture mechanics principals applied
to implant medical devices—A review. Med. Prog. Technol.
1988, 14, 193–203. 19. Ritchie, R.O.; Lubock, P. Fatigue
life estimation procedures for the endurance of a cardiac
valve prosthesis: Stress/life and damage tolerant analyses.
J. Biomech. Eng. 1986, 108, 153–160. 20. Klepetko, W.;
Moritz, A.; Mlczoch, J.; Schurawitzki, H.; Domanig, E.;
Wolner, E. Leaflet fracture in Edwards–Duromedics bileaflet
valves. J. Thorac. Cardiovasc. Surg. 1989, 97, 90–94. 21.
Ritchie, R.O.; Dauskardt, R.H.; Weikang, Yu; Brendzel, A.M.
Cyclic fatigue-crack propagation and fracture toughness
behavior in pyrolytic carbon-coated graphite for heart
valve applications. J. Biomed. Mater. Res. 1990, 24,
189–206. 22. Tada, H.; Paris, P.C.; Irwin, G.R. Stress
Analyses of Cracks Handbook, 2nd Ed.; Paris
Publications/Del research Corp.: St. Louis, MO, 1985. 23.
Schoen, F.J. On the fatigue behavior of pyrolytic carbons
under cyclic loading. Carbon 1973, 11, 413–414. 24. Shim,
H.S. The behavior of isotropic pyrolytic carbons under
cyclic loading. Biomater. Med. Dev. Artif. Organs 1974, 2
(1), 55–65. 25. Kepner, J.; Haubold, A.D.; Beavan, L.A.
Cyclic Fatigue Testing of Pyrolytic Carbon, 41st Pacific
Coast Regional Meeting of the American Chemical Society,
San Francisco, October, 1990 26. Ma, L.; Sines, G.
Unalloyed pyrolytic carbon for implanted mechanical heart
valves. J. Heart Valve Dis. 1999, 8, 578–585. 27. Ma, L.
George Sines. Fatigue behavior of a pyrolytic carbon. J.
Biomed. Mater. Res. 2000, 51 (1), 61–68. 28. Kaae, J.L. The
mechanism of the deposition of a pyrolytic carbon. Carbon
1985, 23 (6), 665–673. 29. Ma, L. Studies on Pyrolytic
Carbons for Biomedical Applications; Dissertation.
University of California: Los Angeles1997. 30. Ryder, J.K.;
Cao, H. Structural integrity assessment of heart valve
prostheses: A damage tolerance analysis of the carbomedics
prosthetic heart valve. J. Heart Valve Dis. 1996, 5 (Suppl
I), S86–S96. P
Rapid Prototyping

20. Cheah, C.M.; Leong, K.F.; Chua, C.K.; Low, K.H.; Quek,
H.S. Characterization of microfeatures in selective laser
sintered drug delivery devices. Proc. Inst. Mech. Engrs. H:
J. Eng. Med. 2002, 216 (H6), 369–383.

21. Wu, B.M.; Borland, S.W.; Giordano, R.A.; Cima, L.G.;


Sachs, E.M.; Cima, M.J. Solid free-form fabrication of drug
delivery devices. J. Controlled Release 1996, 40, 77–87.

22. Lee, M.; Dunn, J.C.D.; Wu, B.M. Scaffold fabrication by


indirect three-dimensional printing. Biomaterials 2005, 26,
4281–4289.

23. Sherwood, J.K.; Riley, S.L.; Palazzolo, R.; Brown,


S.C.; Monkhouse, D.C.; Coates, M.; Griffith, L.G.; Landeen,
L.K.; Ratcliffe, A. A three-dimensional osteochrondral
composite scaffold for articular cartilage repair.
Biomaterials 2002, 23, 4739–4751.

24. Hutmacher, D.W.; Schantz, T.; Zein, I.; Ng, KW.; Teoh,
S.H.; Tan, C.K. Mechanical properties and cell cultural
response of polycaprolactone scaffolds designed and
fabricated via fused deposition modeling. J. Biomed. Mater.
Res. 2001, 55 (2), 203–216.

25. Vozzi, G.; Flaim, C.; Ahluwalia, A.; Bhatia, S.


Fabrication of PLGA scaffolds using soft lithography and
microsyringe deposition. Biomaterials 2003, 24 (14),
2533–2540. 26. Zein, I.; Hutmacher, D.W.; Tan, K.C.; Teoh,
S.H. Fused deposition modeling of novel scaffold
architectures for tissue engineering applications.
Biomaterials 2002, 23 (4), 1169–1185. 27. Tan, W.; Desai,
T.A. Layer-by-layer microfluidics for biomemitic
three-dimensional structures. Biomaterials 2004, 25 (7–8),
1355–1364. 28. King, K.R.; Wang, C.C.J.; Kaazempur-Mofrad,
M.R.; Vacanti, J.P.; Borenstein, J.P. Biodegradable
microfluidics. Adv. Mater. 2004, 16 (22), 2007–2012. 29.
Valerie, A.L.; Sangeeta, N.B. Three-dimesional
photopatterning of hydrogels containing living cells.
Biomed. Microdev. 2002, 4, 257–266. FURTHER READING Curtis,
A.; Wilkinson, C. Nanotechniques and approaches in
biotechnology. Trends Biotechnol. 2001, 19, 97–101. LaVan,
D.A.; Mcguire, T.; Langer, R. Small-scale systems for in
vivo drug delivery. Nat. Biotechnol. 2003, 21 (10),
1184–1191. Lu, Y.; Chen, S.C. Micro and nano-fabrication of
biodegradable polymers for drug delivery. Adv. Drug Deliv.
Rev. 2004, 56, 1621–1633. Yeong, W.Y.; Chua, C.K.; Leong,
K.F.; Chandrasekaran, M. Rapid prototyping in tissue
engineering: challenges and potential. Trends Biotechnol.
2004, 22 (12), 643–652.
Real Time Tomographic Reflection

1. State, A.; Livingston, M.; Garret, W.; Hirota, G.;


Whitton, M.; Pisano, E.; Fuchs, H. Technologies for
Augmented Reality Systems: Realizing UltrasoundGuided
Needle Biopsies. In Computer Graphics Proceedings, Annual
Conference Series, Proceedings of SIGGRAPH, New Orleans,
LA, 1996; 439–446.

2. Fuchs, H.; State, A.; Pisano, E.; Garret, W.; Hirota,


G.; Livingston, M.; Whitton, M.; Pizer, S. Towards
Performing Ultrasound-Guided Needle Biopsies from Within a
Head-Mounted Display. In Visualization in Biomedical
Computing; Springer Verlag: Hamburg, Germany, 1996;
591–600.

3. Fuchs, H.; Livingston, M.; Raskar, R.; Colucci, D.;


Keller, K.; State, A. Augmented Reality Visualization for
Laparoscopic Surgery. In Lecture Notes in Computer Science,
Proceedings of MICCAI, Cambridge, MA, 1998; 934–943.

4. Sauer, F.; Khamene, A.; Bascle, B.; Schimmang, L.;


Wenzel, F.; Vogt, S. Augmented Reality Visualization of
Ultrasound Images: System Description, Calibration, and
Features. In IEEE ACM International Symposium on Augmented
Reality; IEEE Computer Society: New York, 2001; 30–39.

5. Azuma, R.T. A survey of augmented reality. Teleoperators


and virtual environments. 1997, 6 (4), 355–385.

6. DiGioia, A.; Colgan, B.; Koerbel, N. Computer-Aided


Surgery. In Cybersurgery; Satava, R., Ed.; Wiley: New York,
1998; 121–139.

7. Blackwell, M.; Morgan, F.; DiGioia, A. Augmented reality


and its future in orthopaedics. Clin. Orthop. Relat. Res.
1998, 345, 111–122.

8. Stetten, G.; Chib, V.; Tamburo, R. System for


Location-Merging Ultrasound Images with Human Vision. In
Applied Imagery Pattern Recognition (AIPR) Workshop; IEEE
Computer Society: Washington, DC, 2000; 200–205. 9.
Stetten, G.; Chib, V. Real Time Tomographic Reflection with
Ultrasound: Stationary and Hand-Held Implementations;
Carnegie Mellon University, Nov. 2000. Robotics Institute
Technical Report # CMU-RI-TR000-28.
http://www.ri.cmu.edu/pubs/pub_3412.html# text_ref. 10.
Stetten, G.; Chib, V. Overlaying ultrasound images on
direct vision. J. Ultrasound Med. 2001, 20 (1), 235–240.
11. Stetten, G.; Chib, V. Magnified Real-Time Tomographic
Reflection. In Lecture Notes in Computer Science,
Proceedings of MICCAI, Springer Verlag, 2001; 683–690. 12.
Stetten, G.; Shelton, D.; Chang, W.; Chib, V.; Tamburo, R.;
Hildebrand, D.; Lobes, L.; Sumkin, J. Towards a Clinically
Useful Sonic Flashlight. In Proceedings of the IEEE
International Symposium on Biomedical Imaging; IEEE
Computer Society: Washington, DC, 2002. 13. Chang, W.;
Stetten, G.; Lobes, L.; Shelton, D.; Tamburo, R. Guidance
of retrobulbar injection with real time tomographic
reflection. J. Ultrasound Med. 2002, 21, 1131–1135. 14.
Stetten, G.; Chib, V.; Hildebrand, D.; Bursee, J. Real Time
Tomographic Reflection: Phantoms for Calibration and Biopsy.
In IEEE ACM International Symposium on Augmented Reality;
IEEE Computer Society: New York, 2001; 11–18. 15. Masamune,
K.; Masutani, Y.; Nakajima, S.; Sakuma, I.; Dohi, T.;
Iseki, H.; Takakura, K. Three-Dimensional Slice Image
Overlay System with Accurate Depth Perception for Surgery.
In Lecture Notes in Computer Science, Proceedings of
MICCAI, Pittsburgh, PA, 2000; 395–402. 16. Masamune, K.;
Fichtinger, G.; Deguet, A.; Matsuka, D.; Taylor, R. An
Image Overlay System with Enhanced Reality for Percutaneous
Therapy Performed Inside CT Scanner. In Lecture Notes in
Computer Science, Proceedings of MICCAI, Springer Verlag,
2002; 77–84. 17. Hofstein, S.R. Ultrasonic Scope. US Patent
956899, April 29, 1980.
Reconstructive Biomaterials

1. Habal, M.B. Absorbable platting system-discussion of,


resorbable plate fixation in pediatric craniofacial surgery.
Plast. Reconstr. Surg. 1997, 100, 8–11.

2. Habal, M.B.; Pieterzak, W.S. Key points in the fixation


of the craniofacial skeleton with absorbable biomaterial.
J. Craniofac. Surg. 1999, 10, 491–499.

3. Kulkarni, R.K.; Pani, K.C.; Neuman, C.; Leonard, F.


Polylactic acid for surgical implants. Arch. Surg. 1966,
93, 839–843.

4. Gogolewski, S.; Jovanovic, M.; Perren, S.M.; Dillon,


J.G.; Hughes, M.K. Tissue response and in vivo degradation
of selected polyhydroxyacids: polylactides,
poly(3-hydroxybutyrate), and
poly(3-hydroxybutyrateco-3-hydroxyvalerate). J. Biomed.
Mater. Res. 1993, 27, 1135–1148.

5. Van der Elst, M.; Dijkema, A.R.; Klein, C.P.; Patka, P.;
Haarman, H.J. Tissue reaction on PLLA versus stainless
steel interlocking nails for fracture fixation: an animal
study. Biomaterials 1995, 16, 103–106.

6. Mainil-Varlet, P.; Rahn, B.; Gogolewski, S. Long-term in


vivo degradation and bone reaction to various polylactides.
Biomaterials 1997, 18, 257–266.

7. Habal, M.B. Absorbable, invisible and flexible plating


system for the craniofacial skeleton. J. Craniofac. Surg.
1997, 8, 121–126.

Habal, M.B.; Reddi, A.H. Bone Grafts and Bone Substitutes;


WB Saunders: New York, 1992.

Tharanon, W.; Sinn, D.P.; Hobar, P.C.; Sklar, F.H.;


Salomon, J. Surgical outcomes using bioabsorbable plating
systems in pediatric craniofacial surgery. J. Craniofac.
Surg. 1998, 9, 441–444. R
Rotary Cell Culture Systems

26. Nickerson, C.A.; Goodwin, T.J.; Terlonge, J.; Ott,


C.M.; Buchanan, K.L.; Uicker, W.C.; Emami, K.; LeBlanc,
C.L.; Ramamurthy, R.; Clarke, M.S.; Vanderburg, C.R.;
Hammond, T.; Pierson, D.L. Three-dimensional tissue
assemblies: Novel models for the study of Salmonella
enterica serovar Typhimurium pathogenesis. Infect. Immun.
2001, 69, 7106–7120.

27. Margolis, L.B.; Fitzgerald, W.; Glushakova, S.; Hatfill,


S.; Amichay, N.; Baibakov, B.; Zimmerberg, J. Lymphocyte
trafficking and HIV infection of human lymphoid tissue in a
rotating wall vessel. AID Res. Hum. Retroviruses 1997, 13,
1411–1420.

28. Yoffe, B.; Darlington, G.J.; Soriano, H.E.; Krishnan,


B.; Risin, D.; Pellis, N.R.; Khaoustov, V.L. Cultures of
human liver cells in simulated microgravity environment.
Adv. Space Res. 1999, 24, 829–836.

29. Dabos, K.J.; Nelson, L.J.; Bradnock, T.J.; Parkinson,


J.A.; Sadler, I.H.; Hayes, P.C.; Plevris, J.N. The
simulated microgravity environment maintains key metabolic
functions and promotes aggregation of primary porcine
hepatocytes. Biochim. Biophys. Acta 2001, 1562, 119–130.
30. Low, H.P.; Savarese, T.M.; Schwartz, W.J. Neural
precursor cells form rudimentary tissue-like structures in
a rotating wall vessel bioreactor. In Vitro Cell. Dev.
Biol., Anim. 2001, 37, 141–147. 31. Rutsky, L.P.; Bilinski,
S.; Kloc, M.; Phan, T.; Zhang, H.; Katz, S.M.; Stepkowski,
S.M. Microgravity culture conditions reduces immunogenicity
and improves function of pancreatic islets. Transplantation
2002, 74, 13–21. 32. Cameron, D.F.; Hushen, J.J.; Nazian,
S.J.; Willing, A.; Saporta, S.; Sanberg, P.R. Formation of
sertoli cellenriched tissue constructs utilizing simulated
microgravity technology. Ann. N.Y. Acad. Sci. 2001, 944,
420–428.
Self-Assembled Monolayers

18. Porter, M.D.; Bright, T.B.; Allara, D.L.; Chidsey,


C.E.D. Spontaneously organized molecular assemblies. 4.
Structural characterization of n-alkyl thiol monolayers on
gold by optical ellipsometry, infrared spectroscopy, and
electrochemistry. J. Am. Chem. Soc. 1987, 109, 3559–3568.

19. Strong, L.; Whitesides, G.M. Structures of


self-assembled monolayer films of organosulfur compounds
adsorbed on gold single crystals: Electron diffraction
studies. Langmuir 1988, 4, 546–558.

20. Rubinstein, I.; Steinberg, S.; Tor, Y.; Shanzer, A.;


Sagiv, J. Ionic recognition and selective response in
self-assembling monolayer membranes on electrodes. Nature
1988, 332, 426–429.

21. Chidsey, C.E.D.; Liu, G.-Y.; Rowntree, P.; Scoles, G.


Molecular order at the surface of an organic monolayer
studied by low energy helium diffraction. J. Chem. Phys.
1989, 91, 4421–4423.

22. Bain, C.D.; Troughton, B.; Tao, Y.T.; Evall, J.;


Whitesides, G.M.; Nuzzo, R.G. Formation of monolayer films
by the spontaneous assembly of organic thiols from solution
onto gold. J. Am. Chem. Soc. 1989, 111, 321–335.

23. Bain, C.D.; Whitesides, G.M. Formation of monolayers by


the coadsorption of thiols on gold: variation in the length
of the alkyl chain. J. Am. Chem. Soc. 1989, 111, 7164–7175.

24. Whitesides, G.M.; Laibinis, P.E. Wet chemical


approaches to the characterization of organic surfaces:
Self-assembled monolayers, wetting, and the physicalorganic
chemistry of the solid–liquid interface. Langmuir 1990, 6,
87–96.

25. Nuzzo, R.G.; Dubois, L.H.; Allara, D.L. Fundamental


studies of microscopic wetting on organic surfaces. 1.
Formation and structural characterization of a
selfconsistent series of polyfunctional organic monolayers.
J. Am. Chem. Soc. 1990, 112, 558–569.

26. Nuzzo, R.G.; Fusco, F.A.; Allara, D.L. Spontaneously


organized molecular assemblies. 3. Preparation and
properties of solution adsorbed monolayers of organic
disulfides on gold surfaces. J. Am. Chem. Soc. 1987, 109,
2358–2368.
27. Troughton, E.B.; Bain, C.D.; Whitesides, G.M.; Nuzzo,
R.G.; Allara, D.L.; Porter, M.D. Monolayer films prepared by
the spontaneous self-assembly of symmetrical and
unsymmetrical dialkyl sulfides from solution onto gold
substrates: structure, properties, and reactivity of
constituent functional groups. Langmuir 1988, 4, 365–385.

28. Ulman, A. Self-assembled monolayer of


4-mercaptobiphenyls. Acc. Chem. Res. 2001, 34, 855–863.

29. Aslam, M.; Bandyopadhyay, K.; Vijayamohanan, K.;


Lakshminarayanan, V. Comparative behavior of aromatic
disulfide and diselenide monolayers on polycrystalline gold
films using cyclic voltammetry, STM, and quartz crystal
microbalance. J. Colloid Interface Sci. 2001, 234, 410–417.

30. Clegg, R.S.; Reed, S.M.; Hutchison, J.E. Self-assembled


monolayers stabilized by three-dimensional networks of
hydrogen bonds. J. Am. Chem. Soc. 1998, 120, 2486– 2487.
31. Fenter, P.; Eisenberger, P.; Li, J.; Camillone, N.,
III; Bernasek, S.; Scoles, G.; Ramanarayanan, T.A.; Liang,
K.S. Structure of octadecyl thiol self-assembled on the
silver(111) surface: An incommensurate monolayer. Langmuir
1991 , 7, 2013–2016. 32. Ehler, T.T.; Malmberg, N.; Noe,
L.J. Characterization of self-assembled alkanethiol
monolayers on silver and gold using surface plasmon
spectroscopy. J. Phys. Chem., B 1997, 101, 1268–1272. 33.
Laibinis, P.E.; Whitesides, G.M. Self-assembled monolayers
of n-alkanethiolates on copper are barrier films that
protect the metal against oxidation by air. J. Am. Chem.
Soc. 1992, 114, 9022–9028. 34. Lang, P.; Mekhalif, Z.; Rat,
B.; Garnier, F. Self-assembled alkylthiols monolayers onto
platinum; influence of the adsorbed oxygen. J. Electroanal.
Chem. 1998, 441, 83–93. 35. Li, Z.; Chang, S.-C.; Williams,
R.S. Self-assembly of alkanethiol molecules onto platinum
and platinum oxide surfaces. Langmuir 2003, 19, 6744–6749.
36. Brito, R.; Tremont, R.; Feliciano, O.; Cabrera, C.R.
Chemical derivatization of self-assembled
3-mercaptopropionic and 16-mercaptohexadecanoic acids at
platinum surfaces with 3-aminopropyltrimethoxysilane: A
spectroscopic and electrochemical study. J. Electroanal.
Chem. 2003, 19, 53–59. 37. Ulman, A. Formation and
structure of self-assembled monolayers. Chem. Rev. 1996,
96, 1533–1554. 38. Mrksich, M.; Whitesides, G.M. Patterning
self-assembled monolayers using microcontact printing: A
new technology for biosensors. Trends Biotechnol. 1995, 13,
228–235. 39. Chechik, V.; Crooks, R.M.; Stirling, C.J.M.
Reactions and reactivity in self-assembled monolayers. Adv.
Mater. 2000, 12, 1161–1171. 40. Davies, J.; Faulkner, I.
Surface Plasmon Resonance— Theory and Experimental
Considerations. In Surface Analytical Techniques for
Probing Biomaterial Process; Davies, J., Ed.; CRC Press:
Boca Raton, FL, 1996; 67– 87. 41. Ivarsson, B.; Malmqvist,
M. Surface Plasmon Resonance. Development and Use of
Biacore Instruments for Biomolecular Interaction Analysis.
In Biomolecular Sensors; Gizeli, E., Lowe, C.R., Eds.;
Taylor & Francis: London, 2002; 241–268. 42. Hirata, I.;
Hioki, Y.; Toda, M.; Kitazawa, T.; Murakami, Y.; Kitano,
E.; Kitamura, H.; Ikada, Y.; Iwata, H. Deposition of
complement protein, C3b, on mixed self-assembled monolayers
carrying surface hydroxyl and methyl groups studied by
surface plasmon resonance. J. Biomed. Mater. Res. 2003,
66A, 669–676. 43. Nelson, B.P.; Frutos, A.G.; Brockman,
J.M.; Corn, R.M. Near-infrared surface plasmon resonance
measurements of ultrathin films. 1. Angle shift and SPR
imaging experiments. Anal. Chem. 1999, 71, 3928–3934. 44.
Liebermann, T.; Knoll, W. Parallel multispot detection of
target hybridization to surface-bound probe
oligonucleotides of different base mismatch by
surface-plasmon field-enhanced fluorescence microscopy.
Langmuir 2003, 19, 1567–1572. 45. Ferretti, S.; Payner, S.;
Russell, D.A.; Sapsford, K.E. Self-assembled monolayers: A
versatile tool for the formation of bio-surfaces. Trends
Anal. Chem. 2000, 19, 530–540. 46. Schaeferling, M.;
Schiller, S.; Paul, H.; Kruschina, M.; Pavlickova, P.;
Meerkamp, M.; Giammasi, C.; Kambhampati, D. Application of
self-assembly techniques in the design of biocompatible
protein microarray surfaces. Electrophoresis 2002, 23,
3097–3105. 47. Sigal, G.B.; Bamdad, C.; Barberis, A.;
Strominger, J.; Whitesides, G.M. A self-assembled monolayer
for the binding and study of histidine-tagged proteins by
surface plasmon resonance. Anal. Chem. 1996, 68, 490– 497.
48. Hyun, J.; Ahn, S.J.; Lee, W.K.; Chilkoti, A.; Zauscher,
S. Molecular recognition-mediated fabrication of protein
nanostructures by dip-pen lithography. Nano Lett. 2002, 2,
1203–1207. 49. Yousaf, M.N.; Houseman, B.T.; Mrksich, M.
Using electroactive substrates to pattern the attachment of
two different cell populations. Proc. Natl. Acad. Sci. U.
S. A. 2001, 98, 5992–5996. S
Shape Memory Metals

33. Wen, X.; Wang, X.; Zhang, N. Microrough surface of


metallic biomaterials: A literature review. Biomed. Mater.
Eng. 1996, 6, 173–189.

34. Zhao, X.K.; Cai, W.; Tian, Y.; Zhao, L. Microstructure


and hemocompatibility of a phosphorus ion-implanted TiNi
shape-memory alloy. Mater. Sci. Forum 2002, 394–395,
153–156. (Shape Memory Materials and Its Applications). 35.
Steegmueller, R.; Wagner, C.; Fleckenstein, T.; Schuessler,
A. Gold coating of nitinol devices for medical
applications. Mater. Sci. Forum 2002, 394–395, 161–164.
(Shape Memory Materials and Its Applications). 36. Riepe,
G.; Heintz, C.; Kaiser, E.; Chakfe, N.; Morlock, M.;
Delling, M.; Imig, H. What can we learn from explanted
endovascular devices? Eur. J. Vasc. Endovasc. Surg. 2002,
24 (2), 117–122.
Silicones

28. Thomas, D.R. Siloxane Polymers; Clarson, S.J., Semlyen,


J.A., Eds.; Prentise Hall: Englewood, NJ, 1993; 567–615.

29. Rich, J.; Cella, J.; Lewis, L.; Stein, J.; Singh, N.;
Rubinsztajn, S.; Wengrovius, J. Kirk-Othmer Encyclopedia of
Chemical Technology, 5th Ed.; Kroschwitz, J.I., Howe-Grant,
M., Eds.; John Wiley & Sons, Inc.: New York, 1997; Vol. 22,
82–142.

30. Wynne, K.J.; Ho, T.; Johnston, E.E.; Myers, S.A.


Surface science and stability of networks prepared from
hydroxy-terminated polydimethylsiloxane and
methyltriethoxysilane. Appl. Organomet. Chem. 1998, 12,
763–770.

31. Viers, B.D.; Sukumaran, S.; Beaucage, G.; Mark, J.E. On


the observation of novel morphologies in ’’model’’ PDMS
elastomers—Evidence of trapped spinodal structures. Polym.
Preprints 1997, 38, 333–334.

32. Uilk, J.; Bullock, S.; Johnston, E.; Myers, S.A.;


Merwin, L.H.; Wynne, K.J. Surface science of elastomeric
coatings prepared from alpha, omega-dihydroxypoly
(dimethylsiloxane) and the ethoxysiloxane mixture ‘‘ES40’’.
Macromolecules 2000, 33, 8791–8801.

33. Bondurant, S.; Ernster, V.; Herdman, R. Implant


Catalogue. In Safety of Silicone Breast Implants;
Bondurant, S., Ernster, V., Herdman, R., Eds.; National
Academy Press: Washington, DC, 1999; 54–79.

34. Gutowski, K.A.; Mesna, G.T.; Cunningham, B.L.


Salinefilled breast implants: A plastic surgery educational
foundation multicenter outcomes study. Plast. Reconstr.
Surg. 1997, 100, 1019–1027.

35. Mladick, R.A. No-touch submuscular saline breast


augmentation technique. Aesthet. Plast. Surg. 1993, 17,
183–192.

36. Mallam, S.; Hecht, A.-M.; Geissler, E.; Pruvost, P.


Structure of swollen polydimethylsiloxane gels. J. Chem.
Phys. 1989, 91, 6447–6454.

37. Cochrane, H.; Lin, C.S. The influence of fumed silica


properties on the processing, curing, and reinforcement
properties of silicone-rubber. Rubber Chem. Technol. 1993,
66, 48–60.
38. Kwan, K.S.; Harrington, D.A.; Moore, P.A.; Hahn, J.R.;
Degroot, J.V.; Burns, G.T. Synthesis and use of colloidal
silica for reinforcement in silicone elastomers. Rubber
Chem. Technol. 2001, 74, 630–644.

39. Rajan, G.S.; Sur, G.S.; Mark, J.E.; Schaefer, D.W.;


Beaucage, G. Preparation and characterization of some
unusually transparent poly(dimethylsiloxane)
nanocomposites. J. Polym. Sci., Part B, Polym. Phys. 2003,
41, 1897–1901.

40. Barthel, H.; Rosch, L.; Weis, J. Fumed


Silica—Production, Properties, and Applications. In
Organosilicon Chemistry II: From Molecules to Materials;
Auner, N., Weis, J., Eds.; VCH Publishers: Weinheim, 1996;
761–778.

41. Mathias, J.; Wannemacher, G. Basic characteristics and


applications of aerosil 30. The chemistry and physics of
the aerosil surface. J. Colloid Interface Sci. 1988, 125,
61–68.

42. Hertl, W.; Hair, M.L. Reaction of hexamethyldisilazane


with silica. J. Phys. Chem. 1971, 75, 2181–2185.

43. Gun’ko, V.M.; Vedamuthu, M.S.; Henderson, G.L.; Blitz,


J.P. Mechanism and kinetics of hexamethyldisilazane
reaction with a fumed silica surface. J. Colloid Interface
Sci. 2000, 228, 157–170. 44. Boonstra, B.B.; Cochrane, H.;
Dannenberg, E.M. Reinforcement of silicone rubber by
particulate silica. Rubber Chem. Technol. 1975, 48,
558–576. 45. Lucas, G. Process of Treating Silica with a
Siloxane and Product Thereof. U.S. 2,938,009, 1960. 46.
Kobayashi, H.; Ohnishi, M. Method for Modifying the Surface
of Finely Divided Silica. US 4,849,022, 1989. 47. Brown,
E.D.; Hyde, J.F. Process for Rendering Inorganic Powders
Hydrophobic. U.S. 3,334,062, 1967. 48.
http://w1.cabot-corp.com/controller.jsp?N=23+1001+
4294967082+4294967073&entry=product (accessed August 2003).
49. http://www2.sivento.de/sivento/silica/html/e/solutions_
products/datasheet/silica_datasheet.asp?id=44& definition=2
(accessed August 2003). 50.
http://www2.sivento.de/sivento/silica/html/e/solutions_
products/datasheet/silica_datasheet.asp?id=518 &
definition=2 (accessed August 2003). 51.
http://www2.sivento.de/sivento/silica/html/e/solutions_
products/datasheet/silica_datasheet.asp?id=538& definition=2
(accessed August 2003). 52.
http://www2.sivento.de/sivento/silica/html/e/solutions_
products/datasheet/silica_datasheet.asp?id= 57&definition=2
(accessed August 2003). 53.
http://www2.sivento.de/sivento/silica/html/e/solutions_
products/datasheet/silica_datasheet.asp?id= 46&definition=2
(accessed August 2003). 54.
http://www2.sivento.de/sivento/silica/html/e/solutions_
products/datasheet/silica_datasheet.asp?id =522&
definition=2 (accessed August 2003). 55.
http://www.wacker.com/internet/webcache/en_US/
PTM/TM/Wacker_HDK/Hydrophob/WACKER_ HDK_H15.pdf (accessed
August 2003). 56.
http://www.wacker.com/internet/webcache/en_US/
PTM/TM/Wacker_HDK/Hydrophob/WACKER_ HDK_H18.pdf (accessed
August 2003). 57.
http://www.wacker.com/internet/webcache/en_US/
PTM/TM/Wacker_HDK/Hydrophob/WACKER_ HDK_H20.pdf (accessed
August 2003). 58.
http://www.wacker.com/internet/webcache/en_US/
PTM/TM/Wacker_HDK/Hydrophob/WACKER_ HDK_H30.pdf (accessed
August 2003). 59. Burnside, S.D.; Giannelis, E.P. Synthesis
and properties of new poly(dimethylsiloxane)
nanocomposites. Chem. Mater. 1995, 7, 1597–1600. 60. Hron,
P. Hydrophilisation of silicone rubber for medical
applications. Polym. Int. 2003, 52, 1531–1539. 61. Abbasi,
F.; Mirzadeh, H.; Katbab, A.A. Modification of polysiloxane
polymers for biomedical applications: A review. Polym. Int.
2001, 50, 1279–1287. 62. Gunatillake, P.A.; Martin, D.J.;
Meijs, G.F.; McCarthy, S.J.; Adhikari, R. Designing
biostable polyurethane elastomers for biomedical implants.
Aust. J. Chem. 2003, 56, 545–557. 63. Lamba, N.M.K.;
Woodhouse, K.A.; Cooper, S.L. Polyurethanes in Biomedical
Applications; CRC Press: Boca Raton, FL, 1998; 15. 64.
Yilgor, E.; McGrath, J.E. Polysiloxane containing
copolymers: A survey of recent developments. Adv. Polym.
Sci. 1988, 86, 1–86. 65. Garrett, J.T.; Runt, J.; Lin, J.S.
Microphase separation of segmented poly(urethane urea)
block copolymers. Macromolecules 2000, 33, 6353–6359. 66.
Yilgo¨r, E.; Yilgo¨r, I. Hydrogen bonding: A critical
parameter in designing silicone copolymers. Polymer 2001,
42, 7953–7959. 67. Pathiraja, A.; Gunatillake, S.J.;
McCarthy, G.F.; Adhikari, M.; Adhikari, R.
Siloxane-containing polyurethaneurea compositions, AU
PCT/AU00/00345; 2000. 68. Bernacca, G.M.; Straub, I.;
Wheatley, D.J. Mechanical and morphological study of
biostable polyurethane heart valve leaflets explanted from
sheep. J. Biomed. Mater. Res. 2002, 61, 138–145. 69.
Hergenrother, R.W.; Yu, X.-H.; Cooper, S.L. Bloodcontacting
properties of polydimethylsiloxane polyureaurethanes.
Biomaterials 1994, 15, 635–640. 70. Takahara, A.;
Hergenrother, R.W.; Coury, A.J.; Cooper, S.L. In vitro
evaluation of the biostability of segmented polyurethanes.
Artif. Organs 1990, 14, 87–90. 71. Chen, Z.; Ward, R.;
Tian, Y.; Malizia, F.; Gracias, D.H.; Shen, Y.R.; Somorjai,
G.A. Interaction of fibrinogen with surfaces of
end-group-modified polyurethanes: A surface-specific
sum-frequency-generation vibrational spectroscopy study. J.
Biomed. Mater. Res. 2002, 62, 254–264. 72. Ho, T.; Wynne,
K.J. Mechanism and kinetics of hexamethyldisilazane
reaction with a fumed silica surface blends of a
siloxane/urea/urethane-segmented copolymer with an
IPDI-based polyetherurethane. Polym. Adv. Technol. 1994, 6,
25–31. 73. White, K.A.; Ward, R.S.; Gill, R.S.; Lim, F.;
Coviello, S.K. Surface Modification of Polymeric
Biomaterials; Ratner, B.D., Castner, D.G., Eds.; Plenum:
NY, 1997; 27–33. 74. Amitay-Sadovsky, E.; Komvopoulos, K.;
Ward, R.; Somorjai, G.A. Surface reordering of stretched
polyurethane block copolymer films studied by sum frequency
generation vibrational spectroscopy. J. Phys. Chem., B
2003, 107, 6377–6383. 75. Waters, M.; Jagger, R. Improved
wettability of an experimental silicone rubber denture soft
lining material. J. Biomed. Mater. Res. 1999, 48, 765–771.
76. Waters, M.G.J.; Jagger, R.G.; Polyzois, G.L.
Wettability of silicone rubber maxillofacial prosthetic
materials. J. Prosthet. Dent. 1999, 81, 439–443. 77.
Everaert, E.P.; Vandermei, H.C.; Devries, J.; Busscher,
H.J. Hydrophobic recovery of repeatedly plasma-treated
silicone-rubber. 1. Storage in air. J. Adhes. Sci. Technol.
1995, 9, 1263–1278. 78. Kennan, J.J.; Peters, Y.A.;
Swarthout, D.E.; Owen, M.J.; Namkanisorn, A.; Chaudhury,
M.K. Effect of saline exposure on the surface and bulk
properties of medical grade silicone elastomers. J. Biomed.
Mater. Res. 1997, 36 , 487–497. 79. Batich, C.; DePalma,
D.; Marotta, J.; Latorre, G.; Hardt, N.S. Silicone
degradation reactions. Immunol. Silicones 1996, 210, 13–23.
80. Tingey, K.G.; Andrade, J.D. Probing surface
microheterogeneity of poly(ether urethanes) in an aqueous
environment. Langmuir 1991, 7, 2471–2478. 81. Tretinnikov,
O.N.; Ikada, Y. Dynamic wetting and contact-angle
hysteresis of polymer surfaces studies with the modified
Wilhelmy balance method. Langmuir 1994, 10, 1606–1614. 82.
Makal, U.; Wynne, K.J. Unpublished Results. 83. Katano, Y.;
Tomono, H.; Nakajima, T. Surface property of polymer films
with fluoroalkyl side chains. Macromolecules 1994, 27,
2342–2344. 84.
http://www.fda.gov/opacom/morechoices/industry/ guidedc.htm
(accessed August 2003). 85.
http://www.fda.gov/cdrh/international/nonusregulators. html
(accessed August 2003). 86.
http://www.fda.gov/cdrh/ode/guidance/1354.html (accessed
August 2003). 87.
http://www.iso.ch/iso/en/ISOOnline.frontpage. 88. Atkins,
D.C.; Murphy, C.M.; Saunders, C.E. Polymethylsiloxanes:
Thermal and oxidation stabilities. Ind. Eng. Chem. 1947, 39
(11), 1395–1401. 89. Baker, F.S.; Miller, C.E.; Repik,
A.J.; Tolles, E.D. Activated Carbon. In Kirk-Othmer
Encyclopedia of Chemical Technology, 4th Ed.; Kroschwitz,
J.I., Howe-Grant, M., Eds.; John Wiley & Sons, Inc.: New
York, 1997; Vol. 4, 1015–1037. 90. Gilbert, A.K.K.S.W.
Transient catalysts for the polymerization of
organosiloxanes. J. Polym. Sci., Polym. Lett. 1959, 40,
35–58. 91. McMillin, C.R. Elastomers for biomedical
applications. Abstr. Papers Am. Chem. Soc. 1994, 67,
417–446. 92. Yoda, R. Elastomers for biomedical
applications. J. Biomater. Sci., Polym. Ed. 1998, 9,
561–626. 93. Brandon, H.J.; Jerina, K.L.; Wolf, C.J.;
Young, V.L. Biodurability of retrieved silicone gel breast
implants. Plast. Reconstr. Surg. 2003, 111, 2295–2306. S
Silks

3. Demura, M.; Asakura, T. Immobilization of glucose


oxidase with Bombyx mori silk fibroin by only stretching
treatment and its application to glucose sensor.
Biotechnol. Bioeng. 1989, 33, 598–603.

4. Sofia, S.; McCarthy, M.B.; Gronowicz, G.; Kaplan, D.L.


Functionalized silk-based biomaterials for bone formation.
J. Biomed. Mater. Res. 2001, 54 (1), 139–148.

5. Hojo, N., Ed. Zoku Kenshi no Kozo (Structure of Silk


Fibers); Shinshu University: Ueda, Japan, 1980.

6. Zhou, C.-Z.; Confalonieri, F.; Medina, N.; Zivanovic,


Y.; Esnault, C.; Yang, T.; Jacquet, M.; Janin, J.; Duguet,
M.; Perasso, R.; Li, Z.-G. Fine organization of Bombyx mori
fibroin heavy chain gene. Nucleic Acids Res. 2000, 28 (12),
2413–2419.

7. Asakura, T.; Sugino, R.; Yao, J.; Takashima, H.;


Kishore, R. Structural analysis of semi-crystalline Bombyx
mori silk fibroin chain with Silk I and Silk II forms by 13
C, 15 N and 2 H stable isotope labeling,
conformation-dependent chemical shifts and solid state NMR
spectroscopy. Biochemistry 2002, 41 (13), 4415–4424.

8. Asakura, T.; Ashida, J.; Yamane, T.; Kameda, T.;


Nakazawa, Y.; Ohgo, K.; Komatsu, K. A repeated b-turn
structure in poly (Ala-Gly) as a model for silk I of Bombyx
mori silk fibroin studied with two-dimensional
spin-diffusion NMR under off magic angle spinning and
rotational echo double resonance. J. Mol. Biol. 2001, 306
(2), 291–305.

9. Marsh, R.E.; Corey, R.B.; Pauling, L. An investigation


of the structure of silk fibroin. Biochim. Biophys. Acta
1955, 16, 1–34.

10. Asakura, T.; Yao, J.; Yamane, T.; Umemura, K.; Ulrich,
A.S. Heterogeneous structure of silk fibers from Bombyx mori
resolved by 13 C solid-state NMR spectroscopy. J. Am. Chem.
Soc. 2002, 124, 8794–8795.

11. Iizuka, E. Silk thread: Mechanism of spinning and its


mechanical properties. J. Appl. Polym. Sci. 1985, 41,
173–185.

12. Magoshi, J.; Magoshi, Y.; Nakamura, S. Crystallization,


liquid crystal, and fiber formation of silk fibroin. J. Appl.
Polym. Sci. 1985, 41, 187–204.

13. Yamane, T.; Umemura, K.; Nakazawa, Y.; Asakura, T.


Molecular dynamics simulation of conformational change of
poly(Ala-Gly) from repeated b-turn type II to b-sheet in
relation to fiber formation mechanism of Bombyx mori silk
fibroin. Macromolecules 2003, 36, 6766–6772.

14. Altman, G.H.; Diaz, F.; Jakuba, C.; Calabro, T.; Horan,
R.L.; Chen, J.; Lu, H.; Richmond, J.; Kaplan, D.L.
Silk-based biomaterials. Biomaterials 2003, 24 (3),
401–416.

15. Wen, C.M.; Ye, S.T.; Zhou, L.X.; Yu, Y. Silk-induced


asthma in children: A report of 64 cases. Ann. Allergy
1990, 65 (5), 375–378.

16. Zaoming, W.; Codina, R.; Fernandez-Caldas, E.; Lockey,


R.F. Partial characterization of the silk allergens in
mulberry silk extract. J. Investig. Allergol. Clin.
Immunol. 1996, 6 (4), 237–241. 17. Setzen, G.; Williams,
E.F. Tissue response to suture materials implanted
subcutaneously in a rabbit model. Plast. Reconstr. Surg.
1997, 100 (7), 1788–1795. 18. Santin, M.; Motta, A.;
Freddi, G.; Cannas, M. In vitro evaluation of the
inflammatory potential of the silk fibroin. J. Biomed. Mater.
Res. 1999, 46 (3), 382–389. 19. Asakura, T.; Kitaguchi, M.;
Demura, M.; Sakai, H.; Komatsu, K. Immobilization of
glucose oxidase on nonwoven fabrics with Bombyx mori silk
fibroin gel. J. Appl. Polym. Sci. 1992, 46, 49–53. 20.
Demura, M.; Asakura, T.; Nakamura, E.; Tamura, H.
Immobilization of peroxidase with a Bombyx mori silk fibroin
membrane and its application to biophotosensors. J.
Biotech. 1989, 10, 113–119. 21. Qian, J.; Y.; Liu, H.; Yu,
T.; Deng, J. An amperometric new methylene blue N-mediating
sensor for hydrogen peroxide based on regenerated silk
fibroin as an immobilization matrix for peroxidase. Anal.
Biochem. 1996, 236 (2), 208–214. 22. Inouye, K.; Kurokawa,
M.; Nishikawa, S.; Tsukada, M. Use of Bombyx mori silk
fibroin as a substratum for cultivation of animal cells. J.
Biochem. Biophys. Methods 1998 , 37 (3), 159–164. 23.
Furuzono, T.; Taguchi, T.; Kishida, A.; Akashi, M.; Tamada,
Y. Preparation and characterization of apatite deposited on
silk fabric using an alternate soaking process. J. Biomed.
Mater. Res. 2000, 50 (3), 344–352. 24. Krejchi, M.T.;
Atkins, E.D.T.; Waddon, A.J.; Fournier, M.J.; Mason, T.L.;
Tirrell, D.A. Chemical sequence control of b-sheet assembly
in macromolecular crystals of periodic polypeptides.
Science 1994, 265 (5177), 1427–1432. 25. Cappello, J.;
Ferrari, F. Microbial Production of Structural Protein
Polymers. In Plastics from Microbes; Mobley, D.P., Ed.;
Hanser: New York, 1994; 35–92. 26. Asakura, T.; Tanaka, C.;
Yang, M.; Yao, J.; Kurokawa, M. Production and
characterization of a silk-like hybrid protein, based on
the polyalanine region of Samia cynthia ricini silf fibroin
and a cell adhesive region derived from fibronectin.
Biomaterials 2004, 25, 617–624. 27. Yao, J.; Asakura, T.
Synthesis and structural characterization of silk-like
materials incorporated with elastic motif. J. Biochem.
2003, 133, 147–154. 28. van Beek, J.D.; Beaulieu, L.;
Schafer, H.; Demura, M.; Asakura, T.; Meier, B.H.
Solid-state NMR determination of the secondary structure of
Samia cynthia ricini silk. Nature 2000, 405 (6790),
1077–1079. 29. Simmons, A.; Michal, C.; Jelinski, L.
Molecular orientation and two-component nature of the
crystalline fraction of spider dragline silk. Science 1996,
271 (5245), 84–87. 30. Hayashi, C.Y.; Lewis, R.V. Evidence
from flagelliform silk cDNA for the structural basis of
elasticity and modular nature of spider silks. J. Mol.
Biol. 1998, 275 (5), 773–784. S
Small-Angle Light Scattering Methods for
Soft Connective Tissue Structural
Analysis

7. Liu, Z.Q.; Rangayyan, R.M.; Frank, C.B. Statistical


analysis of collagen alignment in ligaments by scalespace
analysis. IEEE Trans. Biomed. Eng. 1991, 38, 580–587.

8. Kastelic, J.; Baer, E. Deformation of Tendon Collagen.


In The Mechanical Properties of Biological Materials;
Vincient, J.F., Currey, J.D., Eds.; Society for
Experimental Biology: Great Britain, 1980; 397–433.

9. Purslow, P.P.; Bigi, A.; Ripamonti, A.; Roveri, N.


Collagen fibre reorientation around a crack in biaxially
stretched materials. Int. J. Macromol. 1984, 6, 21–25.

10. Sasaki, N.; Odajima, S. Stress–strain curve and young’s


modulus of a collagen molecule as determined by the x-ray
diffraction technique. J. Biomech. 1996, 29, 655–658.

11. Baer, E.; Cassidy, J.J.; Hiltner, A. Hierarchical


Structure of Collagen and Its Relationship to the Physical
Properties of Tendon. In Collagen; Nimni, M.E., Ed.; CRC
Press: Boca Raton, 1988; 177–199.

12. Hilbert, S.L.; Ferrans, V.J.; Swanson, W.M. Optical


methods for the nondestructive evaluation of collagen
morphology in bioprosthetic heart valves. J. Biomed. Mater.
Res. 1986, 20, 1411–1421.

13. Tower, T.T.; Tranquillo, R.T. Alignment maps of


tissues: I. Microscopic elliptical polarimetry. Biophys. J.
2001, 81 (5), 2954–2963.

14. Tower, T.T.; Tranquillo, R.T. Alignment maps of


tissues: II. Fast harmonic analysis for imaging. Biophys.
J. 2001, 81 (5), 291f64–2971.

15. Borch, J.; Sundarajan, P.R.; Marchessault, R.H. Light


scattering by cellulose. Iii-Morphology of wood. J. Polym.
Sci. 1971, 9, 313–329.

16. Muggli, R.; Marton, R. Light scattering by cellulose.


V.-Anisotropy scattering by wood fibers. J. Polym. Sci.
1971, 36, 121–139.

17. Raman, C.V.; Bhat, M.R. The structure and optical


behavior of some natural and synthetic fibers. Proc. Acad.
Sci. 1954, A40, 109–116.
18. Stein, R.S.; Erhardt, P.; van Aartsen, J.J.; Clough, S.
Theory of light scattering from oriented and fiber
structures. J. Polym. Sci. 1966, 13, 1–35.

19. Stein, R.S.; Wilson, P.R. Scattering of light by


polymer films possessing correlated orientation fluctuation.
J. Appl. Physiol. 1962, 33, 1914–1922.

20. Chien, J.C.W.; Chang, E.P. Small-angle light scattering


of reconstituted collagen. Macromolecules 1972, 5, 610–617.

21. Moritani, M.; Hayashi, N.; Utsuo, A.; Kawai, H.


Light-scattering patterns from collagen films in relation to
the texture of a random assembly of anisotropic rods in
three dimensions. Polym. J. 1971, 2, 74–87.

22. Kronick, P.L.; Buechler, P.R. Fiber orientation in


calfskin by laser light scattering or x-ray diffraction and
quantitative relation to mechanical properties. J. Am.
Leather Chem. Assoc. 1986, 81, 221–229.

23. Kronick, P.L.; Sacks, M.S.; Dahms, M. Vertical fiber


defect quantified by small angle light scattering. Connect.
Tissue Res. 1991, 27, 1–13.

24. Ferdman, A.G.; Yannas, I.V. Scattering of light from


histologic sections: A new method for the analysis of
connective tissue. J. Invest. Dermatol. 1993, 100, 710–716.
25. McCally, R.L.; Farrell, R.A. Small-angle light
scattering and birefringence properties of chick cornea. J.
Refract. Surg. 1999, 15 (6), 706–710. 26. McCally, R.L.;
Farrell, R.A. Structural implications of small-angle light
scattering from cornea. Exp. Eye Res. 1982, 34 (1), 99–113.
27. Andreo, R.H.; Farrell, R.A. Corneal small-angle
lightscattering theory: Wavy fibril models. J. Opt. Soc. Am.
1982, 72 (11), 1479–1492. 28. Bettelheim, F.A.; Kumbar, M.
An interpretation of small-angle light-scattering patterns
of human cornea. Investig. Ophthalmol. Vis. Sci. 1977, 16
(3), 233–236. 29. Bettelheim, F.A.; Magrill, R. Small-angle
lightscattering patterns of corneas of different species.
Investig. Ophthalmol. Vis. Sci. 1977, 16 (3), 236–240. 30.
Bettelheim, F.A.; Kaplan, D. Small angle light scattering
of bovine cornea as affected by birefringence. Biochim.
Biophys. Acta 1973, 313 (2), 268–276. 31. Sacks, M.S.;
Chuong, C.J. Characterization of collagen fiber architecture
in the canine central tendon. J. Biomech. Eng. 1992, 114,
183–190. 32. Sacks, M.S.; Chuong, C.J.; More, R. Collagen
fiber architecture of bovine pericardium. ASAIO 1994, 40,
M632–M637. 33. Hiester, E.D.; Sacks, M.S. Optimal bovine
pericardial tissue selection sites. Ii. Cartographic
analysis. J. Biomed. Mater. Res. 1998, 39 (2), 215–221. 34.
Hiester, E.D.; Sacks, M.S. Optimal bovine pericardial
tissue selection sites. I. Fiber architecture and tissue
thickness measurements. J. Biomed. Mater. Res. 1998, 39
(2), 207–214. 35. Hamann, M.C.; Sacks, M.S.; Malinin, T.I.
Quantification of the collagen fibre architecture of human
cranial dura mater. J. Anat. 1998, 192 (Pt. 1), 99–106. 36.
Sacks, M.S.; Chuong, C.J.; Petroll, W.M.; Kwan, M.;
Halberstadt, C. Collagen fiber architecture of a cultured
dermal tissue. J. Biomech. Eng. 1997 , 119 (1), 124–127.
37. Sacks, M.S.; Gloeckner, D.C. Quantification of the fiber
architecture and biaxial mechanical behavior of porcine
intestinal submucosa. J. Biomed. Mater. Res. 1999, 46,
1–10. 38. Gloeckner, D.C.; Sacks, M.S.; Billiar, K.L.;
Bachrach, N. Mechanical evaluation and design of a
multilayered collagenous repair biomaterial. J. Biomed.
Mater. Res. 2000, 52 (2), 365–373. 39. Sacks, M.S.; Smith,
D.B.; Hiester, E.D. The aortic valve microstructure:
Effects of transvalvular pressure. J. Biomed. Mater. Res.
1998, 41 (1), 131–141. 40. Sacks, M.S.; Smith, D.B. Effects
of accelerated testing on porcine bioprosthetic heart valve
fiber architecture. Biomaterials 1998, 19 (11–12),
1027–1036. 41. Sacks, M.S.; Schoen, F.J. Collagen fiber
disruption occurs independent of calcification in clinically
explanted bioprosthetic heart valves. J. Biomed. Mater.
Res. 2002, 62 (3), 359–371. 42. Kunzelman, K.S.; Sacks,
M.S.; Cochran, R.P.; Eberhart, R.C. Mitral valve leaflet
collagen distribution by laser analysis. Seventh South.
Biomed. Eng. Conf. 1988, 82–85. Light Scattering 43. Bowes,
L.E.; Jimenez, M.C.; Hiester, E.D.; Sacks, M.S.;
Brahmatewari, J.; Mertz, P.; Eaglstein, W.H. Collagen fiber
orientation as quantified by small angle light scattering in
wounds treated with transforming growth factor-beta2 and
its neutralizing antibody. Wound Repair Regen. 1999, 7 (3),
179–186. 44. Chuong, C.J.; Sacks, M.S.; Johnson, R.L.;
Reynolds, R.C. On the anisotropy of the diaphragmatic
central tendon. J. Biomech. 1991, 24, 563–576. 45. Billiar,
K.L.; Sacks, M.S. A method to quantify the fiber kinematics
of planar tissues under biaxial stretch. J. Biomech. 1997,
30 (7), 753–756. 46. Sacks, M.S. Incorporation of
experimentally-derived fiber orientation into a structural
constitutive model for planar collagenous tissues. J.
Biomech. Eng. 2003, 125 (2), 280–287. 47. Sacks, M.S.;
Chuong, C.J. Orthotropic mechanical properties of
chemically treated bovine pericardium. Ann. Biomed. Eng.
1998, 26 (5), 892–902. 48. Sacks, M.S.; Smith, D.B.;
Hiester, E.D. A small angle light scattering device for
planar connective tissue microstructural analysis. Ann.
Biomed. Eng. 1997, 25 (4), 678–689. 49. Marshall, G.E.
Gaussian Laser Beam Diameters and Divergence. In Optical
Scanning; Marshall, G.E., Ed.; Marcel Dekker: New York,
1991; 1–11. 50. Halliday, D.; Resnick, R. Physics; John
Wiley and Sons: New York, 1960. 51. Guinier, A.; Fournet,
G. Small Angle Scattering of X-Rays; Wiley: New York, 1955.
52. Mendenhall, W.; Sincich, T. Statistics for the
Engineering and Computer Sciences; Dellen: San Francisco,
1988. 53. Cowley, J.M. Principles of Image Formation. In
Introduction to Analytical Electron Microscopy; Hren, J.J.,
Goldstein, J.I., Joy, D.C., Eds.; Plenum Press: New York,
1979; 1–42. 54. Press, W.H.; Flannery, B.P.; Teukolsky,
S.A.; Vetterling, W.T. Numerical Receipes in C; Cambridge
University Press: Cambridge, 1988. S Tissue Structural
Analysis
Spine Biomechanics

13. Kumaresan, S.; Yoganandan, N.; Pintar, F.A. Finite


element modeling approaches of human cervical spine facet
joint capsule. J. Biomech. 1998, 31, 371–376.

14. Linde, F. Elastic and viscoelastic properties of


trabecular bone by a compression testing approach. Dan.
Med. Bull. 1994, 41 (2), 119–138.

15. Shirazi-Adl, S.A.; Suresh, C.S.; Ahmed, A.M. Stress


analysis of the lumbar disc-body unit in compression. Spine
1984, 9 (2), 120–132.

16. Adams, M.A.; Dolan, P. Spine biomechanics. J. Biomech.


2005, 38 (10), 1972–1983.

17. Stokes, I.A.; Gardner-Morse, M.; Churchill, D.; Laible,


J.P. Measurement of a spinal motion segment stiffness
matrix. J. Biomech. 2002, 35 (4), 517–521.

18. Thompson, R.E.; Barker, T.M.; Pearcy, M.J. Defining the


neutral zone of sheep intervertebral joints during dynamics
motions: an in vitro study. Clin. Biomech. 2003, 18 (2),
89–98.

19. Evans, J.H. Clinical biomechanics of the spine. In


Biomechanics in Orthopedics; Niwa, S., Perren, S.M.,
Hattori, T., Eds.; Springer-Verlag: Tokyo, 1992; 31–45.

20. Tibrewal, S.B.; Pearcy, M.J. Lumbar intervertebral disc


heights in normal subjects and patientswith disc
herniation. Spine 1985, 10 (5), 452–454.

21. Urban, J.P.G.; Smith, S.; Fairbank, J.C.T. Nutrition of


the intervertebral disc. Spine 2004, 29 (23), 2700–2709.

22. Thompson, R.E.; Pearcy, M.J.; Barker, T.M. The


mechanical effects of intervertebral disc lesions. Clin
Biomech. 2004, 19 (5), 448–455.

23. Shirazi-Adl, S.A.; Shrivastava, S.C.; Ahmed, A.M.


Stress analysis of the lumbar disc-body unitin compression:
a three-dimensional nonlinear finite element study. Spine
1984, 9 (2), 120–134.

24. Fagan, M.J.; Julian, S.; Siddall, D.J.; Mohsen, A.M.


Patient specific spine models. Part 1: Finite element
analysis of the lumbar intervertebral disc–a material
sensitivity study. Proc. Inst. Mech. Eng. 2002, 216H,
299–314.

25. Ueno, K.; Liu, Y.K. A three-dimensional nonlinear finite


element model of lumbar intervertebral joint in torsion. J.
Biomech. Eng. 1987, 109, 200–209.

26. Pearcy, M.J.; Evans, J.H.; O’Brien, J.P. The load


bearing capacity of vertebral cancellous bone in interbody
fusion of the lumbar spine. Eng. Med. 1983, 12 (4),
183–184.

27. Adam, C.J.; Pearcy, M.J.; McCombe, P. Stress analysis


of interbody fusion–finite element modeling of
intervertebral implant and vertebral body. Clin. Biomech.
2003, 18, 265–272.

28. Carson, W.L.; Asher, M.A. Parameters influencing spine


implant construct axial-flexion stiffness. Proceedings of
the Harrington Spine Symposium, Kansas City, Kansas, July
28–30, 2005; Asher, M.A., Ed.; University of Kansas Medical
Center: Kanas, 2005; 83 pp.

29. Hilibrand, A.S; Robbins, M. Adjacent segment


degeneration and adjacent segment disease: the consequences
of spinal fusion? Spine 2004, 4 (6 suppl), 190S–194S.

30. Huang, R.C.; Girardi, F.P.; Cammisa, F.P.; Wright, T.M.


The implications of constraint in lumbar total disc
replacement. J. Spinal Disord. Tech. 2003, 16 (4), 412–417.
31. Huang, R.C.; Wright, T.M.; Panjabi, M.M.; Lipman, J.D.
Biomechanics of nonfusion implants. Orthop. Clin. N. Am.
2005, 36, 271–280. 32. German, J.W.; Foley, K.T. Disc
arthroplasty in the management of the painful lumbar motion
segment. Spine 2005, 30, S60–S67. 33. Langrana, N.A.;
Harten, R.D.; Lin, D.C.; Reiter, M.F.; Lee, C.K. Acute
thoracolumbar burst fractures: a new view of loading
mechanisms. Spine 2002, 27 (5), 498–508. 34. Shirado, O.;
Kaneda, K.; Tadano, S.; Ishikawa, H.; McAfee, P.C.; Warden,
K.E. Influence of disc degeneration on mechanism of
thoracolumbar burst fractures. Spine 1992, 17 (3), 286–292.
35. Whyne, C.M.; Hu, S.S.; Lotz, J.C. Burst fracture in the
metastatically involved spine: development, validation, and
parametric analysis of a three-dimensional poroelastic
finite-element model.Spine 2003, 28 (7), 652–660. 36.
Huiskes, R.; Ruimerman, R.; van Lenthe, G.H.; Janssen, J.D.
Effects of mechanical forces on maintenance and adaptation
of form in trabecular bone. Nature 2000, 405 (6787),
704–706. 37. Nalla, R.K.; Kruzic, J.J.; Kinney, J.H.;
Ritchie, R.O. Aspects of in vitro fatigue in human cortical
bone: time and cycle dependent crack growth. Biomaterials
2005, 26 (14), 2183–2195. 38. Silva, M.J.; Gibson, L.J.
Modeling the mechanical behavior of vertebral trabecular
bone: effects of agerelated changes in microstructure. Bone
1997, 21 (2), 191–199. 39. Wilcox, R.K. The biomechanics of
vertebroplasty: a review.Proc. Inst. Mech. Eng. H 2004, 218
(1), 1–10. 40. Stokes, I.A.F. How vicious is the ‘vicious
cycle’? Proceedings of the International Research Society
of Spinal Deformities Symposium, Vancouver, Canada, June
10–12, 2004; Sawatsky, B.J., Ed.; International Research
Society of Spinal Deformities: Vancouver, 2004; 331 pp. 41.
Millner, P.A.; Dickson, R.A. Idiopathic scoliosis:
biomechanics and biology. Eur. Spine J. 1996, 5 (6),
362–373. 42. Ogilvie, J.W. Biomechanics. In Moe’s Textbook
of Scoliosis and Other Spinal Deformities; Bradford, D.S.,
Lonstein, J.E., Moe, J.H., Ogilvie, J.W., Winter, R.B.,
Eds.; W.B. Saunders Company: Philadelphia, 1987; 7–24. 43.
Goto, M.; Kawakami, N.; Azegami, H.; Matsuyama, Y.;
Takeuchi, K.; Sasaoka, R. Buckling and bone modeling as
factors in the development of idiopathic scoliosis. Spine
2003, 28 (4), 364–370. 44. Villemure, I.; Aubin, C.E.;
Dansereau, J. Biomechanical simulations of the spine
deformation process in adolescent idiopathic scoliosis from
different pathogenesis hypotheses. Eur. Spine J. 2004, 13
(1), 83–90. 45. Perie, D.; Aubin, C.E.; Petit, Y.
Personalized biomechanical simulations of orthotic
treatment in idiopathic scoliosis. Clin. Biomech. 2004, 19
(2), 190–195. 46. Panjabi, M.M.; Abumi, K.; Duranceau, J.;
Crisco, J.J. Biomechanical evaluation of spinal fixation
devices: II. Stability provided by eight internal fixation
devices. Spine 1988, 13 (10), 1135–1140.
Stem Cells

3. LaBarge, M.A.; Blau, H.M. Biological progression from


adult bone marrow to mononucleate muscle stem cell to
multinucleate muscle fiber in response to injury. Cell 2002,
111 (4), 589–601.

4. Theise, N.D.; Badve, S.; Saxena, R.; Henegariu, O.;


Sell, S.; Crawford, J.M.; Krause, D.S. Derivation of
hepatocytes from bone marrow cells in mice after
radiation-induced myeloablation. Hepatology 2000, 31 (1),
235–240.

5. Trounson, A. Human embryonic stem cells: Mother of all


cell and tissue types. Reprod. Biomed. Online 2002, 4
Suppl. 1, 58–63.

6. Department of Health and Human Services. Stem Cells:


Scientific Progress and Future Research Directions; June
2001. www.nih.gov/news/stemcell/scireport.htm.

7. Grisham, J.W.; Coleman, W.B.; Smith, G.J. Isolation,


culture, and transplantation of rat hepatocytic precursor
(stem-like) cells. Proc. Soc. Exp. Biol. Med. 1993, 204
(3), 270–279.

8. Coleman, W.B.; Smith, G.J.; Grisham, J.W. Development of


dexamethasone-inducible tyrosine aminotransferase activity
in WB-F344 rat liver epithelial stemlike cells cultured in
the presence of sodium butyrate. J. Cell. Physiol. 1994,
161 (3), 463–469.

9. Spees, J.L.; Olson, S.D.; Ylostalo, J.; Lynch, P.J.;


Smith, J.; Perry, A.; Peister, A.; Wang, M.Y.; Prockop,
D.J. Differentiation, cell fusion, and nuclear fusion
during ex vivo repair of epithelium by human adult stem
cells from bone marrow stroma. Proc. Natl. Acad. Sci. U. S.
A. 2003, 100 (5), 2397–2402.

10. Wang, X.; Montini, E.; Al-Dhalimy, M.; Lagasse, E.;


Finegold, M.; Grompe, M. Kinetics of liver repopulation
after bone marrow transplantation. Am. J. Pathol. 2002, 161
(2), 565–574.

11. Presnell, S.C.; Petersen, B.; Heidaran, M. Stem cells


in adult tissues. Semin. Cell Dev. Biol. 2002, 13 (5),
369–376.

12. Henningson, C.T., Jr.; Stanislaus, M.A.; Gewirtz, A.M.


Embryonic and adult stem cell therapy. J. Allergy Clin.
Immunol. 2003, 111 (2 Suppl.), S745–S753.

13. Kirby, S.L.; Bentley, S.; Fry, J.; Walton, W.; Desai,
N.; Latour, A.; Coleman, W.; Anderson, P.; Grisham, J.;
Malouf, N. Hematopoietic cell transdifferentiation of
cloned adult live-derived stem cells. Proc. Am. Soc. Cell
Biol. 2002, A680.

14. Boura, P.; Kountouras, J.; Lygidakis, N.J. Tumor


immunity and immunotherapy. Hepatogastroenterology 2001, 48
(40), 1040–1044.

15. Overturf, K.; al-Dhalimy, M.; Ou, C.N.; Finegold, M.;


Grompe, M. Serial transplantation reveals the stemcell-like
regenerative potential of adult mouse hepatocytes. Am. J.
Pathol. 1997, 151 (5), 1273–1280.

16. Overturf, K.; Al-Dhalimy, M.; Finegold, M.; Grompe, M.


The repopulation potential of hepatocyte populations
differing in size and prior mitotic expansion. Am. J.
Pathol. 1999, 155 (6), 2135–2143.

17. Haralson, M.A.; Hassell, J.R. The Extracellular


Matrix—An Overview. In Extracellular Matrix—A Practical
Approach; Haralson, M.A., Hassell, J.R., Eds.; Oxford
University Press: Oxford, 1995; 1–30. 18. Akiyama, S.K.;
LaFlamme, S.E. Bioadhesion and cell behavior. Colloids
Surf., B: Biointerfaces 1994, 2, 241–250. 19. Dexter, T.M.
Haemopoiesis in long-term bone marrow cultures. A review.
Acta Haematol. 1979, 62 (5–6), 299–305. 20. Rosenzweig, M.;
Pykett, M.; Marks, D.F.; Johnson, R.P. Enhanced maintenance
and retroviral transduction of primitive hematopoietic
progenitor cells using a novel three-dimensional culture
system. Gene Ther. 1997, 4, 928–936. 21. Banu, N.;
Rosenzweig, M.; Kim, H.; Bagley, J.; Pykett, M.
Cytokine-augmented culture of haematopoietic progenitor
cells in a novel three-dimensional cell growth matrix.
Cytokine 2001, 13 (6), 349–358. 22. Dominici, M.; Hofmann,
T.J.; Horwitz, E.M. Bone marrow mesenchymal cells:
Biological properties and clinical applications. J. Biol.
Regul. Homeost. Agents 2001, 15 (1), 28–37. 23. Martin, I.;
Padera, R.; Vunjak-Novakovic, G.; Freed, L.E. In vitro
differentiation of chick embryo bone marrow stromal cells
into cartilaginous and bonelike tissues. J. Orthop. Res.
1998, 16, 181–189. 24. Martin, I.; Shastri, V.P.; Padera,
R.F.; Yang, J.; Mackay, A.J.; Langer, R.; Vunjak-Novakovic,
G.; Freed, L.E. Selective differentiation of mammalian bone
marrow stromal cells cultured on three-dimensional polymer
foams. J. Biomed. Mater. Res. 2001, 55 (2), 229–235. 25.
Young, R.G.; Butler, D.L.; Weber, W.; Caplan, A.I.; Gordon,
S.L.; Fink, D.J. Use of mesynchymal stem cells in a
collagen matrix for Achilles tendon repair. J. Orthop. Res.
1998, 16, 406–413. 26. Awad, H.A.; Butler, D.L.; Harris,
M.T.; Ibrahim, R.E.; Wu, Y.; Young, R.G.; Kadiyala, S.;
Boivin, G.P. In vitro characterization of mesenchymal stem
cell-seeded collagen scaffolds for tendon repair: Effects
of initial seeding density on contraction kinetics. J.
Biomed. Mater. Res. 2000, 51 (2), 233–240. 27. Teng, Y.D.;
Lavik, E.B.; Qu, X.L.; Park, K.I.; Ourednik, J.;
Zurakowski, D.; Langer, R.; Snyder, E.Y. Functional
recovery following traumatic spinal cord injury mediated by
a unique polymer scaffold seeded with neural stem cells.
Proc. Natl. Acad. Sci. U. S. A. 2002, 99 (5), 3024–3029.
28. Park, K.I.; Teng, Y.D.; Snyder, E.Y. The injured brain
interacts reciprocally with neural stem cells supported by
scaffolds to reconstitute lost tissue. Nat. Biotechnol.
2002, 20 (11), 1111–1117. 29. Katz, A.J.; Llull, R.;
Hendrick, M.H.; Futrell, J.W. Emerging approaches to the
tissue engineering of fat. Clin. Plast. Surg. 1999, 26 (4),
587–603. 30. Kral, J.G.; Crandall, D.L. Development of a
human adipocyte synthetic polymer scaffold. Plast.
Reconstr. Surg. 1999, 104, 1732–1738. 31. Patrick, C.W.;
Chauvin, P.P.; Hobley, J.; Reece, G.P. Preadipocytes seeded
PLGA scaffolds for adipose tissue engineering. Tissue Eng.
1999, 5 (2), 139–151. 32. Hurd, D.D. Bone marrow
transplantation for cancer: An overview. Recent Results
Cancer Res. 1993, 132, 1–14. 33. Trofe, J.; Buell, J.F.;
First, M.R.; Hanaway, M.J.; Beebe, T.M.; Woodle, E.S. The
role of immunosuppression in lymphoma. Recent Results
Cancer Res. 2002, 159, 55–66. 34. Orive, G.; Hernandez,
R.M.; Gascon, A.R.; Calafiore, R.; Chang, T.M.; De Vos, P.;
Hortelano, G.; Hunkeler, D.; Lacik, I.; Shapiro, A.M.;
Pedraz, J.L. Cell encapsulation: Promise and progress. Nat.
Med. 2003, 9 (1), 104–107. 35. Ryan, E.A.; Lakey, J.R.;
Paty, B.W.; Imes, S.; Korbutt, G.S.; Kneteman, N.M.; Bigam,
D.; Rajotte, R.V.; Shapiro, A.M. Successful islet
transplantation: Continued insulin reserve provides
long-term glycemic control. Diabetes 2002, 51 (7),
2148–2157. S
Stent Grafts, Endovascular

1. Parodi, J.C.; Palmaz, J.C.; Barone, H.D. Transfemoral


intraluminal graft implantation for abdominal aortic
aneurysms. Ann. Vasc. Surg. 1991, 5, 491–499.

2. Sonesson, B.; Dias, N.; Malina, M.; Olofsson, P.;


Griffin, D.; Lindblad, B.; Ivancev, K. Intra-aneurysm
pressure measurements in successfully excluded abdominal
aortic aneurysm after endovascular repair. J. Vasc. Surg.
Apr 2003, 37 (4), 733–738.

3. Resch, T.; Ivancev, K.; Lindh, M.; Nyman, U.; Brunkwall,


J.; Malina, M.; Lindblad, B. Persistent collateral
perfusion of abdominal aortic aneurysm after endovascular
repair does not lead to progressive change in aneurysm
diameter. J. Vasc. Surg. Aug 1998, 28 (2), 242–249.

4. Harris, P.L.; Vallabhaneni, S.R.; Desgranges, P.;


Becquemin, J.P.; van Marrewijk, C.; Laheij, R.J. Incidence
and risk factors of late rupture, conversion, and death
after endovascular repair of infrarenal aortic aneurysms:
The EUROSTAR experience. European collaborators on
stent/graft techniques for aortic aneurysm repair. J. Vasc.
Surg. 2000, 32 (4), 739–749.

5. Chuter, T.A.; Green, R.M.; Ouriel, K.; DeWeese, J.A.


Infrarenal aortic aneurysm structure: Implications for
transfemoral repair. J. Vasc. Surg. Jul 1994, 20 (1),
44–49. discussion 49–50. 6. Dake, M.D.; Miller, D.C.;
Semba, C.P.; Mitchell, R.S.; Walker, P.J.; Liddell, R.P.
Transluminal placement of endovascular stent-grafts for the
treatment of descending thoracic aortic aneurysms. N. Engl.
J. Med. 2001, 331, 1729–1734. 7. Dake, M.D.; Kato, N.;
Mitchell, R.S.; Semba, C.P.; Razavi, M.K.; Shimono, T.;
Hirano, T.; Takeda, K.; Yada, I.; Miller, D.C. Endovascular
stent-graft placement for the treatment of acute aortic
dissection. N. Engl. J. Med. 1999, 340, 1546–1552. 8.
Chuter, T.A.; Risberg, B.; Hopkinson, B.R.; Wendt, G.;
Scott, R.A.; Walker, P.J.; Viscomi, S.; White, G. Clinical
experience with a bifurcated endovascular graft for
abdominal aortic aneurysm repair. J. Vasc. Surg. Oct 1996,
24 (4), 655–666. 9. Criado, F.J.; Barnatan, M.F.; Rizk, Y.;
Clark, N.S.; Wang, C.F. Technical strategies to expand
stent-graft applicability in the aortic arch and proximal
descending thoracic aorta. J. Endovascular Ther. 2002, 9,
II32–II38. 10. Chuter, T.A.M.; Gordon, R.L.; Reilly, L.M.;
Goodman, J.D.; Messina, L.M. An endovascular system for
thoracoabdominal aortic aneurysm repair. J. Endovasc. Ther.
2001, 8, 25–33. 11. Schneider, D.B.; Curry, T.K.; Reilly,
L.M.; Kang, J.W.; Messina, L.M.; Chuter, T.A.M. Branched
endovascular repair of aortic arch aneurysm. J. Vasc. Surg.
2003, 37, 855. 12. Abraham, C.Z.; Reilly, L.M.; Schneider,
D.B.; Dwyer, S.; Sawhney, R.; Messina, L.M.; Chuter, T.A. A
modular multi-branched system for endovascular repair of
bilateral common iliac artery aneurysms. J. Endovascular
Ther. 2003, 10, 203–207. 13. Hosokawa, H.; Iwase, T.; Sato,
M.; Yoshida, Y.; Ueno, K.; Tamaki, S.; Inoue, K. Successful
endovascular repair of juxtarenal and suprarenal aortic
aneurysms with a branched stent graft. J. Vasc. Surg. May
2001, 33 (5), 1087–1092. 14. Browne, T.F.; Hartley, D.;
Purchas, S.; Rosenberg, M.; van Schie, G.; Lawrence-Brown,
M. A fenestrated covered suprarenal aortic stent. Eur. J.
Vasc. Surg. 1999, 18, 445–449. 15. Makaroun, M.S. The
ancure endografting system: An update. J. Vasc. Surg. 2001,
33 (Suppl. 2), S129–S134. 16. Becquemin, J.P.; Lapie, V.;
Favre, J.P.; Rousseau, H.; for the French Vanguard Study
Group. Midterm results of a second generation bifurcated
endovascular graft for AAA repair. The French vanguard
trial. J. Vasc. Surg. 1999, 30, 209–217. 17. van der Laan,
M.; Prinssen, M.; Bertges, D.; Makaroun, M.S.;
Blankensteijn, J.D. Does the type of endograft affect AAA
volume change after endovascular aneurysm repair? J.
Endovasc. Ther. 2003, 10, 406–410. 18. Zarins, C.K.; for
the AneuRx Clinical Investigators. The US AneuRx clinical
trial: 6-year clinical update 2002. J. Vasc. Surg. 2003,
37, 904–908. 19. Sternbergh, W.C., 3rd; Conners, M.S., 3rd;
Tonnessen, B.H.; Carter, G.; Money, S.R. Aortic aneurysm
sac shrinkage after endovascular repair is
device-dependent: A comparison of Zenith and AneuRx
endografts. Ann. Vasc. Surg. 2003 Jan, 17 (1), 49–53. 20.
Heintz, C.; Riepe, G.; Birken, L.; Kaiser, E.; Chakfe, N.;
Morlock, M.; Delling, G.; Imig, H. Corroded nitinol wires
in explanted aortic endografts: an important mechanism of
failure? J. Endovasc. Ther. 2001, 8, 248–253. 21. Criado,
F.J.; Fairman, R.M.; Becker, G.J.; for Investigators of
Talent LPS Pivotal Clinical Trial Talent LPS AAA
stent-graft: Results of a pivotal clinical trial. J. Vasc.
Surg. 2003, 37, 709–715. 22. Cook Zenith P.M.A. Panel
Presentation. April 10 2003. Gaithersberg, MD. 23. Liffman,
K.; Lawrence-Brown, M.M.; Semmens, J.B.; Bui, A.; Rudman,
M.; Hartley, D.E. Analytical modeling and numerical
simulation of forces in an endoluminal graft. J. Endovasc.
Ther. 2001, 8, 358–371. 24. Zarins, C.K.; Bloch, D.A.;
Crabtree, T.; Matsumoto, A.H.; White, R.A.; Fogarty, T.J.
Stent graft migration after endovascular aneurysm repair:
Importance of proximal fixation. J. Vasc. Surg. 2003, 38,
1264–1272. 25. Matsumura, J.S.; Katzen, B.T.; Hollier,
L.H.; Dake, M.D. Update on the bifurcated EXCLUDER
endoprosthesis: Phase I results. J. Vasc. Surg. 2001, 33
(Suppl. 2), S135–S145. 26. Hodgson, R.; McWilliams, R.G.;
Simpson, A.; Gould, D.A.; Brennan, J.A.; Gilling-Smith,
G.L.; Harris, P.L. Migration versus apparent migration:
Importance of errors due to positioning variation in plain
radiographic follow-up of aortic stent-grafts. J. Endovasc.
Ther. Oct 2003, 10 (5), 902–910. 27. Moore, W.S. The EVT
tube and bifurcated endograft systems: Technical
considerations and clinical summary. J. Endovasc. Surg.
1997, 4, 182–194. 28. White, G.H.; May, J.; Waugh, R.;
Harris, J.P.; Chaufour, X.; Yu, W.; Stephen, M.S.
Shortening of endografts during deployment in endovascular
AAA repair. J. Endovasc. Surg. Feb 1999, 6 (1), 4–10. 29.
Carpenter, J.P.; Neschis, D.G.; Fairman, R.M.; Barker,
C.F.; Golden, M.A.; Velazquez, O.C.; Mitchell, M.E.; Baum,
R.A. Failure of endovascular abdominal aortic aneurysm
graft limbs. J. Vasc. Surg. Feb 2001, 33 (2), 296–302.
discussion 302–3. 30. Fransen, G.A.J.; Desgranges, P.;
Laheij, R.J.F.; Haris, P.L.; Becquemin, J. Frequency,
predicitive factors, and consequences of stent-graft kink
following endovascular AAA repair. J. Endovasc. Ther. 2003,
10, 913–918. 31. Lee, do Y.; Kang, S.G.; Choi, D.; Lee,
G.H.; Maeda, M.; Roh, B.S.; Won, J.W.; Kim, C.W.; Kim,
E.S.; Song, H.Y. Percutaneous modular stent-grafts in the
treatment of abdominal aortic aneurysms. J. Endovasc. Ther.
Aug 2003, 10 (4), 752–759. S
Stents

35. Chorny, M.; Fishbein, I.; Golomb, G. Drug delivery


systems for the treatment of restenosis. Crit. Rev. Ther.
Drug Carr. Syst. 2000, 17, 249–284.

36. Drachman, D.E.; Simon, D.I. Restenosis: Intracoronary


brachytherapy. Curr. Treat. Options Cardiovasc. Med. 2002,
4, 109–118.

37. Serruys, P.W.; Kay, I.P. I like the candy, I hate the
wrapper: The (32)P radioactive stent. Circulation 2000,
101, 3–7.

38. Tamai, H.; Igaki, K.; Kyo, E.; Kosuga, K.; Kawashima,
A.; Matsui, S.; Komori, H.; Tsuji, T.; Motohara, S.;
Uehata, H. Initial and 6-month results of biodegradable
polyL-lactic acid coronary stents in humans. Circulation
2000, 102, 399–404.

39. Heublein, B.; Rhode, R.; Huasdorf, G.; Hartung, W.;


Haverich, R. Biocorrosion, a new principle for temporary
cardiovascular implants? Eur. Heart J. 2000, 21, 286.

40. Peuster, M.; Wohlsein, P.; Brugmann, M.; Ehlerding, M.;


Seidler, K.; Fink, C.; Brauer, H.; Fischer, A.; Hausdorf,
G. A novel approach to temporary stenting: Degradable
cardiovascular stents produced from corrodible
metal-results 6–18 months after implantation into New
Zealand white rabbits. Heart 2001, 86, 563–569. 41.
Rensing, B.J.; Vos, J.; Smits, P.C.; Foley, D.P.; van den
Brand, M.J.; van der Giessen, W.J.; de Feijter, P.J.;
Serruys, P.W. Coronary restenosis elimination with a
sirolimus eluting stent: First European human experience
with 6-month angiographic and intravascular ultrasonic
follow-up. Eur. Heart J. 2001, 22, 2125–2130. 42. Heldman,
A.W.; Cheng, L.; Jenkins, G.M.; Heller, P.F.; Kim, D.W.;
Ware, M., Jr.; Nater, C.; Hruban, R.H.; Rezai, B.; Abella,
B.S.; Bunge, K.E.; Kinsella, J.L.; Sollott, S.J.; Lakatta,
E.G.; Brinker, J.A.; Hunter, W.L.; Froehlich, J.P.
Paclitaxel stent coating inhibits neointimal hyperplasia at
4 weeks in a porcine model of coronary restenosis.
Circulation 2001, 103, 2289–2295. 43. Fox, R. American
Heart Association 2001 scientific sessions: Late-breaking
science–drug-eluting stents. Circulation 2001, 104, E9052.
44. Sousa, J.E.; Costa, M.A.; Abizaid, A.C.; Rensing, B.J.;
Abizaid, A.S.; Tanajura, L.F.; Kozuma, K.; Van Langenhove,
G.; Sousa, A.G.; Falotico, R.; Jaeger, J.; Popma, J.J.;
Serruys, P.W. Sustained suppression of neointimal
proliferation by sirolimus-eluting stents: One-year
angiographic and intravascular ultrasound follow-up.
Circulation 2001, 104, 2007–2011. 45. Liistro, F.; Colombo,
A. Late acute thrombosis after paclitaxel eluting stent
implantation. Heart 2001, 86, 262–264.
Sterilization of Biomedical Materials

19. Lerouge, S.; Wertheimer, M.R.; Yahia, L.H. Plasma


sterilization: A review of parameters, mechanisms and
limitations. Plasmas Polym. 2001, 6, 175–188.

20. Chau, T.T.; Kao, K.C.; Blank, G.; Madrid, F. Microwave


plasmas for low temperature dry sterilization. Biomaterials
1996, 17, 1273–1277.

21. Dillow, A.K.; Dehghani, F.; Hrkach, J.S.; Foster, N.R.;


Langer, R. Bacterial inactivation by using nearand
supercritical carbon dioxide. Proc. Natl. Acad. Sci. USA
1999, 96, 10344–10348.

22. Spilimbergo, S.; Dehghani, F.; Bertucce, A.; Foster,


N.R. Inactivation of bacteria and spores by pulse electric
field and high pressure CO 2 at low temperature. Biotechnol.
Bioeng. 2002, 82, 118–125. 23. Castro, A.J.;
Barbosa-Canovas, G.V.; Swanson, B.G. Microbial inactivation
of foods by pulsed electric fields. J. Food Process. Pres.
1993, 17, 47–73. 24. Vadrot, C.; Darbord, J.C. Quantitative
evaluation of prion inactivation comparing steam
sterilization and chemical sterilants: Proposed method for
test standardization. J. Hosp. Infect. 2006, 64, 143–148.
25. Tessarolo, F.; Caola, I.; Nollo, G.; Antolini, R.;
Guarrera, G.M.; Caciagli, P. Efficiency in endotoxin removal
by a reprocessing protocol for electrophysiology catheters
based on hydrogen peroxide plasma sterilization. Int. J.
Hyg. Environ. Health 2006, 209, 557–565.
Stress Fractures

Fig. 8 Spondylolytic defect in the vertebral arch (black

arrow) causes spondylolisthesis (gray arrow) of the fifth

lumbar vertebra. 9. Ivkovic´, A. Overuse injuries in young


athletes. In Overuse Injuries of the Musculoskeletal
System, 2nd Ed.; Pec´ina, M.M., Bojanic´, I., Eds.; CRC
Press: Boca Raton, 2004; 373–389. 10. Dusek, T; Pecina, M;
Loncar-Dusek, M; Bojanic´, I. Multiple stress fractures in
a young female runner. Acta. Chir. Orthop. Traumatol. Cech.
2004, 71, 308–310. 11. Dusek, T. Influence of high intensity
training on menstrual cycle disorders in athletes. Croat.
Med. J. 2001, 42, 79–92. 12. Brudvig, T.J.S.; Gudger, T.D.;
Obermeyer, L. Stress fractures in 295 trainees. A one-year
study of incidence as related to age, sex, and race. Mil.
Med. 1983, 148, 666–667. 13. Jones, B.H.; Bovee, M.W.;
Harris, J.M.; Cowan, D.N. Intrinsic risk factors for
exercise-related injuries among male and female army
trainees. Am. J. Sports Med. 1993, 21, 705–710. 14. Brunet,
M.E.; Cook, S.D.; Brinker, M.R.; Dickinson, J.A. A survey
of running injuries in 1505 competitive and recreational
runners. J. Sports Med. Phys. Fitness 1990, 30, 307–315.
15. Ivkovic, A.; Bojanic, I.; Ivkovic, M. Trijas
sportasˇica. Lijecˇ. Vjesn. 2001, 123, 200–206. 16. Arendt,
E.A. Stress fractures and the female athlete. Clin. Orthop.
2000, 372, 131–138. 17. Dubravcˇic´-Sˇimunjak, S.; Pec´ina,
M.; Kuipers, H.; Moran, J.; Hasˇpl, M. The incidence of
injuries in elite junior figure skaters. Am. J. Sports Med.
2003, 31, 511–517. 18. Bojanic´, I.; Pec´ina, H.I.;
Pec´ina, M. Prijelomi zamora. Arh. Hig. Rada Toksikol.
2001, 52, 471–482. 19. Krivickas, L.S. Anatomical factors
associated with overuse sports injuries. Sports Med. 1997,
24, 132–146. 20. Walter, S.D.; Hart, L.E.; McIntosh, J.M.;
Sutton, JR. The Ontario cohort study of running-related
injuries. Arch. Intern. Med. 1989, 149, 2561–2564. 21.
Pec´ina, M.M.; Bojanic´, I. Stress fractures. In Overuse
Injuries of the Musculoskeletal System, 2nd Ed.; Pec´ina,
M.M., Bojanic´, I., Eds.; CRC Press: Boca Raton, 2003;
315–349. 22. Knapp, T.P.; Garrett, W.E., Jr. Stress
fractures: general concepts. Clin. Sports Med. 1997, 16,
339–356. 23. Reeder, M.T.; Dick, B.H.; Atkins, J.K.;
Pribis, A.B.; Martinez, J.M. Stress fractures. Current
concepts of diagnosis and treatment. Sports Med. 1996, 22,
198–212. 24. Spitz, D.J.; Newberg, A.H. Imaging of stress
fractures in the athlete. Radiol. Clin. North Am. 2002, 40,
313–331. 25. Deutsch, A.L.; Coel, M.N.; Mink, J.H. Imaging
of stress injuries to bone: radiography, scintigraphy, and
MR imaging. Clin. Sports Med. 1997, 16, 275–290. 26.
Bojanic´, I; Pec´ina, M.M. Traitement conservateur des
fractures de contrainte du scaphoide tarsien chez le
sportif. Rev. Chir. Orthop. 1997, 83, 133–138. 27.
Ishibashi, Y.; Okamura, Y.; Otsuka, H.; Nishizawa, K;
Sasaki, T; Toh, S. Comparison of scintigraphy and magnetic
resonance imaging for stress injuries of bone. Clin. J.
Sport Med. 2002, 12, 79–84. 28. Boden, B.P.; Osbahr, D.C.
High-risk stress fractures: evaluation and treatment. J.
Am. Acad. Orthop. Surg. 2000, 8, 344–353. 29. Pec´ina, M.;
Bojanic´, I.; Dubravcˇic´, S. Stress fractures in figure
skaters. Am. J. Sports Med. 1990, 18, 227–279. 30.
Weinfeld, S.B.; Haddad, S.L.; Myerson, M.S. Metatarsal
stress fractures. Clin. Sports Med. 1997, 16, 319–338. 31.
Nunley, J.A. Fracture of the base of the fifth metatarsal.
The Jones fracture. Orthop. Clin. North Am. 2001, 32,
171–180. 32. Egol, K.A.; Koval, K.J.; Kummer, F.; Frankel,
V.H. Stress fractures of the femoral neck. Clin. Orthop.
1998, 348, 72–78. 33. Moeller, J.L.; Rifat, S.E.
Spondylolysis in active adolescent. Phys. Sports Med. 2001,
29, 27–32. 34. Sys, J.; Michielsen, J.; Bracke, P.;
Martens, M.; Verstreken, J. Nonoperative treatment of
active spondylolysis in elite athletes with normal x-ray
findings: literature review and results of conservative
treatment. Eur. Spine J. 2001, 10, 498–504. 35. Bellah,
R.D.; Summerville, D.A.; Treves, S.T.; Micheli, L.J.
Low-back pain in adolescent athletes: detection of stress
injury to the pars interarticularis with SPECT. Radiology
1991, 180, 509–512. S
Supercritical Fluid Processing

10. Tsevistas, M.; Levy, M.S.; Lo-Yim, M.Y.A.; O’Kennedy,


P.; York, P.; Humphery, G.O.; Hoare, M. The formation of
plasmid DNA loaded pharmaceutical powders using
supercritical fluid technology. Biotechnol. Bioeng. 2001, 72
(1), 12–18.

11. Sievers, R.E.; Karst, U.; Milewski, P.D.; Sellers,


S.P.; Miles, B.A.; Schaefer, J.D.; Stoldt, C.R.; Xu, C.Y.
Formation of small droplet aerosols assisted by
supercritical carbon dioxide. Aerosol. Sci. Technol. 1999,
30, 3–15.

12. Dixon, N.M.; Kell, D.B. The inhibition by CO 2 of the


growthy and metabolism of microorganisms. J. Appl.
Bacteriol. 1989, 67, 109–136.

13. Dı´az-Reinoso, B.; Moure, A.; Domı´nguez, H.; Parajo´,


J.C. Supercritical CO 2 extraction and purification of
compounds with antioxidant activity. J. Agric. Food Chem.
2006, 54 (7), 2441–2469.

14. Howdle, S.M.; Watson, M.S.; Whitaker, M.J.; Popov,


W.K.; Davies, M.C.; Mandeld, F.S.; Shakesheff, K.M.
Supercritical fluid mixing: Preparation of thermally
sensitive polymercomposites containing bioactive materials.
Chem. Commun. 2001, 109–110.

15. Gong, K.; Darr, J.A.; Rehman, I.U. Supercritical fluid


assisted impregnation of indomethacin into chitosan
thermosets for controlled release applications. Int. J.
Pharmaceut. 2005, 315, 93–98.

16. Gong, K.; Viboonkiat, R.; Rehman, I.U.; Buckton, G.;


Darr, J.A. Formation and characterization of porous
indomethacin–PVP coprecipitates prepared using solvent-free
supercritical fluid processing. J. Pharm. Sci. 2005, 94
(12), 2583–2590. 17. Thakur, R.; Gupta, R.B. Formation of
phenytoin nanoparticles using rapid expansion of
supercritical solution with solid cosolvent (RESS–SC)
process. Pharmaceut. Nanotechnol. 2006, 38 (2), 190–199.
18. Quirk, R.A.; France, R.M.; Shakeshe, K.M.; Howdle, S.M.
Supercritical fluid technologies and tissue engineering
scaffolds. Curr. Opin. Solid State Mater. Sci. 2004, 8,
313–321. 19. Partap, S.; Rehman, I.; Jones, J.R.; Darr,
J.A. Supercritical carbon dioxide in water
emulsion-templated synthesis of porous calcium alginate
hydrogels. Adv. Mater. 2006, 18, 501–504. 20. Chaudhry,
A.A.; Haque, S.; Kellici, S.; Boldrin, P.; Rehman, I.;
Khalid, F.A.; Darr, J.A. Instant nanohydroxyapatite: a
continuous and rapid hydrothermal synthesis. Chem. Commun.
2006, 4 (21), 2286–2288. 21. Barry, J.J.A.; Gidda, H.S.;
Scotchford, C.A.; Howdle, S.M. Porous methacrylate
scaffolds: supercritical fluid fabrication and in vitro
chondrocyte responses. Biomater. 2004, 25 (17), 3559–3568.
22. Butler, R.; Davies, C.M.; Cooper, A.I. Emulsion
Templating Using High Internal Phase Supercritical Fluid
Emulsions. Adv. Mater. 2001, 13 (19), 1459–1463.
Surface Coatings

10. Strnad, Z.; Strnad, J.; Urban, K. Effect of


plasmasprayed hydroxyapatite coating on the
osteoconductivity of commercially pure titanium implants.
Int. J. Oral. Maxillofac Implants. 2000, 15 (4), 483–490.

11. Atalar, E.; Haznedaroglu, I.; Aytemir, K.; Aksoyek, S.;


Ovunc, K.; Oto, A.; Ozmen, F. Effects of stent coating on
platelets and endothelial cells after intracoronary stent
implantation. Clin. Cardiol. 2001, 24, 159–164.

12. Grenadier, E.; Roguin, A.; Hertz, I.; Peled, B.;


Boulos, M.; Nikolsky, E.; Amikam, S.; Kerner, A.; Cohen,
S.; Beyar, R. Stenting very small coronary narrowings (o2
mm) using the biocompatible phosphorylcholinecoated
coronary stent. Cath. Cardiovasc. Interv. 2002, 55,
303–308.

13. Kastrati, A.; Schomig, A.; Dirschinger, J.; Mehilli,


J.; von Welser, N.; Pache, J.; Schuhlen, H.; Schilling, T.;
Schmitt, C.; Neumann, F.J. Increased risk of restenosis
after placement of gold-coated stents: results of a
randomized trial comparing gold-coated with uncoated steel
stents in patients with coronary artery disease.
Circulation 2000, 101, 2478–2483.

14. Kalnins, U.; Erglis, A.; Dinne, I.; Kumsars, I.;


Jegere, S. Clinical outcomes of silicon carbide coated
stents in patients with coronary artery disease. Med. Sci.
Monit. 2002, 8, PI16–20.

15. Peng, T.; Gibula, P.; Yao, K.; Goosen, M.F.A. Role of
polymers in improving the results of stenting in coronary
arteries. Biomaterials 1996, 17, 685–694.

16. Serruys, P.W.; Emanuelsson, H.; van der Giessen, W.


Heparin-coated Palmaz-Schatz stents in human coronary
arteries: Early outcome of the Benestent-II pilot study.
Circulation 1996, 93, 412–422.

17. Vrolix, M.C.; Legrand, V.M.; Reiber, J.H.; Grollier,


G.; Schalij, M.J.; Brunel, P.; Martinez-Elbal, L.;
GomezRecio, M.; Bar, F.W.; Bertrand, M.E.; Colombo, A.;
Brachman, J. Heparin-coated Wiktor stents in human coronary
arteries (MENTOR trial). MENTOR trial investigators. Am. J.
Cardiol. 2000, 86, 385–389.

18. Stone, G.W.; Brodie, B.R.; Griffin, J.J.; Costantini,


C.; Morice, M.C.; St. Goar, F.G.; Overlie, P.A.; Popma,
J.J.; McDonnell, J.; Jones, D.; O’Neill, W.W.; Grines, C.L.
Clinical and angiographic follow-up after primary stenting
in acute myocardial infarction: the Primary Angioplasty in
Myocardial Infarction (PAMI) stent pilot trial. Circulation
1999, 99, 1548–1554.

19. Buller, C.E.; Dzavik, V.; Carere, R.G.; Mancini, G.B.;


Barbeau, G.; Lazzam, C.; Anderson, T.J.; Knudtson, M.L.;
Marquis, J.F.; Suzuki, T.; Cohen, E.A.; Fox, R.S.; Teo,
K.K. Primary stenting versus balloon angioplasty in
occluded coronary arteries: the Total Occlusion Study of
Canada (TOSCA). Circulation 1999, 100, 236–242.

20. Mooradian, D.L.; Fernandes, B.; Diglio, C.A.; Lester,


B.R. Angiopeptin (BIM23014C) inhibits vascular smooth
muscle cell migration in vitro through a G-protein-mediated
pathway and is associated with inhibition of adenylyl
cyclase and cyclic AMP accumulation. J. Cardiovasc.
Pharmacol. 1995, 25, 611–618. 21. Sindermann, J.R.;
Skaletz-Rorowski, A.; Bartels, A.; Hohage, H.; Plenz, G.;
Schmidt, A.; Breithardt, G. Paclitaxel and cyclosporine a
show supra-additive antiproliferative effects on smooth
muscle cells by activation of protein kinase C. Basic Res.
Cardiol. 2002, 97, 125–131. 22. Drachman, D.E.; Edelman,
E.R.; Seifert, P.; Groothuis, A.R.; Bornstein, D.A.;
Kamath, K.R.; Palasis, M.; Yang, D.; Nott, S.H.; Rogers, C.
Neointimal thickening after stent delivery of paclitaxel:
change in composition and arrest of growth over six months.
J. Am. Coll. Cardiol. 2000, 36, 2325–2332. 23. Liistro, F.;
Stankovic, G.; Di Mario, C.; Takagi, T.; Chieffo, A.;
Moshiri, S.; Montorfano, M.; Carlino, M.; Briguori, C.;
Pagnotta, P.; Albiero, R.; Corvaja, N.; Colombo, A. First
clinical experience with a paclitaxel derivate-eluting
polymer stent system implantation for in-stent restenosis:
Immediate and long-term clinical and angiographic outcome.
Circulation 2002, 105, 1883–1886. 24. Farb, A.; Heller,
P.F.; Shroff, S.; Cheng, L.; Kolodgie, F.D.; Carter, A.J.;
Scott, D.S.; Froehlich, J.; Virmani, R. Pathological
analysis of local delivery of paclitaxel via a
polymer-coated stent. Circulation 2001, 104, 473–479. 25.
Morice, M.C.; Serruys, P.W.; Sousa, J.E.; Fajadet, J.;
BanHayashi, E.; Perin, M.; Colombo, A.; Schuler, G.;
Barragan, P.; Guagliumi, G.; Molnar, F.; Falotico, R. A
randomized comparison of a sirolimus-eluting stent with a
standard stent for coronary revascularization. N. Engl. J.
Med. 2002, 346, 1773–1780. 26. Sousa, J.E.; Costa, M.A.;
Abizaid, A.C.; Rensing, B.J.; Abizaid, A.S.; Tanajura,
L.F.; Kozuma, K.; Van Langenhove, G.; Sousa, A.G.;
Falotico, R.; Jaeger, J.; Popma, J.J.; Serruys, P.W.
Sustained suppression of neointimal proliferation by
sirolimus-eluting stents: one-year angiographic and
intravascular ultrasound follow-up. Circulation 2001, 104,
2007–2011. 27. Serruys, P.W.; Degertekin, M.; Tanabe, K.;
Abizaid, A.; Sousa, J.E.; Colombo, A.; Guagliumi, G.;
Wijns, W.; Lindeboom, W.K.; Ligthart, J.; de Feyter, P.J.;
Morice, M.C. Intravascular ultrasound findings in the
multicenter, randomized, double-blind RAVEL (RAndomized
study with the sirolimus-eluting VElocity
balloon-expandable stent in the treatment of patients with
de novo native coronary artery Lesions) trial. Circulation
2002, 106, 798–803. S
Surface Modification

39. Maruyama, M.; Capello, W.N.; D’Antonio, J.A.; Jaffe,


W.L.; Bierbaum, B.E. Effect of low-friction iontreated
femoral heads on polyethylene wear rates. Clin. Orthop.
Relat. Res. 2000, 1 (370), 183–191.

40. Frank, J.L.; Garb, J.L.; Halla, B.; Reed, W.P., Jr.
Ionic implantation of silicone chronic venous access
devices does not alter thrombotic complications: A
doubleblinded, randomized clinical trial. Surgery 2001, 129
(5), 547–551.

41. Pignataro, B.; Conte, E.; Scandurra, A.; Marletta, G.


Improved cell adhesion to ion beam-irradiated polymer
surfaces. Biomaterials 1997, 18 (22), 1461–1470.

42. Guzman, L.; Celva, R.; Miotello, A.; Voltolini, E.;


Ferrari, F.; Adami, M. Polymer surface modification by ion
implantation and reactive deposition of transparent films.
Surf. Coat. Technol. 1998, 104, 375–379.

43. Krupa, D.; Baszkiewicz, J.; Kozubowski, J.; Barcz, A.;


Sobczak, J.; Bilinski, A.; Rajchel, B. The influence of
calcium and/or phosphorus ion implantation on the structure
and corrosion resistance of titanium. Vacuum 2001, 63 (4),
715–719.

44. Sato, H.; Tsuji, H.; Ikeda, S.; Ikemoto, N.; Ishikawa,
J.; Nishimoto, S. Enhanced growth of human vascular
endothelial cells on negative ion (Ag )-implanted
hydrophobic surfaces. J. Biomed. Mater. Res. 1999, 44 (1),
22–30.

45. Cui, F.Z.; Luo, Q.L.; Feng, J. Highly adhesive


hydroxyapatite coatings on titanium alloy formed by ion
beam assisted deposition. J. Mater. Sci., Mater. Med. 1997,
8, 403–405.

46. Kurtz, S.M.; Muratoglu, O.K.; Evans, M.; Edidin, A.A.


Advances in the processing, sterilization, and crosslinking
of ultra-high molecular weight polyethylene for total joint
arthroplasty. Biomaterials 1999, 20 (18), 1659–1688.

47. Wetzels, G.M.; Koole, L.H. Photoimmobilisation of


poly(n-vinylpyrrolidinone) as a means to improve
haemocompatibility of polyurethane biomaterials.
Biomaterials 1999, 20 (20), 1879–1887.

48. McPherson, T.B.; Shim, H.S.; Park, K. Grafting of PEO


to glass, nitinol, and pyrolytic carbon surfaces by gamma
irradiation. J. Biomed. Mater. Res. 1997, 38 (4), 289–302.

49. Kidane, A.; McPherson, T.; Shim, H.S.; Park, K. Surface


modification of polyethylene terephthalate using
PEO–polybutadiene–PEO triblock copolymers. Colloid Surf., B
2000, 18 (3–4), 347–353.

50. Curti, P.S.; De Moura, M.R.; Radovanovic, E.; Rubira,


A.F.; Muniz, E.C. Surface modification of polystyrene and
poly(ethylene terephtalate) by grafting
poly(n-isopropylacrylamide). J. Mater. Sci., Mater. Med.
2002, 13 (12), 1175–1180.

51. Ward, J.H.; Bashir, R.; Peppas, N.A. Micropatterning of


biomedical polymer surfaces by novel UV polymerization
techniques. J. Biomed. Mater. Res. 2001, 56 (3), 351–360.

52. Thissen, H.; Hayes, J.P.; Kingshott, P.; Johnson, G.;


Harvey, E.C.; Griesser, H.J. Nanometer thickness laser
ablation for spatial control of cell attachment. Smart
Mater. Struct. 2002, 11 (5), 792–799.

53. Sun, S.; Chong, K.S.; Leggett, G.J. Nanoscale molecular


patterns fabricated by using scanning near-field optical
lithography. J. Am. Chem. Soc. 2002, 124 (11), 2414–2415.
54. Duncan, A.C.; Weisbuch, F.; Rouais, F.; Lazare, S.;
Baquey, C. Laser microfabricated model surfaces for
controlled cell growth. Biosens. Bioelectron. 2002, 17 (5),
413–426. 55. Leng, Y.X.; Chen, J.Y.; Zeng, Z.M.; Tian,
X.B.; Yang, P.; Huang, N.; Zhou, Z.R.; Chu, P.K. Properties
of titanium oxide biomaterials synthesized by titanium
plasma immersion ion implantation and reactive ion
oxidation. Thin Solid Films 2000, 377, 573–577. 56. Lee,
Y.; Han, S.; Lim, H.; Kim, Y.; Kim, H. Surface analysis of
polymers electrically improved by plasmasource
ion-implantation. Anal. Bioanal. Chem. 2002, 373 (7),
595–600. 57. Shin, G.H.; Lee, Y.H.; Lee, J.S.; Kim, Y.S.;
Choi, W.S.; Park, H.J. Preparation of plastic and
biopolymer multilayer films by plasma source ion
implantation. J. Agric. Food. Chem. 2002, 50 (16),
4608–4614. 58. Bilek, M.M.M.; McKenzie, D.R.; Tarrant,
R.N.; Lim, S.H.M.; McCulloch, D.G. Plasma-based ion
implantation utilizing a cathodic arc plasma. Surf. Coat.
Technol. 2002, 156 (1–3), 136–142. 59. Ratner, B.D.;
Chilkoti, A.; Lopez, G.P. Plasma Deposition and Treatment
for Biomedical Applications. In Plasma Deposition,
Treatment, and Etching of Polymers; d’Agostino, R., Ed.;
Academic Press Inc.: San Diego, 1990; Vol. 1, 463–516. 60.
Latkany, R.; Tsuk, A.; Sheu, M.-S.; Loh, I.-H.;
Trinkaus-Randall, V. Plasma surface modification of
artificial corneas for optimal epithelialisation. J. Biomed.
Mater. Res. 1996, 36, 29–37. 61. Dai, L.; Zientek, P.; St.
John, H.A.W.; Pasic, P.; Chatelier, R.C.; Griesser, H.J.
Surface Modification of Contact Lenses: Making Contact
Lenses Wettable. In Surface Modification of Polymeric
Biomaterials; Ratner, B., Castner, D., Eds.; Plenum Press:
New York, 1996; 147. 62. Weikart, C.M.; Matsuzawa, Y.;
Winterton, L.; Yasuda, H.K. Evaluation of plasma
polymer-coated contact lenses by electrochemical impedance
spectroscopy. J. Biomed. Mater. Res. 2001, 54 (4), 597–607.
63. Lumsden, A.B.; Chen, C.; Coyle, K.A.; Ofenloch, J.C.;
Wang, J.H.; Yasuda, H.K.; Hanson, S.R. Nonporous silicone
polymer coating of expanded polytetrafluoroethylene grafts
reduces graft neointimal hyperplasia in dog and baboon
models. J. Vasc. Surg. 1996, 24 (5), 825–833. 64. Yasuda,
H.; Gazicki, M. Biomedical applications of plasma
polymerization and plasma treatment of polymer surfaces.
Biomaterials 1982, 3 (2), 68–77. 65. Hahn, A.W.; Yasuda,
H.K.; James, W.J.; Nichols, M.F.; Sadhir, R.K.; Sharma,
A.K.; Pringle, O.A.; York, D.H.; Charlson, E.J. Glow
discharge polymers as coatings for implanted devices.
Biomed. Sci. Instrum. 1981, 17, 109–113. 66.
Nilsson-Ekdahl, K.; Nilsson, B.; Golander, C.G.; Elwing,
H.; Lassen, B.; Nilsson, U.R. Complement activation on
radio frequency plasma modified polystyrene surfaces. J.
Colloid Interface Sci. 1993, 158, 121–128. 67. Griesser,
H.J.; Chatelier, R.C.; Gengenbach, T.R.; Johnson, G.;
Steele, J.G. Growth of human cells on plasma polymers:
Putative role of amine and amide groups. J. Biomater. Sci.,
Polym. Ed. 1994, 5, 531–554. 68. Haddow, D.B.; Steele,
D.A.; Short, R.D.; Dawson, R.A.; Macneil, S.
Plasma-polymerized surfaces for culture of human
keratinocytes and transfer of cells to an in vitro
wound-bed model. J. Biomed. Mater. Res. 2003, 64 (1),
80–87. 69. Mar, M.N.; Ratner, B.D.; Yee, S.S. An
intrinsically protein-resistant surface plasmon resonance
biosensor based upon a RF-plasma-deposited thin film. Sens.
Actuators, B: Chem. 1999, 54 (1–2), 125–131. 70. Nakamura,
R.; Muguruma, H.; Ikebukuro, K.; Sasaki, S.; Nagata, R.;
Karube, I.; Pedersen, H. A plasmapolymerized film for
surface plasmon resonance immunosensing. Anal. Chem. 1997,
69, 4649–4652. 71. Kiaei, D.; Hoffmann, A.; Horbett, T.
Radio-frequency gas discharge (RFGD) fluorination of
polymers: Protein and cell interactions at RFGD-fluorinated
interfaces. Radiat. Phys. Chem. 1995, 46 (2), 191–197. 72.
Lopez, G.P.; Ratner, B.D.; Tidwell, C.D.; Haycox, C.L.;
Rapoza, R.J.; Horbett, T.A. Glow discharge plasma
deposition of tetraethylene glycol dimethyl ether for
fouling-resistant biomaterial surfaces. J. Biomed. Mater.
Res. 1992, 26, 415–439. 73. Griesser, H.J. Gas plasma
techniques for the preparation of hydrophilic polymer
surfaces. Mater. Forum 1990, 14, 192–202. 74. Lassen, B.;
Malmsten, M. Competitive protein adsorption at radio
frequency plasma polymer surfaces. J. Mater. Sci., Mater.
Med. 1994, 5, 662–665. 75. Tang, L.; Wu, Y.; Timmons, R.B.
Fibrinogen adsorption and host tissue responses to plasma
functionalized surfaces. J. Biomed. Mater. Res. 1998, 42,
156–163. 76. Shen, M.C.; Martinson, L.; Wagner, M.S.;
Castner, D.G.; Ratner, B.D.; Horbett, T.A. PEO-like plasma
polymerized tetraglyme surface interactions with leukocytes
and proteins: In vitro and in vivo studies. J. Biomater.
Sci., Polym. Ed. 2002, 13 (4), 367–390. 77. Bae, J.; Seo,
E.; Kang, I. Synthesis and characterization of heparinized
polyurethanes using plasma glow discharge. Biomaterials
1999, 20, 529–537. 78. Griesser, H.J.; McLean, K.M.;
Beumer, G.J.; Gong, X.; Kingshott, P.; Johnson, G.; Steele,
J.G. Surface immobilization of synthetic proteins via
plasma polymer interlayers. Mater. Res. Symp. Proc. 1999,
544, 9. 79. Hendricks, S.K.; Kwok, C.; Shen, M.; Horbett,
T.A.; Ratner, B.D.; Bryers, J.D. Plasma-deposited membranes
for controlled release of antibiotic to prevent bacterial
adhesion and biofilm formation. J. Biomed. Mater. Res. 2000,
50 (2), 160–170. 80. Narayanan, P. Surface
functionalization by RF plasma treatment of polymers for
immobilization of bioactivemolecules. J. Biomater. Sci.,
Polym. Ed. 1994, 6 (2), 181–193. 81. Sodhi, R.N.S.; Sahi,
V.P.; Mittelman, M.W. Application of electron spectroscopy
and surface modification techniques in the development of
anti-microbial coatings for medical devices. J. Electron
Spectrosc. 2001, 121 (1–3), 249–264. 82. Lens, J.P.;
Terlingen, J.G.A.; Engbers, G.H.M.; Feijen, J. Preparation
of heparin-like surfaces by introducing sulfate and
carboxylate groups on poly(ethylene) using an argon plasma
treatment. J. Biomater. Sci., Polym. Ed. 1998, 9 (4),
357–372. 83. Goessl, A.; Garrison, M.D.; Lhoest, J.B.;
Hoffman, A.S. Plasma lithography-thin film patterning of
polymeric biomaterials by RF plasma polymerization I:
Surface preparation and analysis. J. Biomater. Sci., Polym.
Ed. 2001, 12 (7), 721–738. 84. Ohl, A.; Schroder, K.
Plasma-induced chemical micropatterning for cell culturing
applications: A brief review. Surf. Coat. Technol. 1999,
119, 820–830. 85. Aronsson, B.O.; Lausmaa, J.; Kasemo, B.
Glow discharge plasma treatment for surface cleaning and
modification of metallic biomaterials. J. Biomed. Mater.
Res. 1997, 35 (1), 49–73. 86. Terlingen, J.G.; Feijen, J.;
Hoffman, A.S. Immobilization of surface active compounds on
polymer supports using a gas discharge process. J.
Biomater. Sci., Polym. Ed. 1992, 4 (1), 31–33. 87. Pu,
F.R.; Williams, R.L.; Markkula, T.K.; Hunt, J.A. Effects of
plasma treated PET and PTFE on expression of adhesion
molecules by human endothelial cells in vitro. Biomaterials
2002, 23 (11), 2411–2428. 88. Beyer, D.; Knoll, W.;
Ringsdorf, H.; Wang, J.-H.; Timmons, R.; Sluka, P. Reduced
protein adsorption on plastics via direct plasma deposition
of triethylene glycol monoallyl ether. J. Biomed. Mater.
Res. 1996, 36, 181–189. 89. Pan, Y.V.; Wesley, R.A.;
Luginbuhl, R.; Denton, D.D.; Ratner, B.D. Plasma
polymerized n-isopropylacrylamide: Synthesis and
characterization of a smart thermally responsive coating.
Biomacromolecules 2001, 2 (1), 32–36. 90. Tarducci, C.;
Schofield, W.C.E.; Badyal, J.P.S. Monomolecular
functionalization of pulsed plasma deposited
poly(2-hydroxyethyl methacrylate) surfaces. Chem. Mater.
2002, 14 (6), 2541–2545. 91. Kim, D.D.; Takeno, M.M.;
Ratner, B.D.; Horbett, T.A. Glow discharge plasma
deposition (GDPD) technique for the local controlled
delivery of hirudin from biomaterials. Pharm. Res. 1998, 15
(5), 783–786. 92. Calderon, J.G.; Harsch, A.; Gross, G.W.;
Timmons, R.B. Stability of plasma-polymerized allylamine
films with sterilization by autoclaving. J. Biomed. Mater.
Res. 1998, 42, 597–603. 93. Beck, A.J.; Jones, F.R.; Short,
R.D. Plasma copolymerization as a specific route to the
fabrication of new surfaces with controlled amounts of
specific chemical functionality. Polymer 1996, 37,
5537–5539. 94. Uyama, Y.; Kato, K.; Ikada, Y. Surface
Modification of Polymers by Grafting. In Advances in Polymer
Science; Springer-Verlag: Berlin, 1998; Vol. 137, 3–39. 95.
Kang, E.T.; Zhang, Y. Surface modification of fluoropolymers
via molecular design. Adv. Mater. 2000, 12, 1481–1494. 96.
Claesson, P.M. Poly(ethylene oxide) surface coatings:
Relations between intermolecular forces, layer structure S
and protein repellency. Colloid Surf., A 1993, 77, 109–118.

97. Bamford, C.H.; Al-Lamee, K.G. Studies in polymer


surface modification and grafting for biomedical uses. 2.
Application to arterial blood filters and oxygenators.
Polymer 1996, 37, 4885–4889.

98. Malmsten, M.; Emoto, K.; Van Alstine, J. Effect of


chain density on inhibition of protein adsorption by
poly(ethylene glycol) based coatings. J. Colloid Interface
Sci. 1998, 202, 507–517.

99. Filippini, P.; Rainaldi, C.; Ferrante, A.; Mecheri, B.;


Gabrielli, G.; Bombace, M.; Indovina, P.L.; Santini, M.T.
Modulation of osteosarcoma cell growth and differentiation
by silane-modified surfaces. J. Biomed. Mater. Res. 2001, 55
(3), 338–349.
100. Whitesides, G.M.; Ostuni, E.; Takayama, S.; Jiang, X.;
Ingber, D.E. Soft lithography in biology and biochemistry.
Annu. Rev. Biomed. Eng. 2001, 3, 335–373.

101. Houseman, B.T.; Mrksich, M. The role of ligand density


in the enzymatic glycosylation of carbohydrates presented
in self-assembled monolayers of alkanethiolates on gold.
Angew. Chem. Int. Ed. 1999, 38 (6), 782–785.

102. Mrksich, M. What can surface chemistry do for biology?


Curr. Opin. Biotechnol. 2002, 6, 794–797.

103. Jung, D.R.; Kapur, R.; Adams, T.; Giuliano, K.A.;


Mrksich, M.; Craighead, H.G.; Taylor, D.L. Topographical
and physicochemical modification of material surface to
enable patterning of living cells. Crit. Rev. Biotechnol.
2001, 21 (2), 111–154. 104. Roberts, C.; Chen, C.S.;
Mrksich, M.; Martichonok, V.; Ingber, D.E.; Whitesides,
G.M. Using mixed selfassembled monolayers presenting RGD
and (EG) 3 OH groups to characterize long-term attachment
of bovine capillary endothelial cells to surfaces. J. Am.
Chem. Soc. 1998, 120, 6548–6555. 105. Hubbell, J.A.
Bioactive biomaterials. Curr. Opin. Biotechnol. 1999, 10,
123–129. 106. Tirrell, M.; Kokkoli, E.; Biesalski, M. The
role of surface science in bioengineered materials. Surf.
Sci. 2002, 500, 61–83. 107. Sakiyama-Elbert, S.E.; Hubbell,
J.A. Functional biomaterials: Design of novel biomaterials.
Annu. Rev. Mater. Res. 2001, 31, 183–201. 108. Hermanson,
G.T. Bioconjugate Techniques; Academic Press: London, 1996;
Vol. 1. 109. Peluso, P.; Wilson, D.S.; Do, D.; Tran, H.;
Venkatasubbaiah, M.; Quincy, D.; Heidecker, B.; Poindexter,
K.; Tolani, N.; Phelan, M.; Witte, K.; Jung, L.S.; Wagner,
P.; Nock, S. Optimizing antibody immobilization strategies
for the construction of protein microarrays. Anal. Biochem.
2003, 312, 113– 124.
Surface Topography and Cell Behavior

13. Hamilton, D.W.; Riehle, M.O.; Rappuoli, R.; Monaghan,


W.; Barbucci, R.; Curtis, A.S.G. The response of primary
articular chondrocytes to micrometric topography and
sulfated hyaluronic acid based matrices. Cell Biol. Int.
2005, 29 (8), 605–615.

14. Dow, J.A.T.; Clark, P.; Connolly, P.; Curtis, A.S.;


Wilkinson, C.D. Novel methods for the guidance and
monitoring of single cells and simple networks in culture.
J. Cell Sci. 1986, 8 (suppl.), 55–79.

15. Qu, J.; Chehroudi, B.; Brunette, D.M. The use of


micromachined surfaces to investigate the cell behavioural
factors essential to osseointegration. Oral Dis. 1996, 2,
102–115.

16. Riehle, M.; Ferris, D.; Hamilton, D.; Curtis, A.S.G.


Cell behavior in tubes. Exp. Biol. Online 1998, 3 (2);
http://link.springer.de/link/service/journals/00898/tocs/
t8003100.htm accessed Apr 2005.

17. Hamilton, D.W.; Brunette, D.M. Gap guidance of


fibroblasts and epithelial cells by discontinuous surface
topographies. Exp. Cell Res. 2005, 309 (2), 429–437.

18. Refai, A.K.; Textor, M.; Brunette, D.M.; Waterfield,


J.D. Effect of titanium surface topography on macrophage
activation and secretion of proinflammatory cytokines and
chemokine. J. Biomed. Mat. Res. 2004, 70 (2), 194–205.

19. Weiss, P.; Taylor, A.C. Fish Scales as substratum for


uniform orientation of cells in vitro. Anat. Rec. 1956,
124, 381.

20. Dunn, G.A.; Heath, J.P. A new hypothesis of contact


guidance in tissue cells. Exp. Cell Res. 1976, 101, 1–14.

21. O’Hara, P.T.; Buck, R.C. Contact guidance in vitro: a


light, transmission and scanning electron microscope study.
Exp. Cell Res. 1979, 121, 235–249. 22. Brunette, D.M.
Spreading and orientation of epithelial cells on grooved
substrata. Exp. Cell Res. 1986, 167, 203–217. 23. Oakley,
C.; Brunette, D.M. The sequence of alignment of
microtubules, focal contacts and actin filaments in
fibroblasts spreading on smooth and grooved titanium
substrata. J. Cell Sci. 1993, 106, 343–354. 24. Andersson,
A.M.; Olsson, P.; Lidberg, U.; Sutherland, D. The effects
of continuous and discontinuous groove edges on cell shape
and alignment. Exp. Cell Res. 2003, 288, 177–188. 25.
Brunette, D.M. Fibroblasts on micromachined substrata
orient hierarchically to grooves of different dimensions.
Exp. Cell Res. 1986, 164, 11–26. 26. Chou, L.; Firth, J.D.;
Uitto, V.-J.; Brunette, D.M. Substratum surface topography
alters cell shape and regulates fibronectin mRNA level, mRNA
stability, secretion and assembly in human fibroblasts. J.
Cell Sci. 1995, 108 (4), 1563–1573. 27. Dalby, M.J.;
Riehle, M.O.; Yarwood, S.J.; Wilkinson, C.D.; Curtis, A.S.
Nucleus alignment and cell signaling in fibroblasts:
response to a micro-grooved topography. Exp. Cell Res.
2003, 284 (2), 274–282. 28. Chehroudi, B.; Gould, T.R.;
Brunette, D.M. Effects of a grooved titanium-coated implant
surface on epithelial cell behavior in vitro and in vivo.
J. Biomed. Mater. Res. 1989, 23, 1067–1085. 29. Wieland,
M.; Chehroudi, B.; Textor, M.; Brunette, D.M. Use of
Ti-coated replicas to investigate the effects on fibroblast
shape of surfaces with varying roughness and constant
chemical composition. J. Biomed. Mat. Res. 2002, 60 (3),
434–444. S
Sutures

6. Chu, C.C. Survey of Clinically Important Wound Closure


Biomaterials. In Biocompatible Polymers, Metals, and
Composites; Szycher, M., Ed.; Society for Plastics,
Engineers, Technomic: Westport, CT, 1983; 477–523. Chapter
22.

7. Chu, C.C. Biodegradable Suture Materials: Intrinsic and


Extrinsic Factors Affecting Biodegradation Phenomena. In
Encyclopedic Handbook of Biomaterials and Bioengineering;
Wise, D.L., Altobelli, D.E., Schwartz, E.R., Yszemski, M.,
Gresser, J.D., Trantolo, D.J., Eds.; Marcel Dekker: New
York, 1995; Vol. 1, 543–688.

8. Chu, C.C. Degradation and Biocompatibility of Synthetic


Absorbable Suturematerial: General Biodegradation Phenomena
and Some Factors Affecting Biodegradation. In Biomedical
Applications of Synthetic Biodegradable Polymers;
Hollinger, J., Ed.; CRC Press: Boca Raton, FL, 1995;
103–128. Chapter 5

9. Chu, C.C.; von Fraunhofer, J.A.; Greisler, H.P. Wound


Closure Biomaterials and Devices; CRC Press: Boca Raton,
FL, 1997.

10. Storch, M.; Perry, L.C.; Davison, J.M.; Ward, J.J. A


28-day study of the effect of coated VIcryl Plus
antibacterial suture (coated Polyglactin 910 suture with
triclosan) on wound healing in Guinea pig linear incisional
skin wounds. Surg. Infect. 2002, 3 (Supplement), 89–98.

11. Rothenburger, S.; Spangler, D.; Bhende, S.; Burkley, D.


In vitro antimicrobial evaluation of coated Vicryl Plus
antibacterial suture (coated Polyglactin 910 with riclosan)
using zone of inhibition assays. Surg. Infec. 2002, 3
(Supplement), 79–87.

12. Conn, J., Jr.; Beal, J.M. Coated Vicryl synthetic


absorbable sutures. Surg. Gynecol. Obstet. 1980, 150 (6),
843–844.

13. Mattei, F.V. Absorbable Coating Composition for


Sutures. US Patent 4,201,216, 1980.

14. Casey, D.J.; Lewis, O.G. Absorbable and Nonabsorbable


Sutures. In Handbook of Biomaterials Evaluation: Scientific,
Technical, and Clinical Testing of Implant Materials; von
Recum, A.F., Ed.; Macmillan Publishing: New York, 1986;
86–94. Chapter 7
15. Rodeheaver, G.T.; Thacker, J.G.; Owen, J.; et al.
Knotting and handling characteristics of coated synthetic
absorbable sutures. J. Surg. Res. 1983, 35, 525–530.

16. Rodeheaver, G.T.; Thacker, J.G.; Delich, R.F.


Mechanical performance of polyglycolic acid and polyglactin
910 synthetic absorbable sutures. Surg. Gynecol. Obstet.
1981, 153, 835–841.

17. Kawai, T.; Matsuda, T.; Yoshimoto, M. Coated Sutures


Exhibiting Improved Knot Security. US Patent 4,983,180,
1991.

18. Messier, K.A.; Rhum, J.D. Caprolactone Polymers for


Suture Coating. US Patent 4,624,256, 1986. 19. Bezwada,
R.S.; Hunter, A.W.; Shalaby, S.W. Copolymers of
e-caprolactone, glycolide and glycolic acid for suture
coatings. US Patent 4,994,074, 1991. 20. Wang, D.W.; Casey,
D.J.; Lehmann, L.T. Surgical Suture Coating. US Patent
4,705,820, 1987. 21. Bezwada, R.S.; et al. Monocryl, a new
ultra-pliable absorbable monofilament suture derived from
e-caprolactone and glycolide. 1994, 22. Roby, M.S.; et al.
US Patent, 5,403,347, April 4, 1995. 23. Chu, C.C.; Kizil,
Z. Quantitative evaluation of stiffness of commercial
suture materials. Surg. Gynecol. Obstet. 1989, 168 (3),
233–238. 24. Tomita, N.; Tamai, S.; Morihara, T.; Ikeuchi,
K.; Ikada, Y. Handling characteristics of braided suture
materials for tight tying. J. Appl. Biomater. 1993, 4 (1),
61–65. 25. Abbott, W.M.; Megerman, J.; Hasson, J.E.;
L’Italien, G.; Warnock, D. Effect of compliance mismatch
upon vascular graft patency. J. Vasc. Surg. 1987, 5 (2),
376–382. 26. Megerman, J.; Hamilton, G.; Schmitz-Rixen, T.;
Abbott, W.M. Compliance of vascular anastomoses with
polybutester and polypropylene sutures. J. Vasc. Surg.
1993, 18 (5), 827–834. 27. Herman, J.B. Tensile strength
and knot security of surgical suture materials. Am. Surg.
1971, 37, 209–217. 28. Walton, M. Strength retention of
chromic gut and monofilament synthetic absorbable suture
materials in joint tissues. Clin. Orthop. Relat. Res. 1989
(242), 303–310. 29. Metz, S.A.; Chegini, N.; Masterson,
B.J. In vivo tissue reactivity and degradation of suture
materials: A comparison of Maxon and PDS. J. Gynecol. Surg.
1989, 5, 37–46. 30. Sewell, W.R.; Wiland, J.; Craver, B.N.
A new method of comparing sutures of bovine catgut with
sutures of bovine catgut in three species. Surg. Gynecol.
Obstet. 1955, 100, 483–494. 31. Van Winkle, W.; Salthouse,
T.N. Biological Response to Sutures and Principles of
Suture Selection; Ethicon: Somerville, NJ, 1976; 1–20. 32.
Salthouse, T.N.; Matlaga, B.F. Significance of cellular
enzyme activity at nonabsorbable suture implant sites:
Silk, polyester and polypropylene. J. Surg. Res. 1975, 19,
127–132. 33. Salthouse, T.N. Biocompatibility of Sutures.
In Biocompatibility in Clinical Practice; Williams, D.F.,
Ed.; CRC Press: Boca Raton, FL, 1982; Vol. 1, 12–32. 34.
Madsen, E.T. An experimental and clinical evaluation of
surgical suture materials, I and II. Surg. Gynecol. Obstet.
1953, 97, 73–80. 35. Lee, K.-H.; Chu, C.C. The effect of
superoxide ions in the degradation of five synthetic
absorbable suture materials. J. Biomed. Mater. Res. 2000,
49 (1), 25–35.
Telemedicine

1. Bashshur, R. Technology Serves the People: The Story of


a Cooperative Telemedicine Project by NASA, the Indian
Health Service and the Papago People, Superintendent of
Documents. US Government Printing Office: Washington, D.C.,
1980; 110 p.

2. Benschoter, R.A. Educational Broadcasting;


CCTVPioneering Nebraska Medical Center, Oct. 1971; 1–3.

3. Berek, B.; Canna, M. Telemedicine on the move: Health


care heads down the information superhighway. Hosp.
Technol. Ser. 1994, 13 (6), 1–65.

4. Lobley, D. The economics of telemedicine. J. Telemed.


Telecare 1997, 3 (3), 117–125. 5. Norris, T.E.; Hart, G.L.;
Larson, E.H.; Tarczy-Hornoch, P.; Masuda, D.L.; Fuller,
S.S.; House, P.J.; Dyck, S.M. Low-bandwidth, low-cost
telemedicine consultations in rural family practice. J. Am.
Board Fam. Pract. Mar.– Apr. 2002, 15 (2), 123–127. 6.
Stensland, J.; Speedie, S.M.; Ideker, M.; House, J.;
Thompson, T. The relative cost of outpatient telemedicine
services. Telemed. J. 1999, 5 (3), 245–256. Fall. 7.
Zincone, L.H., Jr.; Doty, E.; Balch, D.C. Financial
analysis of telemedicine in a prison system. Telemed. J.
1997, 3 (4), 247–255. Winter. 8. McCue, M.J.; Mazmanian,
P.E.; Hampton, C.; Marks, T.K.; Fisher, E.; Parpart, F.;
Krick, R.S. The case of Powhatan Correctional
Center/Virginia Department of Corrections and Virginia
Commonwealth University/ Medical College of Virginia.
Telemed. J. 1997, 3 (1), 11–17. Spring. 9. Wilson, M. The
future of telemedicine. Stud. Health Technol. Inform. 2002,
80, 129–136. 10. Picot, J. Telemedicine and telehealth in
Canada: Forty years of change in the use of information and
communications technologies in a publicly administered
health care system. Telemed. J. 1998, 4 (3), 199–205. Fall.
11. Murphy, R.L.; Bird, K.T. Telediagnosis: A new community
health resource; observations on the feasibility of
telediagnosis based on 1000 patient transactions. Am. J.
Public Health Feb. 1974, 64 (2), 113–119. 12. Murphy, R.L.;
Fitzpatrick, T.B.; Haynes, H.A.; Bird, K.T.; Sheridan, T.B.
Accuracy of dermatologic diagnosis by television. Arch.
Dermatol. Jun. 1972, 105 (6), 833–835. 13. Ortner, J.
Spacelab concept and its utilization for science. Acta
Astronaut. Jan.–Feb. 1975, 2 (1–2), 1–13. 14. Doarn, C.R.;
et al. Telemedicine in the US Space Program. Telemed. J.
Spring 1998, 4 (1), 19–30. 15. Rossi Mori, A.; Consorti,
F.; Galeazzi, E. Standards to support development of
terminological systems for healthcare telematics. Methods
Inf. Med. Nov. 1998, 37 (4–5), 551–563. 16. Rovetta, A.;
Bejczy, A.K.; Sala, R. Telerobotic surgery: Applications on
human patients and training with virtual reality. Stud.
Health Technol. Inform. 1997, 39, 508–517. 17. Harnett,
B.M.; Doarn, C.R.; Russell, K.M.; Kapoor, V.; Merriam,
N.R.; Merrell, R.C. Wireless telemetry and internet
technologies for medical management: A Martian analogy.
Aviat. Space Environ. Med. 2001, 72 (12), 1125–1131. 18.
Darkins, A.; Fisk, N.; Garner, P.; Wootton, R.
Pointto-point telemedicine using the ISDN. J. Telemed.
Telecare 1996, 1 (2 Suppl), 82–83. 19. Wu, T.K.; Liu, J.L.;
Tschai, H.J.; Lee, Y.H.; Leu, H.T. An ISDN-based
telemedicine system. Digit. Imaging Aug. 1998, 11 (3 Suppl
1), 93–95. 20. Chan, F.Y.; Whitehall, J.; Hayes, L.;
Taylor, A.; Soong, B.; Lessing, K.; Cincotta, R.; Cooper,
D.; Stone, M.; Lee-Tannock, A.; Baker, S.; Smith, M.;
Green, E.; Whiting, R. Minimum requirements for remote
realtime fetal tele-ultrasound consultation. J. Telemed.
Telecare 1999, 5 (3), 171–176. 21. Turner, J.; Brick, J.;
Brick, J.E. MDTV Telemedicine Project: Technical
considerations in videoconferencing for medical
applications. Telemed. J. 1995, 1 (1), 67–71. Spring. 22.
Broderick, T.J.; Harnett, B.M.; Doarn, C.R.; Rodas, E.B.;
Merrell, R.C. Real-time Internet connections: Implications
for surgical decision making in laparoscopy. Ann. Surg.
Aug. 2001, 234 (2), 165–167. 23. Fedyukin, I.V.; Reviakin,
Y.G.; Orlov, O.I.; Doarn, C.R.; Harnett, B.M.; Merrell,
R.C. Experience in the application of Java technologies in
telemedicine. J. Appl. Health Technol. eHealth Int. 2002,
1, 3. 24. Rosser, J.C.; Bell, R.L.; Harnett, B.M.; Rodas,
E.; Murayama, M.; Merrell, R.C. Use of mobile lowbandwidth
telemedical techniques for extreme telemedicine
application. Am. Coll. Surg. J. 1999, 189 (4), 397–404. 25.
Harnett, B.; Angood, P.; Merriam, N.; Satava, R.; Doarn,
C.R.; Merrell, R.C. The benefits of integrating internet
technology with standard communications for telemedicine in
extreme environments. Aviat. Space Environ. Med. 2001, 72
(12), 1132–1137. 26. Ruffin, M. Telemedicine: Where is
technology taking us? Physician Exec. Dec. 1995, 21 (12),
43–44. 27. Worth, E.R.; Patrick, T.B.; Klimczak, J.C.;
Reid, J.C. Cost-effective clinical uses of wide-area
networks: Electronic mail as telemedicine. Proc. Annu.
Symp. Comput. Appl. Med. Care 1995, 814–818. 28. Della Mea,
V. Internet electronic mail: A tool for lowcost
telemedicine. Telemed. Telecare 1999, 5 (2), 84–89. 29.
High, W.A.; Houston, M.S.; Calobrisi, S.D.; Drage, L.A.;
McEvoy, M.T. Assessment of the accuracy of low-cost
store-and-forward teledermatology consultation. J. Am.
Acad. Dermatol. May 2000, 42 (5 Pt 1), 776–783. 30.
Kennelly, R.J. Improving acute care through use of medical
device data. Int. J. Med. Inf. Feb. 1998, 48 (1–3),
145–149. 31. Rossi Mori, A.; Consorti, F.; Galeazzi, E.
Standards to support development of terminological systems
for healthcare telematics. Methods Inf. Med. Nov. 1998, 37
(4–5), 551–563. 32. Chapman, S. XML and HIPAA: The data
wave of the future? Web-enable your data to strategically
position your organization for future e-health initiatives.
Health Manag. Technol. Feb. 2003, 24 (2), 14, 16. 33.
Wiederhold, G. Future of security and privacy in medical
information. Stud. Health Technol. Inform. 2002, 80,
213–229. 34. Moody, M. HIPAA strengthens business case for
electronic report distribution systems. J. Healthc. Inf.
Manag. 2002, 16 (3), 47–51. Summer. 35. Satava, R.M.
Virtual reality surgical simulator. Surg. Endosc. 1993, 7,
203–205. T
10 Tendons and Ligaments, Mechanical
Testing Of

6. Woo, S.L.; Debski, R.E.; Withrow, J.D.; Janaushet, M.A.


Biomechanics of knee ligaments. Am. J. Sports Med. 1999, 27
(4), 533–543.

7. Bonifasi-Lista, C.; Lake, S.P.; Small, M.S.; Weiss, J.A.


Viscoelastic properties of the human medial collateral
ligament under longitudinal, transverse and shear loading.
J. Orthop. Res. 2005, 23 (1), 67–76.

8. Butler, D.; Kay, M.; Stouffer, D. Comparison of material


properties in the fascicle-bone units from human patellar
tendon and knee ligaments. J. Biomech. 1986, 19, 425–432.

9. Bigliani, L.U.; Pollock, R.G.; Soslowsky, L.J.; Flatow,


E.L.; Pawluk, R.J.; Mow, V.C. Tensile properties of the
inferior glenohumeral ligament. J. Orthop. Res. 1992, 10
(2), 187–197.

10. Gupta, B.N.; Subramanian, K.N.; Brinker, W.O.; Gupta,


A.N. Tensile strength of canine cranial cruciate ligaments.
Am. J. Vet. Res. 1971, 32 (1), 183–190.

11. Hannafin, J.A.; Arnoczky, S.P. Effect of cyclic and


static tensile loading on water content and solute
diffusion in canine flexor tendons: an in vitro study. J.
Orthop. Res. 1994, 12 (3), 350–356.

12. Woo, S.L.; Hollis, J.M.; Adams, D.J.; Lyon, R.M.;


Takai, S. Tensile properties of the human femur-anterior
cruciate ligament-tibia complex. The effects of specimen
age and orientation. Am. J. Sports Med. 1991, 19 (3),
217–225.

13. Woo, S.L.; Lee, T.Q.; Gomez, M.A.; Sato, S.; Field,
F.P. Temperature dependent behavior of the canine medial
collateral ligament. J. Biomech. Eng. 1987, 109 (1), 68–71.

14. Woo, S.L.; Ohland, K.J.; Weiss, J.A. Aging and


sexrelated changes in the biomechanical properties of the
rabbit medial collateral ligament. Mech. Ageing Dev. 1990,
56 (2), 129–142.

15. Woo, S.L.; Orlando, C.A.; Gomez, M.A.; Frank, C.B.;


Akeson, W.H. Tensile properties of the medial collateral
ligament as a function of age. J. Orthop. Res. 1986, 4 (2),
133–141.
16. Weiss, J.A. A Constitutive Model and Finite Element
Representation for Transversely Isotropic Soft Tissues;
Bioengineering, University of Utah: Salt Lake City, 1994.

17. Race, A.; Amis, A.A. The mechanical properties of the


two bundles of the human posterior cruciate ligament. J.
Biomech. 1994, 27 (1), 13–24.

18. Pollock, R.G.; Wang, V.M.; Bucchieri, J.S.; Cohen,


N.P.; Huang, C.Y.; Pawluk, R.J.; Flatow, E.L.; Bigliani,
L.U.; Mow, V.C. Effects of repetitive subfailure strains on
the mechanical behavior of the inferior glenohumeral
ligament. J. Shoulder Elbow Surg. 2000, 9 (5), 427–435.

19. Lynch, H.A.; Johannessen, W.; Wu, J.P.; Jawa, A.;


Elliott, D.M. Effect of fiber orientation and strain rate on
the nonlinear uniaxial tensile material properties of
tendon. J. Biomech. Eng. 2003, 125 (5), 726–731.

20. Lee, T.Q.; Dettling, J.; Sandusky, M.D.; McMahon, P.J.


Age related biomechanical properties of the
glenoid-anterior band of the inferior glenohumeral
ligament-humerus complex. Clin. Biomech. 1999, 14 (7),
471–476. 21. Moore, S.M.; McMahon, P.J.; Azemi, E.; Debski,
R.E. Bi-directional mechanical properties of the posterior
region of the glenohumeral capsule. J. Biomech. 2005, 38
(6), 1365–1369. 22. Moore, S.M.; McMahon, P.J.; Debski,
R.E. Bi-directional mechanical properties of the axillary
pouch of the glenohumeral capsule: implications for
surgical repair. J. Biomech. Eng. 2004, 126 (2), 284–288.
23. Quapp, K.M.; Weiss, J.A. Material characterization of
human medial collateral ligament. J. Biomech. Eng. 1998,
120 (6), 757–763. 24. Woo, S.L.; Akeson, W.H.; Jemmott,
G.F. Measurements of nonhomogeneous, directional mechanical
properties of articular cartilage in tension. J. Biomech.
1976, 9 (12), 785–791. 25. Njus, G.; NM, N. A non-contact
method for determining cross sectional area of soft
tissues. Orthop. Res. Soc. 1986, 11, 126. 26. Race, A.;
Amis, A.A. Cross-sectional area measurement of soft tissue.
A new casting method. J. Biomech 1996, 29 (9), 1207–1212.
27. Allard, P.; Thiry, P.S.; Bourgault, A.; Drouin, G.
Pressure dependence of ‘‘the area micrometer’’ method in
evaluation of cruciate ligament cross-section. J. Biomed.
Eng. 1979, 1 (4), 265–267. 28. Ellis, D. Cross sectional
area measurements for tendon specimens—a comparison of
several methods. J. Biomech. 1969, 2, 175–186. 29. Walker,
L.; Harris, E.; Benedict, J. Stress strain relationship in
human cadaveric plantaris tendon—a preliminary study. Med.
Elect. Biol. Eng. 1964, 2, 31–38. 30. Ellis, D.G.
Cross-sectional area measurements for tendon specimens: A
comparison of several methods. J. Biomech 1969, 2, 175–186.
31. Iaconis, F.; Steindler, R.; Marinozzi, G. Measurements
of cross-sectional area of collagen structures (knee
ligaments) by means of an optical method. J. Biomech. 1987,
20 (10), 1003–1010. 32. Lee, T.Q.; Woo, S.L. A new method
for determining cross-sectional shape and area of soft
tissues. J. Biomech. Eng. 1988, 110 (2), 110–114. 33. Woo,
S.L.; Danto, M.I.; Ohland, K.J.; Lee, T.Q.; Newton, P.O.
The use of a laser micrometer system to determine the
cross-sectional shape and area of ligaments: a comparative
study with two existing methods. J. Biomech. Eng. 1990, 112
(4), 426–431. 34. Chan, S.; Livesay, G.; Morrow, D.; Woo,
S.L-Y. The development of a low cost laser reflectance
system to determine the cross sectional shape and area of
soft tissues. ASME Adv. Bioeng. San Francisco 1995, 31,
123–124. 35. Beynnon, B.D.; Fleming, B.C.; Johnson, R.J.;
Nichols, C.E.; Renstrom, P.A.; Pope, M.H. Anterior cruciate
ligament strain behavior during rehabilitation exercises in
vivo. Am. J. Sports Med. 1995, 23 (1), 24–34. 36. Beynnon,
B.; Howe, J.G.; Pope, M.H.; Johnson, R.J.; Fleming, B.C.
The measurement of anterior cruciate ligament strain in
vivo. Int. Orthop. 1992, 16 (1), 1–12. 37. Harner, C.D.;
Xerogeanes, J.W.; Livesay, G.A.; Carlin, G.J.; Smith, B.A.;
Kusayama, T.; Kashiwaguchi, S.; Woo, S.L. The human
posterior cruciate ligament complex: an interdisciplinary
study. Ligament morphology and biomechanical evaluation.
Am. J. Sports Med. 1995, 23 (6), 736–745. 38. Scheffler,
S.U.; Clineff, T.D.; Papageorgiou, C.D.; Debski, R.E.;
Benjamin, C.; Woo, S.L. Structure and function of the
healing medial collateral ligament in a goat model. Ann.
Biomed. Eng. 2001, 29 (2), 173–180. 39. Lam, T.C.; Frank,
C.B.; Shrive, N.G. Calibration characteristics of a video
dimension analyser (VDA) system. J. Biomech. 1992, 25 (10),
1227–1231. 40. Smutz, W.P.; Drexler, M.; Berglund, L.J.;
Growney, E.; An, K.N. Accuracy of a video strain
measurement system. J. Biomech. 1996, 29 (6), 813–817. 41.
Yin, F.C.; Tompkins, W.R.; Peterson, K.L.; Intaglietta, M.
A video-dimension analyxer. IEEE Trans. Biomed. Eng. 1972,
19 (5), 376–381. 42. Abramowitch, S.D.; Yagi, M.; Tsuda,
E.; Woo, S.L. The healing medial collateral ligament
following a combined anterior cruciate and medial
collateral ligament injury—a biomechanical study in a goat
model. J. Orthop. Res. 2003, 21 (6), 1124–1130. 43.
Whittaker, P.; Canham, P.B. Demonstration of quantitative
fabric analysis of tendon collagen using twodimensional
polarized light microscopy. Matrix 1991, 11 (1), 56–62. 44.
Woo, S.L.; Peterson, R.H.; Ohland, K.J.; Sites, T.J.;
Danto, M.I. The effects of strain rate on the properties of
the medial collateral ligament in skeletally immature and
mature rabbits: a biomechanical and histological study. J.
Orthop. Res. 1990, 8 (5), 712–721. 45. Johnson, G.A.;
Tramaglini, D.M.; Levine, R.E.; Ohno, K.; Choi, N.Y.; Woo,
S.L. Tensile and viscoelastic properties of human patellar
tendon. J. Orthop. Res. 1994, 12 (6), 796–803. 46. Figgie,
H.E., 3rd; Bahniuk, E.H.; Heiple, K.G.; Davy, D.T. The
effects of tibial-femoral angle on the failure mechanics of
the canine anterior cruciate ligament. J. Biomech. 1986, 19
(2), 89–91. 47. Moon, D.; Woo, S.L.; Gabriel, M.;
Abramowitch, S.; Takakura, Y. The effects of refreezing on
the viscoelastic and tensile properties of ligaments.
Journal of Biomechanics 2005. 48. Parry, D.A. The molecular
and fibrillar structure of collagen and its relationship to
the mechanical properties of connective tissue. Biophys.
Chem. 1988, 29 (1–2), 195–209. 49. Debski, R.E.; Moore,
S.M.; Mercer, J.L.; Sacks, M.S.; McMahon, P.J. The collagen
fibers of the anteroinferior capsulolabrum have multiaxial
orientation to resist shoulder dislocation. J. Shoulder
Elbow Surg. 2003, 12 (3), 247–252. 50. Sacks, M.S.; Smith,
D.B.; Hiester, E.D. A small angle light scattering device
for planar connective tissue microstructural analysis. Ann.
Biomed. Eng. 1997, 25 (4), 678–689. 51. Hirokawa, S.;
Tsuruno, R. Three-dimensional deformation and stress
distribution in an analytical/computational model of the
anterior cruciate ligament. J. Biomech. 2000 , 33 (9),
1069–1077. 52. Hurschler, C.; Loitz-Ramage, B.; Vanderby,
R., Jr. A structurally based stress-stretch relationship
for tendon and ligament. J. Biomech. Eng. 1997, 119 (4),
392–399. 53. Kohles, S.S.; Thielke, R.J.; Vanderby, R., Jr.
Finite elasticity formulations for evaluation of
ligamentous tissue. Biomed. Mater. Eng. 1997, 7 (6),
387–390. 54. Lanir, Y. A microstructure model for the
rheology of mammalian tendon. J. Biomech. Eng. 1980, 102
(4), 332–339. 55. Puso, M.A.; Weiss, J.A. Finite element
implementation of anisotropic quasi-linear viscoelasticity
using a discrete spectrum approximation. J. Biomech. Eng.
1998, 120 (1), 62–70. 56. Simbeya, K.W.; Shrive, N.G.;
Frank, C.B.; Matyas, J.R. Recent Advances in Computer
Methods in Biomechanics and Biomedical Engineering; Gordon
and Breach: Swansea, U.K., 1992; 240–249. 57. Weiss, J.A.;
Maker, B.N.; Govindjee, S. Finite element implementation of
incompressible, transversely isotropic hyperelasticity.
Computer Methods Appl. Mech. Eng. 1996, 135, 107–128. 58.
Weiss, J.A.; Gardiner, J.C.; Bonifasi-Lista, C. Ligament
material behavior is nonlinear, viscoelastic and
rateindependent under shear loading. J. Biomech. 2002, 35
(7), 943–950. 59. Gardiner, J.C.; Weiss, J.A.
Subject-specific finite element analysis of the human medial
collateral ligament during valgus knee loading. J. Orthop.
Res. 2003, 21 (6), 1098–1106. 60. Yin, L.; Elliott, D.M. A
biphasic and transversely isotropic mechanical model for
tendon: application to mouse tail fascicles in uniaxial
tension. J. Biomech. 2004, 37 (6), 907–916. 61. Vande
Geest, J.P.; Sacks, M.S.; Vorp, D.A. The effects of
aneurysm on the biaxial mechanical behavior of human
abdominal aorta. J. Biomech. 2005. 62. Vande Geest, J.P.;
Sacks, M.S.; Vorp, D.A. Age dependency of the biaxial
biomechanical behavior of human abdominal aorta. J.
Biomech. Eng. 2004, 126 (6), 815–822. 63. Sacks, M.S.; Sun,
W. Multiaxial mechanical behavior of biological materials.
Ann. Rev. Biomed. Eng. 2003, 5, 251–284. 64. Sacks, M.S.;
Chuong, C.J. Biaxial mechanical properties of passive right
ventricular free wall myocardium. J. Biomech. Eng. 1993,
115 (2), 202–205. 65. Lanir, Y. Constitutive equations for
the lung tissue. J. Biomech. Eng. 1983, 105 (4), 374–380.
66. Debski, R.E.; Weiss, J.A.; Newman, W.J.; Moore, S.M;
McMahon, P.J. Stress and strain in the anterior band of the
inferior glenohumeral ligament during a simulated clinical
examination. J. Shoulder Elbow Surg. 2005, 14 (1 suppl S),
24S–31S. 67. Wang, D.H.; Makaroun, M.S.; Webster, M.W.;
Vorp, D.A. Effect of intraluminal thrombus on wall stress
in patient-specific models of abdominal aortic aneurysm. J.
Vasc. Surg. 2002, 36 (3), 598–604. 68. Song, Y.; Debski,
R.E.; Musahl, V.; Thomas, M.; Woo, S.L. A three-dimensional
finite element model of the human anterior cruciate
ligament: a computational T analysis with experimental
validation. J. Biomech. 2004, 37 (3), 383–390.

69. Stabile, K.J.; Pfaeffle, H.J.; Weiss, J.A.; Gabriel,


M.T.; Tomanio, M.M.; Fischer, K.J. Longitudinal and
transverse mechanical properties of the interosseous
ligament of the forearm. Bioengineering Conference ASME,
2001; 363–364.

70. Hewitt, J.; Guilak, F.; Glisson, R.; Vail, T.P.


Regional material properties of the human hip joint capsule
ligaments. J. Orthop. Res. 2001, 19 (3), 359–364.

71. Gardiner, J.C.; Weiss, J.A. Simple shear testing of


parallel-fibered planar soft tissues. J. Biomech. Eng. 2001,
123 (2), 170–175.

72. Cohen, R.E.; Hooley, C.J.; McCrum, N.G. Viscoelastic


creep of collagenous tissue. J. Biomech. 1976, 9 (4),
175–184.

73. Abramowitch, S.D.; Woo, S.L.; Clineff, T.D.; Debski,


R.E. An evaluation of the quasi-linear viscoelastic
properties of the healing medial collateral ligament in a
goat model. Ann. Biomed. Eng. 2004, 32 (3), 329–335.
74. Provenzano, P.; Lakes, R.; Keenan, T.; Vanderby, R.,
Jr. Nonlinear ligament viscoelasticity. Ann. Biomed. Eng.
2001, 29 (10), 908–914.

75. Thornton, G.M.; Oliynyk, A.; Frank, C.B.; Shrive, N.G.


Ligament creep cannot be predicted from stress relaxation
at low stress: a biomechanical study of the rabbit medial
collateral ligament. J. Orthop. Res. 1997, 15 (5), 652–656.

76. Fung, Y.C. Biomechanics: Mechanical Properties of


Living Tissues, 2nd Ed.; Springer: New York, NY, 1993.

77. Thomopoulos, S.; Williams, G.R.; Gimbel, J.A.; Favata,


M.; Soslowsky, L.J. Variation of biomechanical, structural,
and compositional properties along the tendon to bone
insertion site. J. Orthop. Res. 2003, 21 (3), 413–419.

78. Elliott, D.M.; Robinson, P.S.; Gimbel, J.A.; Sarver,


J.J.; Abboud, J.A.; Lozzo, R.V.; Soslowsky, L.J. Effect of
altered matrix proteins on quasilinear viscoelastic
properties in transgenic mouse tail tendons. Ann. Biomed.
Eng. 2003, 31 (5), 599–605. 79. Kwan, M.K.; Lin, T.H.; Woo,
S.L. On the viscoelastic properties of the anteromedial
bundle of the anterior cruciate ligament. J. Biomech. 1993,
26 (4–5), 447–452. 80. Funk, J.R.; Hall, G.W.; Crandall,
J.R.; Pikley, W.D. Linear and quasi-linear viscoelastic
characterization of ankle ligaments. J. Biomech. Eng. 2000,
122 (1), 15–22. 81. Abramowitch, S.D.; Woo, S.L. An
improved method to analyze the stress relaxation of
ligaments following a finite ramp time based on the
quasi-linear viscoelastic theory. J. Biomech. Eng. 2004,
126 (1), 92–97. 82. Woo, S.L.; Gomez, M.A.; Akeson, W.H.
The time and history-dependent viscoelastic properties of
the canine medical collateral ligament. J Biomech. Eng.
1981, 103 (4), 293–298. 83. Provenzano, P.P.; Lakes, R.S.;
Corr, D.T.; R, R., Jr. Application of nonlinear
viscoelastic models to describe ligament behavior. Biomech.
Model Mechanobiol. 2002, 1 (1), 45–57. 84. Sarver, J.J.;
Robinson, P.S.; Elliott, D.M. Methods for quasi-linear
viscoelastic modeling of soft tissue: application to
incremental stress-relaxation experiments. J. Biomech. Eng.
2003, 125 (5), 754–758. 85. Findley, W.N.; Lai, J.S.;
Onaran, K. Creep and Relaxation of Nonlinear Viscoelastic
Materials; Dover: New York, 1973. 86. Myers, B.S.;
McElhaney, J.H.; Doherty, B.J. The viscoelastic responses
of the human cervical spine in torsion: experimental
limitations of quasi-linear theory, and a method for
reducing these effects. J. Biomech. 1991, 24 (9), 811–817.
87. Nigul, I.; Nigul, U. On algorithms of evaluation of
Fung’s relaxation function parameters. J. Biomech. 1987, 20
(4), 343–352.
Thrombosis

11. Higuchi, D.A.; Wun, T.C.; Likert, K.M.; Broze, G.J.,


Jr. The effect of leukocyte elastase on tissue factor
pathway inhibitor. Blood 1992, 79 (7), 1712–1719.

12. Zillmann, A.; Luther, T.; Muller, I.; Kotzsch, M.;


Spannagl, M.; Kauke, T.; Oelschlagel, U.; Zahler, S.;
Engelmann, B. Platelet-associated tissue factor contributes
to the collagen-triggered activation of blood coagulation.
Biochem. Biophys. Res. Commun. 2001, 281 (2), 603–609.

13. Palabrica, T.; Lobb, R.; Furie, B.C.; Aronovitz, M.;


Benjamin, C.; Hsu, Y.M.; Sajer, S.A.; Furie, B. Leukocyte
accumulation promoting fibrin deposition is mediated in vivo
by P-selectin on adherent platelets. Nature 1992, 359
(6398), 848–851.

14. Taylor, A.D.; Neelamegham, S.; Hellums, J.D.; Smith,


C.W.; Simon, S.I. Molecular dynamics of the transition from
L-Selectin to b2-Integrin dependent neutrophil adhseion
under defined hydrodynamic shear. Biophys. J. 1996, 71 (6),
3488–3500.

15. Schmidtke, D.W.; Diamond, S.L. Direct observation of


membrane tethers formed during neutrophil attachment to
platelets or P-selectin under physiological flow. J. Cell
Biol. 2000, 149 (3), 719–730.

16. Park, E.Y.; Smith, M.J.; Stropp, E.S.; Snapp, K.R.;


DiVietro, J.A.; Walker, W.F.; Schmidtke, D.W.; Diamond,
S.L.; Lawrence, M.B. Comparison of PSGL-1 microbead and
neutrophil rolling: Microvillus elongation stabilizes
P-selectin bond clusters. Biophys. J. 2002, 82 (4),
1835–1847.

17. Laurenzi, I.J.; Diamond, S.L. Monte Carlo simulation of


the heterotypic aggregation kinetics of platelets and
neutrophils. Biophys. J. 1999, 77 (3), 1733–1746.

18. Si-Tahar, M.; Pidard, D.; Balloy, V.; Moniatte, M.;


Kieffer, N.; Van Dorsselaer, A.; Chignard, M. Human
neutrophil elastase proteolytically activates the platelet
integrin alphaIIbeta3 through cleavage of the carboxyl
terminus of the alphaIIb subunit heavy chain. Involvement
in the potentiation of platelet aggregation. J. Biol. Chem.
1997, 272 (17), 11636–11647.

19. Yamamoto, J.; Ishii, I.; Okada, Y.; Yamashita, T.;


Ridler, C.D.; Gorog, P.; Kovacs, I.B. Effect of leukocyte
products on platelet thrombus formation, coagulation and
spontaneous thrombolysis, as measured from native human
blood in vitro. Thromb. Res. 1993, 71 (4), 281–287.

20. Tijburg, P.N.; Ijsseldijk, M.J.; Sixma, J.J.; de Groot,


P.G. Quantification of fibrin deposition in flowing blood with
peroxidase-labeled fibrinogen. High shear rates induce
decreased fibrin deposition and appearance of fibrin
monomers. Arterioscler. Thromb. 1991, 11 (2), 211–220.

21. Ouriel, K.; Donayre, C.; Shortell, C.K.; Cimino, C.;


Donnelly, J.; Oxley, D.; Green, R.M. The hemodynamics of
thrombus formation in arteries. J. Vasc. Surg. 1991, 14
(6), 757–762.

22. Kuijper, P.H.; Gallardo Torres, H.I.; Lammers, J.W.;


Sixma, J.J.; Koenderman, L.; Zwaginga, J.J. Platelet and
fibrin deposition at the damaged vessel wall: Cooperative
substrates for neutrophil adhesion under flow conditions.
Blood 1997, 89 (1), 166–175. 23. Kirchhofer, D.;
Sakariassen, K.S.; Clozel, M.; Tschopp, T.B.; Hadvary, P.;
Nemerson, Y.; Baumgartner, H.R. Relationship between tissue
factor expression and deposition of fibrin, platelets, and
leukocytes on cultured endothelial cells under venous blood
flow conditions. Blood 1993, 81 (8), 2050–2058. 24.
Manodori, A.B.; Barabino, G.A.; Lubin, B.H.; Kuypers, F.A.
Adherence of phosphatidylserineexposing erythrocytes to
endothelial matrix thrombospondin. Blood 2000, 95 (4),
1293–1300. 25. Test, S.T.; Mitsuyoshi, J. Activation of the
alternative pathway of complement by calcium-loaded
erythrocytes resulting from loss of membrane phospholipid
asymmetry. J. Lab. Clin. Med. 1997, 130 (2), 169–182. 26.
Wun, T.; Paglieroni, T.; Field, C.L.; Welborn, J.; Cheung,
A.; Walker, N.J.; Tablin, F. Platelet-erythrocyte adhesion
in sickle cell disease. J. Investig. Med. 1999, 47 (3),
121–127. 27. Hu, W.J.; Eaton, J.W.; Ugarova, T.P.; Tang, L.
Molecular basis of biomaterial-mediated foreign body
reactions. Blood 2001, 98 (4), 1231–1238. 28. Goel, M.S.;
Diamond, S.L. Neutrophil enhancement of fibrin deposition
under flow through platelet-dependent and -independent
mechanisms. Arterioscler. Thromb. Vasc. Biol. 2001, 21
(12), 2093–2098. 29. Allen, D.H.; Tracy, P.B. Human
coagulation factor V is activated to the functional
cofactor by elastase and cathepsin G expressed at the
monocyte surface. J. Biol. Chem. 1995, 270 (3), 1408–1415.
30. Henderson, L.M.; Figueroa, C.D.; Muller-Esterl, W.;
Bhoola, K.D. Assembly of contact-phase factors on the
surface of the human neutrophil membrane. Blood 1994, 84
(2), 474–482. 31. Wachtfogel, Y.T.; Kucich, U.; James,
H.L.; Scott, C.F.; Schapira, M.; Zimmerman, M.; Cohen,
A.B.; Colman, R.W. Human plasma kallikrein releases
neutrophil elastase during blood coagulation. J. Clin.
Invest. 1983, 72 (5), 1672–1677. 32. Goldsmith, H.L.;
Turitto, V.T. Rheological aspects of thrombosis and
haemostasis: Basic principles and applications.
ICTH-report—Subcommittee on rheology of the International
Committee on Thrombosis and Haemostasis. Thromb. Haemost.
1986, 55 (3), 415–435. 33. Selak, M.A. Neutrophil elastase
potentiates cathepsin G-induced platelet activation.
Thromb. Haemost. 1992, 68 (5), 570–576. 34. Goel, M.S.;
Diamond, S.L. Neutrophil cathepsin G promotes
prothrombinase and fibrin formation under flow by activating
fibrinogen-adherent platelets. J. Biol. Chem. 2003, 278
(11), 9458–9463. 35. Sumner, W.T.; Monroe, D.M.; Hoffman,
M. Variability in platelet procoagulant activity in healthy
volunteers. Thromb. Res. 1996, 81 (5), 533–543. 36. Monroe,
D.M.; Hoffman, M.; Roberts, H.R. Platelets and thrombin
generation. Arterioscler. Thromb. Vasc. Biol. 2002, 22 (9),
1381–1389. 37. Goel, M.S.; Diamond, S.L. Adhesion of normal
erythrocytes at depressed venous shear rates to activated
neutrophils, activated platelets, and fibrin polymerized
from plasma. Blood 2002, 100 (10), 3797–3803.
Tinnitus Devices

5. Shulman, A. A final common pathway for tinnitus— The


medial temporal lobe system. Int. Tinnitus J. 1995, 2 (1),
115–126.

6. Vernon, J.A.; Meikle, M.B. Tinnitus Masking. In Tinnitus


Handbook; Tyler, R.S., Ed.; Singular: San Diego, 2000;
313–355.

7. Vernon, J. The history of masking as applied to


tinnitus. J. Laryngol. Otol. Suppl. 1981, 1 (4), 76–79.

8. Warren, R.M. Auditory Perception: A New Analysis and


Synthesis; Cambridge University Press: New York, 1999.

9. Feldmann, H. Homolateral and contralateral masking of


tinnitus by noise bands and pure tones. Audiology 1971, 4
(10), 138–144.

10. Tyler, R.S.; Conrad-Armes, D.; Smith, P. Postmasking


effects of sensorineural tinnitus: A preliminary
investigation. J. Speech Hear. Res. 1984, 1 (27), 466–474.

11. Penner, M.J. Masking of tinnitus and central masking.


J. Speech Hear. Res. 1987, 1 (30), 147–152.

12. Smith, P.A.; Parr, V.M.; Lutman, M.E.; Coles, R.R.A.


Comparative study of four noise spectra as potential
tinnitus maskers. Br. J. Audiol. 1991, 1 (25), 25–34.

13. Kitajima, K.; Kitahara, M.; Kodama, A. Can tinnitus be


masked by band erased filtered masker? Masking tinnitus with
sounds not covering the tinnitus frequency. Am. J. Otol.
1997, 1, 203–206.

14. Holgers, K.M.; Hakansson, B.E. Sound stimulation via


bone conduction for tinnitus relief: A pilot study. Int. J.
Audiol. 2002, 41 (5), 293–300.

15. Cai, Z.; Richards, D.G.; Lenhardt, M.L.; Madsen, A.G.


Response of human skull to bone conducted sound in the
audiometric to ultrasonic range. Int. Tinnitus J. 2002, 8
(1), 1–8.

16. Johnson, R.M.; Hughes, F.M. Diotic Versus Dichotic


Masking of Tinnitus: Tinnitus 91, Proceedings of the Fourth
International Tinnitus Seminar; Aran, J.-M., Dauman, R.,
Eds.; Kugler Publications: New York, 1991; 387–390.
17. Vernon, J.A. Tinnitus Treatment and Relief; Vernon,
J.J., Ed.; Allyn and Bacon: Boston, 1998.

18. Goldstein, B.A.; Shulman, A.; Lenhardt, M.L.; Richards,


D.G.; Madsen, A.G.; Guinta, R. Long-term inhibition of
tinnitus by UltraQuiet therapy: Preliminary report. Int.
Tinnitus J. 2001, 2 (7), 122–127.

19. Hazell, J.W.P.; Wood, S. Tinnitus masking—A significant


contribution to tinnitus management. Br. J. Audiol. 1981, 4
(15), 223–230.

20. Terry, A.M.P.; Jones, D.M.; David, B.R.; Slater, R.


Parametric studies of tinnitus masking and residual
inhibition. Br. J. Audiol. 1983, 1 (17), 245–256.

21. Tyler, R.S. Tinnitus in the profoundly hearing impaired


and the effects of cochlear implants. Ann. Otol. Rhinol.
Laryngol. 1995, 165 (104), 25–30.

22. Ito, J.l.; Sakakihara, L. Suppression of tinnitus bt


cochlear implantation. Am. J. Otolaryngol. 1994, 1 (15),
145–148.

23. Shulman, A.; Tonndorf, J.; Goldstein, B. Electric


tinnitus control. Acta Oto-Laryngol. 1985, 3–4 (99), 3–4,
318–325. 24. Vernon, J.A.; Fenwick, J.A. Attempts to
suppress tinnitus with transcutaneous electrical
stimulation. Otolaryngol. Head Neck Surg. 1985, 3 (93), 9,
385–389. 25. Dobie, R.A.; Hoberg, K.E.; Rees, T.S.
Electrical tinnitus suppression: A double blind crossover
study. Otolaryngol. Head Neck Surg. 1986, 3 (95), 319–323.
26. Steenerson, R.L.; Cronin, G.W. Treatment of tinnitus
with electrical stimulation. Otolaryngol. Head Neck Surg.
1999, 5 (121), 511–513. 27. Al-Jassim, A.H. The use of
Walkman mini-stereo system as a tinnitus masker. J.
Laryngol. Otol. 1988, 102 (1), 27–28. 28. Shulman, A.;
Strashun, A. Descending auditory system/
cerebellum/tinnitus. Int. Tinnitus J. 1999, 5 (1), 92–106.
29. Thomas, M.; Laurell, G.; Lundeberg, T. Vibratory
stimulation as a treatment alternative in patients with
tinnitus. Ear Nose Throat J. 1989, 68 (11), 810–814,
817–818. 30. Kanold, P.; Young, E.D. Proprioceptive
information from the pinna provides somatosensory input to
cat dorsal cochlear nucleus. J. Neurosci. 2001, 19 (21),
7848–7858. 31. Lenhardt, M.L.; Goldstein, B.A.; Shulman,
A.; Guinta, R. Use of high-frequency and muscle vibration
in the treatment of tinnitus. Int. Tinnitus J. 2003, 1 (9),
1, 32–36. 32. Meikle, M.B.; Edlefsen, L.L.; Lay, J.W.
Suppression of Tinnitus by Bone Conduction of Ultrasound.
In Abstracts of the 21st Annual Meeting of the Association
for Research in Otolaryngology; , 1999; 223. 33. Lenhardt,
M.L.; Skellett, R.; Wang, P.; Clarke, A.M. Human ultrasonic
speech perception. Science 1991, 5015 (253), 82–85. 34.
Hakansson, B.; Tjellstrom, A.; Rosenhall, U. Acceleration
levels at hearing threshold with direct bone conduction vs.
conventional bone conduction. Acta OtoLaryngol. 1985, 3–4
(100), 240–252. 35. Reference Equivalent Threshold Force
Levels for Audiometric Bone Vibrators (ANSI S3.26);
American National Standards Institute: New York, 1981. 36.
Corso, J. Bone-conduction thresholds for sonic and
ultrasonic frequencies. J. Acoust. Soc. Am. 1963 , 4 (35),
1738–1743. 37. Lenhardt, M.L.; Richards, D.G.; Madsen,
A.G.; Goldstein, B.A.; Shulman, A.; Guinta, R. Measurement
of bone conduction levels for high frequency. Int. Tinnitus
J. 2002, 1 (8), 1, 9–12. 38. Occupational Safety and Health
Administration Technical Manual; Ultrasonics, U.S.
Department of Labor: Washington, DC, 2002. Section III,
Chapter 5, Subchapter V. 39. Hosoi, H.; Imaizumi, S.;
Sakaguchi, T. Activation of the auditory cortex by
ultrasound. Lancet 1998, 9101 (351), 496–497. 40. Imaizumi,
S.; Hosoi, H.; Sakaguchi, T. Ultrasound activated the
auditory cortex of profoundly deaf subjects. NeuroReport
2001, 12 (3), 583–586. 41. Borth, D.E.; Cain, C.A.
Theoretical analysis of acoustic signal generation in
materials irradiated with microwave energy. IEEE Trans.
Microwave Theor. Tech. 1977, 525 (11), 944–954. 42. Olsen,
R.G.; Lin, J.C. Microwave pulse induced acoustic resonances
in spherical models. IEEE Trans. Microwave Theor. Tech.
1981, 29 (10), 114–117. 43. Ranke, O.F. Physiologie des
Gehors. In Gehor StimmeSprache; Ranke, O.F., Lullies, H.,
Eds.; Springer: Berlin, 1953; 3–110. 44. Tonndorf, J. A new
concept of bone conduction. Arch. Otol. 1968, 2 (87),
49–54. 45. Tonndorf, J. Bone conduction. Studies in
experimental animals. Acta Oto-Laryngol., Suppl. 1966, 213.
46. Magee, T.R.; Davies, A.H. Auditory phenomena during
transcranial Doppler insonation of the basilar artery. J.
Ultrasound Med. 1993, 12 (12), 747–750. 47. Fay, R.R.
Structure and Function in Sound Discrimination Among
Vertebrates. In The Evolutionary Biology of Hearing;
Webster, D.E., Fay, R.R., Popper, A.N., Eds.;
Springer-Verlag: New York, 1992; 245–250. 48. Watson, S.R.;
Halmagyi, G.M.; Colebatch, J.G. Vestibular hypersensitivity
to sound (Tullio phenomenon): Structural and functional
assessment. Neurology 2000, 3 (54), 722–728. 49. Lim, D.
The Development and Structure of the Otoconia. In
Ultrastructural Atlas of the Inner Ear; Friedmann, I.,
Ballantyne, J., Eds.; Butterworth: London, 1984; 245–269.
50. Sheykholeslami, K.; Kaga, K. The otolithic organ as a
receptor of vestibular hearing revealed by
vestibular-evoked myogenic potentials in patients with
inner ear anomalies. Hear. Res. 2002, 165 (1–2), 62–67. 51.
Schwaber, M.K.; Garraghty, P.E.; Kaas, J.H. Neuroplasticity
of the adult primary auditory cortex following cochlear
hearing loss. Am. J. Otol. 1993, 3 (14), 252–258. 52.
Muhlnickel, W.; Elbert, T.; Taub, E.; Flor, H.
Reorganization of auditory cortex in tinnitus. Proc. Natl.
Acad. Sci. U. S. A. 1998, 17 (95), 10340–10343. 53. Mirz,
F.G.; Jedde, A.; Ishizu, K.; Pedersen, C.B. Cortical
networks subserving the perception of tinnitus. Acta
Oto-Laryngol., Suppl. 2000, (543), 241–243. 54. Stegiade,
M. Coherent continuations and short term plasticity in
corticothalamic networks. Tr. Neurosci. 1999, 1 (22),
337–345. 55. Menning, H.; Roberts, L.E.; Pantev, C. Plastic
changes in the auditory cortex induced by intensive
frequency discrimination training. NeuroReport 2002, 4
(11), 817–822. 56. Di Mino, A. Electronic Stimulation
System for Treating Tinnitus Disorders. United States
Patent 6,210,321, April 3, 2001. T
Tissue Engineering

19. Thomson, J.A.; Itskovitz-Eldor, J.; Shapro, S.S.;


Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M.
Embryonic stem cell lines derived from human blastocysts.
Science 1998, 282, 115–117.

20. Bishop, A.E.; Buttery, L.D.; Polak, J.M. Embryonic stem


cells. J. Pathol. 2002, 197 (4), 424–429.

21. Poulsom, R.; Alison, M.R.; Forbes, S.J.; Wright, N.A.


Adult stem cell plasticity. J. Pathol. 2002, 197 (4),
441–456.

22. Auger, F.A.; Lopez-Valle, C.A.; Guignard, R.; Tremblay,


N.; Noe¨l, B.; Goulet, F.; Germain, L. Skin equivalent
produced with human collagens. In Vitro Cell. Dev. Biol.,
Anim. 1995, 31, 432–439.

23. Karasek, M.A.; Charlton, M.E. Growth of postembryonic


skin epithelial cells on collagen gels. J. Invest.
Dermatol. 1971, 56 (3), 205–210.

24. Auger, F.; Rouabhia, M.; Goulet, F.; Berthod, F.;


Moulin, V.; Germain, L. Tissue-engineered human skin
substitutes developed from collagen-populated hydrated
gels: Clinical and fundamental applications. Med. Biol.
Eng. Comput. 1998, 36, 801–812.

25. Auger, F.A.; Pouliot, R.; Tremblay, N.; Guignard, R.;


Noel, P.; Juhasz, J.; Germain, L.; Goulet, F. Multistep
production of bioengineered skin substitutes: Sequential
modulation of culture conditions. In Vitro Cell. Dev.
Biol., Anim. 2000, 36 (2), 96–103.

26. Lopez Valle, C.A.; Auger, F.A.; Rompre´, P.; Bouvard,


V.; Germain, L. Peripheral anchorage of dermal equivalents.
Br. J. Dermatol. 1992, 127, 365–371.

27. Bouvard, V.; Germain, L.; Rompre, P.; Roy, B.; Auger,
F.A. Influence of dermal equivalent maturation on the
development of a cultured skin equivalent. Biochem. Cell.
Biol. 1992, 70 (1), 34–42.

28. Tamariz, E.; Grinnell, F. Modulation of fibroblast


morphology and adhesion during collagen matrix remodeling.
Mol. Biol. Cell 2002, 13 (11), 3915–3929.

29. Goulet, F.; Germain, L.; Caron, C.; Rancourt, D.;


Normand, A.; Auger, F. Tissue-Engineered Ligament. In
Ligaments and Ligamentoplasties; Yahia, L., Ed.;
Springer-Verlag: Berlin, 1997; 367–377.

30. Goulet, F.; Germain, L.; Rancourt, D.; Caron, C.;


Normand, A.; Auger, F.A. Tendons and Ligaments. In Textbook
of Tissue Engineering; Lanza, R., Langer, R., Chick, W.,
Eds.; Academic Press Ltd.: San Diego, 1997; 633–644.

31. Girton, T.S.; Oegema, T.R.; Grassl, E.D.; Isenberg,


B.C.; Tranquillo, R.T. Mechanisms of stiffening and
strengthening in media-equivalents fabricated using
glycation. J. Biomech. Eng. 2000, 122 (3), 216–223.

32. Tranquillo, R.T.; Girton, T.S.; Bromberek, B.A.;


Triebes, T.G.; Mooradian, D.L. Magnetically orientated
tissue-equivalent tubes: Application to a circumferentially
orientated media-equivalent. Biomaterials 1996, 17 (3),
349–357.

33. Seliktar, D.; Black, R.A.; Vito, R.P.; Nerem, R.M.


Dynamic mechanical conditioning of collagen-gel blood
vessel constructs induces remodeling in vitro. Ann. Biomed.
Eng. 2000, 28 (4), 351–362. 34. Niklason, L.E.; Gao, J.;
Abbott, W.M.; Hirschi, K.K.; Houser, S.; Marini, R.;
Langer, R. Functional arteries grown in vitro. Science
1999, 284, 489–493. 35. Yannas, I.V.; Burke, J.F. Design of
an artificial skin: Basic design principles. J. Biomed.
Mater. Res. 1980, 14, 65–81. 36. Yannas, I.V.; Lee, E.;
Orgill, D.P.; Skrabut, E.M.; Murphy, G.F. Synthesis and
characterization of a model extracellular matrix that
induces partial regeneration of adult mammalian skin. Proc.
Natl. Acad. Sci. U. S. A. 1989, 86 (3), 933–937. 37.
Berthod, F.; Hayek, D.; Damour, O.; Collombel, C. Collagen
synthesis by fibroblasts cultured within a collagen sponge.
Biomaterials 1993, 14, 749–754. 38. Berthod, F.; Sahuc, F.;
Hayek, D.; Damour, O.; Collombel, C. Deposition of collagen
fibril bundles by long-term culture of fibroblasts in a
collagen sponge. J. Biomed. Mater. Res. 1996, 32, 87–94.
39. Green, H.; Kehinde, O.; Thomas, J. Growth of cultured
human epidermal cells into multiple epithelia suitable for
grafting. Proc. Natl. Acad. Sci. U.S.A. 1979, 76 (11),
5665–5668. 40. L’Heureux, N.; Paˆquet, S.; Labbe´, R.;
Germain, L.; Auger, F.A. A completely biological
tissue-engineered human blood vessel. FASEB J. 1998, 12,
47–56. 41. Meinhart, J.G.; Deutsch, M.; Fischlein, T.;
Howanietz, N.; Froschl, A.; Zilla, P. Clinical autologous
in vitro endothelialization of 153 infrainguinal ePTFE
grafts. Ann. Thorac. Surg. 2001, 71 (5 Suppl.), S327–S331.
42. Black, A.F.; Berthod, F.; L’Heureux, N.; Germain, L.;
Auger, F.A. In vitro reconstruction of a human
capillary-like network in a tissue-engineered skin
equivalent. FASEB J. 1998, 12, 1331–1340. 43. Young, D.M.;
Greulich, K.M.; Weier, H.G. Speciesspecific in situ
hybridization with fluorochrome-labeled DNA probes to study
vascularization of human skin grafts on athymic mice. J.
Burn Care Rehabil. 1996, 17, 305–310. 44. Auger, F.A.;
Black, A.; Berthod, F.; L’Heureux, N.; Germain, L. In Vitro
Reconstruction of a Human Capillary-Like Network in a
Tissue-Engineered Skin Equivalent. World Congress on
Medical Physics and Biomedical Engineering, Nice, 1997. 45.
Supp, D.M.; Wilson-Landy, K.; Boyce, S.T. Human dermal
microvascular endothelial cells form vascular analogs in
cultured skin substitutes after grafting to athymic mice.
FASEB J. 2002, 16 (8), 797–804. 46. Compton, C.C.; Gill,
J.M.; Bradford, D.A.; Regauer, S.; Gallico, G.G.; O’Connor,
N.E. Skin regenerated from cultured epithelial autografts
on full-thickness burn wounds from 6 days to 5 years after
grafting. A light, electron microscopic and
immunohistochemical study. Lab. Invest. 1989, 60 (5),
600–612. 47. English, K.B.; Stayner, N.; Krueger, G.G.;
Tuckett, R.P. Functional innervation of cultured skin
grafts. J. Invest. Dermatol. 1992, 99 (2), 120–128. 48.
Gingras, M.; Paradis, I.; Berthod, F. Nerve renegeration in
a collagen-chitosan tissue-engineered skin transplanted on
nude mice. Biomaterials 2003, 24, 1653–1661. 49.
Richardson, T.P.; Murphy, W.L.; Mooney, D.J. Polymeric
delivery of proteins and plasmid DNA for tissue engineering
and gene therapy. Crit. Rev. Eukaryot. Gene Expr. 2001, 11
(1–3), 47–58. 50. Rouabhia, M.; Germain, L.; Bergeron, J.;
Auger, F.A. Allogeneic-syngeneic cultured epithelia.
Transplantation 1995, 59 (9), 1229–1235. 51. L’Heureux, N.;
Germain, L.; Labbe, R.; Auger, F.A. In vitro construction
of a human blood vessel from cultured vascular cells: A
morphologic study. J. Vasc. Surg. 1993, 17 (3), 499–509.
52. L’Heureux, N.; Stoclet, J.C.; Auger, F.A.; Lagaud, G.;
Germain, L.; Andriantsitohaina, R. A human tissueengineered
vascular media: A new model for pharmacological studies of
contractile responses. FASEB J. 2001, 15, 515–524. T
Tissue Engineering: AC Electrokinetics

10. Sukhorukov, V.L.; Mussauer, H.; Zimmermann, U. The


effect of electrical deformation forces on the
electropermeabilization of erythrocyte membranes in lowand
high-conductivity media. J. Membrane Biol. 1998, 163 (3),
235–245.

11. Miller, R.D.; Jones, T.B. Electro-orientation of


ellipsoidal erythrocytes. Biophys. J. 1993, 64 (5),
1588–1595.

12. Pohl, H.A. Electrical forming of masses of living


cells. J. Colloid Interface Sci. 1972, 39 (2), 437–438.

13. Huang, Y.; Ho¨lzel, R.; Pethig, R.; Wang, X.B.


Differences in the AC electrodynamics of viable and
non-viable yeast cells determined through combined
dielectrophoresis and electrorotation studies. Phys. Med.
Biol. 1992, 37 (7), 1499–1517.

14. Gascoyne, P.R.C.; Vykoukal, J. Particle separation by


dielectrophoresis. Electrophoresis 2002, 23 (13),
1973–1983.

15. Docoslis, A.; Kalogerakis, N.; Behie, L.A.


Dielectrophoretic forces can be safely used to retain
viable cells in perfusion cultures of animal cells.
Cytotechnology 1999, 30 (1–3), 133–142.

16. Wang, X.B.; Huang, Y.; Burt, J.P.H.; Markx, G.H.;


Pethig, R. Selective dielectrophoretic confinement of
bioparticles in potential energy wells. J. Phys. D: Appl.
Phys. 1993, 26 (8), 1278–1285.

17. Fuhr, G.; Glasser, H.; Muller, T.; Schnell, T. Cell


manipulation and cultivation under a.c. electric field
influence in highly conductive culture media. Biochim.
Biophys. Acta 1994, 1201 (3), 353–360.

18. Mu¨ller, T.; Pfennig, A.; Klein, P.; Gradl G. Ja¨ger,


M.; Schnelle, T. The potential of dielectrophoresis for
single-cell experiments. IEEE Eng. Med. Biol. Mag. 2003, 22
(6), 51–61.

19. Gray, D.S.; Tan, J.L.; Voldman, J.; Chen, C.S.


Dielectrophoretic registration of living cells to a
microelectrode array. Biosens. Bioelectron. 2004, 19 (12),
1765–1774.
20. Heida, T.; Rutten, W.L.C.; Marani, E. Dielectrophoretic
trapping of dissociated fetal cortical rat neurons. IEEE
Trans. Biomed. Eng. 2001, 48 (8), 921–930.

21. Heida, T.; Vulto, P.; Rutten, W.L.; Marani, E.


Viability of dielectrophoretically trapped neural cortical
cells in culture. J. Neurosci. Methods 2001, 110 (1–2),
37–44.

22. Prasad, S.; Yang, M.; Zhang, X.; Ozkan, C.S.; Ozkan, M.
Electric field assisted patterning of neuronal networks for
the study of brain functions. Biomed. Microdevices 2003, 5
(2), 125–137.

23. Yu, Z.; Xiang, G.; Pan, L.; Huang, L.; Yu, Z.; Xing,
W.; Cheng, J. Negative dielectrophoretic force assisted
construction of ordered neuronal networks on cell
positioning bioelectronic chips. Biomed. Microdevices 2004,
6 (4), 311–324.

24. Albrecht, D.R.; Sah, R.L.; Bhatia, S.N. Geometric and


material determinants of patterning efficiency by
dielectrophoresis. Biophys. J. 2004, 87 (4), 2131–2147.

25. Albrecht, D.R.; Tsang, V.L.; Sah, R.L.; Bhatia, S.N.


Photoand electropatterning of hydrogel-encapsulated living
cell arrays. Lab Chip 2005, 5 (1), 111–118. 26. Alp, B.;
Stephens, G.M.; Markx, G.H. Formation of artificial,
structured microbial consortia (ASMC) by dielectrophoresis.
Enzyme Microbiol. Technol. 2002, 31 (1–2), 35–43. 27. Alp,
B.; Andrews, J.S.; Mason, V.P.; Wolowacz, R.; Markx, G.H.
Building structured biomaterials using AC electrokinetics.
IEEE Eng. Med. Biol. Mag. 2003, 22 (6), 91–97. 28. Markx,
G.H.; Andrews, J.S.; Mason, V.P. Towards microbial tissue
engineering? Trends Biotechnol. 2004, 22 (8), 417–422. 29.
Mason, V.P.; Markx, G.H.; Thompson, I.P.; Andrews, J.S.;
Manefield, M. Colonial architecture in mixed species
assemblages affects AHL mediated gene expression. FEMS
Microbiol. Lett. 2005, 244 (1), 121–127. 30.
Verduzco-Luque, C.E.; Alp, B.; Stephens, G.M.; Markx, G.H.
Construction of biofilms with defined internal architecture
using dielectrophoresis and flocculation. Biotechnol.
Bioeng. 2003, 83 (1), 39–44. 31. Matsue, T.; Matsumoto, N.;
Uchida, I. Rapid micropatterning of living cells by
repulsive dielectrophoretic force. Electrochim. Acta 1997,
42 (20–22), 3251–3256. 32. Neil, G.A.; Zimmermann, U.
Electrofusion. Methods Enzymol. 1993, 221, 171–196. 33.
Wang, X.; Yang, J.; Gascoyne, P.R.C. Role of peroxide in AC
electrical field exposure effects on Friend murine
erythroleukemia cells during dielectrophoretic
manipulations. Biochim. Biophys. Acta 1999, 1426 (1),
53–68. 34. Wang, X.B.; Huang, Y.; Gascoyne, P.R.C.; Becker,
F.F.; Ho¨lzel, R.; Pethig, R. Changes in Friend murine
erythroleukaemia cell membranes during induced
differentiation determined by electrorotation. Biochim.
Biophys. Acta 1994, 1193 (2), 330–344. 35. Pethig, R.;
Talary, M.S.; Lees, R.S. Enhanced travelling-wave
dielectrophoresis with signal superposition. IEEE Eng. Med.
Biol. Mag. 2003, 22 (6), 43–50. 36. Huang, Y.; Tame, J.A.;
Pethig, R. Electrokinetic behaviour of colloidal particles
in travelling electric fields: studies using yeast cells. J.
Phys. D: Appl. Phys. 1993, 26 (9), 1528–1535. 37.
Schmeichel, K.L.; Bissell, M.J. Modeling tissue-specific
signaling and organ function in three dimensions. J. Cell
Sci. 2003, 116 (12), 2377–2388. 38. Andrews, J.A.; Mason,
V.P.; Thompson, I.P.; Stephens, G.M.; Markx, G.H.
Construction of artificially structured microbial consortia
(ASMC) using dielectrophoresis: examining bacterial
interactions via metabolic intermediates within
environmental biofilms J. Microbiol. Methods 2005, in press.
FURTHER READING Chethana, S.; Patil, G.; Raghavarao,
K.S.M.S. Magnetic and electrophoretic cell separation.
10.1081/E-EBBE120020410. Madou, M. Microelectromechanical
system (MEMS) manufacturing. 10.1081/E-EBBE-120013935. Tan,
W.; Desai, T. Tissue Engineering, Microscale. 10.1081/
E-EBBE-120012936.
Tissue Engineering of Bladder

28. Machlouf, M.; Atala, A. Emerging concepts for tissue


and organ transplantation. Graft 1998, 1, 31.

29. Atala, A. Tissue Engineering Techniques for Closure of


Bladder Exstrophy: An Experimental Animal Model. In The
Exstrophy-Epispadias Complex; Gearhart, J., Ed.; Plenum
Press: New York, 1998; 63–64.

30. Fauza, D.O.; Fishman, S.; Mehegan, K.; Atala, A.


Videofetoscopically assisted fetal tissue engineering:
Bladder augmentation. J. Pediatr. Surg. 1998, 33, 7.

31. Yoo, J.J.; Lee, I.; Atala, A. Cartilage rods as a


potential material for penile reconstruction. J. Urol.
1998, 160, 1164.

32. Fauza, D.O.; Fishman, S.; Mehegan, K.; Atala, A.


Videofetoscopically assisted fetal tissue engineering: Skin
replacement. J. Pediatr. Surg. 1998, 33, 377.

33. Amiel, G.E.; Atala, A. Current and future modalities


for functional renal replacement. Urol. Clin. North Am.
1999, 26 (1), 235–246.

34. Yoo, J.J.; Park, H.J.; Lee, I.; Atala, A. Autologous


engineered cartilage rods for penile reconstruction. J.
Urol. 1999, 160, 1164.

35. Park, H.J.; Kershen, R.; Yoo, J.J.; Atala, A.


Reconstitution of human corporal smooth muscle and
endothelial cells in vivo. J. Urol. 1999, 162, 1106.

36. Oberpenning, F.O.; Meng, J.; Yoo, J.; Atala, A. De novo


reconstitution of a functional urinary bladder by tissue
engineering. Nat. Biotechnol. 1999, 17, 2.

37. Atala, A. Future perspectives in reconstructive surgery


using tissue engineering. Urol. Clin. North Am. 1999, 26
(1), 157–165. 38. Tobin, M.S.; Freeman, M.R.; Atala, A.
Maturational response of normal human urothelial cells in
culture is dependent on extracellular matrix and serum
additives. Surg. Forum 1994, 45, 786. 39. Freeman, M.R.;
Yoo, J.J.; Raab, G.; Soker, S.; Adam, R.M.; Schneck, F.X.;
Renshaw, A.A.; Klagsbrun, M.; Atala, A. Heparin-binding
EGF-like growth factor is an autocrine factor for human
urothelial cells and is synthesized by epithelial and
smooth muscle cells in the human bladder. J. Clin. Invest.
1997, 99 (5), 1028. 40. Nguyen, H.T.; Park, J.M.; Peters,
C.A.; Adam, R.A.; Orsola, A.; Atala, A.; Freeman, M.R.
Cell-specific activation of the HB-EGF and ErbB1 genes by
stretch in primary human bladder cells. In Vitro Cell. Dev.
Biol. 1999, 35, 371–375. 41. Folkman, J.; Hochberg, M.M.
Self regulation of growth in three dimensions. J. Exp. Med.
1973, 138, 745. 42. de Boer, W.I.; Schuller, A.G.; Vermay,
M.; van der Kwast, T.H. Expression of growth factors and
receptors during specific phases in regenerating urothelium
after acute injury in vivo. Am. J. Path. 1994, 145 , 1199.
43. Baker, R.; Kelly, T.; Tehan, T.; Putman, C.; Beaugard,
E. Subtotal cystectomy and total bladder regeneration in
treatment of bladder cancer. J. Am. Med. Ass. 1955, 168,
1178. 44. Gorham, S.D.; French, D.A.; Shivas, A.A.; Scott,
R. Some observations on the regeneration of smooth muscle
in the repaired urinary bladder of the rabbit. Eur. Urol.
1989, 16, 440.
Tissue Engineering of Blood Vessel

1. American Heart Association Heart and Stroke Statistical


Update; 2001. Dallas.

2. von Segesser, L.K. Arterial Grafting for Myocardial


Revascularization: Indications, Surgical Techniques and
Results; Springer Verlag: New York, 1991.

3. Pelletier, L.C. The Saphenous Vein Graft: What Have We


Learned from the Past 25 Years. In Conduits for Myocardial
Revascularization; Carrier, M., Pelletier, L.C., Eds.; RG
Landes Co.: Austin, 1993; 3–34.

4. Yamada, T.; Shiraishi, R.; Taki, K.; Nakano, S.;


Tokunaga, O.; Itoh, T. Immunohistochemical and
ultrastructural examination of smooth muscle cells in
aortocoronary saphenous vein grafts. Angiology 1997, 48
(5), 381–390. 5. Mavromatis, K.; Fukai, T.; Tate, M.;
Chesler, N.; Ku, D.N.; Galis, Z.S. Early effects of
arterial hemodynamic conditions on human saphenous veins
perfused ex vivo. Arterioscler. Thromb. Vasc. Biol. 2000,
20 (8), 1889– 1895. 6. Predel, H.G.; Yang, Z.;
von_Segesser, L.; Turina, M.; Buhler, F.R.; Luscher, T.F.
Implications of pulsatile stretch on growth of saphenous
vein and mammary artery smooth muscle. Lancet 1992, 340
(8824), 878– 879. 7. Williams, S.K.; Rose, D.G.; Jarrell,
B.E. Microvascular endothelial cell sodding of ePTFE
vascular grafts: Improved patency and stability of the
cellular lining. J. Biomed. Mater. Res. 1994, 28 (2),
203–212. 8. Gelbfish, J.; Jacobowitz, I.J.; Rose, D.M.;
Connolly, M.W.; Acinapura, A.J.; Zisbrod, Z.; Lim, K.H.;
Cappabianca, P.; Cunningham, J.N. Cryopreserved homologous
saphenous vein: Early and late patency in coronary artery
bypass surgical procedures. Ann. Thorac. Surg. 1986, 42
(1), 70–73. 9. Greisler, H.P. New Biologic and Synthetic
Vascular Prosthesis; RG Landes: Austin, TX, 1991. 10.
Dardik, H.; Ibrahim, I.M.; Dardik, I. Modified and unmodified
umbilical vein allografts and xenografts as arterial
substitutes: Morphologic assessment. Surg. Forum 1975, 26,
286–287. 11. Conklin, B.S.; Richter, E.R.; Kreutziger,
K.L.; Zhong, D.S.; Chen, C. Development and evaluation of a
novel decellularized vascular xenograft. Med. Eng. Phys.
2002, 24 (3), 173–183. 12. Ratcliffe, A. Tissue engineering
of vascular grafts. Matrix Biol.: J. Int. Soc. Matrix
Biology 2000, 19 (4), 353–357. 13. Nerem, R.M.; Seliktar,
D. Vascular tissue engineering. Annu. Rev. Bioeng. 2001, 3,
225–243. 14. Salacinski, H.J.; Goldner, S.; Giudiceandrea,
A.; Hamilton, G.; Seifalian, A.M.; Edwards, A.; Carson,
R.J. The mechanical behavior of vascular grafts: A review.
J. Biomater. Appl. 2001, 15 (3), 241–278. 15. Weinberg,
C.B.; Bell, E. A blood vessel model constructed from
collagen and cultured vascular cells. Science 1986, 231
(4736), 397–400. 16. L’Heureux, N.; Paquet, S.; Labbe, R.;
Germain, L.; Auger, F.A. A completely biological
tissue-engineered human blood vessel [see comments]. FASEB
J. 1998, 12 (1), 47–56. 17. Niklason, L.E.; Gao, J.;
Abbott, W.M.; Hirschi, K.K.; Houser, S.; Marini, R.;
Langer, R. Functional arteries grown in vitro [see
comments]. Science 1999, 284 (5413), 489–493. 18. Niklason,
L. Progress in the Engineering of Small Caliber Arterial
Prostheses. In Engineering Tissue Growth International
Conference & Exposition; 2002Pittsburgh 19. Campbell, J.H.;
Efendy, J.L.; Campbell, G.R. Novel vascular graft grown
within recipient’s own peritoneal cavity. Circ. Res.
(Online) 1999, 85 (12), 1173–1178. 20. Cohnheim, J. . Arch.
Pathol. Anat. Physiol. Klin. Med. 1867, 40, 1. 21. Fukuda,
K. Development of regenerative cardiomyocytes from
mesenchymal stem cells for cardiovascular tissue
engineering. Artif. Organs 2001, 25 (3), 187–193. 22.
Arakawa, E.; Hasegawa, K.; Yanai, N.; Obinata, M.; Matsuda,
Y. A mouse bone marrow stromal cell line, TBR-B, shows
inducible expression of smooth musclespecific genes. FEBS
Lett. 2000, 481 (2), 193–196. 23. Noishiki, Y.; Tomizawa,
Y.; Yamane, Y.; Matsumoto, A. Autocrine angiogenic vascular
prosthesis with bone marrow transplantation [see comments].
Nat. Med. 1996, 2 (1), 90–93. 24. Noishiki, Y.; Yamane, Y.;
Tomizawa, Y.; Matsumoto, A. Transplantation of autologous
tissue fragments into an e-PTFE graft with long fibrils.
Artif. Organs 1995, 19 (1), 17–26. 25. Hirschi, K.;
Goodell, M. Common origins of blood and blood vessels in
adults? Differ. Res. Biol. Divers. 2001, 68 (4–5), 186–192.
26. Han, C.I.; Campbell, G.R.; Campbell, J.H. Circulating
bone marrow cells can contribute to neointimal formation.
J. Vasc. Res. 2001, 38 (2), 113–119. 27. Nerem, R.M.;
Girard, P.R. Hemodynamic influences on vascular endothelial
biology. Toxicol. Pathol. 1990, 18 (4 Pt 1), 572–582. 28.
Hamilton, D.W.; Maul, T.M.; Vorp, D.A. Characterization of
the response of bone marrow derived progenitor cells to
cyclic strain: Implications for vascular tissue engineering
applications. Tissue Eng. in press. 29. Sumpio, B.E.
Hemodynamic Forces and Vascular Cell Biology; R.G. Landes
Co.: Austin, TX, 1993. 30. Kanda, K.; Matsuda, T.; Oka, T.
Mechanical stress induced cellular orientation and
phenotypic modulation of 3-D cultured smooth muscle cells.
ASAIO J. 1993, 39 (3), M686–M690. 31. Girton, T.S.; Oegema,
T.R.; Tranquillo, R.T. Exploiting glycation to stiffen and
strengthen tissue equivalents for tissue engineering. J.
Biomed. Mater. Res. 1999, 46 (1), 87–92. 32. Orban, J.M.;
Wilson, L.B.; Kofroth, J.A.; El-Kurdi, M.S.; Maul, T.M.;
Vorp, D.A. Cross linking of collagen gels by
transglutaminase. J. Biomed. Mater. Res. revision under
review. 33. Hirai, J.; Kanda, K.; Oka, T.; Matsuda, T.
Highly oriented, tubular hybrid vascular tissue for a low
pressure circulatory system. ASAIO J. 1994, 40 (3), M383–
M388. 34. Tranquillo, R.T.; Girton, T.S.; Bromberek, B.A.;
Triebes, T.G.; Mooradian, D.L. Magnetically orientated
tissue-equivalent tubes: Application to a circumferentially
orientated media-equivalent. Biomaterials 1996, 17 (3),
349–357. 35. Huynh, T.; Abraham, G.; Murray, J.; Brockbank,
K.; Hagen, P.O.; Sullivan, S. Remodeling of an acellular
collagen graft into a physiologically responsive neovessel.
Nat. Biotechnol. 1999, 17 (11), 1083–1086. 36. Badylak, S.
The extracellular matrix as a scaffold for tissue
reconstruction. Semin. Cell Dev. Biol. 2002, 13 (5), 377.
37. Grassl, E.D.; Oegema, T.R.; Tranquillo, R.T. Fibrin as
an alternative biopolymer to type-I collagen for the
fabrication of a media equivalent. J. Biomed. Mater. Res.
2002, 60 (4), 607–612. 38. Nerem, R.M. Tissue engineering
in the USA. Med. Biol. Eng. Comput. 1992, 30 (4), CE8–CE12.
39. Greisler, H.P.; Gosselin, C.; Ren, D.; Kang, S.S.; Kim,
D.U. Biointeractive polymers and tissue engineered blood
vessels. Biomaterials 1996, 17 (3), 329–336. 40. Vorp,
D.A.; Severyn, D.A.; Steed, D.L.; Webster, M.W. A device
for the application of cyclic twist and extension on
perfused vascular segments. Am. J. Physiol. 1996, 270 (2 Pt
2), H787–H795. 41. Shyy, J.Y. Mechanotransduction in
endothelial responses to shear stress: Review of work in
Dr. Chien’s laboratory. Biorheology 2001, 38 (2–3),
109–117. 42. Williams, B. Mechanical influences on vascular
smooth muscle cell function. J. Hypertens. 1998, 16 (12 Pt
2), 1921–1929. 43. Seliktar, D.; Black, R.A.; Vito, R.P.;
Nerem, R.M. Dynamic mechanical conditioning of collagen-gel
blood vessel constructs induces remodeling in vitro. Ann.
Biomed. Eng. 2000, 28 (4), 351–362. 44. Vorp, D.A.;
Severyn, D.A.; Mears, J. An experimental system to expose
perfused vascular segments to cyclic bending ex vivo. Adv.
Bioeng. 1997, 36, 115–116. 45. Vorp, D.A.; Peters, D.G.;
Webster, M.W. Gene expression is altered in perfused
arterial segments exposed to cyclic flexure ex vivo. Ann.
Biomed. Eng. 1999, 27 (3), 366–371. 46. Tai, N.R.;
Salacinski, H.J.; Edwards, A.; Hamilton, G.; Seifalian,
A.M. Compliance properties of conduits used in vascular
reconstruction. Br. J. Surg. 2000, 87 (11), 1516–1524. T
Tissue Engineering of Bone

1. Vaccaro, A.R. The role of the osteoconductive scaffold


in synthetic bone graft. Orthopedics 2002, 25 (5),
s571–s578.

2. Tomin, E.; Lane, J.; Nakamichi, K.; Hsu, J.; Schneider,


K.; Weiland, A. Performance of molded vascularized bone
grafts induced by BMP-2 and marrow in a rat segmental
defect model. Trans. Orthop. Res. Soc. 1999, 24, 621.

3. Beresford, J.N. Osteogenic stem cells and the stromal


system of bone and marrow. Clin. Orthop. 1989, 240,
270–280.

4. Connolly, J.F. Injectable bone marrow preparations to


stimulate osteogenic repair. Clin. Orthop. 1995, 313, 8–18.

5. Paley, D.; Young, M.C.; Wiley, A.M.; Fornasier, V.L.;


Jackson, R.W. Percutaneous bone marrow grafting of
fractures and bony defects. Clin. Orthop. 1986, 208,
300–312.

6. Liang, C.T.; Barnes, J.; Seedor, J.G.; Quartuccio, H.A.;


Bolander, M.; Jeffrey, J.J.; Rodan, G.A. Impaired bone
activity in aged rats: Alterations at the cellular and
molecular levels. Bone 1992, 13 (6), 435–441.

7. Quarto, R.; Thomas, D.; Liang, C.T. Bone progenitor cell


deficits and the age-associated decline in bone repair
capacity. Calcif. Tissue Int. 1995, 56 (2), 123–129.

8. Kuehnle, I.; Goodell, M.A. The therapeutic potential of


stem cells from adults. BMJ 2002, 325 (7360), 372–376.

9. Bianco, P.; Riminucci, M.; Kuznetsov, S.; Robey, P.G.


Multipotential cells in the bone marrow stroma: Regulation
in the context of organ physiology. Crit. Rev. Eukaryot.
Gene Expr. 1999, 9 (2), 159–173.

10. Vats, A.; Tolley, N.S.; Polak, J.M.; Buttery, L.D. Stem
cells: Sources and applications. Clin. Otolaryngol. 2002,
27 (4), 227–232.

11. Haynesworth, S.E.; Carrino, D.A.; Caplan, A.I.


Characterization of the core protein of the large
chondroitin sulfate proteoglycan synthesized by
chondrocytes in chick limb bud cell cultures. J. Biol.
Chem. 1987, 262 (22), 10574–10581.
12. Jaiswal, N.; Haynesworth, S.E.; Caplan, A.I.; Bruder,
S.P. Osteogenic differentiation of purified,
culture-expanded human mesenchymal stem cells in vitro. J.
Cell. Biochem. 1997, 64 (2), 295–312.

13. Rose, F.R.; Oreffo, R.O. Bone Tissue Engineering: Hope


vs. hype. Biochem. Biophys. Res. Commun. 2002, 292 (1),
1–7.

14. Keller, G.M. In vitro differentiation of embryonic stem


cells. Curr. Opin. Cell. Biol. 1995, 7 (6), 862–869.

15. McLaren, A. Ethical and social considerations of stem


cell research. Nature 2001, 414 (6859), 129–131.

16. Oreffo, R.O.; Triffitt, J.T. Future potentials for using


osteogenic stem cells and biomaterials in orthopedics. Bone
1999, 25 (2), 5S–9S. 17. Thomson, J.A.; Itskovitz-Eldor,
J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.;
Marshall, V.S.; Jones, J.M. Embryonic stem cell lines
derived from human blastocysts. Science 1998, 282 (5391),
1145–1147. 18. Lieberman, J.R.; Daluiski, A.; Stevenson,
S.; Wu, L.; McAllister, P.; Lee, Y.P.; Kabo, J.M.;
Finerman, G.A.; Berk, A.J.; Witte, O.N. The effect of
regional Gene Therapy with bone morphogenetic
protein-2-producing bone-marrow cells on the repair of
segmental femoral defects in rats. J. Bone Jt. Surg., Am.
1999, 81 (7), 905–917. 19. Scaduto, A.A.; Lieberman, J.R.
Gene Therapy for osteoinduction. Orthop. Clin. North Am.
1999, 30 (4), 625–633. 20. Breitbart, A.S.; Grande, D.A.;
Mason, J.M.; Barcia, M.; James, T.; Grant, R.T.
Gene-enhanced tissue engineering: Applications for bone
healing using cultured periosteal cells transduced
retrovirally with the BMP-7 gene. Ann. Plast. Surg. 1999,
42 (5), 488–495. 21. Musgrave, D.S.; Bosch, P.; Ghivizzani,
S.; Robbins, P.D.; Evans, C.H.; Huard, J.
Adenovirus-mediated direct Gene Therapy with bone
morphogenetic protein2 produces bone. Bone 1999, 24 (6),
541–547. 22. Asahara, T.; Kalka, C.; Isner, J.M. Stem cell
therapy and gene transfer for regeneration. Gene Ther.
2000, 7 (6), 451–457. 23. Urist, M.R.; Silverman, B.F.;
Buring, K.; Dubuc, F.L.; Rosenberg, J.M. The bone induction
principle. Clin. Orthop. 1967, 53, 243–283. 24. Gundle, R.;
Joyner, C.J.; Triffitt, J.T. Human bone tissue formation in
diffusion chamber culture in vivo by bone-derived cells and
marrow stromal fibroblastic cells. Bone 1995, 16 (6),
597–601. 25. Vacanti, C.A.; Bonassar, L.J. An overview of
Tissue Engineered bone. Clin. Orthop. 1999, 367, S375–S381.
26. Bruder, S.P.; Fink, D.J.; Caplan, A.I. Mesenchymal stem
cells in bone development, bone repair, and skeletal
regeneration therapy. J. Cell. Biochem. 1994, 56 (3),
283–294. 27. Petite, H.; Viateau, V.; Bensaid, W.; Meunier,
A.; de Pollak, C.; Bourguignon, M.; Oudina, K.; Sedel, L.;
Guillemin, G. Tissue-engineered bone regeneration. Nat.
Biotechnol. 2000, 18 (9), 959–963. 28. Burg, K.J.; Porter,
S.; Kellam, J.F. Biomaterial developments for bone Tissue
Engineering. Biomaterials 2000, 21 (23), 2347–2359. 29.
Pineda, L.M.; Busing, M.; Meinig, R.P.; Gogolewski, S. Bone
regeneration with resorbable polymeric membranes. III.
Effect of poly(L-lactide) membrane pore size on the bone
healing process in large defects. J. Biomed. Mater. Res.
1996, 31 (3), 385–394. 30. Ripamonti, U.; Crooks, J.;
Rueger, D.C. Induction of bone formation by recombinant
human osteogenic protein-1 and sintered porous
hydroxyapatite in adult primates. Plast. Reconstr. Surg.
2001, 107 (4), 977–988. 31. Borden, M.; Attawia, M.; Khan,
Y.; Laurencin, C.T. Tissue Engineered microsphere-based
matrices for bone repair: Design and evaluation.
Biomaterials 2002, 23 (2), 551–559. 32. Andriano, K.P.;
Tabata, Y.; Ikada, Y.; Heller, J. In vitro and in vivo
comparison of bulk and surface hydrolysis in absorbable
polymer scaffolds for tissue engineering. J. Biomed. Mater.
Res. 1999, 48 (5), 602–612. 33. Hollinger, J.O.;
Battistone, G.C. Biodegradable bone repair materials.
Synthetic polymers and ceramics. Clin. Orthop. 1986, 207,
290–305. 34. Dalby, M.J.; Riehle, M.O.; Johnstone, H.;
Affrossman, S.; Curtis, A.S. In vitro reaction of
endothelial cells to polymer demixed nanotopography.
Biomaterials 2002, 23 (14), 2945–2954. 35. Yang, Y.;
Magnay, J.L.; Cooling, L.; El, H.A. Development of a
‘mechano-active’ scaffold for tissue engineering.
Biomaterials 2002, 23 (10), 2119–2126. 36. Badylak, S.F.;
Kropp, B.; McPherson, T.; Liang, H.; Snyder, P.W. Small
intestional submucosa: A rapidly resorbed bioscaffold for
augmentation cystoplasty in a dog model. Tissue Eng. 1998,
4 (4), 379–387. 37. Quirk, R.A.; Chan, W.C.; Davies, M.C.;
Tendler, S.J.; Shakesheff, K.M. Poly(L-lysine)-GRGDS as a
biomimetic surface modifier for poly(lactic acid).
Biomaterials 2001, 22 (8), 865–872. 38. Hrkach, J.S.; Ou,
J.; Lotan, N.; Langer, R. Synthesis of poly(L-lactic
acid-co-L-lysine) graft copolymers. Macromolecules 1995, 28
(13), 4736–4739. 39. Cooper, A.I. Polymer synthesis and
processing using supercritical carbon dioxide. J. Mater.
Chem. 2000, 10 (2), 207–234. 40. Green, D.; Walsh, D.;
Mann, S.; Oreffo, R.O. The potential of biomimesis in bone
Tissue Engineering: Lessons from the design and synthesis
of invertebrate skeletons. Bone 2002, 30 (6), 810–815. 41.
Hauschka, P.V.; Chen, T.L.; Mavrakos, A.E. Polypeptide
growth factors in bone matrix. Ciba Found. Symp. 1988, 136,
207–225. 42. Reddi, A.H. Bone morphogenetic proteins: From
basic science to clinical applications. J. Bone Jt. Surg.,
Am. 2001, 83A (Suppl. 1 (Pt. 1)), S1–S6. 43. Centrella, M.;
Horowitz, M.C.; Wozney, J.M.; McCarthy, T.L. Transforming
growth factor-beta gene family members and bone. Endocr.
Rev. 1994, 15 (1), 27–39. 44. Wozney, J.M. Bone
morphogenetic proteins. Prog. Growth Factor Res. 1989, 1
(4), 267–280. 45. Johnson, E.E.; Urist, M.R.; Finerman,
G.A. Bone morphogenetic protein augmentation grafting of
resistant femoral nonunions. A preliminary report. Clin.
Orthop. 1988, 230, 257–265. 46. Bostrom, M.P.; Lane, J.M.;
Berberian, W.S.; Missri, A.A.; Tomin, E.; Weiland, A.;
Doty, S.B.; Glaser, D.; Rosen, V.M. Immunolocalization and
expression of bone morphogenetic proteins 2 and 4 in
fracture healing. J. Orthop. Res. 1995, 13 (3), 357–367.
47. Einhorn, T.A.; Trippel, S.B. Growth factor treatment of
fractures. Instr. Course Lect. 1997, 46, 483–486. 48. Wang,
E.A.; Israel, D.I.; Kelly, S.; Luxenberg, D.P. Bone
morphogenetic protein-2 causes commitment and
differentiation in C3H10T1/2 and 3T3 cells. Growth Factors
1993, 9 (1), 57–71. 49. Wozney, J.M. Overview of bone
morphogenetic proteins. Spine 2002, 27 (16 Suppl. 1),
S2–S8. 50. Ripamonti, U.; Ramoshebi, L.N.; Matsaba, T.;
Tasker, J.; Crooks, J.; Teare, J. Bone induction by
BMPs/OPs and related family members in primates. J. Bone
Jt. Surg., Am. 2001, 83-A (Suppl. 1 (Pt. 2)), S116–S127.
51. Johnson, E.E.; Urist, M.R. One-stage lengthening of
femoral nonunion augmented with human bone morphogenetic
protein. Clin. Orthop. 1998, 347, 105–116. 52. Aspenberg,
P.; Turek, T. BMP-2 for intramuscular bone induction:
Effect in squirrel monkeys is dependent on implantation
site. Acta Orthop. Scand. 1996, 67 (1), 3–6. 53. Einhorn,
T.A. Problems with delayed and impaired fracture healing
remain a challenge to the orthopedic trauma surgeon.
Orthop. Traumatol. 1997, 11 (4), 243. 54. Einhorn, T.A.;
Lee, C.A. Bone regeneration: New findings and potential
clinical applications. J. Am. Acad. Orthop. Surg. 2001, 9
(3), 157–165. 55. Kaplan, F.S.; Tabas, J.A.; Zasloff, M.A.
Fibrodysplasia ossificans progressiva: A clue from the fly?
Calcif. Tissue Int. 1990, 47 (2), 117–125. 56. Tsuruga, E.;
Takita, H.; Itoh, H.; Wakisaka, Y.; Kuboki, Y. Pore size of
porous hydroxyapatite as the cell-substratum controls
BMP-induced osteogenesis. J. Biochem. (Tokyo) 1997, 121
(2), 317–324. T
Tissue Engineering of Bone Marrow

1. Kaplan, A.; Bruder, S.P. Mesenchymal stem cells:


Building blocks for molecular medicine in the 21st century.
Trends Mol. Med. 2001, 7 (6), 259–264.

2. Madlambayan, G.J.; Rogers, I.; Casper, R.F.; Zandstra,


P.W. Controlling culture dynamics for the expansion of
hematopoietic stem cells. J. Hematother. Stem Cell Res.
2001, 10, 481–492. 3. Abboud, C.N.; Lichtman, M.A.
Structure of the Marrow and the Hemopoietic
Microenvironment. In Hematology; Beutler, E., Ed.;
McGraw-Hill: New York, 2001; 29–58. 4. McNiece, I.;
Briddell, R. Ex vivo expansion of hematopoietic progenitor
cells and mature cells. Exp. Hematol. 2001, 29, 3–11. 5.
Charbord, P. Microenvironmental Cell Populations Essential
for the Support of Hematopoietic Stem Cells.
InHematopoiesis: A Developmental Approach; Zon, L.I., Ed.;
Oxford University Press: New York, 2001; 691–701. 6.
Trentin, J.J. Influence of Hematopoietic Organ Stroma
(Hematopoietic Inductive Microenvironments) on Stem Cell
Differentiation. In Regulation of Hemopoiesis; Gordon,
A.S., Ed.; Appleton-Centurycrofts: New York, 1970; 161–186.
7. Naito, K.; Tamahashi, N.; Chiba, T.; Kaneda, K.; Okuda,
M.; Endo, K.; Yoshinaga, K.; Takahashi, T. The
microvasculature of the human bone marrow correlated with
the distribution of hematopoietic cells. A
computer-assisted three-dimensional reconstruction study.
Tohoku J. Exp. Med. 1992, 166, 439–450. 8. Koller, M.R.;
Palsson, B.O. Tissue engineering: Reconstitution of human
hematopoiesis ex vivo. Biotechnol. Bioeng. 1993, 42,
909–930. 9. Dorshkind, K. Regulation of hemopoiesis by bone
marrow stromal cells and their products. Annu. Rev.
Immunol. 1990, 8, 111–137. 10. Bianco, P.; Riminucci, M.;
Gronthos, S.; Robey, P.G. Bone marrow stromal cells:
Nature, biology, and potential applications. Stem Cells
2001, 19, 180–192. 11. Campbell, A.; Wicha, M.S.
Extracellular matrix and the hematopoietic
microenvironment. J. Lab. Clin. Med. 1988, 112 (2),
140–146. 12. Dao, M.A.; Hashimo, K.; Kato, I.; Nolta, J.A.
Adhesion to fibronectin maintains regenarative capacity
during ex vivo culture and transduction of human
hematopoietic stem and progenitor cells. Blood 1998, 92,
4612–4621. 13. Campbell, A.; Long, M.W.; Wicha, M.S.
Haemonectin, a bone marrow adhesion protein specific for
cells of granulocyte lineage. Nature 1987, 329, 744–746.
14. Weistein, R.; Riordan, M.A.; Wenc, K.; Kreczko, S.;
Zhou, M.; Dainiak, N. Dual role of fibronectin in
hematopoietic differentiation. Blood 1989, 73, 111–116. 15.
Liesveld, J.; Winslow, J.M.; Kempski, M.C.; Ryan, D.H.;
Brennan, J.K.; Abboud, C.N. Adhesive interaction of normal
and leukemic human CD34 + myeloid progenitors-role of
marrow stromal, fibroblast, and cytomatrix components. Exp.
Hematol. 1991, 9, 63–70. 16. Verfaillie, C. Soluble
factor(s) produced by human bone marrow stroma increase
cytokine-induced proliferation and maturation of primitive
hematopoietic progenitors while preventing their terminal
differentiation. Blood 1993, 82, 2045–2053. 17. Weissman,
I. Developmental switches in the immune system. Cell 1994,
76, 207–218. 18. Aiuti, A.; Friedrich, C.; Sieff, C.;
Gutierrez-Ramos, J. Identification of distinct elements of
the stromal microenvironment that control human
hematopoietic stem/progenitor cell growth and
differentiation. Exp. Hematol. 1998, 26, 143–157. 19.
Quesenberry, P.; Crittenden, R.; Lowry, P.; Kittler, E.;
Rao, S.; Peters, S.; Ramshaw, H.; Stewart, F. In vitro and
in vivo studies of stromal niches. Blood Cells 1994, 20,
97–106. 20. Mantalaris, A.; Keng, P.; Bourne, P.; Chang,
A.Y. C.; Wu, J.H.D. Engineering a human bone marrow model:
A case study on ex vivo erythropoiesis. Biotechnol. Prog.
1998, 14, 126–133. 21. Noll, T.; Jelinek, N.; Schmidt, S.;
Biselli, M.; Wandrey, C. Cultivation of hematopoietic stem
and progenitor cells: Biochemical engineering aspects. Adv.
Biochem. Eng. 2002, 74, 111–128. 22. Nielsen, L.K.
Bioreactors for hematopoietic cell culture. Annu. Rev.
Biomed. Eng. 1999, 1, 129–152. 23. Douay, L. Experimental
culture conditions are critical for ex vivo expansion of
hematopoietic cells. J. Hematother. Stem Cell Res. 2001,
10, 341–346. 24. Alley, C.; MacDermott, R.P.; Stewart, C.C.
The effect of serum on human marrow mononuclear cell
proliferation and maturation. J. Reticuloendothel. Soc.
1983, 34, 271–278. 25. Bohmer, R. Interaction of serum and
colony-stimulating factor for survival of a
factor-dependent hemopoietic progenitor cell line. J. Cell.
Physiol. 1989, 139, 531–537. 26. Collins, P.; Miller, W.M.;
Papoutsakis, E.T. Ex vivo culture systems for hematopoietic
cells. Curr. Opin. Biotechnol. 1996, 7 (2), 223–230. 27.
Guba, S.; Gottschalk, L.R.; Jing, Y.H.; Mulligan, T.;
Emerson, S.G. Bone marrow stromal fibroblasts secrete
interleukin-6 and granulocytemacrophage colony-stimulating
factor in the absence of inflammatory stimulation:
Demonstration by serum-free bioassay, enzyme-linked
immunosorbent assay, and reverse transcriptase polymerase
chain reaction. Blood 1992, 80 (5), 1190–1198. 28. Tsukada,
J.; Misago, M.; Kikuchi, M.; Sato, T.; Ogawa, R.; Ota, T.;
Oda, S.; Morimoto, I.; Chiba, S.; Eto, S. Interaction
between recombinant human erythropoietin and serum
factor(s) on murine megakaryocyte colony formation. Blood
1992, 80 (1), 37–45. 29. Lill, M.C.; Lynch, M.; Fraser,
J.K.; Chung, G.Y.; Schiller, G.; Glaspy, J.A.; Souza, L.;
Baldwin, G.C.; Gasson, J.C. Production of functional
myeloid cells from CD34-selected hematopoietic progenitor
cells using a clinically relevant ex vivo expansion system.
Stem Cells 1994, 12, 626–637. 30. Fortunel, N.; Hatzfeld,
A.; Hatzfeld, J.A. Transforming growth factor-B:
Pleiotropic role in the regulation of hematopoiesis. Blood
2000, 96 (6), 2022– 2036. 31. Krystal, G.; Lanm, V.;
Dragowska, W.; Takahashi, C.; Appel, J. Transforming growth
factor beta 1 is an inducer of erythroid differentiation.
J. Exp. Med. 1994, 180, 851–860. 32. Dybedal, I.; Jacobsen,
S.E. Transforming growth factor beta (TGF-beta), a potent
inhibitor of erythropoiesis: Neutralizing TGF-beta
antibodies show erythropoietin as a potent stimulator of
murine burstforming unit erythroid colony formation in the
absence of a burst-promoting activity. Blood 1995 , 86,
949–957. 33. Sandstrom, C.; Miller, W.M.; Papoutsakis, E.T.
Review: Serum-free media for cultures of primitive and
mature hematopoietic cells. Biotechnol. Bioeng. 1994, 43,
706–733. 34. Rosenzwajg, M.; Canque, B.; Gluckman, J.C.
Human dendritic cell differentiation pathway from CD34 +
hematopoietic precursor cells. Blood 1996, 87, 535–544. 35.
Brugger, W.; Mocklin, W.; Heimfeld, S.; Berenson, R.J.;
Mertelsmann, R.; Kanz, L. Ex vivo expansion of enriched
peripheral blood CD34 + progenitor cells by stem cell
factor, interleukin-1 (IL-1), IL-6, IL-3, interferon g and
erythropoietin. Blood 1993, 81, 2579–2584. 36. Brown, R.;
Xu, F.S.; Dusing, S.K.; Li, Q.; Fischer, R.; Patchen, M.
Serum-free conditions for cells capable of producing
long-term survival in lethally irradiated mice. Stem Cells
1997, 15, 237–245. 37. Lansdorp, P.M.; Lansdorp, W.;
Dragowska, W. Longterm erythropoiesis from constant numbers
of CD34 + cells in serum-free cultures initiated with
highly purified progenitor cells from human bone marrow. J.
Exp. Med. 1992, 175, 1501–1509. 38. Mobest, D.;
Mertelsmann, R.; Henschler, R. Serumfree ex vivo expansion
of CD34 + hematopoietic progenitor cells. Biotechnol.
Bioeng. 1998, 60 (3), 341–347. 39. Mobest, D.; Goan, S.;
Junghahn, I.; Winkler, J.; Fichtner, I.; Becker, M.; De
Lima-Hahn, E.; Mertelsmann, R.; Henschler, R. Differential
kinetics of primitive hematopoietic cells assayed in vitro
and in vivo during serum-free suspension culture of CD34 +
blood progenitor cells. Stem Cells 1999, 17, 152–161. 40.
Yamaguchi, M.; Hiramaya, F.; Kanai, M.; Sato, N.; Fukazawa,
K.; Yamashita, K.; Sawada, K.; Koike, T.; Kuwabara, M.;
Ikeda, H.; Ikebuchi, K. Serum-free coculture system for ex
vivo expansion of human cord blood primitive progenitos and
SCID mouse-reconstituting cells using human bone marrow
primary stromal cells. Exp. Hematol. 2001, 29, 174–182. 41.
Almeida-Porada, G.; Brown, R.L.; MacKintosh, F.R.; Zanjani,
E.D. Evaluation of serum-free culture conditions able to
support the ex vivo expansion and engraftment of human
hematopoietic stem cells in human-to sheep xenograft model.
J. Hematother. Stem Cell Res. 2000, 9, 683–693. 42.
McAdams, T.A.; Winter, J.N.; Miller, W.M.; Papoutsakis,
E.T. Hematopoietic cell culture therapies (Part II):
Clinical aspects and applications. Trends in Biotechnol.
1996, 14 (10), 388–396. 43. McAdams, T.; Sandstrom, C.E.;
Miller, W.M.; Bender, J.G.; Papoutsakis, E.T. Ex vivo
expansion of primitive hematopoietic cells for cellular
therapies: An overview. Cytotechnology 1995, 18, 133–146.
44. Verfaillie, C.M. Ex Vivo Expansion of Hematopoietic
Stem Cells. In Hematopoiesis: A Developmental Approach;
Zon, L.I., Ed.; Oxford University Press: New York, 2001;
119–129. T

45. Audet, J.; Zanstra, P.W.; Eaves, C.J.; Piret, J.M.


Advances in hematopoietic stem cell culture. Curr. Opin.
Biotechnol. 1998, 9, 146–151.

46. Broudy, V.C. Stem cell factor and hematopoiesis. Blood


1997, 90, 1354–1364.

47. Lyman, S.D.; Williams, D.E. Biology and potential


clinical applications for flt3 ligand. Curr. Opin. Hematol.
1995, 2, 177–181.

48. Brandt, J.; Bhalla, K.; Hoffman, R. Effects of


interleukin-3 and c-kit ligand on the survival of various
classes of human hematopoietic progenitor cells. Blood
1994, 83 (6), 1507–1514.

49. Lyman, S.D.; Jacobsen, S.E. c-kit ligand and Flt3


ligand: Stem/progenitor cell factors with overlapping yet
distinct activities. Blood 1998, 91, 1101–1134.

50. Shah, A.J.; Smogorzewska, E.M.; Hannum, C.; Crooks,


G.M. Flt3 ligand induces proliferation of quiescent human
bone marrow CD34 + CD38 cells and maintains progenitor
cells in vitro. Blood 1996, 87, 3563–3570.

51. Petzer, A.L.; Zandstra, P.W.; Piret, J.M.; Eaves, C.J.


Differential cytokine effects on primitive (CD34 + CD38 )
human hematopoietic cells: Novel responses to flt3-ligand
and thrombopoietin. J. Exp. Med. 1996, 183, 2551–2558.

52. Won, J.H.; Cho, S.D.; Park, S.K.; Lee, G.T.; Baick,
S.H.; Suh, W.S.; Hong, D.S.; Park, H.S. Thrombopoietin in
synergy with other cytokines for expansion of cord blood
progenitor cells. J. Hematother. Stem Cell Res. 2000, 9,
465–473.
53. Kobayashi, M.; Laver, J.H.; Kato, T.; Miyazaki, H.;
Ogawa, M. Thrombopoietin supports proliferation of human
primitive hematopoietic cells in synergy with steel factor
and/or interleukin-3. Blood 1996, 88, 429–436.

54. Ogawa, M.; Clark, S.C. Synergistic interaction between


interleukin-6 and interleukin-3 in support of stem cell
proliferation in culture. Blood Cells 1988, 14, 329–335.

55. Holyoake, T.L.; Freshney, M.G.; McNair, L.; Parker,


A.N.; McKay, P.J.; Steward, W.P.; Fitzsimons, E.; Graham,
G.J.; Pragnell, I.B. Ex vivo expansion with stem cell
factor and interleukin-11 augments both short-term recovery
posttransplant and the ability to serially transplant
marrow. Blood 1996, 87, 4589–4595.

56. Shih, C.-C.; et al. A secreted and LIF-mediated stromal


cell-derived activity that promotes ex vivo expansion of
human hematopoietic stem cells. Blood 2000, 95, 1957–1966.

57. Bhatia, M.; et al. Bone morphogenetic proteins regulate


the developmental program of human hematopoietic stem
cells. J. Exp. Med. 1999, 189, 1139–1148.

58. Karanu, F.N.; et al. The notch ligand jagged-1


represents a novel factor of human hematopoietic stem
cells. J. Exp. Med. 2000, 192, 1365–1372.

59. Eaves, C.J.; et al. Mechanisms that regulate the cell


cycle status of very primitive hematopoietic cells in
long-term human marrow cultures. II. Analysis of positive
and negative regulators produced by stromal cells within
the adherent layer. Blood 1991, 78, 110–117. 60. Broxmeyer,
H.E.; et al. Comparative analysis of the human macrophage
inflammatory protein family of cytokines (chemokines) on
proliferation of human myeloid progenitor cells.
Interacting effects involving suppression, synergistic
suppression, and blocking of suppression. J. Immunol. 1993,
150, 3448–3458. 61. Verfaillie, C.M.; et al. MIP-1a
combined with IL-3 conserves primitive human LTBMC-IC for
at least 8 weeks in ex vivo ‘‘stroma-non-contact’’
cultures. J. Exp. Med. 1994, 179, 643–649. 62. Zandstra,
P.W.; Eaves, C.J.; Piret, J.M. Expansion of hematopoietic
progenitor cell populations in stirred suspension
bioreactors of normal human bone marrow cells.
Bio/Technology 1994, 12, 909–914. 63. Zandstra, P.W.;
Petzer, A.L.; Eaves, C.J.; Piret, J.M. Cellular
determinants affecting the rate of cytokine depletion in
cultures of human hematopoietic cells. Biotechnol. Bioeng.
1997, 54, 58–68. 64. Koller, M.; Manchel, I.; Palsson,
M.A.; Maher, R.J. Different measures of human hematopoietic
cell culture performance are optimized under vastly
different conditions. Biotechnol. Bioeng. 1996, 50,
505–513. 65. Poloni, A.; Giarratana, M.C.; Kobari, L.;
Firat, H.; Bouchet, S.; Gorin, N.C.; Douay, L. The ex vivo
expansion capacity of normal human bone marrow cells is
dependent on experimental conditions: Role of cell
concentration, serum and CD34 + cell selection in
stroma-free cultures. Hematol. Cell. Ther. 1997, 39, 49–58.
66. Sandstrom, C.E.; et al. Effects of CD34 + cell
selection and perfusion on ex vivo expansion of peripheral
blood mononuclear cells. Blood 1995, 86, 958–970. 67.
Koller, M.R.; Manchel, I.; Newsom, B.S.; Palsson, M.A.;
Palsson, B.O. Bioreactor expansion of human bone marrow:
Comparison of unprocessed, densityseparated and
CD34-enriched cells. J. Hematother. 1995, 4, 159–169. 68.
Martiat, P.; Ferrant, A.; Cogneau, M.; Bol, A.; Rodhain,
J.; Michaux, J.L.; Sokal, G. Assessment of bone marrow
blood flow using positron emission tomography: No
relationship with bone cellularity. Br. J. Haematol. 1987,
66, 307–310. 69. Caldwell, J.; Locey, B.; Clarke, M.F.;
Emerson, S.G.; Palsson, B.O. Influence of medium exchange
schedules on metabolic, growth, and GM-CSF secretion rates
of genetically engineered NIH-3T3 cells. Biotechnol. Prog.
1991, 7, 1–8. 70. Caldwell, J.; Palsson, B.O.; Locey, B.;
Emerson, S.G. Culture perfusion schedules influence the
metabolic activity and granulocyte-macrophage
colony-stimulating factor production rates of human bone
marrow stromal cells. J. Cell. Physiol. 1991, 147, 344–353.
71. Koller, M.R.; Bender, J.G.; Miller, M.E.; Papoutsakis,
E.T. Expansion of primitive human hematopoietic progenitors
in a perfusion bioreactor system with IL-3, IL-6, and stem
cell factor. Bio/Technology 1993, 11, 358–363. 72.
Martinson, J.; Unverzagt, K.; Schaeffer, A.; Smith, S.L.;
Loudovaris, M.; Schneidkraut, M.J.; Bender, J.G.; Van Epps,
D.E. Neutrophil precursor T generation: Effect of culture
conditions. J. Hematother. 1998, 7, 463–471. 73. Schwartz,
R.; Palsson, B.O.; Emerson, S.G. Rapid medium perfusion
rate significantly increases the productivity and longevity
of human bone marrow cultures. Proc. Natl. Acad. Sci.
U.S.A. 1991, 88, 6760– 6764. 74. Patel, S.D.; Papoutsakis,
E.T.; Winter, J.N.; Miller, W.M. The lactate issue
revisited: Novel feeding protocols to examine inhibition of
cell proliferation and glucose metabolism in hematopoietic
cell cultures. Biotechnol. Prog. 2000, 16, 885–892. 75. Oh,
D.; Koller, M.R.; Palsson, B.O. Frequent harvesting from
perfused bone marrow cultures results in increased overall
cell and progenitor expansion. Biotechnol. Bioeng. 1994,
44, 609–616. 76. Haylock, D.N.; To, L.B.; Dowse, T.L.;
Juttner, C.A.; Simmons, P.J. Ex vivo expansion and
maturation of peripheral blood CD34 + cells into the
myeloid lineage. Blood 1992, 80, 1405–1412. 77. Bregni, M.;
Magni, M.; Siena, S.; Di Nicola, M.; Bonadonna, G.; Gianni,
A.M. Human peripheral blood hematopoietic progenitors are
optimal targets of retroviral-mediated gene transfer. Blood
1992, 80, 1418–1422. 78. Piacibello, W.; Sanavio, F.;
Garetto, L.; Severino, A.; Dane, A.; Gammaitoni, I.;
Agglietta, M. Differential growth factor requirement of
primitive cord blood hematopoietic stem cells for
self-renewal and amplification versus proliferation and
differentiation. Leukemia 1998, 12, 718–727. 79. Akatov,
V.; Lezhnev, E.I.; Vexler, A.M.; Kublik, L.N. Low pH value
of pericellular medium as a factor limiting cell
proliferation in dense cultures. Exp. Cell Res. 1985, 160,
412–418. 80. Hevehan, D.; Papoutsakis, E.T.; Miller, W.M.
Physiologically significant effect of pH and oxygen tension
on granulopoiesis. Exp. Hematol. 2000, 28, 267–275. 81.
Carswell, K.; Papoutsakis, E.T. Extracellular pH affects
the proliferation of cultured human T-cells and their
expression of the interleukin-2 receptor. J. Immunol. 2000,
23 (6), 669–674. 82. McAdams, T.A.; Miller, W.M.;
Papoutsakis, E.T. Variations in culture pH affect the
cloning efficiency and differentiation of progenitor cells
in ex vivo haematopoiesis. Br. J. Haematol. 1997, 97,
889–895. 83. Endo, T.; Ishibashi, Y.; Okana, H.; Fukumaki,
Y. Sgnificance of pH on differentiation of human erythroid
cell lines. Leuk. Res. 1994, 18, 49–54. 84. McAdams, T.A.;
Miller, W.M.; Papoutsakis, E.T. pH is a potent modulator of
erythroid differentiation. Br. J. Haematol. 1998, 103,
317–325. 85. Fischkoff, S.; Pollak, A.; Gleich, G.J.;
Testa, J.R.; Misawa, S.; Reber, T.J. Eosinophilic
differentiation of the human promyelocytic leukemia cell
line. J. Exp. Med. 1984, 160, 179–196. 86. Yang, H.;
Miller, W.M.; Papoutsakis, E.T. High pH promotes
megakaryocytic maturation and apoptosis. Stem Cells 2002,
20, 320–328. 87. Tennant, G. Control of pH in human
long-term bone marrow cultures with low-glucose medium
containing zwitterion buffer lengthens the period of
haematopoiesis activity. Br. J. Haematol. 2000, 109,
785–787. 88. Ishikawa, Y.; Ito, T. Kinetics of hematopietic
stem cells in a hypoxic culture. Eur. J. Haematol. 1988,
40, 126–129. 89. Cipolleschi, M.; D’Ippolito, G.; Bernabei,
P.A.; et al. Severe hypoxia enhances the formation of
erythroid bursts from human cord blood cells and the
maintenance of BFU-E in vitro. Exp. Hematol. 1997, 25,
1187–1194. 90. Ivanovic, Z.; Belloc, F.; Faucher, J.; et
al. Hypoxia maintains and interleukin-3 reduces the
pre-colonyforming cell potential of dividing CD34 + murine
bone marrow. Exp. Hematol. 2002, 30, 67–73. 91. Rich, I.;
Kubanek, B. The effect of reduced oxygen tension on colony
formation of erythropoietic cells in vitro. Br. J.
Haematol. 1982, 52, 579–588. 92. Rich, I. A role for the
macrophage in normal hemopoiesis. II. Effect of varying
physiology oxygen tensions on the release of hemopoietic
growth factors from bone-marrow-derived macrophage in
vitro. Exp. Hematol. 1986, 14, 746–751. 93. Cipolleschi,
M.G.; Dello Sbarba, P.; Olivotto, M. The role of hypoxia in
the maintenance of hematopoietic stem cells. Blood 1993,
82, 2031–2037. 94. Ivanovic, Z.; Bartolozzi, B.; Bernabei,
P.A.; et al. Incubation of murine bone marrow cells in
hypoxia ensures the maintenance of marrow-repopulating
ability together with the expansion of committed
progenitors. Br. J. Haematol. 2000, 108, 424–429. 95.
Koller, M.; et al. Reduced oxygen tension increases
hematopoiesis in long-term culture of human stem and
progenitor cells from cord blood and bone marrow. Exp.
Hematol. 1992, 20, 264–270. 96. Mostafa, S.; Miller, W.M.;
Papoutsakis, E.T. Oxygen tension influences the
differentiation, maturation and apoptosis of human
megakaryocytes. Br. J. Haematol. 2000, 111, 879–889. 97.
Pennathur-Das, R.L.; Levitt, L. Augmentation of in vitro
human marrow erythropoiesis under physiological oxygen
tensions is mediated by monocytes and T lymphocytes. Blood
1987, 69 (3), 899–907. 98. Naldini, A.; Carraro, F.;
Silvestri, S.; et al. Hypoxia affects cytokine production
and proliferation responses by human peripheral mononuclear
cells. J. Cell. Physiol. 1997, 173, 335–342. 99. Meagher,
R.; et al. An analysis of the multilineage production of
human hematopoietic progenitors in long-term bone marrow
culture: Evidence that reactive oxygen intermediates
derived from mature phagocytic cells have a role in
limiting progenitor cell self-renewal. Blood 1988, 72,
273–281. 100. Zandstra, P.W.; et al. Cytokine manipulation
of primitive human hematopoietic cell self-renewal. Proc.
Natl. Acad. Sci. U. S. A. 1997, 94, 4698–4703. 101.
Zandstra, P.W.; et al. Ontogeny-associated changes in the
cytokine responses of primitive human haematopoietic cells.
Br. J. Haematol. 1998, 101, 770–778.
Tissue Engineering of Bone Marrow,
Culture Systems

7. Emmons, R.V.; Doren, S.; Zujewski, J.; Cottler-Fox, M.;


Carter, C.S.; Hines, K.; O’Shaughnessy, J.A.; Leitman,
S.F.; Greenblatt, J.J.; Cowan, K.; Dunbar, C.E. Retroviral
gene transduction of adult peripheral blood or
marrow-derived CD34 + cells for six hours without growth
factors or on autologous stroma does not improve marking
efficiency assessed in vivo. Blood 1997, 89, 4040–4046.

8. Barnett, M.J.; Eaves, C.J.; Phillips, G.L.; Gascoyne,


R.D.; Hogge, D.E.; Horsman, D.E.; Humphries, R.K.;
Klingemann, H.G.; Lansdorp, P.M.; Nantel, S.H.; Eaves, A.C.
Autografting with cultured marrow in chronic myeloid
leukemia: Results of a pilot study. Blood 1994, 84,
724–732.

9. Coutinho, L.H.; Testa, N.G.; Chang, J.; Morgenstern, G.;


Harrison, C.; Dexter, T.M. The use of cultured bone marrow
cells in autologous transplantation. Prog. Clin. Biol. Res.
1990, 333, 415–432.

10. Mayani, H. Composition and function of the


hematopoietic microenvironment in human myeloid leukemia.
Leukemia 1996, 10, 1041–1047.

11. Bhatia, M.; McGlave, P.B.; Dewald, G.W.; Blazar, B.R.;


Verfaillie, C.M. Abnormal function of the bone marrow
microenvironment in chronic myelogenous leukemia: Role of
malignant stromal macrophages. Blood 1995, 85, 3636–3645.

12. Chabannon, C.; Olivero, S.; Blaise, D.; Maraninchi, D.;


Viens, P. Ex vivo expansion of human hematopoietic
progenitors and cells to support high-dose chemoradiation
therapy: Five years of clinical experience. Cytokines Cell.
Mol. Ther. 2000, 6, 97–108.

13. Collins, P.C.; Miller, M.E.; Papoutsakis, E.T. Stirred


culture of peripheral and cord blood hematopoietic cells
offers advantages over traditional static systems for
clinically relevant applications. Biotechnol. Bioeng. 1998,
59, 534–543.

14. Douay, L. Culture Conditions for Ex Vivo Expansion of


Hematopoietic Primitive Cells. In Autologous Blood and
Marrow Transplantation X: Proceedings of the Tenth
International Symposium; Dicke, K., Keating, A., Eds.;
Carden Jennings Publishing Co., Ltd.: Charlottesville,
Virginia, 2001; 604–614.
15. Giarratana, M.-C.; Kobari, L.; Neildez Nguyen, T.M.A.;
Firat, H.; Bouchet, S.; Lopez, M.; Gorin, N.-C.; Thierry,
D.; Douay, L. Cell culture bags allow a large extent of ex
vivo expansion of LTCIC and functional mature cells which
can subsequently be frozen: Interest for large-scale
clinical application. Bone Marrow Transplant. 1998, 22 (7),
707–715.

16. Palsson, B.O.; Paek, S.-H.; Schwartz, R.M.; Palsson,


M.A.; Lee, G.-M.; Silver, S.; Emerson, S.G. Expansion of
human bone marrow progenitor cells in a high cell density
continuous perfusion system. Bio/Technology 1993, 11,
368–372.

17. Koller, M.; Emerson, S.G.; Palsson, B.O. Largescale


expansion of human stem and progenitor cells from bone
marrow mononuclear cells in continuous perfusion cultures.
Blood 1993, 82 (2), 378–384.

18. Koller, M.R.; Manchel, I.; Maher, R.J.; Goltry, K.L.;


Armostrong, D.; Smith, A.K. Clinical-scale human umbilical
cord blood cell expansion in a novel automated perfusion
culture system. Bone Marrow Transplant. 1998, 21, 653–663.
19. Collins, P.; Miller, W.M.; Papoutsakis, E.T. Ex vivo
culture systems for hematopoietic cells. Curr. Opin.
Biotechnol. 1996, 7 (2), 223–230. 20. Koller, M.R.;
Bradley, T.R.; Palsson, B.O. Growth factor consumption and
production in perfusion cultures of human bone marrow
correlate with specific cell production. Exp. Hematol. 1995,
23, 1275–1283. 21. Caldwell, J.; Palsson, B.O.; Locey, B.;
Emerson, S.G. Culture perfusion schedules influence the
metabolic activity and granulocyte-macrophage
colony-stimulating factor production rates of human bone
marrow stromal cells. J. Cell. Physiol. 1991, 147, 344–353.
22. Koller, M.R.; Bender, J.G.; Miller, M.E.; Papoutsakis,
E.T. Expansion of primitive human hematopoietic progenitors
in a perfusion bioreactor system with IL-3, IL-6, and stem
cell factor. Bio/Technology 1993, 11, 358–363. 23.
Sandstrom, C.E.; Bender, J.G.; Miller, M.E.; Papoutsakis,
E.T. Development of a novel perfusion chamber to retain
nonadherent cells and its use for comparison of human
‘‘mobilized’’ peripheral blood mononuclear cell cultures
with and without irradiated bone marrow stroma. Biotechnol.
Bioeng. 1996, 50, 493–504. 24. Horner, M.; Miller, M.E.;
Ottino, J.M.; Papoutsakis, E.T. Transport in a grooved
perfusion flat-bed bioreactor for cell therapy applications.
Biotechnol. Prog. 1998, 14, 689–698. 25. Sardonini, C.A.;
Wu, Y.J. Expansion and differentiation of human
hematopoietic cells from static cultures through
small-scale bioreactors. Biotechnol. Prog. 1993, 9,
131–137. 26. Koller, M.R.; Manchel, I.; Newsom, B.S.;
Palsson, M.A.; Palsson, B.O. Bioreactor expansion of human
bone marrow: Comparison of unprocessed, densityseparated
and CD34-enriched cells. J. Hematother. 1995, 4, 159–169.
27. Van Zant, G.; Rummel, S.A.; Koller, M.R.; Larson, D.B.;
Drubachevsky, I.; Palsson, M.A.; Emerson, S.G. Expansion in
bioreactors of human progenitor populations from cord blood
and mobilized peripheral blood. Blood Cells 1994, 20,
482–491. 28. Oh, D.J.; Koller, M.R.; Palsson, B.O. Frequent
harvesting from perfused bone marrow cultures results in
increased overall cell and progenitor expansion.
Biotechnol. Bioeng. 1994, 44, 609–616. 29. Jaroscak, J.;
Goltry, K.; Smith, A.; Waters-Pick, B.; Martin, P.L.;
Driscoll, T.A.; Howrey, R.; Chao, N.; Douville, J.; Burhop,
S.; Fu, P.; Kurtzberg, J. Augmentation of umbilical cord
(UCB) transplantation with ex vivo-expanded UCB cells:
Results of a phase 1 trial using the AastromReplicell
System. Blood 2003, 101 (12), 5061–5067. 30. Zandstra,
P.W.; Nagy, A. Stem cell bioengineering. Annu. Rev. Biomed.
Eng. 2001, 3, 275–305. 31. Zandstra, P.W.; Eaves, C.J.;
Piret, J.M. Expansion of hematopoietic progenitor cell
populations in stirred suspension bioreactors of normal
human bone marrow cells. Bio/Technology 1994, 12, 909–914.
T 32. Pierson, B.A.; Europa, A.F.; Hu, W.S.; Miller, J.S.
Production of human natural killer cells for adoptive
immunotherapy using a computer-controlled stirredtank
bioreactor. J. Hematother. 1996, 5, 474–483. 33. McDowell,
C.L.; Papoutsakis, E.T. Increased agitation intensity
increases CD13 receptor surface content and mRNA levels,
and alters the metabolism of HL60 cells cultures in stirred
tank bioreactors. Biotechnol. Bioeng. 1998, 60, 239–250.
34. Nielsen, L.K. Bioreactors for hematopoietic cell
culture. Annu. Rev. Biomed. Eng. 1999, 1, 129–152. 35. Dao,
M.A.; Hashimo, K.; Kato, I.; Nolta, J.A. Adhesion to
fibronectin maintains regenarative capacity during ex vivo
culture and transduction of human hematopoietic stem and
progenitor cells. Blood 1998, 92, 4612–4621. 36. Chute,
J.P.; Saini, A.A.; Kampen, R.L.; Wells, M.R.; Davis, T.A. A
comparative study of the cell cycle status and primitive
cell adhesion molecule profile of human CD34 + cells
cultured in stroma-free versus porcine microvascular
endothelial cell cultures. Exp. Hematol. 1999, 27, 370–379.
37. Naughton, B.A.; Naughton, G.K. Hematopoiesis on nylon
mesh templates—Comparative long-term bonemarrow culture and
the influence of stromal support cells. Ann. N.Y. Acad. Sci.
1989, 554, 125–140. 38. Naughton, B.A.; Jacob, L.;
Naughton, G.K. A three-dimensional culture system for the
growth of hematopoietic cells. Prog. Clin. Biol. Res. 1990,
333, 435–445. 39. Naughton, B.A.; Tjota, A.; Sibanda, B.;
Naughton, G.K. Hematopoiesis on suspended nylon
screenstromal cell microenvironments. J. Biomech. Eng.,
Trans. ASME 1991, 113, 171–177. 40. Mantalaris, A.; Keng,
P.; Bourne, P.; Chang, A.Y.C.; Wu, J.H.D. Engineering a
human bone marrow model: A case study on ex vivo
erythropoiesis. Biotechnol. Prog. 1998, 14, 126–133. 41.
Wang, T.-Y.; Brennan, J.K.; Wu, J.H.D. Multilineal
hematopoiesis in a three-dimensional murine long-term bone
marrow culture. Exp. Hematol. 1995, 23, 26–32. 42. Wang,
M.; Chen, L.J.; Ni, J.; Weng, C.; Yve, Y. Manufacturing and
evaluation of bioactive and biodegradable materials and
scaffolds for tissue engineering. J. Mater. Sci., Mater.
Med. 2001, 12, 855–860. 43. Xiong, F.; Chen, Z.; Liu, H.;
Xu, Z.; Li, X. Ex vivo expansion of human umbilical cord
blood hematopoietic progenitor cells in a novel
three-dimensional culture system. Biotechnol. Lett. 2002,
24, 1421–1426. 44. Bagley, J.; Rosenzweig, M.; Mark, D.F.;
Pykett, M.J. Extended culture of multipotent hematopoietic
progenitors without cytokine augmentation in a novel
threedimensional device. Exp. Hematol. 1999, 27, 496–504.
45. Banu, N.; Rosenzweig, M.; Kim, H.; Bagley, J.; Pykett,
M. Cytokine-augmented culture of haematopoietic progenitor
cells in a novel three-dimensional cell growth matrix.
Cytokine 2001, 13 (6), 349–358. 46. Knospe, W.; Husseini,
S.G.; Fried, W. Hematopoiesis on cellulose ester membranes.
XI. Induction of new bone and a hematopoietic
microenvironment by matrix factors secreted by marrow
stromal cells. Blood 1989, 74 (1), 66–70. 47. Levee, M.;
Lee, G.-M.; Paek, S.-H.; Palsson, B.O. Microencapsulated
human bone marrow cultures: A potential culture system for
the clonol outgrowth of hematopoietic progenitor cells.
Biotechnol. Bioeng. 1994, 43, 734–739. 48. Tun, T.;
Miyoshi, H.; Aung, T.; Takahashi, S.; Shimizu, R.; Kuroha,
T.; Yamamoto, M.; Ohshima, N. Effect of growth factors on
ex vivo bone marrow cell expansion using three-dimensional
matrix support. Artif. Organs 2002, 26 (4), 333–339. 49.
Li, Y.; Kniss, D.A.; Yang, S.-T.; Lasky, L.C. Human cord
cell hematopoiesis in three-dimensional nonwoven fibrous
matrices: In vitro simulation of the marrow
microenvironment. J. Hematother. Stem Cell Res. 2001, 10,
355–368. 50. Godbey, W.; Atala, A. In vitro systems for
tissue engineering. Ann. N.Y. Acad. Sci. 2002, 961, 10–26.
51. Jelinek, N.; Schmidt, S.; Hilbert, U.; Thoma, S.;
Biselli, M.; Wandrey, C. Novel bioreactors for the ex vivo
cultivation of hematopoietic cells. Eng. Life Sci. 2002, 2
(1), 15–18. 52. Sasaki, T.; Takagi, M.; Yoshida, T. 3D
culture of murine hematopoietic cells with spatial
development of stromal cells in nonwoven fabrics.
Cytotherapy 2002, 4 (3), 285–291. 53. Tomimori, Y.; Takagi,
M.; Yoshida, T. The construction of an in vitro
three-dimensional hematopoietic microenvironment for mouse
bone marrow cells employing porous carriers. Cytotechnology
2000, 34, 121–130. 54. Rosenzweig, M.; Pykett, M.J.; Marks,
D.F.; Johnson, R.P. Enhanced maintenance and retroviral
transduction of primitive hematopoietic progenitor cells
using a novel three-dimensional culture system. Gene Ther.
1997, 4, 928–936. 55. Washburn, N.; Simon, C.G.; Tona, A.;
Elgendy, H.M.; Karim, A.; Amis, E.J. Co-extrusion of
biocompatible polymers for scaffolds with co-continuous
morphology. J. Biomed. Materi. Res. 2002, 60, 20–29. 56.
Liao, C.-J.; Chen, C.-F.; Chen, J.-H.; Chiang, S.-F.; Lin,
Y.-J.; Chang, K.-Y. Fabrication of porous biodegradable
polymer scaffolds using a solvent merging/ particulate
leaching method. J. Biomed. Materi. Res. 2002, 59, 678–681.
57. Lin, H.-R.; Kuo, C.-J.; Yang, C.Y.; Shaw, S.-Y.; Wu,
Y.-J. Preparation of macroporous biodegradable PLGA
scaffolds for cell attachment with the use of mixed salts
as porogen additives. J. Biomed. Materi. Res. 2002, 63,
271–279. 58. Highfill, J.; Haley, S.D.; Kompala, D.S.
Large-scale production of murine bone marrow cells in an
airlift packed bed bioreactor. Biotechnol. Bioeng. 1996,
50, 514–520.
Tissue Engineering of Breast

1. American Cancer Society. Breast Cancer Facts and


Figures. Available at: www.cancer.org. Accessed on
September 2006.

2. Tozeren, A.; Coward, C.W.; Petushi, S.P. Origins and


evolution of cell phenotypes in breast tumors. J. Theor.
Biol. 2005, 233 (1), 43–54.

3. Available at: www.cancer.gov/cancertopics/factsheet/


Therapy/adjuvant-breast. Accessed on May 2006.

4. Taylor, C.W.; Horgan, K.; Dodwell, D. Oncological


aspects of breast reconstruction. Breast 2005, 14 (2),
118–130.

5. Noda, S.; Eberlein, T.J.; Eriksson, E. Breast


reconstruction. Cancer 1994, 74, 376–380.

6. Holder, W.D., Jr; Gruber, H.E.; Moore, A.L.; et al.


Cellular ingrowth and thickness changes in poly-llactide
and polyglycolide matrices implanted subcutaneously in the
rat. J. Biomed. Mater. Res. 1998, 41 (3), 412–421.

7. Gray, H. The Urogenital System, 30th Ed. In Anatomy of


the Human Body; Lea & Febiger: Philadelphia, PA, 1985;
1581–1586.

8. Cavin, A.N. Adipocyte Response to Injectable Breast


Tissue Engineering Scaffolds; Clemson University Press:
Clemson, SC, 2005.

9. Patrick, C.W., Jr; Mikos, A.G.; McIntire, L.V. Tissue


engineered adipose tissue, 1st Ed. In Frontiers in Tissue
Engineering; Pergamon: Oxford, UK, 1998; 369–382.

10. Burg, K.J.L.; Cavin, A.N.; Gomillion, C.T.; et al. 3-D


Test Systems: New Tools for Unlocking the Mysteries of
Breast Cancer. Presented at: Mammary Gland Biology Gordon
Research Conference, Providence, Rhode Island, 2005.

11. Patrick, C.W., Jr; Chauvin, P.B.; Hobley, J.; et al.


Preadipocyte seeded PLGA scaffolds for adipose tissue
engineering. Tissue Eng. 1999, 5 (2), 139–151. 12. Vacanti,
J.P.; Atala, A.; Mooney, D.J.; et al. Breast Tissue
Engineering. U.S. Patent 5,716,404, February 10, 1998. 13.
Burg, K.J.L.; Halberstadt, C.R.; Holder, W.D., Jr.
Absorbable Tissue Expander. U.S. Patent 6,206,930, March
27, 2001. 14. Cui, Q.; Wang, G.J.; Balian, G.
Steroid-induced adipogenesis in a pluripotential cell line
from bone marrow. J. Bone Joint Surg. Am. 1997, 79 (7),
1054–1063. 15. Tabata, Y.; Miyao, M.; Inamoto, T.; et al.
De novo formation of adipose tissue by controlled release
of basic fibroblast growth factor. Tissue Eng. 2000, 6 (3),
279–289. 16. Burg, K.J.L.; Austin, C.E.; Culberson, C.R.;
et al. A novel approach to tissue engineering: injectable
composites. Transactions of the 2000 World Biomaterials
Congress; Kona: Hawaii, 2000. 17. Halberstadt, C.; Austin,
C.; Rowley, J.; et al. A hydrogel material for plastic and
reconstructive applications injected into the subcutaneous
space of a sheep. Tissue Eng. 2002, 8 (2), 309–319. 18.
Patrick, C.W. Breast tissue engineering. Annu. Rev. Biomed.
Eng. 2004, 6, 109–130. 19. Patrick, C.W., Jr. Adipose
tissue engineering: the future of breast and soft tissue
reconstruction following tumor resection. Semin. Surg.
Oncol. 2000, 19 (3), 302– 311. 20. Gomillion, C.T.; Burg,
K.J.L. Evaluation of tissue engineered injectable devices
for breast tissue engineering. In Transactions of the 30th
Annual Meeting of the Society for Biomaterials; Memphis:
Tennessee, 2005. 21. Parzel, C.A.; Cavin, A.N.; Ellis,
S.E.; et al. Analysis of bovine intradermal tissue response
to injectable tissue engineering composites. In
Transactions of the 30th Annual Meeting of the Society for
Biomaterials; Memphis: Tennessee, 2005. 22. Jatoi, I.;
Miller, A.B. Why is breast-cancer mortality declining?
Lancet Oncol. 2003, 4 (4), 251–254. 23. Muss, H.B. Adjuvant
therapy for older women with breast cancer. Breast 2003, 12
(6), 550–557. 24. Singletary, S.E. Surgical margins in
patients with earlystage breast cancer treated with breast
conservation therapy. Am. J. Surg. 2002, 184 (5), 383–393.
25. Available at: www.cancer.org/docroot/ETO/content/
ETO_1_2X_Chemotherapy_What_It_Is_How_It_Helps. asp.
Accessed on May 2006. 26. Dow, K.H. Contemporary Issues in
Breast Cancer: A Nursing Perspective, 2nd Ed.; Jones and
Bartlett Publishers International: London, UK, 2004; 349.
27. O’Regan, R.; Jordan, V.C.; Gradishar, W.J. Tamoxifen
and contralateral breast cancer. J. Am. Coll. Surg. 1999,
188 (6), 678–683. 28. Chung, C.T.; Carlson, R.W. The role
of aromatase inhibitors in early breast cancer. Curr.
Treat. Options Oncol. 2003, 4 (2), 133–140. 29. Maillard,
S.; Ameller, T.; Gauduchon, J.; et al. Innovative drug
delivery nanosystems improve the antitumor activity in
vitro and in vivo of anti-estrogens in human breast cancer
and multiple myeloma. J. Steroid. Biochem. Mol. Biol. 2005,
94 (1–3), 111–121.
Tissue Engineering of Cardiac Muscle

1. Bader, D.; Oberpriller, J.O. Repair and reorganization


of minced cardiac muscle in the adult newt (Notophthalmus
viridescens). J. Morphol. 1978, 155 (3), 349–357.

2. Poss, K.D.; Wilson, L.G.; Keating, M.T. Heart


regeneration in zebrafish. Science 2002, 298 (5601), 2188–
2190.

3. Leor, J.; Patterson, M.; Quinones, M.J.; Kedes, L.H.;


Kloner, R.A. Transplantation of fetal myocardial tissue
into the infarcted myocardium of rat. A potential method
for repair of infarcted myocardium? Circulation 1996, 94 (9
Suppl), II332–II336.

4. Carrier, R.L.; Papadaki, M.; Rupnick, M.; Schoen, F.J.;


Bursac, N.; Langer, R.; Freed, L.E.; Vunjak-Novakovic, G.
Cardiac tissue engineering: cell seeding, cultivation
parameters, and tissue construct characterization.
Biotechnol. Bioeng. 1999, 64 (5), 580–589.

5. Eschenhagen, T.; Fink, C.; Remmers, U.; Scholz, H.;


Wattchow, J.; Weil, J.; Zimmermann, W.; Dohmen, H.H.;
Schafer, H.; Bishopric, N.; Wakatsuki, T.; Elson, E.L.
Three-dimensional reconstitution of embryonic
cardiomyocytes in a collagen matrix: a new heart muscle
model system. Faseb J. 1997, 11 (8), 683–694.

6. Zimmermann, W.H.; Fink, C.; Kralisch, D.; Remmers, U.;


Weil, J.; Eschenhagen, T. Three-dimensional engineered
heart tissue from neonatal rat cardiac myocytes.
Biotechnol. Bioeng. 2000, 68 (1), 106–114. 7. Akins, R.E.;
Boyce, R.A.; Madonna, M.L.; Schroedl, N.A.; Gonda, S.R.;
McLaughlin, T.A.; Hartzell, C.R. Cardiac organogenesis in
vitro: reestablishment of three-dimensional tissue
architecture by dissociated neonatal rat ventricular cells.
Tissue Eng. 1999, 5 (2), 103–118. 8. Kofidis, T.; Akhyari,
P.; Boublik, J.; Theodorou, P.; Martin, U.; Ruhparwar, A.;
Fischer, S.; Eschenhagen, T.; Kubis, H.P.; Kraft, T.; Leyh,
R.; Haverich, A. In vitro engineering of heart muscle:
artificial myocardial tissue. J. Thorac. Cardiovasc. Surg.
2002, 124 (1), 63–69. 9. Leor, J.; Aboulafia-Etzion, S.;
Dar, A.; Shapiro, L.; Barbash, I.M.; Battler, A.; Granot,
Y.; Cohen, S. Bioengineered cardiac grafts: A new approach
to repair the infarcted myocardium? Circulation 2000, 102
(19 Suppl 3), III56–III61. 10. Li, R.K.; Jia, Z.Q.; Weisel,
R.D.; Mickle, D.A.; Choi, A.; Yau, T.M. Survival and
function of bioengineered cardiac grafts. Circulation 1999,
100 (19 Suppl), II63–II69. 11. Shimizu, T.; Yamato, M.;
Isoi, Y.; Akutsu, T.; Setomaru, T.; Abe, K.; Kikuchi, A.;
Umezu, M.; Okano, T. Fabrication of pulsatile cardiac
tissue grafts using a novel 3-dimensional cell sheet
manipulation technique and temperature-responsive cell
culture surfaces. Circ. Res. 2002, 90 (3), e40. 12.
Zimmermann, W.H.; Schneiderbanger, K.; Schubert, P.; Didie,
M.; Munzel, F.; Heubach, J.F.; Kostin, S.; Neuhuber, W.L.;
Eschenhagen, T. Tissue engineering of a differentiated
cardiac muscle construct. Circ. Res. 2002, 90 (2), 223–230.
13. van Luyn, M.J.; Tio, R.A.; Gallego y van Seijen, X.J.;
Plantinga, J.A.; de Leij, L.F.; DeJongste, M.J.; van
Wachem, P.B. Cardiac tissue engineering: characteristics of
in unison contracting twoand threedimensional neonatal rat
ventricle cell (co)-cultures. Biomaterials 2002, 23 (24),
4793–4801. 14. Lanza, R.P.; Chung, H.Y.; Yoo, J.J.;
Wettstein, P.J.; Blackwell, C.; Borson, N.; Hofmeister, E.;
Schuch, G.; Soker, S.; Moraes, C.T.; West, M.D.; Atala, A.
Generation of histocompatible tissues using nuclear
transplantation. Nat. Biotechnol. 2002, 20 (7), 689–696.
15. Langer, R.; Vacanti, J.P. Tissue engineering. Science
1993, 260 (5110), 920–926. 16. Vacanti, J.P.; Langer, R.;
Upton, J.; Marler, J.J. Transplantation of cells in
matrices for tissue regeneration. Adv. Drug. Deliv. Rev.
1998, 33 (1–2), 165–182. 17. Sodian, R.; Hoerstrup, S.P.;
Sperling, J.S.; Daebritz, S.; Martin, D.P.; Moran, A.M.;
Kim, B.S.; Schoen, F.J.; Vacanti, J.P.; Mayer, J. E., Jr.
Early in vivo experience with tissue-engineered trileaflet
heart valves. Circulation 2000, 102 (19 Suppl 3),
III22–III29. 18. Steinhoff, G.; Stock, U.; Karim, N.;
Mertsching, H.; Timke, A.; Meliss, R.R.; Pethig, K.;
Haverich, A.; Bader, A. Tissue engineering of pulmonary
heart valves on allogenic acellular matrix conduits: In
vivo restoration of valve tissue. Circulation 2000, 102 (19
Suppl 3), III50–III55. 19. Fink, C.; Ergun, S.; Kralisch,
D.; Remmers, U.; Weil, J.; Eschenhagen, T. Chronic stretch
of engineered heart tissue induces hypertrophy and
functional improvement. Faseb J. 2000, 14 (5), 669–679. 20.
Eschenhagen, T.; Didie, M.; Munzel, F.; Schubert, P.;
Schneiderbanger, K.; Zimmermann, W.H. 3D engineered heart
tissue for replacement therapy. Basic Res. Cardiol. 2002,
97 (Suppl 1), I146–I152. 21. Gulch, R.W.; Ebrecht, G.
Mechanics of rat myocardium revisited: investigations of
ultra-thin cardiac muscles under high energy demand. Basic
Res. Cardiol. 1987, 82 (Suppl 2), 263–274. 22. Nag, A.C.;
Zak, R. Dissociation of adult mammalian heart into single
cell suspension: An ultrastructural study. J. Anat. 1979,
129 (3), 541–559. 23. Gepstein, L. Derivation and potential
applications of human embryonic stem cells. Circ. Res.
2002, 91 (10), 866–876. 24. Pasumarthi, K.B.; Field, L.J.
Cardiomyocyte cell cycle regulation. Circ. Res. 2002, 90
(10), 1044–1054. 25. Kehat, I.; Kenyagin-Karsenti, D.;
Snir, M.; Segev, H.; Amit, M.; Gepstein, A.; Livne, E.;
Binah, O.; ItskovitzEldor, J.; Gepstein, L. Human embryonic
stem cells can differentiate into myocytes with structural
and functional properties of cardiomyocytes. J. Clin.
Invest. 2001, 108 (3), 407–414. 26. Jiang, Y.; Jahagirdar,
B.N.; Reinhardt, R.L.; Schwartz, R.E.; Keene, C.D.;
Ortiz-Gonzalez, X.R.; Reyes, M.; Lenvik, T.; Lund, T.;
Blackstad, M.; Du, J.; Aldrich, S.; Lisberg, A.; Low, W.C.;
Largaespada, D.A.; Verfaillie, C.M. Pluripotency of
mesenchymal stem cells derived from adult marrow. Nature
2002, 418 (6893), 41–49. 27. Fandrich, F.; Lin, X.; Chai,
G.X.; Schulze, M.; Ganten, D.; Bader, M.; Holle, J.; Huang,
D.S.; Parwaresch, R.; Zavazava, N.; Binas, B.
Preimplantation-stage stem cells induce long-term
allogeneic graft acceptance without supplementary host
conditioning. Nat. Med. 2002, 8 (2), 171–178. 28. Gray,
M.O.; Long, C.S.; Kalinyak, J.E.; Li, H.T.; Karliner, J.S.
Angiotensin II stimulates cardiac myocyte hypertrophy via
paracrine release of TGF-beta 1 and endothelin-1 from
fibroblasts. Cardiovasc Res 1998, 40 (2), 352–363. 29.
Zimmermann, W.H.; Didie, M.; Wasmeier, G.H.; Nixdorff, U.;
Hess, A.; Melnychenko, I.; Boy, O.; Neuhuber, W.L.; Weyand,
M.; Eschenhagen, T. Cardiac grafting of engineered heart
tissue in syngenic rats. Circulation 2002, 106 (12 Suppl
1), I151–I1157. 30. Wurmser, A.E.; Gage, F.H. Stem cells:
Cell fusion causes confusion. Nature 2002, 416 (6880),
485–487. T
Tissue Engineering of Cartilage

15. Browne, J.E.; Anderson, A.F.; Arciero, R.; Mandelbaum,


B.; Moseley J.B., Jr.; Micheli, L.J.; Fu, F.; Erggelet, C.
Clinical outcome of autologous chondrocyte implantation at
5 years in US subjects. Clin. Orthop. Relat. Res. 2005,
(436), 237–245.

16. Peterson, L.; Minas, T.; Brittberg, M.; Nilsson, A.;


Sjogren-Jansson, E.; Lindahl, A. Twoto 9-year outcome after
autologous chondrocyte transplantation of the knee. Clin.
Orthop. Relat. Res. 2000, (374), 212–234.

17. Hasler, E.M.; Herzog, W.; Wu, J.Z.; Muller, W.; Wyss,
U. Articular cartilage biomechanics: theoretical models,
material properties, and biosynthetic response. Crit. Rev.
Biomed. Eng. 1999, 27 (6), 415–488.

18. Archer, C.W.; Francis-West, P. The chondrocyte. Int. J.


Biochem. Cell Biol. 2003, 35 (4), 401–404.

19. Heinegard, D.; Oldberg, A. Structure and biology of


cartilage and bone matrix noncollagenous macromolecules.
FASEB J. 1989, 3 (9), 2042–2051.

20. Roughley, P.J. Articular cartilage and changes in


arthritis: noncollagenous proteins and proteoglycans in the
extracellular matrix of cartilage. Arthritis Res. 2001, 3
(6), 342–347.

21. Giannoni, P.; Siegrist, M.; Hunziker, E.B.; Wong, M.


The mechanosensitivity of cartilage oligomeric matrix
protein (COMP). Biorheology 2003, 40 (1–3), 101–109.

22. Hecht, J.T.; Nelson, L.D.; Crowder, E.; Wang, Y.;


Elder, F.F.; Harrison, W.R.; Francomano, C.A.; Prange,
C.K.; Lennon, G.G.; Deere, M. Mutations in exon 17B of
cartilage oligomeric matrix protein (COMP) cause
pseudoachondroplasia. Nat. Genet. 1995, 10 (3), 325–329.

23. Wong, M.; Carter, D.R. Articular cartilage functional


histomorphology and mechanobiology: a research perspective.
Bone 2003, 33 (1), 1–13.

24. Schumacher, B.L.; Block, J.A.; Schmid, T.M.; Aydelotte,


M.B.; Kuettner, K.E. A novel proteoglycan synthesized and
secreted by chondrocytes of the superficial zone of
articular cartilage. Arch. Biochem. Biophys. 1994, 311 (1),
144–152.
25. Lorenzo, P.; Aman, P.; Sommarin, Y.; Heinegard, D. The
human CILP gene: exon/intron organization and chromosomal
mapping. Matrix Biol. 1999, 18 (5), 445–454.

26. Li, L.; Buschmann, M.D.; Shirazi-Adl, A. The role of


fibril reinforcement in the mechanical behavior of
cartilage. Biorheology 2002, 39 (1–2), 89–96.

27. Oegema T.R., Jr.; Thompson R.C., Jr. The zone of


calcified cartilage: its role in osteoarthritis. In
Articular Cartilage and Osteoarthritis; Kuettner, K., Ed.;
Raven Press: New York, U.S.A., 1992; 319–331.

28. Mente, P.L.; Lewis, J.L. Elastic modulus of calcified


cartilage is an order of magnitude less than that of
subchondral bone. J. Orthop. Res. 1994, 12 (5), 637–647.

29. Park, S.; Hung, C.T.; Ateshian, G.A. Mechanical


response of bovine articular cartilage under dynamic
unconfined compression loading at physiological stress
levels. Osteoarthr. Cartil. 2004, 12 (1), 65–73. 30.
Thambyah, A.; Goh, J.C.; De, S.D. Contact stresses in the
knee joint in deep flexion. Med. Eng. Phys. 2005, 27 (4),
329–335. 31. Mow, V.C.; Holmes, M.H.; Lai, W.M. Fluid
transport and mechanical properties of articular cartilage:
a review. J. Biomech. 1984, 17 (5), 377–394. 32. Carter,
D.R.; Wong, M. Modelling cartilage mechanobiology. Philos.
Trans. R. Soc. Lond. B Biol. Sci. 2003, 358 (1437),
1461–1471. 33. Darling, E.M.; Athanasiou, K.A. Articular
cartilage bioreactors and bioprocesses. Tissue Eng. 2003, 9
(1), 9–26. 34. Vunjak-Novakovic, G.; Martin, I.; Obradovic,
B.; Treppo, S.; Grodzinsky, A.J.; Langer, R.; Freed, L.E.
Bioreactor cultivation conditions modulate the composition
and mechanical properties of tissue-engineered cartilage.
J. Orthop. Res. 1999, 17 (1), 130–138. 35. Knudson, W.;
Loeser, R.F. CD44 and integrin matrix receptors participate
in cartilage homeostasis. Cell Mol. Life Sci. 2002, 59 (1),
36–44. 36. Millward-Sadler, S.J.; Salter, D.M.
Integrin-dependent signal cascades in chondrocyte
mechanotransduction. Ann. Biomed. Eng. 2004, 32 (3),
435–446. 37. Clancy, R.M.; Rediske, J.; Tang, X.; Nijher,
N.; Frenkel, S.; Philips, M.; Abramson, S.B. Outside-in
signaling in the chondrocyte. Nitric oxide disrupts
fibronectin-induced assembly of a subplasmalemmal actin/rho
A/focal adhesion kinase signaling complex. J. Clin. Invest.
1997, 100 (7), 1789–1796. 38. Whitmarsh, A.J.; Davis, R.J.
Regulation of transcription factor function by
phosphorylation. Cell Mol. Life Sci. 2000, 57 (8–9),
1172–1183. 39. Fanning, P.J.; Emkey, G.; Smith, R.J.;
Grodzinsky, A.J.; Szasz, N.; Trippel, S.B. Mechanical
regulation of mitogen-activated protein kinase signaling in
articular cartilage. J. Biol. Chem. 2003, 278 (51),
50,940–50,948. 40. Wang, G.; Woods, A.; Sabari, S.;
Pagnotta, L.; Stanton, L.A.; Beier, F. RhoA/ROCK signaling
suppresses hypertrophic chondrocyte differentiation. J.
Biol. Chem. 2004, 279 (13), 13,205–13,214. 41. Woods, A.;
Wang, G.; Beier, F. RhoA/ROCK signaling regulates Sox9
expression and actin organization during chondrogenesis. J.
Biol. Chem. 2005, 280 (12), 11,626–11,634. 42. Willers, C.;
Chen, J.; Wood, D.; Xu, J.; Zheng, M.H. Autologous
chondrocyte implantation with collagen bioscaffold for the
treatment of osteochondral defects in rabbits. Tissue Eng.
2005, 11 (7–8), 1065–1076. 43. Dell’Accio, F.; Vanlauwe,
J.; Bellemans, J.; Neys, J.; De Bari, C.; Luyten, F.P.
Expanded phenotypically stable chondrocytes persist in the
repair tissue and contribute to cartilage matrix formation
and structural integration in a goat model of autologous
chondrocyte implantation. J. Orthop. Res. 2003, 21 (1),
123–131. 44. Vasara, A.I.; Hyttinen, M.M.; Lammi, M.J.;
Lammi, P.E.; Langsjo, T.K.; Lindahl, A.; Peterson, L.;
Kellomaki, M.; Konttinen, Y.T.; Helminen, H.J.; Kiviranta,
I. Subchondral bone reaction associated with chondral
defect and attempted cartilage repair in goats. Calcif.
Tissue Int. 2004, 74 (1), 107–114. 45. Frenkel, S.R.; Di
Cesare, P.E. Scaffolds for articular cartilage repair. Ann.
Biomed. Eng. 2004, 32 (1), 26–34. 46. Hollister, S.J.
Porous scaffold design for tissue engineering. Nat. Mater.
2005, 4 (7), 518–524. 47. Shin, H.; Jo, S.; Mikos, A.G.
Biomimetic materials for tissue engineering. Biomaterials
2003, 24 (24), 4353–4364. 48. Yarlagadda, P.K.;
Chandrasekharan, M.; Shyan, J.Y. Recent advances and
current developments in tissue scaffolding. Biomed. Mater.
Eng. 2005, 15 (3), 159–177. 49. Altman, G.H.; Diaz, F.;
Jakuba, C.; Calabro, T.; Horan, R.L.; Chen, J.; Lu, H.;
Richmond, J.; Kaplan, D.L. Silk-based biomaterials.
Biomaterials 2003, 24 (3), 401–416. 50. Ma, Z.; Kotaki, M.;
Inai, R.; Ramakrishna, S. Potential of nanofiber matrix as
tissue-engineering scaffolds. Tissue Eng. 2005, 11 (1–2),
101–109. 51. Stayton, P.S.; El Sayed, M.E.; Murthy, N.;
Bulmus, V.; Lackey, C.; Cheung, C.; Hoffman, A.S. ‘Smart’
delivery systems for biomolecular therapeutics. Orthod.
Craniofac. Res. 2005, 8 (3), 219–225. 52. Rosso, F.;
Marino, G.; Giordano, A.; Barbarisi, M.; Parmeggiani, D.;
Barbarisi, A. Smart materials as scaffolds for tissue
engineering. J. Cell Physiol. 2005, 203 (3), 465–470. 53.
Rahfoth, B.; Weisser, J.; Sternkopf, F.; Aigner, T.; von
der Mark, K.; Brauer, R. Transplantation of allograft
chondrocytes embedded in agarose gel into cartilage defects
of rabbits. Osteoarthr. Cartil. 1998, 6 (1), 50–65. 54.
Beekman, B.; Verzijl, N.; Bank, R.A.; von der Mark, K.;
TeKoppele, J.M. Synthesis of collagen by bovine
chondrocytes cultured in alginate; posttranslational
modifications and cell-matrix interaction. Exp. Cell Res.
1997, 237 (1), 135–141. 55. Martin, I.; Obradovic, B.;
Treppo, S.; Grodzinsky, A.J.; Langer, R.; Freed, L.E.;
Vunjak-Novakovic, G. Modulation of the mechanical
properties of tissue engineered cartilage. Biorheology
2000, 37 (1–2), 141–147. 56. Riesle, J.; Hollander, A.P.;
Langer, R.; Freed, L.E.; Vunjak-Novakovic, G. Collagen in
tissue-engineered cartilage: types, structure, and
crosslinks. J. Cell Biochem. 1998, 71 (3), 313–327. 57.
Waldman, S.D.; Grynpas, M.D.; Pilliar, R.M.; Kandel, R.A.
Characterization of cartilagenous tissue formed on calcium
polyphosphate substrates in vitro. J. Biomed. Mater. Res.
2002, 62 (3), 323–330. 58. Kohn, D.H.; Sarmadi, M.; Helman,
J.I.; Krebsbach, P.H. Effects of pH on human bone marrow
stromal cells in vitro: implications for tissue engineering
of bone. J. Biomed. Mater. Res. 2002, 60 (2), 292–299. 59.
Sittinger, M.; Reitzel, D.; Dauner, M.; Hierlemann, H.;
Hammer, C.; Kastenbauer, E.; Planck, H.; Burmester, G.R.;
Bujia, J. Resorbable polyesters in cartilage engineering:
affinity and biocompatibility of polymer fiber structures to
chondrocytes. J. Biomed. Mater. Res. 1996, 33 (2), 57–63.
60. Spiteri, C.G.; Pilliar, R.M.; Kandel, R.A. Substrate
porosity enhances chondrocyte attachment, spreading, and
cartilage tissue formation in vitro. J. Biomed. Mater. Res.
in press. 61. Alhadlaq, A.; Mao, J.J. Tissue-engineered
osteochondral constructs in the shape of an articular
condyle. J. Bone Joint Surg. Am. 2005, 87 (5), 936–944. 62.
Schaefer, D.; Martin, I.; Jundt, G.; Seidel, J.; Heberer,
M.; Grodzinsky, A.; Bergin, I.; Vunjak-Novakovic, G.;
Freed, L.E. Tissue-engineered composites for the repair of
large osteochondral defects. Arthritis Rheum. 2002, 46 (9),
2524–2534. 63. van Susante, J.L.; Buma, P.; Homminga, G.N.;
van den Berg, W.B.; Veth, R.P. Chondrocyte-seeded
hydroxyapatite for repair of large articular cartilage
defects. A pilot study in the goat. Biomaterials 1998, 19
(24), 2367–2374. 64. Sherwood, J.K.; Riley, S.L.;
Palazzolo, R.; Brown, S.C.; Monkhouse, D.C.; Coates, M.;
Griffith, L.G.; Landeen, L.K.; Ratcliffe, A. A
three-dimensional osteochondral composite scaffold for
articular cartilage repair. Biomaterials 2002, 23 (24),
4739–4751. 65. Pilliar, R.M.; Filiaggi, M.J.; Wells, J.D.;
Grynpas, M.D.; Kandel, R.A. Porous calcium polyphosphate
scaffolds for bone substitute applications—in vitro
characterization. Biomaterials 2001, 22 (9), 963–972. 66.
Grynpas, M.D.; Pilliar, R.M.; Kandel, R.A.; Renlund, R.;
Filiaggi, M.; Dumitriu, M. Porous calcium polyphosphate
scaffolds for bone substitute applications in vivo studies.
Biomaterials 2002, 23 (9), 2063–2070. 67. Hamilton, D.J.;
Seguin, C.A.; Wang, J.; Pilliar, R.M.; Kandel, R.A.
Formation of a nucleus pulposus-cartilage endplate
construct in vitro. Biomaterials 2006, 27 (3), 397–405. 68.
Kim, Y.J.; Sah, R.L.; Doong, J.Y.; Grodzinsky, A.J.
Fluorometric assay of DNA in cartilage explants using
Hoechst 33258. Anal. Biochem. 1988, 174 (1), 168–176. 69.
Goldberg, R.L.; Kolibas, L.M. An improved method for
determining proteoglycans synthesized by chondrocytes in
culture. Connect. Tissue Res. 1990, 24 (3–4), 265–275. 70.
Woessner, J.F. Determination of hydroxyproline content in
connective tissues. In The Methodology of Connective Tissue
Research; Hall, D.A., Ed.; JoynsonBruvvers Limited: Oxford,
1976; 227–234. 71. Duda, G.N.; Kleemann, R.U.; Bluecher,
U.; Weiler, A. A new device to detect early cartilage
degeneration. Am. J. Sports Med. 2004, 32 (3), 693–698. 72.
Lyyra, T.; Jurvelin, J.; Pitkanen, P.; Vaatainen, U.;
Kiviranta, I. Indentation instrument for the measurement of
cartilage stiffness under arthroscopic control. Med. Eng
Phys. 1995, 17 (5), 395–399. 73. Newman, A.P. Articular
cartilage repair. Am. J. Sports Med. 1998, 26 (2), 309–324.
74. Stone, K.R.; Ayala, G.; Goldstein, J.; Hurst, R.;
Walgenbach, A.; Galili, U. Porcine cartilage transplants in
the cynomolgus monkey. III. Transplantation of
alpha-galactosidase-treated porcine cartilage.
Transplantation 1998, 65 (12), 1577–1583. 75. Martin, J.A.;
Buckwalter, J.A. Aging, articular cartilage chondrocyte
senescence and osteoarthritis. Biogerontology 2002, 3 (5),
257–264. T

76. Tran-Khanh, N.; Hoemann, C.D.; McKee, M.D.; Henderson,


J.E.; Buschmann, M.D. Aged bovine chondrocytes display a
diminished capacity to produce a collagen-rich,
mechanically functional cartilage extracellular matrix. J.
Orthop. Res. 2005, 23 (6), 1354–1362.

77. Homicz, M.R.; Chia, S.H.; Schumacher, B.L.; Masuda, K.;


Thonar, E.J.; Sah, R.L.; Watson, D. Human septal
chondrocyte redifferentiation in alginate, polyglycolic
acid scaffold, and monolayer culture. Laryngoscope 2003,
113 (1), 25–32.

78. Masuda, K.; Sah, R.L.; Hejna, M.J.; Thonar, E.J. A


novel two-step method for the formation of tissueengineered
cartilage by mature bovine chondrocytes: the
alginate-recovered-chondrocyte (ARC) method. J. Orthop.
Res. 2003, 21 (1), 139–148.

79. Minguell, J.J.; Erices, A.; Conget, P. Mesenchymal stem


cells. Exp. Biol. Med. (Maywood) 2001, 226 (6), 507–520.
80. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal,
R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti,
D.W.; Craig, S.; Marshak, D.R. Multilineage potential of
adult human mesenchymal stem cells. Science 1999, 284
(5411), 143–147.

81. Barry, F.P.; Murphy, J.M. Mesenchymal stem cells:


clinical applications and biological characterization. Int.
J. Biochem. Cell Biol. 2004, 36 (4), 568–584.

82. Tatebe, M.; Nakamura, R.; Kagami, H.; Okada, K.; Ueda,
M. Differentiation of transplanted mesenchymal stem cells
in a large osteochondral defect in rabbit. Cytotherapy
2005, 7 (6), 520–530.

83. Bensaid, W.; Triffitt, J.T.; Blanchat, C.; Oudina, K.;


Sedel, L.; Petite, H. A biodegradable fibrin scaffold for
mesenchymal stem cell transplantation. Biomaterials 2003,
24 (14), 2497–2502. 84. Dowthwaite, G.P.; Bishop, J.C.;
Redman, S.N.; Khan, I.M.; Rooney, P.; Evans, D.J.;
Haughton, L.; Bayram, Z.; Boyer, S.; Thomson, B.; Wolfe,
M.S.; Archer, C.W. The surface of articular cartilage
contains a progenitor cell population. J. Cell Sci. 2004,
117 (Pt 6), 889–897. 85. Murphy, J.M.; Fink, D.J.;
Hunziker, E.B.; Barry, F.P. Stem cell therapy in a caprine
model of osteoarthritis. Arthritis Rheum. 2003, 48 (12),
3464–3474. 86. Waldman, S.D.; Spiteri, C.G.; Grynpas, M.D.;
Pilliar, R.M.; Hong, J.; Kandel, R.A. Effect of
biomechanical conditioning on cartilaginous tissue
formation in vitro. J. Bone Joint Surg. Am. 2003, 85-A
(Suppl. 2), 101–105. 87. Darling, E.M.; Athanasiou, K.A.
Biomechanical strategies for articular cartilage
regeneration. Ann. Biomed. Eng. 2003, 31 (9), 1114–1124.
88. Mauck, R.L.; Nicoll, S.B.; Seyhan, S.L.; Ateshian,
G.A.; Hung, C.T. Synergistic action of growth factors and
dynamic loading for articular cartilage tissue engineering.
Tissue Eng. 2003, 9 (4), 597–611. 89. Waldman, S.D.;
Grynpas, M.D.; Pilliar, R.M.; Kandel, R.A. The use of
specific chondrocyte populations to modulate the properties
of tissue-engineered cartilage. J. Orthop. Res. 2003, 21
(1), 132–138. 90. Klein, T.J.; Schumacher, B.L.; Schmidt,
T.A.; Li, K.W.; Voegtline, M.S.; Masuda, K.; Thonar, E.J.;
Sah, R.L. Tissue engineering of stratified articular
cartilage from chondrocyte subpopulations. Osteoarthr.
Cartil. 2003, 11 (8), 595–602. 91. Kim, T.K.; Sharma, B.;
Williams, C.G.; Ruffner, M.A.; Malik, A.; McFarland, E.G.;
Elisseeff, J.H. Experimental model for cartilage tissue
engineering to regenerate the zonal organization of
articular cartilage. Osteoarthr. Cartil. 2003, 11 (9),
653–664.
Tissue Engineering of Cornea

1. Kruse, F.E. Stem cells and corneal epithelial


regeneration. Eye 1994, 8, 170–183. 2. Thoft, R.A.; Friend,
J. The X, Y, Z hypothesis of corneal epithelial
maintenance. Invest. Ophthalmol. Vis. Sci. 1983, 24 (10),
1442–1443. 3. Schermer, A.; Galvin, S.; Sun, T.-T.
Differentiationrelated expression of a major 64 K corneal
keratin in vivo and in culture suggests limbal location of
corneal epithelial stem cells. J. Cell Biol. 1986, 103 (1),
49–62. 4. Lauweryns, B.; Van Den Oord, J.J.; Missotten, L.
The transitional zone between limbus and peripheral cornea.
An immunohistochemical study. Invest. Ophthalmol. Vis. Sci.
1993, 34 (6), 1991–1999. 5. Kasper, M.; Stosiek, P.; Lane,
B. Cytokeratin and vimentin heterogeneity in human cornea.
Acta Histochem. 1992, 93 (2), 371–381. 6. Germain, L.;
Carrier, P.; Auger, F.A.; Salesse, C.; Gue´rin, S.L. Can we
produce a human corneal equivalent by tissue engineering?
Prog. Retin. Eye Res. 2000, 19 (5), 497–527. 7. Michel, M.;
Torok, N.; Godbout, M.-J.; Lussier, M.; Gaudreau, P.;
Royal, A.; Germain, L. Keratin 19 as a biochemical marker
of skin stem cells in vivo and in vitro: Keratin 19
expressing cells are differentially localized in function
of anatomic sites, and their number varies with donor age
and culture stage. J. Cell. Sci. 1996, 109 (Pt. 5),
1017–1028. 8. Fradette, J.; Germain, L.; Seshaiah, P.;
Coulombe, P.A. The type I keratin 19 possesses distinct and
contextdependent assembly properties. J. Biol. Chem. 1998,
273 (52), 35176–35184. 9. Rheinwald, J.G.; Green, H. Serial
cultivation of strains of human epidermal keratinocytes:
The formation of keratinizing colonies from single cells.
Cell 1975, 6, 331–343. 10. Lindberg, K.; Brown, M.E.;
Chaves, H.V.; Kenyon, K.R.; Rheinwald, J.G. In vitro
propagation of human ocular surface epithelial cells for
transplantation. Invest. Ophthalmol. Vis. Sci. 1993, 34
(9), 2672–2679. 11. Germain, L.; Auger, F.A.; Grandbois,
E.; Guignard, R.; Giasson, M.; Boisjoly, H.M.; Gue´rin,
S.L. Reconstructed human cornea produced in vitro by tissue
engineering. Pathobiology 1999, 67 (3), 140–147. 12.
Gaudreault, M.; Carrier, P.; Larouche, K.; Leclerc, S.;
Giasson, M.; Germain, L.; Guerin, S.L. Influence of sp1/sp3
expression on corneal epithelial cells proliferation and
differentiation properties in reconstructed tissues.
Invest. Ophthalmol. Vis. Sci. 2003, 44 (4), 1447–1457. 13.
Pellegrini, G.; Golisano, O.; Paterna, P.; Lambiase, A.;
Bonini, S.; Rama, P.; De Luca, M. Location and clonal
analysis of stem cells and their differentiated progeny in
the human ocular surface. J. Cell Biol. 1999, 145 (4),
769–782. 14. Joyce, N.C.; Navon, S.E.; Roy, S.; Zieske,
J.D. Expression of cell cycle-associated proteins in human
and rabbit corneal endothelium in situ. Invest. Ophthalmol.
Vis. Sci. 1996, 37 (8), 1566–1575. 15. Engelmann, K.;
Bednarz, J.; Bohnke, M. Endothelial cell transplantation
and growth behavior of the human corneal endothelium.
Ophthalmologe 1999, 96 (9), 555–562. 16. Rama, P.; Bonini,
S.; Lambiase, A.; Golisano, O.; Paterna, P.; De Luca, M.;
Pellegrini, G. Autologous fibrin-cultured limbal stem cells
permanently restore the corneal surface of patients with
total limbal stem cell deficiency. Transplantation 2001, 72
(9), 1478–1485. 17. Auger, F.A.; Lopez-Valle, C.A.;
Guignard, R.; Tremblay, N.; Noe¨l, B.; Goulet, F.; Germain,
L. Skin equivalent produced with human collagens. In vitro
Cell. Dev. Biol., Anim. 1995, 31, 432–439. 18. Bell, E.;
Ehrlich, H.P.; Buttle, D.J.; Nakatsuji, T. Living tissue
formed in vitro and accepted as skin-equivalent tissue of
full thickness. Science 1981, 211 (4486), 10452–11054. 19.
Zieske, J.D.; Mason, V.S.; Wasson, M.E.; Meunier, S.F.;
Nolte, C.J.M.; Fukai, N.; Olsen, B.R.; Parenteau, N.L.
Basement membrane assembly and differentiation of cultured
corneal cells: Importance of culture environment and
endothelial cell interaction. Exp. Cell Res. 1994, 214,
621–633. 20. Minami, Y.; Sugihara, H.; Oono, S.
Reconstruction of cornea in three-dimensional collagen gel
matrix culture. Invest. Ophthalmol. Vis. Sci. 1993, 34 (7),
2316–2324. 21. Michel, M.; L’Heureux, N.; Pouliot, R.; Xu,
W.; Auger, F.A.; Germain, L. Characterization of a new
tissueengineered human skin equivalent with hair. In vitro
Cell. Dev. Biol., Anim. 1999, 35 (6), 318–326. 22.
Prunie´ras, M.; Re´gnier, M.; Woodley, D. Methods for
cultivation of keratinocytes with an air–liquid interface.
J. Invest. Dermatol. 1983, 81 (1), 28s–33s. 23. Giasson,
C.J.; Carrier, P.; Germain, L. Collagen-based corneal
equivalents made with human cells as a model to study
cell–cell contact. Invest. Ophthalmol. Vis. Sci. 2003, 44.
E-Abstract 927. 24. Griffith, M.; Osborne, R.; Munger, R.;
Xiong, X.; Doillon, C.J.; Laycock, N.L.; Hakim, M.; Song,
Y.; Watsky, M.A. Functional human corneal equivalents
constructed from cell lines. Science 1999, 286 (5447),
2169–2172. 25. Griffith, M.; Hakim, M.; Shimmura, S.;
Watsky, M.A.; Li, F.; Carlsson, D.; Doillon, C.J.;
Nakamura, M.; Suuronen, E.; Shinozaki, N.; Nakata, K.;
Sheardown, H. Artificial human corneas: Scaffolds for
transplantation and host regeneration. Cornea 2002, 21
(Suppl. 7), S54–S61. 26. L’Heureux, N.; Paˆquet, S.;
Labbe´, R.; Germain, L.; Auger, F.A. A completely
biological tissue-engineered human blood vessel. FASEB J.
1998, 12, 47–56. 27. Rochon, M.-H.; Roberge, C.J.; Auger,
F.A.; Germain, L. Normal Epithelial Cells Modulate
Capillary Size by Secreting VEGF in a Human In Vitro
Angiogenesis Model Produced by the Self-Assembly Method. J
Invest. Dermatol. 121 (1): p. 1, abst 0002. In
International Investigative Dermatology; April 30–May 4
2003; Miami Beach, Florida. 28. Germain, L.; Carrier, P.;
Giasson, M.; Grandbois, E.; Auger, F.A.; Guerin, S.L. A new
method for in vitro production of human reconstructed
cornea by tissue engineering. Invest. Ophthalmol. Vis. Sci.
1999, 40, S329. 29. Carrier, P.; Deschambeault, A.; Talbot,
M.; Guerin, S.L.; Auger, F.A.; Germain, L. Expression of
extracellular matrix components in reconstructed human
cornea produced by tissue engineering. Invest. Ophthalmol.
Vis. Sci. 2002, 43. E-Abstract 4166. 30. Reim, M.; Kottek,
A.; Schrage, N. The cornea surface and wound healing. Prog.
Retin. Eye Res. 1997, 16 (2), 183–225. 31. Smith, R.J.;
Maloney, R.K. Diffuse lamellar keratitis. A new syndrome in
lamellar refractive surgery. Ophthalmology 1998, 105 (9),
1721–1726. 32. Plow, E.F.; Haas, T.A.; Zhang, L.; Loftus,
J.; Smith, J.W. Ligand binding to integrins. J. Biol. Chem.
2000, 275 (29), 21785–21788. 33. Giancotti, F.G. Complexity
and specificity of integrin signalling. Nat. Cell Biol.
2000, 2 (1), E13–E14. 34. Elner, S.G.; Elner, V.M. The
integrins superfamily and the eye. Invest. Ophthalmol. Vis.
Sci. 1996, 37 (5), 696–701. 35. Zieske, J.D. Extracellular
matrix and wound healing. Curr. Opin. Ophthalmol. 2001, 12
(4), 237–241. 36. Ahmadi, A.J.; Jakobiec, F.A. Corneal
wound healing: Cytokines and extracellular matrix proteins.
Int. Ophthalmol. Clin. 2002, 42 (3), 13–22. 37. Stepp,
M.A.; Zhu, L. Upregulation of a9 integrin and tenascin
during epithelial regeneration after debridement in the
cornea. J. Histochem. Cytochem. 1997, 45 (2), 189–201. 38.
Kenney, M.C.; Nesburn, A.B.; Burgeson, R.E.; Butkowski,
R.J.; Ljubimov, A.V. Abnormalities of the extracellular
matrix in keratoconus corneas. Cornea 1997, 16 (3),
345–351. 39. Tuori, A.; Virtanen, I.; Aine, E.; Uusitalo,
H. The expression of tenascin and fibronectin in
keratoconus, scarred and normal human cornea. Graefe Arch.
Clin. Exp. Ophthalmol. 1997, 235 (4), 222–229. 40. Saika,
S.; Kobata, S.; Hashizume, N.; Okada, Y.; Yamanaka, O.
Epithelial basement membrane in alkaliburned corneas in
rats. Immunohistochemical study. Cornea 1993, 12 (5),
383–390. 41. Latvala, T.; Barraquer-Coll, C.; Tervo, K.;
Tervo, T. Corneal wound healing and nerve morphology after
excimer laser in situ keratomileusis in human eyes. J.
Refract. Surg. 1996, 12 (6), 677–683. 42. Anderson, J.A.;
Binder, P.S.; Rock, M.E.; Vrabec, M.P. Human excimer laser
keratectomy. Immunohistochemical analysis of healing. Arch.
Ophthalmol. 1996 , 114 (1), 54–60. 43. Winter, M.;
Behrendt, S.; Binder, P.S.; Duncker, G.I. Ultrastructural
and immunohistochemical findings after linear excimer laser
keratectomy. J. Refract. Surg. 1997, 13 (1), 60–68. 44.
Schneider, A.I.; Maier-Reif, K.; Graeve, T. Constructing an
in vitro cornea from cultures of the three specific corneal
cell types. In vitro Cell. Dev. Biol., Anim. 1999, 35 (9),
515–526. 45. Chen, K.H.; Azar, D.; Joyce, N.C.
Transplantation of adult human corneal endothelium ex vivo:
A morphologic study. Cornea 2001, 20 (7), 731–737. 46.
Blake, D.A.; Yu, H.; Young, D.L.; Caldwell, D.R. Matrix
stimulates the proliferation of human corneal T endothelial
cells in culture. Invest. Ophthalmol. Vis. Sci. 1997, 38,
1119–1129.

47. Engelmann, K.; Firedl, P. Optimization of culture


conditions for human corneal endothelial cells. In vitro
Cell. Dev. Biol., Anim. 1989, 25 (11), 1065–1072.

48. Pfister, R.R. Corneal stem cell disease: Concepts,


categorization, and treatment by autoand
homotransplantation of limbal stem cells. CLAO J. 1994, 20
(1), 64–72. 49. Holland, E.J.; Schwartz, G.S. The evolution
of epithelial transplantation for severe ocular surface
disease and a proposed classification system. Cornea 1996,
15 (6), 549–556. 50. Pellegrini, G.; Traverso, C.E.;
Franzi, A.T.; Zingirian, M.; Cancedda, R.; De Luca, M.
Long-term restoration of damaged corneal surfaces with
autologous cultivated corneal epithelium. Lancet 1997, 349
(9057), 990–993.
Tissue Engineering of the Esophagus

1. Aprodu, S.G.; Gavrilescu, S.; Savu, B.; Munteanu, V.;


Vlad, A. Indications for esophageal replacement in
children. Rev. Med. Chir. Soc. Med. Nat. Iasi 2004, 108
(4), 805–808.

2. Orringer, M.B.; Marshall, B.; Iannettoni, M.D.


Transhiatal esophagectomy: clinical experience and
refinements. Ann. Surg. 1999, 230 (3), 392–400; discussion
400–403.

3. Sauvanet, A.; Mariette, C.; Thomas, P.; Lozac’h, P.;


Segol, P.; Tiret, E.; Delpero, J.R.; Collet, D.; Leborgne,
J.; Pradere, B.; Bourgeon, A.; Triboulet, J.P. Mortality
and morbidity after resection for adenocarcinoma of the
gastroesophageal junction: predictive factors. J. Am. Coll.
Surg. 2005, 201 (2), 253–262.

4. Orringer, M.B.; Stirling, M.C. Cervical esophagogastric


anastomosis for benign disease. Functional results. J.
Thorac. Cardiovasc. Surg. 1988, 96 (6), 887–893.

5. Yildirim, S.; Koksal, H.; Celayir, F.; Erdem, L.; Oner,


M.; Baykan, A. Colonic interposition vs. gastric pull-up
after total esophagectomy. J. Gastrointest. Surg. 2004, 8
(6), 675–678.

6. Dimick, J.B.; Wainess, R.M.; Upchurch, G.R., Jr.;


Iannettoni, M.D.; Orringer, M.B. National trends in
outcomes for esophageal resection. Ann. Thorac. Surg. 2005,
79 (1), 212–216; discussion 217–218.

7. Iannettoni, M.D.; Whyte, R.I.; Orringer, M.B.


Catastrophic complications of the cervical esophagogastric
anastomosis. J. Thorac. Cardiovasc. Surg. 1995, 110 (5),
1493–1500; discussion 1500–1501. 8. Reavis, K.M.; Chang,
E.Y.; Hunter, J.G.; Jobe, B.A. Utilization of the delay
phenomenon improves blood flow and reduces collagen
deposition in esophagogastric anastomoses. Ann. Surg. 2005,
241 (5), 736–745; discussion 745–747. 9. Chattopadhyay,
T.K.; Shad, S.K.; Kumar, A. Intragastric bile acid and
symptoms in patients with an intrathoracic stomach after
oesophagectomy. Br. J. Surg. 1993, 80 (3), 371–373. 10.
Koh, P.; Turnbull, G.; Attia, E.; LeBrun, P.; Casson, A.G.
Functional assessment of the cervical esophagus after
gastric transposition and cervical esophagogastrostomy.
Eur. J. Cardiothorac. Surg. 2004, 25 (4), 480–485. 11.
Bhasin, D.K.; Sharma, B.C.; Gupta, N.M.; Sinha, S.K.;
Singh, K. Endoscopic dilation for treatment of anastomotic
leaks following transhiatal esophagectomy. Endoscopy 2000,
32 (6), 469–471. 12. Orringer, M.B.; Marshall, B.;
Iannettoni, M.D. Eliminating the cervical esophagogastric
anastomotic leak with a side-to-side stapled anastomosis.
J. Thorac. Cardiovasc. Surg. 2000, 119 (2), 277–288. 13.
Rizk, N.P.; Bach, P.B.; Schrag, D.; Bains, M.S.; Turnbull,
A.D.; Karpeh, M.; Brennan, M.F.; Rusch, V.W. The impact of
complications on outcomes after resection for esophageal
and gastroesophageal junction carcinoma. J. Am. Coll. Surg.
2004, 198 (1), 42–50. 14. Inci, I.; Ozcelik, C.; Nizam, O.;
Balci, A.E.; Eren, N.; Ozgen, G. Traumatic oesophageal
perforation. Scand. Cardiovasc. J. 1997, 31 (2), 97–100.
15. Dunne, D.P.; Paterson, W.G. Acid-induced esophageal
shortening in humans: a cause of hiatus hernia? Can. J.
Gastroenterol. 2000 , 14 (10), 847–850. 16. Huang, J.Q.;
Hunt, R.H. pH, healing rate, and symptom relief in patients
with GERD. Yale J. Biol. Med. 1999, 72 (2–3), 181–194. 17.
Paterson, W.G. Role of mast cell-derived mediators in
acid-induced shortening of the esophagus. Am. J. Physiol.
1998, 274 (2 Pt 1), G385–G388. 18. Sampliner, R.E.
Reduction of acid exposure and regression of Barrett’s
esophagus. Dig. Dis. 2000, 18 (4), 203–207. 19. Zhang, X.;
Geboes, K.; Depoortere, I.; Tack, J.; Janssens, J.; Sifrim,
D. Effect of repeated cycles of acute esophagitis and
healing on esophageal peristalsis, tone, and length. Am. J.
Physiol. Gastrointest. Liver Physiol. 2005, 288 (6),
G1339–G1346. 20. Sampliner, R.E. Prevention of
adenocarcinoma by reversing Barrett’s esophagus with
mucosal ablation. World J. Surg. 2003, 27 (9), 1026–1029.
21. Stein, H.J.; Feith, M.; Siewert, J.R. Malignant
degeneration of Barrett’s esophagus: clinical point of
view. Recent Results Cancer Res. 2000, 155, 42–53. 22.
Giovannini, M.; Bernardini, D.; Moutardier, V.; Monges, G.;
Houvenaeghel, G.; Seitz, J.F.; Derlpero, J.R. Endoscopic
mucosal resection (EMR): results and prognostic factors in
21 patients. Endoscopy 1999, 31 (9), 698–701. 23.
Giovannini, M.; Bories, E.; Pesenti, C.; Moutardier, V.;
Monges, G.; Danisi, C.; Lelong, B.; Delpero, J.R.
Circumferential endoscopic mucosal resection in Barrett’s
esophagus with high-grade intraepithelial neoplasia or
mucosal cancer. Preliminary results in 21 patients.
Endoscopy 2004, 36 (9), 782–787. 24. Katada, C.; Muto, M.;
Manabe, T.; Boku, N.; Ohtsu, A.; Yoshida, S. Esophageal
stenosis after endoscopic mucosal resection of superficial
esophageal lesions. Gastrointest. Endosc. 2003, 57 (2),
165–169. 25. Fitzgerald, R.C. Ablative mucosectomy is the
procedure of choice to prevent Barrett’s cancer. Gut 2003,
52 (1), 16–17. 26. Luman, W.; Lessels, A.M.; Palmer, K.R.
Failure of Nd-YAG photocoagulation therapy as treatment for
Barrett’s oesophagus—a pilot study. Eur. J. Gastroenterol.
Hepatol. 1996, 8 (7), 627–630. 27. Sharma, P.;
Bhattacharyya, A.; Garewal, H.S.; Sampliner, R.E.
Durability of new squamous epithelium after endoscopic
reversal of Barrett’s esophagus. Gastrointest. Endosc.
1999, 50 (2), 159–164. 28. Chen, M.K.; Beierle, E.A. Animal
models for intestinal tissue engineering. Biomaterials
2004, 25 (9), 1675–1681. 29. Kobold, E.E.; Thal, A.P. A
simple method for the management of experimental wounds of
the duodenum. Surg. Gynecol. Obstet. 1963, 116, 340–344.
30. Badylak, S.F.; Vorp, D.A.; Spievack, A.R.; SimmonsByrd,
A.; Hanke, J.; Freytes, D.O.; Thapa, A.; Gilbert, T.W.;
Nieponice, A. Esophageal reconstruction with ECM and muscle
tissue in a dog model. J. Surg. Res. 2005, 128, 87–97. 31.
Grikscheit, T.; Ochoa, E.R.; Srinivasan, A.; Gaissert, H.;
Vacanti, J.P. Tissue-engineered esophagus: experimental
substitution by onlay patch or interposition. J. Thorac.
Cardiovasc. Surg. 2003, 126 (2), 537–544. 32. Grikscheit,
T.; Srinivasan, A.; Vacanti, J.P. Tissueengineered stomach:
a preliminary report of a versatile in vivo model with
therapeutic potential. J. Pediatr. Surg. 2003, 38 (9),
1305–1309. 33. Grikscheit, T.C.; Ochoa, E.R.; Ramsanahie,
A.; Alsberg, E.; Mooney, D.; Whang, E.E.; Vacanti, J.P.
Tissueengineered large intestine resembles native colon
with appropriate in vitro physiology and architecture. Ann.
Surg. 2003, 238 (1), 35–41. 34. Kaihara, S.; Kim, S.S.;
Benvenuto, M.; Choi, R.; Kim, B.S.; Mooney, D.; Tanaka, K.;
Vacanti, J.P. Successful anastomosis between
tissue-engineered intestine and native small bowel.
Transplantation 1999, 67 (2), 241–245. 35. Rashid, S.T.;
Salacinski, H.J.; Hamilton, G.; Seifalian, A.M. The use of
animal models in developing the discipline of
cardiovascular tissue engineering: a review. Biomaterials
2004, 25 (9), 1627–1637. 36. Forse, R.A.; MacDonald, P.H.;
Mercer, C.D. Anastomotic and regional blood flow following
esophagogastrectomy in an opossum model. J. Invest. Surg.
1999, 12 (1), 45–52. 37. Zijlstra, F.G.; Hynna-Liepert,
T.T.; Dinda, P.K.; Beck, I.T.; Paterson, W.G.
Gastrointestinal blood flow in the opossum with special
reference to the esophagus. Can. J. Physiol. Pharmacol.
1990, 68 (9), 1221–1225. 38. Beckstead, B.L.; Pan, S.;
Bhrany, A.D.; Bratt-Leal, A.M.; Ratner, B.D.; Giachelli,
C.M. Esophageal epithelial cell interaction with synthetic
and natural scaffolds for tissue engineering. Biomaterials
2005, 26 (31), 6217–6228. 39. Zhu, Y.; Chan-Park, M.B.;
Chian, K.S. The growth improvement of porcine esophageal
smooth muscle cells on collagen grafted poly
(dl-lactide-co-glycolide) membrane. J. Biomed. Mater. Res.
B Appl. Biomater. 2005, 75B, 193–199. 40. Zhu, Y.; Chian,
K.S.; Chan-Park, M.B.; Mhaisalkar, P.S.; Ratner, B.D.
Protein bonding on biodegradable poly(
L-lactide-co-caprolactone) membrane for esophageal tissue
engineering. Biomaterials 2006, 27 (1), 68–78. 41.
Takimoto, Y.; Okumura, N.; Nakamura, T.; Natsume, T.;
Shimizu, Y. Long-term follow-up of the experimental
replacement of the esophagus with a collagensilicone
composite tube. Asaio J. 1993, 39 (3), M736–M739. 42.
Takimoto, Y.; Nakamura, T.; Teramachi, M.; Kiyotani, T.;
Shimizu, Y. Replacement of long segments of the esophagus
with a collagen-silicone composite tube. Asaio J. 1995, 41
(3), M605–M608. 43. Takimoto, Y.; Nakamura, T.; Yamamoto,
Y.; Kiyotani, T.; Teramachi, M.; Shimizu, Y. The
experimental replacement of a cervical esophageal segment
with an artificial prosthesis with the use of collagen
matrix and a silicone stent. J. Thorac. Cardiovasc. Surg.
1998, 116 (1), 98–106. 44. Yamamoto, Y.; Nakamura, T.;
Shimizu, Y.; Matsumoto, K.; Takimoto, Y.; Kiyotani, T.;
Sekine, T.; Ueda, H.; Liu, Y.; Tamura, N. Intrathoracic
esophageal replacement in the dog with the use of an
artificial esophagus composed of a collagen sponge with a
doublelayered silicone tube. J. Thorac. Cardiovasc. Surg.
1999, 118 (2), 276–286. 45. Yamamoto, Y.; Nakamura, T.;
Shimizu, Y.; Takimoto, Y.; Matsumoto, K.; Kiyotani, T.; Yu,
L.; Ueda, H.; Sekine, T.; Tamura, N. Experimental
replacement of the thoracic esophagus with a bioabsorbable
collagen sponge scaffold supported by a silicone stent in
dogs. Asaio J. 1999, 45 (4), 311–316. 46. Yamamoto, Y.;
Nakamura, T.; Shimizu, Y.; Matsumoto, K.; Takimoto, Y.;
Liu, Y.; Ueda, H.; Sekine, T.; Tamura, N. Intrathoracic
esophageal replacement with a collagen sponge—silicone
double layer tube: evaluation of omental-pedicle wrapping
and prolonged placement of an inner stent. Asaio J. 2000,
46 (6), 734–739. 47. Takimoto, Y.; Teramachi, M.; Okumura,
N.; Nakamura, T.; Shimizu, Y. Relationship between stenting
time and regeneration of neoesophageal submucosal tissue.
Asaio J. 1994, 40 (3), M793–M797. 48. Hayashi, K.; Ando,
N.; Ozawa, S.; Kitagawa, Y.; Miki, H.; Sato, M.; Kitajima,
M. A neo-esophagus reconstructed by cultured human
esophageal epithelial cells, smooth muscle cells,
fibroblasts, and collagen. Asaio J. 2004, 50 (3), 261–266.
49. Akizuki, T.; Oda, S.; Komaki, M.; Tsuchioka, H.;
Kawakatsu, N.; Kikuchi, A.; Yamato, M.; Okano, T.;
Ishikawa, I. Application of periodontal ligament cell sheet
for periodontal regeneration: a pilot study in beagle dogs.
J. Periodontal Res. 2005, 40 (3), 245–251. 50. Nishida, K.;
Yamato, M.; Hayashida, Y.; Watanabe, K.; Yamamoto, K.;
Adachi, E.; Nagai, S.; Kikuchi, A.; Maeda, N.; Watanabe,
H.; Okano, T.; Tano, Y. Corneal reconstruction with
tissue-engineered cell sheets T composed of autologous oral
mucosal epithelium. N. Engl. J. Med. 2004, 351 (12),
1187–1196.
51. Shiroyanagi, Y.; Yamato, M.; Yamazaki, Y.; Toma, H.;
Okano, T. Urothelium regeneration using viable cultured
urothelial cell sheets grafted on demucosalized gastric
flaps. BJU Int. 2004, 93 (7), 1069–1075.

52. Yang, J.; Yamato, M.; Kohno, C.; Nishimoto, A.; Sekine,
H.; Fukai, F.; Okano, T. Cell sheet engineering: recreating
tissues without biodegradable scaffolds. Biomaterials 2005,
26 (33), 6415–6422. 53. Badylak, S.; Meurling, S.; Chen,
M.; Spievack, A.; Simmons-Byrd, A. Resorbable bioscaffold
for esophageal repair in a dog model. J. Pediatr. Surg.
2000, 35 (7), 1097–1103. 54. Brown, B.; Lindberg, K.;
Reing, J.; Stolz, D.B.; Badylak, S.F. The basement membrane
component of biologic scaffolds derived from extracellular
matrix. Tissue Eng. 2006, 12 (3), 519–526.
Tissue Engineering, Fat

Fig. 5 Comparison of cell and tissue development in two

and three-dimensional cell culture systems. Scanning

electron micrograph images of 3T3-L1 cells grown in vitro

in two-dimensional cell culture or three-dimensional tissue

engineered constructs. Cells in two-dimensional monolayers

are loosely connected to each other and ECM sheets are

preferentially deposited on the cell surface. In contrast,

three-dimensional differentiated adipocytes are situated

within a coherent environment featuring three-dimensional

cell–cell and cell–ECM interactions. Bar=20 mm. (From

Ref. [5].) 8. Gregoire, F.M.; Smas, C.M.; Sul, H.S.


Understanding adipocyte differentiation. Physiol. Rev.
1998, 78 (3), 783–809. 9. Rosen, E.D.; Spiegelman, B.M.
Molecular regulation of adipogenesis. Annu. Rev. Cell Dev.
Biol. 2000, 16 (1), 145–171. 10. Smas, C.M.; Sul, H.S.
Control of adipocyte differentiation. Biochem. J. 1995, 309
(3), 697–710. 11. Boudreau, N.J.; Jones, P.L. Extracellular
matrix and integrin signalling: the shape of things to
come. Biochem. J. 1999, 339 (3), 481–488. 12. Guerre-Millo,
M. Adipose tissue hormones. J. Endocrinol. Invest. 2002, 25
(10), 855–861. 13. Sugihara, H.; Yonemitsu, N.; Miyabara,
S.; Toda, S. Proliferation of unilocular fat cells in the
primary culture. J. Lipid Res. 1987, 28 (9), 1038–1045. 14.
Tholpady, S.S.; Aojanepong, C.; Llull, R.; Jeong, J.H.;
Mason, A.C.; Futrell, J.W.; Ogle, R.C.; Katz, A.J. The
cellular plasticity of human adipocytes. Ann. Plast. Surg.
2005, 54 (6), 651–656. 15. Kawaguchi, N.; Toriyama, K.;
Nicodemou-Lena, E.; Inou, K.; Torii, S.; Kitagawa, Y.
Reconstituted basement membrane potentiates in vivo
adipogenesis of 3T3-F442A cells. Cytotechnology 1999, 31
(3), 215–220. 16. Fischbach, C.; Seufert, J.; Staiger, H.;
Hacker, M.; Neubauer, M.; Gopferich, A.; Blunk, T.
Three-dimensional in vitro model of adipogenesis:
comparison of culture conditions. Tissue Eng. 2004, 10
(1–2), 215–229. 17. Kang, X.; Xie, Y.; Kniss, D.A. Adipose
tissue model using three-dimensional cultivation of
preadipocytes seeded onto fibrous polymer scaffolds. Tissue
Eng. 2005, 11 (3–4), 458–468. 18. Patrick, C.W., Jr.;
Chauvin, P.B.; Hobley, J.; Reece, G.P. Preadipocyte seeded
PLGA scaffolds for adipose tissue engineering. Tissue Eng.
1999, 5 (2), 139–151. 19. Patrick, C.W., Jr.; Zheng, B.;
Johnston, C.; Reece, G.P. Long-term implantation of
preadipocyte-seeded PLGA scaffolds. Tissue Eng. 2002, 8
(2), 283–293. 20. von Heimburg, D.; Zachariah, S.; Low, A.;
Pallua, N. Influence of different biodegradable carriers on
the in vivo behavior of human adipose precursor cells.
Plast. Reconstr. Surg. 2001, 108 (2), 411–420. 21.
Halbleib, M.; Skurk, T.; de Luca, C.; von Heimburg, D.;
Hauner, H. Tissue engineering of white adipose tissue using
hyaluronic acid-based scaffolds. I: in vitro
differentiation of human adipocyte precursor cells on
scaffolds. Biomaterials 2003, 24 (18), 3125–3132. 22. von
Heimburg, D.; Kuberka, M.; Rendchen, R.; Hemmrich, K.; Rau,
G.; Pallua, N. Preadipocyteloaded collagen scaffolds with
enlarged pore size for improved soft tissue engineering.
Int. J. Artif. Organs 2003, 26 (12), 1064–1076. 23. Kimura,
Y.; Ozeki, M.; Inamoto, T.; Tabata, Y. Adipose tissue
engineering based on human preadipocytes combined with
gelatin microspheres containing basic fibroblast growth
factor. Biomaterials 2003, 24 (14), 2513–2521. 24. Kral,
J.G.; Crandall, D.L. Development of a human adipocyte
synthetic polymer scaffold. Plast. Reconstr. Surg. 1999,
104 (6), 1732–1738. 25. Halberstadt, C.; Austin, C.;
Rowley, J.; Culberson, C.; Loebsack, A.; Wyatt, S.;
Coleman, S.; Blacksten, L.; Burg, K.; Mooney, D.; Holder,
W., Jr. A hydrogel material for plastic and reconstructive
applications injected into the subcutaneous space of a
sheep. Tissue Eng. 2002, 8 (2), 309–319. 26. Cho, S.W.;
Kim, S.S.; Won Rhie, J.; Mi Cho, H.; Yong Choi, C.; Kim,
B.S. Engineering of volume-stable adipose tissues.
Biomaterials 2005, 26 (17), 3577–3585. 27. Borges, J.;
Mueller, M.C.; Padron, N.T.; Tegtmeier, F.; Lang, E.M.;
Stark, G.B. Engineered adipose tissue supplied by
functional microvessels. Tissue Eng. 2003, 9 (6),
1263–1270. 28. Gimble, J.M. Adipose tissue-derived
therapeutics. Expert. Opin. Biol. Ther. 2003, 3 (5),
705–713. 29. Katz, A.J.; Llull, R.; Hedrick, M.H.; Futrell,
J.W. Emerging approaches to the tissue engineering of fat.
Clin. Plast. Surg. 1999, 26 (4), 587–603. 30. Rodriguez,
A.M.; Elabd, C.; Amri, E.Z.; Ailhaud, G.; Dani, C. The
human adipose tissue is a source of multipotent stem cells.
Biochim. 2005, 87 (1), 125–128. 31. Zuk, P.A.; Zhu, M.;
Mizuno, H.; Huang, J.; Futrell, J.W.; Katz, A.J.; Benhaim,
P.; Lorenz, H.P.; Hedrick, M.H. Multilineage cells from
human adipose tissue: implications for cell-based
therapies. Tissue Eng. 2001, 7 (2), 211–228. 32. Kirkland,
J.L.; Hollenberg, C.H.; Gillon, W.S. Age, anatomic site,
and the replication and differentiation of adipocyte
precursors. Am. J. Physiol. Cell Physiol. 1990, 258 (2),
C206–C210. 33. Tchkonia, T.; Tchoukalova, Y.D.; Giorgadze,
N.; Pirtskhalava, T.; Karagiannides, I.; Forse, R.A.; Koo,
A.; Stevenson, M.; Chinnappan, D.; Cartwright, A.; Jensen,
M.D.; Kirkland, J.L. Abundance of two human preadipocyte
subtypes with distinct capacities for replication,
adipogenesis, and apoptosis varies among fat depots. Am. J.
Physiol. Endocrinol. Metab. 2005, 288 (1), E267–E277. 34.
Neubauer, M.; Hacker, M.; Bauer-Kreisel, P.; Weiser, B.;
Fischbach, C.; Schulz, M.B.; Goepferich, A.; Blunk, T.
Adipose tissue engineering based on mesenchymal stem cells
and basic fibroblast growth factor in vitro. Tissue Eng. in
press. 35. Hong, L.; Peptan, I.; Clark, P.; Mao, J.J. Ex
vivo adipose tissue engineering by human marrow stromal
cell seeded gelatin sponge. Ann. Biomed. Eng. 2005, 33 (4),
511–517. 36. Bruder, S.P.; Jaiswal, N.; Haynesworth, S.E.
Growth kinetics, self-renewal, and the osteogenic potential
of purified human mesenchymal stem cells during extensive
subcultivation and following cryopreservation. J. Cell
Biochem. 1997, 64 (2), 278–294. 37. Gregory, C.A.; Prockop,
D.J.; Spees, J.L. Nonhematopoietic bone marrow stem cells:
molecular control of expansion and differentiation. Exp.
Cell Res. 2005, 306 (2), 330–335. 38. Dani, C.; Smith,
A.G.; Dessolin, S.; Leroy, P.; Staccini, L.; Villageois,
P.; Darimont, C.; Ailhaud, G. Differentiation of embryonic
stem cells into adipocytes in vitro. J. Cell Sci. 1997, 110
(11), 1279–1285. T

39. Dani, C. Embryonic stem cell-derived adipogenesis.


Cells Tissues Organs 1999, 165 (3–4), 173–180.

40. Hacker, M.; Tessmar, J.; Neubauer, M.; Blaimer, A.;


Blunk, T.; Gopferich, A.; Schulz, M.B. Towards biomimetic
scaffolds: anhydrous scaffold fabrication from
biodegradable amine-reactive diblock copolymers.
Biomaterials 2003, 24 (24), 4459–4473.

41. Ibrahimi, A.; Bonino, F.; Bardon, S.; Ailhaud, G.;


Dani, C. Essential role of collagens for terminal
differentiation of preadipocytes. Biochem. Biophys. Res.
Commun. 1992, 187 (3), 1314–1322.

42. Kimura, Y.; Ozeki, M.; Inamoto, T.; Tabata, Y. Time


course of de novo adipogenesis in matrigel by gelatin
microspheres incorporating basic fibroblast growth factor.
Tissue Eng. 2002, 8 (4), 603–613.

43. Kawaguchi, N.; Toriyama, K.; Nicodemou-Lena, E.; Inou,


K.; Torii, S.; Kitagawa, Y. De novo adipogenesis in mice at
the site of injection of basement membrane and basic
fibroblast growth factor. Proc. Natl. Acad. Sci. USA 1998,
95 (3), 1062–1066.

44. Schoeller, T.; Lille, S.; Wechselberger, G.; Otto, A.;


Mowlavi, A.; Piza-Katzer, H. Histomorphologic and
volumetric analysis of implanted autologous preadipocyte
cultures suspended in fibrin glue: a potential new source
for tissue augmentation. Aesthetic Plast. Surg. 2001, 25
(1), 57–63.

45. Helgerson, S.L.; Seelich, T.; di Orio, J.P.; Bittner,


K.; Spaethe, R. Fibrin. In Encyclopedia of Biomaterials and
Biomedical Engineering; Wnek, G., Bowlin, G., Eds.; Marcel
Dekker, Inc.: New York, 2004; 1, 603–610.

46. Meinhart, J.; Fussenegger, M.; Hobling, W.


Stabilization of fibrin-chondrocyte constructs for cartilage
reconstruction. Ann. Plast. Surg. 1999, 42 (6), 673–678.

47. West, D.C.; Kumar, S. Hyaluronan and angiogenesis. Ciba


Found. Symp. 1989, 143, 187–201.

48. Desvergne, B.; Wahli, W. Peroxisome


proliferatoractivated receptors: nuclear control of
metabolism. Endocr. Rev. 1999, 20 (5), 649–688.

49. Neubauer, M.; Fischbach, C.; Bauer-Kreisel, P.; Lieb,


E.; Hacker, M.; Tessmar, J.; Schulz, M.B.; Goepferich, A.;
Blunk, T. Basic fibroblast growth factor enhances PPARgamma
ligand-induced adipogenesis of mesenchymal stem cells. FEBS
Lett. 2004, 577 (1–2), 277–283.

50. Yuksel, E.; Weinfeld, A.B.; Cleek, R.; Wamsley, S.;


Jensen, J.; Boutros, S.; Waugh, J.M.; Shenaq, S.M.; Spira,
M. Increased free fat-graft survival with the long-term,
local delivery of insulin, insulin-like growth factor-I,
and basic fibroblast growth factor by PLGA/ PEG
microspheres. Plast. Reconstr. Surg. 2000, 105 (5),
1712–1720.

51. Yuksel, E.; Weinfeld, A.B.; Cleek, R.; Waugh, J.M.;


Jensen, J.; Boutros, S.; Shenaq, S.M.; Spira, M. De novo
adipose tissue generation through long-term, local delivery
of insulin and insulinlike growth factor-1 by PLGA/PEG
microspheres in an in vivo rat model: a novel concept and
capability. Plast. Reconstr. Surg. 2000, 105 (5),
1721–1729. 52. Walton, R.L.; Beahm, E.K.; Wu, L. De novo
adipose formation in a vascularized engineered construct.
Microsurgery 2004, 24 (5), 378–384. 53. Hausman, G.J.;
Richardson, R.L. Adipose tissue angiogenesis. J. Anim. Sci.
2004, 82 (3), 925–934. 54. Rupnick, M.A.; Panigrahy, D.;
Zhang, C.Y.; Dallabrida, S.M.; Lowell, B.B.; Langer, R.;
Folkman, M.J. Adipose tissue mass can be regulated through
the vasculature. Proc. Natl. Acad. Sci. USA 2002, 99 (16),
10730–10735. 55. Varzaneh, F.E.; Shillabeer, G.; Wong,
K.L.; Lau, D.C.W. Extracellular matrix components secreted
by microvascular endothelial cells stimulate preadipocyte
differentiation in vitro. Metabolism 1994, 43 (7), 906–912.
56. Hutley, L.J.; Herington, A.C.; Shurety, W.; Cheung, C.;
Vesey, D.A.; Cameron, D.P.; Prins, J.B. Human adipose
tissue endothelial cells promote preadipocyte
proliferation. Am. J. Physiol. Endocrinol. Metab. 2001, 281
(5), E1037–E1044. 57. Hutley, L.; Shurety, W.; Newell, F.;
McGeary, R.; Pelton, N.; Grant, J.; Herington, A.; Cameron,
D.; Whitehead, J.; Prins, J. Fibroblast growth factor 1: a
key regulator of human adipogenesis. Diabetes 2004, 53
(12), 3097–3106. 58. Barleon, B.; Borges, J.; Lauchart, M.;
Herzog, C.; Tegtmeier, F.; Walgenbach, K.J.; Andree, C.;
Bittner, K.; Stark, G.B.; Marme, D. Angiogenesis in fat
tissue: characterization of the molecular and cellular
interactions between preadipocytes, adipocytes, and
endothelial cells in vitro. In Cells Tissues Organs 2000,
2nd BioValley Tissue Engineering Symposium, Freiburg,
Germany, Nov 25–27, 1999; Vol. 166, suppl. 1, Abstract
B403, 103. . 59. Dallabrida, S.M.; Zurakowski, D.; Shih,
S.C.; Smith, L.E.; Folkman, J.; Moulton, K.S.; Rupnick,
M.A. Adipose tissue growth and regression are regulated by
angiopoietin-1. Biochem. Biophys. Res. Commun. 2003, 311
(3), 563–571. 60. Dolderer, J.H.; Findlay, M.W.;
Cooper-White, J.; Thompson, E.W.; Trost, N.; Hennessy, O.;
Penington, A.; Morrison, W.A. Making tissue engineering for
breast reconstruction a reality. In International
Conference ‘‘Strategies of Tissue Engineering,’’ Wuerzburg,
Germany, Jun 2004. Cytotechnology 2004, 6 (3) Abstract
O-36, 263–263.
Tissue Engineering of Heart Valves

7. McGrath, L.B.; et al. Perioperative events in patients


with failed mechanical and bioprosthetic valves. Ann.
Thorac. Surg. 1995, 60 (2 Suppl), S475–S478.

8. Bernal, J.M.; et al. Valve-related complications with


the Hancock I porcine bioprosthesis. A twelveto
fourteen-year follow-up study. J. Thorac. Cardiovasc. Surg.
1991, 101 (5), 871–880.

9. Burdon, T.A.; et al. Durability of porcine valves at


fifteen years in a representative North American patient
population. J. Thorac. Cardiovasc. Surg. 1992, 103 (2),
238–251. Discussion 251–252.

10. Grunkemeier, G.L.; Bodnar, E. Comparative assessment of


bioprosthesis durability in the aortic position. J. Heart
Valve Dis. 1995, 4 (1), 49–55.

11. O’Brien, M.F.; et al. The homograft aortic valve: A


29-year, 99.3% follow up of 1,022 valve replacements. J.
Heart Valve Dis. 2001, 10 (3), 334–344. Discussion 335.

12. Sabet, H.Y.; et al. Congenitally bicuspid aortic


valves: A surgical pathology study of 542 cases (1991
through 1996) and a literature review of 2,715 additional
cases. Mayo Clin. Proc. 1999, 74 (1), 14–26.

13. Dunn, J.M. Porcine valve durability in children. Ann.


Thorac. Surg. 1981, 32, 357–368.

14. Vesely, I.; Barber, J.E.; Ratliff, N.B. Tissue damage


and calcification may be independent mechanisms of
bioprosthetic heart valve failure. J. Heart Valve Dis.
2001, 10 (4), 471–477.

15. Sacks, M.S.; Schoen, F.J. Collagen fiber disruption


occurs independent of calcification in clinically explanted
bioprosthetic heart valves. J. Biomed. Mater. Res. 2002, 62
(3), 359–371.

16. Ishihara, T.; et al. Structure and classification of


cuspal tears and perforations in porcine bioprosthetic
cardiac valves implanted in patients. Am. J. Cardiol. 1981,
48, 665–678.

17. Pomar, J.L.; et al. Late tears in leaflets of porcine


bioprostheses in adults. Ann. Thorac. Surg. 1984, 37 (1),
78–83.
18. Stein, P.D.; et al. Relation of calcification to torn
leaflets of spontaneously degenerated porcine bioprosthetic
valves. Ann. Thorac. Surg. 1985, 40 (2), 175–180.

19. Vesely, I.; Boughner, D.; Song, T. Tissue buckling as a


mechanism of bioprosthetic valve failure. Ann. Thorac.
Surg. 1988, 46 (3), 302–308.

20. Krucinski, S.; et al. Numerical simulation of leaflet


flexure in bioprosthetic valves mounted on rigid and
expansile stents. J. Biomech. 1993, 26 (8), 929–943.

21. Thubrikar, M.J.; et al. Role of mechanical stress in


calcification of aortic bioprosthetic valves. J. Thorac.
Cardiovasc. Surg. 1983, 86, 115–125.

22. Schoen, F.J.; Tsao, J.W.; Levy, R.J. Calcification of


bovine pericardium used in cardiac valve bioprostheses.
Implications for the mechanisms of bioprosthetic tissue
mineralization. Am. J. Pathol. 1986, 123 (1), 134–145.

23. Ferrans, V.J.; et al. Calcific deposits in porcine


bioprotheses. Structure and pathogenesis. Am. J. Cardiol.
1980, 46, 721–734. 24. Gabbay, S.; et al. Do heart valve
bioprostheses degenerate for metabolic or mechanical
reasons? J. Thorac. Cardiovasc. Surg. 1988, 95 (2),
208–215. 25. Sabbah, H.N.; Hamid, M.S.; Stein, P.D.
Mechanical stresses on closed cusps of porcine
bioprosthetic valves: Correlation with sites of
calcification. Ann. Thorac. Surg. 1986, 42 (1), 93–96. 26.
Schoen, F.J.; Mitchell, R.N.; Jonas, R.A. Pathological
considerations in cryopreserved allograft heart valves. J.
Heart Valve Dis. 1995, 4 (Suppl 1), S72–S75. Discussion
S75–S76. 27. Vesely, I.; et al. Mechanical testing of
cryopreserved aortic allografts. Comparison with xenografts
and fresh tissue. J. Thorac. Cardiovasc. Surg. 1990, 99
(1), 119–123. 28. Vesely, I.; Casarotto, D.C.; Gerosa, G.
Mechanics of cryopreserved aortic and pulmonary homografts.
J. Heart Valve Dis. 2000, 9 (1), 27–37. 29. Gross, L.;
Kugel, M.A. Topographic anatomy and histology of the valves
in the human heart. Am. J. Pathol. 1931, 7, 445–456. 30.
Vesely, I.; Noseworthy, R. Micromechanics of the fibrosa and
the ventricularis in aortic valve leaflets. J. Biomech.
1992, 25 (1), 101–113. 31. Vesely, I.; Lozon, A. Natural
preload of aortic valve leaflet components during
glutaraldehyde fixation: Effects on tissue mechanics. J.
Biomech. 1993, 26 (2), 121–131. 32. Vesely, I.; Lozon, A.;
Talman, E. Is zero-pressure fixation of bioprosthetic valves
truly stress free? J Thorac. Cardiovasc. Surg. 1993, 106
(2), 288–298. 33. Vesely, I. Reconstruction of loads in the
fibrosa and ventricularis of porcine aortic valves. ASAIO J.
1996, 42 (5), M739–M746. 34. Bashey, R.I.; Torii, S.;
Angrist, A. Age-related collagen and elastin content of
human heart valves. J. Gerontol. 1967, 22 (2), 203–208. 35.
Scott, M.J.; Vesely, I. Morphology of porcine aortic valve
cusp elastin. J. Heart Valve Dis. 1996, 5 (5), 464–471. 36.
Kanke, Y.; et al. Biochemical study of cardiac valvular
tissue. Biosynthesis in vitro of hexosamine-containing
substances in bovine heart valve. Biochem. J. 1971, 124
(1), 207–214. 37. Jimenez, S.A.; Bashey, R.I.
Solubilization of bovine heart-value collagen. Biochem. J.
1978, 173 (1), 337–340. 38. Bashey, R.I.; Bashey, H.M.;
Jimenez, S.A. Characterization of pepsin-solubilized bovine
heart-valve collagen. Biochem. J. 1978, 173 (3), 885–894.
39. Nimni, M.E. The molecular organization of collagen and
its role in determining the biophysical properties of the
connective tissues. Biorheology 1980, 17 (1–2), 51–82. 40.
Viidik, A. Interdependence Between Structure and Function
in Collagenous Tissues. In Biology of Collagen; Academic
Press, 1980; 257–280. 41. Hilbert, S.L.; Ferrans, V.J.;
Swanson, W.M. Optical methods for the nondestructive
evaluation of collagen morphology in bioprosthetic heart
valves. J. Biomed. Mater. Res. 1986, 20 (9), 1411–1421. 42.
Vesely, I. Aortic root dilation prior to valve opening
explained by passive hemodynamics. J. Heart Valve Dis.
2000, 9 (1), 16–20. 43. Vesely, I. The role of elastin in
aortic valve mechanics. J. Biomech. 1998, 31 (2), 115–123.
44. Roach, M.R.; Song, S.H. Arterial elastin as seen with
scanning electron microscopy: A review. Scanning Microsc.
1988, 2 (2), 994–1004. 45. Broom, N.; Christie, G. The
Structure/Function Relationship of Fresh and
Glutaraldehyde-Fixed Aortic Valve Leaflets. In Cardiac
Bioprostheses; Cohn, L., Galluci, V., Eds.; Yorke Medical
Books: New York, 1982. 46. Clark, R.E.; Finke, E.H.
Scanning and light microscopy of human aortic leaflets in
stressed and relaxed states. J. Thorac. Cardiovasc. Surg.
1974, 67 (5), 792– 804. 47. Adamczyk, M.M.; Lee, T.C.;
Vesely, I. Biaxial strain properties of elastase-digested
porcine aortic valves. J. Heart Valve Dis. 2000, 9 (3),
445–453. 48. Mako, W.J.; et al. Loss of Glycosaminoglycans
(GAGs) From Implanted Bioprosthetic Heart Valves. In 70th
Scientific Sessions of the American Heart Association; 1997.
49. Grande-Allen, K.J.; et al. Loss of chondroitin
6-sulfate and hyaluronan accompany structural deterioration
of porcine bioprosthetic valves. J. Biomed. Mater. Res.
2002. accepted. 50. Mulholland, D.L.; Gotlieb, A.I. Cell
biology of valvular interstitial cells. Can. J. Cardiol.
1996, 12 (3), 231–236. 51. Taylor, P.M.; Allen, S.P.;
Yacoub, M.H. Phenotypic and functional characterization of
interstitial cells from human heart valves, pericardium and
skin. J. Heart Valve Dis. 2000, 9 (1), 150–158. 52. Roy,
A.; Brand, N.J.; Yacoub, M.H. Molecular characterization of
interstitial cells isolated from human heart valves. J.
Heart Valve Dis. 2000, 9 (3), 459–464. Discussion 464–465.
53. Della Rocca, F.; et al. Cell composition of the human
pulmonary valve: A comparative study with the aortic
valve—the VESALIO Project. Vitalitate Exornatum Succedaneum
Aorticum labore Ingegnoso Obtinebitur. Ann. Thorac. Surg.
2000, 70 (5), 1594–1600. 54. Love, J.W. Autologus Tissue
Valves; R.G. Landes Company: Austin, TX, 1993. 55.
Marchand, M.; et al. Twelve-year experience with
Carpentier–Edwards PERIMOUNT pericardial valve in the
mitral position: A multicenter study. J. Heart Valve Dis.
1998, 7 (3), 292–298. 56. Cheung, D.T.; et al. Behavior of
vital and killed autologous pericardium in the descending
aorta of sheep. J. Thorac. Cardiovasc. Surg. 1999, 118 (6),
998–1005. 57. Marchion, D.C.; et al. Serum components
stimulate pericardial tissue contraction. J. Heart Valve
Dis. 2002, 11 (2), 283–287. 58. Eastwood, M.; McGrouther,
D.A.; Brown, R.A. Fibroblast responses to mechanical
forces. Proc. Inst. Mech. Eng., H 1998, 212 (2), 85–92. 59.
Chiquet, M. Regulation of extracellular matrix gene
expression by mechanical stress. Matrix Biol. 1999, 18 (5),
417–426. 60. Elder, S.H.; et al. Chondrocyte
differentiation is modulated by frequency and duration of
cyclic compressive loading. Ann. Biomed. Eng. 2001, 29 (6),
476–482. 61. Mudera, V.C.; et al. Molecular responses of
human dermal fibroblasts to dual cues: Contact guidance and
mechanical load. Cell Motil. Cytoskelet. 2000, 45 (1), 1–9.
62. Zund, G.; et al. The in vitro construction of a tissue
engineered bioprosthetic heart valve. Eur. J. Cardiothorac.
Surg. 1997, 11 (3), 493–497. 63. Shinoka, T.; et al. Tissue
engineering heart valves: Valve leaflet replacement study in
a lamb model. Ann. Thorac. Surg. 1995, 60 (6 Suppl),
S513–S516. 64. Hoerstrup, S.P.; et al. Functional living
trileaflet heart valves grown in vitro. Circulation 2000,
102 (19 Suppl 3), III44–III49. 65. Vesely, I.; Noseworthy,
R.; Wilson, G. Development of a hybrid xenograft/autograft
aortic valve bioprosthesis. Can. J. Cardiol. 1991. 66.
Wilson, G.J.; et al. Acellular matrix allograft small
caliber vascular prostheses. ASAIO Trans. 1990, 36 (3),
M340–M343. 67. Courtman, D.W.; et al. Development of a
pericardial acellular matrix biomaterial: Biochemical and
mechanical effects of cell extraction. J. Biomed. Mater.
Res. 1994, 28 (6), 655–666. 68. Goldstein, S.; et al.
Transpecies heart valve transplant: Advanced studies of a
bioengineered xeno-autograft. Ann. Thorac. Surg. 2000, 70
(6), 1962–1969. 69. Livesey, S.; Boerboom, L.; Coleman, C.
Decellularized Porcine Heart Valves as Templates for Host
Cell Repopulation. In Workshop on Prosthetic Heart Valves:
Future Directions; Hilton Head: South Carolina, 1998. 70.
Mitchell, R.N.; Jonas, R.A.; Schoen, F.J. Pathology of
explanted cryopreserved allograft heart valves: Comparison
with aortic valves from orthotopic heart transplants. J.
Thorac. Cardiovasc. Surg. 1998, 115 (1), 118–127. 71.
Human, P.; Zilla, P. Characterization of the immune
response to valve bioprostheses and its role in primary
tissue failure. Ann. Thorac. Surg. 2001, 71 (5 Suppl),
S385–S388. 72. Rajani, B.; Mee, R.B.; Ratliff, N.B.
Evidence for rejection of homograft cardiac valves in
infants. J. Thorac. Cardiovasc. Surg. 1998, 115 (1),
111–117. 73. Vyavahare, N.; et al. Mechanisms of
bioprosthetic heart valve failure: Fatigue causes collagen
denaturation and glycosaminoglycan loss. J. Biomed. Mater.
Res. 1999, 46 (1), 44–50. 74. Bell, E.; et al. Living
tissue formed in vitro and accepted as skin-equivalent
tissue of full thickness. Science 1981, 211 (4486),
1052–1054. 75. Huang, D.; et al. Mechanisms and dynamics of
mechanical strengthening in ligamentequivalent
fibroblast-populated collagen matrices. Ann. Biomed. Eng.
1993, 21 (3), 289–305. T

76. Butler, D.L.; Awad, H.A. Perspectives on cell and


collagen composites for tendon repair. Clin. Orthop. 1999,
(367 Suppl), S324–S332.

77. Barocas, V.H.; Tranquillo, R.T. An anisotropic biphasic


theory of tissue-equivalent mechanics: The interplay among
cell traction, fibrillar network deformation, fibril
alignment, and cell contact guidance. J. Biomech. Eng.
1997, 119 (2), 137–145.

78. Bell, E.; Ivarsson, B.; Merrill, C. Production of a


tissue-like structure by contraction of collagen lattices
by human fibroblasts of different proliferative potential in
vitro. Proc. Natl. Acad. Sci. U. S. A. 1979, 76 (3),
1274–1278.

79. Harris, A.K.; Stopak, D.; Wild, P. Fibroblast traction


as a mechanism for collagen morphogenesis. Nature 1981, 290
(5803), 249–251.

80. Stopak, D.; Harris, A.K. Connective tissue


morphogenesis by fibroblast traction. I. Tissue culture
observations. Dev. Biol. 1982, 90 (2), 383–398.

81. Grinnell, F.; Lamke, C.R. Reorganization of hydrated


collagen lattices by human skin fibroblasts. J. Cell. Sci.
1984, 66, 51–63.
82. Guidry, C.; Grinnell, F. Studies on the mechanism of
hydrated collagen gel reorganization by human skin
fibroblasts. J. Cell. Sci. 1985, 79, 67–81.

83. Tranquillo, R.T.; Durrani, M.A.; Moon, A.G. Tissue


engineering science: Consequences of cell traction force.
Cytotechnology 1992, 10 (3), 225–250.

84. Grinnell, F. Fibroblasts, myofibroblasts, and wound


contraction. J. Cell Biol. 1994, 124 (4), 401–404.

85. Grinnell, F. Fibroblast-collagen-matrix contraction:


Growth-factor signalling and mechanical loading. Trends
Cell Biol. 2000, 10 (9), 362–365.

86. Oakes, B.W.; et al. The synthesis of elastin, collagen,


and glycosaminoglycans by high density primary cultures of
neonatal rat aortic smooth muscle. An ultrastructural and
biochemical study. Eur. J. Cell Biol. 1982, 27, 34–46.

87. Shi, Y.; Vesely, I. The Use of Directed Collagen Gel


Shrinkage in the Development of Tissue Engineered Mitral
Valve Chordae. In 1st Biennial Meeting of the Society for
Heart Valve Disease; 2001. London, England.

88. Girton, T.S.; Oegema, T.R.; Tranquillo, R.T. Exploiting


glycation to stiffen and strengthen tissue equivalents for
tissue engineering. J. Biomed. Mater. Res. 1999, 46 (1),
87–92. 89. Seliktar, D.; et al. Dynamic mechanical
conditioning of collagen-gel blood vessel constructs
induces remodeling in vitro. Ann. Biomed. Eng. 2000, 28
(4), 351–362. 90. Eisenberg, L.M.; Markwald, R.R. Molecular
regulation of atrioventricular valvuloseptal morphogenesis.
Circ. Res. 1995, 77 (1), 1–6. 91. Larsen, N.E.; et al.
Hylan gel biomaterial: Dermal and immunologic
compatibility. J. Biomed. Mater. Res. 1993, 27, 1129–1134.
92. Larsen, N.E.; et al. Biocompatibility of Hylan Polymers
in Various Tissue Compartments. In Polymers in Medicine and
Pharmacy; Mikos, A.G., Ed.; Materials Research Society:
Pittsburgh, 1995; 149–153. 93. Denlinger, J.L.; El-Mofty,
A.A.; Balazs, E.A. Replacement of the liquid vitreus with
sodium hyaluronate in monkeys. II. Long-term evaluation.
Exp. Eye Res. 1980, 31 (1), 101–117. 94. Laurent, T.C.
Biochemistry of hyaluronan. Acta Otolaryngol., Suppl. 1987,
442, 7–24. 95. Balasz, E.A.; Leshchiner, A. Crosslinked
Gels of Hyaluronic Acid and Products; Biomatrix Inc., 1984.
96. Balazs, E.A.; Denlinger, J.L. Clinical uses of
hyaluronan. Ciba Found. Symp. 1989, 143, 265–275.
Discussion 275–280. 97. Vercruysse, K.P.; et al. Synthesis
and in vitro degradation of new polyvalent hydrazide
cross-linked hydrogels of hyaluronic acid. Bioconjug. Chem.
1997, 8 (5), 686–694. 98. Tomihata, K.; Ikada, Y.
Crosslinking of hyaluronic acid with water-soluble
carbodiimide. J. Biomed. Mater. Res. 1997, 37 (2), 243–251.
99. Ma¨lson, T.; Lindqvist, B. Crosslinked hyaluronate gels
their use and method for producing them. 1998. 100. Balazs,
E.A.; et al. Matrix engineering. Blood Coagul. Fibrinolysis
1991, 2 (1), 173–178. 101. Ramamurthi, A.; Vesely, I.
Smooth muscle cell adhesion on crosslinked hyaluronan gels.
J. Biomed. Mater. Res. 2002, 60 (1), 195–205. 102. Bryant,
S.J.; Nuttelman, C.R.; Anseth, K.S. The effects of
crosslinking density on cartilage formation in
photocrosslinkable hydrogels. Biomed. Sci. Instrum. 1999,
35, 309–314. 103. Neitmann, M.; et al. Attachment and
chemotaxis of melanocytes after ultraviolet irradiation in
vitro. Br. J. Dermatol. 1999, 141 (5), 794–801.
Tissue Engineering of the Intestine

22. Thompson, J.S.; Kampfe, P.W.; Newland, J.R.;


Vanderhoof, J.A. Growth of intestinal neomucosa on
prosthetic materials. J. Surg. Res. 1986, 41 (5), 484–492.

23. Thompson, J.S.; Hollingsed, T.C.; Saxena, S.K.


Prevention of contraction of patched intestinal defects.
Arch. Surg. 1988, 123 (4), 428–430.

24. Evans, G.S.; Flint, N.; Somers, A.S.; Eyden, B.;


Potten, C.S. The development of a method for the
preparation of rat intestinal epithelial cell primary
cultures. J. Cell Sci. 1992, 101, 219–231.

25. Choi, R.S.; Riegler, M.; Pothoulakis, C.; Kim, B.S.;


Mooney, D.; Vacanti, M.; Vacanti, J.P. Studies of brush
border enzymes, basement membrane components, and
electrophysiology of tissue-engineered neointestine. J.
Pediatr. Surg. 1998, 33 (7), 991–997.

26. Kim, S.S.; Kaihara, S.; Benvenuto, M.S.; Choi, R.S.;


Kim, B.S.; Mooney, D.J.; Vacanti, J.P. Effects of
anastomosis of tissue-engineered neointestine to native
small bowel. J. Surg. Res. 1999, 87 (1), 6–13.

27. Grikscheit, T.C.; Siddique, A.; Ochoa, E.R.;


Srinivasan, A.; Alsberg, E.; Hodin, R.A.; Vacanti, J.P.
Tissueengineered small intestine improves recovery after
massive small bowel resection. Ann. Surg. 2004, 240 (5),
748–754.

28. Grikscheit, T.C.; Ochoa, E.R.; Ramsanahie, A.; Alsberg,


E.; Mooney, D.; Whang, E.E.; Vacanti, J.P. TissueEngineered
large intestine resembles native colon with appropriate in
vitro physiology and architecture. Ann. Surg. 2003, 238
(1), 35–41.

29. Yamada, T.; Yoshikawa, M.; Takaki, M.; Torihashi, S.;


Kato, Y.; Nakajima, Y.; Ishizaka, S.; Tsunoda, Y. In vitro
functional gut-like organ formation from mouse embryonic
stem cells. Stem Cells 2002, 20 (1), 41–49.

30. Micci, M.A.; Learish, R.D.; Li, H.; Abraham, B.P.;


Pasricha, P.J. Neural stem cells express RET, produce
nitric oxide, and survive transplantation in the
gastrointestinal tract. Gastroenterology 2001, 121 (4),
757–766.

31. Brittingham, J.; Phiel, C.; Trzyna, W.C.; Gabbeta, V.;


McHugh, K.M. Identification of distinct molecular phenotypes
in cultured gastrointestinal smooth muscle cells.
Gastroenterology 1998, 115 (3), 605–617.

32. Kruger, G.M.; Mosher, J.T.; Bixby, S.; Joseph, N.;


Iwashita, T.; Morrison, S.J. Neural crest stem cells
persist in the adult gut but undergo changes in
self-renewal, neuronal subtype potential, and factor
responsiveness. Neuron 2002, 35 (4), 657–669.

33. Li, C.X.; Liu, B.H.; Tong, W.D.; Zhang, L.Y.; Jiang,
Y.P. Dissociation, culture and morphologic changes of
interstitial cells of Cajal in vitro. World J.
Gastroenterol. 2005, 11 (18), 2838–2840.

34. Hori, Y.; Nakamura, T.; Matsumoto, K.; Kurokawa, Y.;


Satomi, S.; Shimizu, Y. Tissue engineering of the small
intestine by acellular collagen sponge scaffold grafting.
Int. J. Artif. Organs. 2001, 24 (1), 50–54.

35. Lee, M.; Dunn, J.C.; Wu, B.M. Scaffold fabrication by


indirect three-dimensional printing. Biomaterials. 2005, 26
(20), 4281–4289.

36. Nishimura, I.; Garrell, R.L.; Hedrick, M.; Iida, K.;


Osher, S.; Wu, B. Precursor tissue analogs as a
tissue-engineering strategy. Tissue Eng. 2003, 9 (suppl 1),
S77–S89. 37. Chen, M.K.; Badylak, S.F. Small bowel tissue
engineering using small intestinal submucosa as a scaffold.
J. Surg. Res. 2001, 99 (2), 352–358. 38. Hori, Y.;
Nakamura, T.; Kimura, D.; Kaino, K.; Kurokawa, Y.; Satomi,
S.; Shimizu, Y. Experimental study on tissue engineering of
the small intestine by mesenchymal stem cell seeding. J.
Surg. Res. 2002, 102 (2), 156–160. 39. Thiesen, A., Jr.;
Drozdowski, L.; Iordache, C.; Neo, C.C.; Woudstra, T.D.;
Xenodemetropoulos, T.; Keelan, M.; Clandinin, M.T.;
Thompson, A.B.R. Adaptation following intestinal resection:
mechanisms and signals. Best Pract. Res. Clin.
Gastroenterol. 2003, 17 (6), 981–995. 40. Dunn, J.C.;
Parungo, C.P.; Fonkalsrud, E.W.; McFadden, D.W.; Ashley,
S.W. Epidermal growth factor selectively enhances
functional enterocyte adaptation after massive small bowel
resection. J. Surg. Res. 1997, 67 (1), 90–93. 41. Huang,
F.S.; Kemp, C.J.; Williams, J.L.; Erwin, C.R.; Warner, B.W.
Role of epidermal growth factor and its receptor in
chemotherapy-induced intestinal injury. Am. J. Physiol.
Gastrointest. Liver Physiol. 2002, 282 (3), G432–G442. 42.
Kuenzler, K.A.; Pearson, P.Y.; Schwartz, M.Z. Hepatoctye
growth factor pretreatment reduces apoptosis and mucosal
damage after intestinal ischemia-reperfusion. J. Pediatr.
Surg. 2002, 37, 1093–1097. 43. Ohda, Y.; Hori, K.; Tomita,
T.; Hida, N.; Kosaka, T.; Fukuda, Y.; Miwa, H.; Matsumoto,
T. Effects of hepatocyte growth factor on rat inflammatory
bowel disease models. Dig. Dis. Sci. 2005, 50 (5), 914–921.
44. Kitchen, P.A.; Goodlad, R.A.; FitzGerald, A.J.; Mandir,
N.; Ghatei, M.A.; Bloom, S.R.; Berlanga-Acosta, J.;
Playford, R.J.; Forbes, A.; Walters, J.R. Intestinal growth
in parenterally-fed rats induced by the combined effects of
glucagon-like peptide 2 and epidermal growth factor. JPEN
2005, 29 (4), 248–254. 45. Shin, E.D.; Estall, J.L.; Izzo,
A.; Drucker, D.J.; Brubaker, P.L. Mucosal adaptation to
enteral nutrients is dependent on the physiologic actions
of glucagon-like peptide-2 in mice. Gastroenterology 2005,
128 (5), 1340–1353. 46. Fiore, N.F.; Ledniczky, G.; Liu,
Q.; Orazi, A.; Du, X.; Williams, D.A.; Grosfeld, J.L.
Comparison of interleukin-11 and epidermal growth factor on
residual small intestine after massive small bowel
resection. J. Pediatr. Surg. 1998, 33 (1), 24–29. 47.
Steeb, C.B.; Trahair, J.F.; Tomas, F.M.; Read, L.C.
Prolonged administration of IGF peptides enhances growth of
gastrointestinal tissues in normal rats. Am. J. Physiol.
1994, 266 (6 Pt 1), G1090–G1098. 48. Lund, P.K. Molecular
basis of intestinal adaptation: the role of the
insulin-like growth factor system. Ann. N Y Acad. Sci.
1998, 859, 18–36. 49. Peterson, C.A.; Gillingham, M.B.;
Mohapatra, N.K.; Dahly, E.M.; Adamo, M.L.; Carey, H.V.;
Lund, P.K.; Ney, D.M. Enterotrophic effect of insulin-like
growth factor-I but not growth hormone and localized
expression of insulin-like growth factor-I, insulin-like
growth factor binding protein-3 and -5mRNAs in jejunum of
parenterally fed rats. JPEN 2000, 24 (5), 288–295. 50.
Theiss, A.L.; Fruchtman, S.; Lund, P.K. Growth factors in
inflammatory bowel disease: the actions and interactions of
growth hormone and insulin-like growth factor-I. Inflamm.
Bowel Dis. 2004, 10 (6), 871–880. 51. Wang, J.; Niu, W.;
Nikiforov, Y.; Naito, S.; Chernausek, S.; Witte, D.;
LeRoith, D.; Strauch, A.; Fagin, J.A. Targeted
overexpression of IGF-I evokes distinct patterns of organ
remodeling in smooth muscle cell tissue beds of transgenic
mice. J. Clin. Invest. 1997, 100, 1425–1439. 52. Knott,
A.W.; Juno, R.J.; Jarboe, M.D.; Profitt, S.A.; Erwin, C.R.;
Smith, E.P.; Fagin, J.A.; Warner, B.W. Smooth muscle
overexpression of IGF-I induces a novel adaptive response
to small bowel resection. Am. J. Physiol. Gastrointest.
Liver Physiol. 2004, 287 (3), G562–G570. 53. Kuemmerle,
J.F.; Zhou, H.; Bowers, J.G. IGF-I stimulates human
intestinal smooth muscle cell growth by regulation of G1
phase cell cycle proteins. Am. J. Physiol. Gastrointest.
Liver Physiol. 2004, 286 (3), G412–G419. 54. Walters, J.R.
Cell and molecular biology of the small intestine: new
insights into differentiation, growth and repair. Curr.
Opin. Gastroenterol. 2004, 20 (2), 70–76. 55. Kleber, M.;
Lee, H.Y.; Wurdak, H.; Buchstaller, J.; Riccomagno, M.M.;
Ittner, L.M.; Suter, U.; Epstein, D.J.; Sommer, L. Neural
crest stem cell maintenance by combinatorial Wnt and BMP
signaling. J. Cell Biol. 2005, 169 (2), 309–320. 56.
Gardner-Thorpe, J.; Grikscheit, T.C.; Ito, H.; Perez, A.;
Ashley, S.W.; Vacanti, J.P.; Whang, E.E. Angiogenesis in
tissue-engineered small intestine. Tissue Eng. 2003, 9 (6),
1255–1261. 57. Duxbury, M.S.; Grikscheit, T.C.;
Gardner-Thorpe, J.; Rocha, F.G.; Ito, H.; Perez, A.;
Ashley, S.W.; Vacanti, J.P.; Whang, E.E. Lymphangiogenesis
in tissueengineered small intestine. Transplantation 2004,
77 (8), 1162–1166. 58. Park, J.; Puapong, D.P.; Wu, B.M.;
Atkinson, J.B.; Dunn, J.C. Enterogenesis by mechanical
lengthening: morphology and function of the lengthened
small intestine. J. Pediatr. Surg. 2004, 39 (12),
1823–1827. 59. Stark, G.B.; Dorer, A.; Walgenbach, K.J.;
Grunwald, F.; Jaeger, K. The creation of a small bowel
pouch by tissue expansion: an experimental study in pigs.
Langenbecks Arch. Chir. 1990, 375 (3), 145–150. 60.
Narayan, D.; Castro, A.; Jackson, I.T.; Herschman, B.
Tissue expanders in the gut: a histological and
angiographic study. J. R. Coll. Surg. Edinb. 1992, 37,
402–404. 61. Kawaguchi, A.L.; Dunn, J.C.; Lam, M.;
O’Connor, T.P.; Diamond, J.; Fonkalsrud, E.W. Glucose
uptake in dilated small intestine. J. Pediatr. Surg. 1998,
33 (11), 1670–1673. 62. Manders, E.K.; Saggers, G.C.;
Diaz-Alonso, P.; Finn, L.; Sipio, J.C.; Glumac, T.; Au,
V.K.; Wong, R.K.M.; Mottaleb, M. Elongation of peripheral
nerve and viscera containing smooth muscle. Clin. Plast.
Surg. 1987, 14 (3), 551–562. 63. Printz, H.; Schlenzka, R.;
Requadt, P.; Tscherny, M.; Wagner, A.C.; Eissele, R.;
Rothmund, M.; Arnold, R.; Goke, B. Small bowel lengthening
by mechanical distraction. Digestion 1997, 58 (3), 240–248.
64. Chen, Y.; Zhang, J.; Qu, R.; Wang, J.; Xie, Y. An
animal experiment on short gut lengthening. Chin. Med. J.
1997, 110 (5), 354–357. T
Tissue Engineering of Ligament

35. Doukas, W.C.; Speer, K.P. Anatomy, pathophysiology, and


biomechanics of shoulder instability. Orthop. Clin. North
Am. 2001, 32. pp. vii, 381–391.

36. Lephart, S.M.; Kocher, M.S. The Role of Exercise in the


Prevention of Shoulder Disorders. In The Shoulder: A
Balance of Mobility and Stability; Matsen, F.A., Fu, F.H.,
Hawkins, R.J., Eds.; American Academy of Orthopaedic
Surgeons: Rosemont, IL, 1993.

37. Schultz, R.A.; Miller, D.C.; Kerr, C.S.; Micheli, L.


Mechanoreceptors in human cruciate ligaments. A
histological study. J. Bone Jt. Surg., Am. 1984, 66,
1072–1076.

38. Schutte, M.J.; Dabezies, E.J.; Zimny, M.L.; Happel,


L.T. Neural anatomy of the human anterior cruciate
ligament. J. Bone Jt. Surg., Am. 1987, 69, 243–247.

39. Myers, J.B.; Lephart, S.M. Sensorimotor deficits


contributing to glenohumeral instability. Clin. Orthop.
2002, 400, 98–104.

40. Fischer-Rasmussen, T.; Jensen, P.E. Proprioceptive


sensitivity and performance in anterior cruciate
ligament-deficient knee joints. Scand. J. Med. Sci. Sports
2000, 10, 85–89.

41. Fremerey, R.W.; Lobenhoffer, P.; Zeichen, J.; Skutek,


M.; Bosch, U.; Tscherne, H. Proprioception after
rehabilitation and reconstruction in knees with deficiency
of the anterior cruciate ligament: A prospective,
longitudinal study. J. Bone Jt. Surg., Br. 2000, 82,
801–806.

42. Friden, T.; Roberts, D.; Ageberg, E.; Walden, M.;


Zatterstrom, R. Review of knee proprioception and the
relation to extremity function after an anterior cruciate
ligament rupture. J. Orthop. Sports Phys. Ther. 2001, 31,
567–576.

43. Black, J. In Biological Performance of Materials:


Fundamentals of Biocompatibility, 2nd Ed.; Marcel Dekker:
New York, NY, 1992.

44. Dunn, M.G.; Liesch, J.B.; Tiku, M.L.; Zawadsky, J.P.


Development of fibroblast-seeded ligament analogs for ACL
reconstruction. J. Biomed. Mater. Res. 1995, 29, 1363–1371.
45. Goulet, F.; Rancourt, D.; Cloutier, R.; Germain, L.;
Poole, A.R.; Auger, F.A. Tendons and Ligaments. In
Principles of Tissue Engineering, 2nd Ed.; Lanza, R.P.,
Langer, R., Vacanti, J., Eds.; Academic Press: San Diego,
2000.

46. Goulet, F.; Germain, L.; Rancourt, D.; Caron, C.;


Normand, A.; Auger, F.A. Tendons and Ligaments. In
Principles of Tissue Engineering; Lanza, R.P., Langer, R.,
Chick, W.L., Eds.; Academic Press: San Diego, 1997.

47. Altman, G.H.; Horan, R.L.; Martin, I.; Farhadi, J.;


Stark, P.R.; Volloch, V.; Richmond, J.C.; VunjakNovakovic,
G.; Kaplan, D.L. Cell differentiation by mechanical stress.
FASEB J. 2002, 16, 270–272.

48. Dunn, M.G.; Maxian, S.H.; Zawadsky, J.P. Intraosseous


incorporation of composite collagen prostheses designed for
ligament reconstruction. J. Orthop. Res. 1994, 12, 128–137.

49. McCarthy, M.R.; Yates, C.K.; Anderson, M.A.;


YatesMcCarthy, J.L. The effects of immediate continuous
passive motion on pain during the inflammatory phase of soft
tissue healing following anterior cruciate ligament
reconstruction. J. Orthop. Sports Phys. Ther. 1993, 17,
96–101. 50. Dunn, M.G.; Avasarala, P.N.; Zawadsky, J.P.
Optimization of extruded collagen fibers for ACL
reconstruction. J. Biomed. Mater. Res. 1993, 27, 1545–1552.
51. Cao, Y.; Vacanti, J.P.; Ma, X.; Paige, K.T.; Upton, J.;
Chowanski, Z.; Schloo, B.; Langer, R.; Vacanti, C.A.
Generation of neo-tendon using synthetic polymers seeded
with tenocytes. Transplant. Proc. 1994, 26, 3390–3392. 52.
Huang, D.; Chang, T.R.; Aggarwal, A.; Lee, R.C.; Ehrlich,
H.P. Mechanisms and dynamics of mechanical strengthening in
ligament-equivalent fibroblast-populated collagen matrices.
Ann. Biomed. Eng. 1993, 21, 289–305. 53. Dunn, M.G.; Tria,
A.J.; Kato, Y.P.; Bechler, J.R.; Ochner, R.S.; Zawadsky,
J.P.; Silver, F.H. Anterior cruciate ligament
reconstruction using a composite collagenous prosthesis. A
biomechanical and histologic study in rabbits. Am. J.
Sports Med. 1992, 20, 507–515. 54. Goldstein, J.D.; Tria,
A.J.; Zawadsky, J.P.; Kato, Y.P.; Christiansen, D.; Silver,
F.H. Development of a reconstituted collagen tendon
prosthesis. J. Bone Jt. Surg. 1989, 71, AM., 1183–1191. 55.
Kato, Y.P.; Christiansen, D.L.; Hahn, R.A.; Shieh, S.J.;
Goldstein, J.D.; Silver, F.H. Mechanical properties of
collagen fibres: A comparison of reconstituted and rat tail
tendon fibres. Biomaterials 1989, 10, 38–42. 56. Law, J.K.;
Parsons, J.R.; Silver, F.H.; Weiss, A.B. An evaluation of
purified reconstituted Type I collagen fibers. J. Biomed.
Mater. Res. 1989, 23, 961–977. 57. Wasserman, A.J.; Kato,
Y.P.; Christiansen, D.; Dunn, M.G.; Silver, F.H. Achilles
tendon replacement by a collagen fiber prosthesis:
Morphological evaluation of neotendon formation. Scanning
Microsc. 1989, 3, 1183–1200. 58. Bellincampi, L.D.;
Closkey, R.F.; Prasad, R.; Zawadsky, J.P.; Dunn, M.G.
Viability of fibroblast-seeded ligament analogs after
autogenous implantation. J. Orthop. Res. 1998, 16, 414–420.
59. Weadock, K.; Olson, R.M.; Silver, F.H. Evaluation of
collagen crosslinking techniques. Biomater. Med. Dev.
Artif. Organs 1983, 11, 293–318. 60. Kato, Y.P.; Dunn,
M.G.; Zawadsky, J.P.; Tria, A.J.; Silver, F.H. Regeneration
of Achilles tendon with a collagen tendon prosthesis.
Results of a one-year implantation study. J. Bone Jt.
Surg., Am. 1991, 73, 561–574. 61. Sugita, T.; Amis, A.A.
Anatomic and biomechanical study of the lateral collateral
and popliteofibular ligaments. Am. J. Sports Med. 2001, 29,
466–472. 62. Staubli, H.U.; Schatzmann, L.; Brunner, P.;
Rincon, L.; Nolte, L.P. Mechanical tensile properties of
the quadriceps tendon and patellar ligament in young
adults. Am. J. Sports Med. 1999, 27, 27–34. 63. Quapp,
K.M.; Weiss, J.A. Material characterization of the human
medial collateral ligament. J. Biomech. Eng. 1998, 120,
757–763.
Tissue Engineering of Liver

1. Scientific Registry of Transplant Recipients Oct. 31,


2002 http://www.ustransplant.org/facts.html. 2. Asonuma,
K.; Gilbert, J.C.; Stein, J.E.; Takeda, T.; Vacanti, J.P.
Quantification of transplanted hepatic mass necessary to
cure the Gunn rat model of hyperbilirubinemia. J. Pediatr.
Surg. 1992, 27 (3), 298–301. 3. Kaihara, S.; Borenstein,
J.; Koka, R.; Lalan, S.; Ochoa, E.R.; Ravens, M.; Pien, H.;
Cunningham, B.; Vacanti, J.P. Silicon micromachining to
tissue engineer branched vascular channels for liver
fabrication. Tissue Eng. 2000, 6 (2), 105–117. 4. Cheung,
W.; Borenstein, J.; Kaazempur-Mofrad, M.; Shin, M.; Sevy,
A.; Kulig, K.; Vacanti, J.P. Development of an Implantable
Tissue-Engineered Liver Device with a Vascular Network of
Channels Co-Cultured with Human Hepatocytes and Human
Microvascular Endothelial Cells. Presented at the American
Transplant Congress, Washington, D.C., May 30–June 4, 2003.
5. Gupta, S.; Malhi, H.; Gorla, G.R. Re-engineering the
liver with natural biomaterials. Yonsei Med. J. 2000, 41
(6), 814–824. 6. Powers, M.J.; Domansky, K.;
Kaazempur-Mofrad, M.R.; Kalezi, A.; Capitano, A.;
Upadhyaya, A.; Kurzawski, P.; Wack, K.E.; Beer Stolz, D.;
Kamm, R.; Griffith, L.G. A microfabricated array bioreactor
for perfused 3D liver culture. Biotechnol. Bioeng. 2002, 78
(3), 257–269. 7. Davis, M.W.; Vacanti, J.P. Toward
development of an implantable tissue-engineered liver.
Biomaterials 1996, 17 (3), 365–372. 8. te Velde, A.A.;
Ladiges, N.C.J.J.; Flendrig, L.M.; Chamuleau, R.A.F.M.
Functional activity of isolated pig hepatocytes attached to
different extracellular matrix substances. Implication for
application of pig hepatocytes in a bioartificial liver. J.
Hepatol. 1995, 23 (2), 184–192. 9. Mitaka, T.; Kojima, T.;
Mizuguchi, T.; Mochizuki, Y. Growth and maturation of small
hepatocytes isolated from adult rat liver. Biochem.
Biophys. Res. Commun. 1995, 214 (2), 310–317. 10. Mitaka,
T.; Mizuguchi, T.; Sato, F.; Mochizuki, C.; Mochizuki, Y.
Growth and maturation of small hepatocytes. J.
Gastroenterol. Hepatol. 1998, 13 (Suppl.), S70–S77. 11.
Kim, S.S.; Sundback, C.A.; Kaihara, S.; Benvenuto, M.S.;
Kim, B.; Mooney, D.J.; Vacanti, J.P. Dynamic seeding and in
vitro culture of hepatocytes in a flow perfusion system.
Tissue Eng. 2000, 6 (1), 39–44. 12. Powers, M.J.; Janigian,
D.M.; Wack, K.E.; Baker, C.S.; Beer Stolz, D.; Griffith,
L.G. Functional behavior of primary rat liver cells in a
three-dimensional perfused micorarray bioreactor. Tissue
Eng. 2002, 8 (3), 499–512. 13. Mooney, D.; Hansen, L.;
Vacanti, J.; Langer, R.; Farmer, S.; Ingber, D. Switching
from differentiation to growth in hepatocytes: Control by
extracellular matrix. J. Cell. Physiol. 1992, 151 (3),
497–505. 14. Sugimoto, S.; Mitaka, T.; Ikeda, S.; Harada,
K.; Ikai, I.; Yamaoka, Y.; Mochizuki, Y. Morphological
changes induced by extracellular matrix are correlated with
maturation of rat small hepatocytes. J. Cell. Biochem.
2002, 87 (1), 16–28. 15. Goulet, F.; Normand, C.; Morin, O.
Cellular interactions promote tissue-specific function,
biomatrix deposition and junctional communication of
primary cultured hepatocytes. Hepatology 1988, 8 (5),
1010–1018. 16. Hansen, L.K.; Mooney, D.J.; Vacanti, J.P.;
Ingber, D.E. Integrin binding and cell spreading on
extracellular matrix act at different points in the cell
cycle to promote hepatocyte growth. Mol. Biol. Cell 1994, 5
(9), 967–975. 17. Ingber, D. Mechanical signaling. Ann.
N.Y. Acad. Sci. 2002, 961, 162–163. 18. Kim, W.J.H.
Cellular signaling in tissue regeneration. Yonsei Med. J.
2000, 41 (6), 692–703. 19. Bhandari, R.N.B.; Riccalton,
L.A.; Lewis, A.L.; Fry, J.R.; Hammond, A.H.; Tendler,
S.J.B.; Shakesheff, K.M. Liver tissue engineering: A role
for co-culture systems in modifying hepatocyte function and
viability. Tissue Eng. 2001, 7 (3), 345–357. 20.
Michalopoulos, G.K.; Bowen, W.C.; Zajac, V.F.; BeerStolz,
D.; Watkins, S.; Kostrubsky, V.; Strom, S.C. Morphogenetic
events in mixed cultures of rat hepatocytes and
nonparenchymal cells maintained in biological matrices in
the presence of hepatocyte growth factor and epidermal
growth factor. Hepatology 1999, 29 (1), 90–100. 21. Mooney,
D.J.; Kaufmann, P.M.; Sano, K.; Schwendeman, S.P.; Majahod,
K.; Schloo, B.; Vacanti, J.P.; Langer, R. Localized
delivery of epidermal growth factor improves the survial of
transplanted hepatocytes. Biotechnol. Bioeng. 1996, 50 (4),
422–429. 22. Mitaka, T.; Mikami, M.; Sattler, G.L.; Pitot,
H.C.; Mochizuki, Y. Small cell colonies appear in the
primary culture of adult rat hepatocytes in the presence of
nicotinamide and epidermal growth factor. Hepatology 1992,
16 (2), 440–447. 23. Xin, X.; Yang, S.; Ingle, G.; Zlot,
C.; Rangell, L.; Kowalski, J.; Schwall, R.; Ferrara, N.;
Gerritsen, M.E. Hepatocyte growth factor enhances vascular
endothelial growth factor-induced angiogenesis in vitro and
in vivo. Am. J. Pathol. 2001, 158 (3), 1111–1120. 24.
Nyberg, S.L.; Remmel, R.P.; Mann, H.J.; Peshwa, M.V.; Hu,
W.; Cerra, F.B. Primary hepatocytes outperform HepG2 cells
as the source of biotransformation functions in a
bioartificial liver. Ann. Surg. 1994, 220 (1), 59–67. 25.
Allen, J.W.; Bhatia, S.N. Engineering liver therapies for
the future. Tissue Eng. 2002, 8 (5), 725–737. 26. Faris,
R.A.; Konkin, T.; Halpert, G. Liver stem cells: A potential
source of hepatocytes for the treatment of human liver
disease. Artif. Organs 2001, 25 (7), 513–521. 27. Noishiki,
Y. Dreams for the future in the field of in vivo tissue
engineering. Artif. Organs 2001, 25 (3), 159–163. 28.
Flendrig, L.M.; la Soe, J.W.; Jo¨rning, G.G.A.; Steenbeek,
A.; Karlsen, O.T.; Bove´e, W.M.M.J.; Ladiges, N.C.J.J.; te
Velde, A.A.; Chamuleau, R.A.F.M. In vitro evaluation of a
novel bioreactor based on an integral oxygenator and a
spirally wound nonwoven polyester matrix for hepatocyte
culture as small aggregates. J. Hepatol. 1997, 26 (6),
1379–1392. 29. Mizuguchi, T.; Hui, T.; Palm, K.; Sugiyama,
N.; Mitaka, T.; Demetriou, A.A.; Rozga. Enhanced
proliferation and differentiation of rat hepatocytes
cultured with bone marrow stromal cells. J. Cell. Physiol.
2001, 189 (1), 106–119. 30. Terada, S.; Sato, M.; Sevy, A.;
Vacanti, J.P. Tissue engineering in the twenty-first
century. Yonsei Med. J. 2000, 41 (6), 685–691. 31. Griffith,
L.G. Emerging design principles in biomaterials and
scaffolds for tissue engineering. Ann. N.Y. Acad. Sci.
2002, 961, 83–95. 32. Wang, Y.; Guillermo, A.A.; Sheppard,
B.J.; Langer, R. A tough biodegradable elastomer. Nat.
Biotechnol. 2002, 20 (6), 602–606. 33. Carlisle, E.S.;
Mariappan, M.R.; Nelson, K.D.; Thomes, B.E.; Timmons, R.B.;
Constantinescu, A.; Eberhart, R.C.; Bankey, P.E. Enhancing
hepatocyte adhesion by pulsed plasma deposition and
polyethylene glycol coupling. Tissue Eng. 2000, 6 (1),
45–52. 34. Irvine, D.J.; Mayes, A.M.; Griffith, L.G.
Nanoscale clustering of RGD peptides at surfaces using comb
polymers. 1. Synthesis and characterization of comb thin
films. Biomacromolecules 2001, 2 (1), 85–94. 35. Irvine,
D.J.; Ruzette, A.V.; Mayes, A.M.; Griffith, L.G. Nanoscale
clustering of RGD peptides at surfaces using comb polymers.
2. Surface segregation of comb polymers in polylactide.
Biomacromolecules 2001, 2 (2), 545–556. 36. Prestwitch,
G.D.; Matthew, H. Hybrid, composite, and complex
biomaterials. Ann. N.Y. Acad. Sci. 2002, 961, 106–108. 37.
Metzger, D.W. Immune responses to tissue-engineered
extracellular matrix used as a bioscaffold. Ann. N.Y. Acad.
Sci. 2002, 961, 335–336. 38. Vacanti, J.P.; Morse, M.A.;
Saltzman, W.M.; Domb, A.J.; Perez-Atayde, A.; Langer, R.
Selective cell transplantation using bioabsorbable
artificial polymers as matrices. J. Pediatr. Surg. 1988, 23
(1 pt 2), 3–9. 39. Wolfe, S.P.; Hsu, E.; Reid, L.M.;
Macdonald, J.M. A novel multi-coaxial hollow fiber
bioreactor for adherent cell types. Part 1: Hydrodynamic
studies. Biotechnol. Bioeng. 2002, 77 (1), 83–90. 40. Wolf,
C.F.; Munkelt, B.E. Bilirubin conjugation by an artificial
liver composed of cultured cells and synthetic capillaries.
Trans. Am. Soc. Artif. Intern. Organs 1975, 21, 16–27. 41.
Arnaout, W.S.; Moscioni, A.D.; Barbout, R.L.; Demetriou,
A.A. Development of bioartificial liver: Bilirubin
conjugation in gunn rats. J. Surg. Res. 1990, 48 (4),
379–382. 42. Nyberg, S.L.; Shirabe, K.; Peshwa, M.V.;
Sielaff, T.D.; Crotty, P.L.; Mann, H.J.; Remmel, R.P.;
Payne, W.D.; Hu, W.S.; Cerra, F.B. Extracorporeal
application of a gel-entrapment, bioartificial liver:
Demonstration of drug metabolism and other biochemical
functions. Cell Transplant 1993, 2 (6), 441–452. 43. Chen,
S.C.; Hewitt, W.R.; Watanabe, F.D.; Eguchi, S.; Kahaku, E.;
Middleton, Y.; Rozga, J.; Demetriou, A.A. Clinical
experience with a porcine hepatocyte-based liver support
system. Int. J. Artif. Organs 1996, 19 (11), 664–669. T

44. Detry, O.; Arkadopoulos, N.; Ting, P.; Kahaku, E.;


Watanabe, F.D.; Rozga, J.; Demetriou, A.A. Clinical use of
a bioartificial liver in the treatment of
acetaminopheninduced fuliminant hepatic failure. Am.
Surgeon 1999, 65 (10), 934–938.

45. Watanabe, F.D.; Mullon, C.J.-P.; Hewitt, W.R.;


Arkadopoulos, N.; Kahaku, E.; Eguchi, S.; Khalili, T.;
Arnaout, W.; Shackleton, C.R.; Rozga, J.; Solomon, B.;
Demetriou, A.A. Clinical experience with a bioartificial
liver in the treatment of severe liver failure—A phase I
clinical trial. Ann. Surg. 1997, 225, 484–491.

46. Sussman, N.L.; Gislason, G.T.; Kelly, J.H.


Extracorporeal liver support. Application to fulminant
hepatic failure. J. Clin. Gastroenterol. 1994, 18, 320–324.

47. Sussman, N.L.; Kelly, J.H. Extracorporeal liver


support: Cell-based therapy for the failing liver. Am. J.
Kidney Dis. 1997, 30 (5), S66–S71.

48. Ellis, A.J.; Hughes, R.D.; Wendon, J.A.; Dunne, J.;


Langley, P.G.; Kelly, J.H.; Gislason, G.T.; Sussman, N.L.;
Williams, R. Pilot-controlled trial of the extracorporeal
liver assist device in acute liver failure. Hepatology
1996, 24, 1446–1451.

49. Gerlach, J.C. Long-term liver cell cultures in


bioreactors and possible application for liver support.
Cell Biol. Toxicol. 1997, 13 (4–5), 349–355.

50. Gerlach, J.C.; Lemmens, P.; Schon, M.; Janke, J.;


Rossaint, R.; Busse, B.; Puhl, G.; Neuhaus, P. Experimental
evaluation of a hybrid liver support system. Transplant.
Proc. 1997, 29 (1–2), 852.

51. Sauer, I.M.; Gerlach, J.C. Modular extracorporeal liver


support. Artif. Organs 2002, 26 (8), 703–706.

52. Mazariegos, G.V.; Kramer, D.J.; Lopez, R.C.; Shakil,


A.O.; Rosenbloom, A.J.; DeVera, M.; Giraldo, M.; Grogan,
T.A.; Zhu, Y.; Fulmer, M.L.; Amiot, B.P.; Patzer, J.F., II.
Safety observations in phase I clinical evaluation of the
excorp medical bioartificial liver support system after the
first four patients. ASAIO 2001, 47, 471–475. 53.
Mazariegos, G.V.; Patzer, J.F., II; Lopez, R.C.; Giraldo,
M.; Devera, M.E.; Grogan, T.A.; Zhu, Y.; Fulmer, M.L.;
Amiot, B.P.; Kramer, D.L. First clinical use of a novel
bioartificial liver support system (BLSS). Am. J. Transplant
2002, 2 (3), 260–266. 54. Patzer, J.F., II; Mazariegos,
G.V.; Lopez, R. Preclinical evaluation of the Excorp
Medical Inc, bioartificial liver support system. J. Am.
Coll. Surg. 2002, 195 (3), 299–310. 55. Kuddus, R.; Patzer,
J.F., II; Lopez, R.; Mazariegos, G.V.; Meighen, B.; Kramer,
D.J.; Rao, A.S. Clinical and laboratory evaluation of the
safety of a bioartificial liver assist device for potential
transmission of porcine endogenous retorvirous.
Transplantation 2002, 73 (3), 740–749. 56. Richardson,
T.P.; Peters, M.C.; Ennett, A.B.; Mooney, D.J. Polymeric
system for dual growth factor delivery. Nat. Biotechnol.
2001, 19 (11), 1029–1034. 57. Peters, M.C.; Polverini,
P.J.; Mooney, D.J. Engineering vascular networks in porous
polymer matrices. J. Biomed. Mater. Res. 2002, 60 (4),
668–678. 58. Fontaine, M.; Schloo, B.; Jenkins, R.; Uyama,
S.; Hansen, L.; Vacanti, J.P. Human hepatocyte isolation
and transplantation into an athymic rat, using
prevascularized cell polymer constructs. J. Pediatr. Surg.
1995, 30 (1), 56–60. 59. Toner, M.; Kocsis, J. Storage and
translational issues in reparative medicine. Ann. N.Y.
Acad. Sci. 2002, 961, 258–262.
Tissue Engineering, Microscale

9. Zilla, P.; Greisler, H.P. Tissue Engineering of Vascular


Prosthetic Grafts; R G Landes Company: Austin, TX, 1999.

10. Nerem, R.M.; Seliktar, D. Vascular tissue engineering.


Annu. Rev. Biomed. Eng. 2001, 3, 225–243.

11. Desai, T.A. Microand nanoscale structures for tissue


engineering constructs. Med. Eng. Phys. 2000, 22, 595–606.

12. Prokop, A. Bioartificial organs in the twenty-first


century: Nanobiological devices. Ann. N.Y. Acad. Sci. 2001,
944, 472–490.

13. Whitesides, G.M.; Ostuni, E.; Takayama, S.; Jiang, X.;


Ingber, D.E. Soft lithography in biology and biochemistry.
Annu. Rev. Biomed. Eng. 2001, 3, 335–373.

14. Ito, Y. Surface micropatterning to regulate cell


functions. Biomaterials 1999, 20, 2333–2342.

15. Xia, Y.; Whitesides, G.M. Soft lithography. Angew.


Chem. Int. Ed. 1998, 37, 550–575.

16. Kane, R.S.; Takayama, S.; Ostuni, E.; Ingber, D.E.;


Whitesides, G.M. Patterning proteins and cells using soft
lithography. Biomaterials 1999, 20, 2363–2376.

17. Patel, N.; Padera, B.; Sanders, G.H.W.; Cannizzaro,


S.M.; Davies, M.C.; Langer, R.; Roberts, C.J.; Tendler,
S.J.B.; Williams, P.M.; Shakesheff, K.M. Spatially
controlled cell engineering on biodegradable polymer
surfaces. FASEB J. 1998, 12, 1447–1454.

18. Mrksich, M.; Whitesides, G.M. Using self-assembled


monolayers to understand the interactions of man-made
surfaces with proteins and cells. Annu. Rev. Biophys.
Biomol. Struct. 1996, 155–178.

19. Chen, C.S.; Mrksich, M.; Huang, S.; Whitesides, G.M.;


Ingber, D.E. Micropatterned surfaces for control of cell
shape, position, and function. Biotechnol. Prog. 1998, 14,
356–363.

20. Kumar, A.; Whitesides, G. Features of gold having


micrometer to centimeter dimensions can be formed through a
combination of stamping with an elastomeric stamp and an
alkanethiol ink followed by chemical etching. Appl. Phys.
Lett. 1993, 63, 2002.
21. Folch, A.; Toner, M. Cellular micropatterns on
biocompatible materials. Biotechnol. Prog. 1998, 14,
388–392.

22. Raty, S.; Davis, J.A.; Beebe, D.J.; Rodriguez-Zas,


S.L.; Wheeler, M.B. Culture in microchannels enhances in
vitro embryonic development of preimplantation mouse
embryos. Theriogenology 2001, 55, 241.

23. Beebe, D.J.; Mensing, G.A.; Walker, G.M. Physics and


applications of microfluidics in biology. Annu. Rev. Biomed.
Eng. 2002, 4, 261–286.

24. Takayama, S.; Ostuni, E.; LeDue, P.; Naruse, K.;


Ingber, D.; Whitesides, G. Laminar flows: Subcellular
positioning of small molecules. Nature 2001, 411, 1016.

25. Takayama, S.; McDonald, J.C.; Ostuni, E.; Liang, M.N.;


Kenis, P.J.A.; Ismagilov, G.V.; Whitesides, G. Patterning
cells and their environments using multiple laminar fluid
flows in capillary networks. Proc. Natl. Acad. Sci. 1999,
96, 5545–5548.

26. Dertinger, S.K.; Jiang, X.; Li, Z.; Murthy, V.N.;


Whitesides, G.M. Gradients of substrate-bound laminin
orient axonal specification of neurons. Proc. Natl. Acad.
Sci. 2002, 99, 12542–12547. 27. Li Jeon, N.; Baskaran, H.;
Dertinger, S.K.; Whitesides, G.M.; Van de Water, L.; Toner,
M. Neutrophil chemotaxis in linear and complex gradients of
interleukin-8 formed in a microfabricated device. Natl.
Biotechnol. 2002, 20, 826–830. 28. Chiu, D.T.; Jeon, N.L.;
Huang, S.; Kane, R.S.; Wargo, C.J.; Choi, I.S.; Ingber,
D.E.; Whitesides, G.M. Patterned deposition of cells and
proteins onto surfaces by using three-dimensional
microfluidic systems. Proc. Natl. Acad. Sci. 2000, 97,
2408–2413. 29. Wang, N.; Ostuni, E.; Whitesides, G.M.;
Ingber, D.E. Micropatterning tractional forces in living
cells. Cell Motil. Cytoskelet. 2002, 52, 97–106. 30.
Curtis, A.; Wilkinson, C. Topographical control of cells.
Biomaterials 1997, 18, 1573–1583. 31. Deutsch, J.; Motlagh,
D.; Russell, B.; Desai, T.A. Fabrication of microtextured
membranes for cardiac myocyte attachment and orientation.
J. Biomed. Mater. Res. (Appl Biomater) 2000, 53, 267–275.
32. Brunette, D.M. Fibroblasts on micromachined substrata
orient hierarchically to grooves of different dimensions.
Exp. Cell Res. 1986, 164, 11–26. 33. Schmidt, J.A.; von
Recum, A.F. Texturing of polymer surfaces at the cellular
level. Biomaterials 1991, 12, 385–389. 34. Van Kooten,
T.G.; Whitesides, J.F.; von Recum, A.F. Influence of
silicone surface texture on human skin fibroblast
proliferation as determined by cell cycle analysis. J.
Biomed. Mater. Res. 1998, 43, 1–14. 35. Kapur, R.; Spargo,
B.J.; Chen, M.S.; Calvert, J.M.; Rudolph, A.S. Fabrication
and selective surface modification of 3-dimensionally
textured biomedical polymers from etched silicon
substrates. J. Biomed. Mater. Res. 1996, 33 (4), 205–216.
36. Den Braber, E.T.; de Ruijter, J.E.; Ginsel, L.A.; von
Recom, A.F.; Jansen, J.A. Orientation of ECM protein
deposition, fibroblasts cytoskeleton, and attachment complex
components on silicone microgrooved surfaces. J. Biomed.
Mater. Res. 1998, 40 (2), 291–300. 37. Meyle, J.; Gultig,
K.; Nisch, W. Variation in contact guidance by human cells
on a microstructured surface. J. Biomed. Mater. Res. 1995,
29 (1), 81–88. 38. Chou, L.; Firth, J.D.; Uitto, V.J.;
Brunette, D.M. Substratum surface topography alters cell
shape and regulates fibronectin mRNA level, mRNA stability,
secretion and assembly in human fibroblasts. J. Cell. Sci.
1995, 108 (4), 1563–1573. 39. Kapur, R.; Calvert, J.M.;
Rudolph, A.S. Electrical, chemical, and topological
addressing of mammalian cells with microfabricated systems.
J. Biomech. Eng. 1999, 121 (1), 65–72. 40. Flemming, R.G.;
Murphy, C.J.; Abrams, G.A.; Goodman, S.L.; Nealey, P.F.
Effects of synthetic microand nano-structured surfaces on
cell behavior. Biomaterials 1999, 20 (6), 573–588. 41.
Drumheller, P.; Hubbell, J. Surface Immobilization of
Adhesion Ligands for Investigations of Cell-Substrate
Interactions. In The Biomedical Engineering Handbook;
Bronzino, J., Ed.; CRC Press: Boca Raton, FL, 1995. 42.
Zhang, M.; Ferrari, M. Hemocompatible polyethylene glycol
films on silicon. Biomed. Microdevices 1998, 1, 81–89. 43.
Hubbell, J.A. Biomaterials in tissue engineering.
Biotechnology 1995, 13, 565–576. 44. Ruoslahti, E. RGD and
other recognition sequences for integrins. Annu. Rev. Cell
Dev. Biol. 1996, 12, 697–715. 45. Britland, S.; Clark, P.;
Connolly, P.; Moores, G. Micropatterned substratum
adhesiveness: A model for morphogenetic cues controlling
cell behaviour. Exp. Cell. Res. 1992, 198, 124–129. 46.
Aldenhoff, Y.B. Studies on a new strategy for surface
modification of polymeric biomaterials. J. Biomed. Mater.
1995, 29, 917–928. 47. Tseng, Y.C.; Park, K. Synthesis of
photoreactive poly(ethyleneglycol) and its application. J.
Biomed. Mater. Res. 1992, 26, 373–391. 48. Zhang, M.;
Desai, T.A.; Ferrari, M. Proteins and cells on PEG
immobilized silicon surfaces. Biomaterials 1998, 19. 49.
Sharma, S.; Popat, K.C.; Desai, T.A. Controlling nonspecific
protein interactions in silicon biomicrosystems with
nanostructured poly(ethylene glycol) films. Langmuir 2002,
18, 8728–8731. 50. Lanza, R.P.; Chick, W.L. Encapsulated
cell therapy. Sci. Am. Sci. Med. 1995, 16–25. 51. Colton,
C.K. Implantable biohybrid artificial organs. Cell
Transplant 1995, 4, 415–436. 52. Fu, X.W.; Sun, A.M.
Microencapsulated parathyroid cells as a bioartificial
parathyroid. In vivo studies. Transplantation 1989, 47,
432–435. 53. Aebischer, P.; Winn, S.R.; Galletti, P.M.
Transplantation of neural tissue in polymer capsules. Brain
Res. 1988, 448, 364–368. 54. Kaihara, S.; Borenstein, J.;
Koka, R.; Lalan, S.; Ochoa, E.R.; Ravens, M.; Pien, H.;
Cunningham, B.; Vacanti, J.P. Silicon micromachining to
tissue engineer branched vascular channels for liver
fabrication. Tissue Eng. 2000, 6, 105–117. 55. Desai, T.A.
Microfabricated interfaces: New approaches in tissue
engineering and biomolecular separation. Biomolecular
Engineering 2000, 17, 23–26. 56. Desai, T.A.
Microfabrication technology for pancreatic cell
encapsulation. Exp. Opin. 2002, 2, 633–646. 57. Leoni, L.;
Desai, T.A. Nanoporous biocapsules for the encapsulation of
insulinoma cells: Biotransport and biocompatibility
considerations. IEEE Trans. Biomed. Eng. 2001, 48 (11),
1335–1341. 58. Tan, W.; Krishnaraj, R.; Desai, T.A.
Evaluation of nanostructured composite collagen–chitosan
matrices for tissue engineering. Tissue Eng. 2001, 7,
203–211. 59. Motlagh, D.; Hartman, T.J.; Desai, T.A.;
Russell, B. Microfabricated grooves recapitulate neonatal
myocyte connexin43 and N-cadherin expression and
localization. Biomaterials 2002, in press. 60. Dike, L.E.;
Chen, C.S.; Mrksich, M.; Tien, J.; Whitesides, G.M.;
Ingber, D.E. Geometric control of switching between growth,
apoptosis, and differentiation during angiogenesis using
micropatterned substrates. In vitro Cell. Dev. Biol. Anim.
1999, 35, 441–448. 61. Tan, W.; Desai, T.A. Microfluidic
patterning of cellular biopolymer matrices for biomimetic
three-dimensional structures. Biom. Microdev. 2003, 5,
235–244. 62. Heiduschka, P.; Thanos, S. Implantable
bioelectric interfaces for lost nerve functions. Prog.
Neurobiol. 1998, 55, 433–461. 63. Branch, D.W.; Wheeler,
B.C.; Brewer, G.J.; Leckband, D.E. Long-term maintenance of
patterns of hippocampal pyramidal cells on substrates of
polyethylene glycol and microstamped polylysine. IEEE
Trans. Biom. Eng. 2000, 47, 290–300. 64. Rajnicek, A.;
Britland, S.; McCaig, C. Contact guidance of CNS neurites
on grooved quartz: Influence of groove dimensions, neuronal
age and cell type. J. Cell Sci. 1997, 110, 2905–2913. 65.
Lauer, L.; Ingebrandt, S.; Scholl, M.; Offenhausser, A.
Aligned microcontact printing of biomolecules on
microelectronic device surfaces. IEEE Trans. Biomed. Eng.
2001, 48, 838–842. 66. Pins, G.D.; Toner, M.; Morgan, J.R.
Microfabrication of an analog of the basal lamina:
Biocompatible membranes with complex topographies. FASEB J.
2000, 14, 593–602. 67. Saltzman, W.M.; Olbricht, W.L.
Building drug delivery into tissue engineering. Nat. Rev.
2002, 1, 177–186. 68. Hartgerink, J.D.; Beniash, E.; Stupp,
S.I. Self-assembly and mineralization of peptide–amphiphile
nanofibers. Science 2001, 294, 1684–1687. T
Tissue Engineering of Microvascular
Networks

4. Langer, R.; Vacanti, J.P. Tissue engineering. Science


1993, 260, 920.

5. Vacanti, J.P.; Langer, R. Tissue engineering: The design


and fabrication of living replacement devices for surgical
reconstruction and transplantation. Lancet 1999, 354 (Suppl
I), 32SI–34SI.

6. Bhatia, S.N.; Yarmush, M.L.; Toner, M. Controlling cell


interactions by micropatterning in co-cultures: Hepatocytes
and 3T3 fibroblasts. J. Biomed. Mater. Res. 1997, 34, 189.

7. Kaihara, S.; Borenstein, J.T.; Koka, R.; Lalan, S.;


Ochoa, E.R.; Ravens, M.; Pien, H.; Cunningham, B.; Vacanti,
J.P. Silicon micromachining to tissue engineer branched
vascular channels for liver fabrication. Tissue Eng. 2000,
6, 105.

8. Bell, E.; Ehrlich, P.; Buttle, D.J.; Nakatsuji, T.


Living tissue formed in vitro and accepted as
skin-equivalent of full thickness. Science 1981, 221,
1052–1054.

9. Parenteau, N.; Sabolinski, N.M.P.; Nolte, S.; Oleson,


C.; Kriwet, M. Biological and physical factors influencing
the successful engraftment of a cultured human skin
substitute. Biotechnol. Bioeng. 1996, 52, 3–14.

10. Niklason, L.E.; Gao, J.; Abbott, W.M. Functional


arteries grown in vivo. Science 1999, 284, 489–493.

11. Richardson, T.P.; Peters, M.C.; Ennett, A.B.; Mooney,


D.J. Polymeric system for dual growth factor delivery. Nat.
Biotechnol. 2001, 19, 1029.

12. Borenstein, J.T.; Terai, H.; King, K.R.; Weinberg,


E.J.; Kaazempur-Mofrad, M.R.; Vacanti, J.P.
Microfabrication technology for vascularized tissue
engineering. Biomed. Microdevices 2002, 4 (3), 167–175.

13. Kaazempur-Mofrad, M.R.; Vacanti, J.P.; Kamm, R.D.


Computational Modeling of Blood Flow and Rheology in
Fractal Microvascular Networks. In Computational Fluid and
Solid Mechanics; Bathe, K.J., Ed.; Elsevier Science, Ltd.:
Oxford, 2001; 864–867.

14. Kaazempur-Mofrad, M.R.; et al. . (to be published).


15. King, K.R.; Wang, C.C.; Shin, M.; Vacanti, J.P.;
Borenstein, J.T. Biodegradable Polymer Microfluidics for
Tissue Engineering Microvasculature, MRS Symp. Proc., MRS
Press, 2002; Vol. 729, U1.3.

16. Borenstein, J.T.; Cheung, W.; Hartman, L.;


Kaazempur-Mofrad, M.R.; King, K.R.; Sevy, A.; Shin, M.;
Weinberg, E.J. Living Three-Dimensional Microfabricated
Constructs For The Replacement Of Vital Organ Function;
IEEE MEMS: Boston, MA, 2003; 1754.

17. Guyton, A.C.; Hall, J.E. Textbook of Medical


Physiology; W.B. Saunders: Philadelphia, PA, 2000.

18. Kassab, G.S.; Lin, D.H.; Fung, Y.C. Morphometry of pig


coronary venous system. Am. J. Physiol. 1994, 267, H2100.

19. Murray, C.D. The physiological principle of minimum


work, I: The vascular system and the cost of blood volume.
Proc. Natl. Acad. Sci. 1926, 12, 207–214.

20. Weinberg, E.J.; Kaazempur-Mofrad, M.R.; Borenstein,


J.T. Numerical Model of Flow in Distensible Microfluidic
Network. In Computational Fluid and Solid Mechanics; Bathe,
K.J., Ed.; Elsevier Science, Ltd.: Oxford, 2003; Vol. 2,
1569–1572. 21. Gabriel, K.J. Microelectromechanical
systems. Proc. IEEE 1998, 86, 1534. 22. Borenstein, J.T.;
Gerrish, N.D.; Currie, M.T.; Fitzgerald, E.A. A New
Ultra-Hard Etch—Stop Layer for High Precision
Micromachining. In Technical Digest. IEEE International
MEMS 99 Conference. Twelfth IEEE International Conference
on Micro Electro Mechanical Systems; IEEE: Piscataway, NJ,
1999; 205. 23. Damji, A.; Weston, L.; Brunette, D.M.
Directed confrontations between fibroblasts and epithelial
cells on micromachined grooved substrata. Exp. Cell. Res.
1996, 228, 114. 24. Den Braber, E.T.; Ruijter, J.E.D.;
Ginsel, L.A.; Recum, A.F.V.; Jansen, J.A. Orientation of
ECM protein deposition, fibroblast cytoskeleton, and
attachment complex components on silicon microgrooved
surfaces. J. Biomed. Mater. Res. 1998, 40, 291. 25. Chen,
C.S.; Mrksich, M.; Huang, S.; Whitesides, G.M.; Ingber,
D.E. Geometric control of cell life and death. Science
1997, 276, 1425. 26. Curtis, A.; Wilkinson, C.
Topographical control of cells. Biomaterials 1997, 18,
1573. 27. Flemming, R.G.; Murphy, C.J.; Abrams, G.A.;
Goodman, S.L.; Nealey, P.F. Effects of synthetic microand
nano-structured surfaces on cell behavior. Biomaterials
1999, 20, 571. 28. McWhorter, P.J.; Frazier, A.B.;
Rai-Choudhury, P. Micromachining and Trends for the
Twenty-First Century. In Handbook of Microlithography,
Micromachining and Microfabrication; Rai-Choudhury, P.,
Ed.; SPIE Press: Bellingham, WA, 1997; Vol. 3. 29. Kendall,
D.L.; Malloy, K.J.; Fleddermann, C.B. Critical Technologies
for the Micromachining of Silicon. In Semiconductors and
Metals; Faber, K.T., Malloy, K.J., Eds.; Academic Press:
New York, 1992; Vol. 37, 193. 30. Madou, M. Fundamentals of
Microfabrication; CRC Press: Boca Raton, FL, 2002. 31.
Ayon, A.A.; Chen, K.-S.; Lohner, K.A.; Spearing, S.M.;
Sawin, H.H.; Schmidt, M.A. Deep reactive ion etching of
silicon. Mat. Res. Soc. Symp. Proc. 1999, 546, 51. 32.
Hynes, A.M.; Ashraf, H.; Bhardwaj, J.K.; Hopkins, J.;
Johnston, I.; Sheperd, J.N. Recent advances in silicon
etching for MEMS using the ASETM process. Sens. Actuators,
A 1999, 74, 13. 33. Jo, B.-H.; Beebe, D.J. Fabrication of
three-dimensional microfluidic systems by stacking molded
PDMS layers. SPIE 1999, 3877, 222. 34. Quake, S.R.;
Scherer, A. From microto nanofabrication with soft
materials. Science 2000, 290, 1536. 35. Whitesides, G.M.;
Stroock, A.D. Flexible methods for microfluidics. Phys.
Today 2001, 54, 42. 36. Thomson, R.C.; Yaszemski, M.J.;
Mikos, A.G. Polymer Scaffold Processing. In Principles of
Tissue Engineering; Lanza, R., Langer, R., Chick, W., Eds.;
Landes & Co., 1997. 37. Wang, Y.; Guillermo, A.; Ameer, A.;
Sheppard, J.; Langer, R. A tough biodegradable elastomer.
Nat. Biotechnol. 2002, 20, 602–606. 38. King, K.R.; Wang,
C.; Vacanti, J.P.; Borenstein, J.T. Submitted for
publication. 39. Davies, P.F. Flow-mediated endothelial
mechanotransduction. Physiol. Rev. 1995, 75, 519–560. 40.
Bhatia, S.N.; Chen, C.S. Tissue engineering at the
microscale. Biomed. Microdevices 1999, 2, 131–144. 41.
Griffith, L.G.; Naughton, G. Science 2002, 295, 1009–1014.
42. Poznansky, M.C.; Evans, R.H.; Foxall, R.B.; Olszak,
I.T.; Plascik, A.H.; Hartman, K.E.; Brander, C.; Meyer,
T.H.; Pykett, M.J.; Chabner, K.T.; Kalams, S.A.;
Rosenzwieg, M.; Scadden, D.T. Efficient generation of human
T cells from a tissue-engineered thymic organoid. Nat.
Biotechnol. 2000, 18, 729. 43. Webb, D.J.; Horwitz, A.F.
New dimensions in cell migration. Nat. Cell Biol. 2003, 5,
690. 44. LeClerc, E.; Sakai, Y.; Fujii, T. ‘‘Cell culture
in 3dimensional microfluidic structure of PDMS.’’ Biomed.
Microdevices. 2003, 5, 109–114. 45. Biomaterials for Tissue
Engineering; Wong, J.Y., Plant, A.L., Schmidt, C.E., Shea,
L., Coury, A.J., Chen, C.S., Eds.; MRS Press: Warrendale
PA, 2004; Vol. EXS-1, F3.5. T
Tissue Engineering of Pancreas

23. Assady, S.; Maor, G.; Amit, M.; Itskovitz-Eldor, J.;


Skorecki, K.L.; Tzukerman, M. Insulin production by human
embryonic stem cells. Diabetes 2001, 50 (8), 1691–1697.

24. Rajagopal, J.; Anderson, W.J.; Kume, S.; Martinez,


O.I.; Melton, D.A. Insulin staining of ES cell progeny from
insulin uptake. Science 2003, 299 (5605), 363.

25. Soria, B. In-vitro differentiation of pancreatic


betacells. Differentiation 2001, 68 (4–5), 205–219.

26. Bonner-Weir, S.; Sharma, A. Pancreatic stem cells. J.


Pathol. 2002, 197 (4), 519–526. Review.

27. Ramiya, V.K.; Maraist, M.; Arfors, KE.; Schatz, D.A.;


Peck, A.B.; Cornelius, J.G. Reversal of insulin-dependent
diabetes using islets generated in vitro from pancreatic
stem cells. Nat. Med. 2000, 6 (3), 278–282.

28. Bonner-Weir, S.; Taneja, M.; Weir, G.C.; Tatarkiewicz,


K.; Song, K.H.; Sharma, A.; O’Neil, J.J. In vitro
cultivation of human islets from expanded ductal tissue.
Proc. Natl. Acad. Sci. U. S. A. 2000, 97 (14), 7999–8004.

29. Chen, R.; Meseck, M.; McEvoy, R.C.; Woo, S.L.


Glucose-stimulated and self-limiting insulin production by
glucose 6-phosphatase promoter driven insulin expression in
hepatoma cells. Gene Ther. 2000, 7 (21), 1802–1809.

30. Thule, P.M.; Liu, J.; Phillips, L.S. Glucose regulated


production of human insulin in rat hepatocytes. Gene Ther.
2000, 7 (3), 205–214.

31. Cheung, A.T.; Dayanandan, B.; Lewis, J.T.; Korbutt,


G.S.; Rajotte, R.V.; Bryer-Ash, M.; Boylan, M.O.; Wolfe,
M.M.; Kieffer, T.J. Glucose-dependent insulin release from
genetically engineered K cells. Science 2000, 290 (5498),
1959–1962. 32. Tang, S.C.; Sambanis, A. Development of
genetically engineered human intestinal cells for regulated
insulin secretion using rAAV-mediated gene transfer.
Biochem. Biophys. Res. Commun. 2003, 303 (2), 645–652. 33.
Ryan, E.A.; Lakey, R.T.; Rajotte, R.V.; Korbutt, G.S.; Kin,
T.; Imes, S.; Rabinovitch, A.; Elliot, J.F.; Bigam, D.;
Kneteman, N.M.; Warnock, G.L.; Larsen, I.; Shapiro, A.M.J.
Clinical outcomes and insulin secretion after islet
transplantation with the edmonton protocol. Diabetes 1999,
50, 710–719. 34. Soon-Shiong, P.; Heintz, R.E.; Merideth,
N.; Yao, Q.X.; Yao, Z.; Zheng, T.; Murphy, M.; Moloney,
M.K.; Schmehl, M.; Harris, M.; Mendez, R.; Mendez, R.;
Sandford, P.A. Insulin independence in a type 1 diabetic
patient after encapsulated islet transplantation. Lancet
1994, 343 (8903), 950–951. 35. Scharp, D.W.; Swanson, C.J.;
Olack, B.J.; Latta, P.P.; Hegre, O.D.; Doherty, E.J.;
Gentile, F.T.; Flavin, K.S.; Ansara, M.F.; Lacy, P.E.
Protection of encapsulated human islets implanted without
immunosuppression in patients with type I or type II
diabetes and in nondiabetic control subjects. Diabetes
1994, 43 (9), 1167–1170. 36. Buchser, E.; Goddard, M.;
Heyd, B.; Joseph, J.M.; Favre, J.; de Tribolet, N.;
Lysaght, M.; Aebischer, P. Immunoisolated xenogenic
chromaffin cell therapy for chronic pain. Initial clinical
experience. Anesthesiology 1996, 85 (5), 1005–1012. 37.
Emerich, D.F.; Winn, S.R. Immunoisolation cell therapy for
CNS diseases. Crit. Rev. Ther. Drug Carr. Syst. 2001, 18
(3), 265–298. 38. Zielinski, B.A.; Lysaght, M.J.
Immunoisolation. In Principles of Tissue Engineering, 2nd
Ed.; Lanza, R.P., Langer, R., Vacanti, J., Eds.; Academic
Press: San Diego, CA, 2000; 321–329.
Tissue Engineering of Peripheral Nerve

10. Fugleholm, K.; Schmalbruch, H.; Krarup, C. Post


reinnervation maturation of myelinated nerve fibers in the
cat tibial nerve: Chronic electrophysiological and
morphometric studies. J. Peripher. Nerv. Syst. 2000, 5,
82–95.

11. Varejao, A.S.P.; Meek, M.F.; Ferreira, A.J.A.;


Patricio, J.A.B.; Cabrita, A.M.S. Functional evaluation of
peripheral nerve regeneration in the rat: Walking track
analysis. J. Neurosci. Methods 2001, 108, 1–9.

12. Kanaya, F.; Firrell, J.C.; Breidenbach, W.C. Sciatic


function index, nerve conduction tests, muscle contraction,
and axon morphometry as indicators of regeneration. Plast.
Reconstr. Surg. 1996, 98, 1264–1271.

13. Mackinnon, S.E. Sciatic function index, nerve


conduction tests, muscle contraction, and axon morphometry
as indicators of regeneration—Discussion. Plast. Reconstr.
Surg. 1996, 98, 1272–1274.

14. De Medinaceli, L.; Freed, W.J.; Wyatt, R.J. An index of


the functional condition of rat sciatic nerve based on
measurements made from walking tracks. Exp. Neurol. 1982,
77, 634–643.

15. Dijkstra, J.R.; Meek, M.F.; Robinson, P.H.;


Gramsbergen, A. Methods to evaluate functional nerve
recovery in adult rats: Walking track analysis, video
analysis and the withdrawal reflex. J. Neurosci. Methods
2000, 96, 89–96.

16. Johnston, R.B.; Zachary, L.; Dellon, A.L.; Seiler,


W.A.; Teplica, D.M. Improved imaging of rat hind foot
prints for walking track analysis. J. Neurosci. Methods
1991, 38, 111–114.

17. Hudson, T.; Evans, G.R.D.; Schmidt, C. Engineering


strategies for peripheral nerve repair. Clin. Plast. Surg.
1999, 26 (4), 617–628.

18. Seal, B.L.; Otero, T.C.; Panitch, A. Review: Polymeric


biomaterials for tissue and organ regeneration. Mater. Sci.
Eng. 2001, 34 (4), 147–230.

19. Evans, G.R.D. Peripheral nerve injury: A review and


approach to tissue engineering constructs. Anat. Rec. 2001,
263, 396–404.
20. Danielsen, N.; Dahlin, L.B.; Thomsen, P. Inflammatory
cells and mediators in the silicone chamber model for nerve
regeneration. Biomaterials 1993, 14 (15), 1180– 1185.

21. Hadlock, T.; Elisseeff, J.; Langer, R.; Vacanti, J.;


Cheney, M. A tissue-engineered conduit for peripheral nerve
repair. Arch. Otolaryngol. Head Neck Surg. 1998, 124 (10),
1081–1086.

22. Lavik, E.B.; Hrkach, J.S.; Lotan, N.; Nazarov, R.;


Langer, R. A simple synthetic route to the formation of a
block copolymer of poly(lactic-co-glycolic acid) polylysine
for the fabrication of functionalized, degradable
structures for biomedical applications. J. Biomed. Mater.
Res. 2001, 58 (3), 291–294.

23. Valentini, R.F.; Sabatini, A.M.; Dario, P.; Aebischer,


P. Polymer electret guidance channels enhance peripheral
nerve regeneration in mice. Brain Res. 1989, 48, 300–304.

24. Langone, F.; Lora, S.; Veronese, F.M.; Caliceti, P.;


Parnigotto, P.P.; Valenti, F.; Palma, G. Peripheral nerve
repair using a poly(organo)phosphazene tubular prosthesis.
Biomaterials 1995, 6 (5), 347–353. 25. den Dunnen, W.F.;
van der Lei, B.; Schakenraad, J.M.; Stokroos, I.; Blaauw,
E.; Bartels, H.; Pennings, A.J.; Robinson, P.H.
Poly(DL-lactide-epsilon-capralactone) nerve guides perform
better than autologous nerve grafts. Microsurgery 1996, 17
(7), 348–357. 26. Aebischer, P.; Valentini, R.F.; Dario,
P.; Domenici, C.; Galleti, P.M. Piezoelectric guidance
channels enhance regeneration in the mouse sciatic nerve
after axotomy. Brain Res. 1987, 435, 165–168. 27. Molander,
H.; Engkvist, O.; Hagglund, J.; Olsson, Y.; Torebjork, E.
Nerve repair using a polyglactin tube and nerve graft: An
experimental study in the rabbit. Biomaterials 1983, 4,
276–280. 28. Nyilas, E.; Chiu, T.H.; Sidman, R.L.; Henry,
E.W.; Brushart, T.M.; Dikkes, P.; Madison, R. Peripheral
nerve repair with bioresorbable prosthesis. Trans. Trans.
Am. Soc. Artif. Int. Organs 1983, 29, 307–313. 29. Seckel,
B.R.; Chiu, T.H.; Nyilas, E.; Sidman, R.L. Nerve
regeneration through synthetic biodegradable nerve guides:
Regulation by the target organ. Plast. Reconst. Surg. 1984,
74 (2), 173–181. 30. Labrador, R.O.; Buti, M.; Navarro, X.
Peripheral nerve repair: Role of agarose matrix density on
functional recovery. Neuroreport 1995, 6 (15), 2022–2026.
31. Madison, R.D.; Da Silva, C.F.; Dikkes, P. Peripheral
nerve repair: Role of agarose matrix density on functional
recovery. Brain Res. 1988, 447 (2), 325–334. 32. Williams,
L.R.; Longo, F.M.; Powell, H.C.; Lundborg, G.; Varon, S.
Spatial-temporal progress of peripheral nerve regeneration
within a silicone chamber: Parameters for a bioassay. J.
Comp. Neurol. 1983, 218, 460–470. 33. Li, S.T. A
Multi-Layered, Semipermeable Conduit for Nerve Regeneration
Comprised of Type I Collagen, Its Method of Manufacture and
a Method of Nerve Regeneration Using Said Conduit. U.S.
Patent No. 4,963,146. 34. Archibald, S.J.; Shefner, J.;
Krarup, C.; Madison, R.D. Monkey median nerve repaired by
nerve graft or collagen nerve guide tube. J. Neurosci.
1995, 15 (5), 4109–4123. 35. Lundborg, G. A 25-year
perspective of peripheral nerve surgery: Evolving
neuroscientific concepts and clinical significance. J. Hand
Surg. Am. 2000, 25 (3), 391–414. 36. Lundborg, G.; Rosen,
B.; Dahlin, L.; Danielsen, N.; Holmberg, J. Tubular versus
conventional repair of median and ulnar nerves in the human
forearm: Early results from a prospective, randomized,
clinical study. J. Hand Surg. 1997, 22A, 99–106. 37. Hall,
S.M. Regeneration in cellular and acellular autografts in
the peripheral nervous system. Neuropathol. Appl.
Neurobiol. 1986, 12 (1), 27–46. 38. Heath, C.A.; Rutkowski,
G.E. The development of bioartificial nerve grafts for
peripheral nerve regeneration. TIBTECH 1998, 16, 163–168.
39. Young, C.; Miller, E.; Nicklous, D.M.; Hoffman, J.R.
Nerve growth factor and neurotrophin-3 affect functional
recovery following peripheral nerve injury differently.
Restor. Neurol. Neurosci. 2001, 18 (4), 167–175. 40. Verdu,
E.; Labrador, R.O.; Rodriguez, F.J.; Ceballos, D.; Fores,
J.; Navarro, X. Alignment of collagen and
laminin-containing gels improve nerve regeneration within
silicone tubes. Restor. Neurol. Neurosci. 2002, 20 (5),
169–180. 41. Ahmed, Z.; Brown, R.A.; Ljungberg, C.; Wiberg,
M.; Terenghi, G. Nerve growth factor enhances peripheral
nerve regeneration in non-human primates. Scand. J. Plast.
Reconstr. Surg. Hand Surg. 1999, 33 (4), 393–401. 42.
Manthorpe, M.; Engvall, E.; Ruoslahti, E.; Longo, F.M.;
Davis, G.E.; Varon, S. Laminin promotes neuritic
regeneration from cultured peripheral and central neurons.
J. Cell Biol. 1983, 97, 1882–1890. 43. Ceballos, D.;
Navarro, X.; Dubey, N.; WendelschaferCrabb, G.; Kennedy,
W.R.; Tranquillo, R.T. Magnetically aligned collagen gel
filling a collagen nerve guide improves peripheral nerve
regeneration. Exp. Neurol. 1999, 158, 290–300. 44. Patel,
N.; Bhandari, R.; Shakesheff, K.M.; Cannizzaro, S.M.;
Davies, M.C.; Langer, R.; Roberts, C.J.; Tendler, S.J.;
Williams, P.M. Printing patterns of biospecificallyadsorbed
protein. J. Biomater. Sci., Polym. Ed. 2000, 11 (3),
319–331. 45. Bonner, J.; O’Connor, T.P. The permissive cue
laminin is essential for growth cone turning in vivo. J.
Neurosci. 2001, 21 (24), 9782–9791. 46. Hammarback, J.A.;
Palm, L.S.; Furcht, L.T.; Letourneau, P.C. Guidance of
neurite outgrowth by pathways of substratum-adsorbed
laminin. J. Neurosci. Res. 1985, 13, 213–220. 47. Clark,
P.; Britland, S.; Connolly, P. Growth cone guidance and
neuron morphology on micropatterned laminin surfaces. J.
Cell Sci. 1993, 105, 203–212. 48. Saneinejad, S.; Shoichet,
S.M. Patterned glass surfaces direct cell adhesion and
process outgrowth of primary neurons of the central nervous
system. J. Biomed. Mater. Res. 1998, 42, 13–19. 49. Patel,
N.; Padera, R.; Giles, S.H.W.; Cannizzaro, S.M.; Davies,
M.C.; Langer, R.; Roberts, C.J.; Tendler, S.J.; Williams,
P.M.; Shakesheff, K.M. Spatially controlled cell
engineering on biodegradable polymer surfaces. FABES J.
1998, 12, 1447–1454. 50. Wheeler, B.C.; Corey, J.M.;
Brewer, G.J.; Branch, D.W. Microcontact printing for
precise control of nerve cell growth in culture. J.
Biomech. Eng. 1999, 121, 73–78. 51. Cuevas, P.; Carceller,
F.; Dujovny, M.; Garcia-Gomez, I.; Cuevas, B.;
Gonzalez-Corrochano, R.; Diaz-Gonzalez, D.; Reimers, D.
Peripheral nerve regeneration by bone marrow stromal cells.
Neurol. Res. 2002, 24 (7), 634–638. 52. Navarro, J.;
Oudrhiri, N.; Fabrega, S.; Lehn, P. Gene delivery systems:
Bridging the gap between recombinant viruses and artificial
vectors. Adv. Drug Deliv. Rev. 1998, 30, 5–11. 53. Gravel,
C.; Gotz, R.; Lorrain, A.; Sendtner, M. Adenoviral gene
transfer of ciliary neurotrophic factor and brain-derived
neurotrophic factor leads to long-term survival of
axotomized motor neurons. Nat. Med. 1997, 3, 765–770. 54.
Baumgartner, B.J.; Shine, H.D. Neuroprotection of spinal
motorneurons following targeted transduction with an
adenoviral vector carrying the gene for glial cell
line-derived neurotrophic factor. Exp. Neurol. 1998, 153,
102–112. 55. Gimenez, Y.R.M.; Revah, F.; Pradier, L.;
Loquet, I.; Mallet, A.P. Prevention of motorneuron death by
adenovirus-mediated neurotrophic factors. J. Neurosci. Res.
1997, 48, 281–285. 56. Lemarchand, P.; Jaffe, H.A.; Danel,
C.; Cid, M.C.; Kleinman, H.K.; Stratford-Perricaudet, L.D.;
Perricaudet, M.; Pavirani, A.; Lecocq, J.; Crystal, R.G.
Adenovirus-mediated transfer of a recombinant human a 1
-antitrypsin cDNA to human endothelial cells. PNAS 1992,
89, 6482–6486. 57. Harris, J.D.; Lemoine, N.R. Strategies
for targeted gene therapy. Trends Genet. 1996, 12, 400–405.
58. Miller, N.; Vile, R. Vectors for gene therapy. FASEB J.
1995, 190–199. 59. Verma, I.M.; Somia, N. Gene
therapy–promises, problems and prospects. Nature 1997, 389,
239–242. 60. Wim, T.J.; Hermens, M.C.; Verhaagen, J. Viral
vectors, tools for gene transfer in the nervous system.
Prog. Neurobiol. 1998, 55, 399–432. 61. Patrick, C.W., Jr.;
Zheng, B.; Wu, X.; Gurtner, G.; Barlow, M.; Koutz, C.;
Chang, D.; Schmidt, M.; Evans, G.R. Muristerone A-induced
nerve growth factor release from genetically engineered
human dermal fibroblasts for peripheral nerve tissue
engineering. Tissue Eng. 2001, 7 (3), 303–311. T
Tissue Engineering of Rotator Cuff Tendons

1. Carpenter, J.E.; Thomopoulos, S.; Flanagan, C.L.;


DeBano, C.M.; Soslowsky, L.J. Rotator cuff defect healing:
A biomechanical and histologic analysis in an animal model.
J. Shoulder Elb. Surg. 1998, 7, 599–605.

2. Butler, D.L.; Goldstein, S.A.; Guilak, F. Functional


tissue engineering: The role of biomechanics. J. Biomech.
Eng. 2000, 122, 570–575.

3. Itoi, E.; Berglund, L.J.; Grabowski, J.J.; Schultz,


F.M.; Growney, E.S.; Morrey, B.F.; An, K.N. Tensile
properties of the supraspinatus tendon. J. Orthop. Res.
1995, 13, 578–584.

4. Halder, A.; Zobitz, M.E.; Schultz, F.; An, K.N.


Mechanical properties of the posterior rotator cuff. Clin.
Biomech. 2000, 15, 456–462.

5. Halder, A.; Zobitz, M.E.; Schultz, F.; An, K.N.


Structural properties of the subscapularis tendon. J.
Orthop. Res. 2000, 18, 829–834.

6. Guilak, F. Functional tissue engineering: The role of


biomechanics in reparative medicine. Ann. N.Y. Acad. Sci.
2002, 961, 193–195.

7. Hyman, J.; Rodeo, S.A. Injury and repair of tendons and


ligaments. Phys. Med. Rehabil. Clin. North Am. 2000, 11
(2), 267–288.

8. Bank, R.A.; TeKoppele, J.M.; Oostingh, G.; Hazleman,


B.L.; Riley, G.P. Lysylhydroxylation and non-reducible
crosslinking of human supraspinatus tendon collagen:
Changes with age in chronic rotator cuff tendonitis. Ann.
Rheum. Dis. 1999, 58 (1), 35–41.

9. Riley, G.P.; Harrall, R.L.; Constant, C.R.; Chard, M.D.;


Cawston, T.E.; Hazleman, B.L. Tendon degeneration and
chronic shoulder pain: Changes in the collagen composition
of the human rotator cuff tendons in rotator cuff
tendonitis. Ann. Rheum. Dis. 1994, 53, 359–366.

10. Woo, S.L.Y.; Hildebrand, K.; Watanabe, N.; Fenwick,


J.A.; Papageorgiou, C.D.; Wang, J.H.C. Tissue engineering
of ligament and tendon healing. Clin. Orthop. 1999, 367S,
S312–S323.

11. Potenza, A.D. Tendon healing within the flexor digital


sheath in the dog: An experimental study. J. Bone Jt. Surg.
[Am.] 1962, 44, 49–64.

12. Gelberman, R.H.; Manske, P.R.; Akeson, W.H.; Woo,


S.L.Y.; Lundborg, G.; Amiel, D. Flexor tendon repair. J.
Orthop. Res. 1986, 4, 119–128.

13. Glaser, D.L.; Ramachandran, R.; Shore, E.M.; Lin, T.W.;


Yeh, P.C.; Beredjiklian, P.; Kaplan, F.S.; Goldhammer,
D.J.; Soslowsky, L.J. The Origin of Cells Within a Healing
Tendon. In Transactions of the Orthopaedic Research
Society, 49th Annual Meeting, New Orleans, Feb. 1–5, 2003.

14. Lin, T.W.; Cardenas, L.; Glaser, D.L.; Soslowsky, L.J.


Tendon Healing in Interleukin-6 Knockout Mice. In
Transactions of the Orthopaedic Research Society, 49th
Annual Meeting, New Orleans, Feb. 1–5, 2003.

15. Thomopoulos, S.; Hattersley, G.; Rosen, V.; Mertens,


M.; Galatz, L.; Williams, G.R.; Soslowsky, L.J. The
localized expression of extracellular matrix components in
healing tendon insertion sites: An in situ hybridization
study. J. Orthop. Res. 2002, 20 (3), 454–463. 16. Woo,
S.L.Y.; Maynard, J.; Butler, D.; Lyon, R.; Torzilli, P.;
Akeson, W.; Cooper, R.; Oakes, B. Ligament, Tendon, and
Joint Capsule Insertions to Bone. In Injury and Repair of
the Musculoskeletal Soft Tissues; Woo, S.L.Y., Buckwalter,
J.A., Eds.; American Academy of Orthopaedic Surgeons:
Savannah, 1987; 129–166. 17. Gelberman, R.H.; Nunley, J.A.,
II; Osterman, A.L.; Breen, T.F.; Dimick, M.P.; Woo, S.L.
Influences of the protected passive mobilization interval on
flexor tendon healing. A prospective randomized clinical
trial. Clin. Orthop. 1991, 264, 189–196. 18. Reichmister,
J.P.; Friedman, S.L. Long-term functional results after
manipulation of the frozen shoulder. Md. Med. J. 1999, 48
(1), 7–11. 19. Strickland, J.W. Development of flexor tendon
surgery: Twenty-five years of progress. J. Hand Surg. [Am.]
2000, 25, 214–235. 20. Lastayo, P.C.; Wright, T.; Jaffe,
R.; Hartzel, J. Continuous passive motion after repair of
the rotator cuff. A prospective outcome study. J. Bone Jt.
Surg. [Am.] 1998, 80 (7), 1002–1011. 21. Thomopoulos, S.;
Williams, G.R.; Soslowsky, L.J. Tendon to bone healing:
Differences in biomechanical, structural, and compositional
properties due to a range of activity levels. J. Biomech.
Eng. 2003, 125 (1), 106–113. 22. Gomez, M.A.; Woo, S.L.;
Amiel, D.; Harwood, F.; Kitabayashi, L.; Matyas, J.R. The
effects of increased tension on healing medial collateral
ligaments. Am. J. Sports Med. 1991, 19 (4), 347–354. 23.
Liechty, K.W.; MacKenzie, T.C.; Shaaban, A.F.; Radu, A.;
Moseley, A.B.; Deans, R.; Marshak, D.R.; Flake, A.W. Human
mesenchymal stem cells engraft and demonstrate site-specific
differentiation after in utero transplantation in sheep.
Nat. Med. 2000, 6 (11), 1282–1286. 24. Prockop, D.J. Marrow
stromal cells as stem cells for nonhematopoietic tissues.
Science 1997, 276, 71–74. 25. Lazarus, H.M.; Haynesworth,
S.E.; Gerson, S.L.; Rosenthal, N.S.; Caplan, A.I. Ex vivo
expansion and subsequent infusion of human bone
marrow-derived stromal progenitor cells (mesenchymal
progenitor cells): Implication for therapeutic use. Bone
Marrow Transplant. 1995, 16, 557–564. 26. Butler, D.L.;
Awad, H.A. Perspectives on cell and collagen composites for
tendon repair. Clin. Orthop. 1999, 367S, S324–S332. 27.
Awad, H.A.; Butler, D.L.; Boivin, G.P.; Smith, F.N.;
Malaviya, P.; Huibregtse, B.; Caplan, A.I. Autologous
mesenchymal stem cell-mediated repair of tendon. Tissue
Eng. 1999, 5, 267–277. 28. Young, R.G.; Butler, D.L.;
Weber, W.; Caplan, A.I.; Gordon, S.L.; Fink, D.J. Use of
mesenchymal stem cells in a collagen matrix for Achilles
tendon repair. J. Orthop. Res. 1998, 16, 406–413. 29.
Hildebrand, K.A.; Jia, F.; Woo, S.L. Response of donor and
recipient cells after transplantation of cells to the
ligament and tendon. Microsc. Res. Tech. 2002, 58, 34–38.
30. Dejardin, L.M.; Arnoczky, S.P.; Ewers, B.J.; Haut,
R.C.; Clarke, R.B. Tissue-engineered rotator cuff tendon
using porcine small intestine submucosa: Histologic and
mechanical evaluation in dogs. Am. J. Sports Med. 2001, 29
(2), 175–184. 31. Dejardin, L.M.; Arnoczky, S.P.; Clarke,
R.B. Use of small intestinal submucosal implants for
regeneration of large fascial defects: An experimental
study in dogs. J. Biomed. Mater. Res. 1999, 46, 203–211.
32. Aiken, S.W.; Badylak, S.F.; Toombs, J.P.; Shelbourne,
K.D.; Hiles, M.C.; Lantz, G.C.; van Sickle, D. Small
intestinal submucosa as an intra-articular ligamentous
graft material: A pilot study in dogs. Vet. Comp. Orthop.
Traumatol. 1994, 7, 124–128. 33. Badylak, S.F.; Tullius,
R.; Kokini, K.; Shelbourne, K.D.; Klootwyk, T.; Voytik,
S.L.; Kraine, M.R.; Simmons, C. The use of xenogeneic small
intestinal submucosa as a biomaterial for Achilles tendon
repair in a dog model. J. Biomed. Mater. Res. 1995, 29,
977–985. 34. Hodde, J.P.; Badylak, S.F.; Shelbourne, K.D.
The effect of range of motion on remodeling of small
intestinal submucosa (SIS) when used as an Achilles tendon
repair material in the rabbit. Tissue Eng. 1997, 3 (1),
27–37. 35. Chang, J.; Most, D.; Stelnicki, E.; Siebert,
J.W.; Longaker, M.T.; Hui, K.; Lineweaver, W.C. Gene
expression of transforming growth factor beta-1 in rabbit
zone II flexor tendon wound healing: Evidence for dual
mechanisms of repair. Plast. Reconstr. Surg. 1997, 100 (4),
937–944. 36. Abrahamsson, S.O.; Lundborg, G.; Lohmander,
L.S. Recombinant human insulin-like growth factor-I
stimulates in vitro matrix synthesis and cell proliferation
in rabbit flexor tendon. J. Orthop. Res. 1991, 9, 495–502.
37. Duffy, F.J., Jr.; Seiler, J.G.; Gelberman, R.H.;
Hergrueter, C.A. Growth factors and canine flexor tendon
healing: Initial studies in uninjured and repair models. J.
Hand Surg. [Am.] 1995, 20, 645–649. 38. Stein, L.E. Effects
of serum, fibroblast growth factor, and platelet-derived
growth factor on explants of rat tail tendon: A
morphological study. Acta Anat. 1985, 123, 247–252. 39.
Evans, C.H.; Robbins, P.D. Genetically augmented tissue
engineering of the musculoskeletal system. Clin. Orthop.
1999, 367S, S410–S418. 40. Gerich, T.G.; Kang, R.; Fu,
F.H.; Robbins, P.D.; Evans, C.H. Gene transfer to the
rabbit patellar tendon: Potential for genetic enhancement
of tendon and ligament healing. Gene Ther. 1996, 3,
1089–1093. 41. Lou, J.; Manske, P.R.; Aoki, M.; Joyce, M.E.
Adenovirus-mediated gene transfer into tendon and tendon
sheath. J. Orthop. Res. 1996, 14, 513–517. T
Tissue Engineering Scaffolds

3. How, T.V.; Guidoin, R.; Young, S.K. Engineering design


of vascular prostheses. Proc. Inst. Mech. Eng. 1992, 206,
61–71.

4. Vacanti, J.P.; Vacanti, C.A. The Challenge of Tissue


Engineering. In Principles of Tissue Engineering; Lanza,
R., Langer, R., Chick, W., Eds.; Academic Press, 1997; 1–5.

5. Mooney, D.J.; Langer, R.S. Engineering Biomaterials for


Tissue Engineering: The 10–100 Micron Size Scale. In The
Biomedical Engineering Handbook; CRC Press, 1995;
1609–1618.

6. Starke, G.R.; Douglas, A.S.; Conway, D.J. An Integral


Mathematical Approach to Tissue Engineering of Vascular
Grafts. In Tissue Engineering of Prosthetic Vascular
Grafts; Zilla, P., Greisler, H.P., Eds.; R.G. Landes
Company, 1999; 441–459.

7. Hsu, S.; Kambic, H. On matching compliance between


canine carotid arteries and polyurethane grafts. Artif.
Organs 1997, 21, 1247–1254.

8. Hynes, R.O. Integrins: Bidirectional, allosteric


signaling machines. Cell 2002, 110, 673–687.

9. Olsen, B.R. Matrix Molecules and Their Ligands. In


Principles of Tissue Engineering; Lanza, R., Langer, R.,
Chick, W., Eds.; R.D. Landes Co., 1997; 47–65.

10. Mikos, A.G.; Bao, Y.; Cima, L.G.; Ingber, D.E.;


Vacanti, J.P.; Langer, R.S. Preparation of poly(glycolic
acid) bonded fiber structures for cell attachment and
transplantation. J. Biomed. Mater. Res. 1993, 27, 183–189.

11. Mooney, D.J.; Mazzoni, C.L.; Breuer, C.; McNamara, K.;


Hern, D.; Vacanti, J.P.; Langer, R.S. Stabilized
polyglycolic acid fiber-based tubes for tissue engineering.
Biomaterials 1996, 17 (2), 115–124.

12. Mikos, A.G.; Lyman, M.D.; Freed, L.E.; Langer, R.S.


Wetting of poly(L-lactic acid) and
poly(DL-lacticco-glycolic acid) foams for tissue culture.
Biomaterials 1994, 15 (1), 55–58.

13. Thomson, R.C.; Yaszemski, M.J.; Powers, J.M.; Mikos,


A.G. Hydroxyapatite fiber reinforced poly(a-hydrosy ester)
foams for bone regeneration. Biomaterials 1998, 19,
1935–1943.

14. Shastri, V.P.; Martin, I.; Langer, R.S. Macroporous


polymer foams by hydrocarbon templating. Proc. Natl. Acad.
Sci. 2000, 97, 1970–1975.

15. Wake, M.C.; Gupta, P.K.; Mikos, A.G. Fabrication of


pliable biodegradable polymer foams to engineer soft
tissues. Cell Transplant 1996, 5 (4), 465–473.

16. Mikos, A.G.; Sarakinos, G.; Leite, S.M.; Vacanti, J.P.;


Langer, R.S. Laminated three-dimensional biodegradable
foams for use in tissue engineering. Biomaterials 1993, 14
(5), 323–328.

17. Thomson, R.C.; Yaszemski, M.J.; Powers, J.M.; Mikos,


A.G. Fabrication of biodegradable polymer scaffolds to
engineer trabecular bone. J. Biomater. Sci., Polym. Ed.
1995, 7, 23–28.

18. Widmer, M.S.; Gupta, P.K.; Lu, L.; Meszlenyi, R.K.;


Evans, G.R.D.; Brandt, K.; Savel, T.; Gurlek, A.; Patrick,
C.W., Jr.; Mikos, A.G. Manufacture of porous biodegradable
polymer conduits by an extrusion process for guided tissue
regeneration. Biomaterials 1998, 19 , 1945–1955. 19.
Mooney, D.J.; Baldwin, D.F.; Suh, N.P.; Vacanti, J.P.;
Langer, R.S. Novel approach to fabricate porous sponges of
poly(D,L-lactic-co-glycolic acid) without the use of
organic solvents. Biomaterials 1996, 17, 1417–1422. 20.
Harris, L.D.; Kim, B.S.; Mooney, D.J. Open pore
biodegradable matrices formed with gas foaming. J. Biomed.
Mater. Res. 1998, 42 (3), 396–402. 21. Nam, Y.S.; Yoon,
J.J.; Park, T.G. A novel fabrication method of macroporous
biodegradable polymer scaffolds using gas foaming salt as a
porogen additive. J. Biomed. Mater. Res. 2000, 53 (1), 1–7.
22. Whang, K.; Thomas, C.H.; Healy, K.E. A novel method to
fabricate bioabsorbable scaffolds. Polymer 1995, 36 (4),
837–842. 23. Hsu, Y.Y.; Gresser, J.D.; Trantolo, D.J.;
Lyons, C.M.; Gangadharam, P.R.J.; Wise, D.L. Effect of
polymer foam morphology and density on kinetics of in vitro
controlled release of isoniazid from compressed foam
matrices. J. Biomed. Mater. Res. 1997, 35, 107–116. 24. Lo,
H.; Guggino, S.E.; Leong, K.W. Poly(L-lactic acid) foams
with cell seeding and controlled-release capacity. J.
Biomed. Mater. Res. 1996, 30, 475–484. 25. Zhang, R.; Ma,
P.X. Poly(a-hydroxyl acids)/hydroxyapatite porous
composites for bone-tissue engineering. I. Preparation and
morphology. J. Biomed. Mater. Res. 1999, 44, 446–455. 26.
Reddy, P.P.; Barrieras, D.J.; Wilson, G.; Bagli, D.J.;
McLorie, G.A.; Khoury, A.E.; Merguerian, P.A. Regeneration
of functional bladder substitutes using large segment
acellular matrix allografts in a porcine model. J. Urol.
2000, 164, 936–941. 27. Badylak, S.F. The extracellular
matrix as a scaffold for tissue reconstruction. Semin. Cell
Dev. Biol. 2002, 13 (5), 377–383. 28. Badylak, S.F.; Lantz,
G.C.; Coffey, A.; Geddes, L.A. Small intestine submucosa as
a large diameter vascular graft in the dog. J. Surg. Res.
1989, 47, 74–80. 29. Yoo, J.J.; Meng, J.; Oberpenning, F.;
Atala, A. Bladder augmentation using allogenic bladder
submucosa seeded with cells. Urology 1998, 51 (2), 221–225.
30. Lee, H.B.; Lew, D.H. De novo induction of island
capsule flap by using two silastic sheets: Part 1.
Generation. Plast. Reconstr. Surg. 1996, 104, 1023–1028.
31. Schoeller, T.; Lille, S.; Stenzl, A.; Ninkovic, M.;
Piza, H.; Otto, A.; Russell, R.C.; Wechselberger, G.
Bladder reconstruction using a prevascularized capsular
tissue seeded with urothelial cells. J. Urol. 2001, 165,
980–985. 32. Campbell, J.H.; Efendy, J.L.; Campbell, G.R.
Novel vascular graft grown within recipient’s own
peritoneal cavity. Circ. Res. 1999, 85 , 1173–1178. 33.
Jacobs, P.F. Stereolithography and Other RP&M Technologies
from Rapid Prototyping to Rapid Tooling; ASME Press: New
York, 1996; 1–26. 34. Sun, W.; Lal, P. Recent development
on computer aided tissue engineering—A review. Comput.
Methods Programs Biomed. 2002, 67 (2), 85–103. 35.
Hutmacher, D.W. Scaffold design and fabrication
technologies for engineered tissues—State of the art and
future prospectives. J. Biomater. Sci., Polym. Ed. 2001,
12, 107–124. 36. Zein, I.; Hutmacher, D.W.; Tan, K.C.;
Teoh, S.H. Fused deposition modeling of novel scaffold
architectures for tissue engineering applications.
Biomaterials 2002, 23, 1169–1185. 37. Sachlos, E.; Reis,
N.; Ainsley, C.; Derby, B.; Czernuszka, J.T. Novel collagen
scaffolds with predefined internal morphology made by solid
freeform fabrication. Biomaterials 2003, 24, 1487–1497. 38.
Bowlin, G.L.; Pawlowski, K.J.; Stitzel, J.D.; Boland, E.D.;
Simpson, D.G.; Fenn, J.B.; Wnek, G.E. Electrospinning of
Polymer Scaffolds for Tissue Engineering. In Tissue
Engineering and Biodegradable Equivalents: Scientific and
Clinical Applications; Wise, D., Eds.; The Humana Press,
Inc. 2002; 65–178. 39. Boland, E.D.; Wnek, G.E.; Simpson,
D.G.; Pawlowski, K.J.; Bowlin, G.L. Tailoring tissue
engineering scaffolds using electrostatic processing
techniques: A study of poly(glycolic acid). J. Macromol.
Sci. 2001, 38, 1231–1243. 40. Stitzel, J.D.; Pawlowski,
K.J.; Wnek, G.E.; Simpson, D.G.; Bowlin, G.L. Arterial
smooth muscle cell proliferation on a novel biomimicking,
biodegradable vascular graft scaffold. J. Biomater. Appl.
2001, 15, 1–12. 41. Matthews, J.A.; Simpson, D.G.; Wnek,
G.E.; Bowlin, G.L. Electrospinning of collagen nanofibers.
Biomacromolecules 2002, 3, 232–238. 42. Matthews, J.A.;
Boland, E.D.; Wnek, G.E.; Simpson, D.G.; Bowlin, G.L.
Electrospinning of collagen type II: A feasibility study.
J. Bioact. Compat. Polym. 2003, 18, 125–134. 43. Wnek,
G.E.; Carr, M.E.; Simpson, D.G.; Bowlin, G.L.
Electrospinning of nanofiber fibrinogen structures. Nano
Lett. 2003, 3 (2), 213–216. 44. Sanders, J.E.; Stiles,
C.E.; Hayes, C.L. Tissue response to single-polymer fibers
of varying diameters: Evaluation of fibrous encapsulation
and macrophage density. J. Biomed. Mater. Res. 2000, 52,
231–237. 45. Zhang, R.; Ma, P.X. Processing of Polymer
Scaffolds: Phase Separation. In Methods of Tissue
Engineering; Atala, A., Lanza, R.C., Eds.; Academic Press
2002; 715–724. T
Tissue Engineering of Skeletal Muscle

35. Wood, S.J.; Pritchard, J.; Sofroniew, M.V. Reexpression


of nerve growth factor receptor after axonal injury
recapitulates a developmental event in motor neurons:
Differential regulation when regeneration is allowed or
prevented. Eur. J. Neurosci. 1990, 2, 650–657.

36. Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of


VEGF and its receptors. Nat. Med. 2003, 9 (6), 669–676.

37. Hopkins, S.P.; Bulgrin, J.P.; Sims, R.L.; Bowman, B.;


Donvan, D.L.; Schmidt, S.P. Controlled delivery of vascular
endothelial growth factor promotes neovascularization and
maintains the limb function in a rabbit model of ishemia.
J. Vasc. Surg. 1997, 27, 886–895.

38. Nabal, E.G. Stem cells combined with gene transfer for
therapeutic vasculogenesis: Magic bullets? Circulation
2002, 105, 672–674.

39. Iwaguro, H.; Yamaguchi, J.; Kalka, C.; Murasawa, S.;


Masuda, H.; Hayashi, S.; Silver, M.; Li, T.; Isner, J.M.;
Asahara, T. Endothelial progenitor cell vascular
endothelial growth factor gene transfer for vascular
regeneration. Circ. 2002, 105, 732–738. 40. Lu, Y.;
Shansky, J.; Del Tatto, M.; Ferland, P.; McGuire, S.;
Marszalkowski, J.; Maish, M.; Hopkins, R.; Wang, X.;
Kosnik, P.; Nackman, M.; Lee, A.; Creswick, B.;
Vandenburgh, H. Therapeutic potential of implanted
tissue-engineered bioartificial muscles delivering
recombinant proteins to the sheep heart. Ann. N. Y. Acad.
Sci. 2002, 961, 78–82. 41. Deasy, B.M.; Huard, J. Gene
therapy and tissue engineering based on muscle-derived stem
cells. Curr. Opin. Mol. Ther. 2002, 4 (4), 382–389. 42.
Pelinkovic, D.; Lee, J.Y.; Engelhardt, M.; Rodosky, M.;
Cummins, J.; Fu, F.H.; Huard, J. Muscle cellmediated gene
delivery to the rotator cuff. Tissue Eng. 2003, 9 (1),
143–151. 43. Young, B.H.; Peng, H.; Huard, J. Muscle-based
gene therapy and tissue engineering to improve bone
healing. Clin. Ortop. 2002, 403 (Suppl.), S243–S251.
Tissue Engineering of Skin

23. Parenteau, N.; Sabolinkski, M.; Prosky, S.; Nolte, C.;


Oleson, M.; Kriwet, K.; Bilbo, P. Biological and physical
factors influencing the successful engraftment of a cultured
human skin substitute. Biotechnol. Bioeng. 1996, 52, 3–14.

24. Eaglstein, W.H.; Falanga, V. Tissue engineering for


skin: An update. J. Am. Acad. Dermatol. 1998, 39 (6),
1007–1010.

25. Nolte, C.F.M.; Oleson, M.A.; Hansbrough, J.F.; Morgan,


J.; Greenleaf, G.; Wilkins, L. Ultrastructural features of
composite skin cultured grafted on athymic mice. J. Anat.
1994, 185, 325–333.

26. Bosca, A.R.; Tinois, E.; Faure, M.; Kanitakis, J.;


Roche, P.; Thivolet, J. Epithelial differentiation of human
skin equivalents after grafting onto nude mice. J. Invest.
Dermatol. 1988, 91 (2), 136–141.

27. English, K.B.; Stayner, N.; Krueger, G.G.; Tuckett,


R.P. Functional innervation of cultured skin grafts. J.
Invest. Dermatol. 1992, 99 (2), 120–128.

28. Wasserman, D.; Sclotterer, M.; Toulon, A.; Cazalet, C.;


Marien, M.; Cherruau, B.; Jaffray, P. Preliminary clinical
studies of biological skin equivalent in burned patients.
Burns 1988, 14, 326–330.

29. Eaglstein, W.H.; Iriondo, M.; Laszlo, K. A composite


skin substitute (graftskin) for surgical wounds. Dermatol.
Surg. 1995, 21 (10), 839–843.

30. Falanga, V.; Margolis, D.; Alvarez, O.; Auletta, M.;


Maggiacomo, J.; Altman, M.; Jensen, J.; Sabolinski, M.;
Hardin-Young, J. Rapid healing of venous ulcers and lack of
clinical rejection with an allogeneic cultured human skin
equivalent. Arch. Dermatol. 1998, 134 (3), 293–300.

31. Cooper, M.L.; Hansbrough, J.F.; Spielvogel, R.L.;


Cohen, R.; Bartel, R.L.; Naughton, G. In vivo optimization
of a living dermal substitute employing cultured human
fibroblasts on a biodegradable polyglycolic acid of
polyglactin mesh. Biomaterials 1991, 12 (2), 243–248.

32. Hansbrough, J.F.; Morgan, J.L.; Greenleaf, G.E.;


Bartel, R. Composite grafts of human keratinocytes grown on
a polyglactin mesh-cultured fibroblast dermal substitute
function as a bilayer skin replacement in fullthickness
wounds on athymic mice. J. Burn Care Rehabil. 1993, 14 (5),
485–494.

33. Purdue, G.F. Dermagraft-TC pivotal safety and efficacy


study. J. Burn Care Rehabil. 1996, 18, S13–S14.

34. Purdue, G.F.; Hunt, J.L.; Still, J.M., Jr.; Law, E.J.;
Herndon, D.N.; Goldfarb, I.W.; Schiller, W.R.; Hansbrough,
J.F.; Hickerson, W.L.; Himel, H.N.; Kealey, P.; Twomey, J.;
Missavage, A.E.; Solem, L.D.; Davis, M.; Totoritis, M.;
Gentzkow, G.D. A multicenter clinical trial of a
biosynthetic skin replacement, Dermagraft-TC, compared with
cryopreserved human cadaver skin for temporary coverage of
excised burn wounds. J. Burn Care Rehabil. 1997, 18, 52–57.

35. Chamberlain, L.J.; Yannas, I.V. Preparation of


Collagen-Glycosaminoglycan Copolymers for Tissue
Regeneration. In Methods of Molecular Medicine; Morgan,
J.R., Yarmush, M.L., Eds.; Humana Press: New Jersey, 1998.
36. Yannas, I.V.; Burke, J.F.; Warpehoski, M.; Stasikelis,
P.; Skrabut, E.M.; Orgill, D.; Giard, D.J. Prompt, longterm
functional replacement of skin. Trans. Am. Soc. Artif.
Intern. Organs 1981, 27, 19–23. 37. Yannas, I.V.; Burke,
J.F.; Orgill, D.P.; Skrabut, E.M. Wound tissue can utilize
a polymeric template to synthesize a functional extension
of skin. Science 1982, 215 (4529), 174–176. 38. Yannas,
I.V.; Burke, J.F.; Orgill, D.P.; Skrabut, E.M. Regeneration
of skin following closure of deep wounds with a
biodegradable template. Trans. Soc. Biomater. 1982, 5,
24–27. 39. Yannas, I.V.; Orgill, D.P.; Skrabut, E.M.;
Burke, J.F. Skin Regeneration with a Bioreplaceable
Polymeric Template. In Polymeric Materials and Artifical
Organs; Gebelein, C.G., Ed.; American Chemical Society:
Washington, DC, 1984. 40. Yannas, I.V.; Lee, E.; Orgill,
D.P.; Skrabut, E.M.; Murphy, G.F. Synthesis and
characterization of a model extracellular matrix which
induces partial regeneration of adult mammalian skin. Proc.
Natl. Acad. Sci. U.S.A. 1989, 86, 933–937. 41. Chang, A.S.;
Yannas, I.V.; Perutz, S.; Loree, H.; Sethi, R.R.; Krarup,
C.; Norregaard, T.V.; Zervas, N.T.; Silver, J.
Electrophysiological Study of Recovery of Peripheral Nerves
Regenerated by a Collagen-Glycoasaminoglycan Copolymer
Matrix. In Progress in Biomedical Polymers; Gebelein, C.G.,
Ed.; Plenum: New York, 1990. 42. Chamberlain, L.J.; Yannas,
I.V.; Hsu, H.-P.; Strichartz, G.; Spector, M. Collagen-GAG
substrate enhances the quality of nerve regeneration
through collagen tubes up to level of autograft. Exp.
Neurol. 1998, 154 (2), 315–329. 43. Hsu, W.-C.; Spilker,
M.H.; Yannas, I.V.; Rubin, P.A.D. Inhibition of
conjunctival scarring and contraction by a porous
collagen-GAG implant. Investig. Ophthalmol. Vis. Sci. 2000,
41 (9), 2404–2411. 44. Yannas, I.V.; Burke, J.F. Design of
an artificial skin I. Basic design principles. J. Biomed.
Mater. Res. 1980, 14 (1), 65–81. 45. Yannas, I.V. Use of
Artificial Skin in Wound Area Management. In The Surgical
Wound; Dineen, P., Ed.; Lea & Febiger: Philadelphia, 1981.
46. Yannas, I.V.; Lee, E.; Orgill, D.P.; Ferdman, A.;
Skrabut, E.M.; Murphy, G.F. De novo synthesis of skin.
Proc. Am. Chem. Soc. Div. Polym. Mater. 1987, 57, 28–32.
47. Butler, C.E.; Orgill, D.P.; Yannas, I.V.; Compton, C.C.
Effect of keratinocyte seeding of
collagen-glycosaminoglycan membranes on the regeneration of
skin in a porcine model. Plast. Reconstr. Surg. 1998, 101
(6), 1572–1579. 48. Butler, C.E.; Yannas, I.V.; Compton,
C.C.; Correia, C.A.; Orgill, D.P. Comparison of cultured
and uncultured keratinocytes seeded into a collagen-GAG
matrix for skin replacements. Br. J. Plast. Surg. 1999, 52
(2), 127–132. 49. Compton, C.C.; Butler, C.E.; Yannas,
I.V.; Warland, G.; Orgill, D.P. Organized skin structure is
regenerated in vivo from collagen-GAG matrices seeded with
autologous keratinocytes. J. Invest. Dermatol. 1998, 110
(6), 908–916. 50. Orgill, D.P.; Butler, C.; Regan, J.F.;
Barlow, M.S.; Yannas, I.V.; Compton, C.C. Vascularized
collagenglycosaminoglycan matrix provides a dermal
substrate and improves take of cultured epithelial
autografts. Plast. Reconstr. Surg. 1998, 102 (2), 423–429.
51. Burke, J.F.; Yannas, I.V.; Quinby, W.C.; Bondoc, C.C.;
Jung, W.K. Successful use of a physiologically acceptable
artificial skin in the treatment of extensive burn injury.
Ann. Surg. 1981, 194, 413–428. 52. Heimbach, D.; Luterman,
A.; Burke, J.; Cram, A.; Herndon, D.; Hunt, J.; Jordan, M.;
McManus, W.; Solem, L.; Warden, G. Artificial dermis for
major burns. Ann. Surg. 1988, 208, 313–320. 53. Stern, R.;
McPherson, M.; Longaker, M.T. Histologic study of artificial
skin used in the treatment of fullthickness thermal injury.
J. Burn Care Rehabil. 1990, 11 (1), 7–13. 54. Michaeli, D.;
McPherson, M. Immunologic study of artificial skin used in
the treatment of thermal injuries. J. Burn Care Rehabil.
1990, 11 (1), 21–26. 55. Sheridan, R.L.; Hegarty, M.;
Hegarty, R.G.; Tompkins, R.G.; Burke, J.F. Artificial skin
in massive burns: Results to ten years. Eur. J. Plast.
Surg. 1994, 17, 91–93. 56. Boyce, S.T.; Hansbrough, J.F.
Biologic attachment and growth of cultured human
keratinocytes onto a graftable collagen and chondroitin
6-sulfate substrate. Surgeon 1988, 103 (4), 421–431. 57.
Boyce, S.T.; Christianson, D.; Hansbrough, J.F. Structure
of a collagen-GAG substitute optimized for cultured
epidermal keratinocytes. J. Biomed. Mater. Res. 1988, 22
(10), 939–957. 58. Cooper, M.L.; Hansbrough, J.F. Use of a
composite skin graft composed of cultured human
keratinocytes and fibroblasts and a collagen-GAG matrix to
cover full-thickness skin wounds on athymic mice. Surgery
1991, 109 (2), 198–207. 59. Cooper, M.L.; Andree, C.;
Hansbrough, J.F.; ZapataSirvent, R.L.; Spielvogel, R.L.
Direct comparison of a cultured composite skin substitute
containing human keratinocytes and fibroblasts to an
epidermal sheet graft containing human keratinocytes on
athymic mice. J. Invest. Dermatol. 1993, 101 (6), 811–819.
60. Hansbrough, J.F.; Boyce, S.T.; Cooper, M.L.; Foreman,
T.J. Burn wound closure with cultured autologous
keratinocytes attached to a collagen-glycosaminoglycan
substrate. JAMA 1989, 262 (15), 2125–2130. 61. Boyce, S.T.;
Greenhalgh, D.G.; Kagan, R.J.; Housinger, T.; Sorrell,
J.M.; Childress, C.P.; Rieman, M.; Warden, G.D. Skin
anatomy and antigen after burn wound closure with composite
grafts of cultured skin cells and biopolymers. Plast.
Reconstr. Surg. 1993, 91 (4), 632–641. 62. Yannas, I.V.
Models of organ regeneration processes induced by
templates. Ann. N.Y. Acad. Sci. 1997, 831, 280–293. 63.
Bianco, P.; Robey, P.G. Stem cells in tissue engineering.
Nature 2001, 414, 118–121. 64. Yannas, I.V. Regeneration
Templates. In The Biomedical Engineering Handbook;
Bronzino, J.D., Ed.; CRC Press: Boca Raton, FL, USA, 2000;
113-1–113-18. T
Tissue Engineering of the Small Intestine

Fig. 8 Illustration of the technique to lengthen small

intestine. (Reproduced from Ref. [36].) Walker, W.A. Human


fetal enterocytes in vitro: Modulation of the phenotype by
extracellular matrix. Proc. Natl. Acad. Sci. U.S.A. 1996,
93, 7717–7722. 12. Patel, H.R.; Tait, I.S.; Evans, G.S.;
Campbell, F.C. Influence of cell interactions in a novel
model of postnatal mucosal regeneration. Gut 1996, 38,
679–686. 13. Sattar, A.; Robson, S.C.; Patel, H.R.; Angus,
B.; Campbell, F.C. Expression of growth regulatory genes in
a SCID mouse-human model of intestinal epithelial
regeneration. J. Pathol. 1999, 187, 229–236. 14. Slorach,
E.M.; Campbell, F.C.; Dorin, J.R. A mouse model of
intestinal stem cell function and regeneration. J. Cell.
Sci. 1999, 112 (Pt. 18), 3029–3038. 15. Tait, I.S.; Flint,
N.; Campbell, F.C.; Evans, G.S. Generation of neomucosa in
vivo by transplantation of dissociated rat postnatal small
intestinal epithelium. Differentiation 1994, 56, 91–100.
16. Tait, I.S.; Penny, J.I.; Campbell, F.C. Does neomucosa
induced by small bowel stem cell transplantation have
adequate function? Am. J. Surg. 1995, 169, 120–125. 17.
Tait, I.S.; Evans, G.S.; Flint, N.; Campbell, F.C. Colonic
mucosal replacement by syngeneic small intestinal stem cell
transplantation. Am. J. Surg. 1994, 167, 67–72. 18. Organ,
G.M.; Mooney, D.J.; Hansen, L.K.; Schloo, B.; Vacanti, J.P.
Transplantation of enterocytes utilizing polymer-cell
constructs to produce a neointestine. Transplant. Proc.
1992, 24, 3009–3011. 19. Organ, G.M.; Mooney, D.J.; Hansen,
L.K.; Schloo, B.; Vacanti, J.P. Enterocyte transplantation
using cellpolymer devices to create intestinal
epithelial-lined tubes. Transplant. Proc. 1993, 25,
998–1001. 20. Vacanti, J.P.; Morse, M.A.; Saltzman, W.M.;
Domb, A.J.; Perez-Atayde, A.; Langer, R. Selective cell
transplantation using bioabsorbable artificial polymers as
matrices. J. Pediatr. Surg. 1988, 23, 3–9. 21. Gibson,
P.R.; van de, P.E.; Maxwell, L.E.; Gabriel, A.; Doe, W.F.
Isolation of colonic crypts that maintain structural and
metabolic viability in vitro. Gastroenterology 1989, 96,
283–291. 22. Karp, J.M.; Dalton, P.D.; Shoichet, M.S.
Scaffolds for tissue engineering. MRS Bull. 2003, 4,
301–306. 23. Hench, L.L.; Polak, J.M. Third-generation
biomedical materials. Science 2002, 295, 1014–1017. 24.
Matthew, H.W.T. Polymers for Tissue Engineering Scaffolds.
In Polymeric Biomaterials; Dumitriu, S., Ed.; Marcel
Dekker: New York, 2002; 167–186. 25. Stock, U.A.; Vacanti,
J.P. Tissue engineering: Current state and prospects. Annu.
Rev. Med. 2001, 52, 443–451. 26. Weng, J.; Wang, M.
Producing chitin scaffolds with controlled pore size and
interconnectivity for tissue engineering. J. Mater. Sci.
Lett. 2001, 20, 1401–1403. 27. Behrend, D.; Schmitz, K.-P.;
Haubold, A. Bioresorbable polymer materials for implant
technology. Adv. Eng. Mater. 2000, 3, 123–125. 28.
Chaignaud, B.E.; Langer, R.; Vacanti, J.P. The History of
Tissue Engineering Using Synthetic Biodegradable Polymer
Scaffolds and Cells. In Synthetic Biodegradable Polymer
Scaffolds; Atala, A., Mooney, D.J., Eds.; Birkhauser:
Boston, 1997; 1–14. 29. Griffith, L.G. Polymeric
biomaterials. Acta Mater. 2000, 48, 263–277. 30. Maquet,
V.; Jerome, R. Design of macroporous biodegradable polymer
scaffold for cell transplantation. Mat. Sci. Forum 1997,
250, 15–42. 31. Boccaccini, A.R.; Roether, J.A.; Hench,
L.L.; Maquet, V.; Jerome, R. A composites approach to
tissue engineering. Ceram. Eng. Sci. Proc. 2002, 23,
805–816. 32. Thomson, R.C.; Yaszemski, M.J.; Mikos, A.G.
Polymer Scaffold Processing. In Principles of Tissue
Engineering; Lanza, R.P., Langer, R., Chick, W.L., Eds.;
R.G. Landes: Austin, TX, 1997; 263–271. 33. Yang, S.;
Leong, K.-F.; Du, Z.; Chua, C.-K. The design of scaffolds
for use in tissue engineering. I. Part, Traditional
factors. Tissue Eng. 2001, 7, 679–689. 34. Conley Wake, M.;
Gupta, P.K.; Mikos, A.G. Fabrication of pliable
biodegradable polymer foams to engineer soft tissues. Cell
Transplant 1996, 5, 465–473. 35. Perez, A.; Grikscheit,
T.C.; Blumberg, R.S.; Ashley, S.W.; Vacanti, J.P.; Whang,
E.E. Tissue-engineered small intestine: Ontogeny of the
immune system. Transplantation 2002, 74, 619–623. 36. Hori,
Y.; Nakamura, T.; Kimura, D.; Kaino, K.; Kurokawa, Y.;
Satomi, S.; Shimizu, Y. Experimental study on tissue
engineering of the small intestine by mesenchymal stem cell
seeding. J. Surg. Res. 2002, 102, 156–160. 37. Hori, Y.;
Nakamura, T.; Matsumoto, K.; Kurokawa, Y.; Satomi, S.;
Shimizu, Y. Tissue engineering of the small intestine by
acellular collagen sponge scaffold grafting. Int. J. Artif.
Organs 2001, 50–54. 38. Tait, I.S.; Evans, G.S.; Kedinger,
M.; Flint, N.; Potten, C.S.; Campbell, F.C. Progressive
morphogenesis in vivo after transplantation of cultured
small bowel epithelium. Cell Transplant 1994, 3, 33–40. 39.
Mooney, D.J.; Mazzoni, C.L.; Breuer, C.; McNamara, K.;
Hern, D.; Vacani, J.P.; Langer, R. Stabilized polyglycolic
acid fibre-based tubes for tissue engineering. Biomaterials
1996, 17, 115–124. 40. Kaihara, S.; Kim, S.S.; Benvenuto,
M.; Choi, R.; Kim, B.S.; Mooney, D.; Tanaka, K.; Vacanti,
J.P. Anastomosis between tissue-engineered intestine and
native small bowel. Transplant. Proc. 1999, 31, 661–662.
41. Kaihara, S.; Kim, S.S.; Benvenuto, M.; Choi, R.; Kim,
B.S.; Mooney, D.; Tanaka, K.; Vacanti, J.P. Successful
anastomosis between tissue-engineered intestine and native
small bowel. Transplantation 1999, 67, 241–245. 42. Kim,
S.S.; Kaihara, S.; Benvenuto, M.S.; Choi, R.S.; Kim, B.S.;
Mooney, D.J.; Vacanti, J.P. Effects of anastomosis of
tissue-engineered neointestine to native small bowel. J.
Surg. Res. 1999, 87, 6–13. 43. Tavakkolizadeh, A.; Berger,
U.V.; Stephen, A.E.; Kim, B.S.; Mooney, D.; Hediger, M.A.;
Ashley, S.W.; Vacanti, J.P.; Whang, E.E. Tissue-engineered
neomucosa: Morphology, enterocyte dynamics, and SGLT1
expression topography. Transplantation 2003, 75, 181–185.
44. Kim, S.S.; Kaihara, S.; Benvenuto, M.; Choi, R.S.; Kim,
B.S.; Mooney, D.J.; Taylor, G.A.; Vacanti, J.P.
Regenerative signals for intestinal epithelial organoid
units transplanted on biodegradable polymer scaffolds T for
tissue engineering of small intestine. Transplantation
1999, 67, 227–233.

45. Grikscheit, T.C.; Ogilvie, J.B.; Ochoa, E.R.; Alsberg,


E.; Mooney, D.; Vacanti, J.P. Tissue-engineered colon
exhibits function in vivo. Surgery 2002, 132, 200–204.

46. Okumura, N.; Nakamura, T.; Shimizu, Y.; Natsume, T.;


Ikada, Y. Experimental study of a new tracheal prosthesis
made from collagen-grafted mesh. ASAIO Trans. 1991, 37,
M317–M319.

47. Okumura, N.; Nakamura, T.; Takimoto, Y.; Natsume, T.;


Teramachi, M.; Tomihata, K.; Ikada, Y.; Shimizu, Y. A new
tracheal prosthesis made from collagen grafted mesh. ASAIO
J. 1993, 39, M475–M479.

48. Yamamoto, Y.; Nakamura, T.; Shimizu, Y.; Matsumoto, K.;


Takimoto, Y.; Kiyotani, T.; Sekine, T.; Ueda, H.; Liu, Y.;
Tamura, N. Experimental replacement of the thoracic
esophagus with a bioabsorbable collagen sponge scaffold
supported by a silicone stent in dogs. ASAIO J. 1999, 45,
311–316.

49. Natsume, T.; Ike, O.; Okada, T.; Takimoto, N.; Shimizu,
Y.; Ikada, Y. Porous collagen sponge for esophageal
replacement. J. Biomed. Mater. Res. 1993, 27, 867–875.

50. Takimoto, Y.; Nakamura, T.; Yamamoto, Y.; Kiyotani, T.;


Teramachi, M.; Shimizu, Y. The experimental replacement of
a cervical esophageal segment with an artificial prosthesis
with the use of collagen matrix and a silicone stent. J.
Thorac. Cardiovasc. Surg. 1998, 116, 98–106.

51. Takimoto, Y.; Okumura, N.; Nakamura, T.; Natsume, T.;


Shimizu, Y. Long-term follow-up of the experimental
replacement of the esophagus with a collagen-silicone
composite tube. ASAIO J. 1993, 39, M736–M739.
52. Yamamoto, Y.; Nakamura, T.; Shimizu, Y.; Matsumoto, K.;
Takimoto, Y.; Kiyotani, T.; Sekine, T.; Ueda, H.; Liu, Y.;
Tamura, N. Experimental replacement of the thoracic
esophagus with a bioabsorbable collagen sponge scaffold
supported by a silicone stent in dogs. ASAIO 1999, 45,
311–316.

53. Potten, C.S.; Hendry, J.H. Radiation and Gut; Elsevier:


Amsterdam, 1995; 45–59.

54. Withers, H.R.; Elkind, M.M. Microcolony survival assay


for cells of mouse intestinal mucosa exposed to radiation.
Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med. 1970,
17, 261–267.

55. Fujimoto, K.; Beauchamp, R.D.; Whitehead, R.H.


Identification and isolation of candidate human colonic
clonogenic cells based on cell surface integrin expression.
Gastroenterology 2002, 123, 1941–1948. 56. Beaulieu, J.F.
Differential expression of the VLA family of integrins
along the crypt-villus axis in the human small intestine.
J. Cell. Sci. 1992, 102 (Pt. 3), 427–436. 57. Kayahara, T.;
Sawada, M.; Takaishi, S.; Fukui, H.; Seno, H.; Fukuzawa,
H.; Suzuki, K.; Hiai, H.; Kageyama, R.; Okano, H.; Chiba,
T. Candidate markers for stem and early progenitor cells,
Musashi-1 and Hes1, are expressed in crypt base columnar
cells of mouse small intestine. FEBS Lett. 2003, 535,
131–135. 58. Potten, C.S.; Booth, C.; Tudor, G.L.; Booth,
D.; Brady, G.; Hurley, P.; Ashton, G.; Clarke, R.;
Sakakibara, S.; Okano, H. Identification of a putative
intestinal stem cell and early lineage marker; musashi-1.
Differentiation 2003, 71, 28–41. 59. Bhatia, M.; Wang,
J.C.; Kapp, U.; Bonnet, D.; Dick, J.E. Purification of
primitive human hematopoietic cells capable of repopulating
immune-deficient mice. Proc. Natl. Acad. Sci. U. S. A. 1997,
94, 5320–5325. 60. Alison, M.R.; Poulsom, R.; Jeffery, R.;
Dhillon, A.P.; Quaglia, A.; Jacob, J.; Novelli, M.;
Prentice, G.; Williamson, J.; Wright, N.A. Hepatocytes from
nonhepatic adult stem cells. Nature 2000, 406, 257. 61.
Gao, Z.; McAlister, V.C.; Williams, G.M. Repopulation of
liver endothelium by bone-marrow-derived cells. Lancet
2001, 357, 932–933. 62. Poulsom, R.; Forbes, S.J.;
Hodivala-Dilke, K.; Ryan, E.; Wyles, S.; Navaratnarasah,
S.; Jeffery, R.; Hunt, T.; Alison, M.; Cook, T.; Pusey, C.;
Wright, N.A. Bone marrow contributes to renal parenchymal
turnover and regeneration. J. Pathol. 2001, 195, 229–235.
63. Eglitis, M.A.; Mezey, E. Hematopoietic cells
differentiate into both microglia and macroglia in the
brains of adult mice. Proc. Natl. Acad. Sci. U.S.A. 1997,
94, 4080–4085. 64. Okamoto, R.; Yajima, T.; Yamazaki, M.;
Kanai, T.; Mukai, M.; Okamoto, S.; Ikeda, Y.; Hibi, T.;
Inazawa, J.; Watanabe, M. Damaged epithelia regenerated by
bone marrow-derived cells in the human gastrointestinal
tract. Nat. Med. 2002, 8, 1011–1017. 65. Brittan, M.; Hunt,
T.; Jeffery, R.; Poulsom, R.; Forbes, S.J.; Goldman, J.M.;
Goldman, J.; Alison, M.R.; Wright, N.A. Bone marrow
derivation of pericryptal myofibroblasts in the mouse and
human small intestine and colon. Gut 2002, 50, 752–757. 66.
Anderson, D.J.; Gage, F.H.; Weissman, I.L. Can stem cells
cross lineage boundaries? Nat. Med. 2001, 7, 393–395.
Tissue Engineering of Tendons

2. Pennisi, E. Tending tender tendons. Science 2002, 295


(5557), 1011.

3. Kubota, H.; Manske, P.R.; Aoki, M.; Pruitt, D.L.;


Larson, B.J. Effect of motion and tension on injured flexor
tendons in chickens. J. Hand Surg. [Am.] 1996, 21 (3),
456–463.

4. Cooper, R.R.; Misol, S. Tendon and ligament insertion. A


light and electron microscopic study. J. Bone Jt. Surg. Am.
1970, 52 (1), 1–20.

5. Pioletti, D.P.; Rakotomanana, L.R. Non linear


viscoelastic law for soft biological tissues. Eur. J. Mech.
2000, 19, 749–759.

6. Pioletti, D.P.; Rakotomanana, L.R.; Benvenuti, J.F.;


Leyvraz, P.F. Viscoelastic constitutive law in large
deformations: Application to human knee ligaments and
tendons. J. Biomech. 1998, 31 (8), 753–757.

7. Juncosa, N.; West, J.R.; Galloway, M.T.; Boivin, G.P.;


Butler, D.L. In vivo forces used to develop design
parameters for tissue engineered implants for rabbit
patellar tendon repair. J. Biomech. 2003, 36 (4), 483–488.

8. Ker, R.F. The implications of the adaptable fatigue


quality of tendons for their construction, repair and
function. Comp. Biochem. Physiol., Part A, Mol. Integr.
Physiol. 2002, 133 (4), 987–1000.

9. Banes, A.J.; Weinhold, P.; Yang, X.; Tsuzaki, M.; Bynum,


D.; Bottlang, M.; Brown, T. Gap junctions regulate
responses of tendon cells ex vivo to mechanical loading.
Clin. Orthop. 1999, 367 Suppl., S356–S370.

10. Mcneilly, C.M.; Banes, A.J.; Benjamin, M.; Ralphs, J.R.


Tendon cells in vivo form a three dimensional network of
cell processes linked by gap junctions. J. Anat. 1996, 189
(Pt. 3), 593–600.

11. Koob, T.J. Biomimetic approaches to tendon repair.


Comp. Biochem. Physiol., Part A, Mol. Integr. Physiol.
2002, 133 (4), 1171–1192.

12. Vogel, K.G. Breakout session 5: Tendon and ligament.


Clin. Orthop. 1999, 367 Suppl., S371–S374.
13. Mooney, D.J.; Mikos, A.G. Growing new organs. Sci. Am.
1999, 280 (4), 60–65.

14. Rowland, J.R.; Tsukazaki, S.; Kikuchi, T.; Fujikawa,


K.; Kearney, J.; Lomas, R.; Wood, E.; Seedhom, B.B.
Radiofrequency-generated glow discharge treatment:
Potential benefits for polyester ligaments. J. Orthop. Sci.
2003, 8 (2), 198–206.

15. Goldstein, J.D.; Tria, A.J.; Zawadsky, J.P.; Kato,


Y.P.; Christiansen, D.; Silver, F.H. Development of a
reconstituted collagen tendon prosthesis. A preliminary
implantation study. J. Bone Jt. Surg. Am. 1989, 71 (8),
1183–1191.

16. Kato, Y.P.; Dunn, M.G.; Zawadsky, J.P.; Tria, A.J.;


Silver, F.H. Regeneration of Achilles tendon with a
collagen tendon prosthesis. Results of a one-year
implantation study. J. Bone Jt. Surg. Am. 1991 , 73 (4),
561–574.

17. Badylak, S.; Arnoczky, S.; Plouhar, P.; Haut, R.;


Mendenhall, V.; Clarke, R.; Horvath, C. Naturally occurring
extracellular matrix as a scaffold for musculoskeletal
repair. Clin. Orthop. 1999, 367 Suppl., S333–S343. 18. Ide,
A.; Sakane, M.; Chen, G.; Shimojo, H.; Ushida, T.;
Tateishi, T.; Wadano, Y.; Miyanaga, Y. Collagen
hybridization with poly( L-lactic acid) braid promotes
ligament cell migration. Mater. Sci. Eng. 2001, 17, 95–99.
19. Koob, T.J.; Hernandez, D.J. Material properties of
polymerized NDGA–collagen composite fibers: Development of
biologically based tendon constructs. Biomaterials 2002, 23
(1), 203–212. 20. Butler, D.L.; Awad, H.A. Perspectives on
cell and collagen composites for tendon repair. Clin.
Orthop. 1999, 367 Suppl., S324–S332. 21. Young, R.G.;
Butler, D.L.; Weber, W.; Caplan, A.I.; Gordon, S.L.; Fink,
D.J. Use of mesenchymal stem cells in a collagen matrix for
Achilles tendon repair. J. Orthop. Res. 1998, 16 (4),
406–413. 22. Awad, H.A.; Butler, D.L.; Boivin, G.P.; Smith,
F.N.; Malaviya, P.; Huibregtse, B.; Caplan, A.I. Autologous
mesenchymal stem cell-mediated repair of tendon. Tissue
Eng. 1999, 5 (3), 267–277. 23. Awad, H.A.; Butler, D.L.;
Harris, M.T.; Ibrahim, R.E.; Wu, Y.; Young, R.G.; Kadiyala,
S.; Boivin, G.P. In vitro characterization of mesenchymal
stem cell-seeded collagen scaffolds for tendon repair:
Effects of initial seeding density on contraction kinetics.
J. Biomed. Mater. Res. 2000, 51 (2), 233–240. 24. Goulet,
F.; Rancourt, D.; Cloutier, R.; Germain, L.; Poole, A.R.;
Auger, F.A. Tendon and ligament. In Principle of Tissue
Engineering, 2nd Ed.; Lanza, R., Langer, R., Vacanti, J.,
Eds.; Academic Press, 2000; 711–722. 25. Hildebrand, K.A.;
Jia, F.; Woo, S.L. Response of donor and recipient cells
after transplantation of cells to the ligament and tendon.
Microsc. Res. Tech. 2002, 58 (1), 34–38. 26. Woo, S.L.;
Hildebrand, K.; Watanabe, N.; Fenwick, J.A.; Papageorgiou,
C.D.; Wang, J.H. Tissue engineering of ligament and tendon
healing. Clin. Orthop. 1999, (367 Suppl.), S312–S323. 27.
Banes, A.J.; Tsuzaki, M.; Hu, P.; Brigman, B.; Brown, T.;
Almekinders, L.; Lawrence, W.T.; Fischer, T. PDGF-BB, IGF-I
and mechanical load stimulate DNA synthesis in avian tendon
fibroblasts in vitro. J. Biomech. 1995, 28 (12), 1505–1513.
28. Ripamonti, U.; Van Den Heever, B.; Sampath, T.K.;
Tucker, M.M.; Rueger, D.C.; Reddi, A.H. Complete
regeneration of bone in the baboon by recombinant human
osteogenic protein-1 (hOP-1, bone morphogenetic protein-7).
Growth Factors 1996, 13 (3–4), 273–289. 29. Bonadio, J.
Tissue engineering via local gene delivery: Update and
future prospects for enhancing the technology. Adv. Drug
Deliv. Rev. 2000, 44 (2–3), 185–194. T
Titanium and Its Alloys

18. Okazaki, Y.; Asao, S.; Rao, S.; Tateishi, T. Effect of


concentration of Zr, Sn, Nb, Ta, Pd, Mo, Co, Cr, Si, Ni, Fe
on the relative growth ratios of bio–cells. J. Jpn. Inst.
Metals 1996, 60, 902–906.

19. Yamamoto, A.; Kobayashi, T.; Maruyama, N.; Nakazawa,


K.; Sumita, M. Fretting fatigue properties of Ti–6Al–4V
alloy in pseudo–body fluid and evaluation of
biocompatibility by cell culture method. J. Jpn. Inst.
Metals 1995, 59, 463–470.

20. Akahori, T.; Niinomi, M.; Fukunaga, K.; Inagaki, I.


Effects of microstructure on the short fatigue crack
initiation and propagation characteristics of biomedical
a/b titanium alloys. Metall. Mater. Trans. A 2000, 31A,
1949–1958.

21. Niinomi, M.; Akahori, T.; Ishimizu, K. Fatigue and


fretting fatigue of biomaterials, Ti–29Nb–13Ta– 45.6Zr, in
air and simulated body environment. In Materials Lifetime
Science and Engineering; Liaw, P.K., Buchana, R.A.,
Klarstrom, D.L., Wei, R.P., Harlow, D.G., Eds.; TMS, 2003;
223–230.

22. Nakazawa, K.; Sumita, M.; Maruyama, N. Fatigue and


fretting fatigue of austenitic stainless steels in psuedo–
body fluid. J. Jpn. Inst. Metals 1999, 63, 1600–1608.

23. Freese, H.L.; Volas, M.G.; Wood, J.R. Metallurgy and


technological properties of titanium and titanium alloys.
In Titanium in Medicine; Brunette, D.M., Tengvall, P.,
Textor, M., Thomsen, P., Eds.; Springer, 2001; 25–51.

24. Niinomi, M. Recent biocompatible metallic biomaterials.


In Structural Biomaterials for the 21st Century; Niinomi,
M., Okabe, T., Tallef, E.H., Lesure, D.R., Lippard, H.E.,
Eds.; TMS, 2001; 15–24.

25. Zwicker, R.; Buehler, K.; Beck, H.; Schmid, H.J.


Mechanical properties and tissue reaction of a titanium
alloy for implant material. In Titanium’80; Kmura, H.,
Izumi, O., Eds.; TMS–AIME: Warrendale, PA, 1980; 2,
505–514.

26. Niinomi, M. Mechanical properties of biomedical


titanium alloys. Mater. Sci. Eng. A 1998, A243, 231–236.

27. Browy, K.–H.; Kramer, K.–H. On the properties of a new


titanium alloy (TiAl5Fe2.5) as implant material. In
Titanium Science and Technology; Lu¨tjering, G., Zwicker,
U., Bunk, W., Eds.; Deutsche Gesellshaft fur Metallkunde E.
V., 1985; Vol 2, 1381–1386.

28. Semlitsch, M. Classic and new titanium alloys for


production of artificial hip joints. Titan 2 1986, 721–740.

29. Implants for Surgery–Metallic Materials–Part 10:


Wrought Titanium 5–Aluminum 2.5–Iron Alloy; ISO 5832–10:
Gene´ve, Swizerland, 1996.

30. Standard Specification for Wrought Titanium 6Al– 7Nb


Alloy for Surgical Implant; ASTM Designation F1295–92,
ASTM: Philadelphia, PA, 1994; 687–689.

31. Implants for Surgery—Metallic Materials—Part II:


Wrought Titanium 6–Aluminum 7–Niobium Alloy; ISO 5832–11:
Gene`ve, Swizerland, 1994.

32. Okazaki, Y. Standardization and activation of industry.


Mater. Jpn. 2004, 43 (3), 182–185.

33. Sasaki, Y.; Doi, K.; Matsushita, T. New titanium alloys


for artificial hip joints. Kinzoku 1996, 66, 812–817.

34. Okazaki, Y.; Ito, Y.; Tateishi, T.; Ito, A. Effect of


heat treatment on microstructure and mechanical properties
of new titanium alloys for surgical implantation. J. Jpn.
Inst. Metals 1995, 59, 108–115. 35. Niinomi, M. Recent
metallic materials for biomedical applications. Matall.
Mater. Trans. A 2002, 33A, 477– 486. 36. Mishra, A.K.;
Davidson, J.A.; Poggie, R.A.; Kovacs, P.; Fitzgerald, T.J.
Mechanical and tribological properties and biocompatibility
of diffusion hardened Ti– 13Nb–13Zr—a new titanium alloy
for surgical implants. In Medical Applications of Titanium
and Its Alloy; Brown, S.A., Lemons, J.E., Eds.; ASTM STP
1272, ASTM, West Conshohocken, PA, 1996; 96–116. 37. Wang,
K.K.; Gustavson, L.J.; Dumbleton, J.H. Microstructure and
properties of a new beta titanium alloy, Ti–12Mo–6Zr–2Fe,
developed for surgical implants. In Medical Applications of
Titanium and Its Alloy; Brown, S.A., Lemons, J.E., Eds.;
ASTM STP 1272, ASTM, West Conshohocken, PA, 1996; 76–87.
38. Zardiackas, L.D.; Mitchell, D.W.; Disegi, J.A.
Characterization of Ti–15Mo beta titanium alloy for
orthopaedic implant. In Medical Applications of Titanium
and Its Alloy; Brown, S.A., Lemons, J.E., Eds.; ASTM STP
1272, ASTM, West Conshohocken, PA, 1996; 60–75. 39.
Bhambri, S.K.; Shetty, R.H.; Gilberton, L.N. Optimization
of properties of Ti–15Mo–2.8Nb–3Al–0.2Si and
Ti–15Mo–2.8Nb–0.2Si–0.26O beta titanium alloys for
application in prosthetic implants. In Medical Applications
of Titanium and Its Alloy; Brown, S.A., Lemons, J.E., Eds.;
ASTM STP 1272, ASTM, West Conshohocken, PA, 1996; 88–95.
40. Ahmed, T.; Long, M.; Silvestri, J.; Ruiz, C.; Rack,
H.J. A new low modulus, biocompatible titanium alloy. In
Titanium’95; Blenkinsop, P.A., Evans, W.J., Flower, H.M.,
Eds.; The Institute of Materials: London, 1996; Vol. II,
1760–1767. 41. Niinomi, M; Hattori, T; Niwa, S.
Biomaterials in orthopedics. In Material Characteristics
and Biocompatibility of Low Rigidity Titanium Alloys for
Biomedical Applications; Yaszemski, M.J., Trantolo, D.J.,
Lewandrowski, K.U., Hasirci, V., Altobelli, D.E., Wise,
D.L., Eds.; Marcel Dekker, Inc., 2004; 41–62. 42. Morita,
A.; Fukui, H.; Tadano, H.; Hayashi, S.; Hasegawa, J.;
Niinomi, M. Alloying titanium and tantalum by cold crucible
levitation melting (CCLM) furnace. Mater. Sci. Eng. A 2000,
A280, 208–213. 43. Fukui, H.; Kunii, T.; Fujishiro, Y.;
Morita, A.; Niinomi, M.; Yamada, A.; Hasegawa, J. Melting
method for biomedical Ti–Ta alloy with high melting point.
J. Jpn. Soc. Dent. Mater. Devices 2000, 19, 49–55. 44.
Kawasaki, H.; Kuroda, D.; Hanawa, T.; Kuroda, H.;
Kobayashi, M.; Kobayashi, T.; Imai, H. Mechanical
properties of Ti–Fe–Ta system alloys for biomedical
applications. Proceedings of the Fall Meeting of the Japan
Institute of Metals, 2003; 130. 45. Ou, A.; Chou, T.;
Asami, K.; Inoue, A. Properties of Ti–Zr–Nb–Ta system
bio–soft titanium alloy. Proceedings of the Annual Meeting
of the Japan Institute of Metals, 2003; 326. 46. Zhou,
Y.L.; Niinomi, M.; Akahori, T. Effect of Ta content on
Young’s moduli and tensile strength of binary Ti–Ta alloys
for biomedical applications. Mater. Sci. Eng. A 2004, A371,
283–290. 47. Zhou, Y.L.; Niinomi, M.; Akahori, T.
Decomposition of martensite a during aging treatments and
resulting mechanical properties of Ti–Ta alloys. Mater.
Sci. Eng. A 2004, A371, 283–290. 48. Zhou, Y.L.; Niinomi,
M.; Akahori, T. Dynamic Young’s modulus and mechanical
properties of Ti–Hf alloys. Mater Trans. 2004, 45 (5),
1549–1554. 49. Uggowitzer, P.J.; Ba¨hre, W.F.; Speidel,
M.O. Metal injection molding of Ni–Free stainless steels.
Proc. Adv. Powder Metall. Part. Mater. 1997, 3,
18–11318–121. 50. Yoneyama, T; Doi, H.; Kobayasi, E.;
Kasuga, J.; Fukushima, O.; Murakami, M. Mechanical
properties and phase transformation of Ti–Ni super elastic
alloy for dental application. Proc. AISB4 FNB 2004,
241–242. 51. Okuyama, M; Kobayashi, J.; Take, S.; Itoi, Y.
Effect of plasma spray and surface sealing treatment of
Ni–Ti shape memory alloy applying to biomaterials. Mater.
Trans. 2004, 45 (4), 1045–1053. 52. Ogata, Y; Takatugu, M.;
Kunimasa, T.; Kobayashi, K.F. Tensile strength and
pseudo–elasticity of YAG laser spot melted Ti–Ni shape
memory alloy wire. Mater. Trans. 2004, 45 (4), 1070–1076.
53. Nitta, K.; Watanabe, S.; Masahashi, N.; Hosoda, H.;
Hanada, S. Ni–free Ti–Nb–Sn shape memory alloys. In
Structural Biomaterials for the 21st Century; Niinomi, M.,
Okabe, T., Taleff, E.M., Lesure, D.R.M., Lippard, H.E.,
Eds.; TMS, 2001; 25–35. 54. Kim, H.Y.; Ohmatsu, Y.; Kim,
J.I.; Miyazaki, S.; Hosoda, H. Mechanical properties and
shape memory behavior of Ti–Mo–Ga alloys. Mater. Trans.
2004, 45 (4), 1090–1095. 55. Inamura, T.; Fukui, Y.;
Hosoda, H.; Wakashima, K.; Miyazaki, S. Relationship
between texture and macroscopic transformation strain in
severely cold– rolled Ti–Nb–Al super elastic alloy. Mater.
Trans. 2004, 45 (4), 1083–1089. 56. Hosoda, H.; Hosoda, N.;
Miyazaki, S. Mechanical properties of Ti–Mo–Al biomedical
shape memory alloys. Trans. MRSJ 2001, 26 (1), 243–246. 57.
Ikeda, M.; Komatsu, S.; Nakamura, Y. Effects of Sn and Zr
additions on phase constitution and aging behavior of Ti–50
mass% Ta alloys quenched from b single phase region. Mater.
Trans. 2004, 45 (4), 1106–1112. 58. Toyoshima, K.;
Hirasawa, T.; Ikeda, M. Effect of Zr addition on phase
constitution and heat treatment behavior of Ti–35 mass% Nb
alloy. Proceedings of the Annual Meeting of the Japan
Institute of Metals, 2003; 130. 59. Maeshima, T; Nishida,
M. Shape memory and mechanical properties of biomedical
Ti–Sc–Mo alloy. Mater. Trans. 2004, 45 (4), 1101–1105. 60.
Maeshima, T.; Nishida, M. Shape memory properties of
biomedical Ti–Mo–Ag and Ti–Mo–Sn alloys. Mater. Trans.
2004, 45 (4), 1096–1100. 61. Sakaguchi, N.; Niinomi, M.;
Akahori, T. Deformation behaviors of Ti–Nb–Ta–Zr system
alloys for biomedical applications. Mater. Trans. 2004, 45
(4), 1113–1119. 62. Hosoda, H.; Miyazaki, S. Recent topics
of shape memory materials and related technology. J. Jpn.
Soc. Mech. Eng. 2004, 107 (1028), 509–520. 63. Niinomi, M.
Recent research and development in titanium alloys for
biomedical applications and healthcare goods. Sci. Technol.
Adv. Mater. 2003, 4, 445–4541. 64. Niinomi, M.
Cyto–toxicity and fatigue performance of low rigidity
titanium alloy, Ti–29Nb–13Ta–4.6Zr, for biomedical
applications. Biomaterials 2003, 24, 2673–2683. 65.
Niinimi, M.; Akahori, T.; Hattopri, Y.; Morikawa, K.;
Kasuga, T.; Fukui, H.; Suzuki, A.; Kyo, K.; Niwqa, S. Super
elastic functional b type titanium alloy with low Young’s
modulus for biomedical applications. J. ASTM Int. 2005, 2
(6), 448–464. 66. Hattori, T.; Morikawa, K.; Niwa, S.;
Sato, K.; Niinomi, M.; Suzuki, A. Bone tissue reaction to
new b titanium low rigidity alloy: rabbit study on bone
healing, remodelling and atrophy in intramedullary fracture
fixation. International Conference on Biomechanics and
Annual Scientific Meeting of Taiwanese Society of
Biomechanics, Nov 9–1, 2001; National Yang Ming University:
Taipei, Taiwan, 2001; 28. 67. Niinomi, M. Fatigue
characteristics and microstructure of titanium alloys for
biomedical applications. Proceedings Fatigue’2002; Blom,
A.F., Ed.; EMAS, 2002; 3/5, 2073–2083. 68. Niinomi, M.
Cyto–toxicity and fatigue performance of low rigidity
titanium alloy, Ti–29Nb–13Ta–4.6Zr, for biomedical
applications. Biomaterials 2003, 24, 2673–2683. 69.
Niinomi, M.; Akahori, T.; Yabunaka, T.; Fukui, H.; Suzuki,
A. Fretting fatigue characteristics in air and simulated
body environment of newly developed b type titanium alloy
for biomedical applications. J. Iron Steel Inst. Jpn. 2002,
88 (9), 553–560. 70. Maruyama, N.; Nkazawa, K.; Sumita, M.;
Sato, M. Effect of stress frequency of fatigue and fretting
fatigue life for commercially pure Ti and Ti–6Al–4V alloy
in pseudo-body fluid. J. Jpn. Inst. Biomater. 2000, 18 (1),
17–23. 71. Li, S.J.; Yang, R.; Li, S.; Hao, Y.L.; Cui,
Y.Y.; Niinomi, M.; Guo, Z.X. Wear characteristics of Ti–Nb–
Ta–Zr and Ti–6Al–4V alloys for biomedical applications.
Wear 2004, 257, 869–876. 72. Ueda, K.; Narushima, T.;
Ouchi, C.; Iguchi, Y. Wear loss and elution of C. P. Ti and
titanium alloys in simulated body fluid. Mater. Sci. Forum
2005, 475– 479, 2333–2336. 73. Tamura, Y.; Yokoyama, A.;
Watari, F.; Uo, M.; Kawasaki, T. Mechanical properties of
surface nitrided titanium for abrasion resistant implant
materials. Mater. Trans. 2002, 43 (12), 3043–3051. 74.
Akahori, T.; Niinomi, M.; Ishimizu, K.; Fkui, H.; Suzuki,
A. Effect of thermomechanical treatment on fatigue
characteristics of Ti–29Nb–13Ta–4.6Zr alloy for biomedical
applications. J. Jpn. Inst. Metals 2003, 67 (11), 652–660.
75. Niinomi, M. Fatigue characteristics and microstructure
of titanium alloys for biomedical applications. T
Proceedings Fatigue’2002; Blom, A.F., Ed.; EMAS, 2002; Vol.
3/5, 2073–2083.

76. Niinomi, M.; Kobayashi, T.; Toriyama, O.; Kawakami, N.;


Ishida, Y.; Matsuyama, Y. Fracture characteristics,
microstructure and tissue reaction of Ti– 5Al–2.5Fe for
orthopedic surgery. Metall. Mater. Trans. A 1996, 27A (12),
3925–3935.

77. Maruyama, N.; Nakazawa, K.; Sumita, M.; Sato, M. Effect


of stress frequency on fatigue and fretting fatigue life
for commercially pure Ti and Ti–6Al–4V alloy in pseudo-body
fluid. J. Jpn. Biomater. Soc. 2000, 18 (1), 17–23.

78. Akahori, T.; Niinomi, M.; Fukui, H.; Suzuki, A.


Fatigue, fretting fatigue and corrosion characteristics of
biocompatible beta type titanium alloy conducted with
various thermo-mechanical treatments. Mater. Trans. 2004,
45 (5), 1540–1548.

79. Niinomi, M.; Akahori, T.; Nakamura, S.; Fkui, H.;


Suzuki, A. Friction wear of surface oxidized newly
developed b type titanium alloy for biomedical applications
in simulated body environment. J. Iron Steel Inst. Jpn.
2002, 88 (9), 567–574.

80. Mano, T.; Sugiyama, O.; Shibuya, Y.; Nakayama, H.;


Osamu Takai, O. Formation of diamond–like carbon based
double-layer film on Ti–6Al–4V substrate by ionization
deposition. Mater. Trans. 2004, 45 (5), 1601–1606.

81. Nishio, K.; Yamaguchi, T.; Era, H.; Katoh, M. Surface


modification of titanium using laser beam. Mater. Trans.
2004, 45 (5), 1613–1619.

82. Yoshinari, M.; Oda, Y. Titanium surface modification


with dry process for dental implants. In Structural
Biomaterials for the 21st Century; Niinomi, M., Okabe, T.,
Taleff, E.M., Lesuer, D.R., Lippard, H.F., Eds.; TMS, 2001;
129–136.

83. Yoshinari, M.; Hayakawa, T.; Wolke, J.G.C.; Nemoto, K.;


Jansen, J.A. Influence of rapid heating with infrared
radiation on RF magnetron–sputtered calcium phosphate
coating. J. Biomater. Res. 1997, 37, 60–67.

84. Wang, C.K.; Chern, J.H.; Ju, C.P.; Ong, H.C.; Chang,
R.P. Structural characterization of pulsed laserdeposited
hydroxyapatite film on titatium substrate. Biomaterials
1997, 18, 1331–1338.

85. Yoshinari, M.; Ohtsuka, Y.; De´rand, T. Thin


hydroxyapatite coating produced by the ion beam dynamic
mixing method. Biomaterials 1994, 15, 529–535.

86. Cui, F.Z.; Luo, Z.S.; Feng, Q. Highly adhesive


hydroxyapatite coatings on titanium alloy formed by ion
beam assisted deposition. J. Mater. Sci. Mater. Med. 1997,
8, 403–405.

87. Nonami, T.; Naganuma, K.; Kameyama, T. Superplastic


bonding of biomaterial-apatite and Ti-alloy. Mater. Jpn.
1998, 37, 856–858.

88. Hanawa, T.; Kon, M.; Ukai, H.; Murakami, K.; Miyamoto,
Y.; Asaoka, K. Surface modification of titanium in
calcium-ion-containing solutions. J. Biomater. Res. 1997,
34, 273.
89. Ban, S.; Maruno, S. Effect of temperature on
electrochemical deposition of calcium phosphate coatings in
a simulated body fluid. Biomaterials 1995, 16, 977–981. 90.
Kim, H.M.; Miyaji, F.; Kokubo, T. Effect of heat treatment
on apatite-forming ability of Ti metal induced by alkali
treatment. J. Mater. Sci.: Mater. Med. 1997, 8, 341–347.
91. Kasuga, T.; Watanabe, M.; Nogami, M.; Niinomi, M.
Calcium phosphate invert glass-ceramic coating joined by
self-development of compositionally gradient layers on a
titanium alloy. Biomaterials 2001, 22, 577–582. 92. Kasuga,
T.; Nogami, M.; Niinomi, M.; Hattori, T.; Hench, L.L.
Enhancing effect of autoclaving on bioactivity of
b–titanium alloy coated with calcium phosphate
glass–ceramic. Key Eng. Mater. 2005, 284– 286, 243–246. 93.
Okazaki, Y. Implant applications of highly corrosion–
resistant Ti–15Zr–4Nb–4Ta alloy. Mater. Trans. 2002, 43
(12), 2936–2942. 94. Okazaki, Y. Dental casting properties
of Ti–15Zr– 4Nb–4Ta alloy. Mater. Trans. 2002, 43 (12),
3134– 3141. 95. Srimaneepong, V., et al. Strain and
displacement of Ti–6Al–7Nb alloy casting for removable
partial denture frameworks. Proc. AISB4 FNB 2004, 237–238.
96. Akahori, T.; Niinomi, M.; Isohama, R.; Suzuki, A.
Improvement of mechanical performance of cast a/b titanium
alloys for dental applications by thermochemical
processing. In Structural Biomaterials for the 21st
Century; Niinomi, M., Okabe, T., Tallef, E.H., Lesure,
D.R., Lippard, H.E., Eds.; TMS, 2001; 91–98. 97. Eylon, D.;
Newman, J.R.; Thome, J.K. Titanium and titanium alloy
castings. InMetals Handbook, 10th Ed.; Abel, L.A., Kiepura,
R.T., Thomas, P., Lampman, H.F., Wheaton, N.D., Eds.; ASM
International: Materials Park, OH, 1990; Vol. 2, 634–646.
98. Yoshitani, S.; Niinomi, M.; Fukunaga, K.; Fukui, H.;
Takeuchi, T.; Katsura, S. Dental precision castability of
biocompatible b–type titanium alloy. CAMP–ISIJ 2000, 13,
1345. 99. Kobayashi, E.; Doi, H.; Takahashi, M.; Nakano,
T.; Yoneyama, T.; Hamanaka, H.J. Castability and mechanical
properties of Ti–6Al–7Nb–alloy dental cast. Jnp. Soc. Dent.
Mater. Dev. 1995, 14, 406–413. 100. Miyazaki, T.; Tani, Y.;
Tamaki, Y.; Suzuki, E.; Miyaji, T. Application of CaO to
dental investments. J. Jpn. Soc. Dent. Mater. Dev. 1987, 6,
437–440. 101. Miyazaki, T.; Tani, Y.; Tamaki, Y.; Suzuki,
E.; Miyaji, T. Casting of titanium with calcia investment
(part2). Improvement of mold treatment and finess on
castings. J. Jpn. Soc. Dent. Mater. Dev. 1987, 6, 633–638.
102. Takeuchi, T.; Niinomi, M.; Akahori, T.; Katsura, S.;
Fukui, H. Development of dental precision casting process
for titanium alloy with high biocompatibility. Mater. Jpn.
2005, 44 (1), 68–70. 103. Niinomi, M.; Akahori, T.;
Takeuchi, T.; Katsura, S. Dental precision casting of
Ti–29Nb–13Ta–4.6Zr using calcia mold. Mater. Sci. Forum
2005, 475–479, 2303–2308. 104. Niinomi, M.; Akahori, T.;
Takeuchi, T.; Katsura, S.; Fukui, H.; Toda, H. Mechanical
properties and cytotoxicity of newly designed beta type
titanium alloys with low melting points for dental
applications. Mater. Sci. Eng. C. 2005, (3), 417–425.
Transdermal Drug Delivery

1. Crank, J.; Park, G.S. Diffusion in Polymers; Academic


Press: London, 1968; 1–39.

2. Mitragotri, S.; Johnson, M.E.; Blankschtein, D.; Langer,


R. An analysis of the size selectivity of solute
partitioning, diffusion, and permeation across lipid
bilayers. Biophys. J. 1999, 77 (3), 1268–1283.

3. Hildebrand, J.H.; Prausnitz, J.M.; Scott, R.L. Regular


and Related Solutions; Van Nostrand Reinhold: New York,
1970.

4. Yalkowsky, S.H.; Dannenfelser, R.-M.; Myrdal, P.;


Simamore, P. Unified physical property estimation
relationships (UPPER). Chemosphere 1994, 28, 1657–1673.

5. Shaw, J.E.; Prevo, M.; Gale, R.; Yum, S.I. Percutaneous


Absorption. In Biochemistry and Physiology of the Skin;
Goldsmith, L.A., Ed.; Oxford University Press: Rochester,
NY, 1991; Vol. II, 1447–1479.

6. Asbill, C.S.; El-Kattan, A.F.; Michniak, B. Enhancement


of transdermal drug delivery: Chemical and physical
approaches. Crit. Rev. Ther. Drug Carr. Syst. 2000, 17 (6),
621–658.

7. Burnette, R.J. Iontophoresis. In Transdermal Drug


Delivery: Development Issues and Research Initiatives;
Hadgraft, J., Guy, R.H., Eds.; Marcel Dekker: New York,
1989; 247–291.

8. Pikal, M.J. The role of electroosmotic flow in


transdermal iontophoresis. Adv. Drug Deliv. Rev. 1992, 9,
201–237.

9. Tamada, J.A.; Garg, S.; Jovanovic, L.; Pitzer, K.R.;


Fermi, S.; Potts, R.O. Noninvasive glucose monitoring.
Comprehensive clinical results. JAMA 1999, 282 (19),
1839–1844.

10. Phipps, J.B.; Padmanabhan, R.V.; Lattin, G.A.


Iontophoretic delivery of model inorganic and drug ions. J.
Pharm. Sci. 1989, 78 (5), 365–369. 11. Scott, E.R.; Phipps,
J.B.; Gyory, J.R.; Padmanabhan, R.V. Electrotransport
Systems for Transdermal Delivery: A Practical
Implementation of Iontophoresis. In Handbook of
Pharmaceutical Controlled Release Technology; Wise, D.L.,
Ed.; Marcel Dekker: New York, 2000; 617–659. 12. Mikszta,
J.A.; Alarcon, J.B.; Brittingham, J.M.; Sutter, D.E.;
Pettis, R.J.; Harvey, N.G. Improved genetic immunization
via micromechanical disruption of skinbarrier function and
targeted epidermal delivery. Nat. Med. 2002, 8 (4),
415–419. 13. Henry, S.; McAllister, D.V.; Allen, M.G.;
Prausnitz, M.R. Microfabricated microneedles: A novel
approach to transdermal drug delivery. J. Pharm. Sci. 1998,
87 (8), 922–925. 14. Lin, W.Q.; Cormier, M.; Samiee, A.;
Griffin, A.; Johnson, B.; Teng, C.-L.; Hardee, G.E.;
Daddona, P.E. Transdermal delivery of antisense
oligonucleotides with microprojection patch (Macroflux s )
technology. Pharm. Res. 2001, 18 (12), 1789–1793. 15.
Matriano, J.A.; Cormier, M.; Johnson, J.; Young, W.A.;
Buttery, M.; Nyam, K.; Daddona, P.E. Macroflux s
microprojection array patch technology: A new and efficient
approach for intracutaneous immunization. Pharm. Res. 2002,
19 (1), 63–70. 16. Cormier, M.; Daddona, P.E. Macroflux
Technology for Transdermal Delivery of Therapeutic Proteins
and Vaccines. In Modified-Release Drug Delivery Technology;
Roberts, M., Hadgraft, J., Rathbone, M.J., Eds.; Marcel
Dekker: New York, 2002; 589–598. 17. Patil, S.M.; Hogan,
D.J.; Maibach, H.I. Transdermal Drug Delivery Systems:
Adverse Dermatologic Reactions. In Dermatotoxicology, 5th
Ed.; Marzulli, F.N., Maibach, H.I., Eds.; Taylor & Francis:
New York, 1996; 389–396. 18. Kydonieus, A.F.; Wille, J.J.
Biochemical Modulation of Skin Reactions: Transdermals,
Topicals, Cosmetics; CRC Press: New York, 2000. 19. Mize,
N.K.; Johnson, J.A.; Hansch, C.; Cormier, M. Quantitative
structure-activity relationship and cytotoxicity. Curr.
Probl. Dermatol. 1995, 23, 224–229. 20. Bronaugh, R.L.;
Maibach, H.I. Percutaneous Absorption.
Drugs–Cosmetics–Mechanism–Methodology, 3rd Ed.; Marcel
Dekker, Inc.: New York, 1999. 21. Walters, K.A.
Dermatological and Transdermal Formulations; Marcel Dekker:
New York, 2002. 22. Couvreur, P.; Duchene, D.; Green, P.;
Junginger, H.E. Minutes, Transdermal Administration, A Case
Study, Iontophoresis; Editions de Sante´, Pubs.: Paris,
1997. 23. Smith, E.W.; Maibach, H.I. Percutaneous
Penetration Enhancers; CRC Press: New York, 1995. T
Transgenic and Gene-Targeted Organs

49. Fodor, W.L.; Williams, B.L.; Matis, L.A.; Madri, J.A.;


Rollins, S.A.; Knight, J.W.; Velander, W.; Squinto, S.P.
Expression of a functional human complement inhibitor in a
transgenic pig as a model for the prevention of xenogeneic
hyperacute organ rejection. Proc. Natl. Acad. Sci. U.S.A.
1994, 91, 11153–11157.

50. McCurry, K.R.; Kooyman, D.L.; Diamond, L.E.; Byrne,


G.W.; Logan, J.S.; Platt, J.L. Human complement regulatory
proteins in transgenic animals regulate complement
activation in xenoperfused organs. Transplant. Proc. 1995,
27, 317–318.

51. Schmoeckel, M.; Bhatti, F.N.K.; Zaidi, A.; Cozzi, E.;


Waterworth, P.D.; Tolan, M.J.; Goddard, M.; Warner, R.G.;
Langford, G.A.; Dunning, J.J.; Wallwork, J.; White, D.J.G.
Orthotopic heart transplantation in a transgenic
pig-to-primate model. Transplantation 1998, 65, 1570–1577.

52. Zaidi, A.; Bhatti, F.; Schmoeckel, M.; Cozzi, E.;


Chavez, G.; Wallwork, J.; White, D.; Friend, P. Kidneys
from HDAF transgenic pigs are physiologically compatible
with primates. Transplant. Proc. 1998, 30, 2465–2466.

53. Schmoeckel, M.; Nollert, G.; Shahmohammadi, M.; Young,


V.K.; Chavez, G.; Kasper-Konig, W.; White, D.J.G;
Muller-Hocker, J.; Arendt, R.M.; WilbertLampen, U.; Hammer,
C.; Reichart, B. Prevention of hyperacute rejection by
human decay accelerating factor in xenogeneic perfused
working hearts. Transplantation 1996, 62, 729–734.

54. Byrne, G.W.; McCurry, K.R.; Martin, M.J.; McClellan,


S.M.; Platt, J.L.; Logan, J.S. Transgenic pigs expressing
human CD59 and decay-accelerating factor produce an
intrinsic barrier to complement-mediated damage.
Transplantation 1997, 63, 149–155.

55. Leventhal, J.R.; Matas, A.J.; Sun, L.H.; Reif, S.;


Bolman, R.M.; Dalmasso, A.P.; Platt, J.L. The
immunopathology of cardiac xenograft rejection in the
guinea pig-to-rat model. Transplantation 1993, 56, 1–8.

56. Lin, S.S.; Platt, J.L. Immunologic barriers to


xenotransplantation. J. Heart Lung Transplant. 1996, 15,
547–555.

57. Porter, K.A. Renal transplantation. In Pathology of the


Kidney, 4th Ed.; Little, Brown and Company: Boston, 1992;
Vol. 3, 1799–1933.

58. Gaca, J.G.; Lee, W.; Aksoy, O.; Braedehoeft, S.J.;


Gonzalez-Stawinski, G.V.; Parker, W.; Davis, R.D. Evidence
for polyreactive xenoreactive antibodies in the repertoire
of human anti-swine antibodies: the ‘next’ humoral barrier
to xenotransplantation. Transplant. Immunol. 2001, 9,
19–27.

59. Blakely, M.L.; Van Der Werf, W.J.; Berndt, M.C.;


Dalmasso, A.P.; Bach, F.H.; Hancock, W.W. Activation of
intragraft endothelial and mononuclear cells during
discordant xenograft rejection. Transplantation 1994, 58,
1059–1066.

60. Malyguine, A.M.; Saadi, S.; Holzknecht, R.A.; Patte,


C.R.; Sud, N.; Platt, J.L.; Dawson, J.R. Induction of
procoagulant function in porcine endothelial cells by human
NK cells. J. Immunol. 1997, 159, 4659–4664. 61. Platt,
J.L.; Lin, S.S.; McGregor, C.G.A. Acute vascular rejection.
Xenotransplantation 1998, 5, 169–175. 62. Bracy, J.L.;
Sachs, D.H.; Iacomini, J. Inhibition of xenoreactive
natural antibody production by retroviral gene therapy.
Science 1998, 281, 1845–1847. 63. Chopek, M.W.; Simmons,
R.L.; Platt, J.L. ABOincompatible renal transplantation:
initial immunopathologic evaluation. Transplant. Proc.
1987, 19, 4553–4557. 64. Bach, F.H.; Ferran, C.;
Hechenleitner, P.; Mark, W.; Koyamada, N.; Miyatake, T.;
Winkler, H.; Badrichani, A.; Cardinas, D.; Hancock, W.H.
Accommodation of vascularized xenografts: expression of
‘‘protective genes’’ by donor endothelial cells in a host
Th2 cytokine environment. Nat. Med. 1997, 3, 196–204. 65.
Bach, F.H.; Ferran, C.; Soares, M.; Wrighton, C.J.;
Anrather, J.; Winkler, H.; Robson, S.C.; Hancock, W.W.
Modification of vascular responses in xenotransplantation:
inflammation and apoptosis. Nat. Med. 1997, 3, 944–948. 66.
Ferran, C.; Badrichani, A.Z.; Cooper, J.T.; Stroka, D.M.;
Bach, F.H. Xenotransplantation: progress toward clinical
development. Adv. Nephrol. Necker Hosp. 1997, 27, 391–420.
67. Ferran, C.; Stroka, D.M.; Badrichani, A.Z.; Cooper,
Z.T.; Bach, F.H. Adenovirus-mediated gene transfer of A20
renders endothelial cells resistane to activation: a means
of evaluating the role of endothelial cell activation in
xenograft rejection. Transplant. Proc. 1997, 29, 879–880.
68. Von Albertini, M.; Ferran, C.; Brostjan, C.; Bach,
F.H.; Goodman, D.J. Membrane-associated lymphotoxin on
natural killer cells activates endothelial cells via an
NFkappaB-dependent pathway. Transplantation 1998, 66,
1211–1219. 69. Murray, A.G.; Khodadoust, M.M.; Pober, J.S.;
Bothwell, A.L.M. Porcine aortic endothelial cells activate
human T cells: direct presentation of MHC antigens and
costimulation by ligands for human CD2 and CD28. Immunity
1994, 1, 57–63. 70. Dorling, A.; Stocker, C.; Tsao, T.;
Haskard, D.O.; Lechler, R.I. In vitro accommodation of
immortalized porcine endothelial cells: resistance to
complement mediated lysis and down-regulation of VCAM
expression induced by low concentrations of polyclonal
human IgG antipig antibodies. Transplantation 1996, 62,
1127–1136. 71. Kirk, A.D.; Li, R.A.; Kinch, M.S.;
Abernethy, K.A.; Doyle, C.; Bollinger, R.R. The human
antiporcine cellular repertoire. In vitro studies of
acquired and innate cellular responsiveness.
Transplantation 1993, 55, 924–931. 72. Waldmann, H.
Therapeutic approaches for transplantation. Curr. Opin.
Immunol. 2001, 13, 606–610. 73. Logan, J.S.; Martin, M.J.
Transgenic swine as a recombinant production system for
human hemoglobin. Methods Enzymol. 1994, 231, 435–445. 74.
Prather, R.S.; Hawley, R.J.; Carter, D.B.; Lai, L.;
Greenstein, J.L. Transgenic swine for biomedicine and
agriculture. Theriogenology 2003, 59, 115–125. 75. Wilmut,
I.; Schnieke, A.E.; McWhir, J.; Kind, A.J.; Campbell, K.H.
Viable offspring derived from fetal and adult mammalian
cells. Nature 1997, 385, 810–813. 76. Kato, Y.; Tani, T.;
Sotomaru, Y.; Kurokawa, K.; Kato, J.; Doguchi, H. Eight
calves cloned from somatic cells of a single adult. Science
1998, 282, 2095–2098. 77. Wakayama, T.; Perry, A.C.;
Zuccotti, M.; Johnson, K.R.; Yanagimachi, R. Full-term
development of mice from enucleated oocytes injected with
cumulus cell nuclei. Nature 1998, 394, 315–316. 78. Phelps,
C.J.; Koike, C.; Vaught, T.D.; Boone, J.; Wells, K.D.;
Chen, S.H.; Ball, S.; Specht, S.M.; Polejaeva, I.A.;
Monahan, J.A.; Jobst, P.M.; Sharma, S.B.; Lamborn, A.E.;
Garst, A.S.; Moore, M.; Demetris, A.J.; Rudert, W.A.;
Bottino, R.; Bertera, S.; Trucco, M.; Starzl, T.E.; Dai,
Y.; Ayares, D.L. Production of alpha
1,3-galactosyltransferase-deficient pigs. Science 2003, 299,
411–414. 79. Dai, Y.; Vaught, T.D.; Boone, J.; Chen, S.;
Phelps, C.J.; Ball, S.; Monahan, J.A.; Jobst, P.M.;
McCreath, K.J.; Lamborn, A.E.; Cowell-Lucero, J.L.; Wells,
K.D.; Colman, A.; Polejaeva, I.A.; Ayares, D.L. Targeted
disruption of the alpha1,3-galactosyltransferase gene in
cloned pigs. Nat. Biotechnol. 2002, 20, 251–255. 80. Lai,
L.; Kolber-Simonds, D.; Park, K.W.; Cheong, H.T.;
Greenstein, J.L.; Im, G.S.; Samuel, M.; Bonk, A.; Rieke,
A.; Day, B.N.; Murphy, C.N.; Carter, D.B.; Hawley, R.J.;
Prather, R.S. Production of a-1,3-galactosyltransferase
knockout pigs by nuclear transfer cloning. Science 2002,
295, 1089–1092. 81. Lai, L.; Prather, R.S. Progress in
producing knockout models for xenotransplantation by
nuclear transfer. Ann. Med. 2002, 34, 501–506. 82. Onishi,
A.; Iwamoto, M.; Akita, T.; Mikawa, S.; Takeda, K.; Awata,
T.; Hanada, H.; Perry, A.C.F. Pig cloning by microinjection
of fetal fibroblast nuclei. Science 2000, 289, 1188–1190.
83. Polejaeva, I.A.; Chen, S.; Vaught, T.D.; Page, R.L.;
Mullins, J.; Ball, S.; Dai, Y.; Boone, J.; Walker, S.;
Ayares, D.L.; Colman, A.; Campbell, K.H.S. Cloned piglets
produced by nuclear transfer from adult somatic cells.
Nature 2000, 407, 86–90. 84. Betthauser, J.; Forsberg, E.;
Augenstein, M.; Childs, L.; Eilertsen, K.; Enos, J.;
Forsythe, T.; Golueke, P.; Jurgella, G.; Koppang, R.;
Lesmeister, T.; Mallon, K.; Mell, G.; Misica, P.; Pace, M.;
Pfister-Genskow, M.; Strelchenko, N.; Voelker, G.; Watt, S.;
Thompson, S.; Bishop, M. Production of cloned pigs from in
vitro systems. Nat. Biotechnol. 2000, 18, 1055–1059. 85.
Sharma, A.; Naziruddin, B.; Cui, C.; Martin, M.J.; Xu, H.;
Wan, H.; Lei, Y.; Harrison, C.; Yin, J.; Okabe, J.;
Mathews, C.; Stark, A.; Adams, C.S.; Houtz, J.; Wiseman,
B.S.; Byrne, G.W.; Logan, J.S. Pig cells that lack the gene
for alpha1-3 galactosyltransferase express low levels of
the gal antigen. Transplantation 2003, 75, 430–436. 86.
Nottle, M.B.; D’Apice, A.J.; Cowan, P.J.; Boquest, A.C.;
Harrison, S.J.; Grupen, C.G. Transgenic perspectives in
xenotransplantation, 2001. Xenotransplantation 2002, 9,
305–308. 87. Brunelli, J.P.; Pall, M.L. Lambda/plasmid
vector construction by in vivo cre/lox-mediated
recombination. BioTechniques 1994 , 16, 1060–1064. 88.
Sandrin, M.S.; Fodor, W.L.; Mouhtouris, E.; Osman, N.;
Cohney, S.; Rollins, S.A.; Guilmette, E.R.; Setter, E.;
Squinto, S.P.; McKenzie, I. Enzymatic remodelling of the
carbohydrate surface of a xenogeneic cell substantially
reduces human antibody binding and complement-mediated
cytolysis. Nat. Med. 1995, 1, 1261–1267. 89. Sharma, A.;
Okabe, J.F.; Birch, P.; Platt, J.L.; Logan, J.S. Reduction
in the level of Gal (a1,3) Gal in transgenic mice and pigs
by the expression of an a(1,2) fucosyltransferase. Proc.
Natl. Acad. Sci. U.S.A. 1996, 93, 7190–7195. 90. Crick, S.;
Sheppard, M.N.; Ho, S.Y.; Gebstein, L.; Anderson, R.H.
Anatomy of the pig heart: comparisons with normal human
cardiac structure. J. Anat. 1998, 193, 105–119. 91. Crick,
S.J.; Sheppard, M.N.; Ho, S.Y.; Anderson, R.H. Localisation
and quantitation of autonomic innervation in the porcine
heart I: conduction system. J. Anat. 1999, 195, 341–357. T
Transparent Ceramics: Bioactive Materials
and Tissue Engineering

11. Ohgushi, H.; Okumura, M.; Tamai, S.; Shors, E.C.;


Caplan, A.I. Marrow cell induced osteogenesis in porous
hydroxyapatite and tricalcium phosphate. J. Biomed. Mater.
Res. 1990, 24, 1563–1570.

12. Ohgushi, H.; Okumura, M.; Yoshikawa, T.; Inoue, K.;


Senpuku, N.; Tamai, S. Bone formation process in porous
calcium carbonate and hydroxyapatite. J. Biomed. Mater.
Res. 1992, 26, 885–895.

13. Kihara, T.; Oshima, A.; Hirose, M.; Ohgushi, H.


Threedimensional visualization analysis of in vitro
cultured bone fabricated by rat marrow mesenchymal stem
cells. Biochem. Biophys. Res. Commun. 2004, 316, 943–948.

14. Mendes, S.C.; Tibbe, J.M.; Veenhof, M.; Bakker, K.;


Both, S.; Platenburg, P.P.; Oner, F.C.; De Bruijn, J.D.;
Van Blitterswijk, C.A. Bone tissue-engineered implants
using human bone marrow stromal cells: effect of culture
conditions and donor age. Tissue Eng. 2002, 8, 911–920.

15. de Bruijn, J.D.; van den Brink, I.; Mendes, S.; Dekker,
R.; Bovell, Y.P.; van Blitterswijk, C.A. Bone induction by
implants coated with cultured osteogenic bone marrow cells.
Adv. Dent. Res. 1999, 13, 74–81.

16. Kasten, P.; Luginbuhl, R.; van Griensven, M.;


Barkhausen, T.; Krettek, C.; Bohner, M.; Bosch, U.
Comparison of human bone marrow stromal cells seeded on
calcium-deficient hydroxyapatite, beta-tricalcium phosphate
and demineralized bone matrix. Biomaterials 2003, 24,
2593–2603.

17. Mayr-Wohlfart, U.; Fiedler, J.; Gunther, K.P.; Puhl,


W.; Kessler, S. Proliferation and differentiation rates of
a human osteoblasts-like cell line (SaOS-2) in contact with
different bone substitute materials. J. Biomed. Mater. Res.
2001, 57, 132–139.

18. Ioku, K.; Kawagoe, D.; Toya, H.; Fujimori, H.; Goto,
S.; Ishida, K.; Mikuni, A.; Mae, H. OH-designed transparent
apatite ceramics prepared by spark plasma sintering. Trans.
Mater. Res. Soc. Jpn. 2002, 27, 447– 449.

19. Kotobuki, N.; Ioku, K.; Kawagoe, D.; Fujimori, H.;


Goto, S.; Ohgushi, H. Observation of osteogenic
differentiation cascade of living mesenchymal stem cells on
transparent hydroxyapatite ceramics. Biomaterials 2005, 26,
779–785.

20. Kotobuki, N.; Kawagoe, D.; Nomura, D.; Kato, Y.;


Muraki, K.; Fujimori, H.; Goto, S.; Ioku, K.; Ohgushi, H.
Observation and quantitative analysis of rat mesenchymal
stem cells cultured in vitro on newly formed transparent
b-tricalcium phosphate. J. Mater. Sci. in press. 21.
Maniatopoulos, C.; Sodek, J.; Melcher, A.H. Bone formation
in vitro by stromal cells obtained from bone marrow of
young adult rat. Cell. Tissue Res. 1988, 254, 317–330. 22.
Uchimura, E.; Machida, H.; Kotobuki, N.; Kihara, T.;
Kitamura, S.; Ikeuchi, M.; Hirose, M.; Miyake, J.; Ohgushi,
H. In-situ visualization and quantification of
mineralization of cultured osteogenetic cells. Calcif.
Tissue Int. 2003, 73 , 575–583. 23. Ohgushi, H.; Okumura,
M. Osteogenic ability of rat and human marrow cells in
porous ceramics. Acta. Orthop. Scand. 1990, 61, 431–434.
24. Okumura, M.; Ohgushi, H.; Tamai, S. Bonding
osteogenesis in coralline hydroxyapatite combined with bone
marrow cells. Biomaterials 1991, 12, 411–416. 25. Goshima,
J.; Goldberg, V.M.; Caplan, A.I. Osteogenic potential of
culture-expanded rat marrow cells as assayed in in vitro
with porous calcium phosphate ceramic. Biomaterials 1991,
12, 254–258. 26. Yoshikawa, T.; Ohgushi, H.; Uemura, T.
Human marrow cell-derived cultured bone in porous ceramics.
J. Biomed. Mater. Eng. 1998, 8, 311–320. 27. Ohgushi, H.;
Yoshikawa, T.; Nakajima, H.; Tamai, S.; Dohi, Y.; Okunaga,
K. Al 2 O 3 doped apatite-wollastonite containing glass
ceramic provokes osteogenic differentiation of marrow
stromal stem cells. J. Biomed. Mater. Res. 1999, 44,
381–388. 28. Yoshikawa, T.; Ohgushi, H.; Dohi, Y.; Davies,
J.E. Viable bone formation in porous hydroxyapatite: marrow
cell-derived in vitro bone on the surface of ceramics.
Biomed. Mater. Eng. 1997, 7, 49–58. 29. Kitamura, S.;
Ohgushi, H.; Hirose, M.; Funaoka, H.; Takakura, Y.; Ito, H.
Osteogenic differentiation of human bone marrow-derived
mesenchymal cells cultured on alumina ceramics. Artif.
Organs 2004, 28, 72–82. 30. Ohgushi, H.; Kitamura, S.;
Kotobuki, N.; Hirose, M.; Machida, H.; Muraki, K.;
Takakura, Y. Clinical application of marrow mesenchymal
stem cells for hard tissue repair. Yonsei Med. J. 2004, 45,
61–67. 31. Kotobuki, N.; Hirose, M.; Takakura, Y.; Ohgushi,
H. Cultured autologous human cells for hard tissue
regeneration: preparation and characterization of
mesenchymal stem cells from bone marrow. Artif. Organs
2004, 28, 33–39. 32. Ohgushi, H.; Kotobuki, N.; Funaoka,
H.; Machida, H.; Hirose, M.; Tanaka, Y.; Takakura, Y.
Tissue engineered ceramic artificial joint -ex vivo
osteogenic differentiation of patient mesenchymal cells on
total ankle joints for treatment of osteoarthritis.
Biomaterials 2005, 26, 4654–4661. Transparent
Ultra-High Molecular Weight Polyethylene
(UHMWPE)

28. Turell, M.; Bellare, A.; Gomoll, A. Fracture Toughness


of Radiation Cross-Linked Ultra-high Molecular Weight
Polyethylene. Transactions of the 48th Annual Meeting of
the Orthopaedic Research Society, Dallas, TX, USA, , 2002;
1034

29. Cole, J.C.; Lemons, J.E.; Eberhardt, A.W. Gamma


irradiation alters fatigue-crack behavior and fracture
toughness in 1900H and GUR 1050 UHMWPE. J. Biomed. Mater.
Res. (Appl. Biomater.) 2002, 63, 559–566. 30. Gsell, R.;
Yao, J.Q.; Laurent, M.P.; Crowninshield, R.D. Improved
Oxidation Resistance of Highly Crosslinked UHMWPE for Total
Knee Arthroplasty. Transactions of the 27th Annual Meeting
of the Society for Biomaterials, St. Paul, MN, USA, , 2001;
84
Ultrasound and Tissue Interaction

1. Goldberg, B.B.; Kimmelman, B.A. Medical Diagnostic


Ultrasound: A Retrospective on its 40th Anniversary; AIUM:
Laurel, MD, 1988.

2. Shung, K.K.; Smith, M.B.; Tsui, B.W.N. Principles of


Medical Imaging; Academic Press: San Diego, 1992.

3. Miller, M.M. Gene transfection and drug delivery.


Ultrasound Med. Biol. 2000, 26, S59–S62.

4. Kossoff, G. Rradiation Force. In Interaction of


Ultrasound and Biological Tissues; Reid, J.M., Sikov, M.R.,
Eds.; FDA: Rockvlle, MD, 1972; 159–161.

5. Nightingale, K.R.; Nightingale, R.W.; Palmeri, M.L.;


Trahey, G.E. Finite Element Analysis of Radiation Force
Induced Tissue Motion with Experimental Validation. In Proc
1999 IEEE Ultrasonics Symp.; Schneider, S.C., Levy, M.,
McVoy, B.R., Eds.; IEEE: New York, 1999; 1319–1323. 6.
Fields, S.; Dunn, F. Correlation of echographic
visualization of tissue with biological composition and
physiological state. J. Acoust. Soc. Am. 1972, 54, 809–811.
7. Shung, K.K.; Thieme, G.A. Ultrasonic Scattering by
Biological Tissues; CRC Press: Boca Raton, FL, 1993. 8.
Greenleaf, J.A. Tissue Characterization with Ultrasound;
CRC Press: Boca Raton, FL, 1986. 9. Morse, P.M.; Ingard,
K.U. Theoretical Acoustics; McGraw Hill: New York, 1968.
10. Wickline, S.A.; Perez, J.E.; Miller, J.G.
Cardiovascular Tissue Characterization in vivo. In
Ultrasonic Scattering in Biological Tissues; Shung, K.K.,
Thieme, G.A., Eds.; CRC Press: Boca Raton, FL, 1993;
313–345. 11. Hamilton, M.F.; Blackstock, D.T. Nonlinear
Acoustics; Academic Press: San Diego, 1998. 12. Tranquart,
F.; Grenier, N.; Eder, V.; Pourcelot, L. Clinical use of
ultrasound tissue harmonic imaging. Ultrasound Med. Biol.
1999, 25, 889–894. 13. Pierce, A. Acoustics: An
Introduction to Physical Principles and Applications;
McGraw Hill: New York, 1986. 14. Lighton, T.G. The Acoustic
Bubble; Academic Press: London, 1994. 15. ter Harr, G.R.
Therapeutic ultrasound—An overview. Europ. J. Ultrasound
1999, 9, 3–9. 16. Mitragotri, S.; Blankschtein, D.; Langer,
R. Ultrasoundmediated transdermal protein delivery. Science
1995, 269, 850–853. U
Ultrasound Doppler

8. Fei, D.Y.; Fu, C.T. New method to obtain angle


independent Doppler color images using a sector transducer.
Ann. Biomed. Eng. 1999, 27, 187–193.

9. Hoskins, P.R. A comparison of singleand dual-beam


methods for maximum velocity estimation. Ultrasound Med.
Biol. 1999, 25, 583–592.

10. Arslan, H.; Sakarya, M.E.; Bozkurt, M.; Unal, O.;


Dilek, O.N.; Harman, M. The role of power Doppler
sonography in the evaluation of superficial soft tissue
abscesses. Eur. J. Ultrasound 1998, 8, 101–106.

11. Ophir, J.; Parker, K.J. Contrast agents in diagnostic


ultrasound. Ultrasound Med. Biol. 1989, 15, 319–333.

12. Mitchell, D.G.; Merton, D.A.; Liu, J.B.; Goldberg, B.B.


Superficial masses with color Doppler imaging. J. Clin.
Ultrasound 1991, 19, 555–560.

13. McDicken, W.N.; Sutherland, G.R.; Gordon, L.N. Color


Doppler velocity imaging of the myocardium. Ultrasound Med.
Biol. 1992, 18, 651–654.

14. National Advisory Neurological and Communicative


Disorders and Stroke Council. Decade of the Brain: Answers
Through Scientific Research; U.S. Dept. of Health and Human
Services, Public Health Service, National Institutes of
Health: Bethesda, MD, 1989. NLM Unique ID: 8912912.

15. European Carotid Surgery Trialists’ Collaborative Group


Interim results for symptomatic patients with severe
(70–99%) or with mild (0–29%) carotid stenosis. MRC
European Carotid Surgery Trial. Lancet 1991, 337,
1235–1243. 16. North American Symptomatic Carotid
Endarterectomy Trial Collaborators Beneficial effect of
carotid endarterectomy in symptomatic patients with
high-grade carotid stenosis. N. Engl. J. Med. 1991, 325,
445–453. 17. Executive Committee for the Asymptomatic
Carotid Atherosclerosis Study. Endarterectomy for
asymptomatic carotid artery stenosis. JAMA 1995, 273,
1421–1428. 18. Aaslid, R.; Lindegaard, K.F. Transcranial
Doppler Sonography; Aaslid, R., Ed.; Wien, Springer-Verlag,
New York, 1986. 19. Yao, S.T. Haemodynamic studies in
peripheral arterial disease. Br. J. Surg. 1970, 57,
761–766. 20. Vogt, M.T.; Cauley, J.A.; Newman, A.B.;
Kuller, L.H.; Hulley, S.B. Decreased ankle/arm blood
pressure index and mortality in elderly women. JAMA 1993,
270, 465–469. 21. Phillips, J.H. Practical Quantitative
Doppler Echocardiography; CRC Press, Inc.: Boca Raton, FL,
1991. 22. White, E.M.; Choyke, P.L. Duplex Sonography of
the Abdomen. In Duplex Sonography; Grant, E.G., White,
E.M., Eds.; Springer-Verlag: New York, 1988; 129–190. 23.
Sallomi, D. The use of contrast-enhanced power Doppler
ultrasound in the diagnosis and follow-up of inflammatory
abdominal masses associated with Crohn’s disease. Eur. J.
Gastroenterol. Hepatol. 2003, 15, 249–251.
Ultrasound Therapy, Bone Healing

23. Reher, P.; Doan, N.; Bradnock, B.; Meghji, S.; Harris,
M. Effect of ultrasound on the production of IL-8, basic
FGF and VEGF. Cytokine 1999, 11 (6), 416–423.

24. Rawool, N.M.; Goldberg, B.B.; Forsberg, F; Winder,


A.A.; Hume, E. Power Doppler assessment of vascular changes
during fracture treatment with low-intensity ultrasound. J.
Ultrasound Med. 2003, 22 (2), 145–153.

25. Yang, K.H.; Parvizi, J.; Wang, S.J.; Lewallen, D.G.;


Kinnick, R.R.; Greenleaf, J.F.; Bolander, M.E. Exposure to
low-intensity ultrasound increases aggrecan gene expression
in a rat femur fracture model. J. Orthop. Res. 1996, 14
(5), 802–809.

26. Wang, S.J.; Lewallen, D.G.; Bolander, M.E.; Chao, E.Y.;


Ilstrup, D.M.; Greenleaf, J.F. Low intensity ultrasound
treatment increases strength in a rat femoral fracture
model. J. Orthop. Res. 1994, 12 (1), 40–47.

27. Hantes, M.E.; Mavrodontidis, A.N.; Zalavras, C.G.;


Karantanas, A.H.; Karachalios, T.; Malizos, K.N.
Low-intensity transosseous ultrasound accelerates osteotomy
healing in a sheep fracture model. J. Bone Joint Surg. Am.
2004, 86 (10), 2275–2282.

28. Mayr, E.; Rudzki, M.M.; Rudzki, M.; Borchardt, B.;


Hausser, H.; Ruter, A. Does low intensity, pulsed
ultrasound speed healing of scaphoid fractures? Handchir.
Mikrochir. Plast. Chir. 2000, 32, 115–122.

29. Banken, R. Low-intensity ultrasound (Exogent) for the


treatment of fractures, Technology brief; Agence
d’e´valuation des technologies et des modes d’intervention
en sante´ (AETMIS). AETMIS 03-05, Montreal, Canada, , 2004;
1–15.

30. Mayr, E.; Frankel, V.; Ruter, A. Ultrasound—An


alternative healing method for nonunions? Arch. Orthop.
Trauma Surg. 2000, 120 (1–2), 1–8.

31. Nolte, P.A.; van der Krans, A.; Patka, P.; Janssen,
I.M.; Ryaby, J.P.; Albers, G.H. Low-intensity pulsed
ultrasound in the treatment of nonunions. J. Trauma 2001,
51 (4), 693–702.

32. Tsumaki, N.; Kakiuchi, M.; Sasaki, J.; Ochi, T.;


Yoshikawa, H. Low-intensity pulsed ultrasound accelerates
maturation of callus in patients treated with opening-wedge
high tibial osteotomy by hemicallotasis. J. Bone Joint
Surg. Am. 2004, 86 (11), 2399–2405.

33. El-Mowafi, H.; Mohsen, M. The effect of low-intensity


pulsed ultrasound on callus maturation in tibial
distraction osteogenesis. Int. Orthop. 2005, 29 (2),
121–124.

34. Tanzer, M.; Kantor, S.; Bobyn, J.D. Enhancement of bone


growth into porous intramedullary implants using
non-invasive low intensity ultrasound. J. Orthop. Res.
2001, 19 (2), 195–199. 35. Harris, M. The conservative
management of osteoradionecrosis of the mandible with
ultrasound therapy. Br. J. Oral Maxillofac. Surg. 1992, 30,
313–318. 36. El-Bialy, T.H.; Royston, T.J.; Magin, R.L.;
Evans, C.A.; Zaki, Ael-M.; Frizzell, L.A. The effect of
pulsed ultrasound on mandibular distraction. Ann. Biomed.
Eng. 2002 , 30 (10), 1251–1256. 37. El-Bialy, T.; El-Shamy,
I.; Graber, T.M. Repair of orthodontically induced root
resorption by ultrasound in humans. Am. J. Orthod.
Dentofacial Orthop. 2004, 126 (2), 186–193. 38. Erdogan,
O.; Esen, E.; Ustun, Y.; Kurkcu, M.; Akova, T.; Gonlusen,
G.; Uysal, H.; Cevlik, F. Effects of lowintensity pulsed
ultrasound on healing of mandibular fractures: An
experimental study in rabbits. J. Oral Maxillofac. Surg.
2006, 64 (2), 180–188. 39. Fedotov, S.N.; Minin, E.A.;
Borisov, I.N. Effect of local cooling and ultrasound on the
reparative processes following mandibular fracture.
Stomatologiia 1986, 65 (4), 4–6. 40. El-Bialy, T.H.;
el-Moneim, Z.A.; Evans, C.A. Effect of ultrasound on rabbit
mandibular incisor formation and eruption after mandibular
osteodistraction. Am. J. Orthod. Dentofacial Orthop. 2003,
124, 427–434. 41. El-Bialy, T.H.; El-Shamy, I.; Graber,
T.M. Growth modification of the rabbit mandible using
therapeutic ultrasound: Is it possible to enhance
functional appliance results? Angle Orthod. 2003, 73,
631–639. 42. Schortinghuis, J.; Ruben, J.L.; Raghoebar,
G.M.; Stegenga, B. Ultrasound to stimulate mandibular bone
defect healing: A placebo-controlled single-blind study in
rats. J. Oral Maxillofacial Surg. 2004, 62, 194–201. 43.
Schortinghuis, J.; Ruben, J.L.; Raghoebar, G.M.; Stegenga,
B. Therapeutic ultrasound to stimulate osteoconduction; A
placebo controlled single blind study using e-PTFE
membranes in rats. Arch. Oral Biol. 2004, 49, 413–420. 44.
Schortinghuis, J.; Bronckers, A.L.; Stegenga, B.;
Raghoebar, G.M.; de Bont, L.G. Ultrasound to stimulate
early bone formation in a distraction gap: A double blind
randomised clinical pilot trial in the edentulous mandible.
Arch. Oral Biol. 2005, 50, 411–420. 45. El-Bialy, T.;
Hassan, A.; Albaghdadi, T.; Fouad, H.A.; Maimani, A.R.
Growth modification of the mandible with ultrasound in
baboons: A preliminary report. Am. J. Orthod. Dentofacial
Orthop. 2006, 130 (4), 435–437. 46. Young, S.R.; Dyson, M.
Effect of therapeutic ultrasound on the healing of
full-thickness excised skin lesions. Ultrasonic. 1990, 28
(3), 175–180.
Urinary Prostheses

16. Falke, G.; Yoo, J.J.; Kwon, T.G.; Moreland, R.; Atala,
A. Formation of corporal tissue architecture in vivo using
human cavernosal muscle and endothelial cells seeded on
collagen matrices. Tissue Eng. 2003, 9, 871.

17. Kwon, T.G.; Yoo, J.J.; Atala, A. Autologous penile


corpora cavernosa replacement using tissue engineering
techniques. J. Urol. 2002, 168, 1754.

18. Holroyd-Leduc, J.M.; Straus, S.E. Management of urinary


incontinence in women: scientific review. JAMA 2004, 291,
986.

19. Brown, J.S.; Nyberg, L.M.; Kusek, J.W.; Burgio, K.L.;


Diokno, A.C.; Foldspang, A.; et al. Proceedings of the
National Institute of Diabetes and Digestive and Kidney
Diseases International Symposium on Epidemiologic Issues in
Urinary Incontinence in Women. Am. J. Obstet. Gynecol.
2003, 188, S77.

20. Burgio, K.L.; Matthews, K.A.; Engel, B.T. Prevalence,


incidence and correlates of urinary incontinence in
healthy, middle-aged women. J. Urol. 1991, 146, 1255.

21. Nygaard, I.E.; Heit, M. Stress urinary incontinence.


Obstet. Gynecol. 2004, 104, 607.

22. Wilson, L.; Brown, J.S.; Shin, G.P.; Luc, K.O.; Subak,
L.L. Annual direct cost of urinary incontinence. Obstet.
Gynecol. 2001, 98, 398.

23. Siegel, S.W. History of the prosthetic treatment of


urinary incontinence. Urol. Clin. North Am. 1989, 16, 99.

24. Martins, F.E.; Boyd, S.D. Artificial urinary sphincter


in patients following major pelvic surgery and/or
radiotherapy: are they less favorable candidates? J. Urol.
1995, 153, 1188.

25. Gousse, A.E.; Madjar, S.; Lambert, M.M.; Fishman, I.J.


Artificial urinary sphincter for post-radical prostatectomy
urinary incontinence: long-term subjective results. J.
Urol. 2001, 166, 1755.

26. Donovan, M.G.; Barrett, D.M.; Furlow, W.L. Use of the


artificial urinary sphincter in the management of severe
incontinence in females. Surg. Gynecol. Obstet. 1985, 161,
17.
27. Webster, G.D.; Perez, L.M.; Khoury, J.M.; Timmons, S.L.
Management of type III stress urinary incontinence using
artificial urinary sphincter. Urology 1992, 39, 499.

28. Haab, F.; Trockman, B.A.; Zimmern, P.E.; Leach, G.E.


Quality of life and continence assessment of the artificial
urinary sphincter in men with minimum 3.5 years of
followup. J. Urol. 1997, 158, 435. 29. Litwiller, S.E.;
Kim, K.B.; Fone, P.D.; White, R.W.; Stone, A.R.
Post-prostatectomy incontinence and the artificial urinary
sphincter: a long-term study of patient satisfaction and
criteria for success. J. Urol. 1975, 156, 1996. 30.
Chancellor, M.B.; Yokoyama, T.; Tirney, S.; Mattes, C.E.;
Ozawa, H.; Yoshimura, N.; et al. Preliminary results of
myoblast injection into the urethra and bladder wall: a
possible method for the treatment of stress urinary
incontinence and impaired detrusor contractility.
Neurourol. Urodyn. 2000, 19, 279. 31. Cannon, T.W.; Lee,
J.Y.; Somogyi, G.; Pruchnic, R.; Smith, C.P.; Huard, J.; et
al. Improved sphincter contractility after allogenic
muscle-derived progenitor cell injection into the
denervated rat urethra. Urology 2003, 62, 958. 32. Lee,
J.Y.; Cannon, T.W.; Pruchnic, R.; Fraser, M.O.; Huard, J.;
Chancellor, M.B. The effects of periurethral muscle-derived
stem cell injection on leak point pressure in a rat model
of stress urinary incontinence. Int. Urogynecol. J. Pelvic
Floor Dysfunct. 2003, 14, 31. 33. Chen, F.; Yoo, J.J.;
Atala, A. Acellular collagen matrix as a possible ‘‘off the
shelf’’ biomaterial for urethral repair. Urology 1999, 54,
407. 34. Yiou, R.; Yoo, J.J.; Atala, A. Restoration of
functional motor units in a rat model of sphincter injury
by muscle precursor cell autografts. Transplantation 2003,
76, 1053. 35. Strasser, H.; Berjukow, S.; Marksteiner, R.;
Margreiter, E.; Hering, S.; Bartsch, G.; et al. Stem cell
therapy for urinary stress incontinence. Exp. Gerontol.
2004, 39, 1259. 36. Girdansky, J.; Newman, H. Use of a
vitallium testicular implant. Am. J. Surg. 2005, 53 37.
Lattimer, J.K.; Vakili, B.F.; Smith, A.M.; Morishima, A. A
natural-feeling testicular prosthesis. J. Urol. 1973, 110,
81. 38. Lipworth, L.; Tarone, R.E.; McLaughlin, J.K.
Silicone breast implants and connective tissue disease: an
updated review of the epidemiologic evidence. Ann. Plast.
Surg. 2004, 52, 598. 39. Turek, P.J.; Master, V.A. Safety
and effectiveness of a new saline filled testicular
prosthesis. J. Urol. 2004, 172, 1427.
Vascular Fluid Dynamics

1. Burton, A.C. Physiology and Biophysics of the


Circulation; Year Book Medical Publishers: Chicago, 1965.

2. Womersley, J.R. An Elastic Tube Theory of Pulse


Transmission and Oscillatory Flow in Mammalian Arteries,
WADC Technical Report TR-56-6l4; Wright Air Development
Center: Dayton, Ohio, 1957.

3. McDonald, D.A. Blood Flow in Arteries, 2nd Ed.; Williams


& Wilkins: Baltimore, 1974.

4. Pedley, T.J. The Fluid Mechanics of Large Blood Vessels;


Cambridge University Press, 1980.

5. Berger, S.A. Flow in large blood vessels. In


Contemporary Mathematics; Cheer, A.Y., van Dam, C.P., Eds.;
American Mathematical Society: Providence, RI, 1993; Vol.
141, 479–518.

6. Taylor, C.A.; Draney, M.T. Experimental and


computational methods in cardiovascular fluid mechanics.
Ann. Rev. Fluid Mech. 2004, 36, 197–231.
Vascular Grafts

1. Zdrahala, R. Small caliber vascular grafts. Part I:


State of the art. J. Biomater. Appl. 1996, 10 (4), 309–329.

2. Niklason, L.E.; Langer, R.S. Advances in tissue


engineering of blood vessels and other tissues. Transpl.
Immunol. 1997, 5 (4), 303–306.

3. National Center for Health Statistics. National Hospital


Discharge Survey. In Vital Health Statistics; Hyattsville,
Massachusetts, 1990–1999.

4. Wesolow, A. Biological Behaviour of Tissue and


Prosthetic Grafts. In Vascular Surgery. Principles and
Techniques; Haimovici, H., Ed.; Appleton-CenturyCrofts:
Norwark, CT, 1984; 93–118.

5. Bos, G.; Poot, A.; Beugeling, T.; van Aken, W.; Feijen,
J. Small-diameter vascular graft prostheses: Current
status. Arch. Physiol. Biochem. 1998, 106 (2), 100–115.

6. Dardik, H. Biological Behaviour of Glutaraldehyde—


Stabilized Human Umbilical Cord Vein Grafts. In Vascular
Surgery. Principles and Techniques; Haimovici, H., Ed.;
Appleton-Century-Crofts: Norwark, CT, 1984.

7. Sauvage, L.; Wesolowski, S. Healing and fate of arterial


grafts. Surgery 1955, (38), 1090.

8. Butt, K. Bovine Heterograft for Arteriovenous Fistula.


In Vascular Grafting; Sawyer, P., Kaplitt, M., Eds.;
Appleton-Century-Crofts: New York, 1978; 278–281.

9. Zdrahala, R. Small caliber vascular grafts. Part II:


Polyurethanes revisited. J. Biomater. Appl. 1996, 11 (1),
37–61.

10. Fox, D.; Vorp, D.; HP, G. Bioresorbable Grafts: A


Counterintuitive Approach. In Tissue Engineering of
Vascular Prosthetic Grafts; Zilla, P., Greisler, H.P.,
Eds.; R.G. Landes; Texas, 1999; 489–503.

11. King, M.; Blais, P.; Guidoin, R.; Prowse, E.; Marcois,
M.; Gosselin, C.; Noel, H. Polyethylene Terephthalate
(Dacron) Vascular Prostheses—Material and Fabric
Construction Aspects. In Biocompatibility of Clinical
Implant Materials; CRC Press, 1981; 177–207. 12. von
Oppell, U.; Zilla, P. Tissue adhesives in cardiovascular
surgery. J. Long-Term Eff. Med. Implants 1998, 8 (2),
87–101. 13. Sladen, J.; Gilmour, J.; Wong, R. Cumulative
patency and actual palliation in patients with claudication
after aortofemoral bypass. Prospective long-term follow-up
of 100 patients. Am. J. Surg. 1986, 152 (2), 190–195. 14.
Veith, F.J.; Gupta, S.K.; Ascer, E.; White-Flores, S.;
Samson, R.H.; Scher, L.A.; Towne, J.B.; Bernhard, V.M.;
Bonier, P.; Flinn, W.R.; Astelford, P.; Yao, J.S.T.;
Bergan, J.J. Six-year prospective multicenter randomized
comparison of autologous saphenous vein and expanded
polytetrafluoroethylene grafts in infrainguinal arterial
reconstructions. J. Vasc. Surg. 1986, 3 (1), 104–114. 15.
Bezuidenhout, D.; Davies, N.; Zilla, P. Effect of
welldefined dodecahedral porosity on inflammation and
neo-vascularization. ASAIO J. 2002, 48, 465–471. 16. Nam,
Y.; Park, T. Porous biodegradable polymeric scaffolds
prepared by thermally induced phase separation. J. Biomed.
Mater. Res. 1999, 47 (1), 8–17. 17. Tiwari, A.; Salacinski,
H.; Seifalian, A.; Hamilton, G. New prostheses for use in
bypass grafts with special emphasis on polyurethanes.
Cardiovasc. Surg. 2002 , 10 (3), 191–197. 18. Eberhart, A.;
Zhang, Z.; Guidoin, R.; Laroche, G.; Guay, L.; De La Faye,
D.; Batt, M.; King, M.W. A new generation of polyurethane
vascular prostheses: Rara avis or ignis fatuus? J. Biomed.
Mater. Res. 1999, 48 (4), 546–558. 19. Cardiovascular
Implants—Tubular Vascular Prostheses; Association for the
advancement of Medical Instrumentation, 1998. 20.
Haimovici, H. History of Vascular Surgery. In Vascular
Surgery. Principles and Techniques; Haimovici, H., Ed.;
Appleton-Century-Crofts: Norwark, CT, 1984; 3–18. 21.
Voorhees, A.; Jaretzki, A.; AH, B. The use of tubes
constructed from Vinyon ‘‘N’’ cloth in bridging arterial
defects. Ann. Surg. 1952, 135, 332. 22. Wesolowski, S.
Foundations of Modern Vascular Grafts. In Vascular Grafts;
Sawyer, P., Kaplitt, M., Eds.; Appleton Century Croft: New
York, 1978. 23. Okoshi, T.; Soldani, G.; Goddard, M.;
Galletti, P. Penetrating micropores increase patency and
achieve extensive endothelialization in small diameter
polymer skin coated vascular grafts. ASAIO J. 1996, 42 (5),
M398–401. 24. Davids, L.; Dower, T.; Zilla, P. The Lack of
Healing in Conventional Vascular Grafts. In Tissue
Engineering of Vascular Grafts; Zilla, P., Greisler, H.,
Eds.; RG Landes: Austin, 1999; 3–44. 25. Seifalian, A.;
Giudiceandrea, A.; Schmitz-Rixen, T. Noncompliance: The
Silent Acceptance of a Villain. In Tissue Engineering of
Vascular Prosthetic Grafts; P, Z., HP, G., Eds.; R.G.
Landes: Texas, 1999. 26. Stefanadis, C.; Stratos, C.;
Boudoulas, H.; Kourouklis, C.; Toutouzas, P. Distensibility
of the ascending aorta: Comparison of invasive and
non-invasive techniques in healthy men and in men with
coronary artery disease. Eur. Heart J. 1990, 11 (11),
990–996. 27. Kawasaki, T.; Sasayama, S.; Yagi, S.; Asakawa,
T.; Hirai, T. Non-invasive assessment of the age related
changes in stiffness of major branches of the human
arteries. Cardiovasc. Res. 1987, 21, 678–687. 28. Anderson,
J.; Gristina, A.; Hanson, S.; Harker, L.; Johnson, R.;
Merritt, K.; Naylor, P.; Schoen, F. Host Reaction to
Biomaterials and Their Evaluation. In Biomaterials Science:
An Introduction to Materials in Medicine; Ratner, B.,
Hoffman, A., Schoen, F., Lemons, J., Eds.; Academic Press:
San Diego, 1996. 29. Yun, J.; DeFife, K.; Colton, E.;
Stack, S.; Azeez, A.; Cahalan, L.; Verhoeven, M.; Cahalan,
P.; Anderson, J. Human monocyte/macrophage adhesion and
cytokine production on surface-modified
poly(tetrafluoroethylene/hexafluoropropylene) polymers with
and without protein preadsorption. J. Biomed. Mater. Res.
1995, 29 (2), 257–268. 30. Park, K.; Kim, Y.; Han, D.; Kim,
Y.; Lee, E.; Suh, H.; Choi, K. Bacterial adhesion on PEG
modified polyurethane surfaces. Biomaterials 1998, 19 (7–9),
851–859. 31. Lee, J.; Ju, Y.; Lee, W.; Park, K.; Kim, Y.
Platelet adhesion onto segmented polyurethane surfaces
modified by PEOand sulfonated PEO-containing block copolymer
additives. J. Biomed. Mater. Res. 1998, 40 (2), 314–323.
32. San Roman, J.; Bujan, J.; Bellon, J.; Gallardo, A.;
Escudero, M.; Jorge, E.; de, H.J.; Alvarez, L.; Castillo,
O.J. Experimental study of the antithrombogenic behavior of
Dacron vascular grafts coated with hydrophilic acrylic
copolymers bearing salicylic acid residues. J. Biomed.
Mater. Res. 1996, 32 (1), 19–27. 33. Phaneuf, M.; Berceli,
S.; Bide, M.; Quist, W.; LoGerfo, F. Covalent linkage of
recombinant hirudin to poly (ethylene terephthalate)
(Dacron): Creation of a novel antithrombin surface.
Biomaterials 1997, 18 (10), 755–765. 34. Aldenhoff, Y.;
van, D.V.F.; ter, W.J.; Habets, J.; Poole, W.L.; Koole, L.
Performance of a polyurethane vascular prosthesis carrying
a dipyridamole (Persantin) coating on its lumenal surface.
J Biomed Mater Res 2001, 54 (2), 224–233. 35. Yoneyama, T.;
Sugihara, K.; Ishihara, K.; Iwasaki, Y.; Nakabayashi, N.
The vascular prosthesis without pseudointima prepared by
antithrombogenic phospholipid polymer. Biomaterials 2002,
23 (6), 1455–1459. 36. Esquivel, C.; Bjorck, C.; Bergentz,
S.; Bergqvist, D.; Larsson, R.; Carson, S.; Dougan, P.;
Nilsson, B. Reduced thrombogenic characteristics of
expanded polytetrafluoroethylene and polyurethane arterial
grafts after heparin bonding. Surgery 1984, 95 (1),
102–107. 37. Kinoshita, Y.; Kuzuhara, T.; Kirigakubo, M.;
Kobayashi, M.; Shimura, K.; Ikada, Y. Soft tissue reaction
to collagen-immobilized porous polyethylene: Subcutaneous
implantation in rats for 20 wk. Biomaterials 1993, 14 (3),
209–215. 38. Krijgsman, B.; Seifalian, A.; Salacinski, H.;
Tai, N.; Punshon, G.; Fuller, B.; Hamilton, G. An
assessment of covalent grafting of RGD peptides to the
surface of a compliant poly(carbonate-urea)urethane
vascular conduit versus conventional biological coatings:
Its role in enhancing cellular retention. Tissue Eng. 2002,
8 (4), 673–680. 39. Kao, W.; Lee, D.; Schense, J.; Hubbell,
J. Fibronectin modulates macrophage adhesion and FBGC
formation: The role of RGD, PHSRN, and PRRARV domains. J.
Biomed. Mater. Res. 2001, 55 (79–88). 40. Hamdan, A.;
Misare, B.; Contreras, M.; LoGerfo, F.; Quist, W.
Evaluation of anastomotic hyperplasia progression using the
cyclin specific antibody MIB-1. Am. J. Surg. 1996, 172 (2),
168–170, discussion 170–161. 41. Zilla, P.; Fasol, R.;
Dudeck, U.; Siedler, S.; Preiss, P.; Fischlein, T.; Muller,
G.W.; Baitella, G.; Sanan, D.; Odell, J.; Reichart, B. In
situ cannulation, microgrid follow-up and low-density
plating provide first passage endothelial cell masscultures
for in vitro lining. J. Vasc. Surg. 1990, 12 (2), 180–189.
42. Sauvage, L.; Berger, K.; Wood, S.; Nakagawa, Y.;
Mansfield, P. An external velour surface for porous arterial
prostheses. Surgery 1971, 70 (6), 940–953. 43. Berger, K.;
Sauvage, L.; Rao, A.; Wood, S. Healing of arterial
prostheses in man: Its incompleteness. Ann. Surg. 1972, 175
(1), 118–127. 44. Hammond, W. Surface Population with
Blood-Borne Cells. In Tissue Engineering of Prosthetic
Vascular Grafts; Zilla, P., Greisler, H., Eds.; Landes
Bioscience: Austin, 1998. 45. Weinberg, C.; Bell, E. A
blood vessel model constructed from collagen and cultured
vascular cells. Science 1986, 231, 397–400. 46. L’Heureux,
N.; Germain, L.; Labbe, R.; Auger, F. In vitro construction
of a human blood vessel from cultured vascular cells; A
mophologic study. J. Vasc. Surg. 1993, 17 (499–509). 47.
Hirai, J.; Matsuda, T. Venous reconstruction using hybrid
vascular tissue composed of vascular cells and collagen
tissue regeneration process. Cell Transplant 1996, 5
(93–105). 48. L’Heureaux, N.; Paquet, S.; Labbe, R.;
Germain, L.; Auger, F. A completely biological
tissue-engineered human blood vessel. FASEB 1998, 12
(47–56). 49. Herring, M.B.; Dilley, R.; Jersild, R.A.;
Boxer, L.; Gardner, A.; Glover, J. Seeding arterial
prostheses with vascular endothelium. The nature of the
lining. Ann. Surg. 1979, 190 (1), 84–90. 50. Zilla, P.;
Fasol, R.; Deutsch, M.; Fischlein, T.; Minar, E.; Hammerle,
A.; Krupicka, O.; Kadletz, M. Endothelial cell seeding of
polytetrafluoroethylene vascular grafts in humans: A
preliminary report. J. Vasc. Surg. 1987, 6 (6), 535–541.
51. Jarrell, B.; Williams, S.; Stokes, G. Use of freshly
isolated capillary endothelial cells for the immediate
establishment of a monolayer on a vascular graft at
surgery. Surgery 1986, 100, 392–399. 52. Zilla, P.;
Deutsch, M.; Meinhart, J. Endothelial cell transplantation.
Semin. Vasc. Surg. 1999, 12 (1), 52–63. 53.
Gillis-Haegerstrand, C. Adhesion Molecule Expression
Following In Vitro Lining. In Tissue Engineering of
Prosthetic Vascular Grafts; Zilla, P., Greisler, H., Eds.;
RG Landes: Austin, 1999. 54. Zilla, P.; Deutsch, M.;
Meinhart, J.; Puschmann, R.; Eberl, T.; Minar, E.; Dudczak,
R.; Lugmaier, H.; V Schmidt, P.; Noszian, I.; Fischlein, T.
Clinical in vitro endothelialization of femoropopliteal
bypass grafts: An actuarial follow-up over three years. J.
Vasc. Surg. 1994, 19 (3), 540–548.

55. van der Lei, B.; Wildevuur, C. Microporous, Compliant,


Biodegradable Small-Calibre Vascular Grafts. In Current
Perspectives on Implantable Devices; Williams, G., Ed.; Jai
Press: London, 1990.

56. Gorna, K.; Gogolewski, S. Biodegradable polyurethanes


for implants. II. In vitro degradation and calcification of
materials from poly(epsilon-caprolactone)-poly(ethylene
oxide) diols and various chain extenders. J. Biomed. Mater.
Res. 2002, 60 (4), 592–606.

57. Bruin, P.; Smedinga, J.; Pennings, A.; Jonkman, M.


Biodegradable lysine diisocyanate-based
poly(glycolideco-epsilon-caprolactone)-urethane network in
artificial skin. Biomaterials 1990, 11 (4), 291–295.

58. Fromstein, J.D.; Woodhouse, K.A. Elastomeric


biodegradable polyurethane blends for soft tissue
applications. J. Biomater. Sci., Polym. Ed. 2002, 13 (4),
391–406. 59. Xue, L.; Griesler, H. Blood Vessels. In
Principles of Tissue Engineering; Lanza, R., Langer, R.,
Vacanti, J., Eds.; Academic Press: San Diego, 2000;
427–446. 60. Sullivan, S.; Brockbank, K.G.M. Small-Diameter
Vascular Grafts. In Principles of Tissue Engineering;
Lanza, R., Langer, R., Vacanti, J., Eds.; Academic Press:
San Diego, 2000; 447–454. 61. Van Wachem, P.; Plantinga,
J.; Wissink, M.; Beernink, R.; Poot, A.; Engbers, G.;
Beugeling, T.; van, A.W.; Feijen, J.; van, L.M. In vivo
biocompatibility of carbodiimide-crosslinked collagen
matrices: Effects of crosslink density, heparin
immobilization, and bFGF loading. J. Biomed. Mater. Res.
2001, 55 (3), 368–378. 62. Campbell, J.; Efendy, J.;
Campbell, G. Novel vascular graft grown within recipient’s
own peritoneal cavity. Circ. Res. 1999, 85 (12), 1173–1178.
63. Edelman, E. Vascular tissue engineering: Designer
arteries. Circ. Res. 1999, 85 (12), 1115–1117.
Vascular Grafts: Development Strategies

4. Groegler, F.M.; Kapfer, X.; Meichelbo¨ck, W. Crural


Prosthetic Revascularization: Randomized, Prospective,
Multicentric Comparison of Standard and Carbon Impregnated
ePTFE Grafts. In Montefiore Annual Symposium on Current
Critical Problems, New Horizons and Techniques in Vascular
and Endovascular Surgery (‘01); New York, NY, Nov 15–18,
2001. Abstract 01–2, I2.1–I2.3.

5. Lumsden, A.B.; Chen, C.; Coyle, K.A.; Ofenloch, J.C.;


Wang, J.H.; Yasuda, H.K.; Hanson, S.R. Nonporous silicone
polymer coating of expanded polytetrafluoroethylene grafts
reduces graft neointimal hyperplasia in dog and baboon
models. J. Vasc. Surg. 1996, 24, 825– 833.

6. Quarmby, J.W.; Burnand, K.G.; Lockhart, S.J.M.; Donald,


A.E.; Sommerville, K.M.; Jamieson, C.W.; Browse, N.L.
Prospective randomized trial of woven versus
collagen-impregnated knitted prosthetic Dacron grafts in
aortoiliac surgery. Br. J. Surg. 1998, 85, 775–777.

7. Phaneuf, M.D.; Quist, W.C.; Bide, M.J.; LoGerfo, F.W.


Modification of polyethylene terephthalate (Dacron) via
denier reduction, effects on material tensile strength,
weight, and protein binding capabilities. J. Appl.
Biomater. 1995, 6, 289–299.

8. Kang, S.S.; Petsikas, D.; Murchan, P.; Cziperle, D.J.;


Ren, D.; Kim, D.U.; Greisler, H.P. Effects of albumin
coating of knitted Dacron grafts on transinterstitial blood
loss and tissue ingrowth and incorporation. Cardiovasc.
Surg. 1997, 5, 184–189.

9. Bos, G.W.; Poot, A.A.; Beugeling, T.; van Aken, W.G.;


Feijen, J. Small-diameter vascular graft prostheses,
current status. Arch. Physiol. Biochem. 1998, 106, 100–115.

10. Eberhart, R.C.; Munro, M.S.; Williams, G.B.; Kulkarni,


P.V.; Shannon, W.A., Jr.; Brink, B.E.; Fry, W.J. Albumin
adsorption and retention on C18-alkyl-derivatized
polyurethane vascular grafts. Artif. Organs 1987, 11,
375–382.

11. Ishikawa, Y.; Sasakawa, S.; Takase, M.; Osada, Y.


Effect of albumin immobilization by plasma polymerization
on platelet reactivity. Thromb. Res. 1984, 35, 193–202.

12. Tsai, C.C.; Huo, H.H.; Kulkarni, P.V.; Eberhart, R.C.


Biocompatible coatings with high albumin affinity. Trans.
Am. Soc. Artif. Intern. Organs 1990, 36, M307–M310.

13. Rumisek, J.D.; Wade, C.E.; Brooks, D.E.; Okerberg,


C.V.; Barry, M.J.; Clarke, J.S. Heat-denatured
albumin-coated Dacron vascular grafts: Physical
characteristics and in vivo performance. J. Vasc. Surg.
1986, 4, 136–143.

14. Kottke-Marchant, K.; Anderson, J.M.; Umemura, Y.;


Marchant, R.E. Effect of albumin coating of the in vitro
blood compatibility of Dacron arterial prostheses.
Biomaterials 1989, 10, 147–155.

15. Phaneuf, M.D.; Dempsey, D.J.; Bide, M.J.; Szycher, M.;


Quist, W.C.; LoGerfo, F.W. Bioengineering of a novel small
diameter polyurethane vascular graft with covalently bound
recombinant hirudin. ASAIO J. 1998, 44, M653–M658. 16.
Dempsey, D.J.; Phaneuf, M.D.; Bide, M.J.; Szycher, M.;
Quist, W.C.; Logerfo, F.W. Synthesis of a novel small
diameter polyurethane vascular graft with reactive binding
sites. ASAIO J. 1998, 44, M506– M510. 17. Mohamed, M.S.;
Mukherjee, M.; Kakkar, V.V. Thrombogenicity of heparin and
non-heparin bound arterial prostheses, an in vitro
evaluation. J. R. Coll. Surg. Edinb. 1998, 43, 155–157. 18.
Park, K.D.; Okano, T.; Nojiri, C.; Kim, S.W. Heparin
immobilization onto segmented polyurethane-urea
surfaces—Effect of hydrophilic spacers. J. Biomed. Mater.
Res. 1988, 22, 977–992. 19. Nojiri, C.; Kido, T.; Sugiyama,
T.; Horiuchi, K.; Kijima, T.; Hagiwara, K.; Kuribayashi,
E.; Nogawa, A.; Ogiwara, K.; Akutsu, T. Can heparin
immobilized surfaces maintain nonthrombogenic activity
during in vivo long-term implantation? ASAIO J. 1996, 42,
M468–M475. 20. Greisler, H.P.; Klosak, J.J.; Dennis, J.W.;
Ellinger, J.; Kim, D.U.; Burgess, W.; Maciag, T.
Endothelial cell growth factor attachment to biomaterials.
ASAIO Trans. 1986, 32, 346–349. 21. Greisler, H.P.; Klosak,
J.J.; Dennis, J.W.; Karesh, S.M.; Ellinger, J.; Kim, D.U.
Biomaterial pretreatment with ECGF to augment endothelial
cell proliferation. J. Vasc. Surg. 1987, 5, 393–399. 22.
Greisler, H.P.; Gosselin, C.; Ren, D.; Kang, S.S.; Kim,
D.U. Biointeractive polymers and tissue engineered blood
vessels. Biomaterials 1996, 17, 329–336. 23. Greisler,
H.P.; Cziperle, D.J.; Kim, D.U.; Garfield, J.D.; Petsikas,
D.; Murchan, P.M.; Applegren, E.O.; Drohan, W.; Burgess,
W.H. Enhanced endothelialization of expanded
polytetrafluoroethylene grafts by fibroblast growth factor
type 1 pretreatment. Surgery 1992, 112, 244–254. 24.
Gosselin, C.; Ren, D.; Ellinger, J.; Greisler, H.P. In vivo
platelet deposition on polytetra-fluoroethylene coated with
fibrin glue containing fibroblast growth factor 1 and heparin
in a canine model. Am. J. Surg. 1995, 170, 126–130. 25.
Zarge, J.I.; Gosselin, C.; Huang, P.; Greisler, H.P.
Pletelet deposition on ePTFE Grafts coated with fibrin glue
with or without FGF-1 and heparin. J. Surg. Res. 1997, 67,
4–8. 26. Gray, J.L.; Kang, S.S.; Zenni, G.C.; Kim, D.U.;
Kim, P.I.; Burgess, W.H.; Drohan, W.; Winkles, J.A.;
Hauderschild, C.C.; Greisler, H.P. FGF-1 affixation
stimulates ePTFE endothelialization without intimal
hyperplasia. J. Surg. Res. 1994, 57, 596–612. 27. Doi, K.;
Matsuda, T. Enhanced vascularization in a microporous
polyurethane graft impregnated with basic fibroblast growth
factor and heparin. J. Biomed. Mater. Res. 1997, 34,
361–370. 28. Wesolowski, S.A.; Fries, C.C.; Domingo, R.T.;
Liebig, W.J.; Sawyer, P.N. The compound prosthetic vascular
graft. A pathologic survey. Surgery 1963, 53, 19–44. 29.
Ruderman, R.J.; Hegyeli, A.F.; Hattler, B.G.; Leonard, F. A
partially biodegradable vascular prosthesis. ASAIO Trans.
1972, 18, 30–37. 30. Bowald, S.; Busch, C.; Eriksson, I.
Arterial regeneration following polyglactin 910 suture mesh
grafting. Surgery 1972, 86, 722–729. 31. Bowald, S.; Busch,
C.; Eriksson, I. Absorbable material in vascular
prostheses: A new device. Acta Chir. Scand. 1980, 146,
391–395. 32. Greisler, H.P. Arterial regeneration over
absorbable prostheses. Arch. Surg. 1982, 117, 1425–1431.
33. Greisler, H.P.; Kim, D.U.; Price, J.B.; Voorhees, A.B.
Arterial regenerative activity after prosthetic
implantation. Arch. Surg. 1985, 120, 315–323. 34. Greisler,
H.P.; Ellinger, J.; Schwarcz, T.H.; Golan, J.; Raymond,
R.M.; Kim, D.U. Arterial regeneration over polydioxanone
prostheses in the rabbit. Arch. Surg. 1987, 122, 715–721.
35. Greisler, H.P.; Schwarcz, T.H.; Ellinger, J.; Kim, D.U.
Dacron inhibition of arterial regenerative activity. J.
Vasc. Surg. 1986, 3, 747–756. 36. Greisler, H.P.; Kim,
D.U.; Dennis, J.W.; Klosak, J.J.; Widerborg, K.A.; Endean,
E.D.; Raymond, R.M.; Ellinger, J. Compound polyglactin
910/polypropylene small vessel prostheses. J. Vasc. Surg.
1987, 5, 572–583. 37. Greisler, H.P.; Endean, E.D.; Klosak,
J.J.; Ellinger, J.; Dennis, J.W.; Buttle, K.; Kim, D.U.
Polyglactin 910/ polydioxanone bicomponent totally
resorbable vascular prostheses. J. Vasc. Surg. 1988, 7,
697–705. 38. Galletti, P.M.; Aebischer, P.; Sasken, H.F.;
Goddard, M.B.; Chiu, T.H. Experience with fully
bioresorbable aortic grafts in the dog. Surgery 1988, 103,
231–241. 39. Yue, X.; van der Lei, B.; Schakenraad, J.M.;
van Oene, G.H.; Kuit, J.H.; Feijen, J.; Wildevuur, C.R.
Smooth muscle cell seeding in biodegradable grafts in rats:
A new method to enhance the process of arterial wall
regeneration. Surgery 1988, 103, 206–212. 40. van der Lei,
B.; Nieuwenhuis, P.; Molenaar, I.; Wildevuur, C.R.
Long-term biologic fate of neoarteries regenerated in
microporous, compliant, biodegradable, small-caliber
vascular grafts in rats. Surgery 1987, 101, 459–467. 41.
Herring, M.; Gardner, A.; Glover, J. A single-staged
technique for seedeing vascular grafts with autogenous
endothelium. Surgery 1978, 84, 498–504. 42. Graham, L.M.;
Burkel, W.E.; Ford, J.W.; Vinter, D.W.; Kahn, R.H.;
Stanley, J.C. Expanded polytetrafluoroethylene vascular
prostheses seeded with enzymatically derived and cultured
canine endothelial cells. Surgery 1982, 91, 550–559. 43.
Jarrell, B.E.; Williams, S.K. Microvessel derived
endothelial cell isolation, adherence, and monolayer
formation for vascular grafts. J. Vasc. Surg. 1991, 13,
733–734. 44. Sipehia, R.; Martucci, G.; Lipscombe, J.
Transplantation of human endothelial cell monolayer on
artificial vascular prosthesis: The effect of growth-support
surface chemistry, cell seeding density, ECM protein
coating, and growth factors. Artif. Cells Blood Substit.
Immobil. Biotechnol. 1996, 24, 51–63. 45. Shindo, S.;
Takagi, A.; Whittemore, A.D. Improved patency of
collagen-impregnated grafts after in vitro autogenous
endothelial cell seeding. J. Vasc. Surg. 1987, 6, 325–332.
46. Prendiville, E.J.; Coleman, J.E.; Callow, A.D.; Gould,
K.E.; Laliberte-Verdon, S.; Ramberg, K.; Connolly, R.J.
Increased in vitro incubation time of endothelial cells on
fibronectin-treated ePTFE increases cell retention in blood
flow. Eur. J. Vasc. Surg. 1991, 5, 311–319. 47. Sentissi,
J.M.; Rambnerg, K.; O’Donnell, T.F., Jr.; Connolly, R.J.;
Callow, A.D. The effect of flow on vascular endothelial
cells grown in tissue culture on polytetrafluoroethylene
grafts. Surgery 1986, 99, 337–343. 48. Pasic, M.;
Muller-Glauser, W.; von Segesser, L.K.; Lachat, M.;
Mihaljevic, T.; Turina, M. Superior late patency of
small-diameter Dacron grafts seeded with omental
microvascular cells, an experimental study. Ann. Thorac.
Surg. 1994, 58, 677–683. 49. Tseng, D.Y.; Edelman, E.R.
Effects of amide and amine plasma-treated ePTFE vascular
grafts on endothelial cell lining in an artificial
circulatory system. J. Biomed. Mater. Res. 1998, 42,
188–198. 50. Schneider, P.A.; Hanson, S.R.; Price, T.M.;
Harker, L.A. Preformed confluent endothelial cell monolayers
prevent early platelet deposition on vascular prostheses in
baboons. J. Vasc. Surg. 1988, 8, 229–235. 51. Rosenman,
J.E.; Kempczinski, R.F.; Pearce, W.H.; Silberstein, E.B.
Kinetics of endothelial cell seeding. J. Vasc. Surg. 1985,
2, 778–784. 52. Vohra, R.; Thomson, G.J.; Carr, H.M.;
Sharma, H.; Welch, M.; Walker, M.G. In vitro adherence and
kinetics studies of adult human endothelial cell seeded
polytetrefluoroethylene and gelatin impregnated Dacron
grafts. Eur. J. Vasc. Surg. 1991, 5, 93–103. 53. Miyata,
T.; Conte, M.S.; Trudell, L.A.; Mason, D.; Wittemore, A.D.;
Brinyi, L.K. Delayed exposure to pulsatile shear stress
improves retention of human saphenous vein endothelial
cells on seeded ePTFE grafts. J. Surg. Res. 1991, 50,
485–493. 54. Sugawara, Y.; Miyata, T.; Sato, O.; Kimura,
H.; Namba, T.; Makuuchi, M. Rapid post-incubation
endothelial retention by Dacron grafts. J. Surg. Res. 1997,
67, 132–136. 55. Prendiville, E.J.; Colemam, J.E.; Callow,
A.D.; Gould, K.E.; Laliberte-Verdon, S.; Ramberg, K.;
Connolly, R.J. Increased in vitro incubation time of
endothelial cells on fibronectin-treated ePTFE incteases
cell retention in blood flow. Eur. J. Vasc. Surg. 1991, 5,
311–319. 56. Ott, M.J.; Ballermann, B.J. Shear
stress-conditioned, endothelial cell-seeded vascular
grafts, improved cell adherence in response to in vitro
shear stress. Surgery 1995, 117, 334–339. 57. Vohra, R.K.;
Thomson, G.J.; Sharma, H.; Carr, H.M.; Walker, M.G. Effects
of shear stress on endothelial cell monolayer on expanded
polytetrefluoroethylene grafts using preclot and fibronectin
matrices. Eur. J. Vasc. Surg. 1990, 4, 33–41. 58.
Giudiceandrea, A.; Seifalian, A.M.; Krijgsman, B.;
Hamilton, G. Effect of prolonged pulsatile shear stress in
vitro on endothelial cell seeded PTFE and compliant
polyurethane vascular grafts. J. Vasc. Endovasc. Surg.
1998, 15, 147–154. V

59. Bowlin, G.L.; Rittgers, S.E.; Milsted, A.; Schmidt,


S.P. In vitro evaluation of electrostatic endothelial cell
transplantation onto 4mm interior diameter expanded
polytetrafluoroethylene grafts. J. Vasc. Surg. 1998, 27,
504–511.

60. Zilla, P.; Preiss, P.; Groscurth, P.; R_semeier, F.;


Deutsch, M.; Odell, J.; Heidinger, C.; Fasol, R.; von
Oppell, U. In vitro-lined endothelium, initial integrity
and ultrastructural events. Surgery 1994, 116, 524–534.

61. Whitehouse, W.M., Jr.; Wakefield, T.W.; Vinter, D.W.;


Ford, J.W.; Swanson, D.P.; Thrall, J.H.; Froelich, J.W.;
Brown, L.E.; Burkel, W.E.; Graham, L.M. Indium-111 oxide
labeled platelet imaging of endothelial seeded Dacron
thoracoabdominal vascular prostheses in a canine
experimental model. ASAIO Trans. 1983, 29, 183–187.

62. Wakefield, T.W.; Lindblad, B.; Graham, L.M.; Whitehouse,


W.M., Jr.; Ripley, S.D.; Petry, N.A.; Spaulding, S.A.;
Burkel, W.E.; Stanley, J.C. Nuclide imaging of vascular
graft-platelet interactions, comparison of indium excess
and technetium subtraction techniques. J. Surg. Res. 1986,
40, 388–394.
63. Stanley, J.C.; Burkel, W.E.; Ford, J.W.; Vinter, D.W.;
Kahn, R.H.; Whitehouse, W.M., Jr.; Graham, L.M. Enhanced
patency of small-diameter, externally supported Dacron
iliofemoral grafts seeded with endothelial cells. Surgery
1982, 92, 994–1005.

64. Allen, B.T.; Long, J.A.; Clark, R.E.; Sicard, G.A.;


Clark, R.D.; Hopkins, K.T.; Welch, M.J. Influence of
endothelial cell seeding on platelet deposition and patency
in small-diameter Dacron arterial grafts. J. Vasc. Surg.
1984, 1, 224–233.

65. Graham, L.M.; Stanley, J.C.; Burkel, W.E. Improved


patency of endothelial-cell-seeded, long, knitted Dacron
and ePTFE vascular prostheses. ASAIO J. 1985, 8, 65–73.

66. Rosenman, J.E.; Kempczinski, R.F.; Berlatzky, Y.;


Pearce, W.H.; Ramalanjaona, G.R.; Bjornson, H.S. Bacterial
adherence to endothelial-seeded polytetrafluoroethylene
grafts. Surgery 1985, 98, 816–823.

67. Graham, L.M.; Fox, P.L. Growth factor production


following prosthetic graft implantation. J. Vasc. Surg.
1991, 13, 742–746.

68. Sapienza, P.; di Marzo, L.; Cucina, A.; Corvino, V.;


Mingoli, A.; Giustiniani, Q.; Ziparo, E.; Cavallaro, A.
Release of PDGF-BB and bFGF by human endothelial cells
seeded on expanded polytetrafluoroethylene vascular grafts.
J. Surg. Res. 1998, 75, 24–29.

69. Magometschnigg, H.; Kadletz, M.; Vodrazka, M.; Dock,


W.; Grimm, M.; Grabenwoger, M.; Minar, E.; Staudacher, M.;
Fenzl, G.; Wolner, E. Prospective clinical study with in
vitro endothelial cell lining of expanded
polytetrafluoroethylene grafts in crural repeat
reconstruction. J. Vasc. Surg. 1992, 15, 527–535.

70. Herring, M.B.; Gardner, A.; Glover, J. Seeding human


arterial prostheses with mechanically derived endothelium.
The detrimental effect of smoking. J. Vasc. Surg. 1984, 1,
279–289.

71. Jensen, N.; Lindblad, B.; Bergovist, D. Endothelial


cell seeded Dacron aortobifurcated grafts, Platelet
deposition and long-term follow-up. J. Cardiovasc. Surg.
1994, 35 , 425–429. 72. Zilla, P.; Deutsch, M.; Meinhart,
J.; Puschmann, R.; Eberl, T.; Minar, E.; Dudczak, R.;
Lugmaier, H.; Schmidt, P.; Noszian, I. Clinical in vitro
endothelialization of femoropopliteal bypass grafts, an
actuarial follow-up over three years. J. Vasc. Surg. 1994,
19, 540–548. 73. Deutsch, M.; Meinhart, J.; Fischlein, T.;
Preiss, P.; Zilla, P. Clinical autologous in vitro
endothelialization of infrainguinal ePTFE grafts in 100
patients: a 9-year experience. Surgery 1999, 126, 847–855.
74. Deutsh, M.; Meinhart, J.; Vesely, M.; Fischlein, T.;
Groscurth, P.; von Oppell, U.; Zella, P. In vitro
endothelialization of expanded polytetrafluoroethylene
grafts, a clinical case report after 41months of
implantation. J. Vasc. Surg. 1997, 25, 757–763. 75. Park,
P.K.; Jarrell, B.E.; Williams, S.K.; Carter, T.L.; Rose,
D.G.; Martinez-Hernandez, A.; Carabasi, R.A., III. Thrombus
free, human endothelial surface in midregion of a Dacron
vascular graft in the splanchnic venous
circuit—Observations after nine months of implantation. J.
Vasc. Surg. 1990, 11, 468–475. 76. Wilson, J.M.; Birinyi,
L.K.; Salomon, R.N.; Libby, P.; Callow, A.D.; Mulligan,
R.C. Implantation of vascular grafts lined with genetically
modified endothelial cells. Science 1989, 244, 1344–1346.
77. Dichek, D.A.; Nussbaum, O.; Degen, S.J.; Anderson, W.F.
Enhancement of the fibrinolytic activity of sheep
endothelial cells by retroviral-mediated gene transfer.
Blood 1991, 77, 533–541. 78. Shayani, V.; Newman, K.D.;
Dichek, D.A. Optimization of recombinant t-PA secretion
from seeded vascular grafts. J. Surg. Res. 1994, 57,
495–504. 79. Brothers, T.E.; Judge, L.M.; Wilson, J.M.;
Burkel, W.E.; Stanley, J.C. Effect of genetic transduction
on in vitro canine endothelial cell prostanoid production
and growth. Surg. Forum 1990, 41, 337–339. 80. Jaklitsh,
M.T.; Biro, S.; Casscells, W.; Dichek, D.A. Transduced
endothelial cells expressing high levels of tissue
plasminogen activator have an unaltered phenotype in vitro.
J. Cell. Physiol. 1993, 154, 207–216. 81. Huber, T.S.;
Welling, T.H.; Sarkar, R.; Messina, L.M.; Stanley, J.C.
Effects of retroviral-mediated tissue plasminogen activator
gene transfer and expression on adherence and proliferation
of canine endothelial cells seeded onto expanded
polytetrafluoroethylene. J. Vasc. Surg. 1995, 22, 795–803.
82. Podrazik, R.M.; Whitehill, T.A.; Komorowski, T.A.;
Karo, K.H.; Messina, L.M.; Stanley, J.C. In vivo fate of
lacZ-transduced endothelial cells seeded ePTFE
thoracoabdominal vascular prostheses in dogs. Surg. Forum
1993, 44, 334–337. 83. Baer, R.P.; Whitehill, T.E.; Sarkar,
R.; Sarkar, M.; Messina, L.M.; Komorowski, T.A.; Stanley,
J.C. Retroviral-mediated transduction of endothelial cells
with the lacZ gene impairs cellular proliferation in vitro
and graft endothelialization in vivo. J. Vasc. Surg. 1996,
24, 892–899. 84. Dunn, P.F.; Newmann, K.D.; Jones, M.;
Yamada, I.; Shayani, V.; Virmani, R.; Dichek, D.A. Seeding
of vascular grafts with genetically modified endothelial
cells, secretion of recombinant tPA resuts in decreased
seeded cell retention in vitro and in vivo. Circulation
1996, 93, 1439–1446. 85. Falk, J.; Townsend, L.E.; Vogel,
L.M.; Boyer, M.; Olt, S.; Wease, G.L.; Trevor, K.T.;
Seymour, M.; Glover, J.L.; Bendick, P.J. Improved adhere of
genetically modified endothelial cells to small-diameter
expanded polytetrafluoroethylene grafts in a canine model.
J. Vasc. Surg. 1998, 27, 902–909. 86. Noishiki, Y.; Yamane,
Y.; Okoshi, T.; Tomizawa, Y.; Satoh, S. Choice, isolation,
and preparation of cells for bioartificial vascular grafts.
Artif. Organs 1998, 22, 50–62. 87. Yue, X.; Van der Lei,
B.; Schakenraad, J.M.; Van Oene, G.H.; Kuit, J.H.; Feijen,
J.; Wildevuur, C.R. Smooth muscle cell seeding in
biodegradable grafts in rats, a new method to enhance the
process of artery wall regeneration. Surgery 1998, 103,
206–212. 88. Shinoka, T.; Shum-Tim, D.; Ma, P.X.; Tanel,
R.E.; Isogai, N.; Langer, R.; Vacanti, J.P.; Mayer, J.E.
Creation of viable pulmonary artery autografts through
tissue engineering. J. Thorac. Cardiovasc. Surg. 1998, 115,
536–546. 89. Weinberg, C.B.; Bell, E. A blood vessel model
constructed from collagen and cultured vascular cells.
Science 1986, 231, 397–400. 90. L’Heurerx, N.; Germain, L.;
LabbJ, T.; Auger, F.A. In vitro construction of a human
blood vessel from cultured vascular cells. A morphologic
study. J. Vasc. Surg. 1993, 17, 499–509. 91. L’Heurerx, N.;
PWquet, S.; LabbJ, T.; Germain, L.; Auger, F.A. A
completely biological tissue-engineered human blood vessel.
FASEB J. 1998, 12, 47–56. 92. Ziegler, T.; Nerem, R.M.
Tissue engineering a blood vessel, regulation of vascular
biology by mechanical stresses. J. Cell. Biochem. 1994, 56,
204–209. 93. Langer, R.; Vacanti, J.P. Tissue engineering.
Science 1993, 260, 920–926. 94. Fuchs, J.R.; Nasseri, B.A.;
Vicanti, J.P. Tissue engineering: A 21st century solution
to surgical reconstruction. Ann. Thorac. Surg. 2001, 72,
577–591. 95. Moldovan, N.I.; Ferrari, M. Prospects for
microtechnology and nanotechnology in bioengineering of
replacement microvessels. Arch. Pathol. Lab. Med. 2002,
126, 320–324. 96. Griffith, L.G.; Naughton, G. Tissue
engineering— Current challenges and expanding
opportunities. Science 2002, 295, 1009–1016. V
Vascular Grafts: Host Interactions

1. Voorhees, A.B., Jr.; Jaretzki, A.; Blakemore, A.H. The


use of tubes constructed of Vinyon N cloth in bridging
arterial defects. Ann. Surg. 1952, 135, 332–336.

2. Conte, M.S. The ideal small arterial substitute, a


search for the Holy Grail? FASEB J. 1998, 12, 43–45.

3. Vroman, L. Methods of investigating protein interaction


on artificial and natural surfaces. Ann. N.Y. Acad. Sci.
1987, 516, 300–305.

4. De La Cruz, C.; Haimovich, B.; Greco, R.S. Immobilized


IgG and fibrinogen differentially affect the cytoskeletal
organization and bactericidal function of adherent
neutrophils. J. Surg. Res. 1998, 80, 28–34.

5. Shepard, A.D.; Gelfand, J.A.; Callow, A.D.; O’Donnell,


T.F., Jr. Complement activation by synthetic vascular
prostheses. J. Vasc. Surg. 1984, 1, 829–838.

6. Toursarkissian, B.; Eisenberg, P.R.; Abendschein, D.R.;


Rubin, B.G. Thrombogenicity of small-diameter prosthetic
grafts. Relative contributions of graft-associated thrombin
and factor Xa. J. Vasc. Surg. 1997, 25, 730–735.

7. Ito, R.K.; Rosenblatt, M.S.; Contreras, M.A.; Beophy,


X.M.; LoGerfo, F.W. Monitoring platelet interactions with
prosthetic graft implants in a canine model. ASAIO Trans.
1990, 36, M175–M178.

8. McCollum, C.N.; Kester, R.C.; Rajah, S.M.; Learoyd, P.;


Pepper, M. Arterial graft maturation, the duration of
thrombotic activity in Dacron aortobifemoral grafts
measured by platelet and fibrinogen kinetics. Br. J. Surg.
1981, 68, 61–64.

9. Stratton, J.R.; Thiele, B.L.; Ritchie, J.L. Platelet


deposition on Dacron aortic bifurcation grafts in man,
quantitation with indium-111 platelet imaging. Circulation
1982, 66, 1287–1293.

10. Stratton, J.R.; Thiele, B.L.; Ritchie, J.L. Natural


history of platelet deposition on Dacron aortic bifurcation
grafts in the first year after implantation. Am. J. Cardiol.
1938, 52, 371–374.

11. Mazur, C.; Tschopp, J.F.; Faliakou, E.C.; Gould, K.E.;


Diehl, J.T.; Pierschbacher, M.D.; Connolly, R.J. Selective
aIIbb3 receptor blockage with peptide TP9201 prevents
platelet uptake on Dacron vascular grafts without
significant effect on bleeding time. J. Lab. Clin. Med.
1994, 124, 589–599.

12. Kelly, A.B.; Maragamore, J.M.; Bourdon, P.; Hanson,


S.R.; Harker, L.A. Antithrombotic effects of synthetic
peptides targeting various functional domains of thrombin.
Proc. Natl. Acad. Sci. U.S.A. 1992, 89, 6040–6044.

13. Bevilacqua, M.P.; Pober, J.S.; Wheeler, M.E.; Cotran,


R.S.; Gimbrone, M.A., Jr. Interleukin-1 acts on cultured
human vascular endothelium to increase the adhesion of
polymorphonuclear leukocytes, monocytes, and related
leukocyte cell lines. J. Clin. Invest. 1985, 76, 2003–2011.

14. Gamble, J.R.; Harlan, J.M.; Klebanoff, S.J.; Vadas,


M.A. Stimulation of the adherence of neutrophils to
umbilical vein endothelium by human recombinant tumor
necrosis factor. Proc. Natl. Acad. Sci. U.S.A. 1985, 82,
8667–8671. 15. Ford-Hutchinson, A.W.; Bray, M.A.; Doig,
M.V.; Shipley, M.E.; Smith, M.J. Leukotriene B, a potent
chemokinetic and aggregating substance released from
polymorphonuclear leukocytes. Nature 1980, 286, 264–265.
16. Deuel, T.F.; Senior, R.M.; Chang, D.; Griffin, G.L.;
Heinrikson, R.L.; Kaiser, E.T. Platelet factor 4 is
chemotactic for neutrophils and monocytes. Proc. Natl.
Acad. Sci. U.S.A. 1981, 78, 4584–4587. 17. Greisler, H.P.;
Ellinger, J.; Henderson, S.C.; Shaheen, A.M.; Burgess,
W.H.; Lam, T.M. The effects of an atherogenic diet on
macrophage/biomaterial interations. J. Vasc. Surg. 1991,
14, 10–23. 18. Greisler, H.P.; Petsikas, D.; Cziperle,
T.M.; Murchan, P.; Henderson, S.C.; Lam, D.J. Dacron
stimulation of macrophage transforming growth factor-beta
release. Cardiovasc. Surg. 1996, 4, 169–173. 19. Swartbol,
P.; Truedsson, L.; Pa¨rsson, H.; Norgren, L. Tumor necrosis
factor-a and interleukin-6 release from white blood cells
induced by different graft materials in vitro are affected
by pentoxifylline and iloprost. J. Biomed. Mater. Res.
1997, 36, 400–406. 20. Mattana, J.; Effiong, C.; Kapasi, A.;
Singhal, P.C. Leukocyte-polytetrafluroethylene interaction
enhances proliferation of vascular smooth muscle cells via
tumor necrosis factor-alpha secretion. Kidney Int. 1997,
52, 1478–1485. 21. Glazebrook, H.; Hatch, T.; Brindle, N.P.
Regulation of insulin-like growth factor-1 expression in
vascular endothelial cells by the inflammatory cytokines
interleukin-1. J. Vasc. Res. 1998, 35, 143–149. 22. Totani,
L.; Cumashi, A.; Piccoli, A.; Lorenzet, R.
Polymorphonuclear leukocytes induce PDGF release from
IL-1beta-treated endothelial cells, role of adhesion
molecules and serine proteases. Arterioscler. Thromb. Vasc.
Biol. 1998, 18, 1534–1540. 23. Clowes, A.W.; Kirkman, T.R.;
Reidy, M.A. Mechanisms of arterial graft healing: Rapid
transmural capillary ingrowth provides a source of intimal
endothelium and smooth muscle in porous PTFE prostheses.
Am. J. Pathol. 1986, 123, 220–230. 24. Greisler, H.P.;
Dennis, J.W.; Endean, E.D.; Ellinger, J.; Burrle, K.F.;
Kim, D.U. Derivation of neointima in vascular grafts.
Circulation 1988, 78, I6–I12. 25. Golden, M.A.; Hanson,
S.R.; Kirkman, T.R.; Schnerder, P.A.; Clowes, A.W. Healing
of polytetrafluoroethylene arterial grafts is influenced by
graft porosity. J. Vasc. Surg. 1990, 11, 838–844. 26.
Onuki, Y.; Kouchi, Y.; Yoshida, H.; Wu, M.H.D.; Shi, Q.;
Wechezak, A.R.; Coan, D.; Sauvage, L.R. Early flow surface
endothelialization before microvessel ingrowth in
accelerated graft healing, with BrdU identification of
cellular proliferation. Ann. Vasc. Surg. 1998 , 12,
207–215. 27. Shi, Q.; Wu, M.H.; Hayashida, N.; Wechezak,
A.R.; Clowes, A.W.; Sauvage, L.R. Proof of fallout
endothelialization of impervious Dacron grafts in the aorta
and inferior vena cava of the dog. J. Vasc. Surg. 1994, 20,
546–556. 28. Kouchi, Y.; Onuki, Y.; Wu, M.H.; Shi, Q.;
Ghali, R.; Wechezak, A.R.; Kaplan, S.; Walker, M.; Sauvage,
L.R. Apparent blood stream origin of endothelial and smooth
muscle cells in the neointima of long, impervious
carotid-femoral grafts in the dog. Ann. Vasc. Surg. 1998,
12, 46–54. 29. Wu, M.H.; Shi, Q.; Wechezak, A.R.; Clowes,
A.W.; Gordon, I.L.; Sauvage, L.R. Definitive proof of
endothelialization of a Dacron arterial prosthesis in a
human being. J. Vasc. Surg. 1995, 21, 862–867. 30.
Greisler, H.P. Regulation of Vascular Graft Healing by
Induction of Tissue Incorporation. In Human Biomaterials
Applications, 1st Ed.; Wise, D.L., Altobelli, D.E.,
Yaszemski, M.J., Greisler, H.P., Eds.; Humana Press, Inc.:
Totowa, NJ, 1996; 227. 31. Hamdan, A.D.; Misare, B.;
Contreras, M.; Logerfo, F.W.; Quist, W.C. Evaluation of
anastomotic hyperplasia progression using the cyclin
specific antibody MIB-1. Am. J. Surg. 1996, 172, 168–171.
32. Pitsch, R.J.; Minion, D.J.; Goman, M.L.; van Aalst,
J.A.; Fox, P.L.; Graham, L.M. Platelet-derived growth
factor production by cells from Dacron grafts implanted in
a canine model. J. Vasc. Surg. 1997, 26, 70–78. 33.
Cameron, B.L.; Tsuchida, H.; Connall, T.P.; Nagae, T.;
Furukawa, K.; Wilson, S.E. High porosity PTFE improves
endothelialization of arterial grafts without increasing
early thrombogenicity. J. Cardiovasc. Surg. 1993, 34,
281–285. 34. Clowes, A.W.; Kohler, T. Graft
endothelialization: The role of angiogenic mechanisms. J.
Vasc. Surg. 1991, 13, 734–736. 35. Greisler, H.P.; Kim,
D.U.; Price, J.B.; Voorhees, A.B., Jr. Arterial
regenerative activity after prosthetic implantation. Arch.
Surg. 1985, 120, 315–323. 36. Greisler, H.P.; Ellinger, J.;
Schwarcz, T.H.; Golan, J.; Raymond, R.M.; Kim, D.U.
Arterial regeneration over a polydioxanone prostheses in
the rabbit. Arch. Surg. 1987, 122, 715–721. 37. Abbott,
W.M.; Cambria, R.P. Control of Physical Characteristics,
Elasticity and Compliance of Vascular Grafts. In Biological
and Synthetic Vascular Prostheses, 1st Ed.; Stanley, J.C.,
Ed.; Grune and Stratton: New York, 1982; 189. 38. Stansby,
G.; Berwanger, C.; Shukla, N.; Schmitz-Rixen, T.; Hamilton,
G. Endothelial seeding of compliant polyurethan vascular
graft material. Br. J. Surg. 1994, 81, 1286–1289. 39.
Stump, D.M.; Hart, V.G.; Tuttle, S.L. Circumferential
flexible vascular grafts. J. Biomech. 1998, 31, 705–710. 40.
Benedetti-Valentini, F.; Gossetti, B.; Irace, I.;
Martinelli, O.; Gattuso, R. Composite grafts for critical
ischaemia. Cardiovasc. Surg. 1994, 4, 372–379. 41. Pappas,
P.J.; Hobson, R.W., II; Meyers, M.G.; Jamil, Z.; Lee, B.C.;
Silva, M.B., Jr.; Goldberg, M.C.; Padberg, F.T., Jr.
Patency of infra-inguinal polytetrafluoroethylene bypass
grafts with distal interposition vein cuffs. Cardiovasc.
Surg. 1998, 6, 19–25. 42. Neville, R.F.; Attinger, C.;
Sidawy, A.N. Prosthetic bypass with a distal vein patch for
limb salvage. Am. J. Surg. 1997, 174, 173–178. 43. Gentile,
A.T.; Mills, J.L.; Gooden, M.A.; Hagerty, R.D.; Berman,
S.S.; Hughes, J.D.; Kleinhert, L.B.; Williams, S.K. Vein
patching reduces neointimal thickening associated with
prosthetic graft implanation. Am. J. Surg. 1998, 176,
601–607. 44. Hasson, J.E.; Megerman, J.; Abbott, W.M.
Increased compliance near vascular anastomoses. J. Vasc.
Surg. 1985, 2, 419–423. 45. Moore, W.S.; Cafferata, H.T.;
Hall, A.D.; Blaisdell, F.W. In defense of grafts across the
inguinal ligaments: An evaluation of early and late results
of aorto-femoral bypass grafts. Ann. Surg. 1968, 168,
207–214. 46. Lind, R.E.; Wright, C.B.; Lynch, T.G.;
Lamberth, W.C., Jr.; Slaymaker, E.E.; Brandt, B., III.
Aortofemoral bypass grafting: Microvel. Am. Surg. 1982, 48,
89–92. 47. Veith, F.J.; Gupta, S.K.; Ascher, E.;
White-Flores, S.; Samson, R.H.; Scher, L.A.; Towne, J.B.;
Bernhard, V.M.; Bonier, P.; Flinn, W.R. Six-year
prospective multicenter randomized comparison of autologous
saphenous vein and expanded polytetraflouroethelene grafts
in infra-inguinal arterial reconstructions. J. Vasc. Surg.
1986, 3, 104–114. 48. Leikweg, W.G.; Greenfield, L.J.
Vascular prostheses graft infections: Collected experiences
and results of treatment. Surgery 1977, 81, 335–342. 49.
Szilagyi, D.E.; Smith, R.F.; Elliot, J.P.; Vrandecic, M.P.
Infection in arterial reconstruction with synthetic
vascular graft. Ann. Surg. 1972, 176, 321–333. 50.
Goldstone, J.; Moore, W.S. Infections in vascular
prostheses. Am. J. Surg. 1974, 128, 225–233. 51.
Goeau-Brissonniere, O.; Mercier, F.; Nicolas, M.H.;
Bacourt, F.; Coggia, M.; Lebrault, C.; Pechere, J.C.
Treatment of vascular graft infection by in situ
replacement with a rifampin-bonded gelatin sealed Dacron
graft. J. Vasc. Surg. 1994, 19, 739–741. 52. Vicaretti, M.;
Hawthorne, W.J.; Ao, P.Y.; Fletcher, J.P. An increased
concentration of rifampicin bonded to gelatin-sealed Dacron
reduces the incidence of subsequent graft infections
following a staphylococcal challenge. Cardiovasc. Surg.
1998, 6, 268–273. V
Vascular Imaging

1. Cochran, S.T.; Bomyea, K.; Sayre, J.W. Trends in adverse


events after IV administration of contrast media. AJR Am.
J. Roentgenol. 2001, 176, 1385–1388.

2. Mistretta, C.A. Relative characteristics of MR


angiography and competing vascular imaging modalities. J.
Magn. Reson. Imaging 1993, 3, 685–698.

3. Valji, K. Standard Angiographic and Interventional


Techniques. In Vascular and Interventional Radiology, 1st
Ed.; W.B. Saunders Company: Philadelphia, 1999; 12–38.

4. Nissen, S.E.; Yock, P. Intravascular ultrasound: Novel


pathophysiological insights and current clinical
applications. Circulation 2001, 103, 604–616.

5. Waller, B.F. ‘‘Crackers, breakers, stretchers, drillers,


scrapers, shavers, burners, welders and melters’’—The
future treatment of atherosclerotic coronary artery
disease? A clinical-morphologic assessment. J. Am. Coll.
Cardiol. 1989, 13, 969–987.

6. Barnes, J.E. Characteristics and control of contrast in


CT. Radiographics 1992, 12, 825–837.

7. Fuchs, T.; Kachelriess, M.; Kalender, W.A. Technical


advances in multi-slice spiral CT. Eur. J. Radiol. 2000,
36, 69–73.

8. Gareth, W.L.; Phillips, M.R.C.P. Review of arterial


ultrasound. World J. Surg. 2000, 24, 232–240.

9. Stewart, J.H.; Grubb, M. Understanding vascular


ultrasonography. Mayo Clin. Proc. 1992, 67, 1186– 1196.

10. Nederkoorn, P.J.; van der, G.Y.; Hunink, M.G. Duplex


ultrasound and magnetic resonance angiography compared with
digital subtraction angiography in carotid artery stenosis:
A systematic review. Stroke 2003, 34, 1324–1332.

11. Dogan, A.; Dempsey, R.J. Diagnostic modalities for


carotid artery disease. Neurosurg. Clin. N. Am. 2000, 11,
205–220. 12. Finn, J.P.; Edelman, R.R. Black-blood and
segmented k-space magnetic resonance angiography. Magn.
Reson. Imaging Clin. N. Am. 1993, 1, 349–357. 13. Owen,
R.S.; Carpenter, J.P.; Baum, R.A.; Perloff, L.J.; Cope, C.
Magnetic resonance imaging of angiographically occult
runoff vessels in peripheral arterial occlusive disease. N.
Engl. J. Med. 1992, 326, 1577–1581. 14. Prince, M.R.;
Yucel, E.K.; Kaufman, J.A.; Harrison, D.C.; Geller, S.C.
Dynamic gadolinium-enhanced three-dimensional abdominal MR
arteriography. J. Magn. Reson. Imaging 1993, 3, 877–881.
15. Willig, D.S.; Turski, P.A.; Frayne, R.; Graves, V.B.;
Korosec, F.R.; Swan, J.S.; Mistretta, C.A.; Grist, T.M.
Contrast-enhanced 3D MR DSA of the carotid artery
bifurcation: Preliminary study of comparison with
unenhanced 2D and 3D time-of-flight MR angiography.
Radiology 1998, 208, 447–451. 16. Goyen, M.; Ruehm, S.G.;
Debatin, J.F. MR-angiography: The role of contrast agents.
Eur. J. Radiol. 2000, 34, 247–256. 17. Yuan, C.; Mitsumori,
L.M.; Beach, K.W.; Maravilla, K.R. Carotid atherosclerotic
plaque: Noninvasive MR characterization and identification
of vulnerable lesions. Radiology 2001, 221, 285–299. 18.
Stuber, M.; Botnar, R.M.; Kissinger, K.V.; Manning, W.J.
Free-breathing black-blood coronary MR angiography: Initial
results. Radiology 2001, 219, 278–283. 19. Huber, A.;
Matzko, M.; Wintersperger, B.J.; Reiser, M. [Reconstruction
methods in postprocessing of CTand MR-angiography of the
aorta]. Radiologe 2001, 41, 689–694. 20. Katzen, B.T. The
future of catheter-based angiography: Implications for the
vascular interventionalist. Radiol. Clin. North Am. 2002,
40, 689–692. 21. Cademartiri, F.; van der, L.A.;
Luccichenti, G.; Pavone, P.; Krestin, G.P. Parameters
affecting bolus geometry in CTA: A review. J. Comput.
Assist. Tomogr. 2002, 26, 598–607. 22. Flohr, T.; Kuttner,
A.; Bruder, H.; Stierstorfer, K.; Halliburton, S.S.;
Schaller, S.; Ohnesorge, B.M. Performance evaluation of a
multi-slice CT system with 16-slice detector and increased
gantry rotation speed for isotropic submillimeter imaging
of the heart. Herz 2003, 28, 7–19. 23. Murphy, K.J.; Rubin,
J.M. Power Doppler: It’s a good thing. Semin. Ultrasound CT
MR 1997, 18, 13–21. 24. Melany, M.L.; Grant, E.G. Clinical
experience with sonographic contrast agents. Semin.
Ultrasound CT MR 1997, 18, 3–12. 25. Knopp, M.V.;
Tengg-Kobligk, H.; Floemer, F.; Schoenberg, S.O. Contrast
agents for MRA: Future directions. J. Magn. Reson. Imaging
1999, 10, 314–316. 26. Ambrose, J.A.; Tannenbaum, M.A.;
Alexopoulos, D.; Hjemdahl-Monsen, C.E.; Leavy, J.; Weiss,
M.; Borrico, S.; Gorlin, R.; Fuster, V. Angiographic
progression of coronary artery disease and the development
of myocardial infarction. J. Am. Coll. Cardiol. 1988, 12,
56–62. 27. Yuan, C.; Mitsumori, L.M.; Ferguson, M.S.;
Polissar, N.L.; Echelard, D.; Ortiz, G.; Small, R.; Davies,
J.W.; Kerwin, W.S.; Hatsukami, T.S. In vivo accuracy of
multispectral magnetic resonance imaging for identifying
lipid-rich necrotic cores and intraplaque hemorrhage in
advanced human carotid plaques. Circulation 2001, 104,
2051–2056. 28. Hatsukami, T.S.; Ross, R.; Polissar, N.L.;
Yuan, C. Visualization of fibrous cap thickness and rupture
in human atherosclerotic carotid plaque in vivo with
highresolution magnetic resonance imaging. Circulation
2000, 102, 959–964. 29. Vogl, T.J.; Balzer, J.O.; Mack,
M.G.; Bett, G.; Oppelt, A. Hybrid MR interventional imaging
system: Combined MR and angiography suites with single
interactive table. Feasibility study in vascular liver
tumor procedures. Eur. Radiol. 2002, 12, 1394–1400. V
Vascularization of Engineered Constructs

1. Patrick, C.W.; Chauvin, P.B.; Hobley, J.; Reece, G.P.


Preadipocyte seeded PLGA scaffolds for adipose tissue
engineering. Tissue Eng. 1999, 5 (2), 139–151.

2. Ochoa, E.R.; Vacanti, J.P. An overview of the pathology


and approaches to tissue engineering. Ann. N.Y. Acad. Sci.
2002, 979, 10–26. Discussion 35–38.

3. Tanaka, Y.; Tsutsumi, A.; Crowe, D.M.; Tajima, S.;


Morrison, W.A. Generation of an autologous tissue (matrix)
flap by combining an arteriovenous shunt loop with artificial
skin in rats: Preliminary report. Br. J. Plast. Surg. 2000,
53 (1), 51–57.

4. Mian, R.; Morrison, W.A.; Hurley, J.V.; Penington, A.J.;


Romeo, R.; Tanaka, Y.; Knight, K.R. Formation of new tissue
from an arteriovenous loop in the absence of added
extracellular matrix. Tissue Eng. 2000, 6 (6), 595– 603.

5. Cronin, K.J.; Messina, A.; Knight, K.R.; Penington,


A.J.; Morrison, W.A. A new murine model of spontaneous
autologous tissue engineering by combining arteriovenous
pedicle with matrix materials. Plast. Reconstr. Surg. 2004,
113, 260–269.

6. Cassell, O.C.; Morrison, W.A.; Messina, A.; Penington,


A.J.; Thompson, E.W.; Stevens, G.W.; Perera, I.M.;
Kleinman, H.K.; Hurley, J.V.; Romeo, R.; Knight, K.R. The
influence of extracellular matrix on the generation of
vascularized, engineered, transplantable tissue. Ann. N.Y.
Acad. Sci. 2001, 944, 429–442.

7. Hirschi, K.K.; Skalak, T.C.; Peirce, S.M.; Little, C.D.


Vascular assembly in natural and engineered tissues. Ann.
N.Y. Acad. Sci. 2002, 961, 223–242.

8. Folkman, J. Angiogenesis in cancer, vascular, rheumatoid


and other disease. Nat. Med. 1995, 1 (1), 27–31.

9. Risau, W. Mechanisms of angiogenesis. Nature 1997, 386


(6626), 671–674.

10. Larsen, W.J. Development of the vasculature. InHuman


Embryology, 3rd Ed.; Scott, W.J., Ed.; Churchill
Livingstone: London, 2001; 195–234.

11. Asahara, T.; Murohara, T.; Sullivan, A.; Silver, M.;


van der Zee, R.; Li, T.; Witzenbichler, B.; Schatteman, G.;
Isner, J.M. Isolation of putative progenitor endothelial
cells for angiogenesis. Science 1997, 275 (5302), 964– 967.

12. Burri, P.H.; Djonov, V. Intussusceptive angiogenesis–


the alternative to capillary sprouting. Mol. Aspects Med.
2002, 23 (Suppl. 6), 1–27.

13. Converse, J.M.; Smahel, J.; Ballantyne, D.L., Jr.;


Harper, A.D. Inosculation of vessels of skin graft and host
bed: A fortuitous encounter. Br. J. Plast. Surg. 1975, 28
(4), 274–282.

14. Darland, D.C.; D’Amore, P.A. Blood vessel maturation:


Vascular development comes of age. J. Clin. Invest. 1999,
103 (2), 157–158.

15. Stegemann, J.P.; Nerem, R.M. Phenotype modulation in


vascular tissue engineering using biochemical and
mechanical stimulation. Ann. Biomed. Eng. 2003, 31 (4),
391–402.

16. Cao, R.; Brakenhielm, E.; Pawliuk, R.; Wariaro, D.;


Post, M.J.; Wahlberg, E.; Leboulch, P.; Cao, Y. Angiogenic
synergism, vascular stability and improvement of hind-limb
ischemia by a combination of PDGF-BB and FGF-2. Nat. Med.
2003, 9 (5), 604–613. 17. Dvorak, H.F.; Dvorak, A.M.;
Manseau, E.J.; Wiberg, L.; Churchill, W.H. Fibrin gel
investment associated with line 1 and line 10 solid tumor
growth, angiogenesis, and fibroplasia in guinea pigs. Role
of cellular immunity, myofibroblasts, microvascular damage,
and infarction in line 1 tumor regression. J. Natl. Cancer
Inst. 1979, 62 (6), 1459–1472. 18. Pepper, M.S.; Ferrara,
N.; Orci, L.; Montesano, R. Potent synergism between
vascular endothelial growth factor and basic fibroblast
growth factor in the induction of angiogenesis in vitro.
Biochem. Biophys. Res. Commun. 1992, 189 (2), 824–831. 19.
Shweiki, D.; Itin, A.; Soffer, D.; Keshet, E. Vascular
endothelial growth factor induced by hypoxia may mediate
hypoxia-initiated angiogenesis. Nature 1992, 359 (6398),
843–845. 20. Shima, D.T.; Deutsch, U.; D’Amore, P.A.
Hypoxic induction of vascular endothelial growth factor
(VEGF) in human epithelial cells is mediated by increases
in mRNA stability. FEBS Lett. 1995, 370 (3), 203–208. 21.
Ferrara, N.; Houck, K.; Jakeman, L.; Leung, D.W. Molecular
and biological properties of the vascular endothelial
growth factor family of proteins. Endocr. Rev. 1992, 13
(1), 18–32. 22. Unemori, E.N.; Ferrara, N.; Bauer, E.A.;
Amento, E.P. Vascular endothelial growth factor induces
interstitial collagenase expression in human endothelial
cells. J. Cell. Physiol. 1992, 153 (3), 557–562. 23.
Iwaguro, H.; Yamaguchi, J.; Kalka, C.; Murasawa, S.;
Masuda, H.; Hayashi, S.; Silver, M.; Li, T.; Isner, J.M.;
Asahara, T. Endothelial progenitor cell vascular
endothelial growth factor gene transfer for vascular
regeneration. Circulation 2002, 105 (6), 732– 738. 24.
Epstein, S.E.; Kornowski, R.; Fuchs, S.; Dvorak, H.F.
Angiogenesis therapy: Amidst the hype, the neglected
potential for serious side effects. Circulation 2001, 104
(1), 115–119. 25. Elcin, Y.M.; Dixit, V.; Gitnick, G.
Extensive in vivo angiogenesis following controlled release
of human vascular endothelial cell growth factor:
Implications for tissue engineering and wound healing.
Artif. Organs 2001, 25 (7), 558–565. 26. Peters, M.C.;
Polverini, P.J.; Mooney, D.J. Engineering vascular networks
in porous polymer matrices. J. Biomed. Mater. Res. 2002, 60
(4), 668–678. 27. Lu, Y.; Shansky, J.; Del Tatto, M.;
Ferland, P.; Wang, X.; Vandenburgh, H. Recombinant vascular
endothelial growth factor secreted from tissue-engineered
bioartificial muscles promotes localized angiogenesis.
Circulation 2001, 104 (5), 594–599. 28. Lee, K.Y.; Peters,
M.C.; Mooney, D.J. Comparison of vascular endothelial
growth factor and basic fibroblast growth factor on
angiogenesis in SCID mice. J. Control. Release 2003, 87
(1–3), 49–56. 29. Nicosia, R.F.; Ottinetti, A. Modulation
of microvascular growth and morphogenesis by reconstituted
basement membrane gel in three-dimensional cultures of rat
aorta: A comparative study of angiogenesis in matrigel,
collagen, fibrin, and plasma clot. In Vitro Cell. Dev. Biol.
1990, 26 (2), 119–128. 30. Schweigerer, L.; Neufeld, G.;
Friedman, J.; Abraham, J.A.; Fiddes, J.C.; Gospodarowicz,
D. Capillary endothelial cells express basic fibroblast
growth factor, a mitogen that promotes their own growth.
Nature 1987, 325 (6101), 257–259. 31. Bikfalv, A.; Han,
Z.C. Angiogenic factors are hematopoietic growth factors
and vice versa. Leukemia 1994, 8 (3), 523–529. 32.
Passaniti, A.; Taylor, R.M.; Pili, R.; Guo, Y.; Long, P.V.;
Haney, J.A.; Pauly, R.R.; Grant, D.S.; Martin, G.R. A
simple, quantitative method for assessing angiogenesis and
antiangiogenic agents using reconstituted basement
membrane, heparin, and fibroblast growth factor. Lab.
Invest. 1992, 67 (4), 519–528. 33. Kawai, K.; Suzuki, S.;
Tabata, Y.; Ikada, Y.; Nishimura, Y. Accelerated tissue
regeneration through incorporation of basic fibroblast
growth factor-impregnated gelatin microspheres into
artificial dermis. Biomaterials 2000, 21 (5), 489–499. 34.
Tabata, Y. The importance of drug delivery systems in
tissue engineering. Pharm. Sci. Technol. Today 2000, 3 (3),
80–89. 35. Rinsch, C.; Quinodoz, P.; Pittet, B.; Alizadeh,
N.; Baetens, D.; Montandon, D.; Aebischer, P.; Pepper, M.S.
Delivery of FGF-2 but not VEGF by encapsulated genetically
engineered myoblasts improves survival and vascularization
in a model of acute skin flap ischemia. Gene Ther. 2001, 8
(7), 523–533. 36. Perets, A.; Baruch, Y.; Weisbuch, F.;
Shoshany, G.; Neufeld, G.; Cohen, S. Enhancing the
vascularization of three-dimensional porous alginate
scaffolds by incorporating controlled release basic
fibroblast growth factor microspheres. J. Biomed. Mater.
Res. 2003, 65A (4), 489–497. 37. Godbey, W.T.; Atala, A. In
vitro systems for tissue engineering. Ann. N.Y. Acad. Sci.
2002, 961, 10–26. 38. Black, A.F.; Berthod, F.; L’Heureux,
N.; Germain, L.; Auger, F.A. In vitro reconstruction of a
human capillary-like network in a tissue-engineered skin
equivalent. Faseb J. 1998, 12 (13), 1331–1340. 39. Frerich,
B.; Lindemann, N.; Kurtz-Hoffmann, J.; Oertel, K. In vitro
model of a vascular stroma for the engineering of
vascularized tissues. Int. J. Oral Maxillofac. Surg. 2001,
30 (5), 414–420. 40. Kaihara, S.; Borenstein, J.; Koka, R.;
Lalan, S.; Ochoa, E.R.; Ravens, M.; Pien, H.; Cunningham,
B.; Vacanti, J.P. Silicon micromachining to tissue engineer
branched vascular channels for liver fabrication. Tissue
Eng. 2000, 6 (2), 105–117. 41. Bos, G.W.; Poot, A.A.;
Beugeling, T.; van Aken, W.G.; Feijen, J. Small-diameter
vascular graft prostheses: Current status. Arch. Physiol.
Biochem. 1998, 106 (2), 100–115. 42. Schmidt, C.E.; Baier,
J.M. Acellular vascular tissues: Natural biomaterials for
tissue repair and tissue engineering. Biomaterials 2000, 21
(22), 2215–2231. 43. Huynh, T.; Abraham, G.; Murray, J.;
Brockbank, K.; Hagen, P.O.; Sullivan, S. Remodeling of an
acellular collagen graft into a physiologically responsive
neovessel. Nat. Biotechnol. 1999, 17 (11), 1083–1086. 44.
Field, P.L. The chemically treated bovine ureter–clinical
performance of a novel biological vascular prosthesis.
Cardiovasc. Surg. 2003, 11 (1), 30–34. 45. Crowe, D.M.;
Mitchell, G.M.; Hurley, J.V.; Olivier, T.V.; O’Loughlin,
K.; Morrison, W.A. Cold stored femoral vessels as
microvascular allografts: A preliminary study. Microsurgery
1994, 15 (10), 712–716. 46. Birchall, I.E.; Lee, V.W.;
Ketharanathan, V. Retention of endothelium on ovine
collagen biomatrix vascular conduits under physiological
shear stress. Biomaterials 2001, 22 (23), 3139–3144. 47.
Salacinski, H.J.; Punshon, G.; Krijgsman, B.; Hamilton, G.;
Seifalian, A.M. A hybrid compliant vascular graft seeded
with microvascular endothelial cells extracted from human
omentum. Artif. Organs 2001, 25 (12), 974–982. 48.
Shum-Tim, D.; Stock, U.; Hrkach, J.; Shinoka, T.; Lien, J.;
Moses, M.A.; Stamp, A.; Taylor, G.; Moran, A.M.; Landis,
W.; Langer, R.; Vacanti, J.P.; Mayer, J.E. Tissue
engineering of autologous aorta using a new biodegradable
polymer. Ann. Thorac. Surg. 1999, 68 (6), 2298–2304.
Discussion 2305. 49. Chu, C.F.; Lu, A.; Liszkowski, M.;
Sipehia, R. Enhanced growth of animal and human endothelial
cells on biodegradable polymers. Biochim. Biophys. Acta
1999, 1472 (3), 479–485. 50. Chupa, J.M.; Foster, A.M.;
Sumner, S.R.; Madihally, S.V.; Matthew, H.W. Vascular cell
responses to polysaccharide materials: In vitro and in vivo
evaluations. Biomaterials 2000, 21 (22), 2315–2322. 51.
Salacinski, H.J.; Tai, N.R.; Punshon, G.; Giudiceandrea,
A.; Hamilton, G.; Seifalian, A.M. Optimal
endothelialisation of a new compliant
poly(carbonate-urea)urethane vascular graft with effect of
physiological shear stress. Eur. J. Vasc. Endovasc Surg.
2000, 20 (4), 342–352. 52. Kaushal, S.; Amiel, G.E.;
Guleserian, K.J.; Shapira, O.M.; Perry, T.; Sutherland,
F.W.; Rabkin, E.; Moran, A.M.; Schoen, F.J.; Atala, A.;
Soker, S.; Bischoff, J.; Mayer, J.E. Functional
small-diameter neovessels created using endothelial
progenitor cells expanded ex vivo. Nat. Med. 2001, 7 (9),
1035–1040. 53. L’Heureux, N.; Paquet, S.; Labbe, R.;
Germain, L.; Auger, F.A. A completely biological
tissue-engineered human blood vessel. Faseb J. 1998, 12
(1), 47–56. 54. Niklason, L.E.; Gao, J.; Abbott, W.M.;
Hirschi, K.K.; Houser, S.; Marini, R.; Langer, R.
Functional arteries grown in vitro. Science 1999, 284
(5413), 489–493. 55. Tranquillo, R.T. The tissue-engineered
small-diameter artery. Ann. N.Y. Acad. Sci. 2002, 961,
251–254. 56. Nerem, R.M. Tissue engineering a blood vessel
substitute: The role of biomechanics. Yonsei Med. J. 2000,
41 (6), 735–739. 57. Griffith, L.G. Emerging design
principles in biomaterials and scaffolds for tissue
engineering. Ann. N.Y. Acad. Sci. 2002, 961, 83–95. 58.
Sieminski, A.L.; Gooch, K.J. Biomaterial-microvasculature
interactions. Biomaterials 2000, 21 (22), 2232–2241. 59.
Sharkawy, A.A.; Klitzman, B.; Truskey, G.A.; Reichert, W.M.
Engineering the tissue which V encapsulates subcutaneous
implants. II. Plasma-tissue exchange properties. J. Biomed.
Mater. Res. 1998, 40 (4), 586–597.

60. Yamamoto, M.T.Y.; Kawasaki, H.; Ikada, Y. Promotion of


fibrovascular tissue ingrowth into porous sponges by
fibroblast growth factor. J. Mater. Sci. Mater. Med. 2000,
11 (4), 213–218.

61. Kawaguchi, N.; Toriyama, K.; Nicodemou-Lena, E.; Inou,


K.; Torii, S.; Kitagawa, Y. De novo adipogenesis in mice at
the site of injection of basement membrane and basic
fibroblast growth factor. Proc. Natl. Acad. Sci. U.S.A.
1998, 95 (3), 1062–1066.

62. Tabata, Y.; Miyao, M.; Inamoto, T.; Ishii, T.; Hirano,
Y.; Yamaoki, Y.; Ikada, Y. De novo formation of adipose
tissue by controlled release of basic fibroblast growth
factor. Tissue Eng. 2000, 6 (3), 279–289.

63. Wang, W.; Gu, Y.; Tabata, Y.; Miyamoto, M.; Hori, H.;
Nagata, N.; Touma, M.; Balamurugan, A.N.; Kawakami, Y.;
Nozawa, M.; Inoue, K. Reversal of diabetes in mice by
xenotransplantation of a bioartificial pancreas in a
prevascularized subcutaneous site. Transplantation 2002, 73
(1), 122–129.

64. Balamurugan, A.N.; Gu, Y.; Tabata, Y.; Miyamoto, M.;


Cui, W.; Hori, H.; Satake, A.; Nagata, N.; Wang, W.; Inoue,
K. Bioartificial pancreas transplantation at prevascularized
intermuscular space: Effect of angiogenesis induction on
islet survival. Pancreas 2003, 26 (3), 279–285. 65. Kimura,
M.Y.; Ozeki, M.; Inamoto, T.; Tabata, Y. Adipose tissue
engineering based on human preadipocytes combined with
gelatin microspheres containing basic fibroblast growth
factor. Biomaterials 2003, 24 (14), 2513–2521. 66. Nor,
J.E.; Peters, M.C.; Christensen, J.B.; Sutorik, M.M.; Linn,
S.; Khan, M.K.; Addison, C.L.; Mooney, D.J.; Polverini,
P.J. Engineering and characterization of functional human
microvessels in immunodeficient mice. Lab. Invest. 2001, 81
(4), 453–463. 67. Koike, N.; Fukumura, D.; Gralla, O.; Au,
J.; Schechner, J.S.; Jain, R.K. Creation of long-lasting
blood vessels. Nature 2004, 428 (March 11), 138–139. 68.
Morrison, W.A.; Dvir, E.; Doi, K.; Hurley, J.V.; Hickey,
M.J.; O’Brien, B.M. Prefabrication of thin transferable
axial-pattern skin flaps: An experimental study in rabbits.
Br. J. Plast. Surg. 1990, 43 (6), 645–654.
Vascular Tissue Engineering

1. Heart Disease and Stroke Statistics-2004 Update;


American Heart Association: Dallas, TX, 2004.

2. Favaloro, R.G. Saphenous vein autograft replacement of


severe segmental coronary artery occlusion: operative
technique. Ann. Thorac. Surg. 1968, 5 (4), 334–339.

3. Fitzgibbon, G.M.; Leach, A.J.; Keon, W.J.; Burton, J.R.;


Kalfa, H.P. Coronary bypass graft fate. Angiographic study
of 1,179 vein grafts early, one year, and five years after
operation. J. Thorac. Cardiovasc. Surg. 1986, 91 (5),
773–778.

4. Weintraub, W.S.; Jones, E.L.; Craver, J.M.; Gyton, R.A.


Frequency of repeat coronary bypass or coronary angioplasty
after coronary artery bypass surgery using saphenous venous
grafts. Am. J. Cardiol. 1994, 73 (2), 103–112.

5. Predel, H.G.; Yang, Z.; von Segesser, L.; Turina, M.;


Buhler, F.R.; Luscher, T.F. Implications of pulsatile
stretch on growth of saphenous vein and mammary artery
smooth muscle. Lancet 1992, 340 (8824), 878– 879. 6.
Williams, S.K.; Rose, D.G.; Jarrell, B.E. Microvascular
endothelial cell sodding of ePTFE vascular grafts: improved
patency and stability of the cellular lining. J. Biomed.
Mater. Res. 1994, 28 (2), 203–212. 7. Brattich, M. Vascular
access thrombosis: etiology and prevention. ANNA J. 1999,
26 (5), 537–540. 8. Johansen, K.; Lyman, D.; Sauvage, L.
Biomaterials for hemodialysis access. Blood. Purif. 1994,
12 (1), 73–77. 9. Greisler, H.P. New Biologic and Synthetic
Vascular Prostheses; R.G. Landes, 1991. 10. Nerem, R.M.;
Seliktar, D. Vascular tissue engineering. Ann. Rev. Biomed.
Eng. 2001, 3, 225–243. 11. Vorp, D.; Maul, T.; Nieponice,
A. Molecular aspects of vascular tissue engineering. Front.
Biosci. 2005, 10, 768–789. 12. Herring, M. Endothelial
seeding blood flow surfaces. In Vascular Grafting: Clinical
Applications and Technique’eds’; PSG Inc., 1983; 275–314.
13. Abbott, W.M.; Megerman, J.; Hasson, J.E.; L’Italien,
G.; Warnock, D.F. Effect of compliance mismatch on vascular
graft patency. J. Vasc. Surg. 1987, 5 (2), 376–382. 14.
Haruguchi, H.; Teraoka, S. Intimal hyperplasia and
hemodynamic factors in arterial bypass and arteriovenous
grafts: a review. J. Artif. Organs 2003, 6 (4), 227–235.
15. Hubbell, J.A. Bioactive biomaterials. Curr. Opin.
Biotechnol. 1999, 10 (2), 123–129. 16. Niklason, L.E.; Gao,
J.; Abbott, W.M.; Hirschi, K.K.; Houser, S.; Marini, R.;
Langer, R. Functional arteries grown in vitro. Science
1999, 284 (5413), 489–493. 17. Grassl, E.D.; Oegema, T.R.;
Tranquillo, R.T. Fibrin as an alternative biopolymer to
type-I collagen for the fabrication of a media equivalent.
J. Biomed. Mater. Res. 2002, 60 (4), 607–612. 18. Cho,
S.W.; Lim, S.H.; Kim, I.K.; Hong, Y.S.; Kim, S.S.; Yoo,
K.J.; Park, H.Y.; Jang, Y.; Chang, B.C.; Choi, C.Y.; Hwang,
K.C.; Kim, B.S. Small-diameter blood vessels engineered
with bone marrow-derived cells. Ann. Surg. 2005, 241 (3),
506–515. 19. Sandusky, G.E.; Badylak, S.F.; Morff, R.J.;
Johnson, W.D.; Lantz, G. Histologic findings after in vivo
placement of small intestine submucosal vascular grafts and
saphenous vein grafts in the carotid artery in dogs. Am. J.
Pathol. 1992, 140 (2), 317–324. 20. Seliktar, D.; Black,
R.A.; Vito, R.P.; Nerem, R.M. Dynamic mechanical
conditioning of collagen-gel blood vessel constructs
induces remodeling in vitro. Ann. Biomed. Eng. 2000, 28
(4), 351–362. 21. Guan, J.; Sacks, M.S.; Beckman, E.J.;
Wagner, W.R. Synthesis, characterization, and
cytocompatibility of elastomeric, biodegradable
poly(ester-urethane)ureas based on poly(caprolactone) and
putrescine. J. Biomed. Mater. Res. 2002, 61 (3), 493–503.
22. Guan, J.; Fujimoto, K.L.; Sacks, M.S.; Wagner, W.R.
Preparation and characterization of highly porous,
biodegradable polyurethane scaffolds for soft tissue
applications. Biomaterials 2005, 26 (18), 3961–3971. 23.
Nieponice, A.S.L.; Guan, J.; Huard, J.; Wagner, W.R.; Vorp,
D.A. Preliminary evaluation of a novel muscle-derived stem
cell-based vascular construct. In ET2005: Engineering
Tissues; Hilton Head, SC, March 9–13, 2005. 24. Seliktar,
D.; Nerem, R.M.; Galis, Z.S. Mechanical strain-stimulated
remodeling of tissue engineered blood vessel constructs.
Tissue eng. 2003, 9 (4), 657–666. 25. Orban, J.M.; Wilson,
L.B.; Kofroth, J.A.; El-Kurdi, M.S.; Maul, T.M.; Vorp, D.A.
Crosslinking of collagen gels by transglutaminase. J.
Biomed. Mater. Res. 2004, 68A (4), 756–762. 26. Cummings,
C.L.; Gawlitta, D.; Nerem, R.M.; Stegemann, J.P. Properties
of engineered vascular constructs made from collagen,
fibrin, and collagen-fibrin mixtures. Biomaterials 2004, 25
(17), 3699–3706. 27. Berglund, J.D.; Nerem, R.M.; Sambanis,
A. Incorporation of intact elastin scaffolds in
tissue-engineered collagen-based vascular grafts. Tissue
Eng. 2004, 10 (9–10), 1526–1535. 28. Boland, E.D.;
Matthews, J.A.; Pawlowski, K.J.; Simpson, D.G.; Wnek, G.E.;
Bowlin, G.L. Electrospinning collagen and elastin:
preliminary vascular tissue engineering. Front. Biosci.
2004, 9, 1422–1432. 29. Badylak, S.F.; Lantz, G.C.; Coffey,
A.; Geddes, L.A. Small intestinal submucosa as a large
diameter vascular graft in the dog. J. Surg. Res. 1989, 47
(1), 74–80. 30. Badylak, S.F. Xenogeneic extracellular
matrix as a scaffold for tissue reconstruction. Transpl.
Immunol. 2004, 12 (3–4), 367–377. 31. Atala, A. Tissue
engineering and regenerative medicine: concepts for
clinical application. Rejuvenation Res. 2004, 7 (1), 15–31.
32. Schaner, P.J.; Martin, N.D.; Tulenko, T.N.; Shapiro,
I.M.; Tarola, N.A.; Leichter, R.F.; Carabasi, R.A.;
Dimuzio, P.J. Decellularized vein as a potential scaffold
for vascular tissue engineering. J. Vasc. Surg. 2004, 40
(1), 146–153. 33. Palecek, S.P.; Loftus, J.C.; Ginsberg,
M.H.; Lauffenburger, D.A.; Horwitz, A.F. Integrin-ligand
binding properties govern cell migration speed through
cellsubstratum adhesiveness. Nature 1997, 385 (6616),
537–540. 34. Seliktar, D.; Zisch, A.H.; Lutolf, M.P.;
Wrana, J.L.; Hubbell, J.A. MMP-2 sensitive, VEGF-bearing
bioactive hydrogels for promotion of vascular healing. J.
Biomed. Mater. Res. A 2004, 68 (4), 704–716. 35. Boura, C.;
Menu, P.; Payan, E.; Picart, C.; Voegel, J.C.; Muller, S.;
Stoltz, J.F. Endothelial cells grown on thin
polyelectrolyte mutlilayered films: an evaluation of a new
versatile surface modification. Biomaterials 2003, 24 (20),
3521–3530. 36. Steffens, G.C.; Yao, C.; Prevel, P.;
Markowicz, M.; Schenck, P.; Noah, E.M.; Pallua, N.
Modulation of angiogenic potential of collagen matrices by
covalent incorporation of heparin and loading with vascular
endothelial growth factor. Tissue Eng. 2004, 10 (9–10),
1502–1509. 37. Lee, M.; Kim, S.W. Polyethylene
glycol-conjugated copolymers for plasmid DNA delivery.
Pharm. Res. 2005, 22 (1), 1–10. 38. Carter, J.; Hristova,
K.; Harasaki, H.; Smith, W.A. Short exposure time
sensitivity of white cells to shear stress. ASAIO J. 2003,
49 (6), 687–691. 39. Dardik, A.; Chen, L.; Frattini, J.;
Asada, H.; Aziz, F.; Kudo, F.A.; Sumpio, B.E. Differential
effects of orbital and laminar shear stress on endothelial
cells. J. Vasc. Surg. 2005, 41 (5), 869–880. 40. Soletti L,
N.A.; Vorp, D.A. A new seeding device for tissue engineered
tubular structures. In ET2005: Engineering Tissues; Hilton
Head, SC, March 9–13, 2005. 41. Shirota, T.; He, H.; Yasui,
H.; Matsuda, T. Human endothelial progenitor cell-seeded
hybrid graft: proliferative and antithrombogenic potentials
in vitro and fabrication processing. Tissue Eng. 2003, 9
(1), 127–136. 42. Mazzucotelli, J.P.; Roudiere, J.L.;
Bernex, F.; Bertrand, P.; Leandri, J.; Loisance, D. A new
device for endothelial cell seeding of a small-caliber
vascular prosthesis. Artif. Organs 1993, 17 (9), 787–790.
43. Bowlin, G.L.; Rittgers, S.E. Electrostatic endothelial
cell transplantation within small-diameter (o6mm) vascular
prostheses: a prototype apparatus and procedure. Cell
transplant. 1997, 6 (6), 631–637. 44. van Wachem, P.B.;
Stronck, J.W.; Koers-Zuideveld, R.; Dijk, F.; Wildevuur,
C.R. Vacuum cell seeding: a new method for the fast
application of an evenly distributed cell layer on porous
vascular grafts. Biomaterials 1990, 11 (8), 602–606. 45.
Chien, S.; Li, S.; Shyy, Y.J. Effects of mechanical forces
on signal transduction and gene expression in endothelial
cells. Hypertension 1998, 31 (1 Pt 2), 162–169(Online). 46.
Owens, G.K.; Kumar, M.S.; Wamhoff, B.R. Molecular
regulation of vascular smooth muscle cell differentiation
in development and disease. Physiol. Rev. 2004, 84 (3),
767–801. 47. Owens, G.K. Molecular control of vascular
smooth muscle cell differentiation. Acta physiol. Scand.
1998, 164 (4), 623–635. 48. Valiunas, V.; Gemel, J.; Brink,
P.R.; Beyer, E.C. Gap junction channels formed by
coexpressed connexin40 and connexin43. Am. J. Physiol.
Heart Circ. Physiol. 2001, 281 (4), H1675–H1689. 49. Owens,
G.K. Regulation of differentiation of vascular smooth
muscle cells. Physiol. Rev. 1995, 75 (3), 487–517. 50.
Mckee, J.A.; Banik, S.S.; Boyer, M.J.; Hamad, N.M.; Lawson,
J.H.; Niklason, L.E.; Counter, C.M. Human arteries
engineered in vitro. EMBO Rep. 2003, 4 (6), 633–638. 51.
Fields, R.C.; Solan, A.; McDonagh, K.T.; Niklason, L.E.;
Lawson, J.H. Gene therapy in tissue engineered blood
vessels. Tissue Eng. 2003, 9 (6), 1281–1287. 52.
Remy-Zolghadri, M.; Laganiere, J.; Oligny, J.F.; Germain,
L.; Auger, F.A. Endothelium properties of a tissue
engineered blood vessel for small diameter vascular
reconstruction. J. Vasc. Surg. 2004, 39 (3), 613–620. 53.
L’Heureux, N.; Germain, L.; Labbe, R.; et al. In vitro
construction of a human blood vessel from cultured vascular
cells: a morphologic study. J. Vasc. Surg. 1993, 17 (3),
499–509. 54. L’Heureux, N.; Paquet, S.; Labbe, R.; et al. A
completely biological tissue engineered human blood vessel.
FASEB J. 1998, 12 (1), 47–56. 55. Niklason, L.E.; Langer,
R.S. Advances in tissue engineering of blood vessels and
other tissues. Transplant. Immunol. 1997, 5 (4), 303–306. V

56. Williams, B. Mechanical influences on vascular smooth


muscle cell function. J. Hypertens. 1998, 16 (12 Pt 2),
1921–1929.

57. Stegemann, J.P.; Nerem, R.M. Phenotype modulation in


vascular tissue engineering using biochemical and
mechanical stimulation. Ann. Biomed. Eng. 2003, 31 (4),
391–402.

58. Bonin, L.R.; Madden, K.; Shera, K.; Ihle, J.; Matthews,
C.; Aziz, S.; Perez-Reyes, N.; McDougall, J.K.; Conroy,
S.C. Generation and characterization of human smooth muscle
cell lines derived from atherosclerotic plaque.
Arterioscler. Thromb. Vasc. Biol. 1999, 19 (3), 575–587.

59. Collins, K.; Mitchell, J.R. Telomerase in the human


organism. Oncogene 2002, 21 (4), 564–579.
60. Shay, J.W.; Bacchetti, S. A survey of telomerase
activity in human cancer. Eur. J. Cancer 1997, 33 (5),
787–791.

61. Minguell, J.J.; Erices, A.; Conget, P. Mesenchymal stem


cells. Exp. Biol. Med. 2001, 226 (6), 507–520.

62. Jackson, K.A.; Majka, S.M.; Wulf, G.G.; Goodell, M.A.


Stem cells: a mini review. J. Cell. Biochem. Suppl. 2002,
38, 1–6.

63. Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res.


1991, 9 (5), 641–650.

64. Aubin, J.E.; Liu, F.; Malaval, L.; Gupta, A.K.


Osteoblast and chondroblast differentiation. Bone 1995, 17
(2 suppl), 77S–83S.

65. Jaiswal, N.; Haynesworth, S.E.; Caplan, A.I.; Bruder,


S.P. Osteogenic differentiation of purified, cultureexpanded
human mesenchymal stem cells in vitro. J. Cell biochem.
1997, 64 (2), 295–312.

66. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal,


R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti,
D.W.; Craig, S.; Marshak, D.R. Multilineage potential of
adult human mesenchymal stem cells. Science 1999, 284
(5411), 143–147.

67. Reyes, M.; Dudek, A.; Jahagirdar, B.; Koodie, L.;


Marker, P.H.; Verfaillie, C.M. Origin of endothelial
progenitors in human postnatal bone marrow. J. Clin.
Invest. 2002, 109 (3), 337–346.

68. Reyes, M.; Verfaillie, C.M. Characterization of


multipotent adult progenitor cells, a subpopulation of
mesenchymal stem cells. Ann. N. Y. Acad. Sci. 2001, 938,
231–233;Discussion 233–235

69. Wakitani, S.; Saito, T.; Caplan, A.I. Myogenic cells


derived from rat bone marrow mesenchymal stem cells exposed
to 5-azacytidine. Muscle Nerve 1995, 18 (12), 1417–1426.

70. Young, R.G.; Butler, D.L.; Weber, W.; Caplan, A.I.;


Gordon, S.L.; Fink, D.J. Use of mesenchymal stem cells in a
collagen matrix for achilles tendon repair. J. Orthop. Res.
1998, 16 (4), 406–413.

71. Terada, N.; Hamazaki, T.; Oka, M.; Hoki, M.; Mastalerz,
D.M.; Nakano, Y.; Meyer, E.M.; Morel, L.; Petersen, B.E.;
Scott, E.W. Bone marrow cells adopt the phenotype of other
cells by spontaneous cell fusion. Nature 2002, 416 (6880),
542–545.

72. Heil, M.; Ziegelhoeffer, T.; Mees, B.; Schaper, W. A


different outlook on the role of bone marrow stem cells in
vascular growth: bone marrow delivers software not
hardware. Circ. Res. 2004, 94 (5), 573–574. 73. Simper, D.;
Stalboerger, P.G.; Panetta, C.J.; Wang, S.; Caplice, N.M.
Smooth muscle progenitor cells in human blood. Circulation
2002, 106 (10), 1199–1204. 74. Abderrahim-Ferkoune, A.;
Bezy, O.; Astri-Roques, S.; Elabd, C.; Ailhaud, G.; Amri,
E.Z. Transdifferentiation of preadipose cells into smooth
muscle-like cells: role of aortic carboxypeptidase-like
protein. Exp. Cell Res. 2004, 293 (2), 219–228. 75.
Planat-Benard, V.; Silvestre, J.S.; Cousin, B.; Andre, M.;
Nibbelink, M.; Tamarat, R.; Clergue, M.; Manneville, C.;
Saillan-Barreau, C.; Duriez, M.; Tedgui, A.; Levy, B.;
Penicaud, L.; Casteilla, L. Plasticity of human adipose
lineage cells toward endothelial cells: physiological and
therapeutic perspectives. Circulation 2004, 109 (5),
656–663. 76. Deasy, B.M.; Huard, J. Gene therapy and tissue
engineering based on muscle-derived stem cells. Curr. Opin.
Mol. Ther. 2002, 4 (4), 382–389. 77. Lee, J.Y.; Qu
Petersen, Z.; Cao, B.; Kimura, S.; Jankowski, R.; Cummins,
J.; Usas, A.; Gates, C.; Robbins, P.; Wernig, A.; Huard, J.
Clonal isolation of muscle-derived cells capable of
enhancing muscle regeneration and bone healing. J. Cell
Biol. 2000, 150 (5), 1085–1100. 78. Qu-Petersen, Z.; Deasy,
B.; Jankowski, R.; Ikezawa, M.; Cummins, J.; Pruchnic, R.;
Mytinger, J.; Cao, B.; Gates, C.; Wernig, A.; Huard, J.
Identification of a novel population of muscle stem cells in
mice: potential for muscle regeneration. J. Cell biol.
2002, 157 (5), 851–864. 79. Kanda, K.; Matsuda, T.; Oka, T.
Mechanical stress induced cellular orientation and
phenotypic modulation of 3-d cultured smooth muscle cells.
ASAIO J. 1993, 39, M686–M690. 80. Niklason, L. Progress in
the engineering of small caliber arterial prostheses.
Engineering Tissue Growth International Conference &
Exposition, Pittsburgh, 2002. 81. Solan, A.; Mitchell, S.;
Moses, M.; Niklason, L. Effect of pulse rate on collagen
deposition in the tissue engineered blood vessel. Tissue
Eng. 2003, 9 (4), 579–586. 82. Seliktar, D.; Nerem, R.M.;
Galis, Z.S. The role of matrix metalloproteinase-2 in the
remodeling of cellseeded vascular constructs subjected to
cyclic strain. Ann. Biomed. Eng. 2001, 29 (11), 923–934.
83. Gross, M.F.; Friedman, M.H. Dynamics of coronary artery
curvature obtained from biplane cineangiograms. J. Biomech.
1998, 31 (5), 479–484. 84. Vorp, D.A.; Severyn, D.A.;
Steed, D.L.; Webster, M.W. A device for the application of
cyclic twist and extension on perfused vascular segments.
Am. J. Physiol. 1996, 270 (2 Pt 2), H787–H795. 85. Jeong,
S.I.; Kim, S.H.; Kim, Y.H.; Jung, Y.; Kwon, J.H.; Kim,
B.S.; Lee, Y.M. Manufacture of elastic biodegradable PLCL
scaffolds for mechano-active vascular tissue engineering.
J. Biomater. Sci. Polym. Ed. 2004, 15 (5), 645–660. 86.
Nomi, M.; Atala, A.; Coppi, P.D.; Soker, S. Principals of
neovascularization for tissue engineering. Mol. Asp. Med.
2002, 23 (6), 463–483. 87. Basilico, C.; Moscatelli, D. The
FGF family of growth factors and oncogenes. Adv. Cancer
Res. 1992, 59, 115– 165. V 88. Gilbert, S.F. Developmental
Biology; Sinauer Associates, 1997. 89. Pickering, J.G.;
Ford, C.M.; Tang, B.; Chow, L.H. Coordinated effects of
fibroblast growth factor-2 on expression of fibrillar
collagens, matrix metalloproteinases, and tissue inhibitors
of matrix metalloproteinases by human vascular smooth
muscle cells. Evidence for repressed collagen production
and activated degradative capacity. Arterioscler. Thromb.
Vasc. Biol. 1997, 17 (3), 475–482. 90. Fu, P.; Sodian, R.;
Luders, C.; Lemke, T.; Kraemer, L.; Hubler, M.; Weng, Y.;
Hoerstrup, S.P.; Meyer, R.; Hetzer, R. Effects of basic
fibroblast growth factor and transforming growth factor-beta
on maturation of human pediatric aortic cell culture for
tissue engineering of cardiovascular structures. ASAIO J.
2004, 50 (1), 9–14. 91. Stiles, C.D.; Capone, G.T.; Scher,
C.D.; Antoniades, H.N.; Van-Wyk, J.J.; Pledger, W.J. Dual
control of cell growth by somatomedins and platelet-derived
growth factor. Proc. Natl. Acad. Sci. U.S.A. 1979, 76 (3),
1279–1283. 92. Corjay, M.H.; Blank, R.S.; Owens, G.K.
Plateletderived growth factor-induced destabilization of
smooth muscle alpha-actin mRNA. J. Cell Physiol. 1990, 145
(3), 391–397. 93. Kinner, B.; Zaleskas, J.M.; Spector, M.
Regulation of smooth muscle actin expression and
contraction in adult human mesenchymal stem cells. Exp.
Cell Res. 2002, 278 (1), 72–83. 94. Iruela-Arispe, M.L.;
Sage, E.H. Endothelial cells exhibiting angiogenesis in
vitro proliferate in response to TGF-beta 1. J. Cell
Biochem. 1993, 52 (4), 414–430. 95. Pepper, M.S.
Transforming growth factor-beta: vasculogenesis,
angiogenesis, and vessel wall integrity. Cytokine Growth
Factor Rev. 1997, 8 (1), 21–43. 96. Eggermann, J.; Kliche,
S.; Jarmy, G.; Hoffmann, K.; Mayr Beyrle, U.; Debatin,
K.M.; Waltenberger, J.; Beltinger, C. Endothelial
progenitor cell culture and differentiation in vitro: a
methodological comparison using human umbilical cord blood.
Cardiovasc. Res. 2003, 58 (2), 478–486. 97. Gehling, U.M.;
Ergun, S.; Schumacher, U.; Wagener, C.; Pantel, K.; Otte,
M.; Schuch, G.; Schafhausen, P.; Mende, T.; Kilic, N.;
Kluge, K.; Schafer, B.; Hossfeld, D.K.; Fiedler, W. In
vitro differentiation of endothelial cells from
AC133-positive progenitor cells. Blood 2000, 95 (10),
3106–3112. 98. Wijelath, E.S.; Rahman, S.; Murray, J.;
Patel, Y.; Savidge, G.; Sobel, M. Fibronectin promotes
VEGFinduced CD34 cell differentiation into endothelial
cells. J. Vasc. Surg. 2004, 39 (3), 655–660. 99. Hood,
J.D.; Meininger, C.J.; Ziche, M.; et al. VEGF upregulates
ECNOS message, protein, and no production in human
endothelial cells. Am. J. Physiol. 1998, 274 (3 Pt 2),
H1054–H1058. 100. He, H.; Venema, V.J.; Gu, X.; Venema,
R.C.; Marrero, M.B.; Caldwell, R.B. Vascular endothelial
growth factor signals endothelial cell production of nitric
oxide and prostacyclin through FLK-1/KDR activation of
C-SRC. J. Biol. Chem. 1999, 274 (35), 25130–25135. 101.
Asahara, T.; Bauters, C.; Pastore, C.; Kearney, M.; Rossow,
S.; Bunting, S.; Ferrara, N.; Symes, J.F.; Isner, J.M.
Local delivery of vascular endothelial growth factor
accelerates reendothelialization and attenuates intimal
hyperplasia in balloon-injured rat carotid artery.
Circulation 1995, 91 (11), 2793–2801. 102. Hoerstrup, S.P.;
Zund, G.; Sodian, R.; Schnell, A.M.; Grunenfelder, J.;
Turina, M.I. Tissue engineering of small caliber vascular
grafts. Eur. J. Cardiothorac. Surg. 2001, 20 (1), 164–169.
103. Williams, C.; Wick, T.M. Perfusion bioreactor for
small diameter tissue-engineered arteries. Tissue Eng.
2004, 10 (5–6), 930–941. 104. Shinoka, T.; Matsumura, K.;
Hibino, N.; Murata, A.; Kosaka, Y.; Kurosawa, H. Clinical
practice of transplantation of regenerated blood vessels
using bone marrow cells. Nippon Naika Gakkai Zasshi 2003,
92 (9), 1776–1780. 105. Lantz, G.C.; Badylak, S.F.; Coffey,
A.C.; Geddes, L.A.; Blevins, W.E. Small intestinal
submucosa as a small-diameter arterial graft in the dog. J.
Invest. Surg. 1990, 3 (3), 217–227. 106. Lantz, G.C.;
Badylak, S.F.; Coffey, A.C.; Geddes, L.A.; Sandusky, G.E.
Small intestinal submucosa as a superior vena cava graft in
the dog. J. Surg. Res. 1992, 53 (2), 175–181. 107.
Sandusky, G.E.; Lantz, G.C.; Badylak, S.F. Healing
comparison of small intestine submucosa and ePTFE grafts in
the canine carotid artery. J. Surg. Res. 1995, 58 (4),
415–420. 108. Chue, W.L.; Campbell, G.R.; Caplice, N.;
Muhammed, A.; Berry, C.L.; Thomas, A.C.; Bennett, M.B.;
Campbell, J.H. Dog peritoneal and pleural cavities as
bioreactors to grow autologous vascular grafts. J. Vasc.
Surg. 2004, 39 (4), 859–867. 109. Matsumura, G.;
Miyagawa-Tomita, S.; Shin-Oka, T.; Ikada, Y.; Kurosawa, H.
First evidence that bone marrow cells contribute to the
construction of tissueengineered vascular autografts in
vivo. Circulation 2003, 108 (14), 1729–1734. 110. Stankus,
J.J.; Guan, J.; Wagner, W.R. Fabrication of biodegradable,
elastomeric scaffolds with sub-micron morphologies. J.
Biomed. Mater. Res. A 2004, 70 (4), 603–614. 111.
Weissberg, P.L.; Qasim, A. Stem cell therapy for myocardial
repair. Heart 2005, 91 (5), 696–702. 112. Isomatsu, Y.;
Shin’oka, T.; Matsumura, G.; Hibino, N.; Konuma, T.;
Nagatsu, M.; Kurosawa, H. Extracardiac total cavopulmonary
connection using a tissue-engineered graft. J. Thorac.
Cardiovasc. Surg. 2003, 126 (6), 1958–1962. 113. Naito, Y.;
Imai, Y.; Shin’oka, T.; Kashiwagi, J.; Aoki, M.; Watanabe,
M.; Matsumura, G.; Kosaka, Y.; Konuma, T.; Hibino, N.;
Murata, A.; Miyake, T.; Kurosawa, H. Successful clinical
application of tissue-engineered graft for extracardiac
Fontan operation. J. Thorac. Cardiovasc. Surg. 2003, 125
(2), 419–420. 114. Shin’oka, T.; Imai, Y.; Ikada, Y.
Transplantation of a tissue-engineered pulmonary artery. N.
Eng. J. Med. 2001, 344 (7), 532–533.
Vitrification

29. Marsland, T.P.; Evans, S.; Pegg, D.E. Dielectic


measurements for the design of an electromagnetic rewarming
system. Cryobiology 1987, 24, 311–323.

30. Robinson, M.P.; Pegg, D.E. Rapid electromagnetic


warming of cells and tissues. IEEE Trans. Biomed. Eng.
1999, 46 (12), 1413–1425.

31. Ruggera, P.S.; Fahy, G.M. Rapid and uniform


electromagnetic heating of aqueous cryoprotectant solutions
from cryogenic temperatures. Cryobiology 1990, 27, 465–478.

32. DeVries, A.L. Antifreeze peptides and glycopeptides in


cold-water fishes. Annu. Rev. Physiol. 1983, 45, 245–260.

33. Taylor, M.J.; Song, Y.C.; Brockbank, K.G.M.


Vitrification in Tissue Preservation: New Developments. In
Life in the Frozen State; Benson, E., Fuller, B., Lane, N.,
Eds.; Taylor and Francis Books: London, 2004; Chapter 22.

34. Walsh, J.R.; Taylor, M.J.; Brockbank, K.G.M. Storage


and Transport Issues for Tissue Engineered Medical
Products. In Tissue Engineered Medical Products (TEMPs),
ASTM STP 1452; Picciolo, G.L., Schutte, E., Eds.; ASTM
International: West Conshohocken, PA, 2003.

35. Kaiser, J. New prospects of putting organs on ice.


Science 2002, 295 (5557), 1015.

36. Karlsson, J.O.M. Cryopreservation: Freezing and


vitrification. Science 2002, 296, 655–656.

37. Taylor, M.J.; Song, Y.C.; Kheirabadi, B.S.; Lightfoot,


F.G.; Brockbank, K.G.M. Vitrification Fulfills its Promise as
an Approach to Reducing Freeze-Induced Injury in a
Multicellular Tissue. In Advances in Heat and Mass Transfer
in Biotechnology; American Society of Mechanical Engineers,
1999; 93–102. Volume number HTD-Vol. 363/BED-Vol. 44.

38. Song, Y.C.; Kheirabadi, B.S.; Lightfoot, F.G.;


Brockbank, K.G.M.; Taylor, M.J. Vitreous cryopreservation
maintains the function of vascular grafts. Nat. Biotechnol.
2000, 18, 296–299.

39. Fahy, G. Vitrification. In Low Temperature Biology;


McGrathDillerEds.; ASME, 1988; 113–146.

40. Fahy, G.M.; Saur, J.; Williams, R.J. Physical problems


with vitrification of large systems. Cryobiology 1990, 27,
492–510.

41. Song, Y.C.; Hagen, P.O.; Lightfoot, F.G.; Taylor, M.J.;


Smith, A.C.; Brockbank, K.G.M. In vivo evaluation of the
effects of a new ice-free cryopreservation process on
autologous vascular grafts. J. Invest. Surg. 2000, 13 (5),
279–288.

42. Brockbank, K.G.M.; Song, Y.C. Mechanisms of


bioprosthetic heart valve calcification. Transplantation
2003, 75 (8), 1133–1135.

43. Malinin, T.I.; Martinez, O.V.; Brown, M.D. Banking of


massive osteoarticular and intercalary bone allografts12
years’ experience. Clin. Orthop. 1985, 197, 44–57.

44. Marco, F.; Leon, C.; Lopez-Oliva, F.; Perez, A.J.;


Sanchez-Barba, A.; Lopez-Duran, S.L. Intact articular
cartilage cryopreservation. In vivo evaluation. Clin.
Orthop. 1992 , 283, 11–20.

45. Ohlendorf, C.; Tomford, W.W.; Mankin, H.J. Chondrocyte


survival in cryopreserved osteochondral articular
cartilage. J. Orthop. Res. 1996, 14, 413–416. 46. Bakay,
A.; Csonge, L.; Papp, G.; Fekete, L. Osteochondral
resurfacing of the knee joint with allograft. Clinical
analysis of 33 cases. Int. Orthop. 1998, 22 (277), 281. 47.
Stone, B.B.; Defranzo, B.E.; Dicesare, C.; Rapko, S.M.;
Brockbank, K.G.M.; Wolfrum, J.M.; Wrenn, C.A.; Grossman,
J.D. Cryopreservation of human articular cartilage for
autologous chondrocyte transplantation. Cryobiology 1998,
37 (abstract), 445–446. 48. Tomford, W.W.; Fredericks,
G.R.; Mankin, H.J. Studies on cryopreservation of articular
cartilage chondrocytes. J. Bone Jt. Surg., Am. 1984, 66,
253–259. 49. Brockbank, K.G.M. Method for Preserving
Musculoskeletal Tissues. Patent 5131850, 1992. 50.
Brockbank, K.G.M.; Lightfoot, F.G.; Song, Y.C.; Taylor,
M.J. Interstitial ice formation in cryopreserved
homografts: A possible cause of tissue deterioration and
calcification in vivo. J. Heart Valve Dis. 2000, 9 (2),
200–206. 51. Song, Y.C.; Lightfoot, F.G.; Chen, Z.; Taylor,
M.J.; Brockbank, K.G.M. Vitreous preservation of rabbit
articular cartilage. Cell Preserv. Technol. 2004, 2 (1),
67–74. 52. Polge, C.; Smith, A.Y.; Parkes, A.S. Revival of
spermatozoa after vitrification and dehydration at low
temperatures. Nature 1949, 164, 666. 53. Lovelock, J.E.;
Bishop, M.W.H. Prevention of freezing damage to living
cells by dimethyl sulfoxide. Nature 1959, 183, 1394. 54.
Muldrew, K.; Hurtig, M.; Schachar, N.; McGann, L.E.
Localization of freezing injury in articular cartilage.
Cryobiology 1994, 31, 31–38. 55. Wu, F.J.; Davisson, T.H.;
Pegg, D.E. Preservation of tissue-engineered articular
cartilage. Cryobiology 1998, 37, 410. 56. Muldrew, K.;
Novak, K.; Studholme, C.; Wohl, G.; Zernicke, R.; Schachar,
N.; McGann, L.E. Transplantation of articular cartilage
following a step-cooling cryopreservation protocol.
Cryobiology 2001, 43, 260–267. 57. Karlsson, J.O.M.; Toner,
M. Long-term storage of tissues by cryopreservation:
Critical issues. Biomaterials 1994, 17, 243–256. 58. Jomha,
N.M.; Anoop, P.C.; Bagnall, K.; McGann, L.E. Effects of
increasing concentrations of dimethyl sulfoxide during
cryopreservation of porcine articular cartilage. Cell
Preserv. Technol. 2002, 1 (2), 111. 59. Mazur, P. Freezing
of living cells: Mechanisms and implications. Am. J.
Physiol. 1984, 247 , 125. 60. Taylor, M.J.; Pegg, D.E. The
effect of ice formation on the function of smooth muscle
tissue following storage at �211C and �601C. Cryobiology
1982, 20, 36–40. 61. Hunt, C.J.; Taylor, M.J.; Pegg, D.E.
Freeze-substitution and isothermal freeze fixation studies
to elucidate the pattern of ice formation on smooth muscle
at 252 K (�21 C). J. Microsc. 1982, 125 (2), 177–186. 62.
Taylor, M.J. Sub-Zero Preservation and the Prospect of
Long-Term Storage of Multicellular Tissues and Organs. In
Transplantation Immunology-Clinical and Experimental;
Calne, R.Y., Ed.; Oxford University Press: Oxford, 1984;
360–390. 63. Boutron, P.; Mehl, P.; Kaufmann, A.; Augibaud,
P. Glass forming tendency and stability of the amorphous
stat in aqueous solutions of polyalcohols with four
carbons. I Binary systems water-polyalcohols. Cryobiology
1986, 23, 453–469. 64. Boutron, P.; Mehl, P. Theoretical
predictions of devitrification tendency: Determination of
critical warming rates using finite expansions. Cryobiology
1990, 27, 359–377. 65. Kroener, C.; Luyet, B. Formation of
cracks during the vitrification of glycerol solutions and
disappearance of the cracks during rewarming. Biodynamica
1966, 10 (201), 47–52. 66. Williams, R.J. Four modes of
nucleation in viscous solutions. Cryobiology 1989, 26 (6),
568. 67. Rabin, Y.; Podbilewicz, B. Temperature-controlled
microscopy for 4-D imaging of living cells: Apparatus,
thermal analysis, and temperature dependency of embryonic
elongation in C. elegans. J. Microsc. 2000, 199 (3),
214–223. 68. Rabin, Y.; Steif, P.S. Thermal stresses in a
freezing sphere and its application to cryobiology. ASME J.
Appl. Mech. 1998, 65 (2), 328–333. 69. Rabin, Y.; Steif,
P.S. Thermal stress modeling in cryosurgery. Int. J. Solids
Struct. 2000, 37, 2363–2375. 70. Rubinsky, B.; Cravalho,
E.G.; Mikic, B. Thermal stress in frozen organs.
Cryobiology 1980, 17, 66–73. 71. Rabin, Y.; Taylor, M.J.;
Wolmark, N. Thermal expansion measurements of frozen
biological tissues at cryogenic temperatures. J. Biomech.
Eng. 1998, 120, 259–266. 72. Song, Y.C.; Taylor, M.J.;
Brockbank, K.G.M. Ice-free cryopreservation of arterial
grafs. Cryobiology 2001, 41, 370 (abstract). 73. Brockbank,
K.G.M.; Song, Y.C. Development of preservation methods for
cellular engineered tissues. Presented at the First World
Congress on Regenerative Medicine, Leipzig, Germany. Int.
J. Artif. Organs 2003, 26, 9, 818 (abstract). 74. Song,
Y.C.; An, Y.H.; Kang, Q.K.; Li, C.; Bogg, J.M.; Chen, Z.;
Taylor, M.J.; Brockbank, K.G.M. Vitreous preservation of
articular cartilage grafts. J. Invest. Surg. 2004, 17, 1–6.
V
Wear Debris

5. Besong, A.A.; Tipper, J.L.; Ingham, E.; Stone, M.H.;


Wroblewski, B.M.; Fisher, J. Quantitative comparison of
wear debris from UHMWPE that has and has not been
sterilised by gamma irradiation. J. Bone Jt. Surg. 1998,
80B, 340–344.

6. Fisher, J.; Dowson, D. Tribology of total artificial


joints. Proc. Inst. Mech. Eng., H 1991, 205, 73–79.

7. Barbour, P.S.M.; Stone, M.H.; Fisher, J. A hip joint


simulator study using new and physiologically scratched
femoral heads with ultra-high molecular weight polyethylene
acetabular cups. Proc. Inst. Mech. Eng., H 2000, 214,
569–576.

8. Blunn, G.; Brach del Preva, E.M.; Costa, L.; Fisher, J.;
Freeman, M.A.R. Ultrahigh molecular weight polyethylene
(UHMWPE) in total knee replacement: Fabrication,
sterilisation and wear. J. Bone Jt. Surg. 2002, 84-B,
946–949.

9. Fisher, J.; Chan, K.L.; Hailey, J.L.; Shaw, D.; Stone,


M.H. Preliminary study of the effect of ageing following
irradiation on the wear of ultrahigh-molecular-weight
polyethylene. J. Arthroplast. 1995, 10 (5), 689–692.

10. Blanchet, L. Serum lubrication absorption by UHMWPE.


Orthopaedic bearing implants. J. Tribol. 2002, 124, 1–4.

11. Bell, J.; Tipper, J.L.; Ingham, E.; Stone, M.H.;


Fisher, J. The influence of phospholipid concentration in
proteincontaining lubricants on the wear of ultra-high
molecular weight polyethylene in artificial hip joints.
Proc. Inst. Mech. Eng., H 2001, 215, 259–263.

12. Pooley, C.M.; Tabor, D. Friction and molecular


structure: The behaviour of some thermoplastics. Proc. R.
Soc. Lond. 1972, 329A, 251–258.

13. Wang, A.; Stark, C.; Dumbleton, J.H. Mechanistic and


morphological origins of UHMWPE wear debris in total joint
replacement prostheses. Proc. Inst. Mech. Eng., H 1996,
210, 137–141.

14. Bennett, D.; Orr, J.; Beverland, D.E.; Baker, R. The


influence of shape and sliding distance of femoral head loci
on the wear of acetabular cups. Proc. Inst. Mech. Eng., H
2002, 216, 393–402.
15. Campbell, P.; Doorn, P.; Dorey, F.; Amstutz, H.C. Wear
and morphology of ultra-high molecular weight polyethylene
wear particles from total hip replacements. Proc. Inst.
Mech. Eng., H 1996, 210, 167–174.

16. Green, T.R.; Fisher, J.; Stone, M.H.; Wroblewski, B.M.;


Ingham, E. Polyethylene particles of a ‘critical size’ are
necessary for the induction of cytokines by macrophages in
vitro. Biomaterials 1998, 19, 2297–2302.

17. Green, T.R.; Fisher, J.; Matthews, J.B.; Stone, M.H.;


Ingham, E. Effect of size and dose on bone resorption
activity of macrophages by in vitro clinically relevant
ultra high molecular weight polyethylene particles. J.
Biomed. Mater. Res. (Appl. Biomater.) 2000, 53, 490–497.

18. Ingram, J.; Matthews, J.B.; Tipper, J.L.; Stone, M.H.;


Fisher, J.; Ingham, E. Comparison of the biological
activity of grade GUR 1120 and GUR 415 HP UHMWPE wear
debris. Bio-Med. Mater. Eng. 2002 , 12, 177–188.

19. Fisher, J.; Bell, J.; Barbour, P.S.M.; Tipper, J.L.;


Matthews, J.B.; Besong, A.A.; Stone, M.H.; Ingham, E. A
novel method for the prediction of functional biological
activity of polyethylene wear debris. Proc. Inst. Mech.
Eng., H 2001, 215, 127–132. 20. Endo, M.; Tipper, J.L.;
Barton, D.C.; Stone, M.H.; Ingham, E.; Fisher, J.
Comparison of wear, wear debris and functional biological
activity of moderately crosslinked and non-crosslinked
polyethylenes in hip prostheses. Proc. Inst. Mech. Eng., H
2002, 216, 111–122. 21. Tipper, J.L.; Firkins, P.J.;
Besong, A.A.; Barbour, P.S.M.; Nevelos, J.; Stone, M.H.;
Ingham, E.; Fisher, J. Characterisation of wear debris from
UHMWPE on zirconia ceramic, metal-on-metal and alumina
ceramic-on-ceramic hip prostheses generated in a
physiological anatomical hip joint simulator. Wear 2001,
250, 120–128. 22. McKellop, H.; Shen, F.; Lu, B.; Campbell,
P.; Salovey, R. Development of extemely wear resistant
UHMWPE for total hip replacement. J. Bone Jt. Surg. 1999,
81A, 157–167. 23. Muratoglu, O.K.; Bragdon, C.R.; O’Connor,
D.O.; Harris, W. Unified wear model for highly crosslinked
UHMWPE. Biomaterials 1999, 20, 1463–1478. 24. Kurtz, S.M.;
Muratoglu, O.K.; Evan, M.; Edidin, A. Advances in
processing, sterilisation and crosslinking of ultra high
molecular weight polyethylene. Biomaterials 1999, 81A,
157–167. 25. Galvin, A.; Tipper, J.L.; Stone, M.H.; Ingham,
E.; Fisher, J. Reduction in wear of crosslinked
polyethylene under different conditions. Refining future
strategies in hip replacement. Proc. Inst. Mech. Eng. 2002,
C601/ 005. 26. Ingram, J.H.; Fisher, J.; Stone, M.H.;
Ingham, E. Effect of crosslinking on biological activity of
UHMWPE wear debris. Trans. 49th Orthop. Res. Soc. 2003, 28,
1439. 27. Boutin, P.; Christel, P.S.; Dorlot, J.M.;
Meunier, A.; deRoquancourt, B.D.; Herman, S.; Sedel, L.;
Witvoet, J. The use of dense alumina–alumina combination in
total hip replacement. J. Biomed. Mater. Res. 1988, 22,
1203–1209. 28. McKellop, H.; Park, S.H.; Chiesa, R.; Doorn,
P.; Normand, B.L.P.; Grigris, P.; Amstutz, H. In vivo wear
of three types of metal on metal hip prostheses during 2
decades of use. Clin. Orthop. Relat. Res. 1966, 329,
S128–S140. 29. Nevelos, J.E.; Prudhommeaux, F.; Hamadouche,
M.; Doyle, C.; Ingham, E.; Meunier, A.; Nevelos, A.B.;
Sedel, L.; Fisher, J. Comparative analysis of two different
types of alumina–alumina hip prosthesis retrieved for
aseptic loosening. J. Bone Jt. Surg. 1999, 83-B (4),
598–603. 30. Nevelos, J.; Ingham, E.; Doyle, C.; Streicher,
R.; Nevelos, A.; Walter, W.; Fisher, J. Microseparation of
the centers of alumina–alumina artificial hip joints during
simulator testing produces clinically relevant wear and
patterns. J. Arthroplast. 2000, 15, 793–795. 31. Nevelos,
J.E.; Ingham, E.; Doyle, C.; Nevelos, A.B.; Fisher, J.
Analysis of retrieved alumina ceramic components for
Mittelmeier total hip prostheses. Biomaterials 1999, 20,
1833–1840. 32. Nevelos, J.E.; Ingham, E.; Doyle, C.;
Nevelos, A.B.; Fisher, J. The influence of acetabular cup
angle on the wear of ‘‘BIOLOX Forte’’ alumina ceramic
bearing couples in a hip joint simulator. J. Mater. Sci.,
Mater. Med. 2001, 12, 141–144. 33. Nevelos, J.E.; Ingham,
E.; Doyle, C.; Nevelos, A.B.; Fisher, J. Wear of HIPed and
non-HIPed alumina– alumina hip joints under standard and
severe simulator testing conditions. Biomaterials 2001, 22,
2191–2197. 34. Tipper, J.L.; Hatton, A.; Nevelos, J.E.;
Ingham, E.; Doyle, C.; Streicher, R.; Nevelos, A.B.;
Fisher, J. Alumina–alumina artificial hip joints. Part II:
Characterisation of the wear debris from in vitro hip
joints simulations. Biomaterials 2002, 23, 3441–3448. 35.
Hatton, A.; Nevelos, J.E.; Nevelos, A.A.; Banks, R.E.;
Fisher, J.; Ingham, E. Alumina–alumina artificial hip
joints. Part I: A histological analysis and
characterisation of wear debris by laser capture
microdissection of tissues retrieved at revision.
Biomaterials 2002, 23, 3429–3440. 36. Stewart, T.; Tipper,
J.L.; Streicher, R.; Ingham, E.; Fisher, J. Long-term wear
or HIPed alumina on alumina bearings for THR under
microseparation conditions. J. Mater. Sci., Mater. Med.
2001, 12, 1053–1056. 37. Ingham, E.; Hatton, A.; Matthews,
J.B.; Nevelos, J.E.; Fisher, J. Osteolytic potential of
cliically relevant alumina ceramic wear debris. Trans. 49th
Orthop. Res. Soc. 2003, 28, 212. 38. Tipper, J.L.;
Matthews, J.B.; Ingham, E.; Stewart, T.D.; Fisher, J.;
Stone, M.H. Wear and Functional Biological Activity of Wear
Debris Generated from UHMWPEon-Zirconia Ceramic,
Metal-on-Metal, and Alumina Ceramic-on-Ceramic Hip
Prostheses During Hip Simulator Testing. In Friction,
Lubrication and Wear of Artificial Joints; PEP: London;
2002; 7–28. 39. Doorn, P.F.; Mirra, J.M.; Campbell, P.A.;
Amstutz, H.C. Tissue reaction to metal on metal total hip
prostheses. Clin. Orthop. 1996, 329, S187–S205. 40. Jacobs,
J.J.; Skipor, A.K.; Patterson, L.M.; Hallab, N.J.;
Paprosky, W.G.; Black, J.; Galante, J.O. Metal release in
patients who have had a primary total hip arthroplasty. A
prospective, controlled, longitudinal study. J. Bone Jt.
Surg. 1998, 80A, 1447–1458. 41. Tipper, J.L.; Firkins,
P.J.; Ingham, E.; Stone, M.H.; Farrar, R.; Fisher, J.
Quantitative analysis of the wear and wear debris from low
and high carbon content cobalt chrome alloys used in metal
on metal total hip replacements. J. Mater. Sci., Mater.
Med. 1999, 10, 355–362. 42. Firkins, P.J.; Tipper, J.L.;
Saadatzadeh, M.R.; Ingham, E.; Stone, M.H.; Farrar, R.;
Fisher, J. Quantitative analysis of wear and wear debris
from metal-on-metal hip prostheses tested in a
physiological hip joint simulator. Bio-Med. Mater. Eng.
2001, 11, 143–157. 43. Firkins, P.J.; Tipper, J.L.; Ingham,
E.; Stone, M.H.; Farrar, R.; Fisher, J. Influence of
simulator kinematics on the wear of metal-on-metal hip
prostheses. Proc. Inst. Mech. Eng., H 2001, 215, 119–121.
44. Medley, J.B.; Chan, F.W.; Krygier, J.J.; Bobyn, J.D.
Comparison of alloys and designs in a hip simulator study
of metal on metal implants. Clin. Orthop. Relat. Res. 1996,
329, S148–S159. 45. Stewart, T.; Butterfield, M.; Stone,
M.H.; Williams, S.; Ingham, E.; Fisher, J. Wear of metal
metal and ceramic ceramic hip prostheses under swing phase
microseparation. Trans. 48th Orthop. Res. Soc. 2002, 48,
128. 46. Jin, Z.M.; Dowson, D.; Fisher, J. Analysis of fluid
film lubrication in artificial hip joint replacements with
surfaces of high elastic modulus. Proc. Inst. Mech. Eng., H
1997, 211, 247–256. 47. Germain, M.A.; Hatton, A.;
Williams, S.; Matthews, J.B.; Stone, M.H.; Fisher, J.;
Ingham, E. Comparison of cytoxicity of clinically relevant
cobalt chrome and alumina ceramic wear particles.
Biomaterials 2003, 24, 469–479. 48. Goodfellow, J. Knee
prostheses—One step forward two steps back. J. Bone Jt.
Surg. 1992, 74B, 1–2. 49. Jones, S.M.G.; Pinder, I.M.;
Moran, C.A.; Malcolm, A.J. Polyethylene wear in uncemented
knee replacements. J. Bone Jt. Surg. 1992, 74B, 18–22. 50.
Fisher, J. Wear of ultra high molecular weight polyethylene
in total artificial joints. Curr. Orthop. 1994, 8, 164–169.
51. Bell, C.J.; Walker, P.G.; Blunn, G.W. The effect of
oxidation on the delamination of UHMWPE tibial components.
J. Arthroplast. 1998, 13, 280–290. 52. Reeves, E.A.;
Barton, D.C.; FitzPatrick, D.; Fisher, J. Comparison of gas
plasma and gamma irradiation in air sterilization on the
delamination wear of the ultrahigh molecular weight
polyethylene used in knee replacements. Proc. Inst. Mech.
Eng., H 2000, 214, 249–255. 53. Fisher, J.; McEwen, H.M.J.;
Barnett, P.I.; Bell, C.J.; Stewart, T.D.; Stone, M.H.;
Ingham, E. Mini-symposium: Total knee replacement—Practical
considerations (i) Wear of polyethylene in artificial knee
joints. Curr. Orthop. 2001, 15, 399–405. 54. Schai, P.;
Thornhill, T.; Scott, R.D. Total knee arthroplasty with the
PFC system. J. Bone Jt. Surg. 1998, 80B, 850–851. 55.
Howling, G.I.; Barnett, P.I.; Tipper, J.L.; Stone, M.H.;
Fisher, J.; Ingham, E. Quantitative characterization of
polyethylene debris isolated from periprosthetic tissue in
early failure knee implants and early and late failure
Charnley hip implants. J. Biomed. Mater. Res. Appl.
Biomater. 2001, 58, 415–420. 56. Walker, P.S.; Blunn, G.W.;
Perry, J.P.; Bell, C.J.; Campbell, P. Methodology for long
term wear testing of knee replacements. Clin. Orthop.
Relat. Res. 2000, 372, 290–301. 57. Benson, L.C.; Des
Jardins, J.; Harman, M.K.; LaBerge, M. Effect of stair
descent loading on UHMWPE wear in a force controller knee
simulator. Proc. Inst. Mech. Eng., H 2002, 216, 409–418.
58. Burgess, I.; Kolan, M.; Cunningham, J.; Unsworth, A.
Development of a six station knee wear simulator. Proc.
Inst. Mech. Eng., H 1997, 211 , 37–47. 59. McEwen, H.M.J.;
McNulty, D.E.; Auger, D.D.; Farrar, R.; Liao, Y.S.; Stone,
M.H.; Fisher, J. Wear—Analysis of Mobile Bearing Knee. In
LCS Mobile Bearing Knee Arthroplasty—A 25 Years Worldwide
Review; Hamelynck, S., Ed.; Springer: Amsterdam; 2002;
67–73. Chapter 8.1. W

60. Barnett, P.I.; McEwen, H.M.J.; Auger, D.D.; Stone,


M.H.; Ingham, E.; Fisher, J. Investigation of wear of knee
prostheses in a new displacement/force-controlled
simulator. Proc. Inst. Mech. Eng., H 2002, 216, 51–61.

61. Barnett, P.I.; Fisher, J.; Auger, D.D.; Stone, M.H.;


Ingham, E. Comparison of wear in a total knee replacement
under different kinematic conditions. J. Mater. Sci.,
Mater. Med. 2001, 12, 1039–1042.

62. McEwen, H.M.J.; Fisher, J.; Goldsmith, A.A.J.; Auger,


D.D.; Hardaker, C.; Stone, M.H. Wear of fixed bearing and
rotating platform mobile bearing knees subjected to high
levels of internal and external tibial rotation. J. Mater.
Sci., Mater. Med. 2001, 12, 1049–1052. 63. Besong, A.A.;
Tipper, J.L.; Stone, M.H.; Ingham, E.; Fisher, J. The
influence of joint kinematics on the number and morphology
of polyethylene wear particles in models of hip and knee
prostheses. Trans. IMechE Conf. Knee Replace. C 1999,
70–73.
Wear Debris, Bone Resorption Animal Models

27. Yang, S.Y.; Mayton, L.; Wu, B.; Goater, J.J.; Schwarz,
E.M.; Wooley, P.H. Adeno-associated virus-mediated
osteoprotegerin gene transfer protects against particulate
polyethylene-induced osteolysis in a murine model.
Arthritis Rheum. 2002, 46 (9), 2514–2523.

28. Schwarz, E.M.; Benz, E.B.; Lu, A.P.; Goater, J.J.;


Mollano, A.V.; Rosier, R.N.; Puzas, J.E.; O’Keefe, R.J.
Quantitative small-animal surrogate to evaluate drug
efficacy in preventing wear debris-induced osteolysis. J.
Orthop. Res. 2000, 18 (6), 849–855.

29. Li, P.; Schwarz, E.M.; O’Keefe, R.J.; Ma, L.; Looney,
R.J.; Ritchlin, C.T.; Boyce, B.F.; Xing, L. Systemic tumor
necrosis factor alpha mediates an increase in peripheral
CD11bhigh osteoclast precursors in tumor necrosis factor
alpha-transgenic mice. Arthritis Rheum. 2004, 50 (1),
265–276.

30. Wong, P.K.; Campbell, I.K.; Egan, P.J.; Ernst, M.;


Wicks, I.P. The role of the interleukin-6 family of
cytokines in inflammatory arthritis and bone turnover.
Arthritis Rheum. 2003, 48 (5), 1177–1189.

31. Poli, V.; Balena, R.; Fattori, E.; Markatos, A.;


Yamamoto, M.; Tanaka, H.; Ciliberto, G.; Rodan, G.A.;
Costantini, F. Interleukin-6 deficient mice are protected
from bone loss caused by estrogen depletion. EMBO J. 1994,
13 (5), 1189–1196.

32. Sacomen, D.; Smith, R.L.; Song, Y.; Fornasier, V.;


Goodman, S.B. Effects of polyethylene particles on tissue
surrounding knee arthroplasties in rabbits. J. Biomed.
Mater. Res. 1998, 43 (2), 123–130.

33. Marnett, L.J.; Rowlinson, S.W.; Goodwin, D.C.;


Kalgutkar, A.S.; Lanzo, C.A. Arachidonic acid oxygenation
by COX-1 and COX-2. Mechanisms of catalysis and inhibition.
J. Biol. Chem. 1999, 274 (33), 22903–22906.

34. Kobayashi, T.; Narumiya, S. Function of prostanoid


receptors: Studies on knockout mice. Prostaglandins Other
Lipid Mediat. 2002, 68–69, 557–573.

35. Zhang, X.P.; Morham, S.G.; Langenbach, R.; Young, D.A.;


Xing, L.; Boyce, B.F.; Puzas, J.E.; Rosier, R.N.; O’Keefe,
R.J.; Schwarz, E.M. Evidence for a direct role of
cyclo-oxygenase 2 in implant wear debris-induced
osteolysis. J. Bone Miner. Res. 2001, 16 (4), 660–670.

36. Li, X.; Okada, Y.; Pilbeam, C.C.; Lorenzo, J.A.;


Kennedy, C.R.; Breyer, R.M.; Raisz, L.G. Knockout of the
murine prostaglandin EP2 receptor impairs
osteoclastogenesis in vitro. Endocrinology 2000, 141 (6),
2054–2051.

37. Suzawa, T.; Miyaura, C.; Inada, M.; Maruyama, T.;


Sugimoto, Y.; Ushikubi, F.; Ichikawa, A.; Narumiya, S.;
Suda, T. The role of prostaglandin E receptor subtypes
(EP1, EP2, EP3, and EP4) in bone resorption: An analysis
using specific agonists for the respective EPs.
Endocrinology. 2000, 141 (4), 1554–1559.

38. Paralkar, V.M.; Borovecki, F.; Ke, H.Z.; Cameron, K.O.;


Lefker, B.; Grasser, W.A.; Owen, T.A.; Li, M.;
DaSilva-Jardine, P.; Zhou, M.; Dunn, R.L.; Dumont, F.;
Korsmeyer, R.; Krasney, P.; Brown, T.A.; Plowchalk, D.;
Vukicevic, S.; Thompson, D.D. An EP2 receptor-selective
prostaglandin E2 agonist induces bone healing. Proc. Natl.
Acad. Sci. USA 2003, 100 (11), 6736–6740. 39. Yoshida, K.;
Oida, H.; Kobayashi, T.; Maruyama, T.; Tanaka, M.;
Katayama, T.; Yamaguchi, K.; Segi, E.; Tsuboyama, T.;
Matsushita, M.; Ito, K.; Ito, Y.; Sugimoto, Y.; Ushikubi,
F.; Ohuchida, S.; Kondo, K.; Nakamura, T.; Narumiya, S.
Stimulation of bone formation and prevention of bone loss
by prostaglandin E EP4 receptor activation. Proc. Natl.
Acad. Sci. USA 2002, 99 (7), 4580–4585. 40. Shanbhag, A.S.;
Hasselman, C.T.; Rubash, H.E. Inhibition of wear debris
mediated osteolysis in a canine total hip arthroplasty
model. Clin. Orthop. Relat. Res. 1997, 344, 33–43. 41.
Bobyn, J.D.; Jacobs, J.J.; Tanzer, M.; Urban, R.M.;
Aribindi, R.; Sumner, D.R.; Turner, T.M.; Brooks, C.E. The
susceptibility of smooth implant surfaces periimplant
fibrosis and migration of polyethylene wear debris. Clin.
Orthop. Relat. Res. 1995, 311, 21–39. 42. Skurla, C.P.;
James, S.P. Postmortem retrieved canine THR: Femoral and
acetabular component interaction. Biomed. Sci. Instrum.
2004, 40, 255–260. 43. Rahbek, O.; Overgaard, S.; Lind, M.
Sealing effect of hydroxyapatite coating on peri-implant
migration of particles. An experimental study in dogs. J.
Bone Joint Surg. Br. 2001, 83 (3), 441–447. 44. Goodman,
S.; Aspenberg, P.; Song, Y.; Knoblich, G.; Huie, P.;
Regula, D.; Lidgren, L. Tissue ingrowth and differentiation
in the bone-harvest chamber in the presence of
cobalt-chromium-alloy and high-densitypolyethylene
particles. J. Bone Joint Surg. Am. 1995, 77A (7),
1025–1035. 45. Goodman, S.B.; Davidson, J.A.; Song, Y.;
Martial, N.; Fornasier, V.L. Histomorphological reaction of
bone to different concentrations of phagocytosable
particles of high-density polyethylene and Ti-6Al-4V alloy
in vivo. Biomaterials 1996, 17 (20), 1943–1947. 46. Pap,
G.; Machner, A.; Rinnert, T.; Horler, D.; Gay, R.E.;
Schwarzberg, H.; Neumann, W.; Michel, B.A.; Gay, S.; Pap,
T. Development and characteristic of a synovial-like
interface membrane around cemented tibial
hemiarthroplasties in a novel rat model of aseptic
prosthesis loosening. Arthritis Rheum. 2001, 44 (4),
956–963. 47. Howie, D.W.; Vernon-Roberts, B.; Oakeshott,
R.; Manthey, B. A rat model of resorption of bone at the
cement–bone interface in the presence of polyethylene wear
particles. J. Bone Joint Surg. Am. 1988, 70 (2), 257–263.
48. von Knoch, M.; Jewison, D.E.; Sibonga, J.D.; Sprecher,
C.; Morrey, B.F.; Loer, F.; Berry, D.J.; Scully, S.P. The
effectiveness of polyethylene versus titanium particles in
inducing osteolysis in vivo. J. Orthop. Res. 2004, 22 (2),
237–243. 49. Wooley, P.H.; Morren, R.; Andary, J.; Sud, S.;
Yang, S.Y.; Mayton, L.; Markel, D.; Sieving, A.; Nasser, S.
Inflammatory response to orthopaedic biomaterials in the
murine air pouch. Biomaterials 2002, 23 (2), 517–526. 50.
Yang, S.Y.; Ren, W.; Park, Y.; Sieving, A.; Hsu, S.;
Nasser, S.; Wooley, P.H. Diverse cellular and apoptotic
responses to variant shapes of UHMWPE particles in a murine
model of inflammation. Biomaterials 2002, 23 (17),
3535–3543. 51. Ren, W.; Yang, S.Y.; Fang, H.W.; Hsu, S.;
Wooley, P.H. Distinct gene expression of receptor
activation of nuclear factor-kappaB and rank ligand in the
inflammatory response to variant morphologies of UHMWPE
particles. Biomaterials 2003, 24 (26), 4819–4826. 52. Sud,
S.; Yang, S.Y.; Evans, C.H.; Robbins, P.D.; Wooley, P.H.
Effects of cytokine gene therapy on particulate-induced
inflammation in the murine air pouch. Inflammation 2001, 25
(6), 361–372. 53. Epstein, N.J.; Warm, B.A.; Spanogle, J.;
Ma, T.; Bragg, B.; Smith, R.L.; Goodman, S.B. Interleukin-1
modulates periprosthetic tissue formation in an
intramedullary model of particle-induced inflammation. J.
Orthop. Res. 2005, 23 (3), 501–510. 54. Yang, S.Y.; Nasser,
S.; Markel, D.C.; Robbins, P.D.; Wooley, P.H. Human
periprosthetic tissue implanted in severe combined
immunodeficient mice respond to gene transfer of a cytokine
inhibitor. J. Bone Joint Surg. Am. 2005, 87 (5), 1088–1097.
W
Wear Debris from Joint Arthroplasties,
Biological Effects Of

9. Rae, T. A study on the effects of particulate metals of


orthopaedic interest on murine macrophages in vitro. J.
Bone Joint Surg. 1975, 57B, 444–450.

10. Rae, T. The toxicity of metals used in orthopaedic


prostheses. An experimental study using cultured human
synovial fibroblasts. J. Bone Joint Surg. 1981, 63B,
435–440.

11. Rae, T. The biological response to titanium and


titanium-aluminum-vanadium particles. II. Long term animal
studies. Biomaterials 1986, 7, 37–40.

12. Buchert, P.K.; Bradley, K.V.; Mallory, T.M.; Engh,


C.A.; Bobyn, J.D. Excessive metal release due to loosening
and fretting of sintered particles on porous-coated hip
prostheses. J. Bone Joint Surg. 1986, 68A, 606–609.

13. Agins, H.S.; Alcock, N.W.; Bansal, M.; Salvatti, E.A.;


Wilson, P.D.; Pellici, P.M.; Bullough, P.G. Metallic wear
in failed titanium-alloy total hip replacements. J. Bone
Joint Surg. 1988, 70A, 347–356.

14. Howie, D.W.; Vernon-Roberts, B.; Oakeshott, R.;


Manthey, B. A rat model of resorption of bone at the
cemented bone interface in the presence of polyethylene
wear particles. J. Bone Joint Surg. 1988, 70A, 257–263.

15. Doorn, P.F.; Mirra, J.M.; Campbell, P.A.; Amstutz, H.C.


Tissue reaction to metal on metal total hip prostheses.
Clin. Orthop. Relat. Res. 1996, 329 (suppl), S187–S205.

16. Jones, L.C.; Hungerford, D.S. Cement disease. Clin.


Orthop. 1987, 225, 193–206.

17. Goodman, S.B.; Fornasier, V.L.; Kei, J. The effects of


bulk versus particulate ultra-high-molecular-weight
polyethylene on bone. J. Arthroplasty 1988, 3 (suppl),
41–46.

18. Elfick, A.P.; Green, S.M.; Krikler, S.; Unsworth, A. The


nature and dissemination of UHMWPE wear debris retrieved
from periprosthetic tissue of THR. J. Biomed. Mater. Res.
A. 2003, 65 (1), 95–108.

19. Friedman, I.R.; Black, J.; Galante, J.O.; Jacobs, J.J.;


Skinner, H.B. Current concepts in orthopaedic biomaterials
and implant fixation. J. Bone Joint Surg. 1993, 75A,
1086–1109.

20. Learmonth, I.D. Biocompatibility: a biomechanical and


biological concept in total hip replacement. Surgeon 2003,
1 (1), 1–8.

21. Ingham, E.; Fisher, J. Biological reactions to wear


debris in total joint replacement. Proc. Instn. Mech.
Engrs. Part H J. Eng. Med. 2001, 214, 21–37.

22. Margevicius, K.J.; Bauer, T.W.; McMahon, I.T.; Brown,


S.A.; Merritt, K. Isolation and characterisation of debris
from around total joint prostheses. J. Bone Joint Surg.
1994, 76A, 1664–1675.

23. Shanbhag, A.S.; Jacobs, J.J.; Glant, T.T.; Gilbert,


J.L; Black, J.; Galante, J.O. Composition and morphology of
wear debris in failed uncemented total hip replacement. J.
Bone Joint Surg. 1994 , 76B, 60–67.

24. Huddleston, H.D. Femoral lysis after cemented hip


arthroplasty. J. Arthroplasty 1988, 3 (4), 285–297.

25. Anthony, P.P.; Gie, G.A.; Howie, C.R.; Ling, R.S.M.


Localized endosteal bone lysis in relation to the femoral
components of cemented total hip arthroplasties. J. Bone
Joint Surg. 1990, 72B, 971–979.

26. Maloney, W.J.; Jasty, M.; Rosenberg, A.; Harris, W.H.


Bone lysis in well-fixed cemented femoral components. J.
Bone Joint Surg. 1990, 72B, 966–970. 27. Maloney, W.J.;
Peters, P.; Engh, C.A.; Chandler, H. Severe osteolysis of
the pelvis in association with acetabular replacement
without cement. J. Bone Joint Surg. 1993, 75A, 1627–1635.
28. Willert, H.G.; Bertram, H.; Buchhorn, G.H. Osteolysis
in alloarthroplasty of the hip. The role of ultra-high
molecular weight polyethylene wear particles. Clin. Orthop.
1990, 258, 95–107. 29. Lombardi, A.V.; Mallory, T.H.;
Vaughn, B.K.; Drouillard, P. Aseptic loosening in total hip
arthroplasty secondary to osteolysis induced by wear debris
from titanium-alloy modular femoral heads. J. Bone Joint
Surg. 1989, 71A, 1337–1342. 30. Witt, J.D.; Swann, M. Metal
wear and tissue response in failed titanium alloy total hip
replacements. J. Bone Joint Surg. 1991, 73B, 559–563. 31.
Buly, R.L.; Huo, M.H.; Salvati, E.; Brien, W.; Bansal, M.
Titanium wear debris in failed cemented total hip
arthroplasty. An analysis of 71 cases. J. Arthroplasty
1992, 7 (3), 315–323. 32. Salvati, E.A.; Betts, F.; Doty,
S.B. Particulate metallic debris in cemented total hip
arthroplasty. Clin. Orthop. 1993, 293, 160–173. 33. Yoon,
T.R.; Rowe, S.M.; Jung, S.T.; Seon, K.J.; Maloney, W.J.
Osteolysis in association with a total hip arthroplasty
with ceramic bearing surfaces. J. Bone Joint Surg. Am.
1998, 80 (10), 1459–1468. 34. Kim, K.J.; Chiba, J.; Rubash,
H.E. In vivo and in vitro analysis of membranes from hip
prostheses inserted without cement. J. Bone Joint Surg. Am.
1994, 76, 172–180. 35. Jiranek, W.A.; Machado, M.; Jasty,
M. Production of cytokines around loosened cemented
acetabular components: analysis with immunohistochemical
techniques and in situ hybridization. J. Bone Joint Surg.
Am. 1993, 75, 863–879. 36. Lassus, J.; Waris, V.; Xu, J.W.;
Li, T.F.; Hao, J.; Nietosvaara, Y.; Santavirta, S.;
Konttinen, Y.T. Increased interleukin-8 (IL-8) expression
is related to aseptic loosening of total hip replacement.
Arch. Orthop. Trauma Surg. 2000, 120 (5–6), 328–332. 37.
Xu, J.W.; Li, T.F.; Partsch, G.; Ceponis, A.; Santavirta,
S.; Konttinen, Y.T. Interleukin-11 (IL-11) in aseptic
loosening of total hip replacement (THR). Scand. J.
Rheumatol. 1998, 27 (5), 363–367. 38. Goldring, S.R.;
Jasty, M.; Roelke, M.S.; Rourke, C.M.; Bringhurst, F.R.;
Harriw, W.H. Formation of a synoviallike membrane at the
bone-cement interface. Its role in bone resorption and
implant loosening after total hip replacement. Arthritis
Rheum. 1986, 29 (7), 836–842. 39. Nakashima, Y.; Sun, D.H.;
Maloney, W.J.; Goodman, S.B.; Schurman, D.J.; Smith, R.L.
Induction of matrix metalloproteinase expression in human
macrophages by orthopaedic particulate debris in vitro. J.
Bone Joint Surg. Br. 1998, 80, 694–700. 40. Puskas, B.L.;
Menke, N.E.; Huie, P.; Song, Y.; Ecklund, K.; Trindade,
M.C.; Smith, R.L.; Goodman, S.B. Expression of nitric
oxide, peroxynitrite, and apoptosis in loose total hip
replacements. J. Biomed. Mater. Res. A. 2003, 66 (3),
541–549. 41. Mandelin, J.; Li, T.F.; Liljestrom, M.; Kroon,
M.E.; Hanemaaijer, R.; Santavirta, S.; Konttinen, Y.T. Wear
Imbalance of RANKL/RANK/OPG system in interface tissue in
loosening of total hip replacement. J. Bone Joint Surg. Br.
2003, 85 (8), 1196–1201. 42. Xu, J.W.; Konttinen, Y.T.;
Waris, V.; Patiala, H.; Sorsa, T.; Santavirta, S.
Macrophage-colony stimulating factor (M-CSF) is increased
in the synovial-like membrane of the periprosthetic tissues
in the aseptic loosening of total hip replacement (THR).
Clin. Rheumatol. 1997, 16 (3), 243–248. 43. Miyanishi, K.;
Trindade, M.C.; Ma, T.; Goodman, S.B.; Schurman, D.J.;
Smith, R.L. Periprosthetic osteolysis: induction of
vascular endothelial growth factor from human
monocyte/macrophages by orthopaedic biomaterial particles.
J. Bone Miner Res. 2003, 18 (9), 1573–1583. 44. Santerre,
J.P.; Labow, R.S.; Boynton, E.L. The role of the macrophage
in periprosthetic bone loss. Can. J. Surg. 2000, 43 (3),
173–179. 45. Kobayashi, Y.; Mizoguchi, T.; Take, I.;
Kurihara, S.; Udagawa, N.; Takahashi, N. Prostaglandin E2
enhances osteoclastic differentiation of precursor cells
through protein kinase A-dependent phosphorylation of TAK1.
J. Biol. Chem. 2005, 280 (12), 11395–11403. 46. Suzawa, T.;
Miyaura, C.; Inada, M.; Maruyama, T.; Sugimoto, Y.;
Ushikubi, F.; Ichikawa, A.; Narumiya, S.; Suda, T. The role
of prostaglandin E receptor subtypes (EP1, EP2, EP3, and
EP4) in bone resorption: an analysis using specific agonists
for the respective EPs. Endocrinology 2000, 141 (4),
1554–1559. 47. Heersche, J.N.; Aubin, J.E. Regulation of
cellular activity of bone forming cells. In Bone; Hall,
B.K., Ed.; Telford Press: Caldwell, NJ, 1990; 327–349. 48.
Horiki, M.; Nakase, T.; Myoui, A.; Sugano, N.; Nishii, T.;
Tomita, T.; Miyaji, T.; Yoshikawa, H. Localization of RANKL
in osteolytic tissue around a loosened joint prosthesis. J.
Bone Miner Metab. 2004, 22 (4), 346–351. 49. Takei, H.;
Pioletti, D.P.; Kwon, S.Y.; Sung, K.L. Combined effect of
titanium particles and TNF-alpha on the production of IL-6
by osteoblast-like cells. J. Biomed. Mater. Res. 2000, 52,
382–387. 50. Xu, J.W.; Konttinen, Y.T.; Lassus, J.; Natah,
S.; Ceponis, A.; Solovieva, S.; Aspenberg, P.; Santavirta,
S. Tumor necrosis factor-alpha (TNF-alpha) in loosening of
total hip replacement (THR). Clin. Exp. Rheumatol. 1996,
14, 643–648. 51. Neale, S.D.; Sabokbar, A.; Howie, D.W.;
Murray, D.W.; Athanasou, N.A. Macrophage colony-stimulating
factor and interleukin-6 release by periprosthetic cells
stimulates osteoclast formation and bone resorption. J.
Orthop. Res. 1999, 17 (5), 686–694. 52. Al-Saffar, N.;
Khwaja, H.A.; Kadoya, Y.; Revell, P.A. Assessment of the
role of GM-CSF in the cellular transformation and the
development of erosive lesions around orthopaedic implants.
Am. J. Clin. Pathol. 1996, 105, 628–639. 53. Waris, V.; Xu,
J.W.; Nordsletten, L.; Sorsa, T.; Santavirta, S.;
Konttinen, Y.T. Basic fibroblast growth factor (bFGF) in the
synovial-like membrane around loose total hip prostheses.
Scand. J. Rheumatol. 1996, 25, 257–262. 54. Konttinen,
Y.T.; Waris, V.; Xu, J.W.; Jiranek, W.A.; Sorsa, T.;
Virtanen, I.; Santavirta, S. Transforming growth
factor-beta 1 and 2 in the synovial-like interface membrane
between implant and bone in loosening of total hip
arthroplasty. J. Rheumatol. 1997, 24 , 694–701. 55.
Al-Saffar, N.; Mah, J.T.L; Kadoya, Y.; Revell, P.A.
Neovascularisation and the induction of cell adhesion
molecules in response to degradation products from
orthopaedic implants. Ann. Rheum. Dis. 1995, 54, 201–208.
56. Perry, M.J.; Mortuza, F.Y.; Ponsford, F.M.; Elson,
C.J.; Atkins, R.M. Analysis of cell types and mediator
production from tissues around loosening joint implants.
Br. J. Rheumatol. 1995, 34 (12), 1127–1134. 57. Tallroth,
K.; Eskola, A.; Santavirta, S.; Konttinen, Y.T.; Lindholm,
T.S. Aggressive granulomatous lesions after hip
arthroplasty. J. Bone Joint Surg. 1989, 71B, 571–575. 58.
Santavirta, S.; Kontinnen, Y.T.; Hoikka, V.; Eskola, A.
Immunopathological response to loose cementless acetabular
components. J. Bone Joint Surg. 1991, 73B, 38–42. 59.
Farber, A.; Chin, R.; Song, Y.; Huie, P.; Goodman, S.
Chronic antigen-specific immune-system activation may
potentially be involved in the loosening of cemented
acetabular components. J. Biomed. Mater. Res. 2001, 55 (3),
433–441. 60. Li, T.F.; Santavirta, S.; Waris, V.; Lassus,
J.; Lindroos, L.; Xu, J.W.; Virtanen, I.; Konttinen, Y.T.
No lymphokines in T-cells around loosened hip prostheses.
Acta Orthop. Scand. 2001, 72 (3), 241–247. 61. Sandhu, J.;
Waddell, J.; Henry, M.; Boynton, E.L. The role of T-cells
in polyethylene induced inflammation. J. Rheumatol. 1998,
25, 1794–1799. 62. Nakashima, Y.; Sun, D.H.; Trindade,
M.C.D.; Maloney, W.J.; Goodman, S.B.; Schurman, D.J.;
Smith, R.L. Titanium alloy particle activation of signaling
pathways for tumor necrosis factor-alpha and interleukin-6
expression in human macrophages in vitro. J. Bone Joint
Surg. 1999, 81A, 603–615. 63. Vermes, C.; Chandrasekaran,
R.; Jacobs, J.J.; Galante, J.O.; Roebuck, K.A.; Glant, T.T.
The effects of particulate wear debris, cytokines, and
growth factors on the functions of MG-63 osteoblasts. J.
Bone Joint Surg. 2001, 83A, 201–211. 64. Vermes, C.; Glant,
T.T.; Hallab, N.J.; Fritz, E.A.; Roebuck, K.A.; Jacobs,
J.J. The potential role of the osteoblast in the
development of periprosthetic osteolysis. Review of in
vitro osteoblast responses to wear debris, corrosion
products, and cytokines and growth factors. J. Arthroplasty
2001, 16 (suppl. 1), 95–100. 65. Trindade, M.C.; Lind, M.;
Sun, D.; Schurman, D.J.; Goodman, S.B.; Smith, R.L. In
vitro reaction to orthopaedic biomaterials by macrophages
and lymphocytes isolated from patients undergoing revision
surgery. Biomaterials 2001, 22 (3), 253–259. 66. Goldring,
S.R.; Schiller, A.L.; Roilke, M.; Rourke, C.M.; O’Neil,
D.A.; Harris, W.H. The synovial-like membrane at the
bone-cement interface in loose total hip replacement and
its proposed role in bone lysis. J. Bone Joint Surg. Am.
1983, 65 (5), 575–584. 67. Haynes, D.R.; Rogers, S.D.; Hay,
S.; Pearcy, M.J.; Howie, D.W. The differences in toxicity
and release of bone-resorbing mediators induced by titanium
and W Of cobalt-chromium-alloy wear particles. J. Bone
Joint Surg Am. 1993, 75 (6), 825–834.

68. Horowitz, S.M.; Doty, S.B.; Lane, J.M.; Burstein, A.H.


Studies of the mechanism by which the mechanical failure of
polymethylmethacrylate leads to bone resorption. J. Bone.
Joint. Surg. Am. 1993, 75 (6), 802–813.
69. Harada, Y.; Wang, J.; Doppalapudi, V.A.; Willis, A.A.;
Jasty, M.; Harris, W.H. Differential effects of different
forms of hydroxyapatite and hydroxyapatite/tricalcium
phosphate particulates on human monocyte/macrophages in
vitro. J. Biomed. Mater. Res. 1996, 31, 19–26.

70. Wang, X.Y.; Li, S.; Salovey, R. Processing of UHMWPE.


J. Appl. Polym. Sci. 1988, 35, 2165–2171.

71. Labow, R.S.; Meek, E.; Santerre, J.P. Differential


synthesis of cholesterol esterase by monocyte-derived
macrophages cultured on poly(ether or ester)-based
poly(urethane)s. J. Biomed. Mater. Res. 1998, 39 (3),
469–477.

72. Sun, D.H.; Trindade, M.C.; Nakashima, Y.; Maloney,


W.J.; Goodman, S.B.; Schurman, D.J.; Smith, R.L. Human
serum opsonization of orthopedic biomaterial particles:
protein-binding and monocyte/macrophage activation in
vitro. J. Biomed. Mater. Res. A 2003, 65 (2), 290–298.

73. Czajkowska, B.; Blazewica, M. Phagocytosis of


chemically modified carbon materials. Biomaterials 1997, 18,
69–74.

74. Hunt, J.A.; Flanagan, B.F.; McLaughlin, P.J.;


Strickland, I.; Williams, D.F. Effect of biomaterial
surface charge on the inflammatory response: evaluation of
cellular infiltration and TNF alpha production. J. Biomed.
Mater. Res. 1996, 31, 139–144.

75. Bi, Y.; Seabold, J.M.; Kaar, S.G.; Ragab, A.A.;


Goldberg, V.M.; Anderson, J.M.; Greenfield, E.M. Adherent
endotoxin on orthopedic wear particles stimulates cytokine
production and osteoclast differentiation. J. Bone Miner
Res. 2001, 16 (11), 2082–2091.

76. Clohisy, J.C.; Teitelbaum, S.; Chen, S.; Erdmann, J.M.;


Abu-Amer, Y. Tumor necrosis factor-alpha mediates
polymethylmethacrylate particle-induced NF-kappaB
activation in osteoclast precursor cells. J. Orthop. Res.
2002, 20 (2), 174–181.

77. Kadoya, Y.; Revell, P.A.; al-Safar, N.; Kobayashi, A.;


Scott, G.; Freeman, M.A. Bone formation and resorption in
failed total joint arthroplasties: histomorphometric
analysis with histochemical and immunohistochemical
technique. J. Orthop. Res. 1996, 14, 473–482.
78. Kadoya, Y.; Kobayashi, A.; Ohashi, H. Wear and
osteolysis in total joint replacements. Acta Orthop. Scand.
1998, 69 (suppl. 278), 1–14.

79. Kadoya, Y.; al-Saffar, N.; Kobayashi, A.; Revell, P.A.


The expression of osteoclast markers on foreign body giant
cells. Bone Miner 1994, 27 (2), 85–96.

80. Quinn, J.; Joyner, C.; Triffitt, I.T.; Athanasou, N.A.


Polymethylmethacrylate-induced inflammatory macrophages
resorb bone. J. Bone Joint Surg. Br. 1992, 74, 652–658.

81. Lam, J.; Takeshita, S.; Barker, J.E.; Kanagawa, O.;


Ross, F.P.; Teitelbaum, S.L. TNF-alpha induces
osteoclastogenesis by direct stimulation of macrophages
exposed to permissive levels of RANK ligand. J. Clin.
Invest. 2000, 106, 1481–1488. 82. Ma, T.; Miyanishi, K.;
Suen, A.; Epstein, N.J.; Tomita, T.; Smith, R.L.; Goodman,
S.B. Human interleukin1-induced murine osteoclastogenesis
is dependent on RANKL, but independent of TNF-alpha.
Cytokine 2004, 26 (3), 138–144. 83. Suda, K.; Udagawa, N.;
Sato, N.; Takami, M.; Itoh, K.; Woo, J.T.; Takahashi, N.;
Nagai, K. Suppression of osteoprotegerin expression by
prostaglandin E2 is crucially involved in
lipopolysaccharideinduced osteoclast formation. J. Immunol.
2004, 172 (4), 2504–2510. 84. Baumann, B.; Rader, C.P.;
Seufert, J.; Noth, U.; Rolf, O.; Eulert, J.; Jakob, F.
Effects of polyethylene and TiAlV wear particles on
expression of RANK, RANKL and OPG mRNA. Acta Orthop. Scand.
2004, 75 (3), 295–302. 85. Kim, K.J.; Kotake, S.; Udagawa,
N.; Ida, H.; Ishii, M.; Takei, I.; Kubo, T.; Takagi, M.
Osteoprotegerin inhibits in vitro mouse osteoclast
formation induced by joint fluid from failed total hip
arthroplasty. J. Biomed. Mater. Res. 2001, 58 (4), 393–400.
86. Jimi, E.; Nakamura, K.; Duong, L.T.; Ikebe, T.;
Takahashi, N.; Rodan, G.A. Interleukin-1 induces
multinucleation and bone resorbing activity of osteoclasts
in the absence of osteoblasts/stromal cells. Exp. Cell Res.
1999, 247, 84–93. 87. Sacomen, D.; Smith, R.L.; Song, Y.;
Fornasier, V.; Goodman, S.B. Effects of polyethylene
particles on tissue surrounding knee arthroplasties in
rabbits. J. Biomed. Mater. Res. 1998, 43, 123–130. 88.
Lohmann, C.H.; Dean, D.D.; Bonewald, L.F.; Schwartz, Z.;
Boyan, B.D. Nitric oxide and prostaglandin E2 production in
response to ultra-high molecular weight polyethylene
particles depends on osteoblast maturation state. J. Bone
Joint Surg. 2002, 84A, 411–419. 89. Kwon, S.Y.; Lin, T.;
Takei, H.; Ma, Q.; Wood, D.J.; O’Connor, D. Alterations in
the adhesion behavior of osteoblasts by titanium particle
loading: inhibition of cell function and gene expression.
Biorheology 2001, 38, 161–183. 90. Kwon, S.Y.; Takei, H.;
Pioletti, D.P.; Lin, T.; Ma, Q.J.; Akeson, W.H. Titanium
particles inhibit osteoblast adhesion to fibronectin-coated
substrates. J. Orthop. Res. 2001, 18, 203–211. 91.
Pioletti, D.P.; Takei, H.; Kwon, S.Y.; Wood, D.; Sung, K.L.
The cytotoxic effect of titanium particles phagocytosed by
osteoblasts. J. Biomed. Mater. Res. 1999, 46, 399–407. 92.
Engh, C.A., Jr.; Moore, K.D.; Vinh, T.N.; Engh, G.A.
Titanium prosthetic wear debris in remote bone marrow. A
report of two cases. J. Bone Joint Surg. Am. 1997, 79 (11),
1721–1725. 93. Wang, M.L.; Tuli, R.; Manner, P.A.; Sharkey,
P.F.; Hall, D.J.; Tuan, R.S. Direct and indirect induction
of apoptosis in human mesenchymal stem cells in response to
titanium particles. J. Orthop. Res. 2003, 21 (4), 697–707.
94. Wang, M.L.; Sharkey, P.F.; Tuan, R.S. Particle
bioreactivity and wear-mediated osteolysis. J. Arthroplasty
2004, 19 (8), 1028–1038. Wear
Wheeled Mobility: Wheelchairs and
Personal Transportation

24. Fass, M.V.; Cooper, R.A.; Fitzgerald, S.G.; Schmeler,


M.; Boninger, M.L.; Algood, S.D.; Ammer, W.A.; Rentschler,
A.J. Durability, value, and reliability of selected
electric powered wheelchairs. Arch. Phys. Med. Rehabil.
2004, 85 (5), 805.

25. Fitzgerald, S.G.; Cooper, R.A.; Boninger, M.L.;


Rentschler, A.J. Comparison of fatigue life for 3 types of
manual wheelchairs. Arch. Phys. Med. Rehabil. 1982, 82
(10), 1484.

26. Pearlman, J.L.; Cooper, R.A.; Karnawat, J.; Cooper, R.;


Boninger, M.L. Evaluation of the safety and durability of
low-cost electric powered wheelchairs. Arch. Phys. Med.
Rehabil. 2005, 86 (12), 2361–2370.

27. Rentschler, A.J.; Cooper, R.A. A comparison of the


dynamic and static stability of power wheelchairs versus
scooters. Proceedings of the First Joint BMES/EMBS
Conference, 1999. 28. Rentschler, A.J.; Cooper, R.A.;
Fitzgerald, S.G.; Boninger, M.L.; Song-Feng, G.; Ammer, W.;
Vitek, M.; Algood, D. Evaluation of selected
electric-powered wheelchairs using the ANSI/RESNA
standards. Arch. Phys. Med. Rehabil. 2004, 85 (4), 611–619.
29. Centers for medicare and medicaid services. In Part B
Physician/Supplier Nat’l Data, CY 2003 Top 200 Level II
Healthcare Common Procedure Coding System; , 2004. 30.
India, G.O. Scheme of Assistance to Disabled Persons for
Purchasing/Fitting of Aids/Appliances (ADIP Scheme);
Ministry of Social Justice and Empowerment: New Delhi. 31.
Guan, T. Information Regarding Chinese Reimbursement for
Assistive Technology; Pearlman, J.L., Ed.; 2005, Dalian,
China.
Xenografts

14. Cooper, D.K.C.; Human, P.A.; Lexer, G.; Rose, A.G.;


Rees, J.; Keraan, M.; Du Toit, E. Effects of cyclosporine
and antibody adsorption on pig cardiac xenograft survival
in the baboon. J. Heart Transplant. 1988, 7, 238–246.

15. Leventhal, J.R.; Dalmasso, A.P.; Cromwell, J.W.; Platt,


J.L.; Manivel, C.J.; Bolman, R.M.; Matas, A.J. Prolongation
of cardiac xenograft survival by depletion of complement.
Transplantation 1993, 55, 857–866.

16. Byrne, G.W.; McCurry, K.R.; Martin, M.J.; McClellan,


S.M.; Platt, J.L.; Logan, J.S. Transgenic pigs expressing
human CD59 and decay-accelerating factor produce an
intrinsic barrier to complement-mediated damage.
Transplantation 1997, 63, 149–155.

17. Sharma, A.; Okabe, J.F.; Birch, P.; Platt, J.L.; Logan,
J.S. Reduction in the level of Gal (a1,3) Gal in transgenic
mice and pigs by the expression of an a(1,2)
fucosyltransferase. Proc. Natl. Acad. Sci. U.S.A. 1996, 93,
7190–7195.

18. Lai, L.; Kolber-Simonds, D.; Park, K.W.; Cheong, H.T.;


Greenstein, J.L.; Im, G.S.; Samuel, M.; Bonk, A.; Rieke,
A.; Day, B.N.; Murphy, C.N.; Carter, D.B.; Hawley, R.J.;
Prather, R.S. Production of a-1,3-galactosyltransferase
knockout pigs by nuclear transfer cloning. Science 2002,
295, 1089–1092.

19. Phelps, C.J.; Koike, C.; Vaught, T.D.; Boone, J.;


Wells, K.D.; Chen, S.H.; Ball, S.; Specht, S.M.; Polejaeva,
I.A.; Monahan, J.A.; Jobst, P.M.; Sharma, S.B.; Lamborn,
A.E.; Garst, A.S.; Moore, M.; Demetris, A.J.; Rudert, W.A.;
Bottino, R.; Bertera, S.; Trucco, M.; Starzl, T.E.; Dai,
Y.; Ayares, D.L. Production of alpha
1,3-galactosyltransferase-deficient pigs. Science 2003, 299,
411–414.

20. Sharma, A.; Naziruddin, B.; Cui, C.; Martin, M.J.; Xu,
H.; Wan, H.; Lei, Y.; Harrison, C.; Yin, J.; Okabe, J.;
Mathews, C.; Stark, A.; Adams, C.S.; Houtz, J.; Wiseman,
B.S.; Byrne, G.W.; Logan, J.S. Pig cells that lack the gene
for alpha1–3 galactosyltransferase express low levels of
the gal antigen. Transplantation 2003, 75, 430–436.

21. Holzknecht, Z.E.; Platt, J.L. Identification of porcine


endothelial cell membrane antigens recognized by human
xenoreactive antibodies. J. Immunol. 1995, 154, 4565–4575.
22. Hattori, R.; Hamilton, K.K.; McEver, R.P.; Sims, P.J.
Complement proteins C5b-9 induce secretion of high
molecular weight multimers of endothelial von Willebrand
factor and translocation of granule membrane protein
GMP-140 to the cell surface. J. Biol. Chem. 1989, 264,
9053–9060.

23. Inverardi, L.; Samaja, M.; Motterlini, R.; Mangili, F.;


Bender, J.R.; Pardi, R. Early recognition of a discordant
xenogeneic organ by human circulating lymphocytes. J.
Immunol. 1992, 149, 1416–1423.

24. Gaca, J.G.; Lee, W.; Aksoy, O.; Braedehoeft, S.J.;


Gonzalez-Stawinski, G.V.; Parker, W.; Davis, R.D. Evidence
for polyreactive xenoreactive antibodies in the repertoire
of human anti-swine antibodies: The ‘next’ humoral barrier
to xenotransplantation. Transpl. Immunol. 2001, 9, 19–27.
25. Malyguine, A.M.; Saadi, S.; Holzknecht, R.A.; Patte,
C.R.; Sud, N.; Platt, J.L.; Dawson, J.R. Induction of
procoagulant function in porcine endothelial cells by human
NK cells. J. Immunol. 1997, 159, 4659– 4664. 26. Chopek,
M.W.; Simmons, R.L.; Platt, J.L. ABO-incompatible renal
transplantation: Initial immunopathologic evaluation.
Transplant. Proc. 1987, 19, 4553–4557. 27. Lin, Y.;
Goebles, J.; Xia, G.; Ji, P.; Vandeputte, M.; Waer, M.
Induction of specific transplantation tolerance across
xenogeneic barriers in the T-independent immune
compartment. Nat. Med. 1998, 4, 173–180. 28. Bach, F.H.;
Ferran, C.; Hechenleitner, P.; Mark, W.; Koyamada, N.;
Miyatake, T.; Winkler, H.; Badrichani, A.; Cardinas, D.;
Hancock, W.H. Accommodation of vascularized xenografts:
Expression of ‘‘protective genes’’ by donor endothelial
cells in a host Th2 cytokine environment. Nat. Med. 1997,
3, 196–204. 29. Murray, A.G.; Khodadoust, M.M.; Pober,
J.S.; Bothwell, A.L.M. Porcine aortic endothelial cells
activate human T cells: Direct presentation of MHC antigens
and costimulation by ligands for human CD2 and CD28.
Immunity 1994, 1, 57–63. 30. Gunsalus, J.R.; Brady, D.A.;
Coulter, S.M.; Gray, B.M.; Edge, A.S.B. Reduction of serum
cholesterol in Watanabe rabbits by xenogeneic
hepatocellular transplantation. Nat. Med. 1997, 3, 48–53.
31. Deacon, T.; Schumacher, J.; Dinsmore, J.; Thomas, C.;
Palmer, P.; Kott, S.; Edge, A.; Penney, D.; Kassissieh, S.;
Dempsey, P.; Isacson, O. Histological evidence of fetal pig
neural cell survival after transplantation into a patient
with Parkinson’s disease. Nat. Med. 1997, 3, 350–353. 32.
Gritsch, H.A.; Glaser, R.M.; Emery, D.W.; Lee, L.A.; Smith,
C.V.; Sablinski, T.; Arn, J.S.; Sachs, D.H.; Sykes, M. The
importance of nonimmune factors in reconstitution by
discordant xenogeneic hematopoietic cells. Transplantation
1994, 57, 906–917. 33. Aebischer, P.; Schluep, M.; Deglon,
N.; Joseph, J.M.; Hirt, L.; Heyd, B.; Goddard, M.; Hammang,
J.P.; Zurn, A.D.; Kato, A.C.; Regli, F.; Baetge, E.E.
Intrathecal delivery of CNTF using encapsulated genetically
modified xenogeneic cells in amyotrophic lateral sclerosis
patients. Nat. Med. 1996, 2, 696–699. 34. Patience, C.;
Takeuchi, Y.; Weiss, R.A. Infection of human cells by an
endogenous retrovirus of pigs. Nat. Med. 1997, 3, 282–286.
35. Van der Laan, L.J.W.; Lockey, C.; Griffeth, B.C.;
Frasier, F.S.; Wilson, C.A.; Onions, D.E.; Hering, B.J.;
Long, Z.; Otto, E.; Torbett, B.E.; Salomon, D.R. Infection
by porcine endogenous retrovirus after islet
xenotransplantation in SCID mice. Nature 2000, 407,
501–504. 36. Paradis, K.; Langford, G.; Long, Z.; Heneine,
W.; Sandstrom, P.; Switzer, W.M.; Chapman, L.E.; Lockey,
C.; Onions, D. Search for cross-species transmission of
porcine endogenous retrovirus in patients treated with
living pig tissue. Science 1999, 285, 1236–1241. 37. Ryan,
K.J.; Brady, J.V.; Cooke, R.E. The Belmont Report; U.S.
Department of Health, Education and Welfare, 1979. 38.
Institute of Medicine. Xenotransplantation: Science,
Ethics, and Public Policy; National Academy Press:
Washington, DC, 1996. 39. Cascalho, M.; Platt, J.L.
Xenotransplantation and other means of organ replacement.
Nat. Rev., Immunol. 2001, 1, 154–160. 40. Platt, J.L. New
directions for organ transplantation. Nature 1998, 392
(Suppl.), 11–17. X
Zirconia Ceramics

41. Lathabai, S.; Mai, Y.W.; Lawn, B.R. Cyclic fatigue


behavior of an alumina ceramic with crack-resistance
curves. J. Am. Ceram. Soc. 1989, 72, 1760–1763.

42. Qasim, T.; Ford, C.; Bush, M.B.; Hu, X.; Malament,
K.A.; Lawn, B.R. Margin failures in brittle dome
structures: Relevance to failure of dental crowns. J.
Biomed. Mater. Res. B Appl. Biomater. January 2006, 80 (1),
78–85.

43. Deng, Y.; Lawn, B.R.; Lloyd, I.K. Characterization of


damage modes in dental ceramic bilayer structures. J.
Biomed. Mater. Res. 2002, 63 (2), 137–145.

44. Suliman, A.H.; Swift, E.J., Jr.; Perdigao, J. Effects


of surface treatment and bonding agents on bond strength of
composite resin to porcelain. J. Prosthet. Dent. 1993, 70
(2), 118–120. 45. Zhang, Y.; Lawn, B. Long-term strength of
ceramics for biomedical applications. J. Biomed. Mater.
Res. B Appl. Biomater. 2004, 69 (2), 166–172. 46. Zhang,
Y.; Lawn, B.R.; Rekow, E.D.; Thompson, V.P. Effect of
sandblasting on the long-term performance of dental
ceramics. J. Biomed. Mater. Res. B Appl. Biomater. 2004, 71
(2), 381–386. 47. Blatz, M.B. Cementation of
zirconium-oxide ceramic restorations. Pract. Proced.
Aesthet. Dent. 2004, 16 (1), 14. This page intentionally
left blank
3/6/24, 1:11 PM SR University

Student Result
Student Basic Information

Enrollment Number 2305C40040

Student Name PARVATHAM SATHISH

Father Name

Degree PHD-PHD-ECE(ECE)

S.No Year- Course Code Course Name Total Grades Credits


Sem

1 1-1 23EC400PC713 BIO MEDICAL INSTRUMENTATION 80 B 4

2 1-1 23EC400PC715 Advanced Technologies in Electronics and 93 A 4


Communication Engineering

3 1-1 23PD301PC101 Research Methodology and Publication Ethics 81 B 4

SGPA : 8.67

https://sraap.in/student/student_sem_wise_result.php 1/1
S No Degree Branch Spl Sec Year Sem Emp Id HallTicket No Name
1 PHD ECE ECE A 1 1 2519 2305C40040 PARVATHAM SATHISH
Total No. of Students : 1
Award List Submitted on : 12-02-2024

Academic Year : 2023-24, Bra


23EC400PC713-BIOMEDICAL
Mid Term Examination(20) Assignment or Certification(20) Assignment or Writing Research paper(20)
18 20 18

I PHD I Sem Mid Marks Award List


Academic Year : 2023-24, Branch:ECE, Yr-Sem : 1-1
23EC400PC713-BIOMEDICAL INSTRUMENTATION
Re Mid Term Examination(20) CIE (60) ETE (40)
56 24
Course Coordinator

Signature of HOD with Date

You might also like