Biopharmaceutics and Pharmacokinetics - A Treatise - Brahmankar, Jaiswal - Pharma Dost

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 206

,•


...

D.M. BRAHMANKAR
SUNIL B. JAISWAL

10

·,

D. M. BRAHMANKAR
M.Sc., Ph.D.
&
SUNIL B. ]AISWAL
M.Pharm., Ph.D .

... .
4. .. ~
••. .,..._

;
..
.
"'• : ·~ .,... .
...
..
.
~

... •"'"
...,... . ..,.
!-:.. · •~ '""'

. .'
... #' . .. •
...
.

'

V ALLABH PRAKASH AN
description of methods usually employed to enhance the bioavailability of
a drug fron1 its formulation has been included at the end of chapter 12.
In addition to ~overing various aspects of design of dosage regimens and
application of pharmacokinetic principles in clinical situations, the text Contents
contains a final chapter on Controlled Release Medication to familiarize
the students with the principles .involved in the , design of innovative •••
Preface Ill
formulations.
We feel that a concept is more readily appreciated and understood 1. Introduction 1-4
when illustrated by a simple figure or a table. In order to exemplify the 2. Absorption of Drug~ 5-75
text, illustrations have been used liberally throughout the book. At the Gastromtestinal Absorption of Drugs 6
end of every chapter, a set of questions including numericals have been Cell Membrane Structure and Physiology 6
provided which are strategically designed to complement the text of each Mechanisms of drug absorptic!l 7
chapter and test the student's grasp of the text and their ability to analyze Factors influencing drug absorption and bioavailability 16
- and solve problems. If this book can help to provide the reader with a Physicochemical factors affecting drug absorption 19
clear understanding of the basic concepts of biopharmaceutics and phattna- Theories of drug dissolution 20

cokinetics, then we would consider our object as fulfilled. Factors affecting drug dissolution and dissolution rate 25
We acknowledge the contribution of hundreds of scientists who are pH-partition hypothesis 32
dedicated to improvement of drug therapy. We are also indebted to the ·Dosage fonn factors affecting drug absorption 39
authors of the various books and articles mentioned in bibliography which Patient related factors affecting. drug absorption 51
became a major source of information for writing this text. Special Absorption of Drugs from Non Per Os Extravascular Routes 63
thanks are due to Professor W. A. Ritschel for his timely help and Buccal/sublingual administration 64
inspiration. We are also grateful to all those who helped in different ways Rectal administration 65
during the preparation of this book especially for their support, encourage- Topical administration 66
ment and constructive criticism, particularly to our students for their Intramuscular administration 67
suggestions. Further, we are appreciative of the publishers who graciously Subcutaneous. administration 68
volunteered to make the publication of this book a reality. Pulmonary & Intranasal administration 69
Intraocular & Vaginal administration 70
Lastly, the authors woulq very much appreciate receiving critical sug-
gestions for improvement and refinement of the text in future. . 3. Distribution of Drugs 76-90
Tissue Penneability of Drugs 76
Physicochemical properties of the drug 77
Nagpur D. M. Brahmankar
Physiological barriers ~o distribution of drugs 79
May 15, 1995 Sunil B. Jaiswal
Organ/tissue Size and Perfusion Rate 83
Binding of Drugs and Perfusion Rate 85
Miscellaneous Factors Affecting Drug Distribution 85
, Volume of Distribution 86
4. Protein Binding of Drugs 91-110
Binding of Drugs to Blood Components 92


I
Plasma protein-drug binding 92
Binding of drug to blood cells 95
Tissue Binding of Drugs (Tissue Localization of Drugs) 96
(iv) (v)
Factors Affecting Protein- Drug Binding 97
7. Excretion of Drugs 178-203
Drug related factors 98
Renal Excretion of Drugs \ 178
Protein/tissue related factors 98

\
' Glomerular filtration 179
Drug interactions 99
Active' tUbular secretion \ 180
Patient related factors 101 Tubular reabsorption 181
Significance of Protein/Tissue Binding of Drugs 103 Concept of clearance 185
Kinetics of Protein Drug Binding 106 18.7
Factors affecting renal excretion
5. Biotransformation of Drugs 111-158 Dose adjustment in renal failure 192
Drug Metabolizing Organs & Enzymes. 113 Dialysis.and ·hemoperfusion 192
Chemical Pathways of Drug Biotransforn1ation 114 Nonrenal Routes of Drug Excretion 194
.Phase 1' Reactions 117 Biliary excretion of drugs-enterohepatic cycling 195
Oxidative reactions 117 Pulmonary excretion 198
Reductive reactions 130 Sal~vary excretion 199
Hydro lytic re~ctions 134 ~ammary & Skin excretion 200
'
Phase II Reactions 138 Gastrointestinal ·&. Genital excretion 201
r

Conjugation with glucuronic acid 139 204-211


· 8. Pharmacokinetic Drug Interactions
Conjugation with sulfate moieties . 141 I

Mechanisms of Drug Interactions 204


Conjugation with alpha-amino acids 142 206
Interactions Affecting Absorption & Distribution of Drugs
Conjugation with glutathione· & mercapturic acid formation 143 207
Interactions Affecting Metabolism & Excretion of Drugs
Acetylation 144
Methylation 145 .9. Pharma(fokinetics: Basic Considerations 212-229
Miscellaneous conjugation reactions 146 Plasma Drug Concentration-Time profile 213
Factors Affecting Biotransfotmation of Drugs 147 Rates, Rate Constants and Orders of Reaction 215
~

Physicochemical properties of the drug 147 Zero-Order Kinetics 216


Chemical factors 148 First-Order Kinetics 217
Biological factors 150 Mixed-Order Kinetics 219
Bioactivation and Tissue Toxicity 154 Pharmacokinetic Models 220
Compartment models 221
6. Prodrugs 159-177
Noncompartmental analysis 224
Applications of Prodrug Design 162
Physiologic models 226
Improvement of taste 162
Change of physical fortn of the drug 163 10. Compartment Modeling 230-272

Reduction of GI irritation 163 One-Compartrnent Open Model 230


Reduction of pain on injection 164 Intravenous bolus administration 231
Enhancement of drug solubility and dissolution rate 164 Intravenous infusion 239
Enhancement of chemical. stability of drug 165 Extravascular administration 244
Enfiancemenr,·o f bioavailability (lipophilicity) 166 Urinary Excretion Data 253
P~~vention of presystemic metabolism 167 Multicompartment Mo<;lels 259
Prl> longation of d.uration of actiotf · 167 Two-Compartinent Open Model 260
Reduction of toxicity . 168 \
Intravenous bolus administration 260
Si;e~specific drug delivery (drug targeting) 169 Intravenous infusion 265
r
Limit-ations of Prodrug Design 176 Extravascular administration ~ · 266
.~

(vi) •
(vii)
'


11. Nonlinear Pharmacokinetics
Causes of Nonlinearity 273-281
Michaelis-Menten Equation 274
Esiimation of Km and y max 215
277
12. Bioavailability -end Bioequivalence
Objectives & Considerations in BioaVailability Studi
Measunnent of Bioavailability es
282-305
283 1 I ' '

Plasma level-time studies 285 •

Urinary excretion studies 286.


288
Introduction
A~ute phannacologic & Therapeutic response
290
In v~tro J?ru~ DiSsolution Testing Models
In Vttro-m vtvo Correlation 290
- 292 Drugs, whether obtained from plant, animal _ or mineral soul'ces or
Bioequiva1ence Studies
2-94 synthesized chemically, are rarely administered in their pure chemical
Methods for Enhancement -of Bioavailability form. Often, they are combined with ~a number of inert substances
"296
13. Appli~~ations of Pbarmacokinetic. Principles (excipients/adjuvants) and transfonned into a convenient dosage fonn that
Design of Dosage Regimens · 306-324 can be administered by a ,suitable route. Earlier, it \Vas believed . that the .
Individualization 307 therapeutic response to a drug is an attribute of its intrinsic phannacologic
.
Monitoring Drug Therapy 315 activity. But tqday, it is very much understood that the dose-response
320 relationship obtained after drug administration by different routes for
14. Drug Conc~ntration and Pharmacologic Response
325-33.4 example. oral and parenteral, are not the same. Variations are also
Therapeutic Concentration Range observed when the same drug is administered as different qosage fonns or
326
Concentrati~n-Response Relationships similar__ dosage forms produced by different manufacturers, which in tum
327
Onse~duratiOn of Effect-Concen~ation Relationship 330 depend upon the physicochemical properties of the drug, the excipients
Intenstty of Effect-Concentration Relationship present in the dosage form, the method of for&nulation and the manner of
332
15. Controlled Release Medication administration. A new and separate discipline called biopharmaceutics
335-371 has therefore been developed to account for all such factors that influence
Design of ~on~oll~d Drug Delivery Systems
337 the therapeutic effectiveness of a drug.
: Phannacokmetic Principles in the Design and Fabrication
of Controlled Drug Delivery Systems Biopharmaceutics is defined as the study of factors influencing the
34~
Drug Release Patterns of Controlled Delivery Dosage Fonns rate and amount of drug that reaches the systemic circulation (Jnd the use
Oral Controlled Release Systems 343 -'

of this information to optimize the therapeutic efficacy of the drug prod-


Parenteral Con~olled Release Systems
347 ucts. The process of m~vement of drug from its site of administra~ion to · ·- _·
Transdettnal Drug Delivery Systems -357 the systemic circulalion is called as absorption. The concentr~tion of ·
Ophthalmic Drug Delivery Systems
365 drug in plasma and hence the onset of action, and . the intensity and -
368
I~trava~in~l. and Intrauterine Drug Delivery Systems . 369
duration of response depend upon the bioavailability of drug from it~
B~oavadabthty Testing of Controlled Release Formulations 371
dosage form. Bioavailability .-is defined as the rate and extent (amount)
BIBLIOGRAPHY of drug absorption. Any alteration in the drug's bioavailability is re-
376-378 flected in its pharrnacologic effects. Other processes that play a role in
INDEX the therapeutic activity of a drug are distribution and elimination. To-
379-400 gether, .they are known as drug disposition. The . movement of drwg
\ . .
r
between one compartment and the ' other· (generally blood and the· ex-
travascular tissues) is referred to as drug distribution. Since the site of
action is usually located in the extravascular tissues, the onset, intensity
(viii)

I
2 BIOPHARMACEUTICS AND PHARMACOKINETICS
INTRODUCTION J
. ~

- arid sometimes duration of action depend upon the distribution behavior of designed to deliver the active principle at an optimal rate and amount,
the drug. The magnitude (intensity) and the duration of action depend
depending upon the patient's needs. A knowledge of the factors affecting
- largely upon the effective concentration and the time period for which this
the bioavailabllity of drug helps in designing such an optimum formula-
concentration is maintained at the site of action which in tum depend
tion and saves many drugs that may be discarded as useless. On the
upon the elimination processes. Elimination is defined as the process
that tends to remove the drug from the body and terminate its action. I' .
r
Elimination o<xur-s by two processes biotransformation (metabolism), Drug + Excipients
' which usually inactivates the drug, and excretion which is responsible for
the exit of drug/metabolites from the body. PHARMACEUTICS FORMULATION

In order' to administer drugs optimally, knowle~ge is needed not only


of the mechanisms of drug absorption, distribution, metabolism and excre- Drug not
Drug in Dosage Form Released
tion (ADM£) but also of the rate (kinetics) at which they occur i.e.
pharmacokinetics. Pharmacokinetics is defined as the study_ of time en
course of drug ADM.&_ af!d'' th~ir relatio_nship_ with its_ ther:.qf!f}.Hti' and toxiG -u -.- --- ....... --- -- _ Oral Administration
f- c:
( '!,fjects oj the drug. Simply-speaktng, pharmacokinetics is the kinetics o._f :J
w ··.......~-
0
Drug Decomposed.
ADM£ or KADME. The use of pharmacokinetic principles in optimizing ()
411( ...
:e
-c:
.._
Cl)
Drug Reiease and
Dissolution ---t Metabolized, Bound
the drug dosage to suit individual patient needs and achieving maximum a: ·- or Excreted

therapeutic utility is called as clinical pharmacokinetics. < · ~ Absorption site


:I: ~ ABSORPTION
Q. in the GIT
· Drug admin-istration and therapy can now be conveniently divided into 0
-
m
four phases or processes: . Drug in Systemic
1: The Pharmaceutic
r Proce~s : It is concerned with the formulation
of an effective dosage fonn of the drug for administration by a suitable
1 Circulation

f
DISPOSITION

I

route. PHARMACOKINETICS DISTRIBUTION ELIMINATION
.' •I en
u
-w
' I

2. The Pbarmacokinetic Process : It is concerned with the ADME f-


Extravascular Tissues
. of drugs as elicitedJ?y the plasma drug concentration-time profile and its z Metabolism

: relati<;>nship with the dose, dosage fonn and frequency and route of ad-
-
~ issues at the and
0 Site of Action Excretion
ministration. In short~t is the sum of all the processes inflicted by the u
<
body on the drug. :e
a:------..-.-~ . . . --- -
3. The Pharmacodynamic Process : It is concerned with the bio-
<
:t:
Q.
chemical and physiologic effects of the drug and its mechanism of action. ~ PHARMACO-
It is characterized by the concentration of drug at the site of action and its U DYNAMICS Pharmacologic Response
-z
rel(ltion to the magnitude of effects observed. Simply speaking, -u....J ___________ ..,.. __ ._. __
pharmacodynamics deals with what the drug does to the body in contrast
to pharmacokinetics which is a study of what the body does to the drug. .
4. The Therapeutic Process : It is concerned with the translation of THERAPEUTICS Therapeutic/To.xic Effects
pharmacologic effect-into clinical benefit._
A schematic representation of the various processes involved in the Fig. 1.1 Schematic representation of the processes involved in drug therapeutics
· therapy with a drug is given in Fig. I. I.
other hand, rational use of the drug· or the therapeutic objective can only
· To achieve optimal therapy with a drug, the drug product must be be achieved through a better understanding of phannacokinetics (in addi--
tion to phannacodynamics of the drug), which helps in design ing a proper
-
4 BIOPHARMACEUTICS AND PHARMACOKINETICS
\

- dosage regimen (the manner in which the drug should be taken). This
obviates the use of the empirical approach where a considerable experi-
mentation is needed to arrive at the balance between the desired therapeutic
.,
and the undesired toxic effects in order to define an appropriate dosage • ,


regtmen. 2 ·. ...

~

.
'

" The knowledge and concepts of biophannaceutics and phannacokinet- .

Absorption· of Drugs
• •

ics thus have an integral role in the design and development of new drugs
and their dosage fonns and improvement of therapeutic efficacy of exist-
ing drugs. .

A drug injected intravascularly (intravenously and/or in.tra-arterially)


• directly enters the systemic circulation and exerts its pharrnacologic ef-
fects. However, majority of drugs are administered extravascularly, generally
orally. If intended to act systemically, such drugs can exert their
phannacologic actions only when they come into blood circu1ation from
their site of application, and for this, absorption is an important prerequi-
site step. · -..
.... .
Drug absorption is defined as the process of movement of uncfzanged .
drug from the site of administration to systemic circulation. Following
absorption, the effectiveness of a drug can only be assessed by its concen-
tration at the site of action. However, it is difficult to measure the drug
concentration at such a site. Instead, the concentration can be measured .
more accur~tely in plasma. There always exist a correlation between- the

plasma concentration of a drug and the therapeutic response and thus, ....._

c Therapeutic success
--
Q ,
\ ·-
«) of a rapidly and THERAPEUTIC LEVEL
'-
c completely absorbed drug
Q) •
u
c MINIMUM EFFECTIVE
0

u0>1 ~~------~----------------- CONCENTRATION


::J
~
Therapeutic failure
0 of a slowly absorbed
C'O
E drug
ff)

-0..
C'O SUBTHERAPEUTIC
LEVEL

--
.. Time ~
Fig/.Plots showing signific~nce of rate and •

extent of absorption in drug therapy


'
bsorption can also be defined as the process of movement.of unchanged
........,.~O"Jrom the site of administration to the site of measurement i.e. plasma.
/

5 '
I
I

. .
"
'
·1
.I J

6 BIOPHARMACEUTICS AND PHARMAS/OKINETICS ABSORPTION OF DRUGS 7


This definition takes into account the loss of drug that occurs after oral membranes/barriers at various locations. Such a movement of drug across
administration due to presystemic metabolism or first-pass effe~t.
-- the membrane is called as drug transport. -- ·
. Not only the magnitude of drug that comes into the systemic circula- The basic structure of cell membrane is shown in Fig. 2.2.
tion but also the rate at which it is absorbed is important. This is clear
from Fig. 2. 1 Water-filled
polar pore _Polar end
' }---H

A drug that is completely but slowly absorbed may· faiJ to show Lipid Q)
c
therapeutic response as the plasma concentration for desired effect is - - Nonpolar end _ ('Q
....
.0
never achieved. On the contrary, a rapidly absorbed ·, drug attains the E
4)
~
therapeutic leve-l easily to elicit phannacologic effect. Thus, both the rate Non polar end }
Lipid ~
and the extent of drug absorption are ~mportant. __ Such an absorption J--++--Polar end
pattern has several advantages: · •

1. Lesser susceptibility··of the drug for degradation or interaction due


to rapid absorption Fig. 2.2 Basic structure of functional cell membrane

2. Higher blood levels and rapid onset of action The cellular membrane consists of a ,.double layer of amphiphilic .phos-
3. More uniform, greater and reproducible therapeutic response pholipid molecules arranged in such ~ fashion that their hydrocarbon
chains are oriented inwards to form the hydropho.bic or lipophilic phase
Drugs that have to enter the systemic circulation to exert tfleir effect and their polar heads oriented to form the outer and inner hydrophilic
can be administered by three major routes: boundaries of the cellular membrane that face the surrounding aqueous
'
1... The Enteral Route : includes peroral i.e. gastrointestinal, sublin- environment~ , Globular protein molecules are associated on either side of
,.
gual/buctal and rectal routes. The G I route is the most common for these hydrophilic boundaries and also interspersed within the membrane r

administration of majority of drugs. structur~. In short, the membrane is a mayonnaise~ sandwich where a
birnolecular layer of lipids is contained between two parallel monomo-
2. The Parenteral Route : includes all routes of administration
lecular layers of proteins. The hydrophobic core of the membrane is
through or under one or more layers of skin. While no absorption is
responsi~le for the relative impermeability of polar molecules. Aqueous
required when the drug is administered i.v., it is necessary for extravascu-
filled pores or perforations of 4 to 10 A in diameter are also present in the
lar parenteral routes like the subcutaneous and the intramuscular routes.
membrane structure through which inorganic ions and small organic wa-
3) The Topical Route : includes skin, eyes or other specific mem- ter-soluble molecules like urea can pass n general, the biomembrane
br~nes. The intranasal, inhalation, intravaginal and transdennal routes acts like a semipermeable barrier pennitt piQ and limited passage of
may be considered enteral or topical according to different definitions. some compounds while restricting that of othe,rs.;-/
\ •
The G I lining constituting the absorption barrier ~llows most nutrients
\

GASTROINTESTINAL ABSORPTION OF DRUGS like glucose, amino acids, fatty acids, vitamins, etc. to pass rapidly through
The oral route of drug administration is the most common for systemi- it into the systemic, circulation but prevents the entry of certain toxins and
cally acting drugs and therefore, more emphasis will be give ointestinal . medicaments. Thus, for a drug to get absorbed after oral administration,
(GI) absorption of_drugs:- Moreover, it covers all the aspects of variability it must first pass through this biological barrier.
,I
observed in drug absorption. Before proceeding r'to Biscuss absorption

aspects, a brief description of cell membr~e structure and physiology is MECHANISMS OF DRUG ABSORPTION
necessary.
• The principal mechanisms for transport of drug molecules across the
Cell Membrane : Structure and Physiology ,. cell membrane in order of their importance are:
r
For a drug to be absorbed and distributed into organs and tissues and 1. Passive diffusion t)
eliminated from the body, it must pass through one or more biological 2 ~ -Pore transport 0 •

I
.
8 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 9
3. Facilitated diffusion Based on the above equation, certain characteristics of passive diffu-
4. Active transport sion can be generalized:
5. Ionic or electrochemical diffusion 1. The drug moves down the concentration gradient indicating down-
6. Ion-pair transport hill transport
.. 7. Endocytosis 2. The rate of drug transfer is directly proportional to the concentra-
Passive Diffusion tion gradient between GI fluids and the blood compartrnent

. Also called nonionic diffusion, it is the major process for absorption 3. G~er the area and lesser the thickness oft~ membrane, faster
of more than 90% of the drugs. The driving force for this process is the --the diffusion; thus, more rapid is the rate of drug absorption from
concentration or electrochemical gradient. It is defined as the differ- the intestine than from the stomach
ence in the drug concentration on either side of the membrane. Drug 4. Equilibrium is attained when. the concentration on either sipe of
movement is a result of the kinetic energy of molecules. Since no energy .._/ the membrane becomes equal
source is required,_ the process is called as passive diffusion . During 5. Drugs which can exist in both ionized and unionized fonns ap-
passive diffusion, ·the drug present in the aqueous solution at ·'the absorp- proach equilibrium primarily by the transfer of the unionized
tion site partitions and dissolves in the lipid material of the membrane and species; the rate of transfer of unionized species is 3 to 4 times
finally leaves it by dissolving again i~ an aqueous rnedium, this time at the rate for ionized drugs
the inside of the membrane. •
I
• 6. Greater the membrane/water partition coefficient of drug, faster
Passive diffusion is best expressed by Ficl{'s first law of diffusion, the absorption; since the membrane is lipoidal in nature, a li-
which states that the drug molecules diffuse from a region of higher pophilic drug diffuses at a faster rate by solubilizing in the lipid
concentration to one of lower concentration until equilibrium is attained layer of the membrane
and that the rate of diffusion is directly proportional to the concentration 7. The drug diffuses rapidly when the volume of G I fluid is low;
· gradient across the membrane. It can be mathematically expressed by the conversely, dilution of G I fluids decreases the drug concentration
following equation: in these fluids (CGIT) and lower the concentration gradient-· (CGIT
- dQ DAKm1w - C). This phenomena is, however, made use of in treating cases
l = = - - - (CGIT - C) (2.1) of oral overdose or poisoning.
dt h
Passive diffusion process is energy ~ndependent and nonsaturable but
where,
dependent, to a lesser extent, on the square root of the molecular size of
dQ/dt = rate of drug diffusion (amount/time). It also r~presents the the drug. The molecular weights of most drugs lie between I 00 to 400
rate of appearance of drug in blood daltons which can be effectively absorbed passively. The diffusion gener-
'
D = diffusion coefficient of the drug through the membrane ally decreases with increase in the molecular weight of the compound.
(area/time) However, there are exceptions for example, cyclosporin A, a peptide of
A , '
I

= surface area of the absorbing membrane for drug diffusion molecular weight 1200, is absorbed orally much better than any other
(area). peptide.

Krn1w = partition coefficient of ·the drug between the lipoidal mem- Initially, when the drug is ingested, CGIT >> C and a large concentra-
brane and the aqueous GI fluids (no units) tion gradient exists thereby acting as the driving force for absorption. As
equilibrium approaches, the drug diffusion should stop and consequently a
(Ca1T - C) = C:ifference in the concentration of drug in the GI fluids large fraction of drug may remain unabsorbed. But this is not the case;
and the plasma, called as the concentration gradient (amount/ once the passively absorbed drug enters blood, it is rapidly swept away
volume) and distributed into a much larger volume of -body fluids and hence, the
h = thickness of the membrane (length) , concentration of drug at the absorption site, CGIT, is maintained greater
J

'
10 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS II
.. .
than the concentration of drug in ' plasma. Such a condition is called as involve a component of the membrane called as the carrier that binds
sink condition for drug absorption. reversibly or noncovalently with the solute molecules to be transported.
\

Since under usual conditions of absorption, D, A, Km/w and h are This carrier-solute complex traverses across the membrane to the other
constants, the term DAKrn~wlh can be replaced by a combined constant P side where it dissociates and discharges the solute molecule. The carrier
called as permeability coefficient. Permeability refers to the ease with then returns to its original site to complete the cycle by accepting a fresh
which a drug can penetrate or diffuse through a membrane. Moreover, molecule of solute. The carrier n1ay be an enzyme or some other compo-
due to sink conditions, the concentration of drug· in plasma C is very nent of the membrane.
small in comparison to CaiT· As a re.sult, equation 2.1 . may be simpli- Important characteristics of carrier-mediated transport are:
fied to: I. The transport· process is structure-specific i.e. the carriers have
'
dQ special affinity for and transfer a drug of specific chemical struc-
~·- = P CaiT (2.2)
dt ture only; generally the carriers have special affinity for essential
nutrients.
Equation 2.2 is an expression for a first-order process. Thus, passive
2. Since the system is structure-specific, drugs having structure simi-
diffusion follows first-order kinetics. Since a large concentration gradient
lar to essential nutrients, called as false nutrients, are absorbed by
always exist at the absorption site for passive diffusion, the rate of drug
the same carrier system. This mechanism is of particular impor-
absorption is usually more rapid than the rate of elimination. Besides, . . tance in the absorption of several antineoplastic agents like
dilution and distribution of th~ absorbed drug into a large pool of body ,
5-tluorou.racil and 5-bromouracil which serve as false nutrients.
fluids and its subsequent binding to various tissues are other reasons for
elimination being slower than absorption. 3. As the number of carriers are limited, the transport system is
subject to competition between agents having similar structure.
Pore Transport 4. Since the number of carriers is limited, ~he system is capacity-
It is also called as convective transport, bulk flow or filtration. limited i.e. at higher drug concentration, the system becomes
The process is important in the absorption of low mo!ecular weight .(less saturated and approaches an asymptote. It is important to note
than l.Q.O), low molecular size (smaller than the diameter of the pore) and that for a drug absorbed by passive diffusion, the rate of absorp-
generally water-soluble drugs through narrow, aqueous-filled channels or tion increases linearly with the concentration but in case of
pores in the membrane structure for example, urea, water and sugars. carrier-medi~ted processes, the drug absorption increases linearly
Chain-like or lirrear compounds of m?lecular weight upto 400 daltons can with concentration until the carriers become saturated after which -
be absorbed by ;filtration. The. driving force is constituted by the hydro- ..
-
static pressure jlr the osmotic differences across the membrane due to

which bulk flow of water alongwith small solid molecules occurs through c Passive diffusion
such aqueous channels. Water flux that promotes such a transport is -
0
0 ..
..._
0
called as ::olvent drag. f/)
.0
ro
Drug penneation through water-filled channels is of particular impor- 0>
::J
..._
tance in renal excretion, removal of drug from the cerebrospinal fluid and
-
-o
0
entry of drugs into the liver.
-
C1>
ro
0:::
Carrier-Mediated Transport
Some polar drug~ cross the membrane more readily than can be
predicted from their concentration gradient and partition coefficient val-
Concentration of drug at the absorption site
ues. This suggests presence of specialized transport mechanisms without I

which many essential water-soluble nutrients like monosaccharides, amino Fig. 2.3 Comparison of rate of absorption versus drug concentration
acids and vitamins will be po~rly absorbed. The mechanism is thought to plots for passive and carrier-mediated transport processes

( \
12 BJOPHARMACEUTICS AND PHARMACOKH ~TICS ABSORPTION OF DRUGS 13

it becomes curvilinear and approach a constant value a. higher interfere with energy production. Facilitated diffusion is of limited impor-
'•
doses (see Fig. 2.3). tance in the absorption of drugs. Examples of such a transport system
' Such a capacity-limited process can be adequately described by ... . include entry of glucose into RBCs and intestinal absorption of yitamins
mixed order kinetics, also called as Michaelis-Menten, satura- B1 and 82. A classic example of passive facilitated .diffusion is the GI
tion -or non-linear kinetics. The process is called mixed order absorption of vitamin B 12 . An intrinsic factor (IF), a glycoprotein pro-

because it is first-order at subsaturation drug concentrations and duced by the gastric parietal cells, fonns a complex with vitamin B 12
apparent zero-or~er at and above saturation levels. Moreover, the which is then transported across the intestinal membrane by a carrier
capacity ... Jimited characteristics of such a system suggest that the system (Fig. 2.4 ).
bioavailability of a drug absorbed by such a system decrease with
Active Transport
increasing dose for example, vitamins like Bt, B2 and B 12. Hence,
administration of a large single oral dose of such vitamins is Active transport is a more important process than facilitated diffusion
irrational. · in the absorption of nutrients and drugs and differs from it in several
respects:
5. Specialized absorption or carrier-mediated absorption generally ·
occurs from specific sites of the intestinal tract which are rich in 1. The drug is transported from a region of lower to one of higher
number of carriers. Such an area in which the carrier system is concentration i.e. against the concentration gradient or uphill transport,
most d~nse is called as absorption w.indow. Drugs absorbed without any regard for equilibrium
through such absorption windows are poor candidates for con- 2. Since the process is uphill, energy is required in the work done
trolled release formulations. by the carrier
Two types of carrier-mediated transport sy·stems have been identified. 3. As the process requires expenditure of energy, it can be inhibited
They are facilitated diffusion and active transport. · by metabolic poisons that interfere with energy production like
fluorides, cyanide and dinitr~phenol and lack of oxygen, etc.
Facilitated Diffusion .
Endogenous substances that are transported actively include sodium,
It is a carrier-mediated transport system that operates down the con- potassium, calcium, iron, glucose, certain amino acids and vitamins like
centration gradient (downhill transport) but at a much a faster rate than niacin, pyridoxin and ascorbic acid. Drugs having structural similarity to
can be accounted by simple passive diffusion. The driving force is •

concentration gradient (hencea passive process). Since no energy expeQ- Gl Lumen Membrane Blood
diture is involved, the process is not inhibited by metabolic poisons that •

Intestinal Membrane Blood ·


Lumen

Drug

.-.,..;.....~ Dissociation Free


Drug-Carrier
complex • drug

Free vitamin 8 ,2

8 12-IF-Carrier
complex Dissociation of
complex
,
Fig. 2.4 Facilitated diffusion of vitamin 8 12 Fig. 2.5 Active absqrption of a drug
,--- ) ~


ABSORPTION OF DRUGS 15
14 BIOPIIARMACEUTICS AND PHARMACOKINETICS ·'

such agents are absorbed actively, particularly the agents useful in cancer Ion-Pair Transport
chemotherapy. Examples include absorption of 5-fluorouracil and 5- Yct another mechanism that explains the absorption of drugs like
bromouracil via the _pyrimidine transport system, absorption of methyldopa quaternary amn1onium compounds and sulfonic acids, which ionize under

and levodopa via an L-amino acid transport system and a_bsorption of all pl l ondit;ons, is ion-pair transport. Despite their low o/w partition
ACE inhibitor enalapril via the small peptide carrier system. A good coefficient values, such agents penetrate the membrane by forming revers-
I

example of competitive inhibition of drug absorption via active transport ible neutral complexes with endogenous ions of the GIT like mucin.
is the impaired absorption of levodopa when ingested with meals rich in Such neutral complexes have both the required lipophilicity as well as
proteins. Active transport is also Jmportant in renal and biliary excretion aqueous solubility for passive diffusion. Such a phenomena is called as
--
of many drugs and their metabolites and secretion of certain acids out of ion-pair transport (Fig. 2.6).
.. #

the CNS (Fig. 2.5). I


.?End cytosis
Ioriic or Electrochemical Diffusion I is a minor transport mechanism which involves engulfing extracellu-
The charge on the membrane influences the permeation of drugs. lar;materials within a segment of the cell membrane to form a saccule or
Molecular forms of solutes are unaffected by the membrane charge and a vesicle (hence also called as corpuscular or vesicular transport) which
' permeate faster than ionic forms. Of the ionic forms. the anionic solute is then pinched-off intracellularly (Fig. 2. 7).
permeates faster than the cationic form. r__ Thus, at a given pH. the rate of Membrane Inside
Outside
permeation is in the following order uni;ni~ed ni()/ucules > anions > Cell
Cell
cations.
/
The permeation of ionized drugs, particularly the cationic drugs, de- Macromolecule
pend on the potential, difference 9r electrical gradient as the driving force
across the membrane. A cationic drug is repelled due to positive charge
. --
,- on 'the outside of the membraAe. As a result, only those cations with a
'I

high kinetic energy pene!rate the ionoc barrier. However, once inside the
membrane, the . cations ·are attracted to negatively charged intracellu~ar
Process of
membrane thereby creating an electrical gradient. Such a drug is then , engulfing
said to be moving downhill with electrical gradient. If the same drug
moves from a higher to lower concentration, it is said to be moving down

the electrical gradient and the phenomena is called as electrochemical
diffusion. Like passive diffusion, the process continues until equilibrium Fig. 2. 7 Endocytic uptake of macromolecules
is reached. This phenomena is responsible for the cellular uptake of macromolecu-
lar nutrients like fats and starch, oil soluble vitamins like A, D, E and K
Gl Lumen Membrane Blood and drugs such as insulin. Another significance of such a process is that
the drug is absorbed into the lymphatic circulation thereby bypassing first-
pass hepatic metabolism .
..
Endocytosis includes two types of processes:
I. Phagocytosis (cell eating) : adsorptive uptake of solid particu-
lates, and
Free 2. Pinocytosis (cell drinking) : uptake of fluid solute.
Cationic Endogenous Neutral
. . drug
I

drug an1on 1on-pa1r Orally administered Sabin polio vaccine and large protein molecules
complex are thought to be absorbed by pinocytosis. Sometimes, an endocytic
Fig. 2 6 Ion-pair transport of a cationic drug
\
16 BIOPHARMACEUTICS AND PHARMACOKINETICS
,
ABSORPTION OF DRUGS
17-
'
••

vesicle is transferred
. .
from one extracellular compartment to another. Such biophannaceutic design, the rate and extent of drug absorption (also called
a phenomenon is called as transcytosis. as. ~.ioavailabili~y) or the -systemic delivery of drug to the body can be
A drug might be absorbe·d by more th~n just one mechanism for ~aned from .raptd and comple~e absorption to slow and sustained absorp-
example, cardiac glycosides are absorbed both passively as well as by tion dependrng upon the desJrect therapeutic objective. The chain of
active transport. T~e transport mechanism also depends, upon the site of .. - events that occur following administration of a solid dosage fo 1111 such as
drug administration (see Table 2.8). a tablet or a capsule until its absorption into systemic circulation are
> depicted in Fig. 2.9.
Absorption of drugs by various mechanisms is summarized in Fig. I

-· ... -·· .
2.8. - G I Lumen G I Barrier . Blood
Absorption Drugs Gl Lumen Membrane Blood solid dosage fW41 Dissolution
Mechanism Absorbed forms . minQr Non ionic
Passive Most drugs having Disintegration (3) drug ~
(1) .
Diffusion high lipophilicity and Drug in solution
granules or 0 00 0 Dissolutio
.. molecular weight in 0o o at the Absorption (4)
the range 100-4.0 0 aggregates oc::s ~ major absorption site
Deaggregation- (3)
Pore Water-soluble drugs (2) Dissolution · Ionic Ionic
Transport of molecular weight ..., .... major drug drug
. '\,.

less than 100 fine particles ... ~:?(~}. (3)

Carrier- Structure-specific
Mediated drugs with affinity for Fig. 2.9 Sequence of events in the absorption of drugs-
Transport carriers transported from orally administered solid dosage forms
from specific sites
The process consists of four steps:
I. Disintegration of the drug product
Ion-pair Drugs that ionize at
Transpo·rt all pH conditions 2. Deaggregation and subsequent release of the drug
absorbed after 3. Dissolution of the drug in the aqueous fluids at the absorption site
complexing with
oppositely charged
.
1ons
08 4. Movement of the dissolved drug through the Gl membrane into ,
the systemic circulation and away from the absorption site ..

Endo- Macromolecular
As illustrated in Fig. 2.9, the drug may also dissolve before disinte-
cytosis nutrients and drugs gration or ~eagg~egation of the dosage fot 111, and before or ~fter reaching
as solid particles or the absorption srte. Unless the drug goes into solution, it cannot be
oily droplets absorbed into the systemic circulation. . .
In a series of ~ine.tic or . rat~ processes, the rate at tvhich the drug
reaches the system1c c1rcu/at1on 1s determined by the slowest of the vari-
Fig. 2.8 Summary of important transport processes
ouJ steps involved in the sequence. Sucn ·. a step ·.is called as the
and drugs absorbed through them
rate-determining or rate-limiting -~tep (RDS). . The rafe . and extent of
drug ab.sorption from its dosage form can be. influenced by ·.a ~umber of
FACTORS INFLUENCING
. DRUG. ABSORPTION ~actors 1n all these st~ps. The vari?us factors, t~a\ i!lfluence drug absorp-
AND BIOA VAI LABILITY tion (also ca.lled as baopha~maceut1c factors ig tiJ_e do8age--. form design) .
Biopharmaceutic Considerations in Dosage Form Design can be classified. as shown Jn Table 2.1. ·
To achieve the desired therapeutic objective, the drug product must
deliver the active drug at an optimal rate and amount. By proper
..

BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 19
• •

TABLE 2.1 PHYSICOCHEMICAL FACTORs-AFFECTING DRUG ABSORPTION


_..Factor:s influencing GI Absorption of a Drug from its Dosage Form Dr ug Solubility and 'Dissolution Rate
A. PHARMACEUTIC FACTORS : include factors relating to the physicochemi- Consider the events that occur following oral administration of solid
cal properties of the drug, and dosage form characteristics and pharmaceutic dosage fonn as shown in Fig. 2.9. Except in case of controlled release
ingredients form~lations, disintegration and deaggregation occur rapidly if it is a well
formulated dosage form. Thus, the two critical slower rate-dete11nining
I. Ph ysicochemical Properties of Drug Substances
processes in the absorption of orally administered drugs are:
L:...,.Drug solubility and dissolution rate
y Particle size and effective surface area 1. Rate of dissolution, and
J' 3. Polymorphism and amorphism 2. Rate of drug perrneation through the biomembrane.
<- f Pseudopolymorphism (hydrates/solvates) •
Dissolution is the RDS for hydrophobic, poorly aqueous soluble drugs
5. Salt form of the drug like griseofulvin and spironolactone; absorption of such drugs is often said
6. Lipophilicity of the drug } pI-I -partition
. . hypothests
. to be dissolution rate-limited. If the drug is hydrophilic with high
7. pKa of the drug and pH 1 aqueous solubility for example, cromolyn sodium or neomycin, then
8. Drug stability dissolution is rapid and the RDS in the a_bsorption of such drugs is rate of
permeation through the biomem brane. In otht?r words, absoll?,t~on. of such
II. Dosage Form Characteristics and Pharmaceutic
..
Ingredients
drugs is said to be permeation rate limited or transmembrane rate.:-
I. Disintegration time (tablets/capsules)
limited (Fig. 2.10).
2. Dissolution time
permeation
3. Manufacturing variables Solid dissolution Drug in Solution across the Drug
Solid disintegration!
4. Pharmaceutic ingredients (excipien~s/adj u vants) Dosage • Drug a at the in the
Form deaggregation Particles Absorption Site biomembrane Body
5. Nature and ty pe of dosage form •

6. Product age and storage conditions


B. PATIENT RELATED FACTORS : include factors relating to the anatomic,
RDS for
Lipophilic Drug
'
RDS for
Hydrophilic Drugs
physiologic and pathologic characteristics of the patient
t. _Age Fig. 2.10 The two rate-determining steps in the absorption

of drugs from orally administered formulations
I 2. Gastric emptying time
~3. Intestinal transit time An important prerequisite for the absorption of a drug by all mecha-
4. Gastrointestinal pH nisms except endocytosis is that it must be present in aqueous solution.
' 5. Disease states This in tum depends on the drug's aqueous solubility and its dissolution
6. Blood flow through the GIT rate. Absolute or intrinsic solubility is defined
,- as the maximum amount
of solute dissolved in a given solvent under standard conditions of temper-
V'Gastrointestinal contents:
a. Other drugs ature, pressure and pH. It is a static property. Dissolution rate is
b. Food d.efined as the amount of solid substance that goes into solution per unit
c. Fluids time under standard conditions of temperature, pH and solvent composi-
d. Other normal GI contents tion and constant solid surface area. It is a dynamic process. Several
8. ·Presystemic metabolism by: drugs have poor aqueous solubility to have a bearing on dissolution rate.
a. Lumenal enzymes The matter is of great concern when the solubility is less than 1 to 2 mgl
b. Gut wall enzymes ml in the pH range of 2 to 8. However, there are well known examples
c. Bacterial enzymes of drugs such as cisapride which inspite of low aqueous solubility have
d. Hepatic enzymes sufficient ~r~l bioavailability. Two reasons can be a~buted to this one,
.
-~-

ABSORPTION OF DRUGS
. . 21 . .
ll 20 BIOPHARMACEUTICS AND PHAR.MACOKINETICS

the rapid rate of -dissolution despite low intrinsic solubility and two, the .
therapeutic dose of drug may be so small that the GI transjt time is
sufficient for adequate dissolution ~nd absorption -· to occur. Thus, in
-contrast to absolute solubility, the dynamic process of drug dissolution is
better related to drug absorption and bioavailability.

Theories of 'D rug Dissolution


Dissolution is · a. process in which a solid substance solubi/izes in a
·given $Olvent i.e. mass transfer from the solid surface to the liquid phase.
as the '

Several theories to explain drug dissolution have been proposed. . Some of


, the important Qnes are: ~

1. Diffusion layer model/Film theory '

2. Danckwert's modeVPenetration or Surface renewal theory, and · Equation 2.3 was based on Fick's second law of diffusion. Brunner
. . incorporat~d Fick's first law of diffusion and modified the Noyes-Whitney's
3. Interfacial barrier modeVDouble b~ier or Limited solvation theory.
equation to:
. . I

Diffusion Layer Model/Film Theory \ dC .DAKw/o (C 5 -- Cb)


This i~ the simplest and the most comm.o n theory for dissolution.I
- (2.4) ·-
dt Vh
.Here, the process of dissolution of solid particles in a liquid, in the where,

absence of reactive or chemical forces, con~ists of two con~ecutive steps:


•.
.. ..
D = diffusion coefficient (diffusivity) of the drug
1. Solution of the solid to foun a thin film or layer at the solid/
A = surface area of the dissolving solid
liquid interface called as the stagnant . film or diffusion layer ••

which is saturated with the drug; this step is usually rapid, and Kw/o = water/oil partition coefficient of the drug considering the fact

• that d~ssolutiqn body fluids are aqueous. Since the rapidity
--Solid/liquid interface with which a drug dissolves depends on the Kw;0 , it is also
called as the in!rinsic dissolution rate constant. It is a
characteristfc of drugs.
V · = volume of dissolution medium.
h = thickness, of the stagnant layer.

- (C 5 - Cb) = concentration gradient for diffusion of drug~


The influence of various parameters in equation 2.4 on drug dissolu-
-Bulk of the solution tion is depicted in Table 2.2. . , ·
-with concentration Cb -
- I .::.. .... - TABLE 2.2 Influence of Some Parameters on Dissolution Rate of Drug
Parameters Symbol Influence on drug dissolution ..

Dissolving Solid Drug Gl Lumen Gl Barrier Blood ' '


Diffusion coefficient D Greater the value, faster the dissolution.
Fig. 2.11 _Diffusion layer model for drug dissolution of drug Diffusion decreases as the viscosity of
'
dissolution medium increases.
. . L>iffusion of the soluble solute from the stagnant layer to the bulk •
'

.of the solution; this step is slower and· is therefore the rate- Surface area of solid Greater the surface area, faster the
'
drug dissolution; .can be increased by
J" ;,,.. r "1ining step in drug dissolution. The model is depicted in
micronization of drug.
... continued ·..
-

22 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 23

4. Adding a water miscible solvent such as alcohol to the dissolution


Parameters Symbol Influence on drug dissolution
fluid, or
Water/oil partition Kw/o Higher the value, more the 5. By adding selected adsorbents to remoye the dissolved drug.
coefficient of drug hydrophilicity and faster the
0>
dissolution in aqueous fluids. :J
'-
0
Concentration gradient (C 5 - Cb) Greater the concentration gradient, 'U
Q)
Zero-order dissolution
faster the diffusion and drug dissolu-
-0> under sink conditions
tion; can be increased by increasing t/)

·-0
t/)
First-order dissolution
drug solubility and the volume of _£....-----under non-sink conditions
......
dissolution medium. 0
c
Thickness of ~tagnant h More the thickness, lesser the diffusion ·-oco .·
~ .

layer and drug dissolution; can be decreased


by increasing agitation.
-'-
c
Q)
0
c:
0
(.)
' Equation 2.4 represents first-order dissolution rate process, the driving
force for which is the concentration gradient (Cs - Cb). Under such· a T
sitUation, dissolution is said to be under nonsink conditions. This is true -~) Time
in case of in vitro dissolution in a ·limited dissolution medium. Dissolu- Fig. 2.12 Dissolution rate under nonsink and sink conditions .
. tion in such a situation slows down after sometime due to build-up in the
concentration of drug in the bulk of the solution. The in vivo dissolution The in vitro sink conditions are so maintained that Cb is always less
is always rapid than in vitro dissolution because the moment the drug than 10% of Cs.
dissolves, it is absorbed into the systemic circulation. As a result, Cb = 0, The Noyes-Whitney's equation assumes that the surface area of the
and dissolution is at its maximum. Thus, under in vivo conditions, there dissolving solid remains constant during dissolution, which is practically
is no concentration b~ild-up in the bulk .o f the solution and hence no not possible for dissolving particles. Hence, dissolution methods that
retarding effect on .the dissolution rate of the drug i.e. Cs >> Cb and sink involves use of constant surface area discs are employed to determine the
conditions are maintained. Under sink conditions, if the volume and rate of dissolution.
surface area of solid are kept constant, then equation 2.4 reduces to: To account for the particle size decrease and change in surface area
accompanying dissolution, Hixson and Crowell's cubic root law of dis-
dC
--=K (2.5) solution is used:
dt
(2.6)
where K incorporates all the constants in equation 2.4. Equation 2.5 where,
represents that the dissolution rate is constant under sink conditions and W0 = original mass of the drug
follows zero-order kinetics i.e. yields a linear plot (Fig. 2.12).
W = mass of the drug remaining to dissolve at time t
To obtain good in vitro-in vivo dissolution rate correlation, the in vitro ·
K = dissolution rate constant
dissolution must always be carried under sink conditio~s. This can be
achieved by: Danckwert's Model (Penetration or Surface Renewal Theory)
'
1. Bathing the dissolving splid in fresh solvent from time to time Danckwert did not approve of the existence of a stagnant layer and
2. Increasing the volume of dissolution fluid suggested that turbulence in the dissolution medium exists at the solid/
liquid interface. As a result, the agitated fluid consisting of macroscopic
3. Removing the dissolved drug by partitioning it from the aqueous mass of eddies or packets reach the solid/liquid interface in a random
phase of the dissolution fluid into an organic phase placed either fashion due to eddy currents, absorb the solute by diffusion and carry it to
above or below the dissolution fluid for example, hexane or the bulk of the solution. Such solute containing packets are continuously
chlorofortn

I
-. /

24 BIOPHARMACEUTICS AND .·PHARMACOKINETICS ABSORPTION OF DRUGS 25


,
.
·- '

replaced with new packets of fresh solvent due to which the drug concen- _ In this theory, the diffl'sivity D may not be independent of saturation
tration at the solid/liquid interface never reaches Cs and has a lower concentration Cs. The interfacial barrier model can be extended to both
limiting value of Ci. Since the solvent packets are exposed to new solid diffusion layer model and the Danckwert' s model o(jor in vitro drug
I

surface each time, the theory is called as surface renewal theory. dis~olution models refer chapter 12).
-
t
The Danckwert's model is expressed by equation: F~~tors Affecting Drug Dissolution and Dissolution Rate
--- ~a-ctorsof in vivo_importance that can ~ffect dissolution and hence
• dm = A(Cs- Cb) ..{yD (2.7) .. . --
dt absorption can be categerized into 2 classes:
--·· .. -
where, . •
- 1. Physicochemical properties of the drug, and -

m = mass of solid dissolved, and - _ 2. Dosage fonn factors.
y = rate of surface renewal (or the interfaci~l tension). .. The various physicochemical properties of drug that affect drug disso-
The model is depicted in Fig. 2. 13. lution and its rate are solubility, particle size, polymorphism, salt fonn,
pseudopolymorphism, complexation, wettability, etc. Dosage form factors
.
,...__ Sol_
id/Liquid interface having include several formulation factors and excipients incorporated in the
concentration Ci < Cs dosage fonn. Each of these factors will be discussed in detail in the latter
0

- 0.
Fresh packet of solvent part of this chapter.
Of the various factors listed above, the factor of prime importance is
~·- Packet of solvent saturated with drug solubility. Almost every factor that affects dissolution rate, influ-
-A C:-
-
-~ drug leaving the interface ences the drug solubility in one way or the other. From several equations
pertaining to dissolution rate, it is clear that it is directly related to drug
- - ~- Bulk .of the solution having
~~-~ _ ~ _ C:: - concentration Cb < Ci solubility. An empirical relation which is useful to predict the dissolution
rate of a drug from its solubility is:
Fig. 2.13 Danckwert's model for drug dissolution dC
R = = 2.24 Cs (2.9)
Interfacial Barrier Model (Double Barrier or Limited Solvation Theory) dt
The diffusion layer model oand the Danckwert's model were based on I where R = dissolution rate of the drug.
two assumptions: •
It has been shown that a drug should have a mtntmum aqueous

"
1. The rate-determining step that controls -dissolution is -the .mass - solubility of I o/o to avoid bioavailability probl~ms.
transport. _. ---
Particle Size and Effective Surface Area of the Drug
2. Solid-solution ·equilibrium is achieved at the solid/liquid interface.
Particle size and surface area of a solid drug are inversely related to
According to the interfacial barrier model, an intermediate concentra- each other. Smaller the drug particle, greater the surface area. Two types

tion can exist at the interface as a result of solvation mechanism and is a of surface area of interest can be defmed:
function of solubility rather than diffusion. When considering the dissolu- 1. Absolute surface area which is the total area of solid surface of
.tion of a crystal, each face of the crystal will have a different interfacial
any particle, and
barrier. ·Such a concept is given by the following equation:
2. Effective surface ·a rea which is the area of solid surface exposed
'•
(2.8) to the dissolution medium.
.
where,
From the modified Noyes-Whitney equation 2.4, it is clear that larger
G = dissolution rate per unit area, and - the surface area, higher the dissolution rate. Since the surface area
Ki = effective inte~facial transp~rt constant. increases with decreasing particle size, a decrease in particle size, which
26 BIOPHARMACEUTICS AND PHARMACCKINETiCS ABSORPTION OF DRUGS 27

can be accomplished by micronization, will result in higher disso tion 2. Adding hydrophilic diluents such as PEG, PVP, dextrose, etc.
rates. However, it 'is important to note that it is not the absolute ~ttiface which coat the surface of hydrophobic drug particles and render
area but the effective surface area that is proportional to the dissolution · them hydrophil~c.
rate. Greater the effective surface area, more intimate the contact between Particle size reduction and subsequent increase in the surface area and
the solid surface and the aqueous solvent and faster the dissolution. But it dissolution rate is. not always advisable especially when the drugs are
is only when micronization reduces the size of particles below 0.1 mi- unstable and degrade in solution form (penicillin G and erythromycin),
crons that there is an increase in the intrinsic solubi1itv and dissolution produce undesirable effects (gastric irritation caused by nitrofurantoin) or
rate of the drug. The surface of such small particles have energy higher when a sustained effect is desired.
than the bulk of the solid resulting in an increased interaction with the In addition to increasing the dissolution rate, the second mechanism by
solvent. This is particularly true in case of drugs which are nonhydrophobic, which a reduction in particle ~ize improves drug dissolution is through an
.for example, micronization of poorly aqueous soluble drugs like griseofulvin, increase in its solubility. Flowever, such an effect can only be achieved
chloramphenicol and several salts of tetracycline results in superior disso- by reducing the particle size to a submicron level which is possible by use
lution rates in comparison to the simple milled fonn of these drugs. of one of the following specialized techniques such as formation of:
Micronization has in fact enabled the fonnulator to decrease the dose 1. Molecular dispersion/solid solution where the sparingly soluble
of certain drugs because of increased absorption efficiency for example, drug is molecularly trapped in the lattice of a .hydrophilic agent
the griseofulvin dose was reduced to half and that of spironolactone was such as cyclodextrins, or
decreased 20 times following micronization. However, in case of hydro- I

2. Solid dispersion where such a drug is dispersed in a soluble


phobic drugs like aspirin, phenacetin and phenobarbital, micronization
carrier such as PVP, PEG, urea, etc.
actually results in a decrease in the effective surface area of such powders
and thus a fall in the dissolution rate. Three reasons have been suggested (Refer chapter 12 for methods used in enhancing the bioavailability of
'
for such an outcome:- drugs)..
1. The hydrophobic surface of the drugs adsorb air onto their surface
Polymorphism and Amorphism
which inhibit their wettability; such powders float on the dissolu-
Depending upon the internal structure, a solid can exist either in a
tion medium.
crystalline or amorphous form (Fig. 2.14 ). When a substance exists in
2. The particles reaggregate to form larger particles due to their high 1nore than one crystalline .form, the different forms are designated as
surface free energy, \\'h ich either float on the surface or settle at polymorphs and the phenomenon as polymorphism. PoJymorphs are of
the bottom of the dissolution medium. two types:
3. Extreme particle size reduction may impart surface charges that t. Enantiotropic polymorph is the Of!e which can be reversibly
may prevent wetting; moreover electrically induced agglomeration changed into another form by alteri'!g the temperature or pres-
may prevent intimate contact of the drug with the dissolution sure e.g. sulfur, and
medium. ·
t 2. Monotropic polymorph is the one which is unstable at all tern-
The net result of these effects is that there is a decrease in the .. peratures and pressures e.g. glyceryl stearates.
effective surface area available to the dissolution medium and therefore a
The polymorphs differ from each other with respect to · their physical
fall in the dissolution rate.
properties such as solubility, melting point, density, hardness and com-
The absolute surface area of hydrophobic drugs can be converted to pression characteristics. They can be prepared by crystalli~ing the drug
their effective surface area by: from different solvents under diverse conditions. The existence of the
1. Use of surfactant as a wetting agent that decreases the interfacial polymorphs can be detennined by using techniques such as optical crys-
tension and displaces the adsorbed air with the solvent for ex- tallography, X-ray diffraction, differential scanning calorimetry, etc.
ample, tween 80 increases the bioavailability of phenacetin. by
' •
promoting its wettability, and
I
28 BIOPHARMACEUTICS AND PHARMACOKINETICS o ABSORPTION OF DRUGS 29
Internal Structure of Compound exist in as many as 6 polymorphic forms and cortisone acetate in 8 forms.

Crystalline
Some ·drugs can exist in amorphous form (i.e. having no internal
Noncrystalline '
crystal structure). Such drugs represent the highest energy state and can
(Amorphous)
-- be considered as supercooled liquids. They have greater aqueous solubil- ~­
Polymorphs Molecular Adducts
ity than the crystalline fot n1s because the energy required to transfer a
(Single Molecules) molecule from crystal lattice is greater than that required ·for noncrystalline
. , _ _....!_ _--.,
I I
I I (amorphous) solid -for example, the amorphous fonn of novobiocin is 10
Enantiotropic Monotropic Non stoichiometric Stoichiometric times more soluble than the crystalline form. Chloramphenicol palmitate,
Complexes Complexes cortisone acetate and phenobarbital are other examples where the amor-
(Pseudopolymorphs) phous forms exhibit higher water solubility. Thus, the order for dissolution
of different solid fonns of drugs is -- amorphous > metastable > stab_le.
Organic Solvates Hydrates
Hydrates/Solvates (Pseudopolymorphism)
Fig. 2.14 Classification of internal structure of a compound
The crystalline foun of a drug can either be a polymoQJh or a molecu-
Depending on their relative stability, one of the several polymorphic lar adduct or both. The stoichiometric type of adducts where the solvent
forms- will be physically more stable than the others. Such a stable molecules are incorporated in the crystal lattice of the solid are called.as
polymorph represents the lowest energy state, has highest melting point the solvates, and the trapped solvent as solvent of crystallization. The
and least aqueous solubility. The remaining polymorphs are called as so/vales can exist in different crystalline forms called as pseudopolymorphs.
metastable forms which represent the higher energy state, have lower • This phenomenon is called as psetidopolymorphism. When the solvent in
melting points and higher aqueous solubilities. Because of their higher association with the drug is water, the solvate is known as a hydrate.
energy state, the metastable forms have a thermodynamic tendency to Hydrates are most common solvate fonus of drugs.
convert to the stable form. A metastable form cannot be called unstable Generally, the anhydrous fonn of a drug has greater aqueous solubility
because if it is kept dry, it wiiJ remain stable for years. than the hydrates. This is because the hydrates are already in interaction
Since the metastable forms have greater aqueous solubility, they show with water and therefore have less energy for crys~al break-up in compari-
better bioavailability and are therefore preferred in formulations for ex- son to the an hydrates (thermodynamically higher energy- state) for further
ar:nple, of the three polymorphic forms of chloramphenicol palmitate -A, interaction with water. The anhydrous fonn of theophylline and ampicillin
B · and C, the B form shows best availability and the A form is virtually have higher ~queous solubilities, dissolv~af a faster rate and show better
,,
inactive biologicaiJy. The polymorphic fonn , III of riboflavin is 20 times I bioavailability in comparison to their monohydrate and trihydrate forms
· more water-soluble than the fonn I. Only 10% of the pharmaceuticals are respectively. On the other hand, the organic (nonaqueous) solvates have
present in their metastable forms. However, because of their poor thenno- greater aqueous solubility than the nonsolvates for example, the n-pentanol
---
dynam ic stability, aging of dosage forms containing such metastable forms solvate of fludrocortisone and succinylsulfathiazole and the chloroform
>
usually result in fonnation of less soluble, stable polymorph for ex- solvate of griseofulvin are more water-soluble than their nonsolvated forms.
ample, the more soluble crystalline form II of cortisone acetate converts to Like polymorphs, the solvates too differ from each other in tenns of their
the less soluble fonn V in an aqueous suspension resulting in caking of physical properties. In case of organic solvates, if the solvent is toxic,
solid. Such a transfonnation of metastable to stable fot 111 can be inhibited they are not of therapeutic use.
by dehydrating the molecule envi'ronment or by adding viscosity building
macromolecules such as PVP, CMC, pectin or gelatin that prevent such a Salt Form of the Drug
conversion by adsorbing onto the surface of the crystals. Most drugs are either weak acids or weak bases. One of the easiest
approach to enhance the sol\lbility and dissolution rate of such drugs is to
About 40% of all organic compounds can exist in various polymorphic
convert them into their salt forms. Generally, with weakly acidic drugs, a
forms . Seventy percent of the barbitur~tes and 65% of sulfonamides
strong base salt is prepared such as the sodium and potassium salts of
exhibit polymorphism . Barbital, methyl paraben and sulfapyridine can I

barbiturates and sulfonamides. In case of weakly basic drugs, a strong


...
10 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 31

acid salt is prepared like the hydrochloride or sulfate salts of several into the bulk of the solution whose pH is low, it is transformed into its
alkaloidal drugs. free acid form having lesser aqueous solubility at the lower pH of the
At a given pH, the solubility of a drug, whether acidic/basic or its salt bulk solution. Consequently, this free acidic fonn of the drug is precipi-
form, is a constant. The influence of salt founation on the drug solubility, tated in the form of fine particles. The resultant increase in the surface
rate of dissolution and absorption can be explained by considering the pH area is then responsible for the rapid dissolution and absorption in com-
of the diffusion layer and not the pH of the bulk of the solution (refer parison to the drug administered in just the acidic form (Fig. 2.15).
diffusion layer theory of drug dissolution). Consider the case of a salt of The principle of in situ salt formation has been utilized to enhance the
a weak acid. At any given pH of the bulk of the solution, the pH of the dissolution and absorption rate of certain drugs like aspirin and penicilJin
diffvsion layer (saturation solubility of the drug) of the salt fonn of a from buffered alkaline tablets. The approach is to increase the pH of the
weak acid will be higher than that observable with the free acid fonn of microenvironment of the drug by incorporating buffer agents and promote
the drug (can be practically observed in the laboratory). Owing to the dissolution rate. Apart from the enhanced bioavailability, buffered aspirin
increased pH of the diffusion layer, the solubility and dissolution rate of a tablets have two more advantages: firstly, the gastric irritation and ulcerogenic
weak acid in this layer is promoted, since it is a known fact that higher tendency of the drug is greatly reduced, and secondly, the problem with
pH favors the dissolution of weak acids. Thus, if dissolution is faster, the use of sodium salt of aspirin (to enhance the solubility) which other-
absorption is· bound to be rapid. In case of salts of weak bases, the pH of wise has poor hydrolytic stability, is overcome by in situ salt fonnation.
the diffusion layer will be lower in CQmparison to that found with the free The selection of appropriate salt form for better dissolution rate is also
base form of the drug. Consequently, the solubility of a basic drug at this important. It has been shown that the choline and the isopropanolamine
lower pH is enhanced. Thus, if: salts of theophylline dissolve 3 to 4 times more rapidly than the
[H+]d = hydrogen ion concentration of the diffusion layer, and ethylenediamine salt and show better bioavailability.

[H+]b = hydrogen ton concentration of the bulk of the solution, then , A factor that influences the solubility of salt forms of the drug is the
size of the counter ion. Generally speaking, smaller the size of the
for salts of weak acids, [H+]d < [H+]b, and counter ion, greater the solubility of salt for example, the bioavailability
for salts of weak bases, [H+]d > [H+lb· . of novobiocin from its sodium salt, calcium salt and free acid form was
found to be in the ratio 50 : 25 : 1. Where the counter ion is very large
The increase and decrease in pH of the diffusion layer by the salts of in size and/or has poor ionic strength (as in the case of ester form of
weak acids and bases have been attributed to the buffering action of drugs), the solubility may be much lower than the free drug itself for
strong base cation and strong acid anion respectively. example, the pamoates, stearates and palmitates of weak bases have poor
Gl Lumen Gl Barrier Blood aqueous solubility. These fonns are, however, useful in several ways
Diffusion layer. Bulk of the solution. such as to prolong the duration of action (steroidal salts), to overcome bad
higher pH (5-6) rel~tively lower pH ( 1-3)
soluble form taste (chloramphenicol palmitate), to enhance GI stability (erythromycin
,...~of the drug - - - estolate) or to decrease the side effects, local or systemic.
.,.....•• diffusion of soluble Absorplion Drug in There are exceptions where the so called more soluble salt form of the
drug particles blood drug showed poor bioavailability. One such study was the comparative
....' .....·
·:;;:;:::~
Rapid
Drug in - ~--+
dissolution of sodium phenobarbital and free phenobarbital from their
•• •t ! *• ...,...

fine precipitate dissolution sqlutien tablets. Slower dissolution with sodium salt was observed and the reason
of weak acid attributed to it was that its tablet swelled but did not disintegrate and thus
Fig. 2.15 Dissolution and absorption of an acidic drug administered in a salt form
dissolved slowly. An identical result was obtained with hydrochloride
salts of several tetracycline analogs and papaverine; better dissolution and
Yet another convincing reason for enhanced solubility of salts of weak bioavailability was observed with the free bases. The reason for poor
acids is the precipitation of the drug as very fme particles. When the solubility and dissolution rate was the suppression action of the common
soluble ionic fotrn of the drug diffuses from the stagnant diffusion layer ion effect.
32 BI()PHARMACEUTICS AND PHARMACOKINETICS
ABSORPTION OF DRUGS
- -·
33
Drug pK 3 and Lipopbilicity and GJ pH pH Partition Hypothesis for weak acids, ionized drug concentration
The pH partition theory (Brodie et al.) explains _in simple terms, the pH = .pKa + l o g - - - - - - - - - - (2.1 0)
unionized drug concentration
process of drug absorption__ from the G IT and its distribution a_cross aU ,
biologic membranes. The .- theory states that for drug compounds of mo- JOPH- pKa
lecular weight greater than 100, which are primarily transpor~ed across % Drug Ionized = x 100 (2.11)
1 + 1OPH .- pKa
the biomembrane by passive diffusion, the process of absorption is gov-
erned by: for weak bases, unionized drug concentration
I. The dissociation constant (pKa) of the drug. •: pH = pKa + log ---,....- - - - - - - - - (2.12)
ionized drug concentration
2. The lipid solubility of the unionized drug (a function of drug
,. topKa- pH
Ko/w). o/o Drug lo~tzed = x 100 (2.13)
3. The pH at the absorption site. 1 + topKa- pH
Since most drugs are weak elec~rolytes (weak acids or weak base~),
I · ·- ······ ··- - · - - When the concentration of ionized and unionized drug becomes equal,
their degree of ionization depends upon the pH of the biological fluid. If the second term of equations 2.10 and 2.12 reduces to zero (since l_og I =
the pH on either side on the membrane is different, then the CO[lpartmerit zero), and thus pH = pKa. The pKa is a characteristic of the drug.
whose pH favors greater ionization of the drug will contain greater amount
I .. . ...,_. I

If there is a membrane barrier that separates the aqueous solutions of


of drug, and only the unionized or undissociated fraction of drug, · if
different pH such as the G IT \and the plasma, then the theoretical ratio R
suf~ciently lipid soluble, can permeate the membrane passively until the.
of drug concentration on either side of the membrane can · be given by
concentration of unionized drug on either side of the membrane becomes
equations derived by Shore et a/: ·
equal i.e. until equilibrium is attained.
for weak acids, ColT
1 + 1QPHGIT - pKa-
The above statement of the hypotl}esis was based on the assumptions Ra = -- (2.14)
that: Cplasn1a
1 + 1QPHplasma - pKa
1. The G IT is a simple lipoidal barrier to the transport of drug.
for weak bases, Co IT
2. Larger the fraction of unionized drug, faster the absorption. Rb = -- (2.15)
3. Greater the lipophilicity (Ko/w) of the unionized drug, better the Cplasma
absQrption.

Drug · pK8 and Gastrointestinal pH


The amount of drug that ·exists in unionized fonn is a function of
dissociation constant (pKa) of the drug and pH of the fluid at the absorp- .
For Weak Acids:
tion site.
I. Very weak acids (pKa > 8) ·such as phenytoin, ethosuximide and
It is customary to express the·dissociation constants of both acidic and
several barbiturates are essentially unionized at all pH values and
basic drugs· by pKa values. The ~ower the pKa of an acidic drug, stronger
,• therefore their absorption is rapid and independent of GI pH . .
the acid i.e. greater the proportion of ionized fonn at a particular pH. The
, higher the pKa of a __basic ~~' the . ~~~~ger the base. Thus, from th'"' . 2. Acids in the pKa range 2.5 to 7.5 are greatly affected by changes
knowledge of J?Ka of the drug and pH at tlle absorption site (or biological in pH and therefore their absorption is pH-dependent; e.g. several
flui~), th~ relative amount .of ionized and unionized drug in solution at a NSAIOs like aspirin, ibuprofen, phenylbutazone, and a number of
particular pH and the percent of drug ionized at this pH can be deter-~ penicillin analogs. Sue~ drugs are better absorbed from acidic
mined by Henderson-Hasselbach equations: conditio~s of stomach (pH < pKa) where they largely exist in
unionized foun.
'

J '

I '
34 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 35

3. · Stronger acids with pK3 < 2.5 such as cromolyn sodium are
Drugs pH/site of absorption
ionized in the entire pH range of G IT and therefore remain poorly
absorbed. Moderately weak bases (pK8 5 to 11.0)
Reserpine 6.6 Ionized at gastric pH,
·For Basic Drugs:
lleroin 7.8 relatively · unionized at intestinC&l pH;
I. Very weak bases (p'Ka < 5.0) such as caffeine, theophylline and a Codeine 8.2 · better absorbed from intestine.
number of benzodiazepines like diazepam, oxazepam and nitrazepam Amitriptyline 9.4
are essentially unionized at all pH values and therefore their Stronger bases (pK8 > 11.0)
absorption is rapid and pH-independent. Mecamylamine 11.2 Ionized at all pH values;
Guanethidine 'II. 7 poorly absorbed from GIT.
2: Bases in the pK3 range 5 to 11.0 are greatly affected by changes
in pH and hence their absorption is pH-dependent; e.g. several By using equations from 2J 0 to 2.15, one can calculate the. relative
morphine analogs, chloroquine, imipramine and amitriptyline. Such amounts of unionized (absorbable) and ionized ( unabsorbable) fonns of
drugs are better absorbed from the relatively alkaline conditions the drug and predict the extent of absorption at a given pH of GIT. An
of the intestine where they largely exist in unionized form. example of this is illustrated in Fig. 2.16.
3. Stronger bases with pKa > 11.0 like mecamylamine and guanethidine
I~ ~-Membrane Barrier---1
are ionized in the entire pH range of G IT and therefore poorly '

absorbed. Drug Stomach Plasma Intestine


pH = 1.5 pH = 7.4 pH = 5.0
A summary of above discussion is given in Table 2.3 .
Weak Acid [HA] = 100 , ~ [HA] -- 100'
h.
[HA] -- 100
e.g. Ibuprofen Jl Jl Jf'
TABLE 2.3 pKa = 4 .4 [A-] -- 0.13 [A-] - 100,000 [A-] -- 398.1
Influence of drug pK 3 and GI pH on Drug Absorption -
. [Total] - 100.13 [Total] = 100,100 [Total) - 498.1
.. Drugs pH/site of absorption
Weak Base [BOH] - 100 '\ ~ [BOH] = 100 '\
l\.
[BOH] = 100 .
Very weak acids (pK8 > 8.0) e.g . Nitrazepam Jl Jl Jl
Pentobarbital 8.1 Unionized at all pH values ~
pKa = 3.2 [8+] = 5012 [B+] -- 0.006 [B+] -- 1.6
.
Hexobarbital 8.2 absorbed along the entire length of GIT [Total] = 5112 [Total] = 100.006 [Total] = 101.6
Phenytoin 8.3
Ethosuximide 9.3 '. Fig. 2.16 Influence of pH on ionization of drug. [HA] and [BOH] are

Moderately weak acids (pK8 2.5 to 7.5) concentration of unionized acid and base, and [A·] and [B•]
Cloxacillin 2. 7 Unionized in gastric pH and are concentration of ionized acid and base respectively
Aspirin 3.. 5 ionized in intestinal pH; better
Lipophilicity and Drug Absorption
Ibuprofen 4.4 absorbed from stomach
Phenylbutazone 4.5 As mentioned earlier, it is the pK 3 of a drug that deteunines the
degree of ionization at a particular pH and that only the unionized drug, if
Stronger acids (pK8 < 2.5)
sufficiently lipid soluble, is absorbed into the systemic circulation. Thus,
Disodium cromoglycate 2.0 Ionized at all pH values;
even if the drug exists in the unionized fotlll, it will be poorly absorbed if
poorly absorbed from GIT.
it has poor lipid solubility (or low Kotw). Ideally, for optimum absorp-
Very weak bases (pK8 < 5.0)
Theophylline . 0. 7
tion, a drug should have sufficient aqueous solubility to dissolve in the
Unionized at all pH values;
Caffeine 0.8 absorbed along the entire length of G IT.
fluids at the absorption site and lipid solubility (l<otw) hi~~ . enough to
Oxazepam I. 7 facilitate the partitioning of the drug in the lipoidal biomembrane and . into
Diazepam 3. 7 the systemic circulation. In other words, a perfect hydrophilic -lipophilic
I
balance (HLB) should be there in the structure of the drug for optimum
, .. .continued
. .
, bioavailability.


BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 37

The lipid solubility of a drug is determined from its oil/water partition I. Presence of virtual membrane pH
coefficient {K0 ;w) value. This value is a measure of the degree of 2. Absorption of ionized drug
distribution of drug between one of the several organic, water immiscible,
3. Influence of G I surface area and residence time of drug
lipophilic solvents such as n-octanol, chlorofonn, n-heptane, etc. and an
4. Presence of aqueous unstirred diffusion layer
aqueous phase (water or a suitable buffer). In general, the octanollpH 7.4
'
buffer partition coefficient value in the range of I to 2 of a drug is 1. Presence of Virtual Membrane pH : The pH-partition hypoth-
sufficient for passive absorption across lipoidal membranes. . A direct esis suggested that only the unionized drug at a given GI lumen pH is
correlation between a drug's K0 ;w and extent of absorption is illustrated in absorbed. An S-shaped curve, called as the pH-absorption curve denot-
Table 2.4. ing the dissociation of drug, is obtained when pH is plotted versus rate of
drug absorption (Fig. 2.17).
TABLE 2.4
Comparison between Intestinal Absorption of Some Drugs through the
Rat Intestine and K 0 ;w of the Ionized Form of the Drugs ', I
/

Drugs Kheptane/water %Absorbed


c
·-0.
0 ' \\ Acidic Basic 1
I1
~

'- \ drugs drugs


Rapid rate · of absorption
0
f/) \
I .0 \ I
<t:
Phenylbutazone 100.0 54 0) \ I
Thiopental 3.3 67 :::s
'-
\ I
0 \
Benzoic acid 0.19 54 ._ \ I
0
\ I
Sal icy Iic acid 0.12 60
-a::
Cl)
ro \ I
Moderate rate of absorption
Aspirin 0.03 21 i
Theophylline 0.02 30
Theobromine < 0.002 22 ____..~ pH of Gl lumen
Sulfanilamide < 0.002 24 Fig . 2.17 pH-absorption curve for acidic and basic drugs. Dotted lines
Slow rate of absorption indicate curves predicted by pl-I-partition hypothesis
Barbituric acid < 0.002 5 and bold lines indicate th~ practical curves.
I
Sulfaguanidine < 0.002 2
' Ho\\ever, differences in the extent of absorption of salicylic acid has
Source: Schanker, J. Med. Pharm., 2, 343 ( 1960). been observed at a given GI pH than that predicted by pH-partition
hypothesis. The experimental pH-absorption curves are tess steep and
In yet another study by Schanker on a series of barbituric acid deriva-
shift to the left (lower pH values) for a basic drug and to the right (higher
tives havmg .s ame pKa, the percent absorbed increased with an increase in
pH values) for an acidic drug. This led to the suggestion that a virtual
the partition coefficient of the drug. Thus, to enhance the bioavailability
pH, also called as the microclimate pH, different from the lumenal pH
of a drug, not only its dissolution rate but also its rate of penn.eability
exists at the membrane surface. This virtual membrane pH actually
should be considered. Rate of dissolution can be increased by altering the
determines the extent of drug ionization and thus, drug absorption.
· physical properties such as particle size or crystalline structure but its

pet rneability can only be promoted by modification of chemical structure 2. Absorption of Ionized Drugs : An important assumption of the
.·- (see chapter 6 on prodrugs ). theory was that only unionized form of the drug is absorbed and pettne-
ation of the ionized drug is negligible since its rate of absorption is 3 to 4
Limitations of pH-Partition Hypothesis times less than that of unionized drug. This is true to a large extent as
The pH-partition hypothesis over-simplified the otherwise complicated ionized drugs have low lipid solubility and relatively poor penneability.
process of drug absorption and therefore has its own limitations. Some of However, the pH-absorption curve shift suggested that ionized fonns of
the deviations fro1n the theory are: some drugs also get absorbed to a considerable extent. If such drugs have
'
33. BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 39
a large lipophili~ group in their structure, despite their ionization, they Thus, drugs having large partition coefficient can rapidly penetrate the
will be absorbed passively for example, morphinan derivatives. Other lipid membrane but diffusion through unstirred water layer is the rate-
me~hanisms are also involved in the absorption of ionized drugs such as limiting step in their absorption. This applies in particular to high molecular ·
. '

-activ.e transport, ion-pair transport and convective flow. weight fatty acids and bile acids.
~-
Influence of GI Surface Area and Residence T~me of Drug : Despite its limitations, the pH-partition theory is still useful in the
. .
According to the pH-partition theory, acidic drugs are best ·absorbed from basic understanding of drug absorption and movement of drug between
stomach (acidic pH) and basic drugs from intestine (alkaline pH) in which various body compartlnents.
.
conditions they are unionized to a large extent. This could be true under
• Drug Stability
. conditions where the surface area of stqmach and intestine are same. It
could also ·m ean that once an acidic · drug reaches the intestine, the remain- A drug for oral use may destabilize either during its shelf-life or in the
. ing fractjon will -be poorly absorbed and that unless a basic drug reaches GIT. Two major stability problems resulting in poor :gioavailability of an
.
, ,~he intestine and gets absorbed :considerably, it may not be able to attain orally administered drug are degradation of the drug into inactive fonn,
'its therapeutic level. But, irrespective of the GI pH and the degree of and interaction with one or more different compodent(s} either of the ,
ionization, both acidic and .b~sic drugs are more rapidly absorbed from the dosage fonn or those present in the GIT to fonn a complex that is poorly
intestine, primari~y· because of its large surface area and secondly, because soluble or is unabsorbable. Destabilization of a drug during its -shelf-life
of long residenfe time of ~he drug in the intestine. and in the GIT will be discussed in detail under formulation factors and
!
patient related factors respectively.
,. 4 . . -Pre~ence of Aqueous Unstirred Diffusion Layer : The pH-shift
in th~ absorption of acidic and basic drugs, as discussed earlier, also
·.accounts· for the fact that the bulk of the lumenal fluid is not in direct DOSAGE FORM FACTORS AFFECTING DRUG ABSORPTION
contact· with the membrane but a barrier called as aqueous unst irred 1
Disintegration Time
. ·, diffusi'orllayer is interposed between them. Such a model is depicted in Disintegration time (DT) is of particular importance in case of solid
Fig. 2.18. dosage fonns like .tablets and capsules. In vitro disintegration test Is by
no means a guarantee of drug's bioavailability because if the disintegrated
~- Aqueous bulk fluid
drug particles do not dissolve, absorption is not possible. However, if a
of the GIT solid dosage fonn does not conform to the DT, it portends bioavailability
_ _ Aqueous unstirred
problems because the subsequent process of dissolution will be much
diffusion layer slower and absorption may be insufficient. Coated tablets, especially

sugar coated ones have long DT. Rapid disintegration is thus important in ·
L
the therapeutic s~ccess of a solid dosage fotrn. DT of a tablet is directly
Drug related to the amount of binder present and the compression force (hard-
molecule _ _ Lipoidal biomembrane
ness) of a tablet. A harder tablet with large amount of binder has a long
_ _ _ _ Blood DT. Disintegration can be aided by incorporating disintegrants in suitable
amounts during fonnulation.
After disintegration of a solid dosage fonn into granules, the granul~
Fig. 2.18 Presence of aqueous unstirred diffusion layer on the membrane surfac~ must deaggregate into fme particles as dissolution from such tiny particles '
'
is faster than that frpm granules.
Such a layer has a real thickness and is .a barrier to absorption of
drugs. In the original pH-partition theory, the rate-limiting step in . 'th~ Manufacturing/Processing Variables
' I

· absorption of drugs was the partitioning in the lipid barrier. With the · Drug dissolution is the single most important factor in the absorption
. incorporation of unstirred aqueous diffusion layer, a drug must diffuse of drugs, especially from the most widely used conventional solid dosage
· frrst through this aqueous barrier and then through the lipoidal barrier. fonns, tablets and capsules. The dosage fonn related factors that influ- ...
40 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 41

ence dissolution and hence absorption of a drug from such formulations vent into the tablet, retards wettability by forming a firmer and more
are: effective sealing layer by the lubricant, and in many cases, promotes
tighter bonding between the particles, all of which result in slowing of the
1. Excipients (fonnulation ingredients apart from the active prin-
ciples), and
El c: A. B. C. D.
2. Manufacturing processes. .-1

-
..... 0
0 ·-
The influence of excipients such as binders, lubricants, disintegrants, '0.2
etc. on drug dissolution will be discussed in the subsequent section of this . -
0.) ~
( t l ,(/)
~0
_

chapter.

Several manufacturing processes influence drug dissolution from solid


dosage forms. Processes of such importance in the manufacture of tablets -4~ Compression Force
are: Fig. 2.19 Influence of compression force on dissolution rate of tablets
1. Method of granulation, and dissolution rate of tablets (curve A of Fig. 2.19). On the other hand,
2. Compression force. higher compression forces cause deformation, crushing or fracture of drug
The processing factor of importance in the manufacture of capsules particles into smaller ones or convert a spherical granule into a disc
that can influence its dissolution is the intensity of packing of capsule shaped particle with a large increase in the effective surface area. This
contents. results in an increase in the dissolution rate of the tablet (curve B of Fig.
2.19). A combination of both the curves A and B is also possible as
Method of Granulation : The wet granulation process is the most shown in curves C and D. In short, the influence of compression force on
conventional technique in the manufacture of tablets and was once thought . the dissolution rate is difficult to predict and a thorough study on each
to yield tablets that dissolve faster than those made by other granulation formulation should be made to ensure better dissolution and bioavailability.
methods. The limitations of this method include-(i) formation of crystal
bridge by the presence of liquid, (ii) the liquid may act as a medium for Intensity of Packing of Capsule Contents : Like the compression
affecting chemical reactions such as hydrolysis and (iii) the drying step force for tablets, packing density in case of capsule dosage form can
may halllL_the thennolabile drugs. The method also involves a large either inhibit or promote dissolution. Diffusion of GI fluids into the
number of steps each of- which can influence drua dissolution method tightly filled capsules creates a high pressure within the capsule resulting
- - 0
and duration of blending, method, time and temperature of drying, etc. in rapid bursting and dissolution of contents. Opposite is also possible. It
The method of direct compression has been utilized to yield tablets that has been shown that capsules with finer particles and intense packing have
dissolve at a faster rate. One of the more recent methods that has resulted poor drug release and dissolution rate due to a decrease in pore size of the
in superior product is agglomerative phase of communition (APOC). compact and poor penetrability by the GI fluids.
The process involves grinding of drugs in a ball mill for time long enough
Pharmaceutic Ingredients/Excipients (Formulation factors)
to affect spontaneous agglomeration. The tablets so produced were stron-
A drug is rarely administered in its original form. Almost always, a
ger and showed rapid rate of dissolution in comparison to tablets made by
convenient dosage form to be administered by a suitable route is prepared.
other methods. The reason attributed to it was an increase in the internal
surface area of the granules prepared by APOC method. Such a formulation contains a number of excipients (non-drug compo-
nents of a formulation). Excipients are added to ensure acceptability,
Compression Force : The compression force employed in tableting physicochemical stability during the shelf-life, unifotrnity of composition
process influence density, porosity, hardness, disintegration time and dis- and dosage, and optimum bioavailability and functionability of the drug

solution of tablets. The curve obtained by plotting compression force product. Despite their inertness and utility in the dosage form, excipients
versus rate of dissolution can take one of the 4 possible shapes shown in can influence absorption of drugs. The m9re the number of excipients in
Fig. 2.19. a dosage fonn, the more complex it is and greater the potential for
On the one hand, higher compression force increases the density. and absorption and bioavailability problems. Commonly used excipients in
hardness of tablet, decreases porosity and hence penetrability of the sol-
42 BIOPHARMACEUTICS AND PHARMACOKINE~'ICS ABSORPTION 'OF DRUGS 43

various dosage forms are vehicles, diluents (fillers), binders and grc:~r ulat- · hydrophilic (aqueous) binders show better dissolution profile with poorly
ing agents, disintegrants, lubricants, coatings, suspending agents, emclsifiers, wettable drugs like phenacetin by imparting hydrophilic properties to the
surfactants, buffers, complexing agents, colorants, sweeteners, crystal growth granule surface. However, the proportion of strong binders in the tablet
inhibitors, etc. formulation is very critical. Large amounts of such binders increase ·
hardness and ·decrease disintegration/dissolution rates of tablets. PEG
Vehicle : Vehicle or solvent system is the major' component of liquid 6000 was found to be a deleterious binder for phenobarbital as it forms a
orals and parenterals. The 3 categories of vehicles in use are aqueous poorJy soluble complex with th~ ·drug. Non-aqueous binders like ethyl
vehicles (water, syrup, etc.), nonaqueous water miscible vehicles (pro- ·
cellulose also retard drug dissolution.
l pylene glycol, glycerol, sorbitol) and nonaqueous water immiscible vehicles
(vegetable oils). Bioavailability of a drug from vehicles depend to a large Disintegrants : These agents overcome· the cohesive strength of tablet
· extent on its miscibility with biological fluids. Aqueous and w.ater mis- and break them·' up on contact with water which is an important prerequi-
cible vehicles are miscible with the body fluids and drugs from them are site. to tablet dissolution.
.
Almo.st all the disintegrants are hydrophilic in
rapi~ly absorbed. Quite often, a drug is more soluble in water miscible nature. A decrease in the amount of disintegrant can significantly lower
,vehicles like propylene glycol (serving as a co-solvent) and show better bioavailability. Adsorbing disin~e~rants like bentonite and veegum should
' ·bioavailability. Sometimes dilution of such vehicles with the body fluids be avoided with low dose drugs like digoxin, alkaloids and steroids since
results in precipitation of drug as fine particles which, ho~ever, dissolve a large amount of dose is permanently adso~b,e~ a_!lj. . ~J?.lY _c:_!taction is
rapidly. Solubilizers such as tween 80 are sometimes used to promote available for absofption. Microcl)':stalline cellulose is a very g?od disintegran
solubility of a drug in aqueous vehicles. In case of water. immiscible . (and a binder too) but at high compression forces, it may retard drug
vehicles, the rate of drug absorption depends upon its partitioning from dissolution.
the oil phase to the aqueous body ·fluids, whifh could be a rate-limiting
Lubricants/Antifrictional Agents : These agents are added to tablet _
step. Viscosity of the vehicles is another factor in the absorption of
rormulations to aid flow of granules, to reduce iriterparticle friction ant:
drugs. Diffusion into the bulk of GI fluids and thus absorption of a drug
sticking or adhesion of particles to dies and punches. The commonly
from a viscous vehicle may be slower.
used lubricants are hydrophobic in nature (several metallic stearates and
Diluents (Fillers) : Diluents are commonly added to tablet (and cap- waxes) . and known to inhibit wettability, penetration · of water into tablet ·
sule) fonnulations if the required dose is inadequate to produce the necessary and their disintegration and dissolution. This is because the disintegrant
bulk. A diluent may be organic or inorganic. Among organic diluents, gets coated with the lubricant if blended simultaneously which however
carbohydrates are very widely used for example, starch, lactose, micro- can be ·prevented by adding the lubricant in the final stage. The best
crystalline cellulose, etc. These hydrophilic powders are very useful in alternative is use of soluble lubricants like SLS and carbowaxes which
promoting the dissolution of peorly water-soluble, hydrophobic drugs like promote drug dissolution.
spironolactone and triamter,ene by fonning a coat onto the hydrophobic
Coatings : In geu_·eraJ, the deleterious effect of various coatings on
surface of drug particles and rendering them hydrophilic. Amo_ng the
drug dissolution from a ·tabiet dosage fonn is in the following order:
inorganic diluents, dicalcium phosphate (DCP) is · most commo . One
enteric coat > sugar coat > nonenteric film coat. The dissolution profile
cl ~ssic example of drug-d~luent interaction resulting in poor bioav ilability
of certain coating materials change on aging; e.g. shellac coated tablets,
is that of tetracycline and DCP. The cause is formation of divalent I '•

on prolonged storage, dissolve more slowly in the intestine. This can,


calcium-tetracycline complex which is poorly soluble and thus, unab orbable.
however, be prevented by incorporating little PVP in the coating formulation ..
Binders and Granulating Agents : These materials are used Ito hold
Suspending AgentsNiscosity Imparters : Popular suspending agents
powders together to form . granules or promote cohesive compacts for •
are hydrophilic polymers like vegetable gums (acacia, tragacanth, etc.),.~ ·
directly compressible inaterials and to ensure that the tablet remains intact
semisynthetic gums (CMC, MG) and synthetic gums which primarily
after compression. Popular binders include polymeric materials (natural,
semisynthetic and synthetic) like starch, cellulose derivatives, acacia, PVP, I
stabilize the solid drug particles by reducing their rate of settling throug~
an increase in ·the vi~cosity of the medium. These agents and some
etc. Others include gelatin and sugar solution. In general, like fillers, the
I
'
44 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 45
I

sugars are also used as viscosity imparters to affect palatability and pourability Complexing Agents : Complex formation has been used to alter the
of solution dosage fonns. Such agents can influence drug absorption in physicochemical and biophannaceutical properties of a drug. A complexed
"evera1 ways. The macromolecular gums often fonn unabsorbable com- drug may have altered stability, solubility, molecular size, partition coeffi-
~texes with drugs for example, sodium CMC forms a poorly soluble cient and diffusion coefficient. Basically, such con1plexes are
complex with amphetamine. An increase in viscosity by these agents pharmacologically inert and must dissociate either at the absorption site or
acts as a mechanical barrier to the diffusion of drug from the dosage fo11n following absorption into the systemic circulation. Several examples
into the bulk of GI fluids .and from GI fluids to the mucosal lining by where complexation has been used to enhance drug bioavailability are:
'
fonning a viscid layer on the G I mucosa. They also retard the GI transit · 1. Enhanced \dissolution through fonnation of a soluble complex e.g.
of drugs. ,
ergotamine tartarate-caffeine complex and hydroquinone-digoxin
Surfactants : Surfactants are widely used in fonnulations as wetting complex,
agents, solubilizers, emulsifiers, etc. Their influence on drug absorption is 2. Enhanced lipophilicity for better membrane. permeability e.g. caf-
very complex. They may enhance or retard drug absorption either by feine-PABA complex, and
interacting with the drug or the membrane or both. Mechanisms involved 3. Enhanced membrane penneability e.g. enhanced GI absorption of
in the increased absorpti~n of drug by use of surfactants include: heparin (normally not absorbed from the G IT) in presence of
1. Promotion of wetting (through increase in effective surface area) EDTA which chelates calcium and magnesium ions of the mem-
and dissolution of drugs e.g. tween 80 with phenacetin brane.
2 . Better membrane contact of the drug for absorption Complexation can be deleterious to drug absorption due to fonnation
3. Enhanced membrane penneability of the drug of poorly soluble or poorly absorbable complex e.g. complexation of
tetracycline with divalent and trivalent cations like calcium (milk, antac-
The beneficial effects of surfactants have beer, observed at pre-critical
ids), iron (hematinics), magnesium (antacids) and aluminium (antacids).
micelle concentration levels. However, physiologic surfactants like the
Reasons for poor bioavailability of some complexes are failure to dissoci-
bile sal~~ (anionic) and lysolecithin (nonionic) promote absorption of hy-
ate at the absorption site and large molecular size of the complex that
drophobic drugs like steroids, oil soluble vitamins and griseofulvin by
cannot diffuse through the cell membrane for example, drug-protein complex.
their micellar solubilizing property.
Decreased absorption of drug in the presence of surfactants has been Colorants : Even a very low concentration of water-soluble dye can
suggested to be due to: have an inhibitory effect on dissolution rate of several crystalline drugs.
The dye molecules get adsorbed onto the crystal faces and inhibit drug
1. Fonnation of unabsorbable drug-micelle complex at surfactant
dissolution for example, brilliant blue retards dissolution of sulfathiazole.
concentrations above critical micelle concentration
Dyes have also been found to inhibit micellar solubilization effect of bile
2. Laxative action induced by a large surfactant concentration acids w.hich may impair the absorption of hydrophobic drugs like steroids.
Buffers : Buffers are sometimes useful in creating the right atmo- Cationic dyes are more reactive than the anionic ones due to their greater
sphere for drug dissolution as was observed for buffered aspirin tablets. power for adsorption on primary particles.
However, certain buffer systems containing potassium cations inhibit the Crystal Growth Inhibitors : In addition to maintaining the initial
drug absorption as seen with vitamin B2 and sulfanilamide.· The reason phys~cal properties of a drug in suspension; crystal growth inhibitors like
attributed to it was the uptake of fluids by the intestinal epithelial cells PVP and PEG inhibit conversion of a high energy metastable polymorph
due to which the effective drug concentration in the tissue is reduced and into stable, less soluble polymorph.
the absorption rate is .decreased. Such an inhibitory effect of the various
buffer cations on the drug transfer rate is in the fol1owing order: K+ > Nature and Type of Dosage Form
NH4 + > Li+ > Na+ > TRIS+. Hence, the buffer system for a salt of a Apart from the proper selection of drug, clinical success often depends
drug should contain the same cation as the drug salt and introduce no to a great extent on the proper selection of dosage fonn of that drug. For
additional cations. a given drug, a 2 to 5 fold or perhaps more difference could be observed
46 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 47
in the oral bioavailability of a drug depending upon the nature and type of . Several factors, especially the excipients which influence bioavailability
dosage fonn. Such a difference is due to the relative rate at which a of a drug from its dosage fottn, have been discussed earlier.
particular dosage fonn releases the drug to the biological fluids and the
membrane. The relative rate at which a drug from a dosage fonn is Solutions : A drug in a solution (syrups, _elixirs, etc.) is most rapidly
presented to the body depends upon the complexity of dosage fonn. The absorbed since the major rate-limiting step, drug dissolution~ is absent.
more complex a dosage fottn, greater the number of rate-limiting steps Factors that influence bioavailability of a drug from solution dosage form
and greater the potential for bioavailability problems. include the nature of solvent (aqueous, water miscible, etc.), viscosity,
- surfactants, solubilizers, stabilizers, etc. Quite often, dilution of a drug in
Slowest solution with GI fluids results in precipitation of drug as fme particles
Disintegration
which generally dissolve rapidly. Factors that limit the fonnulation of a
- TABLETS
drug in solution fonn include stability, solubility, taste, cost of the prod-
uct, etc.
Dissolution
CAPSULES --------~-~ GRANULES Emulsions-: Emulsion dosage fonns have been found to be superior to
of s/Je/1
suspensions in administering poorly aqueous soluble lipophilic drugs. It
was observed with indoxole (an NSAID) that when it_is dissolved in a
z POWDERS
Deaggregation
vegetable oil and emulsified in water, absorption _increases 3 fold over its
0 {COARSE)
-
)-
I

aqueous suspension. Emulsion dosage fonn present a large surface area


n.
cr: of oil to the GIT for absorption of a drug. Scientists have claimed that ~
0
drug administered in oily vehicle (emulsified and solubilized in the GIT

(/)
0)
SUSPENSIONS FINE PARTICLES
~ by bile salts to form mixed micelles) can direct the distribution of drug
directly into the lymphatic system thereby avoiding the hepatic ROrtal vein
and first-pass metabolism.
EMULSIONS ----·~ DISSOLUTION
Suspensions : The major rate-limiting step in the absorption of a drug
from suspension dosage form is drug dissolution which is generally rapid
due to the large surface area of the particles. ·Important factors in the
SOLUTIONS ----•• DRUG IN SOLUTION bioavailability of a drug from suspensions include particle size, -polymor-
Fastest phism, wetting agents, viscosity of the medium, suspending agents, etc.
Biomembrane Absorption
Powders : Though powders are superior to tablets and capsules, they
DRUG IN BLOOD . are not in use nowadays due to handling and palatability problems. Major
factors to be considered in the absorption of a drug from powders are
Fig. 2.20 Course of events that occur following oral particle size, polymorphism, wettability, etc.
administration of various dosage forms
Capsules : Powders and granules are popularly administered in hard
The rate at which a particular dosage fonn releases the drug following
gelatin capsules whereas viscous fluids and oils in soft elastic shells.
administration is given in Fig. 2.20.
Factors of importance in case of hard gelatin capsules include drug p~r­
.__!.\~~~
~~n~ral rule~ th_e bi_QavailabiJltr £.>[_a ._flr_:ug from various dosage ticle size, density, polymorphism, intensity of packing and influence of
forrns decreases in the followin_g order :__S~Jlftiof!~ >.-c.._ -'l:..mul"".sj_f!n~ _>- Suspen- diluents and excipients. Hydrophilic diluents like lactose improve wettability,
sions > Capsules > Tablets > Coated Tablets > Enteric Coated 'J~blets > · deaggregation and dispersion of poorly aqueous soluble drugs whereas
Sustained Release Products. Thus, absorption of a drug from solution is inhibitory effect is observed with hydrophobic lubricants like magnesium
fastest with least potential for bioavailability problems whereas absorption stearate. A hydrophobic drug with a fine particle ~ize in capsule r~~~ts in
from a sustained release product is slowest with greatest bioavailability a decrease in porosity of powder bed and thus, decreased penetrability b~.
risk . - ,..,... -- - . ~ ............. - -

.,
ABSORPTION OF DRUGS
49
48 BIOPHARMACEUTICS AND PHARMACOKINETICS '
J
I

sugar coat which though soluble, is g~~ra~ly tough and takes longer ~o ·
the solvent with the result that clumping of particle oecurs. This can be
tissolve. The sealing coat which is generally .of shellac, is most d~letert­
overcome by incorporating a large amount of hydrophilic diluent (up to
JUS. Press coated tablets may be superior to sugar coated tablets tn such
50%), a small amount of wetting agent cutn lubricant such as SLS (upto
1%) and/or by wet granulation to convert an impenneable powder bed to cases.
the one having good permeability. Other factors of importance include
possible interaction between the drug and the diluent (e.g. tetracycline-
DCP) and between drug and gelatin shell. The influence of capsule Disintegration
.._,__ _
GRANULES
____.'
~

Deaggregation
processing factors on drug dissolution and bioavai lability have already
been discussed.
/ ~
COMPRESSED FINE PARTICLES
Soft elastic capsules as such dissolve faster than hard gelatin capsules
and tablets and show better drug availability from oily solutions, emul-
TABLET
'------------
I Dissolution
sions or suspensions of medicaments (especially hydrophobic drugs). One
of the best examples of this is the faster dissolution of indoxole (equiva- ~ l .t
lent to that of an emulsion dosage form) when fonnulated as softgel in DRUG IN SOLUTION
comparison to hard gelatin capsule and aqueous suspension. Such poorly
soluble drugs can be dissolved in PEG or other suitable solvent with the Biomembrane
aid of surfactants and-encapsulated without difficulty. Softgels are thus of
particular use where the drug dose is low, drug is lipophilic or when oily Absorption
or lipid based medicaments are to be administered. A problem with
softgels is the high water content of the shell (above 20%). This moisture DRUG IN THE BODY
migrates into the shell content and crystallization of drug occurs during
the drying stage resulting in altered drug dissolution characteristics. Fig. 2.2 t Sequence of events in the absorption of a drug from tablet dosage form

-
Tablets : Compressed tablets are the most widely used convenienct
'
Enteric Coated Tablets : Enteric coated tablets have great potential in
and cost effective dosage forms. A schetnatic repr~sentation of disintegra- creating bioavailability problems because the c·1at dissolves only in the
tion, deaggregation, dissolution and absorption of a drug from a tablet alkaline pH of the intestine and it may take as long as 2 to 4 hours for
dosage fonn is shown in Fig. 2.21 . such a tablet to empty from the stomach into the intestine depending upon
The bioavailability problems with tablets arise from the reduction in the 'meals and the QI motility. Hence, the phaunacologic response may
the effective surface area due to granulation and subsequent compression eventually be delayed by as much as 6 to 8 hours. The problem of gastric
into a dosage form. Since dissolution is most rapid from primary drug emptying can, however, be overcome by enteric coating the granules or
particles due to their large surface area, disintegration of a tablet into pellets and presenting them in a capsule or compressing in~o a table~. The
granules and subsequent deaggregation of granules into fine particles is thickness of enteric coat is yet another deterrninant factor tn drug dtssolu- ,
very important. A number of formulation and processing factors influenc- ti.on, increasing thickness being more problematic. Aging of the dosage
ing these steps and also the physicochemical properties of drug substance for 111 also affects drug release, especially with shellac. In one of the
that intluence bioavailabthty have already been discussed in the earlier studies, shellac coated tablets of salicylic acid stored for 2 years showed a
sections of this chapter. 60o/o decrease in the peak plasma level.

Coated Tablets : Tn addition to factors that influence drue: releas~


• & - - -
Sustained Release Products : Drug release from such products is
from compressed tablets, the coating 4cts as yet another barrier which most unpredictable, the problems ranging from dose dumping to unsatis-
must first dissolve or disrupt to_give way to disint~gration and dissolution factory or no drug release at all. However, with the de~elopm~t of
of tablet. Of the two types of coatings, the film coat, which is thin, newer innovations and technologies, it is becoming increasingly rehable
- ---
dtsso1ves rapidly and does not significantly affect drug ;}bsorption. 1'he and the results reproducible with little intersubject variations.
50 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 51

Product Age and Storag~ ~~nditi_!?~_s ______ pH aids dissolution of basic drugs due to salt fonnation and subsequent
A number of changes, especially in the physicochemical properties ionization which are therefore absorbed to a lesser extent from stomach
of a drug in dosage form, can result due to aging and alterations in because of the same reason.
1
storage conditions which can adversely affect bioavailao-ility. With solu-
tion dosage fonn, precipitation of drug due to altered solubility, especially
-due. to conversion of metastable into poorly soluble, stable polymorph can
'
- occur during the shelf-life of the product. Changes in particle size ·
· distribution have been observed with a number of suspension dosage
1·, for1~1s resulting in decreas_
ed rate of drug dissolution and absorption. In Mouth : pH 6.8, small
case of solid ·dosage fonns, especially t~blets, disintegration and dissolu- surface area; Lipophilic,
~~~ - - neutral and basic drugs
tion rates are greatly affected due to aging and storage conditions. An
absorbed directly into
increase in these parameters of tablets has been attributed to excipients the systemic circulation .
that harden on storage (e.g. PVP, acacia, etc.) while the decrease is mainly
due to softening/crumbling of the binder _during storage (e.g. CMC). Drug absorbed
Liver : Major site into systemic
Changes that occur during the shelf-life of a dosage fonn are affected
I , for drug metabol- circulation Stomach : pH 1-3, not
mainly by large variations in 'temperature and humidity. In one of the ism (including first- too large a surface area;
studies conducted on prednisone tablets containing lactose as the filler, pass metabolism) ~------ - - -...... _ Lipophilic, neutral and
acidic drugs absorbed
high temperature and high humidity resulted in harder tablets that disinte-
but lesser than that from
grated and dissolved slowly. intestine
Hepatic
\
portal
PATIENT RELATED FACTORS AFFECTING DRUG ABSORPTION vein
Small lnstestine : pH 5-
Before dealing with the patient related factors influencing bioavailability 7.5, very large _surface
·1 of a drug, the anatomy and physiology of the gastrointestinal tract will be '---1---L----,_ _ _ area, major site for
discussed briefly. absorption of all types of
drugs (lipophilic, neutral,
Gastrointestinal tract Bile : pH 7 .8-8.6, acidic or basic)
aids absorption of
The gastrointestinal tract {GIT) comprises of a number of components,
lipophilic drugs~
their pri_mary function being secretion, digestion and absorption. The route for active Large Intestine : pH
mean length of the entire GIT is 450 1 Cm. The major functional compo- secretion of polar 7 .9-8.0, small surface
drugs/metabolites ----area, all types of drugs
nents of the GIT are stomach, small intestine (duodenum, Jejunum and
(enterohepatic are absorbed but to a
ileum) and large intestine (colon) which grossly differ from each other in lesser extent.
. ' cycling)
ter n1s of anatomy, function, ~ecreti9ns and pH (Fig. 2.22 and Table 2.5).
The entire length of the G I mucosa from stomach to. lar~e _int~s~ine is
lined by a thin layer of mucopolysaccharides (mucus/mucin} which ··ii~r- . ...
L..Jr-- Rectum : pH 7.5-8. 0, much
tnally acts as · an impetrneable barrier to the particulates su~~ _ a~~ Daeretia: ~ '\·. smaller surfa~\ area, all types of
cells or food particles. •• · . .. ••. _f- _ _, · _; drugs are ab~'&tbed, abo.ut half of
". .,. · ,.. the absorbed drug goes directly
Stomach : The ·stomach is a bag like structure having -··a -smootlt." ·
"f • • • into the systemic circulation.
mucosa and thus small surface area. Its acidic pH, due . t6 . s_¢.~t~~ion of
t{Cl, favors absorption· of acidic drugs if they are ·soluble in the gastric Fig.
.. 2 .22 Schematic representation of the GIT and
I .. .

fluids since they are unionized to a large extent in such a pH. The gastric different sites of drug absorption
..
, ,
52 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRl.JGS 53 .

TABLE 2.5 Anatomical and Functional Differences Between is several times that of stomach. The blood flow to the small intestine is
the Important Regions of the GIT 6 to 10 times that of stomach. Moreover, the pH range of 5 to 7.5 is
most favorable for most drugs to remain unionized. The p,eristaltic move-.
Stomach Small
.
Large Rectum ment of intestine is slow, transit time is long, and penneability is high .
Intestine Intestine Thus, a contribution of all the above factors make intestine the best site
pH range 1-3 5-7.5 7.9-8.0 7.5-8.0 for absorption of tnost drugs.
Length (ems) '
20 285 110 20 Large Intestine : Its length and mucosal surface area is very small
Diameter (ems) 15 2.5 5 2.5 (villi and microvilli are absent) in comparison to small intestine and thus
Surface area (sq.M) 0.1-0.2 200 0.15 0.02 absorption of drugs from this region is insignificant. Its contents are
Blood flow (L/min) 0.15 1.0 0.02
neutral or alkaline. The main role of large intestine is in the absorption of
..
water and electrolytes. However, because of the long residence time (6 to
Transit time (hours) 1-5 3-6 6-12 6-12
12 hours), colonic transit may be important in the absorption of some
Absorptive role Iipophil ic, all types some drugs, all types poorly soluble drugs and sustained release dosage fonns.
acidic and of drugs water and of drugs
neutral drugs electrolytes Some of the Patient related factors that influence drug absorption are
. . . discussed below.
Absorptive passive all absorption passtve passive
mechanisms diffusion, mechanisms diffusion, diffusion, Age
convective convective convective
In infants; the gastric pH is high and intestinal surface and blood flow
transport transport transport.
to the GIT is low resulting in altered absorption pattern in comparison to
endocytosis
adults. In elderly persons, causes of impaired drug absorption include
Small Intestine : It is the major site for absorption of most drugs due altered gastric emptying, decreased intestinal surface area and GI blood
to its large surface area. The folds in the intestinal mucosa, called as the flow, higher incidents of achlorhydria and bacterial overgrowth in small
folds of ·Kerckring, result in 3 fold increase in the surface area. The intestine.
surface of these folds possess finger like projections called as villi which
Gastric Emptying
increase the surface area 30 times. From the surface of villi protrude
several microvilli (about 600 from each absorptive cell that lines the villi) Apart from dissolution of a drug and its penne~tion through the ~
resulting i~ 600 times increase in the surface area (Fig. 2.23). bipmembrane, the passage from stomach to the small intestine, called as
gastric emptying, can also be a rate-limiting step in drug absorption
because the major site of druo absorption is. intestine. Thus, generally
speaking, rapid gastric emptyin~ increases bioavailability of a drug .
..

Rapid gastric emptying is advisable where:

Small intestine
1. A rapid onset of action is desired e.g. sedatives
--
as a cylinder Folds of Kerckring Villi Microvilli 2. Dissolution of drug occurs in the intestine e.g. enteric coated ·
~
t,
" P.

Surface Area (m 2
) 0.33 1 10
~

.. 200 "" dosage forms

Relative increase 1 3
;.
30 600 3. The drugs are not stable in the gastric fluids e.g. penicillin G and
•'
in surface area erythromycin
. ' .
.
Fig. 2.23 Representation of the cotnponents of intestinal 4. The drug is best absorbed from the 9istal part of the small intes-
epithelium that accounts for its large surface area tine e.g. vitamin B 12
All these, combined with the great length of small intestine (approxi- For better drug dissolution and absorptio~, the gastric emptying can be·
mately 285 ems), result in more than 200 square meters of surface which promoted by taking the drug on empty stomach. Since gastric e_mptying
54 BIOPHARMACEt)TICS AND PHARMACOKINETICS
ABSORPTION OF DRUGS 55
·- is altered by several factors due to which large intersubject variations are
2. Composition of meal : Predictably, the rate of gastric emptyin.g
· observed, all biophannaceutic studies that require the drug to be taken
for various food materials is in the following order: carbohydrates. .
orally, are performed in volunteers on empty stomach.
> proteins > fats. Fats promote secretion of bile whic? too- has.. ·
Gastric emptying of a drug is delayed by co-administering food be- an inhibitorv effect on gastric emptying. Delayed gastric empty-
t ·cause unless the gastric contents are fluid enough or the size of the solid ing as observed with fatty meals, is beneficial for the absorptior
particles is reduced below 2 mm, its passage through the pylorus into the of poorly soluble drugs like griseofulvin.
intestine is not possible. Delay in gastric emptying is recommended in 3. PhYsical state and viscosity of meal : Liquid meals take · les~
particul~r where:
I than an hour to empty whereas a solid meal may tak~ as long ~s
I. The food promotes drug dissolution and absorption e.g. griseofulvin to 7 hours. Viscous materials empty at a slow rate In comparison
2. Disintegration and dissolution of dosage form is promoted by to less viscous materials. ·
gastric fluids ~ 4. Temperature of the meal : High or low temperature of t~e
'
3. The drugs dissolve slowly e.g. griseofulvin ingested fluid (in comparison to body temperature) reduce t e
gastric emptying rate.
4. The drugs irritate the gastric mucosa e.g. aspirin, phenylbutazone
and nitrofurantoin 5. Gastrointestinal pH : Gastric emptying is retarded at low stohm-
ach pH and promoted at hrgher or alkaline pH. Chemica~s-~ ~t
5. The drugs are absorbed from the· proximal part of the small intes-
affect gastrointestinal pH also alter ga~tric ~mp!Ying. The Inhi~I­
. tine and prolonged drug-absorption site contact is desired e.g.
tory effect of various acids on gastric emptyrng decreases With
vitamin 82 and vitamin C
increase in molecular weight and is in the following order: HCl >
Gastric emptying is a first-order process. Several parameters are used acetic ·> lactic > tartaric > citric. .-With alkaline solu~ions, a l~w
to quantify gastric emptying: bas·e concentration (1% NaHC03) mcreases the gastric emptymg
1. Gastric emptying rate is the speed at which the stomach con- rate more than the one of higher concentration (5%). ·
tents empty into the intestine. 6. Electrolvtes and osmotic pressure : Water, isotonic soluti?dnt,
2. Gastric emptying time is the time required for the gastric con- and solutions of low salt concentration empty the stomach rapt .~ Y
tents to empty into~the small intestine. Longer. the gastric emptying whereas a higher electrolyte concentration decreases gastric. emp- _,

time, lesser the gastric emptying rate. tying rate. .


3. Gastric emptying tYI is the time taken for half the stomach 7. · Body posture : .Gastric emptying is favored while standing and
contents to empty. by lying on the right side since the nortna.l curvature of ~he
' stomach provides a downhill path whereas lymg on the left side
In vivo gastric emptying of a drug (so also the disintegration of dosage
or in supine position retards it.
fonn and drug release) can be studied by using radio-opaque contrast
materials like barium sulfate or tagging the drug with a radioisotope and 8. Emotional state : Stress and anxiety promote gastric motility
scanning the stomach at regular intervals of tim~. whereas depression retards it. _
9. Exercise : Vigorous physical training retards gastric emptying.
A large number of factors influence gastric emptying as discussed
below. 10. Disease states : Diseases like gastroenteritis, gastric ulcer, pylo~icr
stenosis diabetes and hypothyroidism retard gastric emptying. Partia
1. Volume of meal : Larger the bulk of the meals, longer the gastric
or total' gastrectomy, duodenal ulcer and hyperthyroidism promote
emptying time. However, an initial rapid rate of emptying is
gastric emptying rate. _
observed with a large meal volume and an initial lag phase in
11. Drugs : Drugs that retard ga~tr~c empty~g in~lude .poorly sol~ble
I

emptying of a small volume meal. Since gastric emptying is a


first-order process, a plot of log of volume of contents remaining antacids (aluminium hydroxide), anttchohnergics_ (at~optn~,
in the stomach versus time yields a straight line. · propantheline), narcotic analg~sics (morphine) and trtcychc antt-

I
57
56 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS

Like gastric emptying, intestinal transit is influenced by several factors


dep_ressants (i~ipra~ine, amitriptyline). Metoclopramide, dom-
like food, drugs and diseases. Food, decreased digestive secretions and
pertdone and ctsaprtde (".ntiemetics) stimulate gastric emptying.
pregnancy retard intestinal transit whereas diarrhea promotes it. Drugs
T~e J!a~sage of ~rug through the esophagus, called as esophageal like metoclopramide that promote gastric emptying and intestinal transit
transit? ts tm.portant .~~ persons who swallow the solid dosage form lying enhance absorption of rapidly soluble drugs. Laxatives also promote the
down tn suptne position or with little or no water. In such cases the rate of intestinal transit. Drugs such as antichol inergics that retard gastric
· do~age fotm rema~ns lodged in the esophagus and disintegrates sl~wly and intestinal transit promote absorption of poorly soluble drugs for
which may result In delayed absorption or local damage to the mucosa
I
example, propantheline shows a I 00%, 50% and 30% rise in the absorp-
from drugs like NSAIDs. tion of vitamin B2 , nitrofurantoin and hydrochlorothiazide respectively .
.
Intestinal Transit
Gastrointestinal pH
Si~ces.mall inte~t~e is. the major site for absorption of most drugs, A tremendous 107 fold difference in the hydrogen ion concentration is
long tntesttnal transit time ts desirable for complete drug absorption (see observed between the gastric and colon fluids. The Gl pH generally
Table 2.6). .. increases gradually as one moves down the stomach tc the colon and
• TABLE 2.6 rectum (see Fig. 2.22). GI fluid pH influence drug absorption in several
Transit Time for Contents from Different Regions of Intestine
ways:
Intestinal region Transit time 1. Disintegration : The disintegration of some dosage forms is pH
Duodenum 5 minutes sensitive. With enteric coated fonnulations , the coat dissolves only in the
Jejunum 2 hours intestine followed by disintegration of the tablet.
Ileum 3 to 6 hours 2. Dissolution : A large number of drugs are either weak acids or
weak bases whose solubility is greatly affected by pi-I. A pH that favors
Cecum 0.5 to I hour
formation of salt of the drug enhances the dissolution of that drug. Since
Colon 6 to 12 hours
drug dissolution is one of the important rate-determining steps in drug
. The resid~n~e time depends upon the intestinal motility or contrac- absorption, GI pH is of great significance in the oral bioavailability of
tions. ~e mzxzng movement of the intestine that occurs due to peristaltic drugs. Weakly acidic drugs dissolve rapidly in the alkaline pH of the
~ontr~cttons promote drug absorption, firstly, by increasing the drug-
intestine whereas basic drugs dissolve in the acidic pH of the stomach.
tntest~nal membrane contact, and secondly, by enhancing drug dissolution Since the primary site for absm:ption of most drugs is small intestine, the
espectally of poorly soluble drugs, through induced agitation. poorly water-soluble basic drugs must first dissolve in the acidic pH of
stomach before moving into the intestine.
Delayed intestinal transit is desirable for:
3. Absorption : Depending upon the drug pKa and whether its an
1. Drugs that dissolve or release slowly from their dosage form
acidic or a basic drug, the GI pH influences drug absorption by determin-
(sustained release products)
ing the amount of drug that would exist in the unionized form at the site
2. ~rugs that dissolve only in the intestine (enteric coated fonnula- of absorption. This topic has already been dealt with in sufficient details
ttons) under pH-partition hypothesis.
3· ~rugs_absorbed from specific sites in the intestine (several B 4. Stability : GI pH also influences the chemical stability of drugs.
vttamms) The acidic stomach pH is known to affect degradation of penicillin G and
lnt~stin~l · transit tin_le is relatively short in comparison to the gastric erythromycin. This can be overcome by preparing prodrugs of such drugs
~mp~tng .time and as t~e
contents move down the intestine into the colon, that do not degrade or dissolve in acidic pH e.g. carindacillin and erythro-
Its vtscost~y gr~d~ally mcreases due to absorption of water and electro- mycin estolate. With basic drugs, formation of insoluble drug hydroxide
lyte~ whtch _hmtts the design of sustained release products of drugs in the alkaline pH of the intestine has been observed.
havmg short biological half-lives.
58 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 59
Disease States 3. Hepatic diseases : Disorders such as hepatic cirrhosis influence
· . Several dis~ase states can influence the rate and extent of drug absorp- bioavailability mainly of drugs that undergo considerable first-·pass he-
tion. The 3 maJor classes of disease states that can influence the bioavailability patic metabolism e.g. propranolol.
of a drug are:
I. Gastrointestinal diseases, Blood Flow to the GIT
The GIT is extensively supplied by blood capillary network and the
2. Cardiovascular diseases, and -
lymphatic system. The absorbed drug can thus be taker~ up by the blood
3. Hepatic diseases. or the lymph. Since the bl~od flow rate to the GIT (splanchnic circula-
1. Gastrointestinal diseases : A number of pathologic conditions of tion) is 500 to 1000 times (28o/o of cardiac output) more than· the lymph
the GIT can influence changes in drug absorption pattern, namely: / flow, most drugs r~ach the systemic circulation via blood whereas only a
(a) Altered GI motility : (discussed earlier) few drugs, especially low molecular weight, lipid soluble compounds are

removed by lymphatic system .
(b) Gastrointestinal diseases and infections: The influence of achlo-
rhydria (decreased gastric acid secretion and increased stomach The high perfusion rate of GIT ensures that once the drug has crossed
pH) on gastric emptying and drug absorption, especially that of the membrane, it is rapidly removed from the absorption site thus main-
acidic drugs (decreased absorption, e.g. aspirin) has been stud- taining the sink conditions and concentration gradient for continued drug
ied. Two of the intestinal disorders related with malabsorption absorption.
syndrome that influence drug _availability are celiac disease For drugs that have high permeation rates, e.g. highly lipid soluble
(characterized by qestruction of villi and microvilli) and Crohn 's drugs or drugs absorbed through pores, the G I perfusion rate could be a
disease. Abnormalities associated with celiac disease include rate-limiting step in the absorption, e.g. tritiated water. This is not so in
increased gastric emptying rate and GI permeability, altered the case of drugs having poor permeability coefficient, e.g. ribitol. ~lood
intestinal drug metabolism, steatorrhea (impair~d secretion of flow is also important for actively absorbed drugs since oxygen and
bile thus affecting absorption of lipophilic drugs and vitamins) energy is required for transportation.
and reduced enterohepatic cycling of bile salts, all of which can
Food influences blood flow to the G IT. The perfusion rate increases
significantly impair drug absorption. Conditions associated with
after meals and persists for few hours but drug absorption is not influ-
Crohn' s disease that can alter absorption pattern are altered gut
enced significantly.
wall microbial flora, decreased gut surface area and intestinal
transit rate. Malabsorption is also induced by drugs such as Gastrointestinal Contents
antineoplastics and alcohol which increase permeability of agents A number of GI contents can influence drug absorption as discussed
not normally absorbed. GI infections like shigeJlosis, gastroenteritis, below:
cholera and food poisoning also result in malabsorption. Co-
1. Food-drug interactions : Presence of food may either - delay,
lonic diseases such as colitis, amebiasis and constipation can
reduce, increase or may not affect drug absorption (Table 2. 7).
also alter drug absorption.
(c) Gastrointestinal surgery : Gastrectomy can result in drug dump- TABLE 2.7 Influence of Food on Drug Absorption
ing in the intestine, osmotic diarrhea and reduced intestinal Delayed Decreased Increased Unaffected
transit time. Intestinal surgery also influences drug absorption
for predictable reasons. Aspirin Penicillins Griseofulvin Methyldopa

2. Cardiovascular diseases : Several changes associated with con- Paracetamol · Erythromycin Nitrofurantoin Propylthiouracil
gestive cardiac failure influence bioavailability of a drug viz. edema of the Diclofenac Ethanol Diazepam Sulfasomidine
intestine, decreased blood flow to the GIT and gastric emptying rate and Nitrofurantoin Tetracyclines Actively absorbed
altered GI pH, secretions and microbial flora. water soluble
Digoxin Levodopa
• vitamins •
Iron

60 BIOPHARMACEUTICS AND PHARMACOKINE, ICS
ABSORPTION OF DRUGS 61
Food-drug interactions may be due to the influence of food on :·· Iysi-
enzymes which influence drug absorption . Mucin, a protective mucopo-
ologic functions (alterations in the GI emptying rate, GI fluid sec:-etions,
lysaccharide that lines the G I mucosa, interacts with streptomycin and
pH, blood flow and absorptive processes) and/or a consequence of physi-
certain quaternary ammonium compounds and retards their absorption. It
cochemical interaction with the drug (alteration in drug dissolution profile,
also acts as a barrier to diffusion of drugs. The bile salts aid solubiliza-
c~mplexation and adsorption).
tion and absorption of lipid soluble drugs like griseofulvin and vitam ins
As a general rule, drugs are better absorbed under fasting conditions A D E and K on one hand and on the other, decreases absorption of
' '
. rmd presence of food reta!_ds or prevents it. Food does not significantl; . neomycin and kanamycin by forming water insoluble cotnplexes .
illfluence absorption of a drug taken half an hour or more before meals The influence of GI enzymes on absorption will be discussed later.
and two hours or more after meals. Delayed or decreased drug absorption
by the food could be due to one or more of the several mechanisms: 4. Drug-Drug interactions in the G IT : Like food-drug interac-

tions, drug-drug interactions can either be physicochemical or physiological.
(a) Delayed gastric emptying, affecting drugs unstable in the stomach
e.g. peniciJiins, or preventing the transit of enteric coated tablets (a) Physicochemical drug-drug interactions can be due to
into the intestine which may be as long as 6 to 8 hours Adsorption : antidiarrhe.al preparations containing adsorbents like
(b) Formation of a poorly soluble, unabsorbable complex e.g. tetracy- attapulgite or kaolin-pectin retard/prevent absorption of a number
cline-calcium of drugs co-administered with them e.g. promazine and lincomycin.
(c) Increased viscosity due to food thereby preventing drug dissolu- Complexation : antacids and/or mineral substitutes containing heavy
tion and/or diffusion towards the absorption site metals such as aluminium, calcium, iron, magnesium or zinc re-
Increased drug absorption following a meal could be due to one or tard absorption of tetracyclines through formation of unabsorbable
more of the under mentioned reasons: complexes. The anion exchange resins, cholestyramine and colestipol,
bind cholesterol metabolites, bile salts and a nun1ber of drugs in
(a) Increased tirrie for dissolution of a poorly soluble drug
the intestine and prevent their absorption.
(b) Enhanced solubility due to GI secretions like bile
pH change : basic drugs dissolve in gastric pH; co-administration
(c) Prolonged residence time and absorption site contact of the da~g of such drugs, e.g. tetracycline with antacids such as sodium
e.g. water-soluble vitamins bicarbonate results in elevation of stomach pH and hence de-
(d) Increased lymphatic absorption e.g. acitretin creases dissolution rate or causes precipitation of drug.
The specific meal components also have an influence on drug absorp- (b) Physiologic drug-drug interactions can be due to following
tion. Meals high in fat aid solubilization of poorly aqueous soluble drugs mechanisms.-
e.g. isotretinoin. Food high in proteins inhibit absorption of levodopa. Decreased G! transit : an,ti~holinergics such as propantheline retard
An interesting example of influence of high protein meal is that of in- GI motility and promo.t e absorption of drugs like ranitidine and
creased oral availability of propranolol to which two reasons were digoxin, whereas delay absorption of paracetamol and sulfa-
attributed firstly, such a meal increases the hepatic blood flow due to methoxazole.
which the drug can bypass frrst-pass hepatic metabolism (propranolol is a
Increased gastric emptying : metoclopramide promotes G I motil-
drug with high hepatic extraction), and secondly, it promotes blood flow
to the GIT thus aiding drug absorption. ity and enhances absorption of tetracycline, pivampicillin and
levodopa.
2. Fluid volume : Administration of a drug with large fluid volume Altered Gl metabo/isn1 : antibiotics inhibit bacterial metabolism of
results in better dis$olution , rapid gastric emptying and enhanced drugs, e.g. erythromycin enhances efficacy of digoxin by this
absorption for example, erythromycin is better absorbed when taken mechanism . Co-administration of antibiotics with ,oral contracep-
with a glass of water under fasting condition than when taken with meals. tives like ethinyl estradiol decreases the efficacy of latter by
3. Interaction of drug with normal GI constituents : The GIT decreasing enterohepatic cycling ·o f steroid conjugates which
contains a number of nounal constituents such as mucin, bile salts and otherwise are hydrolyzed by gut bacteria after biliary excretion.
ABSORPTION OF DRUGS 63
62 BIOPHARMACEUTICS A.ND PHARMACOKINETICS
via bile such as glucuronides of digoxin and oral contraceptives. The free
Presystemic Metabolism/First-Pass Effects
drugs are reabsorbed into the systemic circulation.
For a drug adm-inistered orally, the 2 main reasons for its decreased
'
bioavailability are: ' To systemic circulation .
a. Decreased absorption, and
b. First-pass/presystemic metabolism.
I

Before a drug reaches blood circulation, it has to pass for the first time
through organs of elimination namely the GIT and the liver. The loss of
drug_ through biotransformation by such eliminating organs during its
passage to systemic circulation is called as first-pass or presystemic First-pass Liver
metabolism. The diminished drug concentration or rarely, complete absence
hepatic metabolism
~
of the drug in plasma after oral administration is indicative of first-pass
I
\••••
effects. The 4 primary systems which affect presystemic metabolism of a Portal Vein •••
• •z
drug are (Fig. 2.24): Metabolism by · @ ... :.:. I
gut wall enzymes •...• ...
........... :
• • • • • 0 ~

1. Lumenal enzymes,
2. Gut wall enzymes/mucosal enzymes, Unabsorbed
drug in faeces Colon Small Intestine Stomach
3. Bacteria1 enzymes, and I
I

4. Hepatic enzymes.
1. Lumenal e~zymes : These are the -enzymes present in the gut G) G)
fluids and include enzymes from intestinal artd pancreatic secretions. The Metabolism by Metabolism by
latter contains hydrolases which hydrolyze ester drugs like chloramphenicol bacterial enzyrt:~es lumenal enzymes
of the colon (intestinal and pancreatic)
palmitate into active chloramphenicol, and peptidase~ which split amide
linkages and inactivate protein/polypeptide drugs. Thus, .one of the approaches ..
Fig. 2.24 Different sites ofipresystemic metabolism
to effect oral absorption of peptides is to deliver them to colon ·which lack
peptidases. 4. Hepatic enzymes : Several drugs undergo first-pass ·hepatic me-
2. Gut wall enzymes : Also called as muc~sal enzymes, they are tabolism, the highly extracted ones being isoprenaline, propranolol, alprenolol,
present in stomach, intestine and colon. Alcohol dehydrogenase (ADH) is pentoxifylline, nitroglycerine, diltiazem, nifedipine, lidocaine, morphine,
an enzyme of stomach .mucosa that inactivates ethanol. Intestinal mucosa etc.
contains both phase I and phase II (predominant) enzymes, e.g. sulfat.ion
of ethinyl estradiol and isoprenaline. The colonic mucosa also contain ABSORPTION OF DRUGS FROM NON PER OS '

both phase I and phase II enzymes. However, it is only the enzymes of EXTRAVASCULAR ROUTES
the pro~im~l small intestine that are most active. Drug absorption from all nonoral extravascular sites is governed uy
the same factors that influence absorption from G IT viz. the physico-
3. Ba~terial enzymes : The GI microflora is . scantily present in
chemical properties of drug, formulation factors, and anatomic, physiolog-
stomach and small intestine and is rich in colon. Hence, most orally
ic and pathologic characteristics of the patient. This is so because the
administered drugs remain unaffected by them. The colonic microbes
barrier to transport of drugs irito the systemic circulation from all such
generally render a dr1:1g more· active or toxic on ·biotransformation for . '

sites is a lipoidal membrane .similar to · the GI barrier and the major


example sulfasalazine, a drug used in ulcerative colitis, is hydrolyzed to .
mechanism in the ~bsorption is passive diffusion. One of the major
sulfapyridine and 5-amino salicylic acid by the microbial enzymes of the
advantages of administering drugs by noninvasive transmucosal routes
colon. An important role of int.estinal microflora is that in enterohepatic
cycling. Their .enzymes hydrolyze the conjugates of drugs actively secreted such as nasal, buccal, rectal, etc. is that greater systemic availability is
64 BIOPHARMACEUTICS AND PHARMACOKINETICS
.'

ABSORPTION OF DRUGS 65
attainable for drugs normally subjected to extensive presystemic elimina-
Some of the advantages of these routes are:
tion due to GI degradation and/or hepatic metabolism (Fig. 2.25). Moreover,
1. Rapid absorption and higher blood levels due to high va~c.ulari~a­
Per Os Delivery tion of the region and therefore particularly useful for admtntstration
of antianginal drugs
Routes to Bypass 2. No first-pass hepatic metabolism
Pre~ystemic Elimination
3. No degradation ~f drugs such as that encountered in the G IT
OCULAR
DELIVERY 4. Presence of saliva facilitates.. both drug dissolution and its subse-
GIT ~~~~~~~~ · quent penneation by keeping the oral mucosa moist
Notable factors to be considered in the oral mucosal delivery of drugs
NASAL
DELIVERY are:
first-pass
1. Lipophilicity of drug : Slightly higher lipid s?lubility th~n that
PULMONARY required for GI absorption is necessary for passive penneatton.
LIVER ~ DELIVERY
SYSTEMIC ~~~~~~
2. Salivary secretion : In addition to high lipid solubili~, the ~~g
CIRCULATION should be soluble in aqueous buccal fluids i.e. biphastc solubthty
BUCCAUSUBLINGUAL of drug is necessary for absorption; absorption is delayed if the
DELIVERY mouth is dry.
3. pH of the saliva : Usually around 6, the buccal pH favors absorp-
RECTAL tion of drugs which remain unionized.
DELIVERY 4. Binding to oral mucosa : Systemic availability of drugs that bind
I

to oral mucosa is poor.


VAGINAL 5. Storage compartment : For some drugs such as bupreno~hine.' a
DELIVERY storage compartment in the buccal mucosa appears to extst wh1ch .
is responsible for the slow absorption of drugs.
Fig. 2.25 Various transmucosal noninvasive routes of drug adminis-
tration to bypass presystemic elimination in GIT/Iiver 6. Thickness of oral epithelium ·: Sublingual absorption is faster t~an
buccal since the epithelium of fonner region is thinner and Im-
peptide and protein drugs can also be delivered by such routes. Some of
mersed in a larger volume of saliva.
the more important biopharmaceutic and pharmacokinetic principles that
must be considered for nonoral absorption will be discussed here. Factors that limit drug ·administration by these routes are: limited
mucosal surface area (thus only a small dose can be administered), con-
Buccal/Sublingual Administration cern for taste of the medicament and discomfort (since the region is
r

The two sites for oral mucosal delivery of drugs are: highly innervated).
1. Sublingual route : The drug is placed under the tongue and Examples of drugs administered by oral mucosal route include anti~gin.als
allowed to dissolve like nitrites and nitrates, antihypertensives like nifedipine, analgesics hke
morphine and bronchodilators like fenoterol. Certain steroids like estra-
2. Buccal route : The medicament is placed'b._et"»'een
....,
the cheek and • diol and peptides like oxytocin can also be adinioistered. , Apa~ from
the gum ·
tablets, the drugs may be administered as a buccal spraY- especially to
The barrier to drug absorption from these routes is the epithelium of children.
oral mucosa. Passive diffusion is the major mechanism for absorption of
most drugs; nutrients may be absorbed by carrier-mediated processes. Rectal Administration
Despite its diminished popularity, the rectal route of drug administra-
tion is still an important route for children and old patients. The drugs
\

66 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 67

~may be administere~ as solutions (microenemas) or suppositories. Ab- 3. Trauma cuts, rashes, inflammation, mild bums or other condi-
sorption is more rapid from solutions than from suppositories but is more tions in which the stratum corneum is destroyed, promote dnig
variable in comparison to oral route. Irritating suppository bases such as absorption. ,
.PEG promotes defecation and drug loss. Presence of fecal matter retards
I , 4. Hydration of skin : soaking the skin in water or occluding: it by
drug absorption. Though highly vascularized, absorption is slower because using emollients, plastic film or dressing, promote hydration of
of- limited surface area. The pH of rectal fluids (around 8) also influences ·-
'
skin and drug absorption.
drug absorption according to pH-partition hypothesis. Absorption of drugs
5. Environment humidity and temperature : higher humidity and
from the lower half of rectum bypasses presystemic hepatic .m etabolism.
temperature increase both the rate of hydration as well as local
Drugs administered by this route include aspirin; paracetamol, theophylline,
blood flow and hence drug absorption.
few barbiturates, etc.

6. Age : gross histological changes take place as the skin ages .
Topical Administration Aged skin is more prone to allergic and irritant effects of topi-
Excluding the respiratory tract's contact with the inhaled air, the cally contacted chemicals as a result of hardening of blood vessels.
skin is virtually the sole human surface directly interfacing. the body with Infants absorb drug through skin as efficiently as adult~. Their
,_the external environment. It is the largest organ of the body weighmg ratio of surface area to body weight is 3 times that of adults;
approximately 2 Kg and 2 meter square in area and receives about 1/3rd hence, systemic toxicity of topically applied drugs is of particular
of total blood circulating through the body. Though tolerant to many concern in infants.
chemicals, topically contacted x_enobiotics can evoke both local and sys- 7. Grooming : the frequency and vigor· with which one bathes and
temic effects. When topically applied drugs are meant to exert their the type of soap that is used also contribute to variability in drug

effects systemically, the mode of administration is called as percutaneous absorption.
or transdermal delivery. 8. Exposure to chemicals : occupational exposure to solvents can
I Anatomically, the skin is made of 3 distinct layers the epidermis, the accelerate shedding of epidermal cells and enhance drug absorption.
dennis and the subcutaneous fat tissue. Epidennis is the nonvascular, 9. Vehicle or base : the vehicle in which the drug is incorporated
multilayered outer region of the skin. The dermis or true skin is a highly influences drug absorption; the one in which the drug is dissolved
vascular region; drugs penneating to .this region are taken up into the • rather than dispersed promotes absorption .
systemic circulation and sink conditions are maintained.
10. Permeation enhancers : incorporation of certain chemicals such as
The principal barrier to 'the entry of xenobiotics is the most superficial DMSO, propylene glycol, azone, etc. in the topical fonnulations
layer of epidermis called as stratum corneum. It is composed of dead, aid drug penetration.
keratinized, metabolically inactive horny cells that act as the major rate- 11. Chronic use of certain drugs : long term use of cortisol or keratolytics
limiting qarrier to passive diffusion of drugs. In order to act either locally
like salicylic acid results in enhanced drug penetration.
or systemically, a topically applied drug may diffuse through the skin by
hair follicles, sweat glands or sebaceous glands but permeation through Drugs that are administered percutaneously include nitroglycerine,
the multiple lipid bilayers of stratum corneum is the dominant pathway lidocaine, betamethasone, estradiol, testosterone, etc. The route is particu- ·
\ though the rate is very_slow. Several factors influence passive percutane- larly useful for drugs with low oral availability and short duration of
. ous absorption of drugs: action; the effect of the latter category of drugs is prolonged because
percutaneous absorption is a slow process.
1. Thickness of stratum corneum : absorption is very slow from
'
regions such as foot and palm where the skin has thickened Intramuscular Administration
stratum corneum. Absorption of drugs from i.m. sites is relatively rapid but much slower
2. Presence of hair follicles : absorption is rapid from regions where in comparison to i.v. injections. Factors that determine rate of drug
numerous hair follicle§ exist e.g. scalp. absorption from i.m. sites are:

68 BIOPHARMACEUTICS AND PHARMACOKINETICS
ABSORPTION OF DRUGS 69

1. Vascularity of the injection site : the decreasing order of blood local cooling or co-injection of a vasoconstrictor like adrenaline or by
flow rate to muscular tissues in which drugs are usually injected immobilization of limb. Because of relatively slow drug absorption from
s.c. tissues, the region is very popular for controlled release medication
is: arm (deltoid) > thigh (vastus latera/is) > buttocks (gluteus
maxim us). Since blood flow rate is often the rate-limiting step in
like implants.
absorption of drugs from i.m. sites, most rapid absorpti~n is from Pulmonary Administration
deltoid muscles and slowest from gluteal region. The absorption
In principle, all drugs intended for systemic effects can be adminis-
rate decreases in circulatory disorders such as hypotension.
tered by inhalation since the large surface area of the alveoli, high penneability
2. Lipid solubility and ionization of drug : highly lipophilic drugs of the. alveolar epithelium and rich perfusion pennit extremely ra~id ~b­
are absorbed rapidly by passive diffusion whereas hydrophilic and sorption just like exchange of gases between the blood and the msprred
ionized drugs are slowly absorbed through capillary pores. air. However, the route has been limited for administering drugs that
3. Molecular size of the drug : small molecules and Ions gain arrect affect pulmonary system such as bronchodilators (salbutamol), anti-in-
access into capillaries through pores whereas macromolecules are flammatory steroids (beclomethasone) a~d antiallergics (cromolyn). Lipid
taken up by the lymphatic system. soluble drugs are rapidly absorbed by passive diffusion and polar drugs by
4. Volume of injection and drug concentration : a drug in concen- pore transport. Absorption of drugs whose ionization is pH sensitive is
trated injection and large volume is absorbed faster than when dependent upon pH of pulmonary fluids. The drugs are generally admin-
given in dilute form and small volume. istered by inhalation either as gases (volatile/gaseous anesthetics ) or
\
aerosol. In the latter case, drug delivery to lungs is largely dependent
'5. pH, composition and viscosity of injection vehicle : a solution of
upon the particle size of the aerosolized droplets particles larger than 10
drug in acidic or alkaline pH (e.g. phenytoin, pH 12) or in a
microns impact on the mouth, throat or upper respiratory tract mucosa and
nonaqueous solvent such as propylene glycol or alcohol (e.g.
do not reach the pulmonary tree whereas very small particles (0.6 mi-
digoxin) when injected intramuscularly result in precipitation of
crons) from which drug absorption is rapid, penetrate rapidly but are
drug at the injection site followed by slow and prolonged absorp-
susceptible to easy exhalation. Sometimes, the patients inability to inhale
tion. Viscous vehicles such as vegetable oils also slow drug a sufficient amount of drug limits drug delivery to lungs.
absorption. The principle can however be utilized to control rate
of drug delivery. Intranasal Administration
Subcutaneous Administration , The nasal route is becoming increasingly popular for systemic delivery
I
especially of some peptide and protein drugs. Drug absorption from nasal
All factors that influence :.m .. drug absorvtion are also applicable to
- - - - - mucosa is as rapid as observed after parenteral administration because of
absorption from subcutaneous- site.
- -
Generally, absorption_of drugs trom a
~
its rich vasculature and high permea_!?ility. The route is otherwise used for
s.c. sit'e is slower than that from i.m. sites due- to poor perfusion, but this
drugs to treat local symptoms like nasal congestion, rhinitis, etc. ~
fact js of particular importance for the administration of drugs for which a
rapid response is not desired and for drugs that degrade when taken orally Two mechanisms for drug transport across the nasal mucosa have been
e.g. insulin and ~odium heparin. . The rate of absorption of a drug from suggested a faster rate that is dependent upon drug lipophilicity, and a
subcutaneous site can be increased in 2 ways:

slower rate which is dependent upon drug molecular weight. In case of
1. Enhancing blood flow to the injection site : by massage, applica- lipophilic drugs, rapid absorption by diffusion is observed upto 400
.
I tion of heat, co-administration of vasodilators locally, or by exercise, daltons and satisfactory absorption upto 1000 daltons. By use of penne-
and ability enhancers such as surfactants, even a drug with molecular weight
of 6000 daltons shows reasonable bioavailability. For polar compounds
2. Increasing the drug-tissue contact area : by co-administering the primarily absorbed by pore transport, an upper threshold of 200 dalt.ons is
enzyme hyaluronidase that breaks down the connective tissue and the limiting factor. Other factors that may influence nasal penneatton of
permits spreading of drug solution over a wide area. drugs include pH of nasal secretions (5.5 to 6.5) and its viscosity, and
Absorption can be slowed down by causing vasoconstriction through pathological conditions such as common cold and rhinitis. Drugs known
I

• I
70 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 71

to in't luence cleansing function of nasal cilia should not be administered TABLE 2.8
by this route. Absorption of Drugs from Non per os Extravascular Routes
Route Absorption Mechanism(s) Drugs Delivered
Intraocular Administration ..

Topical application of drugs to the eyes is mainly meant for local Buccal/ passive diffusion, nitrites and nitrates
effects such as mydriasis, miosis, anesthesia or treatment of infections, Sublingual carrier-mediated antianginals, nifedipine,
transport morphine, fenoterol, etc.
glaucoma, etc. Sterile aqueous solutions of drugs are widely used oph-
thalmic fonnulations and administered in the conjunctival cul-de-sac. The Rectal passive diffusion aspirin, paracetamol, theo-
barrier to intraocular penetration of drugs is the cornea which posseses phylline, few barbiturates, etc.
both hydrophilic and lipophilic characteristics. Thus, for optimum intraocular Transdermal passive diffusi0n nitroglycerine, lidocaine,
penneation, drugs should possess biphasic solubility. The pH of lacrimal scopolamine, testosterone,
fluid influences· absorption of weak electrolytes such as pilocarpine. On betamethasone, etc.
:he other hand, pH of the formulation influences lacrim~ll output higher Intramuscular pas.sive diffusion, phenytoin, digoxin, several
pH decreases tear flow and promotes drug -absorption whereas lower pH endocytosis, pore steroids and antibiotics
solutions increase lacrimation and subsequent drug loss due to drainage. transport and many other drugs.
· Rate of blinking also influences drainage loss. The volume of fluid Subcutaneous passive diffusion insulin, heparin, C.R. implants.
· .instilled into the eyes also affect bioavailability·· ~nd effectiveness of the Inhalation passive diffusion, salbuterol, cromolyn,
/ \
1

drug. Nonnally, the human eye can hold around 10 J.!l of fluid; hence, pore transport becloniethasone. I

instillation of small volume of drug solution in concentrated form increases Intranasal passive diffusion, pheny Ipropanol amine,
its effectiveness than when adqlinistered in large volume in dilute fonn. pore transport antihistamines.
Viscosity imparters in the fonnulation increase bioavailability by prolong- Intraocular passive diffusion atropine, pilocarpine,
ing drug's contact time with the eye. Oily solutions, ointments and gels adrenaline, antibiotics, etc.
show sustained drug action for the same reason. Sometimes systemic steroidal drugs and contracep-
Vaginal passive diffusion
a9sorption of a drug with low therapeutic index such as timolol may tives,' metronidazole, ·etc.
__precipitate undesirable toxic effects. -Systemic entry of drugs occur by
I

j way of absorption into lacrimal duct which drains lacrimal fluid into the
QU~STIONS
nasal cavity and fmally into_the GIT. This can be prevented by simple
1. Define drug absorption. What are the various routes from which absorption
eyelid closure or nasolacrimal occlusion by pressing the fmger tip to the
of a drug is necessary for pharmacologic action? , '
inside comer of the eye after drug instillation.
2. Why are both rapidity and completeness of drug absorption . important?
Vaginal Administration What is their significance in drug therapy?
'
\

· Drugs meant for intravaginal application are generally intended to act 3. What is the major mechanism for absorption of most drugs? What is the
\
locally in the treatment of bacterial or fungal infections ·o r prevent concep- driving force for such a process?
tion. The route is now used for systemic delivery of contraceptives and 4. What do you understand by sink conditions? How is it maintained and
other steroids, without the disadvantage of first-pass metabolism. Con- responsible for complete passive absorption of drugs from the GIT?
. trolled delivery and tertnination of drug action \Vhen desired, is possible 5. List the characteristics of passive diffusion of drugs.
with this route. Factors that may influence drug absorption from intravaginal 6. Why is the absorption rate of a sufficiently water-soluble but lipophilic drug
site include pH of lumen fluids (4 to 5), vaginal secretions and the always greater than its rate of.elim~nation? ,,
microorganisms present in ·the vaginal lumen which may metabolize the 7. What are the characteristics of specialized transport systems? How can the
drug. kinetics of such processes by described?
1
~ ,A summary of mechanisms /and drugs 'absorbed from various noninvasiv~ 8. It is ahyays advisable to administer B vitamins in small multiple doses
routes (other than the GI) is gi~en in Table 2.8. · rather than as a single large dose. Why?
72 BIOPHARMACEUTICS AND PHARMACOKINETICS ABSORPTION OF DRUGS 73
9. Discuss the similarities and differences between passive and facilitated diffusion. 29. Buffered aspirin tablets are more suitable than sodium salt form of aspirin.
10. Why is active transport not a predominant mechanism for absorption of Why?
drugs? What could be the reason for active .a bsorption of several antineoplastics 30. What is the influence of the size of counter ion on solubility of salt forms
or nutrient analogs? of the drugs?
11. How are ionic/ionizable drugs absorbed? 31. State the pH-partition hypothesis briefly. On what assumptions this state-
12. What is the only absorption mechanism for which aqueous solution of a ment is based?
drug · is not a prerequisite? What is the signific·a nce of such a transport 32. Based on pH-partition theory, predict the degree of ionization and absorp-
process? tion of very weak, weak and strong acidic and basic drugs -from stornach
13. Suggest the likely mecQanism for oral absorption of following agents: lithium and intestine.
carbonate, ibuprofen, cyanocobalamin, methotrexate, quaternary an1monium 33. For optimum absorption, a drug should have biphasic solubility or perfect
compounds and insulin. HLB in its structure. Explain.
14. Enlist and illustrate the .steps involved in the absorption of a drug from 34. Discuss the limitations and significance of pH-partition hypothesis.
orally a~ministered sol:id dosage forms.
35. Why is disintegration test not considered a guarantee of a drug's bioavailability
15. Define the rate-det~rmining step. What are the two major RDS' in the from its solid dosage form?
absorption of ()rally administered drugs? Based on the solubility profile, to
36. The influence of compression force on drug dissolution and absorption from
which drugs they apply?
• tablets is unpredictable. Explain .
16., Classify. and enumerate the biopharmaceutic factors influencing bioavailability
37. Discuss briefly the influence of phannaceutical excipients on drug bioavailability.
• r of a drug from its dosage form.

38. How do the surfactants promote bioavailability of a poorly water-soluble


17. Justify the statement dissolution rate is better related to drug absorption
drug? What is their influence when used in higher concentrations?
and bioavailability than solubility.
39. Quote examples of complexation used to enhance bioavailability of a drug.
18. ·Name the various theories that explain drug dissolution.
40. How does the nature and type of dosage form influence drug absorption?
19. Using Noyes-Whitney's equation, discuss the diffusion layer theory and the
variables that influence drug dissolution. , 41. List the orally adn1inistered dosage forms in order of decreac;;ing bioavailability.
42. What are the anatomical and p~•ysiological
reasons for differences in the rate
20. Why is in vivo drug dissolution always faster than in vitro dissolution?
What conditions , should be simulated in order to obtain a good relationship? and extent of absorption of a drug from various regions of the G IT?

21. What assumptions are made in diffusion layer and Danckwert' s models? 43. Why are all types of drugs, whether acidic, basic or neutral, better absorbed
from small intestine?
22. What is the difference between absolute and effective surface area? How
can the latter of a hydrophobic drug be increased? 44. Fo.r which drugs rapid GE is desirable and when should it be slow?

23. Micronization of hydrophobic drugs actually results in reduction- in effective 45. Discuss briefly the factors affecting GE of drugs.
surface area and dissolution rate. Why? 46. What are the possible reasons for delayed and for enhanced absorption of a
24. For which drugs an increase in surface area by micronization is not -advisable? drug after meals? Why biopharmaceutic studies that require the drug to be
taken orally, be performed in volunteers on empty stomach?
25. Classify the internal structure of a solid compound . and define: polymor-
. phism, amorphism and pseudopolymorphism. 47. Delayed intestinal transit is sometimes desirable. Why?
'
26. How will you account for the rapid dissolution of amorphs, metastable 48. What are the consequences of various disease states on ·oral bioavailability
polymorphs and organic solvate$ in comparison to their respective counter- ,of a drug?
\

parts? 49. What are the various mechanisms for drug-drug interactions in the G IT?
/ 27. Why is the pH of the diffusion layer high and low respectively for salts of 50. What are the different sites of presystemic metabolism' of orally adminis-
weak acids and weak ·bases in comparison to that observed with free forms tered drugs?
of the drugs? 51. How can the metabolic role of colonic microflora be utilized for drug
28. Give two reasons for higher solubility and better dissolution of salt forms of targeting to large intestine?
\
the drug in comparison to their free acidic or basic forms. 52. What are the advantages of administering drugs by non per os noninvasive
transmucosal routes? Name such routes.
74 BIOPHARMACEUTICS AND PHARMACOKINETICS

53. Discuss the factors in the absorption of drugs from various non per os
transmucosal and transdermal routes. ·
54. Transdermal delivery as well as most of the transmucosal routes other than '

the -GI are limited for systemic administration of low dose drugs only.
Explain. 3
55. How can the absorption of drugs from subcutaneous sites be promoted?
56. What factors limit drug administration by pulmonary route? Why is this
route restricted for administration of drugs affecting pulmonary function?
Distribution of Drugs
57. Name ·the physicochemical and biological factors that limit drug administra-
tion by oral route. ·
After entry into the systemic circulation, either by intravascular injection
58. Given below in the Table are 3 basic drugs together with some of their
physicochemical and biological properties:
or by absorption from any of the various extravascular sites, the drug is
subjected to a number of processes called as disposition processes that
Properties Diazepam Loratidine Guanethidine tend to lo·ltver its plasma concentration. The two major drug disposition .
Molecular weight 285.0 383.0 296.0 processes are:
pKa 3.7 5.0 11.7 1. Distribution which involves reversible transfer of a drug bet1tveen
Aqueous solubility slight insoluble high compartments, and
Ko/w high 27.0
I
low·
. I
2. Elimination which causes irreversible loss of drug from the body.
•,

% Presystemic metabolism nil 90.0 40.0 Elitnination is further divided into two processes viz. biotransformation
(metabolism) and excretion.
a. Based on the aqueous solubility and K 0 ; w, what could be the rate-
The interrelationship between drug distribution, biotransfonnation and
limiting step in the passive absorption of drugs?
excretion and the drug in plasma is shown in Fig. 3 .1.
b. Determine the per cent drug ionization in stomach pH 3.0 and intes-
tine pH 5.0.
Answer : Diazepam : 83% and 4.8o/o, Loratidine : 99% and 50% and Drug
Guanethidine : 100% and 100°/o. Distributed
in the
c. Based on pH-partition hypothesis, ..from which site the drugs will be
best absorbed? Which drug will be absorbed along the entire length
Body
~ Drug ~
(and meta-
of the GIT?
bolites) in
d. Assuming that only Lhe unionized drug is absorbed, what per cent of Plasma
drug will be absorbed from the intestine at pH 7.4?
Answer : Diazepam : 100%, Loratidine : 99.6% and Guanethidine ·
oo/o.
e. Determine the relative amount of drug present in the intestine at pH Drug
5.0 and plasma at pH 7.4. Excreted
Answer : Diazepam-]: 1, Loratidine-2: 1 and Guanethidine-500: I.
f. Delayed GI transit and food intake will be beneficial to absorption of
Fig. 3.1 Interrelationship between different processes of drug disposition
which drug?
g. From tlfe above results and from presystemic metabolism data, a As stated above, distribution is defined as the reversible transfer of a
change in the route of administration is advisable for which drug(s)? drug between on~ 'compartment and another. Since the process is carried
h. If guanethidine shows an oral availability of 20°/o: what could be the
out by the circulation of blood, one of the compa~ments is always the
possible mechanism for its absorption ?
75
76 BIOPHARMACEUTICS AND PHARMAC OKJNT.:.TICS 77
DISTRIBUTION OF DRUGS
blood or the plasma and the other represents extravascular fluids ar. other be tissue pern1eability rate-limited. The tissue permeability of a drug
body tissues. In other words, distribution is reversible transfer cf a drug
depends upon the physicoc~ef!Iical properties of _the ~ru_& a" well as the
befl-veen the blood and the extravascular fluids and tissues. Distribution is
physiologic barriers that restrict diffusion of drug mto ttssues.
a passive process, for which, the driving force is concentration gradient
between the blood and the extravascular tissues. The process occurs by r'hysicochemicai Properties of the Drug . . .
diffusion of free drug only until equilibrium is achieved. As the phannacologic Important physicochemical properties that i~~uence dru_g dtstnbution
action of a drug depends upon its concentration at the site of action, are molecular size, degree of ionization and partition coefficient.
distribution plays a significant role in the onset, intensity and sometimes
Almost all drugs having molecular weight less than 500 to 600 d~ltons
duration of drug action.
easily cross the capillary membrane to diffuse into the extracellular mte:-
Distribution_ of a drug is not uniform throughout the body because stitial fluids. However, penetration of drugs from the extracellular flutd
different tissues receive the drug from . plasma at different rates and to into the cells is a function of molecular size, ionization c?nsta~t and
different extents. Differences in drug distribution among the/~issues es- lipophilicity of the drug. Only small, water-soluble molecules and tons of
sentially arise as result of a number of factors as enumerated below: size below 50 daltons enter the cell through aqueous filled channels
1. Tissue pen11eability of the drug: whereas those of larger size are restricted unless a specialized transport
a. Physicochern ical properties of the drug Iike molecular size, system exists for them.
pKa and o/w partition coefficient The degree of ionization of a drug is an important de term in ant in its
b. Physiological barriers to diffusion of drugs tissue penetrability. The pH of the blood and the ~xtravascular fluid also
2. Organ/tissue size and perfusion rate
play a role in the ionization and diffusion of drugs tnto cells. A drug_ that
remains unionized at these pH values can permeate the cells relatively
3. Binding of drugs to tissue components: more rapidly. Since the blood and the ECF pH normally re~ain.s constant

a. Binding of drugs to blood components at 7.4, they do not have much of an influence on dru~ dtffuston unless
b. Binding of drugs to extravascular tissue proteins altered in conditions such as systemic acidosis or alkalosis.
4. Miscellaneous factors: Most drugs are either weak acids or weak bases and their degree of
a. Acre ionization at plasma or ECF pH depends upon their pKa· All drugs that
b

b. Pregnancy ioni=e at plas 1na pH (i.e. polar. hydrophilic. ~ru~s), cannot ~en~~rate the
lipoidal cell membrane and tissue permeabthty ts the rate-hmatlng step
c. Obesity
d. Diet
capillary wall cell membrane
e. Disease states Intracellular
f. Dtug interactions Blood ECF Fluid

TISSUE PERMEABILITY OF DRUGS


Unionized Drug
Of the several factors listed above, the two major rate-determining (low Ka and large Ko/w)

steps in the distribution of drugs are:
1. Rate of tissue permeability, and
2. Rate of blood .perfusion.
(large Ka and low Ko/w)
If the blood flow to the entire body tissues were rapid and uniform,
differences in the degree of distribution between tissues will be indicative Fig. 3.2 Permeation of unionized and ionized drugs
of differences in the tissue penetrability of the drug and the process will across the capillary and the cell membrane
DISTRIBUTION OF DRUGS 79
78 BIOPHARMACEUTICS AND PHARMACOKINETICS
Physiologic Barriers to Distribution of Drugs
in the distribution of such drugs. Only unionized drugs which are gener- A membrane (or a barrier) with special structural features can be a
ally lipophilic, rapidly cross the cell membrane. Among the drugs that
permeability restriction to dis~i~ution of ~gs. to so~e tiss~es. Some of
have same o/w partition coefficient but differ in the extent of ionization at the important simple and spectahzed phystologtc barrters are.
blood pH, the one that ionizes to a lesser extent will have greater penetra-
1. Sfmpfe capillary endothelial barrier
bility than that which ionizes to a larger extent; for example, pentobarbital
'
and salicylic acid have almost the same K 0 ;w but the fo11ner is more 2. Simple cell membrane barrier
unionized at blood pH and therefore distributes rapidly. The influence of .· _/. Blood-brain barrier
. - -
drug pKa and K 0 ;w on distribution is illustrated by the example that 4. Cerebrospinal fluid barrier
thiopental, a nonpolar, lipophilic drug, largely unionized at plasma pH, ~ . Placental barrier
readily diffuses into the brain whereas penicillins which are polar, water- 6. Blood-testis barrier
soluble and ionized at plasma pH do not cross the blood-brain barrier The Simple Capillary Endothelial Barrier : The membrane of capil-
(Fig. 3 .2). laries that supply blood to most tissues is, practically speak~ng: not a
Since the extent to which a drug exists in unionized fonn governs the barrier to moieties which we call drugs. Thus, all drugs, Jontzed or
distribution pattern, situations that result in alteration of blood pH affect unionized, with a molecular size less than 600 daltons, diffuse through the
such a pattern; for example, acidosis (metabolic or respiratory) results in capillary endothelium and into the interstitial fluid. Only drugs bou.nd to
decreased ionization of acidic drugs and thus increased intracellular drug the blood components are restricted because of the large molecular stze of
concentration and phannacologic action. Opposite is the influence of the complex.
alkalosis. Sodium bicarbonate induced alkalosis is sotnetimes useful in
' The Simple Cell Membrane Barrier : Once a drug diffuses from the
the treatment of barbiturate (and other acidic drugs) poisoning to drive the capillary wall into the extracellular fluid, its further entry into cell~ of
drug out and prevent further entry into the CNS and promote their urinary most tissues is limited by its permeability through the membrane that hnes
excretion by favoring ionization. Converse is true for basic drugs; acido- such cells. Such a simple cell membrane is similar to the lipoidal barrier
sis favors extracellular whereas alkalosis, intracellular distribution. in the GI absorption of drugs (di!~cussed in chapter 2).
In case of polar drugs where permeability is the rate-limiting step in The physicochemical properties that influence permeation of drugs
the distribution, the driving force is the effective partition coefficient of across such a barrier are summarized in Fig. 3.3 .
drug. It is calculated by the following fortnula:
capillary wall cell membrane
:c: • . _Fraction unionized K0 ;w of unionized Intracellular
Ef tectJve K0 ;w - at pH 7 _4 X drug (3.1)
ECF Fluid
Blood

The extent to which the effective partition coefficient influences rapid- Drugs of size less @-~;-+- ® Bulk flow
ity of drug distribution can be seen from ~he example given in Table 3 .1. than 50 daltons

TABLE 3.1
Distribution of Acidic Drugs in CSF
Drug Relative acidity Effective K0 ;w Relative rate
at pH 7. 4 of distribution
@•_.""'""~• Polar/ionized drugs (§/
of size ~ 50 daltons .
Thiopental weaker acid 2.0 80 ~arner

@ @)~
Salicylic acid stronger acid 0.0005 1 U///

Thus, thiopental distributes in CSF at a rate 80 times faster than


Fig. 3.3 Plasma membrane barrier and drug diffusion across it
salicylic acid.
80 BIOPHARMACEUTICS AND PHARMACOKINETICS DISTRIBUTION OF DRUGS 81

/ Blood-Brain Barrier (BBB) : Unlike the capillaries found in other (l11e selective permeability of lipid soluble moieties through th~ BBB
parts of the body, the capillaries in the brain are highly specialized and makes appropriate choice of a drug to treat CNS disorders an essential
tnuch less penneable to water-soluble drugs. The brain capillaries consist part of therapy; for example, Parkinsonism, a disease characterized by
c~f endothelial cells which are joined to one another by continuous tight depletion of dopamine in the brain, cannot be treated by administration nf
Intercellular junctions comprising what is called as the blood-brain bar- dopamine as it does not cross the BBB.1 Hence, levodopa, which car·
- -
rie~ (Fig. 3.4). Moreover, the presence of special cells called as astrocytes, penetrate the CNS where it is metaboli.ied to dopamine, ts used in its
which are the elements of the supporting tissue found at the base of treatment. Targeting of polar drugs to brain in certain conditions such as
endothelial membrane, fonn a solid envelope around the brain capillaries. tumor ha always been a problem. Three different approaches have been
As a result, the intercellular passage is blocked and for a drug to gain utilized successfully to promote crossing the BBB by drugs:
access from the capillary circulation into the brain, it has to pass through j--- Use of penneation enhancers such as dimethyl sulfoxide (DMSO)
the cel~s rather than between them) (However, there are specific sites in
the ~rain where the BBB does not exist, namely, the trigger area and the iY. Osmotic disruption of the BBB by infusing internal carotid
median hypothalamic eminence. Moreover, drugs administered intrana- artery with mannitol

sally may diffuse directly into the CNS because of the continuity between ~- Use of dihydropyridine redox system as drug carriers to the
submucosal areas of the nose and the subarachnoid ~pace of the olfactory brain
lobe).
In the latter case, the lipid soluble dihydropyridine is linked as a
Brain carrier to the polar drug to form a prodrug that readily crosses the BBB.
In the brain, the CNS enzymes oxidize the dihydropyridine moiety to the
polar pyridinium ion form that cannot diffuse back out of the brain. As a
_ _ ECF of brain result, the drug gets trapped in the CNS. Such a redox system has been
used to deliver steroidal drugs to the brain (see chapter 6 on Prodrugs).
_ _ Glial cell
Blood-Cerebrospinal Fluid Barrier : The cerebrosp~nal
fluid (CSF)
is formed mainly by the choroid plexus of the lateral, third and fourth
_ _ Basement
ventricles and is similar in composition to the ECF of brafu. The capil-
membrane
lary endothelium that lines the choroid plexus have open junctions or gaps
Capillary
endothelium CSF
Blood
Tight intercellular
Highly lipid- junction ~-Choroidal cells
soluble drugs
..___ _ _ _ _ _ _ Tight intercellular
Fig. 3.4 Blood-brain barrier
ECF junction
. Since the 888 is a lipoidal barrier, it allows only the drugs having
I I [ Ill...__,_l Capillary endothelium
hptd ~olub!e ~~d partially ionized molecules penetrate at a slow rate The Highly lipid- ·Open junction
effe~ttve partition coefficient of thiopental, a highly lipid soluble -drug is -
Blood e~ e soluble drugs

50 times that of pentobarbital and crosses tht?-.888 much more rapidly. Fig. 3.5 The blood-CSF barrier
Pol~r nat~ral substances such as sugars and amino acids are transported to
bram act1ve1J Thus, structurally similar foreign molecules can also pen- and drugs can flow freely into the extracellular space between the .capil-
etr~te the 888 by the same mechanism. Most antibiotics such as penicillins lary wall and the choroidal cells. However, the choroidal cells are joined
which are polar, water-soluble and ionized at plasma pH, do not cross the to each other by tight junctions founing the blood-CSF barrier which has
· 888 under nonnal circumstances. pettneability characteristics similar to that of the BBB (Fig. 3 .5).
82 BIOPHARMACEUTICS AND PHARMACOKINETICS DISTRIBUTION OF DRUGS 83 . .
As in the case of BBB, only highly lipid so.luble drugs can cross the Drugs are particularly dangerous to the fetus during 2 stages.-
~lood-CSF barrier with relative ease whereas moderately lipid soluble and '
i. In the frrst trimester when the fetal organs develop; it is during
partially ionized drugs penneate slowly. A drug that enters the CSF
this stage that most drugs show their teratogenic effects (con.gerii~ ·
slowly cannot achieve a high concentration as the bulk flow of CSF
~tal defects) e.g. thalidomide, phenytoin, isotretinoin, 7testosterone,
continuously removes the drug. For any given drug, its concentration in
methotrexate, etc.
rhe brain will always be higher than in the CSF.
1

ii. In the latter stages of pregnancy when drugs are known to affect
Although the mechanisms for diffusion of drugs into the CNS and
physiologic functions, e.g. respiratory depression by morphine. :
· CSF are similar, the degree of uptake may ·vary significantly. In some
__ cases, ·CSF drug concentration m~y be higher than its cerebral concentration It is, therefore, always better to restrict all drugs during pregnancy
e.g. sulfamethoxazole and trimethoprim, and vice versa in other cases, e.g. because of the uncertainty of their hazardous effects.
.
certain ~-blockers. .. Blood-Testis Ba~rier : . This barrier is located not at the -capillary
v / ·Placental Barrier : The maternal and the fetal blood vessels are endothelium level but at sertoli-sertoli cell junction. It is the tight junc-
separated by a number of tissue layers made of fetal trophoblast basement tions between the neighboring sertoli cells that act as the blood-testis
me~brane and the endo~helium which together constitute the placental barrier. This barrier restricts the passage of drugs to spennatocytes and
barrier. The flow of blood in the maternal and the fetal blood . vessels is spermatids.
shown in Fig. 3 .6.
ORGANffiSSUE SIZE AND PERFUSION RATE
As discussed until now, distribution is permeability rate-Umited in
the following cases:
_ _ _ Fetal artery .
I •
i. When the drug under consideration is ionic, polar or water soluble

___
...._ Fetal membrane
ii. Where the highly selective physiologic barriers restrict the diffu-
sion of such drugs to the inside of the cell
(trophoblast
Drug movement + endothelium) In contrast, distribution will be perfusion rate-limited when:

~---~ _ _ Maternal artery i. The drug is highly lipophilic


· ii. The membrane across which the drug is supposed to diffuse is
--Maternal vein highly permeable such as those of the capillaries and the muscles.
Wher~as only highly lipophilic drugs such as thiopental can cross the
Fig. 3.6 Placental barrier and blood flow across it most selective of the barriers like the BBB, highly permeable capillary
,
- wall pennits passage of almost all drugs (except those bound to plasma _
The human placental barrier has a mean thickness of 25 microns in proteins). In both circumstances, the rate-limiting step is the rate of blood
early p~~nancy that reduces to 2 microns at full term .which however flow or perfusion to the tissue. Greater the blood flow, faster the: , --
does not reduce its e~fectiveness. Many drugs having molecular weight d istri bu ti on.
less than 1000 daltons' and moderate to high lipid solubility e.g. ethanol,
sulfonamides, barbiturates, gaseous anesthetics, steroids, narcotic analgesics, Perfusion rate is defined as the .volu_m e of blood that flows per unit
anticonvulsants and some antibiotics, cross the barrier by simple diffusion time per unit volume of the tissue. It is expressed in mVminlml of the-·--
quite rapidly. This shows that the placental barrier is not as effective a - tissue. 'The perfusion rate of various tissues are given in- Table 3.2.
barrier as BBB. Nutrients essential for the fetal growth are transported by In Table 3 .2, the various tissues are listed in decreasing order of their
carrier-mediated processes. Immunoglobulins are transported by endocytosis. perfusion rate which indicates the rapidity with which the drug will be ,
distributed to the tissues. Highly perfused tissues such as lungs, kidneys,

. -
.
'

-
r ••

"'
84 BIOPHARMACEUTICS AND PHARMACOKINETICS DISTRIBUTION OF DRUGS -·--··85 .

,adrenal, liver, heart and brain are rapidly equilibrated with lipid soluble ·towards equilibrium, the drug rapidly diffuses out of ~he brain and local-
drugs. · · izes in the adipose tissue whose volume is more than 5 times that of brain
and has greater affinity for the drug. The result is rapid tennination of
TABLE 3.2
action of thiopental due to such a tissue redistribution.
~~lative
Volume of Different Organs, Blood Flow and Perfusion Rate
· ·under Basal Conditions Assuming the Total Bod·y 'Volume to be 70 liters
Organ/tissue · % of Body
·----------------------
Blood Flow % of Cardiac Perfusion
BINDING OF DRUGS .TO TISSUE COMPONENTS
A drug in the body can bind to several components such as the plasma
. Volume (ml/min) Output Rate
proteins, blood cells and hemoglobin (i.e. blood components) and ex-
(ml/minlml)
travascular proteins and other tissues. This topic is dealt comprehensively
I. Highly Perfused in chapter 4 on Protein Binding of Drugs.
I. Lungs 0.7 5000 100.0 10.2
2. Kidneys 0.4 1250 25.0 4.5
''· MISCELLANEOUS FACTORS AFFECTING DRUG DISTRIBUTION .
3. Adrenals 0.03 25 0.5 1.2
4. Liver 2.3 1350 27.0 ~
0.8 Age. ·.
5. Heart 0.5 200 4.0 ' 0.6 Differenees in distribution pattern of a 'd rug in different age groups are
6. Brain 2.0 700 14.0 0.5
\
mainly d~e to differences in-
II. Moderately Perfused a. Total body water (both intracellular and extracellular) is much
7. Muscles 4.2.0 1000 20.0 0.034
greater in infants ,..
8. Skin 15.0 350 7.0 0.033
b. Fat content is also higher in infants and elderly
. III. Poorly Perfused ..
9. Fat (adipose) 10.0 200 4.0 0.03 c. Skeletal muscles are lesset itl infants and in elderly
'
10. Bone (skeleton) 16.0 250 5.0 0.02 d. Organ composition the BBB is poorly developed in infants, the
'
myelin content is low and cerebral blood flow is high, hence
If ~tlb. is t~e tissue/blood partition coefficient of drug then the frr~t­ greater penetration of drugs in the brain
-- f order dtstr1but1on rate constant, Kt, is given by following equation:
e. Plasma protein content - loW albumin content in both infants
Perfusion Rate and in elderly
Kt=----- (3.2) •
_ Kt!b Pregnancy
The tissue distribution half-life is given by equation: During pregnancy, the growth of uterus, placenta and fetus· increases

. .b .· 0.693 0.693 Kt/b


the volume available for distribution of drugs. The fetus represents a.
DIStri ution half-life= = --- · --- 33 separate compartment in which a drug can distribute. The plasma and the·
K1 Perfusion Rate- ( . ) ECF volume also increase but there is a fall in albumin content.
- The extent to wh_ich a drug is distributed in a particular tissue or organ
Obesity , .·-
depends upon the Size of the tissue (i.e. tissue volume) and the tissue/
In obese persons, the high adipose tissue content can take up a large ' ·.

bl?o~-- partition coefficient of the drug. Consider the classic example of


fraction of lipophilic drugs despite the fact that perfusion through it is
t~Iopental. This lipophilic drug has a high tissue/blood partition coeffi-
low. The high fatty acid levels in obese persons alter the bindmg charac-
cient towards the brain and still higher for adipose tissue. Since the brain
teristics of acidic drugs.
(site _of a~tion) is.a highly per~sed organ, following i.v. injection, thiopental
r~adily dti(fuses Into the brain showing a rapid onset of action. Adipose Diet
tls~ue being poorly perfused, takes longer to get distributed with the same A diet high in fats will increase the free fatty acid levels in circulation
drug. But as the concentration of thiopental in the adipose proceeds '
thereby affecting binding of acidic drugs such as NSAIDs to albumin.
86 BIOPHARMACEUTICS AND PHARMACOKINETICS DISTRIBUTION OF DRUGS 87
Disease States
A number of mechanisms may be involved in the alteration of drug or (3.5)
distribution characteristics in disease states:
The apparent volume of distribution bears no direct relationship with
a. Altered albumin and other drug-binding protein concentration
the real volume of distribution.
b. Altered or·reduced perfusion to organs or tissues
The real volume of distribution has direct physiologic meaning and
c. Altered tissue pH
is related to the body water. The body water is made up of 3 distinct
. An interesting example of altered penneability of the physiologic bar-

com pat ttnents as shown in the Table 3.3.
riers is that of BBB. In meningitis and encephalitis, the BBB becomes
TABLE 3.3
more pertneable and thus polar antibiotics such as penicillin G and ampi-
cillin which do not nonnally cross it, gain access to the brain. In a patient Fluid Compartments of a 70 Kg Adult
.
suffering from CCF, the perfusion rate to the entire body decreases affect- Body Fluid Volume %of Body % ofTBW
ing distribution of all drugs. (liters) Weight
Drug Interactions 1. Vascular fluid/blood 6 9 15
· Drug interactions that affect distribution are mainly due to differences (Plasma) (3) (4.5) (7.5)
in plasma protein or tissue binding of drugs. This topic is discussed under 2. Extracellular fluid 12 17 28
the same heading in chapter 4. (excluding plasma)
3. Intracellular fluid 24 34 57
(excluding blood cells)
VOLUME OF DISTRIBUTION I
/

'
I
' . A drug in circulation distributes to various organs and tissues. When Total Body Water (TBW) 42 60 100
the process of distribution is complete (at distribution equilibrium), differ-
ent organs and tissues contain varying concentrations of drug which can The volume of each of these real physiologic compartments c.an be
be detertnined by the volume of tissues in which the drug is present. detenn ined by use of specific tracers or markers (Table 3.4). Tbe plasma
Since different tissues have different concentrations of drug, the volume volume can be detettnined by use of substances of high molecular weight
of distribution cannot have a true physiologic meaning. However, there or substances that are totally bound to plasma albumin, for e.g. high
exists a constant relationship between the concentration of drug in plasma, molecular weight dyes such as Evans blue, indocyanine green and 1-131
C, and the amount of drug in the body, X. albumin. When given i.v., these remain confmed to the plasma. The total
blood volume can also be detertnined if the hematocrit is known. The
XocC extracellular fluid (ECF) volume can be detennined by substances that
or easily penetrates the capillary membrane and rapidly distribute throu~h-
(3.4)
out . the ECF but do not cross the cell membranes, for e.g. the Na+, Cl-,
where V d = proportionality constant
, having the unit of volume and popu- Br, SCN- and SO/- ions and inulin, mannitol and raffmose. However,
larly called as apparent volume of distribution. It is defined as the none of these substances are completely kept out of the cells. The ECF
hypothetical volume of body fluid into which a drug is dissolved or volume, excluding plasma is approximately 15 liters. The total body
distributed. It is called as apparent volum~ because all parts of the water (TBW) volume can be determined by use of substances that dis-
body equilibrated with the drug do not have equa~ concentration. tribute equally in all water compartments of the body (both intra- and
Thus, from equation 3:4, V d is given by the ratio: extracellular), for e.g. heavy water (D20), tritiated water (HTO) and lipid
soluble substances such as antipyrine. The intracellular fluid volume is
/"'A pparent Volume _Amount of Drug in the Body detennined as the difference between the TBW and ECF volume. The
.
J
I

of Distribution Plasma Drug Concentration intracellular fluid volume including those of blood cells is approximately
27 liters.
88 BIOPHARMACEUTICS AND PHARMACOKINETICS DISTRIBUTION OF DRUGS 89

TABLE 3.4 Apparent volume of distribution is expressed in liters and so~etimes


Markers Used to Mear;ure the Volume of Real Physiologic Compartments in liters/Kg body weight. The Vd of various drugs range from as low as 3
Physiologic Fluid Markers Used Approximate liters (plasma volume) to as high as 40,000 liters· (much above the total
Compartment Volume (liters) body size). Many drugs have Vd greater than 30 liters. The Vd is a
characteristic of each drug under normal conditions and is altered under
Plasma Evans blue, indocyanine 3 conditions that affect distribution pattern of the drug.
green, I-131 albumin
Erythrocytes Cr-51 2 QUESTIONS
Extracellular fluid Nonmetabolizable saccharides 15 1. Define-(a) disposition, and (b) distribution of drugs. What is the major
like raffinose, inulin, mannitol mechanism for distribution of drugs and what is its driving force?
and radioisotopes of selected 2. Unless distribution occurs, the drug may not elicit pharmacologic response.
ions : Na+, Cl-, Br- , SO/-
Explain.
Total body water D20, HTO, antipyrine 42 3. Why is distribution of a drug not uniform throughout the body? List the
.. . factors influencing drug distribution .
Sin~e the tracers are not bound or negligibly bound to plasma or tissue 4. What are the two major rate-litniting steps in the distribution of drugs?
proteins, their apparent volume of distribution is same as their true vol- Under what circumstances are they applicable?
ume of distribution. The situation is different with most drugs which bind 5. Which physicochemical properties of the drug limit its distribution?
to plasma proteins or extravascular tissues or both. Certain generaliza- 6. Phenobarbital and salicylic acid have almost the same Ko/w but the former
t_ions can be made regarding the apparent volume of distribution· of such shows extensive distribution. Why?
drugs: 7. What is the influence of change in plasma pH on distribution pattern of a
1. Drugs which bind selectively to plasma proteins or other blood drug? Based on pKa values, which drugs are most affected and which will
components, e.g. warfarin (i.e. those that are less bound to extravascular be least affected by a change in plasma pH?
tissues), have apparent volume of distribution smaller than their true 8. What parameter is considered to be the driving force for distribution of
volume of distribution. The V d of such drugs lies between blood volume polar drugs?
and TBW volume (i.e. between 6 to 42 liters); for example, warfarin has a 9. Why cannot the capillary endothelium be considered a barrier to distribution
Vd of about 10 liters. of unbound drugs? What would be the consequence or fate of drugs had it
been a selective barrier?
2. Drugs which bind selectively to extravascular tissues, e.g. chloro-

10. Name the specialized barriers to distribution of drugs .
quine (i.e. those that are less bound to blood components), have apparent
11. Describe the anatomy and physiology of blood brain barrier. What charac-
volume of distribution larger than their real volume of distribution. The
teristics of a drug are necessary to penetrate such a barrier?
Vd of such drugs is always greater than 42 liters or TBW volume; for
example, chloroquine has a Vd of approximately 15,000 liters. Such 12. How do nutrients which are generally polar, make their way into the brain?
drugs leave the body slowly and are generally more toxic than drugs that 13. Polar drugs such as penicillin normally do not cross BBB but do so in
do not distribute deeply into body tissues. meningitis. Explain.
14. Name the three approaches by which a polar drug can be targeted to brain.
Thus, factors that produce an alteration in binding of drug to blood
components, result in an increase in Vd and those that influence drug 15. Drugs that penetrate the CNS slowly may never achieve adequate therapeu-
·- tic brain concentrations. Why?
binding to extravascular components result in a decrease in Vd. Other
fCJ,ctors that may influence Vd are changes in tissue perfusion and penne- 16. Why is the placental barrier not as effective as BBB?
ability, changes in the physicochemical characteristics of the drug e.g. 17. In which periods drugs are particularly harmful to fetus in pregnant women?
ionization, changes in the body weight and age and several disease states. 18. How are body tissues classified on the basis of perfusion rate?
19. Thiopental is a highly lipophilic, centrally acting drug. By which route
should it be administered for rapid onset of action? Why? What is the
reason for its rapid termination of action?
'
90 BIOPHARMACEUTICS AND PHARMACOKINETICS

20. Which are the factors responsible for the differences in drug distrib1 .on in
persons of different age groups?
21. What are the various mechanisms involved in the alteration of drug distribu-
tion characteristics in disease states?
22. Define apparent volume of distribution. Why cannot the volume of distribu- 4
tion of a drug have a true physiologic meaning?
23. What ~e the various physiologic fluid compartments of the body?
are thetr volumes and how are they estimated?
What • Protein Binding of Drugs
24. The tracers used to determine the volume of body fluids have Vd same as
their true volume. Why?
A drug in the body can interact with several tissue components of
25. How are the binding characteristics of a drug related to its V d? Explain why
some drugs have V d value larger than TBW volume?
which the two major categories are blood and extravascular tissues. The
interacting molecules are generally the macromolecules such as proteins,
26. It is better to express V d in .liters/Kg body weight. Why?
DNA or adipose. The proteins are particularly responsible for such an
27. Can a drug have two or more Vd values. Explain why? interaction. The phenomena of complex formation with proteins is called
28. The tissue/blood partition coefficient values of a drug and tissue perfusion as protein binding of drugs. The importance of such a binding derives
rates are given in table below: from the fact that the bound drug is both phartnacokinetically as well as
pharmacodynamically inert i.e. a protein bound drug is neither metabo-
Tissues K,;b Perfusion Rate K, Distribution t ~
lized nor excreted nor is it pharmacologically active. A bound drug is
Liver I 0.8 also restricted since it remains confmed to a particular tissue for which it
Brain 4 0.5 has greater affinity. Moreover, such a bound drug, because of its enor-
Muscle 8 0.035 mous size, cannot undergo membrane transport and t~us its half-life is
Fat 40 0 .03 increased.
BLOOD
lJrug binding to
Determine the rate constants for distribution and distribution half-lives. blood components
~~ ~--~

Answer : Kt values (per minute) : liver - OJ~, brain - 0 .125, muscle - 0.0044
and fat - 0.00075 ; distribution t11z values (in minutes): liver - 0.86,

Blood 1-D
cells 1\ f . D- Plasma
proteins TISSUE Tissue
brain - 5.54, muscle - 158.4 and fat - 924. LIVER localisation

29. The V d of fluoxetine is 3000 liters. Calculate -


P-D ~=~· ,:t====l~=======' D-F ree drug ;:::,==t:===t='
D;::::::: o~ ,=====-- P~D
Drug/metabolite + "in plasma 1l
a. The amount of drug in the body when the plasma .c oncentration is 1ng/ml.
Answer : 3 mg.
binding to liver
tissues 1
Enzymes

P-D , ' D + Carrier



D
IReceptor I
b. The plasma concentration when the amount of drug in the body is 2 mg. P-M ' Metabolites Drug binding j
to renal
Answer : 0 .67 ng/ml. tissues D-C
c. The % of drug that is present in plasma.
I
Answer : 0.1 %. ' Active
KIDNEY secretion
30. The V d of three drugs- A, B and C are 12, 42 and 400 liters respectively. \(;
'IV 'IV
a. Determine the % drug present outside plasma. Biliary • Excretion ; Pharmacologic
~------~, ~~
Response
Answer : drug A - 75.% , drug B - 93% ~nd drug C - 99.3%. Excret1on in Urine

b. Which drug is most extensively distributed in e. v. tissues? Fig. 4.1 Protein-drug binding: Binding of drugs to various tissue components
and its influence on disposition and clinical response. Note that only
c. If binding of drugs is negligible, which of the three can be used as markers
and for which fluid compartments? the unbound drug moves reversibly between the compartments.

91

I
92 BIOPHARMACEUTICS AND PHARMACOKINETICS· PROTEIN BINDING OF DRUGS 93
Binding of drugs generally involves weak chemical bonds such as
Protein Molecular Concentration Drugs that bind
hydrogen bonds, hydrophobic bonds, ionic bonds or van der Waal's forces
Weight (g%)
and, therefore, is a reversible process. Irreversible drug binding, though
rare, arise_s as a result of covalent binding and is often a reason for the at-Globulin 59,000 0.003-0.007 steroids like corticoster-
carcinogenicity or tissue toxicity of the drug; for example, covalent bind- one, and thyroxine and
ing of chloroform and paracetamol metabolites to liver results in hepatotoxicity. cyanocobalamin
Binding of drugs falls 1,34,000 0.015-0.06 vitamins A, D, E and K
.. into 2 classes: and cupric ions
1. Binding of dtugs to blood components like-
Hemoglobin 64,500 11-16 phenytoin, pentobarbital,
a. Plasma proteins
and phenothiazines
b. Blood cells ..
2. Binding of drugs to extravascular tissue proteins, fats, bones, etc. Binding of Drugs to Human Serum Albumin
The influence of binding on drug disposition and clinical response is The human serum albumin (HSA), having a molecular ·weight of
shown in Fig. 4.1. 65,000, is the most abundant plasma protein (59% of total plasma and 3.5
Of all types of binding, the plasma protein-drug binding is the most to 5.0 g%) with a large drug binding capacity. The therapeutic doses of
significant and most widely studied. most drugs are relatively much smaller and their plasma concentration do
not norrnally reach equimolar concentration with HSA. The HSA can
bind several compounds having varied structures. Both endogenous com-
BINDING OF DRUGS TO BLOOD COMPONENTS pounds such as fatty acids, bilirubin and tryptophan as well as drugs bind
Plasma Protein-Drug Binding to HSA. A large variety of drugs ranging from weak acids, neutral
compounds to weak bases bind to HSA. Four different sites on HSA have
Following entry of a drug into the systemic circulation, the first thing
been identified for drug-binding (Fig. 4.2). They are:
with which it can int~ract are blood components like plasma proteins,
· :Jlood cells and hemoglobin (see Table 4.1). The main interaction of drur. Site I : Also called as warfarin and azapropazone binding site, it
in the blood compartment is with the plasma proteins which are present h. represents the region to which large number of drugs are bound, e.g.
abundant amounts and in large variety. The binding of drugs to plasma several NSAIDs (phenylbutazone, naproxen, indotnethacin), sulfonamides
proteins is reversible. The extent or order of binding of drugs to various (sulfadimethoxine,
.
sulfamethizole), phenytoin, sodium valproate and bil-
plasma proteins is: albt:min > a1-acid glycoprotein > lipoproteins > irubin.
globulins.
Site II : It is- also called as the diaz~pam_ ~ind!~ ~ite. - Drugs
- which
TABLE 4.1 ....

bind to this region include benzodiazepines, medium- . chain - _...fatty acids,


Blood Proteins to which Drugs Bind
ibuprofen, ketoprofen, tryptophan, cloxacillin, probenicid, etc.
Protein Molecular. Concentration Drugs that bind Site I and site II are n!sponsible for the binding of most drugs.
Weight (g%)
Site III : is also called as digitoxin binding site.
Human Serum 65,000 3.5-5.0 large variety of all types
Albumin of drugs Site IV : is also called as tamoxifen binding site.
a1-Acid 44,000 0.04-0.1 basic drugs such as Very few drugs bind to .sites III and IV.
Glycoprotein imipramine, lidocaine, A drug can bind to more than one site in which case the main binding

quinidine, etc. site is called as the primary site and the other as the secondary site; for
Lipoproteins 200,000 to variable basic, lipophilic drugs example, site I is the primary site for dicoumarol and site II the secondary
3,400,000 like chlorpromazine site. Groups of drugs that bind to the same site, compete with each other
.. .continued for binding, but drugs that bind to one site do not competitivery inhibit
94 BIOPHARMACEUTICS AND PHARMACOKINETICS PROTEIN BINDING OF DRUGS 95

binding of drugs to other sites. However, they may either promote or and proteins). Predictably, VLDL is rich in triglycerides and HDL is rich
retard binding of a drug to another site by energetic coupling mechanisms. in apoproteins.
The binding of drugs to lipoproteins is noncompetitive. A number of .,
acidic ( diclofenac), neutral (cyclosporin A) and basic drugs (chlorpromazine)
Warfarin Binding Site
bind to lipoproteins. Basic, lipophilic drugs have relatively more affinity.
Lipoprotein binding becomes significant in cases of drugs that predomi-
Diazepam Binding Site nantly bind to them, and secondly, when levels of HSA and AAG in
plasm a are decreas·ed.
Digitoxin Binding Site
Binding of Drugs to Globulins
Several plasma globulins have been identified and are labeled as a 1-,
T amoxifen Binding Site a2-, Pt-, P2- andy-globulins.
1. a 1-globulin : also called as transcortin or CBG (corticosteroid
Fig. 4.2 Four major drug binding sites on human serum albumin binding globulir:), it binds a number of steroidal drugs such ~s cortisone
and prednisone. It also binds to thyroxine and cyanocoba1amm.
Binding of Drugs to a 1-Acid Glycoprotein (a 1-AGP or AAG)
2. a 2-globulin :- -also called as cerulopl~min, it binds vitam ins A,
Also called as the orosomucoid, it has a molecular weight of 44,000 D, E and K and cupric ions.
and a plasma concentration range of 0.04 to 0.1 g%. It binds to a
number of basic drugs like imipramine, amitriptyline, nortriptyline, lidocaine, 3. p1-globulin : also called as transferrin, it binds to ferrous ions.
propranolol, quinidine and disopyramide. 4. p2;..globulin : binds to carotinoids .

Binding of Drugs to Lipoproteins 5. y-globulin : binds specifically to antigens.


Binding of drugs to HSA and AAG involve hydrophobic bonds. Since Binding of Drugs to Blood Cells
only lipophilic drugs can undergo hydrophobic bonding, lipoproteins can
More than 40% of the blood comprises of blood cells of which the
also bind to such drugs because of their high lipid content. However, the
major cell component is the RBC. The RBCs constitute 95% of the total
plasma concentration of lipoproteins is much less in comparison to HSA
and AAG. blood cells. Thus, significant RBC drug binding is possible. The red cell
is 500 times in diameter as the major plasma protein binding component,
A drug that bind~ _~o lipoproteins does so by dissolving i~__the lipiC. albumin. The RBC comprises of 3 components each of which can bind to
core of the protein and thus its capacity to bind depends_. upon its lipi<. drugs:
l.:Ontent. The molecular weight of lipoproteins vary from 2 lakhs to 3L;
1. Hemoglobin : It has a molecular weight of 64,500 (almost equal
lakhs depending on their chemical composition. They are classified on
the basis of their density. to that of HSA) but is 7 to 8 times the concentration of albumin in blood.
Drugs like phenytoin, pentobarbital and phenothiazines bind to hemoglo-
The 4 classes of lipoproteins are: bin.
1. Chylomicrons -
2. Carbonic Anhydrase : Drugs known to bind to it are acetazolamide
2. Very low density lipoproteins (VLDL) and chlorthalidone (i.e. carbonic anhydrase inhibitors).
3. Low density lipoproteins (LDL) (predominant in humans) 3. Cell Membrane : Imipramine and ~hlorpromazine are reported to
4. High density lipoproteins (HDL) bind with the RBC membrane.
The lipid core pf these macromolecules consists of triglycerides and It has been shown that the rate and extent of entry into RBC is more
cholesteryl esters and the outside is made of apoproteins (free cholesterol for lipophilic drugs, e.g. phenytoin. Hydrophilic drugs like ampicillin do
not enter RBC .


96 BIOPHARMACEUTICS AND PHARMACOKINETICS
97
PROTEIN BINDING OF DRUGS

TISSUE BINDING OF DRUGS 6. Hairs : . Arsenicals, chloroquine and phenothiazines are . reported
(TISSUE LOCALIZATION OF DRUGS) to deposit in hair shafts.
·-
The body tissues, apart from HSA, comprise 40% of the body weight . 7. Bones : Tetracycline is a well known example of a drug that
-- - - - - - --
which is 100 times that of HSA. Hence, tissue-drug binding is much binds to bones and teeth. Administration of this antibiotic · to infa~ts or
more significant than thought to be. children during odontogenesis results in pennanent brown-yellow dt~col­
A drug can bind to one or more of the several tissue components. oration of teeth. Lead is known to replace calcium .~om bones and cause
Tissue-drug binding is important in distribution from two viewpoints: their brittleness.
,,

firstly, it increases the apparent volume of distribution of drugs in contrast 8. Fats : Lipophilic drugs such as thiopental and the pest.ici~e DDT
to plasma protein binding which decreases it; this is because the parameter accumulate in adipose tissues by partitioning inlo it. . Howe~e~,. ht~h o/Yf
is related to the ratio of amount of drug in the body to the plasma · partition coefficient is not the only criteria for ad_tpose dtstnb~tiOn of
concentration of free drug and the latter is dec.reased under conditions of drugs since several highly lipophilic (more than .th1op~ntal) basic _~gs
extensive tissue binding of drugs, and secondly, tissue-drug binding re- like imipramine and chlorpromazine are not locahzed tn fats. The poor
sults in localization of a drug at a specific site in the body (with a perfusion of adipose could be the reason for _such an ambi~ui~. Reports
subsequent increase in biological half-life). This is .more so because a have stated that adipose localization of drugs IS a resul~ of btn~tng compe-
number of drugs bind irreversibly with the tissues (contrast to plasma tition between adipose and non-adipose tissues (lean ttssues hke muscles,
protein-drug binding); for example, oxidation products of paracetamol, skin and viscera) and not partitioning.
phenacetin, chloroform, carbon tetrachloride and bromobenzene bind co- 9. Nucleic Acids : Molecular components of cells such as . ON~
valently to hepatic tissues. interact strongly with drugs like chloroquine and quinacrine resulting In
Factors infl~encing localization of drugs in tissues include lipophilicity distortion of its double helical structure .
. and structural features of the drug, perfusion rate, pH differences, etc.
£xtensive tissue-drug binding suggests that a tissue can act as the storage FACTORS AFFECTING PROTEIN-DRUG BINDING
site for drugs. Dru~ _t~~t bind to both tissue and plasma components
Factors affecting protein-drug binding can be broadly categorized as
result in competition between drug binding sites. -
1. Factors relating to the drug
. For majority of drugs that bind to extravascular tissues, the order of ·
a. Physicochemical characteristics of the drug
binding is: liver > kidney > lung > muscle. Several examples of ex-
travascular tissue-drug binding are: b. Concentration of drug in the body

c. Affmity of a dNg for a particular binding component
1. Liver : As stated earlier, epoxides of a number of halogenated
2. Factors relating to the protein and other binding components .
hydrocarbons and paracetamol bind irreversibly to liver tissues resulting in
a. Physicochen1ical characteristics of the protein or binding agent
hepatotoxicity.
b. Concentration of protein or binding component {
2. _L ungs : .Basic drugs like imipramine, chlorpromazine and anti- c. Number of binding sites on the binding agent
histamines accumulate in iungs.
3. Drug interactions . . . .
3. Kidneys : Metallothionin, a protein present in kidneys, binds to a. Competition between drugs for the bmdtng stte (d1~placement
heavy metals such as lead, mercury, and cadmium and results in their
. interactions)
renal accumulation and toxicity. b.
Competition between drugs and norrnal body constituents
4. Skin : Chloroquine and phenothiazines accumulate in skin by ' c. Allosteric changes in protein molecule
'
interacting with melanin. 4.' Patient related factors
5. Eyes : The retinal pigments of the eye also contain melanin. a. Age
Binding of chloroquine an<t phenothiazines to it is responsible for retinopathy. b. Intersubject variations

c. Disease states

..
98 BIOPHARMACEUTICS AND PHARMACOKINETICS
PROTEIN BINDING OF DRUGS
DRUG RELATED FACTORS .
Number of Bindine Sites on the Protein
PhysicochemicaJ Characteristics of the Drug Albumin has a large number of binding sites as compared to other
As mentio~ed earlier, protein binding is directly related to the lipophilicity proteins and is a high capacity binding component. Several <Jrugs are
of drug. An mcrease in lipophilicity increases the extent of binding; for capable of binding at more than one site on albumin, e.g. fluocloxacillin,
exam~le, ~h~ s~ow .absorption of cloxacillin in comparison to ampicillin flurbiprofen, ketoprofen; tamoxifen and dicoumarol bind to both prunary
a~er.I.m. InJectt~n Is attributed to its higher lipophilicity and larger (95%) and secondary sites on albumin. Indomethacin is known to· bind to 3
btndtng to protems while the latter is less lipophilic and just 20% bound different sites. AAG is a protein with limited binding capacity beca1:1se of
to proteins. Highly lipophilic drugs such as thiopental tend to localize in · its low concentration and low molecular size. Though pure AAG. has
adipose tissues. Anionic or acidic drugs such .as penicillins and sulfon- only one binding site for lidocaine, in presence of HSA, two binding sites
amides bind more to HSA whereas cationic or basic drugs such as imipramine have been reported which was suggested to be due to direct interac~ion
and alprenolol bind to AAG. Neutral, unionized drugs bind more to between HSA and AAG.
lipoproteins.
Concentration of Drug in the Body DRUG INTERACTIONS
The extent of protein-drug binding can change with both changes in Competition Between Drugs for the Binding Sites
drug as well as protein concentration. The concentration of drugs that (Disp.lacement Interactions)
bind to HSA does not have much of an influence as the therapeutic When two or more drugs can bind to the same si~e, competition
_concentration of any drug is irisufficient to saturate it. However, thera- between them for interaction with the binding site results. If one of the
peutic concentration of lidocaine can saturate AA G with which it binds as drugs (drug A) is bound to such a site, then administration of another
the concentration of AAG is much less in comparison to that of HSA in drug (drug B) having affinity for the same site results in displacement of
blood. drug A from its binding site. Such a drug-drug interaction for the
common binding site is called as displacement interaction. The drug A
Drug-Proteinffissue Affinity here is called as the displaced drug and drug B as the displacer. Warfa-
Lidocaine has greater affinity for AAG than for HSA. Digoxin has rin and phenylbutazone have same degree of affmity for HSA. Adminis- .
more affinity for proteins of cardiac muscl~s than those of skeletal muscles tration of phenylbutazone to a patient on warfarin therapy results in
or plasma. Iophenoxic acid, a radioopaque medium, has so great an displacement of latter from its binding site. The free warfarin may cause
affmity for plasma proteins that it has a half-life of 2~ years. adverse hemorrhagic reactions which may be lethal. Phenylbutazone is
also known to displace sulfonamides from their HSA binding sites. Dis-
PROTEINffiSSUE RELATED FACTORS placement interactions can result in unexpected rise in free concentration
of the displaced drug which may enhance clinical response or toxicity.
Physicochentical Properties of Protein/Binding Component
Even a drug metabolite can affect displacement interaction.
· Lipoprotein~ and adipose tissue tend to bind lipophilic drugs by dis-
solving them in their lipid core. The physiologic pH determines the
presence of active aniotiic and cationic groups on the albumin molecules Clinically significant interactions will result when:
to bind a variety of drugs. I. The displaced drug (e.g. warfarin) -
a. is more than 95% bound
Concenttati~n of Protein/Binding Component
b. has a small volume of distribution (less than 0.15 L/Kg)
Among the plasma proteins, binding predominantly occurs with albu- c. shows a rapid onset of therapeutic or adverse effects
min as it is present in ·a higher concentration in comparison to other ••
d. has a narrow therapeutic index
plasma proteins. The amount of several proteins and tissue .components
available for binding, changes during dtsease states. This effect will be II. The displacer drug (e.g. phenylbutazone)- .
discussed in the subsequent sections. a. has a high degree of affmity as the drug to be displaced
\
b. competes for the same binding sites·
100- BIOPHARMACEUTICS AND PHARMACOKINETICS PROTEIN BINDING OF DRUGS 101

c. ·the drug/protein·· concentration ratio is high (above 0.10), and pathologic (diabetes, myocardial infarction, alcohol · abstinence) and
'

. d~ s~ows a rapid and large increase in plasma drug concentration. pharmacologically induced conditions (after heparin and caffeine

administration). The fatty acids, which also bind to albumin, influence
It will be worthwhile to mention here that, both the concentration of I

binding of several benzodiazepines and prop~:anolol (decreased binding)


\t he· (iisplacer drug and its. affinity for the binding site. with respect to that
and warfarin (increased . binding). Bilirubin bindiD.g to HSA can be . ~paired
of the drug to be displaced, will detetrnine the extent to which displace-
by .certain drugs and is of great concern ~ neonates ·whose BBFf and "
.ment will occur.
bilirubin metabolizing ~apacity are not very efficient. Acidic drugs such
For a drug that is 95% bound, a displacement of just 5% of the bound as sodium salicylate, sodium benzoate and sulfonamides displace bilirubin
drug results in a I 00% rise in free drug concentration. If the displaced from its albumin binding site. The free bilirubin is not conjugated by the
drug has a small volume of distribution, it remains confmed to the blood ~iyer of the neonates and thus crosses the ·BBB and precipitates the
compartment and shows serious toxic responses. On the contrary, if such condition called as kernicterus (characterized by degeneration of brain
a drug has a large Vd, it redistributes into a large volume of body fluids and mental retardation).
and clinical effects may be negligible or insignificant. The increase in
free drug concentration following displacement also makes it more avail- All.o steric Changes in Protein Molecule
able for elimination by the liver and the kidneys (Fig. 4.3). If the drug is This is y~t another mechanism by which drugs can affect ·prote·in
easily metabolizable or excretable, its displacement results in significant. binding interactions. The process involves alteration of the protein stnic- .
reduction in elimination half-life. ture by the drug or its metabolite thereby modifying its binding capacity_! .

The agent that produces such an effect is called as allosteric effector, e.g.
'
Enhanced aspirin acetylates the lysine fraction of albumin thereby modifying its
SITE OF ACTION pharmacologic
capacity to bind NSAIDs like phenylbutazone (increased affinity) and
RECEPTOR . response
flufenamic acid (decreased affinity).

PATIENT RELATED FACTORS


STORAGE .Tissue
BLOOD TISSUE redistribution Age
Increase in
I free drug .
concentraJion
Modification in protein-drug binding as influenced by age of the pa-.
D
'
,I P-0
t •
(Disglaced
tient is mainly due to differences in the protein content in various age
'

D1 (Displaeer) Drug) grOUP.S.


Hepatic
LIVER clearance i. Neonates : Albumin content is low in newborn; as a result, the
. unbound concentration of drug that primarily bind to albumin, for ex-
, _ample phenytoin and diazepam, is increased.
\

Renal ii. Young infants : An interesting example of differences in protein-


KIDNEY
clearance drug binding in infants is that of digoxin. Infants suffering from congestive
cardiac failure are given a digitalizing dose 4 to 6 times the adult dose on
0 •
- ·- -· -~-' --
. · - ~··

Fig. 4.3 Fate of a drug after displacement interaction body weight basis. This is contrary to one's belief that infants should be
given low doses considering their poorly developed drug eliminating sys-
DiSplacement also becomes insignificant with the use of more selec-
tem. The reason attributed for use of a large digoxin dose is greater
tive·, potent, low dose drugs.
binding of the drug in infants (the other reason is abnotrnally large renal
Competition Between Drugs and Normal Body Constituents · clearance of digoxin in infants).
Among. the vario~s not1nal body constituents, the free fatty acids are iii. Elderly : In old age, the albumin content is lowered and free
~o~ to mteract wtth a number of drugs t~at bind primarily to H.SA. concentration of drugs that bind primarily to it is increased. Old age is
The free fatty acid level is increased in several physiologic (fasting), also characterized by an increase in the levels of AAG and thus decreased
I

...··

102 BIOPHARMACEUTICS AND PHARMACOKINETICS PROTEIN BINDING OF DRUGS 103
free c~pcen~ation is observed for ~drugs that bind to it. The situation is SIGNIFICANCE OF PROTEINffiSSUE BINDING OF DRUGS
complex and difficult to generalize · for drugs that bind to both HSA and
AAG, e.g lidocaine and propranolol. Absorption '

, The absorption equilibrium is attained by transfer of free drug from


Intersubject Variations the site of administration into the systemic circulation and when the
Intersubject variability in drug binding as studied with few drugs concentration in these two compartments become equal. Following equi-
showed that the difference is small and no more than two fold. These librium, the)Jrocess may stop. However, binding of the absorbed drug to
djfferences have been attributed to genetic and environmental factors. plasma proteins decreases free drug concentration and disturbs such an
equilibrium. Thus, sink conditions and the concentration gradient are re-
Disease States
established which now act as the driving force for further absorption.
Several pathologic conditions are associated with alteration in protein This is particularly useful in case of ionized drugs which are transported
· content. Since albumin is the major drug binding protein, hypoalbuminemia with difficulty.
can severely impair protein-drug binding. Hypoalbuminemia is caused by
Systemic Solubility of Drugs
several conditions like aging, CCF, trauma, bums, inflammatory states,
renal .a nd hepatic disorders, pregnancy, surgery, cancer, etc. Almost every Water insoluble drugs, neutral endogenous macromolecules such as
serious chronic illness is characterized by decreased albumin content. heparin and several steroids and oil soluble vitamins are circulated and
Some of the diseases that modify protein-drug binding are depicted in distriouted to tissues by binding especially to lipoproteins which act as a
Table 4.2. Hyperlipoproteinemia, caused by hypothyroidism, obstructive vehicle for such hydrophobic compounds.
liver disease, alcoholism, etc., affects binding of lipophilic drugs.
'. - . ~· - - . - Distribution
• I •

·- - •

TABLE 4.2 Influence of Disease States on Protein-Drug Binding Plasma protein binding restricts the entry of drugs that have specific
affinity for certain tissues. This prevents accumulation of a large fraction
Disease Influence on Influence on Protein-Drug
of drug in such tissues and thus, subsequent toxic reactions. Plasma
Plasma Protein Binding protein-drug binding thus favors unifortn distribution of drugs throughout
1. Renal failure decreased decreased binding of acidic drugs; the body by its buffer function (maintains equilibrium between the free
(uremia) albumin content neutral and basic drugs unaffect~d. and the bound drug). A protein bound drug in particular does not cross
2. Hepatic failure decreased albumin decreased binding of acidic drugs; the BB.a , the placental barrier and the glomerulus.
synthesis binding of basic drugs is normal or
reduced depending on AAG levels.
Tissue Binding, Apparent Volume of Distribution and Drug Storage
3. Inflammatory increased AAG
A drug that is extensively bound to blood components remains con-
increased binding of basic drugs;
states (trauma, levels neutral and acidic drugs unaffected. fined to blood. Such a drug has a small volume of distribution. A drug
surgery, bums, that shows extravascular tissue binding has a large volume of distribution.
infections, etc.) A tissue or blood component that has great affmity for a particular drug
·-------------------------------------------------------
Putting in a nutshell, all factors, especially drug interactions and pa-
acts as a depot or storage site for that drug; for example, RBC is a storage
site for the lipophilic compound tetrahydrocannabinol.
tient related factors that affect protein or tissue binding of drugs, influence:
The relationship between tissue-drug binding and apparent volume of
1. Pharmacokinetics of drugs : A decrease in plasma protein--drug distribution can be established as follows:
binding i.e. an increase in unbound drug concentration, favors
tissue redistribution and/or clearance of drugs from the body (en- Amount of drug in the body X
vd = - (4.1)
hanced biotrailsfonnation and excretion). Plasma drug concentration C
2. Pharmacodynamics of drugs : An increase in concentration of free
or, the amount of drug in the body, (4.2)
or unbound dru,& results in increased intensity of action (therapeu-
tic/toxic).
104 BIOPHARMACEUTICS AND PHARMACOKINETICS PROTEIN BINDING OF DRUGS 105
Similarly, we can write, •

from getting filtered through the glo~erulus. Thus, dfl:lgs which are more
Amount of drug in plasma = VP C (4.3) than 95% bound are eliminated slowly i.e. they have long elimination
half-lives; for example, tetracycline, which is only 65% bound, has 3:n.
and, Amount of drug in extravascular tissues = V1 C (4.4)
elimination half-life of 8.5 hours in comparison to 15.1 hours of doxycycline
, The total amount of drug in the body is the sum of amount of drug in which is 93% bound to plasma proteins. However, penicillins have short
plasma and the amount of drug in extravascular tissues. Thus, elimination half-lives despite being extensively bound to plasma proteins.
This is because rapid equilibration occurs between the free and the bound
Vd C = Vp C + V1 C1 (4.5) drug and the free drug is equally rapidly excreted by active secretion in
where Vd = apparent volume of distribution of drug renal tubules.
V p = volume of plasma
Vt = volume of extravascular tissues Displacement Interactions- and Toxicity
C1 = tissue drug concentration As stated earlier, displacement interactions are significant in case of
drugs which are more than 95% bound. This is explained fr()m the
Dividing equation 4.5 with C we get: example given in Table 4.3. A displacement of just 1% of a 99% bound
Vd = Vp+V1.C1/C drug results in doubling of the free drug concentration i.e. a 100% rise.
(4.6)
. For a drug that is bound to a lesser extent e.g. 90%, displacement of 1%
• . ;,tte fraction of drug unbound (fu) in plasma is given as: results in only a 10% rise in free drug concentration which may be
:.. ./~
.
. ---... . Concentration of Unbound Drug in Plasma Cu insignificant clinically.
• fu = - - - - - - - - - - - - - - - - - - (4.7)
Total Plasma Drug Concentration c TABLE 4.3 Influence of Percent Binding and Displacement
on Change in Free Concentration of Drugs
Similarly, fraction of drug unbound to tissues is:
Drug A Drug B
Cut
fut = Ct (4.8) % drug before displacement
I
bound 99 90
Assum~g _that at distribution equilibrium, the unbound or free drug free 1 10
conce_ntration In plasma equals that in extravascular tissues i.e. Cu = Cut' 0
/o drug after displacement
equations 4.7 and 4.8 can be combined to give: bound 98 89
Ct fu free 2 11
--- (4.9) 0
/o increase in free drug concentration 100 10
c fut
Substitution of equation 4.9 in 4.6 yields: Kernicterus in infants is an example of a disorder caused by displace-
ment of bilirubin -from albumin binding sites by the NSAIDs. Another
Vt.fu example discussed earlier was that of interaction between warfarin and
V d=Vp+-- (4.1 0)
fut . phenylbutazone. Yet another example of displacement is that of digoxin
with quinidine. Digoxin represents a drug with a large volume of distri-
From equation 4.10 it is clear that greater the unbound or free concen-
tration of drug in plasma, larger its V d. bution (i.e. shows extensive extravascular tissue binding). Since displacement
interactions may precipitate toxicity of displaced · drug, a reduction in its
Elimination dose may be called for. This may become necessary for a ·drug having a
Only the unbound or free drug is capable of being eliminated. This is small V d such as warfarin since displacement can result in a large increase
1
because _the drug-protein complex cannot penetrat_e into the metabolizing in free drug concentration in plasma. With a drug of large V d such as
organ (hver). The large molecular size of the complex also prevents it digoxin, even a substantial increase in the degree of displacement of drug
in plasma may not effect a large increase in free drug concentration and
PROTEIN BINDING OF DRUGS
107
106 BIOPHARMACEUTICS AND PHARMACOKINETICS

dose adjustment may not be required. This is for two reasons one, only Ka > Kd indicates forward reaction i.e. protein-drug binding is fa- :

a small fraction of such a drug is present in plasma whereas most of it is vored. If PT is the total concentration of protein present, bound and ..
localized in extravascular tissues, and secondly, following displacement, unbound, then:
the free drug, because of its large V d' redistributes in a large pool of PT = [PD] + [P] (4.14)
extravascular tissues. The extent to which t~free plasma drug concen-
If r is the number of moles of drug bound to total moles of protein,
tration of drugs with different V d values will change when displaced,
can be computed from Equation 4.1 0. then,
[PO] [PD]
r=-------- (4.15)
Diagnosis " [PT] [PO] + [P]
The chlorine atom of chloroquine when replaced with radiolabeled I-
131 can be used to visualize melanomas of the eye since chloroquine has Substituting the value of [PO] from equation 4.13 in equation 4.15 we
a tendency to interact with the melanin of eyes. The thyroid gland has get: .
great affmity for iodine containing compounds, hence any disorder of the Ka [P] [D] Ka [D]
r = ------------ (4.16)
same can be detected by tagging such a compound with a radioisotope of Ka [0] + 1
Ka [P] [D] + [P]
iodine.
Equation 4.16 holds when there is only one _b~ding site on the protein
Therapy and Drug Targeting and the protein-drug complex is a 1: 1 complex. If more than one or N .
The binding of drugs to lipoproteins can be used for site-specific number of binding sites are available per mole of the protein then: : ·
delivery of hydrophilic moieties. This is particularly useful in cancer
therapies since certain tumor cells have greater affinity for LOL than N Ka [D]
r= (4.17)
nonnal tissues. Thus, binding of a suitable antineoplastic to it can be Ka [D] + 1
used as a therapeutic tool. HOL are similarly transported more to adrenal
and testes. An example of site-specific drug delivery in cancer treatment The value of association constant, Ka and the number of binding sites
is that of estramustine. Estradiol binds selectively and strongly to pros- N can be obtained by plotting equation 4.17 in three different ways as
trate and thus prostrate cancer can be treated by attaching nitrogen mustard shown below.
to estradiol for targeting of prostrate glands. Drug targeting prevents 1. Direct Plot is made by plotting r versus [D] as shown in Fig. 4.4.
normal .cells from. getting . pestroyed.

Plateau
KINETICS OF PROTEIN-DRUG BINDING N
-----
If P represents proteins and D the drug, then applying law of mass
action to reversible protein-drug binding, we can write:
Ka N/2
P + D~=~PD (4.11) ---
KI r I
I
At equilibrium, [PD] I
Ka = (4.12)
[P] [D] ~ 1/Ka
_____.~ (D]
[PD] = Ka [P] [D] (4.13)
where [P] = concentration of free protein Fig. 4.4 Direct plot of r versus [D]. Note that wheri all the binding sites
[D] = concentration of free drug are occupied by the drug, the protein is saturated and plateau is
[PD] = concentration of protein-drug complex reached. At the plateau, r = N . When r = N/2, [D] = 1/Ka.
Ka = association rate constant •

Kd = dissociation rate constant


108 BIOPHARMACEUTICS AND PHARMACOKil'. -TICS
PROTEIN BINDING OF DRUGS 109
2. Scatchard Plot is made by transfonnil;lg equation 4.17 into a
linear fonn. Thus, · 3. Double Reciprocal Plot (Lineweaver-Burk Plot) : The recipro-
cal of equation 4.17 yields:
N Ka [D]
r=----- (4.17)
Ka [D] + I 1 1 1
----+- (4.19)
r N Ka [D] N
r + r Ka [D] = N Ka [D]
A plot of 1/r versus 1/[D] yields a straight line with slope 1/NKa and
r = N Ka [D] - r Ka [D] y-intercept 1/N (Fig. 4.6).
The~efore, r
--= N Ka -r Ka (4.18) QUESTIONS
[D]
l. A protein bound drug is both pharmacokinetically as well as pharmaco-
. A plot of ~/[D] versus r yields a straight line (Fig. 4.5). Slope of the dynamically inert. Explain.
hne = -Ka, y-Intercept = NKa and x-intercept = N.
2. When is drug-binding considered irreversible? What could be the conse-
quence of such an interaction?
3. Classify the body components to which drugs normally bind.
NKa 4. Why does binding of a drug to plasma proteins occur to a large extent in
comparison to binding to other tissue components?
5. Why is HSA considered a versatile protein for drug binding?
r 6. With examples, name the various drug binding sites on HSA.
- Slope= -Ka
[OJ
7. Binding of drugs to erythrocytes could be as significant as binding to HSA.
i Explain.
'
8. From distribution viewpoint, what is the significance of tissue-drug binding?
N
9. Though imipramine and chlorpromazine are more lipophilic than thtopental,
--+• r
they do not localize in fats. Why?
Fig. 4.5 Scatchard plot for protein-drug binding . 10. List the factors influencing protein binding of drugs .
. 11. Define displacement interaction. Wh~t characteristics of the displacer and
the displaced drug are important for displacement interactions to be clini-
cally significant?
12. Displacement of a drug with a large Vd from its plasma protein binding site
1 may not produce significant toxic reactions. Why?
r 13. How do the acidic drugs such as sulfonamides/NSAIDs precipitate kernicterus
r Slope =1/NKa
in neonates?
14. What is the influence of protein binding and displacement interaction on the
elimination half-life of a drug?
15. Give two reasons for administering large digitalizing dose of digoxin to
infants suffering from CCF? \
'
1 16. What is the influence of various disease states on plasma protein level and
-----+) -
[OJ drug-binding?
17. Hovv would the plasma protein-drug binding influence sink conditions and
Fig. 4.6 Double reciprocal plot for protein-drug binding
absorption of a drug from the G IT?
110 BIOPHARMACEUTICS AND PHARMACOKINETICS

18. Rena~ excretion of penicillins is unaffected by protein-drug binding. Why?


19. Give examples where binding of an agent to a specific tissue can be used
for diagnostic purpose. .
20. How can the principle of binding be used for drug -targeting?
21. Derive the relationship showing that greater the free concentration of drug
5
in plasma, larger its Vd·
22. Warf~in has_ a Vd ot' Mliters in a 70 Kg man and is 90% bound to plasma Biotransformation of Drugs
proteins. Gtven that the plasma/ volume is 3 liters and tissue volume 39
liters, determine -
a. the fraction of drug unbound to tissues. •

Answer : 0. 78. The onset of pharmacologic response depends upon two pharmacoki-
netic processes-drug absorption, and drug distribution (since most sites
b. the % increase in free plasma drug concentration if 10% of bound warfarin of action are in the extravascular tissues). The duration and intensity of
is displaced by phenylbutazone. .
action depend upon the rate of drug removal from the body/site of action
Answer : 90%.
or simply speaking, on the rate of elimination and tissue redistribution of
c. the new V d after the displacement assuming that the tissue binding is drug. Elimination is the major process for removal of a drug from the
unaffected.
body and tertnination of its action. It. is defined as the irreversible loss of
Answer : I2. 5 liters. drug from the body. Elimination occurs by two processes viz. biotransfor-
d. from the answer of question (a), suggest whether the displacement will be mation and excretion.
clinically significant or not.
/ Biotransformation of drugs is defined as the conversion from one
23. For a drug showing protein binding, the two points on the Scatchard plot of chemical form to another. The tenn is used synonymously wi_th metabolism.
r versus r/[D] are: ( 0.6, 1.2 x 104) and ( 1.2, 0. 9 x I 04).
The chemical changes are usually affected enzymically in the body and
a. Determine the association constant. chus, the definition excludes chemical instability of a drug within th~
Answer : Ka = 0.5 x I 04 . · body; for e.g. conversion of penicillin to penicilloic acid by the bacterial '
b. How many binding sites are present on the protein molecule? penicillinase and mammalian enzymes is metabolism but its degradation
Answer : N = 3. by the stomach acid to penicillenic acid is chemical instability.
c. If the points of Scatchard plot are transformed onto the Lineweaver-Burk All chemical substances that are not nutrients for the body and enter
plot (1/r versus 1/[D]), what will be the slope of the line? the body through, ingestion, inhalation or absorption are called as xenobiotics
Answer : Slope = liNKa = 0.67 x I o-4 (Greek: xenos = foreign) or exogenous compounds. Drugs are also
d. Compute the x-axis intercept of Lineweaver-Burk plot. xenobiotics which enter the body by virtue of their lipophilicity. It is
Answer : 1/N = 0.33. interesting to note that for effective absorption, a drug needs to be suffi-
ciently lipid soluble but it is this same physicochemical property that
e. Are the values of Ka ~nd N COf!1puted from both the plots same?
enables it to bypass excretion. This is because only water-soluble agents
undergo renal excretion (major route for exit of drugs from the body)
whereas lipid soluble substances are passively reabsorbed from the renal
tubules into the blood after glomerular filtration. Thus, if such a phenom-
enon continues, drugs would accumulate in the body and precipitate toxic
reactions. However, to prevent such a consequence, the body is armed
with the metabolic system which transfot nts the water insoluble, lipophilic,
nonpolar drugs into polar and water-soluble products that can be easily
excreted by the kidneys and are poorly reabsorbed; for · instance, hippuric

111
112 .
BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 113
acid, the metabolite of benzoic acid, is 2.5 times more water-soluble.
Drug biotransformation is thus a detoxification process. However, ex- Drugs Metabolites
ceptions are there when biotransfonnation leads to products with decreased Pharmacologic Activation
water solubility. TheN-acetyl derivatives of sulfonamides are less water- Inactive (Prodrugs) Active
soluble than the parent drug and thus have a tendency to cause crystalluria. Aspirin Salicy lie acid
Biotransformation : ·= Phenacetin.. Paracetamol
Sulfasalazin.e · Mesalamine and Sulfapyridine
.
normally results in pharmacologic inactivation of drugs, i.e. it results Pivampicillin
'

Ampicillin
in fonnation of metabolites with little or no phannacologic activity; e.g. Enalapril . Enalapri Iat
f

conversion of phenytoin to p-hydroxy phenytoin, Chloramphenicol pal~itate Chloramphenicol


occasionally yields metabolites with equal activity; e.g. conversion of
Change in Pharmacologic ;·A ctivity
phenylbutazone to oxyphenbutazone, and
Iproniazid (antidepressant) · Isoniazid (antitubercular) /
rarely leads to toxicologic activation of drugs, i.e. it results in fotlna- Diazepam (tranquilizer) .Oxazepam (anticonv·ulsant) r \

tion of metabolites with high tissue reactivity; e.g. conversion ofparacetamol


to reactive metabolites that cause hepatic necrosi~. In comparison with xenobiotics, the natural endogenous s_ubst~ces
such as neurotransmitters (dopamine, GABA, epinephrine, norepmephrine, ,
Inactive drugs (prodrugs) also depend upon biotransfonnation for acti-
etc.), sterojds (testosterone, progesterone, cortisol, etc.) and' insulin which
vation, the process being called as pharmacologic activation; e.g. conversior
' 0f enalapril to enalaprilat. A change in pha.nnacologic activity of the drug are also used as therapeutic agents, are inactivated rapidly because of the
on metabolism has also been observed (see Table 5 .1 .). body's well developed system for metabolizing such agents. These' ~ub- . .
stances are therefore called as soft drugs. Such soft drugs do not precipita e

TABLE 5.1 Metabolites and.Relative Activity of Drugs · levels. /


Drugs Metabolites
Drug Metabolizing Organs
Pharmacologic Inactivation
Active Liver is the priniary site for metabolism of almost a!l drugs (and o~er
Inactive
Amphetamine xenobiotics) because of its relative richne~s in possesstng a large v~ety
Phenylacetone
Phenobarbital of enzymes in large amounts. Meta~o/ism ?Y o~gans other .~han· lwer :
Hydroxyphenobarbital
Phenytoin (called as extrahepatic metabolism) ts of minor I~portance. smce l~wer
p-Hydroxy phenytoin
Salicylic acid Salicyluric acid level . of drug metabolizing enzymes are present In such tissues. The
decreasing order of drug metabolizing ability of various ~rgans i~: liver >
No Change in Pharmacologic Activity
lungs > kidneys > intestine > placenta > adrenals > skzn. Brain, t~stes,
Active Active
muscles, spleen, etc. also metabolize drugs to a small extent. _
Amitriptyline Nortriptyline
Imipramine Desipramine
Drug Metabolizing .Enzymes
Codeine Morphine
Phenylbutazone Oxyphenbutazone The enzymes that biotransfonn xenobiotics differ ~om _those that. ~e­
Diazepam Temazepam tabolize food materials. They are versatil~ and nons~ctfic m metabohzmg
Digitoxin Digoxin a large number of drugs..t ~ ,enzymes: . ar~:.~ro~Jy. , divid~t-'~to ~
Toxicologic Activation categories:. microsomal. ~d· ·~on~micr~so~~~:~ ·-.~~.. mtc~~s~~~~.·.;~~YI~~s
Active catalyze a majority of ~~·· btotr~~fottnatto~, ~f~~_tt~n~. . _·~~ -~~~~~~~~s
Reactive Intermediates
Isoniazid are basically artifacts which resulted when attemp~:·· ~ere , .f~t__ . ~a~e , to
Tissue acylating intermediate
Paracetamol isOlate endoplas~ic reticulum of the liver homogen~te, Jbe~e ,ve~tcular
Imidoquinone of N-hydroxylated
metabolite fragments or microsomes are deriyed from ~~gh endoplasmiC ,retiCUlum
.. .continued

."'-..·
114 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS t 115

(rough due to the presence of RNA rich ribosomes on the membrane Phase II Reactions
surface wh~se function is protein synthesis) which shed their ribosomes to
~~ 0 '
These reactions generally involve covalent attachment of smaH pol~r _·
become smooth surfaced. The large variety of microsomal enzymes endogenous molecules such as glucuronic acid, sulfate, glycine, etc. to
catalyze a number of oxidative, reductive and hydrolytic and glucuronidation either unchanged drugs or phase I products having suitable functional
reactions. groups viz.' -OH, -COOH, -NH2 and -SH and fonn highly water-soluble.
Some important characteristics of microsomal enzyme system are: conjugates which are readily excretable by the kidneys (or bile). Thus;
these reactions are called as conjugation reactions. : Since the outcome
1. The intact nature of lipoidal membrane bound enzyme of the
of such processes are generally products with increased molecular size
microsomes is essential for its selectivity towards lipid-soluble
(and altered physicochemical properties), they are als? called as sy~the~ic
substrates.
reactions. Quite often, a phase I reaction may not yteld a metabolite that
-2. A number of lipid-soluble substrate~
(xenobiotics in general) can is sufficiently hydrophilic or phannacologically inert but conjugation reac-
interact nonspecifically with the microsomal enzymes; natural en- tions generally result in products with total loss of phattnacologic activity
dogenous substances which are generally water-soluble do ·n ot and high polarity. Hence, phase II reactions are better known as true
interact. detoxification reactions. Since these reactions generally involve transfer
3. The lipid soluble substrate is biotransfonned into a water-soluble of moieties to the substrate to be conjugated, the enzymes responsible are
metabolite by the microsomal enzymes which can be readily ex- called as transferases.
creted. The biotransfor1nation of drug metabolites, particularly the glutathione
The nonmicrosomal enzymes include those that are present in soluble conjugates which are excreted via bile in the gut, by the intestinal micro-
fonn in the cytoplasm and those attached to the mitochondria but not to flora, is considered by few researchers as phase III reactions.
--- endoplasmic reticulum. These are also nonspecific enzymes that catalyze Quite commonly, the biotransfonnation reactions proceed sequentially
. few oxidative reactions, a number of reductive and hydrolytic reactions and the combination of several phase I and phase II reactions yield a
and conjugation reactions other than glucuronidation. It is interesting to
range of metabolites (Fig. 5.1 ).
note that, in· contrast to microsomal enzymes, the nonmicrosomal en-
zymes, especially the soluble enzymes, act on relatively water-soluble
xenobiotics (as well as endogenous compounds), e.g. oxidases, peroxi-
fi:\
~ __
Phase 1/11
_____,
® 8--~
Phase II
®
. .
dases, dehydrogenases, esterases, etc.
1
Phase I
r
Phase 1111
I

CHEMICAL fATHW~YS OF DRUG BIOTRANSFORMATION


l Phase 1/11 I
I

R. T Williams divided the pathways of drug metabolism reactions into Phase II


two general categories: phase I and phase II reactions. DRUG
®
D _ _ __,.
® E--~
®
Phase I Reactions J .\
These reactions generally precede phase II reactions and include o~i­ Phase I Phase 1111
I

da~ve, reductive and hydrolytic reactions. By way of these reactions, a


polar functional group is either introduced or unmasked if already present l Phase 1/11
l Phase II ____..
H _ _
on the otherwise lipid soluble substrate, e.g. -OH, -COOH, -NH2 and -SH. @)
G--~
®
Thus, phase I reactions are also called as functionalization reactions.
,..fhese transfottnations are also called as asynthetic reactions, opposite to Fig. 5.1 Sequence of phase I and phase II reactions yielding a range of p~~ducts
the synthetic phase II reactions. The resulting product of phase I reaction The various phase I and phase II reactions are listed in Table 5.2
is susceptible to phase II reactions. ..
I .
. . .~.

I
~IOTRANSFORMATION OF DRUGS 117 -
:_· ~t16 BIOPHARMACEUTICS AND pyiARMACOKINETICS
fHASE I REACTIONS
TABLE 5.2-Chemical Pathways of Drug Biotransformat:ion--(M) and (N)
OXIDATIVE REACTIONS
Indicate Reactions Catalyzed by Microsomal and Nonmic rosomal Enzymes
Oxidative reactions are the most important and most common meta-
PHASE I REACTIONS ~o lie reactions. Almost all drugs that undergo phase I biotransfo~ Illation ·
• I

A. Oxidative Reactions ~ndergo oxidation at some stage or the other. A simple reas!ln~ for ·
1·. · Oxidation of aromatic carbon atoms (M) ~xidation being a predominant reaction is that energy in animals is prima-
2. Oxidation of olefins (C=C bonds) . (M) rilY derived by oxidative combustion of organic molecules containing
· 3. ~idat'ion ·of benzylic, allylic carbon atoms varbon and hydrogen atoms.
and carbon atoms alpha to carbonyl and imines (M)
Oxidative reactions increase hydrophilicity of xenobiotics b-y introduc-
4. Oxidation of aliphatic carbon atoms (M)
jng polar functional groups such as-OH. Such a polar metabolite can
5. Oxidation of alicyclic carbon atoms (M)
,hus rapidly undergo phase II reaction or is excretable by the kidneys.
6. Oxid~tion of carb(>n-het~roatom systems: -
a. Carbon-Nitrogen systems (aliphatic and aromatic amines) Oxidation of xenobiotics is nonspecifically catalyzed by a number of ·.

i. .N-Dealkylation _ ~ (M) ~nzymes located in the microsomes. Such enzymes require both molecu-
ii. Oxidative. deamination (M),(N) far oxygen (02) and the reducing agent NADPH to effect reaction. They
iii. N-Oxide formation I (M) ~ire therefore referred to as the mixed function oxidases. The overall
iv. N~Hydroxylation (M) ~toichiometry of this reaction involving the substrate RH which yields the.
b. Carbon~Sulfu~ systems: product ROH, is given by the following equation:
i. S-Dealkylation (M)
ii. Desulfuration (M) RH + 02 + NADPH + H+ ) ROH + H20 + NADP+
iii. S-oxidation ~ (M)
(M) where NADPH = reduced nicotinamide adenine dinucleotide phosphat~. ' .
c. Carbon-Oxygen systems (0-dealkylation)
7. Oxidation of alcohol, carbonyl and acid functions (M),(N) Since only one oxygen atom from the molecular oxygen (dioxygen or .
8. Miscellaneous oxidative reactions o 2) is incorporated in the product for tned, the mixed function oxidases
are also called as monooxygenases. Quite often, the product of such a
B. .Reductive - Reactions · ·
(M),(N) reaction contains a hydroxyl function, hence, the enzymes are sometimes
I. Reduction of carbonyl functions .(aldehydes/ketones)
(M) also called as hydroxylases.
2. Reduction of alcohols and C=C ·bonds ·
3. Reduction .o f N-compounds (nitro, azo and N-oxide) (M),(N) The multienzyme mixed function oxidase system, located in the endo-
4. Miscellane~s reductive reactions - plasmic reticultim of hepatic cells, is composed ofan electron transf{.f
· chain consisting of 3 components:
C. Hydrolytic Reactions · --
1. Hydrolysis of esters and ethers (M),(N) 1. A heme protein known asi cytochrome P-450, which is actually a
2. Hydrolysis of amides '•
(M),(N) family of enzymes. It is a tenninal oxidase and plays the impor-
3. Hydroiytic cleavage _.o f nonaromatic heterocycles (M),(N) tant role of transferring an oxygen atom to the substrate RH and
.•

4. Hydrolytic dehalogenation convert it to ROH.


5 .. Miscellaneous hydrolytic _reactions 2. A second enzyme,· the flavoprotein known as cytochrome P-450
'
reductase (or-cytochrome c reductase) which i~ NADPH dep~n­
PHASE II REACTiONS dent. It functions as an electron carrier, catalyzing the reducti~n ·
1. Conjugation with glucuronic acid . (M)
of cytochrome P-450 to the ferrous form by transferring an el~c- ·
2. Conjugation with sulfate moieties (N)

tron from NADPH. "
I • 3. C9njugation with alpha amino acids (N) -

(N) 3. A heat stable lipid component known as phosphatidylcholine_. --


·4. C,onjugation with glutathione and mercapturic acid fpnnation Its function is to facilitate electron transfer from NADPH to
5. Acetylation reactions (N)
(N) cytochrome P-450.
6. Methylation reactions
7. Miscellaneous conjugati~n reactions (N)
/.'


. - -· 118 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 119
-- .

Magnesium ions are also required for maximal activity of mixed func- Oxidation of Arom~tic Carbon Atoms (Aromatic Hydroxylation)
;>
tion oxidases. This reaction proceeds via fot n1ation of a reactive intettnediate arene .
_I
The most important component of mixed function oxidases is the oxide (epoxide) which in most cases undergoes rearrangement to . yield
cytochrome P-450 since it binds to substrate and activates oxygen. The arenols and in some cases catechols and glutathione conjugates .
.reduced forn1 of this enzyme (Fe++) binds with carbon monoxide to fonn
R
a complex that shows maximum absorption at 450 nm, hence the name.
The mechanism of cytochrome P-450 catalyzed metaboJism of xenobiotics

is depicted in the redox cycle in Fig. 5 .2. Arenol
(major)
(P-450) RH . OH R
Oxidized R R R
r;:F~e3l:"
.. --...............
substrate 0
epoxide hydrase OH
.. .
(P-450)ROH (P-450)RH Catechol
Arene oxide Dihydrodiol
A rene (Minor product)
Fe3 • (Highly reactive
CYTOCHROME P-450 GSH 5-epoxide transferase
electrophi!,e)
OXIDATION-REDUCTION e-(NADPH)
CYCLE Cytochrome P-450 R OH
'• reductase
Activated ternary
Tissue toxicity in • ..·SG
complex
hv (P-450)(CO)RH instances when
(P-450)(0l-)RH (P-450){02-)RH (P-450)RH Glutathione conjugate
glutathione is
~ Fe2• Fe2•
CO Fe2•
depleted
(minor product)
Fe3• chromophore,
. absorbs at The arene oxide intennediate is highly reactive and known to be
Cytochrome b5
450 nm
reductase ... (P-450)(0 2)RH carcinogenic or cytotoxic in some instances, e.g. epoxides of bromobenzene
(NADH)e- Fe2 • and benzo(a)pyrene .
. Ternary complex
Monosubstituted benzene derivatives can be hydroxylated at ortho-,
Fig. 5.2 Cytochrome P-450 oxidation-reduction cycle meta- or para-positions but para-hydroxylated product is most common,
..

The various steps in the oxidation of xenobiotics are: e.g. conversion of acetanilide to paracetamol, and phenylbutazone to
I. Binding of the substrate (RH) to the oxidized fonn of the cyto- oxyphenbutazone. '

chrome P-450 (Fe+++) to forn1 a complex.


2. A one electron transfer from NADPH to the complex by cyto-
chrome P-450 reductase to fonn reduced (Fe++) P-450 substrate
Acetanilide Paracetamol
complex; this step is considered as the rate-limiting step in the
overall oxidation of xenobiotics.
HO--<
3. The reduced enzyme-substrate complex combines with a molecule
0 0
of oxygen to fonn a ternary ~omplex.
• H
4. The ternary compl~x combines with a second electron supplied
by NADH in presence of enzyme cytochrome b5 reductase to Phenylbutazone Oxyphenbutazone

fonn a ternary activated oxygen P-450 substrate complex.


I
Such a reaction is favored if the substituent is an activating group
5. One atom of oxygem from .the activated oxygen complex is trans- (electron rich) like the amino group. Deactivating or electron withdr~w­
ferred to the substrate to yield the oxidized_product and the other ing groups such as carboxyl and sulfonamide retard or prevent aromatic
atom fonns water. The free oxidized fotnl of cytochrome P-450 hydroxylation, e.g. probenicid.
is now ready to attach to yet another molecule of substrate.

I
/,.
120 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 121

Oxidation of Olefins Carbon atoms adjacent to olefmic double bonds (are allylic carbon
Oxidation of nonaromatic carbon-carbon double bonds is analogous to atoms) also undergo hydroxylation in a_manner similar to benzylic car-
aromatic hydroxylation i.e. it proceeds via fortnation of epoxides to yield bons, e.g. hydroxylation of hexobarbital to 3'-hydroxy hexobarbital.
I ,2-dihydrodiols. A better known example of olefmic oxidation is con-
Oxidation of Carbon Atoms Alpha to Carbonyls and Imines
version of carbamazepine to carbamazepine-1 0, 11-epoxide; the latter is
Several benzodiazepines contain a carbon atom (C-3) alpha to both
converted to corresponding trans-1 0, 11-dihydrodiol.
CH, CH3
10 11 0 OH I ~ 0 I
_,
N N
H20
3 OH
epoxide hydrase
Cl N Cl -N

CONH 2

Carbamazepine Carbamazepine-1 0, Trans-1 0, 11-dihydroxy


11-epoxide carbamazepine
Diazepam 3-Hydroxy diazepam
(N-methyl oxazepam)
Olefmic hydroxylation differs from aromatic hydroxylation in that
their epoxides are stable and detectable which also _indicate that they are carbonyl (C=O) and imino (C=N) functions which readily undergoes hy-
not as reactive as aromatic epoxides. However, : an important example
droxylation, e.g. diazepam.
where the olefm epoxide is highly reactive is that of aflatoxin B 1. It _is
known as ~e most potent carcinogen (causes hepatic cancer). Oxidation of Aliphatic Carbon Atoms (Aliphatic Hydroxylation)
Alkyl or aliphatic carbon atoms can be hydroxylated at two positions -
Oxidation of' Benzylic Carbon Atoms at the terminal methyl group (called as co-oxidation) and the penultimate
Carbon atoms attached directly to the aromatic rings (benzylic carbon carbon atom (called as ro-1 oxidation) of which the latter accounts for the
atoms) are hydroxylated to corresponding carbinols. If the product is a major product, e.g valproic acid. Hydroxylation at other carbon atoms in
Cft 2OH CHO coo~ long chain compounds is less common.
Alcohol
d.tlydrogtnast
CH3-CH2-CH2, 5-Hydroxy
CH-COOH I . .d
HOCH -c.~-CHt'....-
• 2 va prole aCI
(minor product)
SO 2 NHC<?NHC4 Hg S0 2 NHC.ONHC 4 Hg

. Tolbutamide Primary Carbinol Corresponding Corresponding


aldehyde carboxylic acid OiJ- C.H2-C.H2, 4-Hydroxy
CH-<OOH
CH.J-CH _ CH
2/ valproic acid
primary carbinol, it is further oxidized to aldehydes and then to carboxylic 1 (major product)
Valproic acid
·acids, e.g. tolbutamide. A secondary carbinol is converted to ketone. OH

Oxidation of Allylic Carbon Atoms Terrninal hydroxylation of methyl group yields primary alcohol which
undergoes further oxidation to aldehyde and then to carboxylic acid quite
OH
j ~AIIylic
H CH3 OH
i carbon atom
I I I
CHJ-C CH2 ----4: CH-COOH _..,. CH3-C- CHz~

0 I I
CH 3 CH3
H~ _,N
Ibuprofen Tertiary alcohol metabolite
1f
0
'cH3 · '

rapidly. Penultimate carbon at'om can be secondary or tertiary of which


Hexobarbital 3'-Hydroxy hexobarbital the latter type is also equally reactive, e.g. ibuprofen.
122 BIOPHARMACEUTICS AND PHARMACOKf ~TICS BIOTRANSFORMATION OF DRUGS 123

The ro- ~ oxidation of secondary and tertiary penultimate carbons yield ·.t is however the removed alkyl group that is oxidized. Mechanism of N-
-- -- .._
correspon~mg. alcohols. The secondary alcohol products seldom undergo dealkylation involve oxidation of a-carbon to generate an intennediate
fu~e~ oxtdatio~ to ketones as the latter are less hydrophilic. Further carbinolamine which rearranges by cleavage of C-N bond to yield the N·
oxidation of tertiary alcohol products is improbable. dealkylated product and the corresponding carbonyl of the alkyl group (a
Hydroxylation of aliphatic side chains attached to an aromatic ring primary alkyl is transfonned to aldehyde and a secondary alkyl to ketone).
H OH
\ lex:: \ I \ /
N CH2- CH2- CH2- CH3 • N-C- N-C-
I
N-H + o=c . . . . ._
R~N f 2' )' t.' - - - - . R
N
1 I
H
I I
H H
Parbendazole Carbinolamine N-Dealkylated Carbonyl
1'-Hydroxy parbendazole intermediate metabolite
generally occurs at benzylic methylene groups (1') which can be consid-
ered as ro-1 oxidation, e.g parbendazole. A tertiary nitrogen is more rapidly dealky lated in comparison to sec-
ondary nitrogen because of its higher lipid solubility. Thus, one alkyl
Oxidation of Alicyclic Carbon Atoms (Alicyclic Hydroxylation) from a tertiary nitrogen compound is removed rapidly and the second one
Cyclohexane (alicy~lic) and piperidine (nonaromatic heterocycle) rings slowly. Small alkyl groups like methyl, ethyl, n-propyl, isopropyl, etc.
are commonly found In a number of molecules, e.g. acetohexamide and are removed rapidly. N-dealkylation of t-butyl group is. not possible
because the a-carbon cannot be hydroxylated. Such groups are, however,
removed via initial hydroxylation of one of the methyl groups to alcohol.
OH CH3 CHJ
~HJ
\ I (OJ
... \ . [OJ
... ' N-C-COOH
I -COz ..
NH2 N-C- CH3 N-C-C~OH
I I . I I I
Minoxidil 4'-Hydroxy minoxidil
(HJ !HJ CH3

N-t-Butyl Alcohol Acid


minoxidil respectively. Such rings are generally hydroxylated at C-3 or
C-4 positions.
THJ
[OJ /CHJ
Oxidation of Carbon-Heteroatom Systems 'N-C-H • + o=c
• I I 'cHJ
Biotransfottnation of C-N, C-0 and C-S systems proceed in one of the CHJ
two ways: _

N-isopropyl N-dealkylated Acetone
I. Hydroxylation of carbon atom attached to the heteroatom and product
subsequent cleavage at carbon-heteroatom bond, e.g. N-, 0- and A tertiary nitrogen attached to different alkyl groups undergoes dealkylation
S- dealkylation, oxidative deamination and desulfuration. by removal of smaller alkyl group first. A representative example of each
2. Oxidation of the heteroatom itself, e.g. N- and S-oxidation. of the chemical classes of compounds capable of undergoing N-dealkylation
is given below.
Oxidation of Carbon-Nitrogen Systems
Secondary aliphatic amine e.g. methamphetamine
. .1. N-Deal~ylation : Alkyl groups attached directly to nitrogen atom
tn nt~rogen beanng compounds are capable of undergoing N-dealkylation fHJ.
reactions, e.g. secondary and tertiary aliphatic and aromatic amines ter- CH2-CH-NH2 + HCHO
tiary alicyclic amines and N-substituted amides and hydrazines. Since
N-de~lky.latio~ of amines yield amines and amides yield amides, the Methamphetamine Amphetamine Formaldehyde
reaction ts satd to undergo without any change in the state of oxidation.
I
124 BIOPHARMACEUTICS AND PHARMACOKlNETICS
BIOTRANSFORMATION OF DRUGS 125
Tertiary aliphatic amine e.g. imipramine

(o]
+ HCHO

Carbinolamine . where R=H or alkyl


Imipramine Desipramine Formaldehyde Thus, oxidative deamination is converse of N-dealkylation in tenns of
product fonned the carbonyl product retains a large portion of the parent
Secondary and tertiary aromatic amines are rare among therapeutic
agents. structure and the amines fonned are simple, e.g. NH3. Actually speaking,
both the reactions are same, the criterion for differentiation being the
Tertiary alicyclic amine (nonaromatic heterocycle) e.g. hexobarbital relative size of the products.
Primary aliphatic amines readily undergo deamination, e.g. amphet-
amine, while secondary and tertiary amines are deaminated only when
bulky groups are attached to nitrogen, e.g. propranolol.
0 0 + HCHO
fHJ
HN'-,./N
II 'cHJ HNYH )-- CH2- (::. 0 + NH3
0 0
Hexobarbital Amphetamine Phenylacetone Ammonia
N-demethylated Formaldehyde
metabolite
Primary aniin~ metabolites fonned by N-dealkylation or decarboxyla-
Amides e.g. diazepam tion also undergo deamination. Deamination is inhibited by absence of
CH3 a-hydrogen, e.g. phentettnine. Aromatic and alicyclic amines are resistant
I o 0 to deamination.

+ HCHO 3. N-Oxide Formation : N-oxides are fottned only by nitrogen


Cl atoms h~ving basic properties. Thus,. amines can fortn N-oxides but
amides cannot. Generally, the tertiary amines yield N-oxides. Four
categories of tertiary amines that fottn N -oxides are aliphatic amines
(e.g. imipramine), alicyclic amines (e.g. nicotine), nitrogen atoms of aro-
Diazepam Desmethyldiazepam Formaldehyde matic heterocycles (e.g. trimethoprim) and amines attached to aromatic
rings (e.g. N,N-dimethyl aniline). .
Hydrazines e.g. iproniazid
0 CH)
0
II I II
.,__(- NHNH-C-H
C-NHNH2 +
I
CH3

. Iproniazid N-Dealkylated Acetone Imipramine Imipramine N-oxide


product

2. Oxidative Deamination : Like N~dealkylation -this reaction also


~1 N
proceeds via the carbinolamine pathway ·but here ,tfie C:.N bond cleavage CHJ 0; ' CHJ
. occurs at the bond that links amino group to the larger portion of the drug
molecule. Nicotine Nicotine-1 '-N-oxide
'
/
BIOTRANSFORMATION OF DRUGS 127
126 BIOPHARMACEUTICS AND PHARMACOKINETICS

(~0 CH]O
NH2 so2 NHOH
.
NH2 so 2 NH2
CH30 CH2 CH]O CH2 N-Hydroxy dapsone
Dapsone
CHJO NH2 c~o NH2
NH2 5~ N=O
Trimethoprim Trimethoprim-1-N-oxide
N itroso product
-- ' -- • • , __
-
.. . - _ .. I .. - •:......,.. -1 - ~

CH3
/CH3 l The N-hydroxy dapsone can oxidize ferrous fonn of hemoglobin to ·
N N .. o ferric form and cause methemoglobinemia. A secondary aromatic amine
'CHJ l ;ields a nitrone subsequent to fonnation of secondary hydroxylamine
CH3

, which is further hydrated to primary hy~roxylamine. ~
N, N-Dimethyl aniline N-Oxide metabolite -- .

The N-oxide products are highly water-soluble . and excreted in urine. NHOH
They are however susceptible to reduction to the corresponding amine.
Seco'ndary Secondary Nitrone Primary
4. N-Hydroxylation : Converse to basic compounds that form N- hydroxyl-
aromatic hydroxyl- .
oxides, N-hydroxy fonnation is usually displayed by nonbasic nitrogen am me am me am me
atoms such as amide nitrogen, e.g lidocaine.
N-hydroxylation is also possible at basic nitrogen, e.g. primary and
secondary amines. Phentermine, a primary aliphatic amine, cannot un-
dergo deamination as the a-carbon does not contain hydrogen and the
drug is therefore N-hydroxylated.
CHJ CH3 CH3
lex: I
Lidocaine N-Hydroxy lidocaine CH 2-C- NH 2 CHzC-NHOH ...
I
N-Hydroxylation of amides often leads to generation of chemically
reactive intennediates capable of binding covalently with macromolecules,
CH3 ' CH3

, Phentermine N-Hydroxy phentermine


e.g paracetamol. Paracetamol is safe in therapeutic doses since its reactive
metabolite imidoquinone is neutralized by glutathione; however, in high
doses, the glutatpione level becornes insufficient and significant covalent
tissue binding thus occurs resulting in hepatotoxicity. Phenacetin is also
toxic for the same reason.
· - Jt - ..
-- - ___ ,_ -. - ----~ '
Nitroso metabolite Nitro metabolite
OH 0 0
I II -~0 II '
HO ----HO N- C-CHJ ~... 0 N-C-CHJ Oxidation of Carbon-Sulfur Systems
1. S-Dealkylation : The mechanism of S-dealkylation of thioethers
Paracetamol N-Hydroxy metabolite Reactive electrophilic
SH
imidoquinone

Hepatic necrosis ~<-


l
Covaient binding to liver
N)
N
+ HCHO
H
~onbasicaromatic- amines 'also undergo hydroxyla~ion. A primary
6-Methyl Hydroxylated 6-Mercaptopurine
aromatic amine is further converted to nitroso derivative, e.g. dapsone. mercaptopurine intermediate
128 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 129
(RSR') .is analogous to N-dealkylation i.e. it proceeds via a-carbon hy- hemiketal intennediate which spontaneously undergoes C-0 bond cleav-
droxylation. The C-S bond cleavage results in fonnation of a thiol (RSH"' age to fonn alcohol (arenol or alkanol) and a carbonyl moiety.
and a carbonyl product, e.g. 6-methyl mercaptopurine.
I The 0-containing functional groups (analogous to amines and amides,
2. Desulfuration : This reaction also involves cleavage of carbon- the substrates which undergo N-dealkylation) are ethers and esters. How-
sulfur bond {C=S or thiono). The product is the one with C=O bond. ever, only the ethers undergo 0-dealkylation reaction. Aliphatic ether
Such a desulfuration reaction is colnmonly observed in thioamides (RCSNHR') drugs are rare and aromatic ethers (phenolic) are common. Methyl ethers
such as thiopental. are rapidly dealky lated in comparison to longer chain ethers such as the
one containing n-butyl group. The reaction generally leads to fortnation _
CHJ CH3
I of active ·metabolites, e.g. phenacetin to paracetamol, and codeine to
CHCH2CH2CHJ
0
lH-C~~H2CH3 morphine.
-~~0 ..

s
Phenacetin Paracetamol
Thiopental Pentobarbital
CH3-N CHJ-N
Desulfuration also occurs witlt compounds containing P=S bonds such
as the organophosphate pesticides, e.~ parathion .

0
IJ
C2Hs-,-O---< ___, )--N02 H
CH]O OH
C2Hs
Codeine Morphine
Parathion Paraoxon
Oxidation of Alcohol, Carbonyl and Carboxylic Acid
3. S-Oxidation : Apart from S-dealkylation, thioethers can also un...
dergo .S-oxidation reactions to yield sulfox~des which may be further These reactions are mainly catalyzed by nonmicrosomal enzymes, de-
hydrogenases. Primary and secondary alcohols and aldehydes undergo
·O 0 0
t oxidation relatively easily but tertiary alcohols, ketones and carboxylic
s s \ I
s acids are resistant as such a reaction involves cleavage of C-C bonds.
Primary alcohols are rapidly metabolized to aldehydes (and further to
N
I carboxylic acids) but oxidation of secondary alcohols to ketones proceeds
R slowly. This is because primary alcohols are better substrates for dehy-
Phenothiazine Phenothiazine sulfoxide Phenothiazine sulfone drogenases due to their acidic nature, and secondly, their oxidation
products, carboxylic acids, are more water-soluble than ketones. Since
oxidized to sulfones (RS02R). Several phenothiazines, e.g. chlorpromazine,
undergo S-oxidation. primary alcohols are inactivated rapidly, drugs bearing such groups are
,

Oxidation of Carbon-Oxygen Systems alcohol aldehyde

0-Dealkylation : This reaction is also ~irnilar to N-dealkylation and


;;=:=========-
dehydrogenase
CHJ Ct-K> - - - - - CH3COOH (enters Kreb'scycle)
dehydrogenase
proceeds by a-carbon hydroxylation to fonn an unstable hemiacetal or
Ethanol Acetaldehyde Acetic acid (enters Kreb's cycle)
OH H
~ • I , I
R-0-CH2R R-0-CH-R - - - ·.. R-OH + Q:.C-R
I
rare. In case of ethanol, oxidation to acetaldehyde is reversible and
.
. . further oxidation of the latter to acetic acid is very rapid since acetalde~
Ether · Hemiacetal/hemiketal Alcohol Aldehyde/ketone hyde is highly toxic and should not accumulate in the body. Co~pounds
\ .
,

130 BIOPHARMACE.UTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 131


-- --
with two primary alcohol functions are oxidized stepwise and not simulta- Reduction of Carbonyls (Aldehydes and Ketones) ..
neously. With ·se~ondary alcohols, the rate of oxidation increases with an . Depending on, their reactivity towards bioreduction, carbonyls can be
. - -··
increase in alkyl chain length. Compounds with both primary and second- divided into 3 categories:
ary alcohol groups are oxidized preferentially at the primary group. 1. The aliphatic aldehydes and ketones. (

Miscellaneous Oxidative Reactions 2. The aromatic aldehydes and ketones~ - · · I • :


· a.. Oxidative Aromatization/Dehydrogenation : An example of meta- 3. The esters, acids and am ides.
. '

· bolic aromatization of drugs is nifedipine. The order of reactivity of these categories of drugs in undergoing
reduction is. 1 >. 2 ~ 3 i.e. aliphatic aldehydes and ketones unde_!S9-
extensive bioreduction whereas esters, acids ana amides are least reactive.
Few aldehydes undergo reduction because such a reaction is usually
reversible, and secondly, they are susceptible towards oxidation which 1

COOCH3 CHJ
yields more polar products. Several ketones undergo reduction as it
Nifedipine Pyridine metabolite results in more polar metabolites. Reduction of aldehydes and ketones
yields primary and secondary alcohols respectively. The reaction is cata- ..
b. . Oxidative Dehalogenation : This reaction is common with halo-
lyzed by nonmicrosomal enzymes called as aldo-keto-reductases.
gen containing drugs such as chlorofonn. Dehalogenation of this drug
. yields phosgene w~ich may result in .electrophiles capable of covalent A representative example of compounds undergoing reductive reac-
binding to tissues. · tions is given below.
Cl 0 Aliphatic aldehydes e.g. chloral hydrate
I [o] II covalent binding
Cl- C-Cl Ct- C- Cl Cl 3 C.- CHO · H20 CI3C- C~OH
I - HCl to tissues
H ChloraJ hydrate Trichloroethanol
Chloroform Phosgene
Aliphatic ketones e.g. methadone
'
Oxidative ring cleavage, oxidation .of arenols to quinones, etc. are
other oxidative reactions.

0 OH
I
• >-- C- CH-C2H5
REDUCTIVE REACTIONS I -
/CH3
CH2-CH-N
Bioreductions are also capable of generating polar functional groups I '-...(HJ
.· such as hydroxy and amino which can undergo further biotransfonnation CH3
or conjugation. A number of reductive reactions are exact opposite of
Methadone Methadol
oxidation .. For example:
Alcohol dehydrogenation --~)
4-< Carbonyl reduction Alicyclic ketone e.g. naltrexone

N-Oxidation 4-(--~) Amine oxide reduction [>--c~-N

Thus, in this sen~~¢/


,
bioreduction comprises one-half of reversible reac-
lions. Such . reactions may be catalyzed by the same enzyme (true
reversible reaction) or by different enzymes (apparent reversible reac-
-- .. _ tion). Since reversible reactions usually lead to conversion of inactive · OH OH OH

1
metabolites into active drug, they may r~sult in delay of drug removal lsomorphine derivative .
Naltrexone
fr~m the body and hence prolon&~tion of action. ·.

--
- 133
- - 132 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS

Aromatic ketone e.g. acetophenone For example, reduction of nitrazepam.

0 OH 0
II I
>--C-CHJ l--CH-CHJ
N~
Acetophenone Methyl phenyl carbinol

.J .. , ••

Reduction of Alcohols and Carbon-Carbon Double Bonds Nitrazepam 7-Aminometabolite


These two · reductions are considered together because the groups are
Reduction of azo compounds yield pr.imary amines via fonnation of
interconvertible by simple addition or loss of a water molecule. Before an
hydrazo intennediate (-NH-NH-) which undergoes cleav~ge at N-N bond.
alcohol is reduced, it is · dehydrated to C=C bond, e.g. bencyclane
(antispasmodic). R-N=N-R - - - R-NH- NH-R - - - RNH2
I
+ NHz -R
I

Azo Hydrazo Amines


-H2<> An important example of azo reduction is prontosil. It .is reduced to
active sulfanilamide.

Bencyclane Bencyclanol Benzylidine cycloheptane


\--N~ + N~

Reduction of norethindrone, an a,p-unsaturated carbonyl compound,


results in both reduction of C=C double bond and fonnation of alcohol. Prontosil 1,2,4-Triaminobenzene Sulfanilamide

OH Aliphatic and aromatic tertiary amine N-oxides are reduced to the


. . . c: CH
corresponding amines, e.g. imipramine N-oxide to imipramine.

Misce~laneous Reductive Reactions


t. Reductive Dehalogenation : This- reaction involves replacement
H of halogen attached to the carbqn with the H-atom, e.g. halothane.
'
Norethindrone 3p,5p-Tetrahydro norethindrone
Br
I
I
CF3-C-H
Reduction of N-compounds (Nitro, Azo and N-Oxide) I
Ci
The N-containing functional groups that commonly undergo bioreduction
Halothane 1,1, 1-Trifluoroethane Trifluoroacetic acid
are nitro, .a~o and N-oxide. It is important to note that such a reaction is
reverse of oxidation. The C-F bond is resistant to reduction.
Reduction .of nitro group proceeds via fonnation of nitro so and hy- 2. Reduction of Sulfur Containing Functional Groups : An ex-
droxylamine intennediates to yield amines. an1ple of S-S reductive cleavage is disulfiram.
s 5 .
RN02 - - - R-N=O --r R-NHOH ------ RNH2 C2Hs '-.... ·II II /C2H5
·-.. - "
N- c- s-s-c-N,
C2H5 / C2H5
Nitro Nitroso Hydroxylamine Amine .
Disulfiram Diethyldithiocarbamic acid
---
Sulfoxides are sometimes reduced to sulfides. e.g. sulindac.
134 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 135

HYDROLYTIC REACTIONS Aromatic esters are hydrolyzed by arylesterases and aliphatic esters by
carboxy,lesterases. Examples of various classes of esters undergoing hy-
These reactions differ from oxidative and reductive reactions in 3
drolysis are given below.
respects: - . .

1. The reaction does rrot involve change fu- the state of oxidation of Organic acid (carboxylic acid) esters
the substrate. Ester with a large acidic (and small alcohol) group e.g. clofibrate
2. The reaction results in a large chemical change in the substrate ,
COOC2H5 COOH
brought about by loss of relatively large fr~gments of the mole- •
I I
cule. Ct 0-C-CHJ 0-c-CHJ + C2H50H
I I
CHJ
3. The hydrolytic enzymes that metabolize xenobiotics are the ones CH3
that also··act on endogenous substrates; moreover, their activity is Clofibrate Free acid m'etabolite
- not confmed to liver as they are found in many other organs like (active)
kidney, intestine, etc.
Ester with large alcoholic (and small acidic) group e.g. aspirin. -
-. A number of _functional groups are hydrolyzed viz. esters, ethers,
amides, hydrazides, etc. COOH
I
OCOCH3 >-- OH + CH3 COOH
· Hydrolysis of Esters and Ethers
Esters on hydrolysis yield alcohol and carboxylic acid. The reaction is Aspirin Salicylic acid (active)
catalyzed by esterases.
Ester with large acidic and alcoholic groups (generally amine alcohols)
0 0
II II e.g. succiny.lcholine.
I

R-C-OR - - - - R-C-OH-· + ROH


+
CH2- COOCH2CHz-N (CHJ) 2 pseudo CH2COOH
The ester substrates undergoing hydrolysis can be classified as under: I • - ---------------- I + 2HOCH2CH2N+(CH3)2
cH2-C.OOC.H2 C~-N ( CH3) 2 cholinesterase CH2COOH

ESTERS Succinylcholine Succinic acid Choline

Inorganic Acid Esters


-. • Organic Acid Esters Inorganic Acid Esters •

I
Phosphates e.g. stilbestrol diphosphate
I . \I OH OH C2H5
Esters with a Esters with both Phosphates Sulfates Nitrates I I
I ,__ ( =- c ---(
large and a large acidic and O=P-0 0-P=O - - H O +
I
small moiety alcoholic groups
/ bH OH t2H5
I.-- ;.
Stilbestrol diphosphate Stilbestrol
Esters with a large Esters with a large
acidic group alcoholic group Sulfates e.g. i.sopropyl methanesulfonate

Organic esters with both large acidic and alcoholic groups on hydroly- H 0 0
I II II
sis results in metabolites with complete loss of activity. Esters where one CHrC-0-S-(HJ - - - - + HD-5-CHJ
of the groups is relatively large, retain much of their activity when 1 II · II
CH3 0 0
hydrolyzed since such a group is generally a pharmacophore (having
pharmacologic activity). In many cases, such esters\ are prodrugs which Isopropyl methanesulfonate Isopropanol Methanesulfonic
rely on hydrolysis for their transfortnation into active fortn, e.g. · acid
chloramphenicol palmitate.
136 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS .,,t
i 137
I

Nitrates e.g. pentaerythritol tetranitrate Hydrazides are also a class of amides e.g. isocarboxazide

CH2-0N02 Pentaerythritol + 4HN03 0 . I

II I

'
o 2 NO-CH 2 -~-CH2 -0N0z ~ Pentaerythritol + 3HN0
3
CH2NH-NH-.C--r.=-N --~
d
C~NHNH 2 + HOOC

CH2-0N02 mono nitrate


CH 3
Pentaerythritol tetranitrate
lsoGarboxazide Benzyl hydrazine Acid
Ethers undergoing hydrolysis are glycosides such as digoxin and digi-
toxin and 0-glucuronides. Hydrolytic Cleavage of Nonaromatic Heterocycles
Nonaromatic heterocycles also contain amide functions, e.g. lactams
Hydrolysis of Amides (C-N bond cleavage) .
(cyclic amides). Several lactams that undergo hydrolysis are:
Amides are hydrolyzed slowly in comparison to esters. The reaction,
catalyzed by amidases, involves C-N cleavage to yield carboxylic acid 1. Four-membered lac tams (/3-lactam) e.g. penicillins
and amine. 0 0
II II
0
II I
0
II , CH2·-C-NH -----r
CHJ _ _
CH3
CH2-C-NH II 5 /CH3
CH3
R-C- NHR - - - - R-C-OH + RNH2 COOH N-...... COOH
~-N-....._ COOH
0 H
Primary amides are rare. Secondary amides fotrn the la:rg~st group of Penicilloic acid metabolite
Penicillin G
amide drugs. Examples of amide hydrolysis are given below.
Secondary amide with aliphatic substituent on N-atom e.g. procainamide 2. Five-membered lactams e.g. succinimides
(hydrolyzed slowly in comparison to procaine)

0
~
CH3

Phensuximide Phenyl succinamic acid


Procainamide PABA
.
Secondary amide with aromatic substituent on N-atom (ani/ides) e.g. 3. Six-membered lactams e.g. thalidomide
lidocaine '
0
Phthalimide
derivative
(known to be
0 COOH teratogenic)
0
NH2

0
Lidocaine 2,6-Xylidine N, N-Diethylg lycine 0 0
H Glutarimide
Tertiary amide (N-atom contained in a ring) e.g. carbamazepine Thalidomide N derivative
0
0


Carbamazepine lminostilbene
138 BIOPHARMACEUTICS AND PHARMACOKINET'~S BIOTRANSFORMATION OF DRUGS l.39
4. Seven-membered lactams e.g. chlordiazepoxide when doses of drugs are highe~ than nonnal levels of conjugating mol-
NHCH3 NH2 COOH
ecules, saturation of metabolism occurs and the unconjugated drug/metabolite
precipitates toxicity. The order of capacities of important conjugation
-N
) reactions is: Glucuronidation > Amino acid conjugation > Sulfaiion and
Cl Cl
Cl Glutathione conjugation. The increase in the molecular weight of the
\ drug following conjugation with glucuronic acid, sulfate and glutathione is
176, 80 and 300 daltons respectively. The molecular .weight of the
conjugate is important in dictating its route of excretion. High molecular
Chlordiazepoxide Lactam metabolite Open-lactam metabolite weight conjugates are excreted predominantly in bile while the low
molecular weight ones are excreted in urine.
Hydrolytic Dehalogenation
Chlorine atoms attached to aliphatic carbons are dehalogenated easily,
e.g. DDT. CONJUGATION WITH GLUCURONIC ACID
'
Also called as glucuronidation, it is the most common and most
>--Cl
fT important phase II reaction for several reasons:
CCl2
·1. Readily available source of conjugating moiety, b-glucuronic acid
DDT DOE
(Dichloro diphenyl dichloro ethylene)
which is derived from D-glucose.
(Dichloro diphenyl trichloroethane)
2. Several functional groups viz. alcohols, acids, amines, etc. can.
Miscellaneous Hydrolytic Reactions combine easily with D-glucuronic acid.
Include hydration of epoxides and arene oxides, hydrolysis of sulfonyl 3. Quantitatively, conjugation with D-glucuronic acid occurs to a
ureas, carbamates, hydroxamates and of glucuronide and sulfate conjugates. high degree.
.'
4. All mammals have the common ability to produce glucuronides,
PHASE II REACTIONS 5. The free carboxy I function of glucuronic acid has a pK 3 in the
Phase II reactions involve transfer of a suitable moiety such as glucu- range 3.5 to 4.0 and hence ionisable at both plasma and urine pH
ronic acid, sulfate, glycine, etc. in presence of enzyme transferase to drugs thereby greatly increasing· the water solubility of ·the conjugated
or metabolites of phase I reactions having suitable functional groups to substrate.
form highly polar, readily excretable and pharmacologically inert conju- 6. The glucuronidation enzymes are in close association with the
gates. Tissue reactive and .. carcinogenic metabolites are rendered harmless microsomal mixed function oxidases, the major phase I drug me-
by conjugation with glutathione. The two phase II reactions viz. acetyla- tabolizing enzyme system; thus, a rapid conjugation of phase I
tion and methylation that do not generate polar products, also terminate metabolites is possible.
the pharmacologic activity of xenobiotics. Thus, phase II reactions are
7. Lastly, glucuronidation can take place in most body tissues since
the real drug detoxication pathways.
' the glucuronic acid donor, UDPGA· is produced in pro~esses re-
The moieties transferred to the substrates in a phase II reaction possess lated to glycogen synthesis and thus, will never be deficient
3 characteristics: unlike those involved in other phase II reactions.
I. They are simple endogenous molecules.
Glucuronide fonnation occurs in 2 steps:
2. They are of large molecular size.
1. Synthesis of an activated coenzyme uridine-5'-diphospho-a-D-glu-
3. They are strongly polar or ionic in nature in order to render the curonic acid (UDPGA) from UDP-glucose (UDPG). The coenzyme
substrate water-soluble. UDPGA acts as the donor of glucuronic acid. UDPG is synthe-
An important aspect of conjugation reaction is that they are capacity- sized by interaction of a-D-glucose-1-phosphate with uridine
limited due to the availability of endogenous conjugating molecules. Thus, triphosphate (UTP).
142 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 143
The steps are summarized in the equations below: Conjugation occurs commonly· with glycine. Glutamine conjugation
2- ATP-sulfurylase/Mg++ · occurs to a lesser extent. Conjugation with other amino acids like aspartic
ATP + So4 -------~ APS + PPi
acid, serine and taurine is still uncommon. The substrate is generally an
APS + A TP APS-phosphokinase/Mg++ acid (aromatic in particular) and the reaction ·product is an amide.
PAPS+ ADP
su lfotransferase Examples of drugs fotnting glycine or glutamine Gonjugates are:
PAPS+ RXH ------+ RX-S03 +PAP
Aliphatic acids isopropoxyacetic acid
where · X= 0, NH Alicyclic acids cholic acid
Aryl acids salicylic acid
Furrctional groups capable of forming sulfate conjugates include phenols,
Arylacetic acids phenylacetic acid
alcohols, arylamines, N-hydroxyiamines and N-hydroxyamides. The reac-
tion product is a sulfate ester, also called as ethereal sulfate. . Heterocyclic aryl acids nicotinic acid
Amino acid conjugation occurs extensively in the liver mitochondria·
Examples of compounds undergoing sulfation are:
and thus can be used to estimate hepatic function. The diagnostic marker
Phenols · paraceta:mol, salbutamol used is benzoic acid which on conjugation with glycine yields hippuri~ ·
Alcohols aliphatics C-1 to C-5 acid. Hippuric acid is rapidly excreted in urine. Thus, the rate and extent
Arylamines aniline of urinary excretion of hippuric acid following oral or i.v. administration
Sulfoconjugates ·can be tissue reactive, e.g. the 0-sulfate conjugate of of benzoic acid indicates functioning of liver. A decreased output indi-
N -hydroxy phenacetin covalently binds to hepatic and renal tissues. cates hepatic disorder.
Endogenous substances can also undergo sulfation, e.g. steroids, bio-
logic amines, etc. CONJUGATION WITH G'LUTATHIONE AND
MERCAPTURIC ACID FORMATION
CONJUGATION WITH ALPHA AMINO ACIDS Glutathione (y-glutamyl cysteinyl glycine . or GSH) is a tripeptide with
a strongly nucleophilic character due to · the presence of a -SH (thiol)
This reaction also occurs to a limited extent because of limited
group in its structure.
availability of amino acids. The reaction occurs. in two steps:
I ~HI-+ Nucleophile •
1. Activation of carboxylic acid drug substrate with ATP and coen-
zyme A (CoA) to fonn an acyl CoA intermediate. Thus, the NH2 0 ·CH2 0

I II I I II I
reaction is a contrast of glucuronidation and sulfation where the HOOC-CH-CH2-CH2-C._._I NH-CH-C I NH-CH2-COOH
I I
donor coenzyrne is activated and not the substrate. I I
.
Glutamic acid Cysteine Glycine
2. A'cylation of the a-amino acid by the acyl CoA in presence of
enzyme N-acyl transferase. Thus, it has great affinity for electrophilic ·substrates, a number of
The reaction is summarized below. which are potentially toxic compounds. It is important to note that a
highly electrophilic metabolite has a tendency to react with tissue nucleo-
ltCOOH + ATP acyl synthetase RCOAMP + H20 + PPi philic groups such as -OH, -NH2 and -SH and precipitate toxicities such
as tissue necrosis, carcinogenesis, mutagenesis, teratogenesis, etc. Conju-
RCOAMP + CoASH acyl CoA transferase RCOSCoA + AMP gation with glutathione protects the tissue from such reactive moieties and
- , RCOSCoA + H2N-R'-COOH N-acyl transferase thus the reaction is an important detoxication route.

RCONH-R'-COOH + CoASH GSH conjugation differs from ot~~r conjugation reactions in that the
process does not require initial activation of the coenzyme or the substrate
where R' = -CH2- (if glycine) or >CH-CH2-CH2-CONH2 (if glutamine) since the GSH, which is a nucleophile .itself, is highly reactiv~ towards an
electrophilic substrate. The interaction between the substrate and the GSH



• •
144 BIOPHARMACEUTICS AND PHARMACOKINETICS
• BIOTRANSFORMATION OF DRUGS 145
is catalyzed by enzyme glutathione-S-transferase to fot 111 S-substituted both reactions yield amide products. Acetylation however differs from
glutathione conjugate. This conjugate is not excreted as such in the urine a-a1n ino acid conjugation in that the substrates are exogenous amines (and_
but undergoes cleavage, first by the enzyme y-glutamyl transpeptidase to not carboxylic acids) and the acy lating agent is endogenous acetyl Co A
release the free glutamyl residue and cysteinyl glycine derivative; the (CH 3COSCoA). The general sequence of reaction is similar to that for
latter is cleaved by enzyme cysteinyl glycinase to produce free glycine a-amino acid conjugation. The enzyme invqlved is the nonmicrosomal _.
and the cysteine conjugate. Finally, N-acetylation of this conjugate by N-acetyl transferase.
the enzyme N-acetylase yields S-substituted-N-acetyl cysteine products
called as mercapturic acids. Acetylation is an important metabolic pathway for drugs containing
P\imary amino groups. Alcohols (e.g. choline) and thiols (e.g. CoASH)
CONH-CH2 -COOH Glutathione-S-transferase a1so undergo acetylation but only the endogenous ones. _
I
RX + HS-CH2-CH
I
NHCO-CH2CH2-CH-COOH
NH2
I I
R-S-CH2-CH
CONH- CH2- COOH
I
NH2
Examples of drugs undergoing acetylation are :
Primary aliphatic amines histamine, mescaline
'
- 1

Substrate GSH
I I Primary aromatic amines procainamide, PAS, PABA, ,dapsone
NHCOCH2CH2-CHCOOH
(electrophile) (nucleophile) Sulfonamides sulfanilamide, sulfapyridine
Glutathione conjugate Hydrazines/hydrazides hydralazine, isoniazid, phenelzine
COOH OH OH
y-glutamyr
I Mercapturic acid ·Glutamine - trans peptidase N-Acetyl
R-s-c~-CH derivative NH2-t- CH3COSCoA - - - - - - - NHCOCH3
I --- -- . transferase
NHCDCFfJ CO~H
CONH-c~ -COOH
Cysteinyl glycinase . I Paraaminosalicylic acid N-Acetylated PAS
N-acetylase clH
L - - - - - R-5- CH2- l • R-S-CH2-CH

NH2
7
Glycine ~H2 Acetylation may sometimes lead to toxic products, e.g. acetyl deriva-
tives of some sulfonamides (cause renal toxicity due to decreased water
Cysteinyl glycine conjugate
solubility of the metabolites formed) and reactive arylacetamides.
GSH conjugation occurs by one of the two mechanisms: One of the interesting facts about acetylation is phannacogenetic dif-
1. Nucleophilic substitution at an electron deficient carbon or het- ferences in the rate at which it proceeds in man, (called as acetylation
eroatom such as alkyl halides, alkyl nitrates (e.g. glyceryl trinitrate), polymorphism). The distribution of population in acetylating certain
sulfates, sulfonates, organophosphates, epoxides, lactones, etc. substrates is bimodal viz. slow acetylator and rapid acetylator phenotypes.
As a result, a large inter-ethnic group variations in the therapeutic and
RX + GSH - - + ) R-S-G + H+ + x- toxic levels of drugs that undergo acetylation have been observed, e.g.
'

2. Nucleophilic addition at the electron deficient double bond such isoniazid.


as the a,p-unsaturated carbonyl compounds, e.g. ethacrynic acid.
ME THY LA TION
R-CH=CH-R' + GSH ---..) RCH-S-G-CH2-R'
This reaction differs from general characteristics of phase II re~tions .
where R = electron withdrawing group. in several ways:
I
The hepatotoxicity from paracetamol .overdose ~s due to depletion of I. The metabolites formed are not polar or water-soluble.
GSH. . ,
2. The metabolites, in a number of instances, have equal or greater
phaunacologic activity than the parent drug, e.g. morphine founed
ACETYLATION
'I from nonnorphine.
This reaction is basically an· acylation reaction and thus similar to
3. The reaction is of lesser importance in metabolism of xenobiotics.
conjugation with a~amino acids. The analogy also lies i~ the fact that
It is more important in the biosynthesis (e.g. adrenaline, melato- ·
1


.146 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS -·147

nin) and inactivation of endogenous amities (e.g. noradrenaline, cyanide ion in presence of enzyme rhodanese to fonn inactive thiocya-
serotonin, histamine). nate.
Methylation can be considered as intennediate of phase I and phase II
· · reactions. It can be called as a phase I reaction · as it is reverse of
-=-dem·~thylation reaction and can be classed as a phase U reaction because Conjugation with Ribos~
of i.t-S mechanism. - .. -Endogenous purine and pyrimidine bases - conjugate with ribose to
form nucleotides.
Methylation of substrates proceeds in two steps:
1. Synthesis of an -activated coenzyme S-adenosyl ~ethionine (SAM), Conjugation with Taurine : .- __
the donor of methyl group, from L.-methionin.e an·d A TP. Taurine, a P-·amino sulfonic acid, conjugates the endogenous bile acids
2. Transfer of the methyl group from · SAM to the substrate in to produce components of l:)ile. ·
presence of nonmicrosomal enzyme methyl transferase.
FACTORS AFFECTING BIOTRANSFORMATION OF DRUGS
.
L- ~ M h' · . .
et tontne +
ATP methionine adenosyl transferase
· SAM · pp· p·
+ · 1+ 1
The therapeutic efficacy, toxicity and biological half-life of a drug
·RXH + SAM ~~thyl transferase RX-CH3 + S-adenosyl homocysteine greatly depend upon its metabolic rate. A number of factors. may influ- · '

ence the rate of drug metabolism; they are:


where X = Q, N.H, S. · /': .

1. Physicochemical properties of the drug .' .

Important methyl transferases that catalyze methylation of xenobiotics 2. Chemical factors:


are catechol-0-methyl transferase (COMT}; phenyl-0-methyl transferase
a. Induction of drug m~tabolizing enzymes
(POMT}, phenyl ethanolamine-N-methyl transferase (PNMT}, nonspecific \
. .
transferases, etc. - b. Inhibition of drug metabolizing .enzymes
.. '.

- Examples of substrates undergoing methylatiop are: c. Environmental chemicals


3. Biological factors:
0-Methylation ·
Phenols morphine a. Species differences ' .
Ca:techols .. a-methyl dopa, L-DOPA., isoprena~ine b. Strain differences
N-Methylation • c. Sex differences
Primary aliphatic ~mines norephedrine d. Age
Secondary alicycHc ainines normorphine e. Diet
Aromatic heterocycles nicotine, .histamine '

f. Altered physiologic factors:


S-Methylation 1 i. Pregnancy
Thiols propylthiouracil, 6-mercaptopurine · ii. HonnonaJ imbalance
iii. Disease states
I .
MISCELLANEOUS CONJUGATION REACTIONS g. Temporal factors:
Some of the rare conjugation ~actions are mentioned below. i. Circadian rhythm
' ii. Circannual rhythm
Conjugation of Cya":ide .
'\ ' I

" .
•· ..
·· T~e toxicity of cyanide ion is due to its ability to arrest enzym_e s · PHYSICOCHEMICAL PROPERTIES.
OF THE DRUG .
. I

involved in cellular respir.ation and convert hemoglobin to cyanomethe.=


/ '
Just as the absorption and distribution of a drug are influenced by -its .
I .

qJ.oglobiil which· lack:~· Jhe ability to transport oxygen to tissues. Corijugati~n .physicochemical properties, so is its interaction with the drug metaboliz-
/of ~yanide ion . involves transfer of sulfur atom from thiosulfate: to the ing enzymes. Mol~cular size and ·shape, pK8 , acidity/basicity, lipophilicity
, I
• ,
.. ! • ••

. / '
·/
..


I

148 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OE DRUGS .• 149


J
and steric and electronic characteristics of a drug ; influence its interaction Enzyme induction results in decreased phannacologic activity of most
with the active sites of enzymes and the biotran\sformation processes to drugs, increased activity where the metabolites 4ilre active and altered
which it is subjected. However, sush an interrelationship is not clearly physiologic status due to enhanced metabolism of endogenous compounds
understood. such as sex hottnones. Some examples of inducers and drugs affected by
them are given in Table 5.3.
CHEMICAL FACTORS
Inhibition of Drug Metabolizing Enzymes
Induction of Drug Metaboliz'i ng Enzymes
A decrease in· the drug metabolizing ability of an enzyme is called as
The phenomenon of increased drug metabolizing ability of the enzymes
enzyme inhibition. The process of inhibition may be c;iirect or indirect.
(especially of microsomal monooxygenase system) by several drugs and
chemicals is called as enzyme induction and the agents which bring 1. Direct Inhibition : may result from interaction at the enzymic
about such an effect are known as inducers. Most inducers are in general site, the net outcome being a change in enzyme activity. Direct enzyme
lipophilic compounds with long elimination half-lives. inhibition may be competitive or noncompetitive. ·
Two categories of inducers have been defmed: a. Competitive Inhibition : results when structurally similar com-
1. Phenobarbital type inducers : includes several drugs and pesti- pounds compete for the same site on an enzyme. Such an inhibition
cides which increase the rate of metabolism of a large number of due to substrate competition is reversible and can be overcome by
drugs. high concentration of one of the substrates, e.g. methacholine
inhibits metabolism of acetyl choline by competing with it for
2. Polycyclic hydrocarbon type inducers : such as 3-methyl
cholinesterase.
cholanthrene and cigarette smoke which stimulate the metabolic
rate of few drugs. b. Noncompetitive Inhibition : arises when a structurally unrelated
agent interacts with the enzyme and prevents the metabolism ?[
Sof!Ze drugs such as carbamazepine, meprobamate, cyclophosphamide, drugs. Since the interaction is not structure- specific, metals like
rifampicin, etc. stifh:ulate their own metabolism, the phenomenon being lead, mercury and arsenic and organophosphorus insectiCides in-
called as auto-induction or self-induction. hibit the enzymes noncompetitively. Isoniazid inhibits the metabolism
· Mechanisms involved _in enzyme induction may be increased enzyme of phenytoin by the same mechanism.' "
synthesis, decreased rate of enzyme aegradation; enzyme stabilization or Direct inhibition also results 'H'hen the metabolic product competes
enzyme activation. 'H'ith the substrate for the sante en::yme (called as product inhibi-

' tion). Certain specific inhibitors such as xanthine oxidase inhibitors
TABLE 5.3 (e.g. allopurinol) and MAO inhibitors (e.g. phenelzine) also in-
/
Inducers of Drug Metabolizing Enzyme System h ibit the enzyme activity directly.
and Drugs Commonly Affected by them
2. Indirect Inhibition : is brought about by one of the two mecha-

Inducers Drugs with Enhanced Metabolism ntsms:
Barbiturates Coumarins, Phenytoin, Cortisol, Testosterone, Oral a. Repression : is defined as the decrease in enzyme content. It may
Contraceptives be due to a fall in the rate of enzyme synthesis as affected by
Alcohol Pentobarbital, Coumarins, Phenytoin ethionine . puron1ycin and actinomycin D or because of rise in the
Phenytoin-- - -
rate of enzyn1e degradation such as by carbon tetrachloride. car-
Cortisol, Coumarins, Oral Contraceptives,
bon disulfide. disulfiram. etc.
Tolbutamide
Rifampicin Coumarins, Oral Contraceptives, Tolbutamide,
b. Altered Physiolo~~· : due to nutritional deficiency or honnonal
Rifampicin -. in1 balance.
Cigarette Smoke Nicotine, Amino Azo Dyes Direct enzyrne inhibition is usually rapid (a single dose of inhibitor
rnay be sufficient) and therefore n1ore in1portant than enzyn1e induction.
.
150 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 151
Enzyme inhibition generally results in prolonged phartnacologic action of Strain Differences/Pharmacogenetics
a drug. Some examples of inhibitors and drugs affected by them are · EnfYmes influencing metabolic reactions are under the genetic control.
given in Table 5.4. Just as the differences in drug metabolizing ability between different
species are attributed to genetics, so also are the differences observed
TABLE 5.4 between strains of the same animal species. A study of intersubject
Enzyme Inhib~tors and Drugs Affected by them variability in -drug response (due to differences in~ for example, rate of
Inhibitors Drugs with Decreased Metabolism biotramformation) is called as pharmacogenetics. · The intersubject variations
in drug biotransfonnation may either b~ monogenically or polygenically
MAO Inhibitors Barbiturates, Tyramine controlled. A polygenic control has been observed in studies in twins. In
Coumarins Phenytoin identical twins (monozygotic), very little or no difference in the metabo-
Allopurinol 6-Mercaptopurine - lism of phenylbutazone, dicoumarol and antipyrine was detected but large
PAS Phenytoin, Hexobarbital variations were apparent in fraternal twins (dizygotic; twins developed
'
from two different eggs) for the same drugs.
:&nvironmental Chemicals Differences observed in the metabolism of a drug among different
Several environmental agents influence the drug metabolizing ability races are called as ·e thnic variations. Such a variation may be mono-
of enzymes. Halogenated pesticides such as DDT and polycyclic aromatic morphic or polymorphic. When a unimodal frequency distribution is
hydrocarbons contained in cigarette smoke have enzyme induction effect. observed in the entire population, the variations are called as continuous
Organophosphate insecticides and heavy metals such as mercury, tin, or monomorphic; for example, the entire human race acetylate PABA
nickel, cobalt and arsenic inhibit drug metabolizing ability of enzymes. and PAS to ,only a small extent. A polymodal distribution is indicative of
/

discontinuous variation (polymorphism). · An example of polymorphism


Other environmental factors that may influence drug metabolism are
is the. acetylation of isoniazid (INH) in humans. A bimodal population
temperature, altitude, pressure, atmosphere, etc.
distribution was observed comprising of slow acetylator or inactivator
phenotypes (metabolize INH slowly) and rapid acetylator or inactivator
BIOLOGICAL FACTORS
phenotypes (metabolize INH rapidly) (see Table 5.5.).
, Species Differences
Screening of new therapeutic molecules to ascertain their activity and TABL£5 .5
toxicity requires study in s~veral laboratory animal species. Differences Ethnic Variations in theN-Acetylation of Isoniazid
in .drug response due to species differences are taken into account while Ethnic Group % S/olt' Acetylators % Rapid Acetylators
-extrapolating the data to man.
Whites (USA and Canada) 45 55
-Species differences have been observed in both phase I and phase II
· reactions. In phase I reactions, both qualitative and quantitative variations Blacks (USA) - 48 52
in the enzyme and their activity have been observed. An example of this Latin Americans 67 33
' . is the metabolism of amphetamine and ephedrine. In men and rabbit, American Indians 79 21 __.-

these drugs are predominantly metabolized by oxidative deamination whereas Japanese 87 13 • "
in rats the aromatic oxidation is the m~jor route. In phase II reactions, the
Eskimos 95 05
variations are mainly qualitative and characterized either. by the presence
of, or complete lack of certain conjugating enzymes; for example, in pigs, Source : Kalo\V. W.: Pharnzacogenetics: Heredity and the Response to Drugs.
the phenol is excreted mainly as glucuronide
/
whereas its sulfate conjugate Saunders. Philadelphia. 1962.
dominates in cats. Certain birds utilize ornithine for conjugating aromatic Approximately equal percent of slow and rapid acetylators are found
acids instead of glycine. among whites and blacks whereas the slow acetylators dominate 1apanese
and Eskimo populations. Dos~ adjustments are therefore necessary in the ...
152 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS •
/ 153

l~U~ groups since high levels of INH may cause peripheral neuritis. synthesis is promoted by protein diet which also raises the level of amino
Other drugs known to exhibit pharmacogenetic differences in metabolism acids for conjugation with drugs. The protein-carbohydrate ratio in the
are debrisoquine, succinyl choline, phenytoin, dapsone and sulfadimidine. diet is also important; a high ratio increases the microsomal mixed func-
tion oxidase activity. Fat free diet depresses ~ytochrome P-450 levels
Sex Differences since phospholipids, which are important components of microsomes, be-
Sex related differences in the rate of metabolism can be attributed to come deficient. Dietary deficiency of vitamins (e.g. vitamin A, B2, B3, C
regulation of such processes by sex honnones since variations among
- , _ .. - * I "'" •
and E) and minerals such as Fe, Ca, Mg, Cu and Zn retard the metabolic
I male and female are generally observed following puberty. Such sex activity of enzymes. ' Starvation results in decreased amount of glucu-
differences are widely studied in rats; the ~ale rats have greater drug r.o nides fonned than under nonnal conditions. Malnutrition in women
metabolizing capacity. In humans, women metabolize benzodiazepines results in enhanced metabolism of sex honnones. Alcohol ingestion
slowly than men and several studies show that women on contraceptive results in a short tettn decrease followed by an increase in the enzyme
pills metabolize a nutnber of drugs at a slow rate. activity.
,.

Age Altered Physiologic Factors


Differences in the drug metabolic rate in different age groups is Pregnancy : Studies in animals have shown that the maternal drug
mainly due to variations
- -.
in the enzyme content, enzyme activity and __,.J,
metabolizing ability (of both phase I and phase II reactions) is reduced
hemodynamics. In neonates (upto 2 months), the microsomal enzyme during the later stages of pregnancy. This was suggested as due to high
system is not fully developed and many drugs are biotransfonned slowly; levels of steroid honnones in circulation during pregnancy. In women,
for example, caffeine has a half-life of 4 days in neonates in comparison the metabolism of promazine and pethidine is reduced during pregnancy
to 4 hours in adults. A major portion of this drug is excreted unchanged or when receiving oral contraceptives. Higher rate of hepatic metabolism
in urine by the neonates. Conjugation with sulfate is well developed of anticonvulsants during pregnancy is thought to be due to induction of
(paracetamol is excreted mainly as sulfate) but glucuronidation occurs to a drug metabolizing enzymes by the circulating progesterone.
very small extent. As a result, hyperbilirubinemia precipitates kernicterus
and chloramphenicol leads to cyanosis- or Gray baby syndrome in new Hormonal Imbalance : The influence of sex hormones on drug me-
born. Similarly, sulfonamides cause renal toxicity and paracetamol causes tabolism has already been discussed. The effect of other hormones is
1
hepatotoxicity. Infants (between 2 months and one year) show almost a equally complex. Higher levels of one hormone may inhibit the activity
' similar profile as neonates in metabolizing drugs with improvement in the of few enzymes while inducing that of others. Adrenalectomy, thyroidectomy
capacity as age advances a:nd enzyme activity increases. Children (be- and alloxan induced diabetes in animals showed an impairment in the
tween one year and 12 years) and older infants metabolize several drugs enzyme activity with a subsequent fall in the rate of metabolism. ·A
much more rapidly than adults as the rate of metabolism reaches a maxi- similar effect was observed with pituitary growth hormone. Stress related
mum somewhere between 6 months and 12 years of age. As a result, changes in ACTH levels also influence drug biotransformation.
they require large mg/Kg doses in comparison to adults; for example, the Disease States _: As liver is the primary site for metabolism of most
theophylline half-life in children is two-third of that in adults. In very drugs, all pathologic conditions associated with it result in enhanced half-
elderly persons, the liver size is reduced, the microsomal enzyme activity lives of almost all drugs. Thus, a reduction in hepatic drug metabolizing
is decreased and hepatic blood flow also declines as a result of reduced ability is apparent in conditions such as hepatic carcinoma, hepatiti~
--·--- - -
cardiac output all of which contribute to decreased metabolism of drugs. 1
.::irrhosis, obstructive jaundice, etc. Biotransfonnations such as glycine
Drug conjugation however remains unaffected. conjugation of salicylates, oxidation of vitamin D and hydrolysis of procaine
which occur in kidney, are impaired in renal diseases. Congestive cardiac
Diet failure and myocardial infarction which result il) a decrease in the blood
The enzyme content and activity is altered by a number of dietary flow to the liver, impair metabolism of drugs having high hepatic extrac-
components. In general, low protein diet decreases and high protein diet tion ratio e.g. propranolol and lidocaine. In diabetes, glucuronidation is
increases the drug metabolizing ability. This is because the enzyme reduced due to decreased availability of UDPGA.

f
-
154 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOTRANSFORMATION OF DRUGS 155

Temporal Factors covalent binding to nucleophilic tissue components such as macromol-


Circadian Rhythm : Diurnal variations or variations in the enzyme ecules (proteins, nucleic acids and lipids) or low molecular weight cellular
activity with light cycle is called as circadian rhythm in drug metabo- constituents . . ·c ovalent binding to DNA is responsible for carcinogenicity
lism. It has been observed that the enzyme activity is maximum durmg and tumor formation. The body's defence against electrophiles is their
early morning (6 to 9 a.m.) and minimum in late afternoon (2 to 5 p.m.) inactivation by conjugation with glutathione, the most abundant cellular
which was suggested to correspond with the high and low serum levels of nucleophile with -SH group. An example of tissue toxicity due to electrophiles
corticosterone (the serum corticosterone level is dependent upon the light- is hepatotoxicity of paracetamol metabolites.
dark sequence of the day). Clinical variation in therapeutic effect of a
COCH3
drug at different times of the day is therefore apparent. The study of H COCH 3 HO COCH 3 f
Conjugation
" N/ 'N/ ' N with GSH
variations in drug response as influenced by time is called as
chronopharmacology. Time dependent change in drug kinetics is known
I

N~hydroxylation -H20 /
as chronokinetics. Drugs such as aminopyrine, hexobarbital and imipramine
showed diurnal variations in rats. The half-life of metyrapone was shown
OH
I
OH 0
"" Covalent binding
to liver tissues
to be 2.5 tin1es longer during the night than in the day, in rats. when GSH is
depleted

Paracetamol lmidoquinone derivative


BIOACTIVATION AND TISSUE TOXICITY (electrophile)
. Formation of highly reactive metabolites (from relatively inert chemi-
Free Radicals : are species containing an odd number of electrons.
cal compounds) }t'hich interact with the tissues to precipitate one or more
They may b~ positively charged (cation radical), negatively charged (an-
of the several forms of toxicities such as carcinogenesis and teratogenesis
is called as bioactivation or toxicological activation. The reactive, ion radical) or neutral (neutral radical).
chemically unstable species, capable of toxication, are broadly divided R.+ R.- R.
into two categories electroph iles and free radicals (Fig. 5.3. ). Cation Radical Anion Radical Neutral Radical
Covalent binding · Free radicals are generally fonned via NADPH cytochrome P-450
Electrophiles Tissue
to tissues Necrosis reductase or other flavin containing reductases. Xenobiotics that on meta-

Reactive
/ Cancer
bolic activation yield free radicals are quinones, arylamines, nitroaryls and
carbon tetrachloride. Endogenous compounds such as epinephrine and
Xenobiotic
• ·Metabolites
Mutation
DOPA can also .generate free radicals. Most free radicals are organic.
They produce toxicity b~ peroxidation of cellular components. An impor-
Peroxidationl
tant class of. free radic~ls is inorganic free radicals such as hydrogen
' Free
Radicals
Oxidative
degeneration of
Teratogenic
Effects
l

peroxide (H 20 2) and superoxide anion (02-). These oxidative mo1ettes


• •

cell components can cause tremendous tissue damage leading to mutations or cancer. The
potential toxicity of free radicals is far greater than that of the electrophiles.
Fig. 5.3 Mechanisms of tissue toxicity by bioactivation of drugs Cellular defence mechanisms against free radicals include control imposed
Electrophiles are species deficient in electron pair. The enzyme by membrane structure, neutralization by glutathione, control exerted by
system through which they are generated is cytochrome P-450. Carbon, nonenzymatic antioxidant scavengers such as vitamins A, E and C and
nitrogen or sulfur-containing compounds can be metabolically activated to enzymatic inactivation of oxygen derived free radicals.
yield electrophiles. Important electro.philes are epoxides (e.g. epox ide of Generation of reactive ..metabolites is indicated by modification in
benzo(a)pyrene prese~t in cigarette smoke which causes cancer), hydroxy- enzyme activities, formation of glutathione conjugates (or mercapturic
lamines, nitroso , and azox~~rivatives, nitrenium ions and elementai sulfur.
- )!:t • .;' ~
acids) and depletion in tissue levels of glutathione. Since the availability
I The mechanism by whicn· ~ electrophile.s precipitate toxicity is through of glutathione in the body determine the threshold'· for toxic response,

.
..
~~
-· . -
I
BIOTRANSFORMATION OF DRUGS 157
156 BIOPHARMACEUT1CS AND PHARMACOKINEr 'ICS
14. Phase II reactions are called as true detoxication reactions. Ex'plain.
thiols (e.g. N-acetyl cysteine) can be· used to treat poisoning by drugs such
15. Why oxidative reactions are predominant in comparison to other phase II
· as paracetamol that yi,eld reactive metabolites.
reactions?
Table 5.6 lists some of the compounds whose· metabolites are tissue
16. Explain why the oxidative ~nzymes are-called by different names - mixed
reactive. function oxidases, monooxygenases and hydroxylases.
TABLE 5.6 17. How was -the name cytochrome P-450 derived? Why is it considered to be
Compounds and their Metabolic Reaction the most important component of mixed~ function oxidases?
that Generate Toxic Intermediates 18. Outline the steps _involved in the oxidation of xenobiotics. What is th~ rate-
' limiting step · in such a process? '
Compounds Metabolic Pathway Toxicity
19. Unlike aromatic hydroxylation, oxidation of, olefins do not generate tissue
Benzo(a)pyrene Aromatic epoxidation Lung cancer reactive metabolites. Explain.

Afl~toxin 81 Olefin epoxidation Hepatic cancer 20. Why are secondary and tertiary .alcohols resistant to oxidation?
Thalidomide Hydrolytic cleavage of lactam Teratogenesis 21. Why is N-dealkylation of tertiary nitrogen rapid in comparison to that of
Chlorinated hydro- Oxidative dehalogenation Nephrotoxicity secondary nitrogen?
carbons e.g. CHCl3 22. N-dealkyl'a tion of hbutyl group is not possible. Why?
23. Explain the analogy and distinction between N-dealkylation and oxidative
deamination reactions.
QUESTIONS
. 24. What is the reason for hepatotoxicity of paracetamol, an otherwise safe
1. Name and define th e pharmacokinetic processes involved in the termination · drug, when consumed in lar&e doses? Why is it. that the t~ssu.e at the
of drug action. greatest risk for toxicity is liver when a tissue react1ve metabohte ts gener-
2. Why are drugs referred to as xenobiotics? ated?
3. Ho\v doe s biotran sforn1ation of a drug differ from its chemical instability? 25. Cite examples of 0-dealkylation reactions that yield active metabolites.
4. What \Vill happen if a lipophilic drug that is absorbed into the systemic 26. Why is it that drugs containing primary alcohol groups are rare?
circulation is not metabolized? 27. Justify the statement - bioreductions are one-half of reversible reactions.
5. What is the m~jor
function of metabolic reactions? What \Vould be the Explain why such reactions may prolong the drug action.
con sequence if biotransformation of a drug leads to generation of a less 28. In what respects the hydrolytic reactions differ from oxidative and reductive
soluble metabolite? reactions?
6. Wh'\t are soft drugs? · Why are they considered safe ( \\·.r.t. the metabolites
\
29. Why does hydrolysis of esters with one large and one small moiety leads to
formed) and have short half-lives? metabolites that retain much of their activity?
--
7. What arc the various sites of drug metabolism in the body ? Why is liver 30. List the characteristics of moieties transferred to the substrate in conjugation
considered the major site for such a process? reactions. Why are such reactions capacity-limited?
8. Depending upon the relative activity of the metabolites fonned . quote \\·ith 31. Explain why glucuronidation is the commonest' ~nd most important of all
examples the various end products of biotransformation pr()cesscs. phase II reactions .

9. On \vhat category of drugs do the microsomal and nonn1icroson1al (soluble) 32. What is the similarity between glucuronidation, sulfation and methylation
enzymes act? reactions? How does conjugation with amino acids differ from them? ·
I 0. What arc the characteristics of n1icrosomal enzvrnes?
., 33. What aspect of conjugation with amino acids can be put to diagnostic use? -
II. Classify the chemical path\vays of drug metabolism . 34. Why is it that glucuronidation can take place in most body tissues? ..
12. Which metabo lic reactions I arc considered phase III reactions? 35. The type of conjugation reaction a drug/metabolite undergoes determines its ' ' I

13. Why are phase I reaction s called as functionalization reactions? route of excretion. Comment.
158 BIOPHARMACEUTICS AND PHARMACOKINETICS

36. What is the special significance of glutathione conjugation in comparis?n to


other phase II reactions? Why does such a reaction not . require initial
activation of coenzyme or the substrate?
37. Glutathione conjugates are not detectable in urine. Why?
38. How does acetylation reaction differ from conjugation with amino acids?
What class of drugs undergo .such a reaction?
6
39. In what respects methylation reaction differs from conventional phase II
reactions? What is the biochemical importance of such a react'i on?
Prodrugs
40. How is cyanide inactivated in the body?
41. What are the possible mechanisms of enzyme induction and enzyme inhibi- .
tion by xenobiotics? What are their consequences? Why is enzyme inhibition . Almost all drugs possess some undesirable physicochemical .a~d. b.io-
considered more dangerous than enzyme induction? logical properties. Their therapeutic effic~cy can .be impr.o~ed by mm~IZmg
42. What is the interesting phenomenon observed with acetylation reactions in or eliminating the undesirable properties while retaining the des1r~ble
human race? ones. This can be achieved through biological, physical or chemical
means. The biological approach is to alter the route of administration,
43. Define pharmacogenetics. What is the major cause of intersubject variability
in drug response? which may or may not be acceptable to the patient. The physical
approach is to modify the design of the dosage fonn .such as c?ntrolled
44. A drug eliminated primarily by hepatic biotransformation shows greater
delivery of drugs. The third and the best approach m enhancing drug
intersubject "Variability than those eliminated by urinary excretion. Explain.
selectivity while minimizing toxicity is the chemical approach.
45. To what factors are the sex related differences in drug metabolism attribut-
ed? The three chemical means of optimizing the drug therapeutics are:
46. Why do children require large mg/Kg doses of some drugs in comparison to 1. Design and development of new drugs with desirable :eatures;
adults? this approach, however, requires screening of thousan~s. of chemicals for
.
47. , Neonates are at greater risk from drug intoxication than infants and children. biological activity of which only one may become a chn1cally useful drug .
Explain?
48. How does diet influence drug metabolism?
2. Design of hard and soft drugs (which are basically analogs of
existing drugs) with desirable characteristics.
49. Define chronokinetics. What factors govern the diurnal variations in drug
metabolism?
3. Design of prodrugs.
50. Define toxicologic activation. Classify tissue reactive metabolites and ex-
plain how they are generated. In the latter two approaches, the aim of the design is to develop
moieties having predictable biotransfonnation or excretion in contrast to
51. What mechanisms are involved in carcinogenesis or tissue toxicity with
electrophiles and free radicals? conventional new drug development methods.
52.. What is the biochemical indication of generation of tissue reactive metabo- A hard drug is one which is resistant to biotransformation and,
lites? therefore, has a long biological half-life.. Such a drug will be. eliminated
53. What is the body's defence in inactivating potential carcinogens? How can by the body through excre~ion in unchanged form only. Destgn of hard
dietary habits prevent tissue toxicity? druis involve metabolic stabiliza~ion of t?e existing ~g ~olecules by
' replacing functional groups susceptible to biotransfonnati~n with the sta?le
54. Outline metabolic pathways,
.' With structures, for follolw ing drugs:
a. Diazepam ' ones. Apart from enhanced duration of action and less wastage of active
b. Ibuprofen •
moiety, such a hard drug also ~void~ generation. of pot~ntially hat ~n~l
c. Lidocaine .'
metabolites decreases _tendency for drug-drug tntera~tlons and hmtts
d. Hexobarbital intersubject' variations. An example of this approach is chlorpropamide

159
I
160 BIOPHARMACEUJ'ICS AND PHARMACOKINETICS 161
PRODRUGS I

(not really a true ha~d drug since it is metabolized, though slowly), an Carrier-linked prodrugs (or simple prodrugs) are the ones where .
analog of tolbut~mide with p-methyl replaced with a chloro group. . the active drug is covalently linked to an inert ca~rier or transport
I H. moiety. They are generally esters or amides. Such prodrugs have greatly
/
502NHCON"'- modified lipophilicity due to the attached carrier. The active drug is
nBut
released by hydrolytic cleavage, either chemically or enzymically (Fig.
Tolbutamide (tY2 ·= 6H) Chlorpropamide (tY2 =33H)
' 6.1 ).

Nevertheless, hard drugs are not wit~out drawbacks; too long half- Bioprecursors or metabolic precursors are inert molecules obtained ·
lives and a potential risk of accumulafio·n· are their limitations. . by chemical modification of the. active drug but do not contain a carrier.
In · contrast to metabolic stabilization involved in hard drug design, Such a moiety has almost the same lipophilicity as the parent drug and is
metabolic switching or metabolic promotion concept, which involves bioactivate~ generally by redox biotransfottnation, only enzymatically; for
introduction in a phann~cophore, a functional group of predictable meta- example, arylacetic acid NSAID such as fenbufen from aroyl propionic
bolic reactivity, is used in soft drug and prodrug design. acid precursors. Mixed type prodrugs are also possible;_.--
A soft drug is a biologically active compound that is biotransformed· In some cases of carrier type prodrugs which are required to be
in vivo in a rapid and predictable manner into non-toxic moieties. Such fortnulated as ophthalmic, parenteral or oral liquid preparations, the con- ·
an agent has therefore a very short duration of action. Several natural version to active drug is chemically (nonenzymatically) triggered by a ·
endogenous agents such as insulin and adrenaline, which are also used as change in pH thereby creating stability problems. This can be overcome /
therapeutic compounds, are soft drugs. Design of synthetic soft drugs by use of double prodrug, pro-prodrug or cascade latentiati~n concept.
involves introduction of a group or a bond susceptible to rapid metabolic Here, the prodrug is further derivatized in a fashion such that only enzy-
action; for example, replacement of a part of the alkyl side chain of the matic conversion to prodrug is possible before the latter can cleave to
drug wi~h an ester group that can be readily hydrolyzed in vivo. An release the active drug for example, diesters of pilocarpic acid.
important advantage of soft drugs is formation of relatively inert metabolites.
enzymatic·· chemical/enzymatic
A prodrug is a chemically modified inert drug precursor which upon PRO-PRODRUG PRODRUG DRUG
cleavage hydrolysis
biotransformation liberates--the pharmacologically active parent compound.
Thus, in contrast to soft drugs, prodrugs .are inactive per se and biotransformed (stable in liquid (chemically
formulation) unstable)
in a predictable manner into active metabolites. A prodrug is also called '
'
as pro-agent, bioreversible derivative or latentiated drlJg. The design In contrast to simple prodrugs, where the carrier used is biologically
approach is also referred to as drug latentiation. I

inert, there are instances where the prodrug comprises of two pharmaco-
,I

Depending upon the constitution, lipophilicity, method of bioactivation logically active agents coupled tog~ther to fottn a single molecule such
and the catalyst involved in bioactivation, prodrugs are class-ified into two that each acts as the carrier fof the· other. Such prodrugs of two active
categories: carrier-linked prodrugs and· bioprecursors. compounds are called as mutual prodrugs e.g. benoryhite is a mutu~l ·--
prodrug of NSAIDs aspirin and paracetamol.
. In the subsequent discussion, the general tenn prodrug will be used
ACTIVE
Chemical Prodrug Formulation for all categories of such agents to avoid confusion.
DRUG
' '
I· - --.._ ...__...,.,..._ _... Covalent One of the ·earliest prodrugs to be ~covered accidentally was prontosil
+ ...--..-{166.-<-... bond which is converted to the antibacterial a&_ent sulfanilamide in the body .
INERT ' . Chemical/Enzymatic
. .
Cleavage , Nowadays, prodrugs are intentionally desi~ed to achieve specific goals.
CARRIER In VIVO
\ Some of the earlier developed prodrugs are aspirin and hexamine. Aspirin
~ PRODRUG is a prodrug of salicylic acid designed to decrease GI irritation. Hexamine
or methenamine is a prodrug which when excreted in urine is converted to
Fig. 6.1 Carrier type prodrug : formulation and drug release fottualdehyde in the acidic urine pH and acts as urinary tract antibacterial.
. .
162 BIOPHARMACEUTICS AND PHARMACOKINETICS PRODRUGS 163

An ideal prodrug should possess following properties: imperceptible. Examples of drugs with improved taste are given in Table
I. It should not have intrinsic phannacologic activity. 6.1.
TABLE 6.1
2. It should rapidly transform, chemically or enzymatically, into the
Prodrugs with Improved Taste
active fonn where desired.
3. The metabolic fragments, apart from the active drug, should be Parent Drug Prodrug with Improved Taste
nontoxic. Chloramphenicol Palmitate ester
A drug candidate for chemical modification into a prodrug is generally Clindamycin Palmitate ester
a therapeutic agent having a specific disadvantage which can be overcome
Sulfisoxazole Acety I ester
through such an approach. Since the shortcomings or insufficiencies are
Triamcinolone Diacetate ester
correcte,d through chemical conversion of the active drug into an accept-
able form, the principle of prodrug design is also ca//ed as chemical
Improvement of Odor
formulation.
The odor of a compound _depends upon its vapor pressure (and hence
1
The various applications of this approach are:
boiling point); a liquid with high vapo~ pressure (and low b.p.) will have a
I. Pharmaceutical Applications : The undesirable organoleptic proper- strong odor. Ethyl mercaptan ( ethanethiol) is one such drug which is a
ties and the physicochemical problems .as~ociated with drug formulations foul smelling liquid of b.p. 35°. Th~ drug, useful in the treatment of
can be resolved .• leprosy, is converted into its phthalare ester, diethyldithio-isophthalate
I. Improvement of taste I

2. Improvement of odor
3. Change of physical fonn for preparation of solid dosage forms .cosc2 Hs
4. Reduction of G I irritation Ethyl mercaptan Phthalate ester
5. Reduction of pain on injection which has higher b.p. and is odorless. The prodrug is administered by
6. Enhancement of drug solubility and dissolution rate (hydrophilic- rubbing on the skin. After absorption, the ester is metabolized to parent
ity of drug) drug by thioesterases.
7. Enhancement of chemical stability of drug
Change of Physical Form of the Drug
II. Pharmacokinetic Applications: Some drugs which are in liquid fonn , are unsuitable for fonnulation as
I. Enhancement of bioavailability (lipophilicity) a tablet especially if their dose is high. The method of converting such
2. Prevention of presystemic metabolism COSC2Hs
3. Prolongation of duration of action
4.. Reduction of toxicity
5. Site-specific drug delivery (drug targeting)

Impro-vement of Taste Ethyl mercaptan 1,3-Diester


I

One of the re~sons for poor patient compliance, particularly in case of


children, is the bitterness, acidity or causticity of the drug. Two ap- Cl3C- CH20H
proaches can be utilized to overcome the bad taste of drug. The frrst is
reduction of drug solubility in saliva and the other is to lower the affmity Trichloroethanol p-Acetamidobenzoic acid ester
of drug towards taste receptors, thus making the bitterness or causticity '
164 BIOfPHARMACEUTICS AND PHARMACOKINETICS PRODRUGS 165 '

·liquid drugs into solid prodrugs involves fottnation of symmetrical mol- potassium or amine salts and render the moiety water soluble. For -
~cules having a higher tendency to crystallize; for example, esters of ethyl phenolic drugs and some alcohols as in the case of steroidal drugs such as.
mercaptan and trichloroethanol. cortisol, prednisolone, betamethasone and dexamethasone, the sodium suc-
cinate salts have poor chemical stability and hence phosphate esters are
Reduction of GI Irritation preferred. Glycosidic prodrugs of some agents and L-lysine esters of
Several drugs cause irritation and damage to the gastric mucosa through benzodiazepines are also water soluble. Such hydrophilic promoieties
direct contact, increased stimulation of acid secretion or through interfer- when meant for parenteral use are advantageous over their propylene
ence with the protec_tive mucQsal layer. The NSAIDs, especially the glycol solutions which are toxic or painful. Examples of hydrophilized
salicylates, have such a tendency. They lower the gastric pH and induce prodrugs are given in Table 6.3.
or aggravate ulceration. Examples of prodrugs designed to overcome
such problems of gastric distress are given in Table 6.2. TABLE 6.3
Hydrophilic Prod rugs of Poorly Aqueous _S oluble Drugs
TABLE
. 6.2
Parent Drug Prodrugs with Enhanced Hydrophi/icity
Prodrugs to Reduce Gastric Irritation
Chloramphenicol Sodium succinate ester
Parent Drug Prodrugs That Cause Little/
No Gastric Distress Tocopherols Sodium succinate ester
Corticosteroids 21-sodium succinates, 21-phosphate esters
Salicylic acid Salsalate, Aspirin
Testosterone Phosphate ester
Diethyl stilbestrol Fosfestrol
Menthol P-Glucoside
Kanamycin Kanamycin pamoate
Sul fanilamide Glucosyl sulfanilamide
Phenylbutazone N-methyl piperazine salt
Tetracycline Tetralysine
Nicotinic acid Nicotinic acid hydrazide
Diazepam L-lysirte ester
Oleandrin 0 leandrin acetate
Metronidazole Amino acid esters
Reduction of Pain on Injection Many of the water-soluble prodrugs for oral use act as substrates for
Intramuscular injections are particularly painful when the drug precipi- enzymes in the brush border region of microvilli where they are biotransfonned
tates or penetrates into the surrounding cells or when the solution is into free active drug having lipophilic property facilitating permeation
-strongly acidic, alkaline or alcoholic; for example, th.e low aqueous solu- across the biomembrane and rapid absorption.
bility of clindamycin hydrochloride and the alkaline solution of phenytoin
are responsible for pain on injection. This can be overcome by use of Enhancement of Chemical Stability
more water soluble prodrugs of such agents, for example, the 2'-phosphate A drug may destabilize either during its shelf-life or in the GIT when
ester of clindamycin. used orally. Shelf-life stability is particularly important in case of drugs
for intravenous use . The conventional approach is to lyophilize such
Enhancement of Solubility and Dissolution Rate (Hydropbilicity) of Drug solutions into a powder which can be reconstituted before use. The

. Hydrophilic or water-soluble drugs are desired where , dissolution is prodrug design of such agents is also a good alternative to improve
the rate-limiting step in the absorption of poorly aqueous soluble agents or stability. An example of this is the antineoplastic drug, azacytidine. The
when parenteral or ophthalmi~ fonnulation of such agents are desired. aqueous solution of this drug is readily hydrolyzed but the bisulfite prodrug
Drugs ·With hydroxyl function can be converted into their hydrophilic is stable to such a degradation at acidic pH and is more water soluble
fonns by use of half-esters such as hemisuccinates, hemiglutarates or than the-parent drug~ The prodrug -co"nverts to active drug at the physio-
hemiphthalates; the other half of these acidic carriers can forrn sodium logic pH of 7 .4.
'

'
PRO DRUGS 167
166 ' BIOPHARMACEUTICS AND PHARMACOKINETICS
\
A big advanta£e
. ·- .
of increased.. bioavailability through increased lipophilicity
is reduction in drug dosage; for example, bacampicillin is as effective as
ampicillin in just one-third the dose of latter.
The bioavailability of topically administered drugs also depend upon
lipid solubility. Skin penetrability of polar drugs can be improved by
esterification to fonn lipid soluble compounds. One of the best ap-
Azacytidine Stable bisulfite prodrug proaches in enhancing topical availability of drugs with carboxyl function~ ·
The dry powder of nafate ester prodrug of cefamandole has improved is their esterification with one of the hydroxyl groups of propylene glycol
shelf-life stability over the parent drug. The prodrug rapidly converts to or glycerol. The latter are common penetration enhancer components of
. the active drug upon reconstitution for parenteral administration. topical formulations, e.g. glyceryl ester of naproxen .

A class of drugs susceptible to hydrolysis and destabilization in gastric Prevention of Presystemic Metabolism
acid is penicillins. Carbenicillin, a broad spectrum penicillin, cannot be Several corticosteroids undergo extensive first-pass hepatic metabolism
given orally for the same reason. Its ester prodrugs carindacillin (a- which can be prevented by :use of their ester or ether prodrugs - for
indanol ester) and carfecillin (a-phenyl ester) are however stable at gastric example, triamcinolone acetonide. Propranolol is another drug with high ·
pH. In the intestine, hydrolysis of these agents release carbenicillin at pH first-pass hepatic extraction; its hemisuccinate prodrug is resistant to ester- _
above 7. 0. The latter is stable under such a condition and is absorbed ases of liver. The first-pass hepatic metabolism of opiate narcotic agonists
intact. Erythromycin also has a similar acid instabi_lity problem. Its , or an tag on ists can be reduced by protecting the enzyme labile phenolic
stearate, ethyl succinate and estolate (lauryl sulfate salt of erythromycin hydroxy group (that undergoes rapid glucuronidation) by fonning a prodrug
propionate) ester prodrugs are stable to hydrolysis in stomach. with aspirin. The parent drug is regenerated during first-pass tran~it of the
prodrug through the liver.
Enhancement of Bioavailability (Lipophilicity)
Most drugs are absorbed by passive diffusion for which lipophilicity is Prolongation of Duration of Action
an important prerequisite. Two reasons can be attributed to the enhanced Frequent dosing is required for drugs having short biological half-
oral bioavailability of lipophilic compounds: lives~ this can be overcome by use of both controlled release and prodrug
1. The lipophilic fortn of a drug has enhanced membrane/water approaches. The two rate-controlling steps (Fig. 6.2) in the enhancement
partition coefticient as compared to the hydrophilic forrn thus
'
of duration of drug action ace:
favoring p·assive diffusion; for example, the pivampicillin, t. The rate of release of prod rug from the site of application or
bacampicillin and talampicillin prodrugs of ampicillin are more administration into the systemic circulation, and
lipophilic, better absorbed (above 98%) and rapidly hydrolyzed to 2. The rate of conversion of prodrug into active drug in the blood.
the parent drug in blood, and
2. The lipophilic prodrugs, for example, the esters of erythromycin, PRODRUG DRUG
have poor solubility in gastric fluids and thus greater stability and IN TISSUES IN TISSUES

better absorption.
1l 1l
The dipalmitoyl glycerol ester of the NSAID naproxen produces less G) PRODRUG 0 DRUG
gastric irritation and higher plasma concentration. The intraocular pen- PRODRUG IN BLOOD IN BLOOD
controlled con trolled
etration of polar drugs such as ~-blockers and epinephrine, in the treatment
of glaucoma, can be promoted by use of lipophilic carrier prodrugs of
release
~
p
activation 'II
,
such agents; for example, the diacetate ester of nadolol is 20 times more PRODRUG DRUG
ELIMINATED EL~MINATED
lipophilic and 10 times more readily absorbed ocularly. The dipivalyl
ester of epinephrine has good ocular penetrability (8 to 17 times) in Fig. 6.2 Ratc-lirniting steps in the release of a drug .from prodrug_s
comparison to the parent drug.
168 BIOPHARMACEUTICS AND PHARMACOKINETICS PRODRUGS 169

The control of either of these two steps would result in prolongation of bolic rate in ocular tissue~.). lbuterol, the diisobutyrate ester of terbutaline,
drug action. The easier approach, that of controlling the release rate of a selective ~ 2 -agonist, is also useful in glaucoma. This prodrug is 100
prodrug, is ulseful when in vivo conversion of the latter into active drug is times more potent, has longer duration of action and is free of both local
rapid. Examples include ~he i.m. depot injections of lipophilic ester and systemic toxicity (tachyphylaxis).
prodrugs of steroids (testosterone cypionate and propionate, estradiol pro-
pionate) and antipsychotics (fluphenazine enanthate and decanoate). Since Site-Specific Drug Delivery (Drug Targeting)
, I

testosterone and estradiol are natural soft drugs, their lipo_philic prodrugs After its absorption into the systemic circulation, the drug is distrib-
are sometimes called as prodrug-soft drugs. uted to the various parts of the body including the target site as well as
'
the nontarget tissues. Such a distribution pattern has several disadvan-
The second approach of controlled conversion of prodrug to active
drug -_tl~ough difficult, was successfully utilized to deliver pilocarpine to tages:
.e yes 4ri the treatment of glaucoma. The diesters of the drug when applied 1. The drug may lead to undesirable toxic effects in the nontarget
as ophthalmic solution showed better intraocular penetration due to im- tissues (especially if its therapeutic index is low)~
. proved lipophilicity, and slow conversion of the ester prodrug to active 2. A smaller fraction of the drug will reach its target site because of
pilocarpine prolonged the therapeutic effect. The rate of conversion dilution due to distribution which may be insufficient to evoke the
however, is greatly dependent upon the ester group. Bitolterol, a di-p- therapeutic response.
toluate ester prodrug of N-t-butyl noradrenaline has a longer duration of 3. If the target site has a long distribution time, the drug may get
bronchodilator activity than the parent drug. Moreover, the drug preferen- eliminated without reaching such a site.
tially distributes in the pulmonary tissues and, therefore, does not show
4. Even if the drug reaches the target cells in sufficient amounts, it
adverse cardiovascular effects.
may not be able to penetrate into them.
Reduction of Toxicity These problems can be overcome by targeting the drug to its site of
An important objective of drug design· is to develop one with high action by altering its disposition characteristics. There are several ap-
(lctivity and low toxicity. Examples of drugs for systemic use with local proaches to drug targeting and prodrug design is one of them. The
side effects such as gastric distress with NSAIDs, which can be overcome prodrug is converted into its active form only in the target organ/tissue by
by prodrug design, have already been discussed. Another example of this utilizing either specific enzymes or a pH value different from the normal
is the bioprecursor sulindac. As a sulfoxide, it does not cause any gastric pH for activation. Some of the important examples of site specific drug
irritation and is absorbed better. In blood, it is converted to its active delivery are discussed below.
sulfide forrn.
1. Selective Uptake Systems
The utility of some drugs for local use is limited by the incidence of
[A] Dopamine, a neurotransmitter, produces vasodilation of renal
systemic side effects such as those with timolol and epinephrine which are
tissues by binding to specific receptors in the kidney and thus can be
used in the treatment of glaucoma. Therapy of such a condition requires
used to treat renal hypertension. However, the therapeutic index of
instillation of higher concentration of drug since the agents have poor
dopamine is small as it precipitates high blood pressure by interaction
intraocular penetration. But higher doses of such drugs cause irritation to
with the a-adrenergic receptors. This can be overcome by taking advan-
eyes and systemic absorption precipitates undesirable cardiovascular ef-
tage of the fact that the y-glutamyl derivatives of amino acids and peptides
fects. Lipophilic ester prodrugs of such agents on the other hand have
selectively accumulate in the kidneys. Such a derivative of dopamine, on
better intraocular penetration enabling a reduction in the instilled dose and
reaching the kidneys, is acted upon successively by two enzymes that are
thus adverse effects are limited; for example, the therapeutic index of
present in high concentration in the renal tissues, y-glutamyl transpeptidase
alkyl ester prodrugs of ~imolol improved 16 times while that of dipivalyl
and L-aromatic amino acid decarboxylase to release the active drug do-
epinephrine or dipivefrin (a diester of epinephrine with pivalic acid)
pamine locally. The increase in dopamine levels produces a marked
increased 10 times. The epinephrine prodrug also has improved chemical
increase in renal blood flow.
(i.e. resistance to oxidation) and biochemical stab~lity (decreased meta-

r .'•
170 BIOPHARMACEUTICS AND PHARMACOKINF ·;~ ICS
HO • PRO DRUGS 171
COOH 0 NH2
I II I ketone prodrug, diisovaleryl adrenalone is administered, the sequential
, NH-C-CH2-CH 2 CH- COOH
CH2- CH-
reduction-hydrolysis by the two enzymes regenerate adrenaline that shows
y-G iutamyl DOPA (prodrug) its phannacologic · action at the target site. The drug is generated only
I when reduction precedes hydrolysis. If hydrolysis occurs frrst, the adrenalone
y-Giutamyl
fonned will not be reduced to epinephrine.
trans peptidase

HO I (C) Another example of site-specific delivery is that of acyclovir, the


antiviral drug useful in herpes infections. After entry into the infected
HO 0 :+ Gtu tomic oc id cells, the drug is acted upon by the viral enzyme thymidine kinase to form
acyclovir monophosphate that cannot diffuse back out of the cell. The
DOPA ( B1oprecursor) Acyclovir cellular Acyclovir -~) Destroys
)
monophosphate kinases triphosphate Viral DNA
HO
A Bioprecursor
HO Renal vasodilation monophosphate is further converted to the active triphosphate fonn by the
cellular enzymes. The triphosphate then destroys the viral DNA. Thus,
Dopamine (DOPA = Dihydroxyphenylalanine)
activation of acyclovir occurs only in the cells infected with the virus.
. The same principle can be used to deliver sulfonamides selectively to
k1dneys; the prodrugs used are N-acyl-y-glutamyl suJfonamides. (D) Mesalamine (mesalazine, 5-amino salicylic acid) is a drug useful
in the treatment of inflammatory bowel disease (ulcerative colitis) since it
I BJ Enzymatic activation of the prodrug at the site of action or target is not absorbed into the systemic circulation. However, following oral
is also utilized to deliver epinephrine to eyes in the treatment of glau- administration, the drug is inactivated before reaching the lower intestine,
coma. The disadvantages of direct ocular administration of adrenaline are the site of action. Covalent binding of this agent to sulfapyridine yields
known. The drug is also known to cause corneal staining. Use of a the prodrug sulfasalazine, an azo compound. 'This prodrug reaches the
simple ester prodrug has the disadvantage of being hydrolyzed rapidly colon intact where cleavage by the bacterial enzyme azo reductase re-
because of easy availability of esterases in several tissues. Specific deliv- leases the active mesalamine for local action.
ery to the eyes necessitates presence of a specific ocular enzyme different
from esterases that acts on a specific prodrug to activate it. The iris- • COOH
ci I iary body of the eyes where the epinephrine acts, contai~s an enzyme colonic bacteria
ketone reductase in addition to the usual esterase. Hence, when a diester HO N=-N
azoreductase
RO
0 ()i Mesalamine
II ketone reductase I
RO----< C-CH2NHCH3 RO---( >--CH- CH2NHCH3
Sulfasalazine
+
I I
Diester adrenalone Diester adrenaline
j esterase j esterase Sulfapyridine
HO
0 OH
II I A disadvantage of the carrier moiety sulfasalazine, is its systemic
HO C- CHzNHCHJ HO CH-CH2-NHCH3 toxicity. More inert carriers can be used to deliver mesalamine to colon
e.g. 4-amino benzyl alanine (prodrug is basalazide). Another much im-
Adrenalone Adrenaline proved prod rug is a dimer of mesalam ine known as olsalazine which is
(generates only at the target site)
also an azo compound
172 BIOPHARMACEUTICS AND PHARMACOKINETICS
PRODRUGS 173
. [E) Hexamine (methenamine) is a prodrug of the urinary tract antisep-
chemical
tic, founaldehyde. After oral absorption, hexamine remains inert and [0] + [OHC] [0-0HC] lipophilic prodrug
stable in blood at pH 7 .4. When excreted in urine, the prodrug decom- coupling
poses in the acidic pH to generate active formaldehyde which exerts its i.v. administration
antibacterial action in the urinary tract.
N
((N~ or N4(CH2)6 1-
+ ..
6 H20 + 4H - - 4NH~t + HCHO
NJ N BBB
'-.....-/

Hexamine Formaldehyde

Since 10 to 30% of the prodrug decomposes in the stomach and causes


gastric distress, it is generally formulated as enteric coated/buffered tab- [0-DHC]
lets. (D-DHC]- Systemic oxidation
Circulation Brain lock-in
2. Redox System for Drug Delivery to Brain [0-QC•]
The high selectivity and poor permeability of the blood-brain barrier enzymatic
clearage
limits the delivery of hydrophilic drugs to the brain and thus, therapeutic
concentration is difficult to achieve. Conversion into their lipophilic oxidation
forms leads to simultaneous enhancement of transport of drug to other
tissues thereby greatly increasing the chances of systemic toxicity. More-
over, to achieve therapeutic levels in the brain, the plasma concentration
of the drug has to be very high. Thus, specific delivery of the drug to the
brain is very difficult. enzymatic
cleavage
A more recent, novel and smart approach for delivery of drugs to
brain is use of dihydropyridine <:::> pyridinium salt redox system . The
drug to be delivered to the brain is covalently linked to the lipoidal
dihydropyridine carrier group to form a prodrug which will partition
across the otherwise highly selective BBB. Following administration, the e I i m i n a t i on
\.

prodrug is rapidly distributed throughout the body as well as' in the brain.
The reduced or the dihydro form of the carrier prodrug is oxidized by Fig. 6.3 Dihydropyridine ¢::> pyridinium salt redox system for drug deliv-
NAD-NADH system , both, in the brain and the body, to form lipid ery to brain. [D] = drug, [DHC] = lipophilic dihydro form of the
carrier, [D-DHC] = lipophilic drug-dihydro carrier conjugate, [D-
insoluble, polar quaternary pyridinium salt fonn (which is inactive). Due
QC+] = hydrophilic drug-quaternary carrier conjugate, and [QC+]
to high hydrophilicity of such an ionic prodrug, the amount present in the
= quaternary form of the carrier.
systemic circulation is subjected to rapid renal excretion but the similar
polar for rn present in the brain is prevented from diffusing out of the The choice of the carrier in such a redox system is important; it must
lipophilic BBB resulting in its lock-in in the CNS. The drug is slowly be nontoxic, both, when alone and as a conjugate with the drug. The
released from such an oxidized prodrug into the CSF by chemical/enzy- carrier that was used successfully is trigonelline (N-methyl nicotinic acid).
matic process, allowing the therapeutic concentration to be maintained • The technology holds great potential in treating various conditions of
over a prolonged perfod of time. Thus, the drug is preferentially targeted brain caused by neurotransmitter disorders such as Parkinsonism as was
to the brain whereas its systemic concentration is negligible (Fig. 6.3.). proved by successful delivery of dopamine to the brain of r~ts.

• •
174 BIOPHARMACEUTICS AND PHARMACOKINETICS
PRO DRUGS 175
HO
(C) Hepatic carcinoma is rich in azoreductase levels. Azobenzene
coo- COOH 0
II
OH

Reduction C-NH·C~-CH2 · OH
mustard prodrug can thus be targeted to such a tissue where it is cleaved
Dopamine (polar, to release the active drug.
cannot cross BBB)
[D] A novel approach of the above technology in the treatment of
cancer is use of Antibody Directed Enzyme Prodrug Therapy (ADEPT).
Trigonelline Dihydro form of Lipophillic drug-dihydro
(inert carrier) carrier (lipid soluble) carrier conjugate
The method involves conjugating to the antitumor antibody, a specific
enzyme that selectively activates the prodrug of a cytotoxic agent. When
A similar approach was utilized to treat poisoning with organophos- such an enzyme~antibody ·conjugate is administered, the antibody selec-
phorus compounds (nerve gases) which are cholinesterase inhibitors. The tively attaches to tumor cells. Such an antibody that carries the enzyme
antidote, N-methyl pyridinium-2-carbaldoxinle (2-PAM or pralidoxime), a specifically to the carcinoma/target site is called as homing device. Sub-
potent reactivator of cholinesterase, penetrates the BBB poorly due to its sequent administration of the prodrug of cytotoxic agent results in its
activation only by the enzyme at the target site thus destroying only the .
reduction tumor cells (Fig. 6.4.). In mice, the ADEPT technique was found to be
oxidation
~ CH= N-OH
more effective than the conventional chemotherapy.
CH=N-OH ,
CH3
ADMINISTRATION
2-PAM Pro-2-PAM
(Hydrophilic, cannot (lipophilic,
LEGEND
cross BBB) crosses BBB)

quaternary nitrogen. However, the reduced dihydropyridine form of 2-


B Enzyme
TUMOR
CELLS
PAM, called as pro-2-PAM, readily enters the CNS. Once inside the
'brain, it is oxidized to polar 2-PAM and thus trapped in the CNS.
~ Antibody

Enzyme-
3. Site-Specific Drug Delivery in Cancer Antibody ACTIVE ANTI-
Several cancerous tissues and tumors are rich in certain enzymes as Conjugate NEOPLASTIC
AGENT
compared to those found in nonnal tissues. Thus, a prodrug can be ~ Antigen
designed to selectively target such tumorous cells where it can be acti- •

vated to parept antineoplastic agent. The approach protects the normal Fig. 6.4 ADEPT system for targeting carcinoma
cells from the cytotoxic effects of the drug.
The foregoing discussion on the several applications of prodrug design
[A] Prostrate carcinomas are particularly rich in enzymes acid phos- suggests that the gain in therapeutic benefit from such an approach may
phatases. Thi~ fact was used to design stilbestrol diphosphate to treat either be modest or marked. For well-accepted useful drugs that display
such ~onditions. The prodrug is activated to yield stilbestrol exclusively some unwanted property which can be ameliorated by prodrug design, the I

in the target organ·. gain is usually modest but real. Prodrugs of such agents are called as
post hoc designed. On the other hand, for active compounds that suffer
[B] Prostrate cancer can also be treated by utilizing the fact that from some severe limitation, for example, high hydrophilicity restricting
'
estrogen has strong and specific affinity for such a tissue. Thus, nitrogen bioavailability, prodrug design leads to a marked therapeutic gain. Prodrugs
mustard linked to estradiol can be used for selective targeting of prostrate of such agents are called as ad hoc designed.
where hydrolysis of the. prodrug will release the,. cytotexic mustard to
destroy the cancer cells.

\

t
••

PRODRUGS 177
176 BIOPHARMACEUTICS AND PHARMACOKINETICS
4. The prodrug design approach is called as drug latentiation or chemical
Limitations of Prodrug Design formulation. Explain.
One of the biggest problems that can arise in prodrug design is the 5. Name, define and compare the two categories of prodrugs. Why sometimes. ,
toxicity which may be due to: it becomes necessary to design a double prodrug? What do you understand
1. Forn1ation of an unexpected metabolite from the total prodrug that by mutual prodrugs?
may be toxic.
0 •

6. Enumerate ·the properties of an ideal prodrug.


2. The inert carrier generated following cleavage of prodrug may 7. What criteria are necessary for a drug to be a candidate for prodrug design?
also transfonn into a toxic metabolite. 8. List the various pharmaceutical and pharmacokinetic applications of prodrug _
3. During its activation stage, the prodrug might consume a vital design. .,
cell constituent such as glutathione leading to its depletion. 9. What principles are utilized in prodrug design for taste masking and odor
An example of generation of reactive intertnediates from prodrug is improvement of drugs?
.~

phenacetin. The activity of this agent is due to fonnation of paracetamol 10. Cite examples of prodrugs used to overcome gastric distress.
'
by de-ethylation. Compared to phenacetin, paracetamol is relatively safe 11. How are· poorly water soluble drugs containing hydroxyl function converted
unless used in high doses. Phenacetin, apart from generating acetamino- into hydrophilic prodrugs? Cite ·examples.
phen, is also hydrolyzed at the acetamido linkage to fonn p-phenetidine '
12. How does chemical modification leading to increased lipophilicity enhances
which is further metabolized to yield products that precipitate bioavailability and therapeutic efficacy and decreases toxicity of existing
·-
methemoglobinemia, hemolysis and renal toxicity. Moreover, the N- drugs?
hydroxylated metabolites of phenacetin generate reactive intennediates. 13. How can the prodrug design approach be utilized for controlled delivery and
prolonged action of active principles?
NHCOCH3 14. What are the shortcomings of a conventional drug w.r.t. ~its distribution
pattern? What are the two principles behind enhancing specific delivery of
Paracetamol ---~) . Analgesic/antipyretic
drugs to the target site using the prodrug approach?
15. Discuss the metabolic role of tissues in selective drug targeting using the
prodrug principle.
NHCOCH3
16. The pH at a specific target site can be utilized for activation of prodrugs.
p-Phenetidine · ---~)Toxic metabolites Elaborate with examples.
17. How is the redox system used successfully for drug targeting to brain?
18. Comment on the ADEPT system for targeting of antineoplastics to cancer
cells.
19. What are post hoc and ad hoc designed prodrugs?
• 0

-~> Reactive intermediates 20. What could be the most important drawback of prodrug design?

QUESTIONS

1. What are the various approaches to overcome the undesirable physiochemical,


phannacokinetic and pharmacodynamic properties of a drug?
2. \Vhat are hard drugs? What are their advantages over a conventional drug?

3. How do prodrugs differ from soft drugs?


EXCRETION OF DRUGS 179·

Rate of = Rate of + Rate of - Rate of (7.1)


,. excretion filtration secretion reabsorption
..

7 BLOOD
--......

1 Glomerular filtration of

Excretion of Drugs GLOMERULUS _ _. water and unbound


drugs and metaboJites
'
G) 2 Active tubular secretion
of acidic and basic
Drugs and/or their._ metabolites are removed from the body by excre- PROXIMAL TUBULE ----.
12'++-
\!:.)
drugs and metabolites
'

. tion. Excretion is defined as the process whereby drugs and/or their_ 3 Active reabsorption
· metabolites are irreversibly transferred from internal to external environ- ® '---::...,.. BLOOD of acidic and basic
--inent. Excretion of unchanged or intact drug is important in the tennination endogenous

compounds and
of its pharmacologic aet~on. The principal organs of excretion are kid- '
passive reabsorption
LOOP OF HENLE--4
neys. Excretion by organs other than kidneys such as lungs, biliary · of lipophilic d,rugs.
-. system, intestine, salivary glands and sweat glands is known as nonrenal
4 Urinary excretion of
: excretion. DISTAL drugs and metaboli~es
TUBULE .· that are filtered and/or
actively secreted and
RENAL EXCRETION OF DRUGS COLLECTING __....~ URINE not reabsorbed.
Almost all drugs and their metabolites are excreted by the kidneys to TUBULE

some extent or the other. Some drugs such as gentamicin are--eKclusively Fig. 7.1 A simplified diagram illustrating processes
~ eliminated by renal route only. Agents that are water-soluble, nonvolatile, involved in the urinary excretion of drugs
_,s mall i.n mol~cular siie (less than 500 daltons) and which are ' metabolized
slowly, are excreted in the urine. Glomerular Filtration
The bas·ic functional unit of the kidney involved in excretion is the Glomerular filtration is a nonselective, unidirectional process whereby
nephron. Each kidney comprises of one million nephrons. Each nephron most compounds, ionized or unionized, are filter~d except those that are
is ~ad~ up of the glomerulus, the proximal tubule, the loop of Henle, the hound to plasma proteins or blood cells and thus beha~e as macromolecules.
distal tubule and the collecting tubule. Tbe glomerulus also acts as a negatively charged selective barrier promoting
' retention of anionic compounds. The driving force for filtration through
_The principal processes that detet 111 ine the urinary excretion of a drug
the glomerulus is the hydrostati¢ pressure of -the blood flowing in the
are:
capilh;tries. Out of the 25% of cardiac output or 1.2 liters of blood/min
1. Glomerular filtration, that goes to the kidneys via renal .a rtery, only 10°/o or 120 to 130 mllmin ._
.
2. Active tubular secretion, and is filte·r ed through .the glomeruli, the rate being called as the glomerular

J.. Active or passiv~ tubular reabsorption. filtration rate (GFR). Though some 180 liters of protein and cell free
\ ultrafiltrat'e pa~s through the glomeruli each day, only about 1.5 liters. is
These processes· are depicted in Fig. 7. 1. _
excreted as ~rine, the remainder- being reabsorbed from the_tub1:_d es.
Glomerular filtration
. and active tubular secretion tend to.., increase the The GFR can be detennined by an agent that is ·excreted e~clusively ·
concentration of drugs in lumen and h~nce facilitate excretion whereas
-- - - - by filtration and is neither secreted ..nor reabsorbed
-. - ·- - . . . .. .
in the tubules. Tht.
tubular reabsorption decreases it and prevents the movement of drug out ~ .-.. ............

. :xcretion rate value of such an agent is 120 to 130 ml/min. Creatinine,


_ of the body, Thus, the rate of excretion can be given by equation:
- . : j nulin, mannitol and sodium thiosulfate are used to estimate GFR of which
I
·' .· .
the· fotrner two are widely used to estimate renal function .
178
180 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS 181.

Active !ubular Secretion · large doses as ·in gonococcal infections. The actively se,c reted and filtered
It is a carrier-mediated process which requires energy for transporta- probenecid, if unionized in tubular fluid, is highly lipid soluble and ther~­
tion of compounds against the concentration gradient. The system is fore will get reabsorbed passively. Inhibition of drug secretion by probenectd
capacity-limited and saturable. Two active tubular secretion mechanisms · is undesirable in case of nitrofurantoin since the latter is used as a urinary
have 'been identified: tract antiseptic (organic ba~es can also interfere with tubular secretion of
'
cationic drugs but are not in therapeutic use). While inhibiting the active
1. System for secretion of organic acids/anions like penicillins, secretion of anionic drugs on one hand, probenecid is known to suppress / .
. salicylates, glucuronides, sulfates, etc. It is the same system by
the carrier-mediated reabsorption of the endogenous metabolite, uric acid
which endogenous acids such as uric acid are secreted.
- - and is thus of therapeutic value as a uricosuric agent in the treatment of
2. System for secretion of organic bases/cations like morphine, gout.
mecamylamine, ·hexamethonium and endogenous amines

such· as
catecholamines, choline, histamin.e, etc. Tubular Reabsorption
Tubular reabsorption occurs after the glomerular filtration of drugs. It .
Both the systems are relatively nonselective and inrlependent of each
- - .. -. takes place all along the renal tubule. Reabsorption of a dru~ is indicated ·
other but both can be bidirectional i.e. agents may both be secreted as
when the excretion rate values are less than the GFR of 130\mllmin. An
well as reabsorbed actively for example, uric acid.
agent such as glucose that is completely reabsorbed after filtration has a
Active secretion is unaffected by changes in pH and protein binding clearance value of zero. Contrary to tubular secretion, reabsorption
sine~ th-e bound drug rapidly dissoCiates the moment the unbount I dru~ results in an increase in the half life of a drug .
. gets excreted. But in contrast to glomerular filtration, -it ~ is -dependent
Tubular reabsorption can either be an:
upon renal ·blood flow. Drugs undergoing active secretion have excretion
rate values greater than the nonnal GFR value of 130 mllmin; for example, 1. Active process, or
penicillin has renal clearance value of 500 rnllmin. Such a high value is 2. Passive process.
indicative of both glomerular filtration as well as tubular secretion. Active tubular reabsorption is commonly seen with high threshold
Agents that are used to measure active tubular secretion are the ones endogenous substances or nutrients that the body needs to conserve such
that are filtered as well as secreted t'o such an extent that they are ·as electrolytes, glucose, vitamins, amino acids, etc. Uric acid is also
removed from the blood in a singl~ pass ·through the kidneys i.e. their actively reabsorbed (inhibited by the uricosuric agents). Very few. ~gs ·
clearance reflects the renal plasma flow rate which is 600 to 700 ml/min. are known to undergo reabsorption actively e.g. oxopurinol.
Para amino hippuric acid (PAH), a highly polar agent and iodopyracet are Passive tubular reabsorption is common for a large number of exog-:'
used to detettnine active secretion. Active secretion occurs predominantly enous substances including drugs. The driving force for such a process
in the proximal tubule region of the nephron. i.e. the concentration gradient is established by the back diffusion or
-- Any two structurally similar drugs having similar ionic charge and reabsorption of water along with sodium and other inorg~nic ions . t?n-
employing the same carrier-mediated process for excretion, enter into derstandably, if a drug is neither secreted nor reabsorbed, tts .concentration
competition. A drug with greater rate of clearance will retard the excre- in the urine will be 100 times that of free drug in plasma due to water
tion of the other drug with which it competes. The half-life of both the reabsorption since less than 1o/o of glomerular filtrate is excreted as urine.
drugs is increased since the total sites for active secretion are limited. The primary detenninant in the passive reabsorption of drugs is their
This-·may result in accumulation of drugs and thus, precipitation of toxic- , . lipophilicity. Lipophilic substances are extensively reabsorbed while polar
. ity. However, the principle of competition can be exploited for therapeutic molecules are not. Since a majority of drugs are weak electrolytes (weak
benefits. An int~resting exampJe of this is the anionic age~t probenecid.
1A • \
acids or weak _,~ases), diffusion of such agents through the lipoidal tubular
Probenecid inhi/~(t tl he active tu~,ular secretion of organic acids such as r . .embrane depend upon the degree of ionization ~~~ i_!l_ _~ -~~£~nd_s_on
penicillins, PAS,~·~h~~H, 17-keto steroids, etc. thus increasing their concen- two Important factors:
tration in pl~sma by at least two fold. A 50% reduction in penicillin G I

1. pH of the urine
dose is suggested, especially when the drug is meant to be consumed in •
2. pK 3 of the drug.
182 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS 183

Urine pH : It is an important factor in the sense that it is not constant drugs at various values of urine pH, assuming that the drug does not bind
like the plasma pH but varies between 4.5 to 7.5, the two extremes. Thus, to plasma ·proteins, urine flow of 1 ml/min, plasma pH_ 1.4 and that
a large pH gradient may exist between urine and plasma. equilibrium is achieved by diffusion of unionized drug only. The renal
The pH of the urine is dependent upon diet, drug intake and patho- clearance values ClR are computed by use of equation 7.8.
physiology of the patient. Food rich in carbohydrates result in higher
TABLE 7.1
urinary pH whereas proteins lower it. Drugs such as acetazolamide and
Percent Drug Ionized and Renal Clearance Values (in ml/min)
·antacids such as sodium bicarbonate produce alkaline urine while ascorbic
acid makes it acidic. More significant alteration in urine pH is brought Drugs pKa Nature Urine pH Values
1bout by i.v. infusion of solutions of sodium bicarbonate and ammonium 4.5 6.3 7.5
chloride which are used in the treatment of acid-base imbalance. Respira-
tory and metabolic acidosis and alkalosis result in acidification and % % %
alkalinization of the urine respectively. Ionized CIR Ionized CIR Ionized CIR
I'•

The relativ~ amount of ionized and unionized drug in the urine at a Acids
particular pH and the percent of drug ionized at this pH can be computed A 2.0 Strong 99.7 0.001 99.99 0.8 100.0 1.26
from the Henderson-Hasselbach equations: B 6.0 Weak 3.0 0.04 66.6 0.115 97.0 1.25
c 10.0 V.Weak 0.0 0.99 0.02 0.99 0.3 1.0
[ionized]
for weak acids, pH= pKa + l o g - - - - - (7.2)
[unionized] Bases
D 12.0 Strong 100.0 794.3 100.0 12.6 99.99 0.79
10PH-pKa
% Drug ionized = - - - - - x 100 (7.3) E 8.0 Weak 99.9 635.2 98.0 10.26 76.0 0.83
1 + 10PH-pKa 24.0 1.32 0.0 1.0 0.0 1.0
F 4.0 V.Weak
'
[unionized] u
for weak bases, pH = pKa + log - - - - - (7.4) Re,n al Clearance = - X Urine Flow Rate (7.8)
[ionized]
ClR (ml/min) P (ml/min)
10PKa-pH
% Drug ionized = x 100 (7.5) Drug pKa : The significance of pH dependent excretion for any
1 + 10PKa-pH particular compound is greatly dependent upon its pK~ and lipid solubil-
ity. A characteristic of d(ugs, pKa values govern the degree of ionization
The concen~ati?n ratio R of the drug in urine to that in plasma (U:P)
at a particular pH. A polar and ionized drug will be poorly reabsorbed
can be given by equations derived by Shore et a/:
passively and excreted rapidly (see Table ?.1 ). Reabsorption is also
for weak acids, Ra =
u --
1 + 1opHurine-pKa
(7.6)
affected by the lipid solubility of drug; an ionized but lipophilic drug will
p 1 + 1opHplasma-pKa be reabsorbed while an unionized but polar one will be excreted.
The combined effect of urine pH and drug pKa and lipid solubility on
u
'
1 + 1OPKa-pHurine
foi:/weak bases, Rb = -- (7.7) reab~orption of drugs is summqtized as follows:
p 1 + 1OPKa-pHplasma
1. An acidic drug such as penicillin or a basic drug such as gentamicin
-Note : The above equations from 7.2 to 7.7 are identical to equations which is polar in its unionized form, is not reabsorbed passively, irrespec-
2.10 to 2.15 of chapter 2. tive of the extent of ionization in urine. Excretion of such drugs is
independent of pH of urine and its flow rate. Their rate of excretion is
The relationship between drug pKa, urine pH, degree of ionization and
the sum of rate of filtration and rate of active secretion.
renal clearance is illustrated in Table 7 .1. Table 7.1 shows percent drug - '

ionized and renal clearance values (in ml/min) of several acidic and basic 2. Very weakly acidic, nonpolar drugs (pKa > 8.0) such as phenytoin
or very weakly basic, nonpolar drugs (pKa < 6.0) such as propoxyphene I
. -184 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS
185
-
· are mostly, unionized throughout the entire range of urine pH and are excretion is independent of urine pH and are not reabsorbed, are unaf-
. ~ therefore extensively reabsorbed passively at all values of urine pH. The fected by urine flow rate. An increase in urine flow in case of such drugs
- rate of excretion of such drugs is always low and insensitive to urine pH. will only produce a more dilute urine. Only those drugs whose reabsorption
3. A ~trongly acidic drug (pK3 ~ 2.0) such as cromoglycic acid or a is pH-sensitive, for example, weak acids and weak bases, show depen-
strongly basic drug (pK3 ~ 12.0) such as guanethidine, is completely dence on urine flow rate. For such agents, reabsorption is inversely
ionized at all values of urine pH and are, therefore, not reabsorbed. Their proportional to the urinary flow. These compounds can be divided into
rate of excretion is always high and insensitive to pH of urine. two types based on their extent of reabsorption in relation to that of water:

4. Only for an acidic drug in the pK3 range 3.0 to 8.0 (e.g. several 1. Drugs which are reabsorbed to an extent equal to or greater than
NSAIDs) and for a basic drug in the pK3 range 6.0 to 12.0 (e.g. morphine the reabsorption of water e.g. phenobarbital. In such cases, the relation-
analogs, 1 tricyclic antidepressants, etc.) the extent of reabsorption is greatly ship between renal clearance and urinary excretion is linear.
dependent upon urine pH and varies from negligible to aiiDost complete; 2. Drugs which are reabsorbed to an extent lower than the reabso~tion
for example, the amount of dexamphetamine excreted in the urine varies of water e.g. theophylline and many more drugs. In these cases, the
from 3 to 55% of the administered dose as the urine pH varies from 8.0 to relationship between renal clearance and urinary excretion is convex cur-
5.0. Fig. 7.2 illustrates the influence of urine pH on drug excretion. vilinear.
In addition to modification of pH, urinary elimination of an agent can
t-- Tubule Wall ---+
DRUG ACIDIC FILTRATE PLASMA KALINE FILTRATE a18o be enhanced by forced diuresis. Forced diuresis is the increase in .
Ut ine floW induced by large fluid intake OY administration Oj mannitol Or
.. Weak Acid Negligible Ionization Significant Ionization
H+ +A- HA HA H+ +A-
other diuretics. The prfnciple can be used in an intoxicated oerson to
HA -----HA - - -
1~
remove excessive drug by promoting- its excretion and decreasing the time
pKa 3.0 - 8.0
H+ +A-
Drug Reabsorbed
t
Drug lost
for reabsorption.
Passively in Urine
Both urine pH control and forced diuresis can be used to treat toxicity
Weak Base Significant Ionization Negligible Ionization with drug overdose when:
BOH
J ,,
s+ + oH-· ·soH +--- BOH --.w BOH
·'
a-+ OH- 1. Urinary excretion is the major route for elimination of drug
pKa 6.0. - 12.0
I
1l 1 2. The drug is extensively reabsorbed passively from the renal tu-
bules
Drug lost in Urine
3. The reabsorption is sensitive to urine pH (and urine flow rate)
Fig. 7.2 Influence of urinary pH on excretion of weakly acidic and weakly
Apart from the foregoing discussion on the passive reabsorption of
basic drugs. Bold -arrows indicate that the process is predominant.
drugs, the process is also important in the reabsorption of low threshold
The toxicity due to overdosage of drugs whose excretion is sensitive to substances such as urea, certain phosphates and sulfates, etc.
pH change can be treated by acidification or alkalinization of the urine
with ammonium chloride or sodium bicarbonate respectively. Thus, crystalluria
CONCEPT OF CLEARANCE
caused by precipitation of sulfonamides in the renal -tubules and subse-
quent kidney damage can be overcome by alkalinizing the urine. Excretion The clearance concept was first introduced to describe renal excretion
of basic drugs can be promoted by acidification of urine. The therapeutic of endogenous compounds in order to measure the kidney function. The
activity of the urinary antiseptic hexamine also depends on the urine pH. tet rn 'is no-" ' applied to all organs involved in drug elimination such as
It is not converted to active fonn i.e. fotrnaldehyde unless the urine is liver, lungs, the biliary system, etc. and referred to as hepatic clearance,
acidic. pulmonary clearance, biliary clearance and so on. The sum of individual
clearances by all eliminating organs is called as total body clearance or
Urine Flow Rate : In ad~ition to urine pH and drug pK 3 , the rate of total systemic clearance. It is sometimes expressed as a sum of renal
urine flow also influences the extent of reabsorption. Polar drugs whose
clearance and nonrenal clearance.
187
186 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS

The· contribution of each of the above physiologic processes in clear-


Clearance is defined as the hypothetical volume of body fluids con-
ing a drug cannot be determined by direct measurem~nt. It can howe~er
taining drug from which the drug is removed or cleared completely in a
be determined by comparing the clearance values obtatned for a drug wtth:.
specific period of time. It is expressed in ml/min and is a constant for
that of an agent such as creatinine or inulin which .is cleared by glomeru-
any given plasma drug concentration. In comparison to apparent volume
lar filtration only. The ratio of these two values is called as renal
of distribution which relates plasma drug concentration to the amount of
drug in the body, clearance relates plasma concentrati~n Jo the rate 'o f clearance ratio or excretion ratio.
drug elimination. ClR of drug
Renal Clearance Ratio = - - - - - - - (7 .12)
Elimination rate ClR of creatinine
Clearance (Cl) = - - - - - - - - - (7.9)
Plasma drug concentration Thus, depending upon whether the drug is only filtered, filtered and
Renal Clearance (CIR) : It can be defined as the volume of blood or secreted or filtered and reabsorbed, the clearance ratio will vary (Table
plasma which is completely cleared of the unchanged drug by the kidney 7 .2. ). The renal clearance values range from zero to 650 m 1/m in and the
per unit time. It is expressed mathematically as: clearance ratio from zero to five.
Rate of urinary excretion
ClR = (7.10) FACTORS AFFECTING RENAL EXCRETION
Plasma drug concentration OR RENAL CLEARANCE
Physiologically speaking, renal clearance is the ratio of "sum of rate Apart from the three physiologic processes that govern tPe urinary
of glomerular filtration and active secretion minus rate of reabsorption" excretion, other factors influencing renal clearance of drugs and metabo-
to Hplasma drug concentration C". lites are:
Rate of filtration + Rate of secretion- Rate of reabsorption 1. Physicochemical properties of the drug
(7 .II) 2. Plasma concentration of the drug
c •
3. Distribution and binding characteristics of the drug

TABLE 7.2 4. Urine pH


Relationship between Renal Clearance Values 5. Blood flow to the kidneys
and Mechanism of Clearance 6. Biolpgical factors
I 7. -Drug· inter~ctions
Renal Clearance Renal Clearance Mechanism of Renal Example(s)
(ml/min) Ratio Clearance 8. Disease states

Physicochemical Properties of the Drug


0
(least value)
0 Drug filtered and
reabsorbed con1pletely
Glucose I

'•I Important physicochemical factors affecting renal excretion o: a drug


are - molecular size, pKa and lipid solubility. The molecular wetght of a
·< 130 Above 0, Drug filtered and Lipophilic
Below 1 reabsorbed partially
drug is very critical in its urinary elimination. An agent of small molecu-
drugs
lar size can be easily filtered through the glomerulus. Compounds of
130 (GFR) 1 Drug is filtered only Creatinine, weights below 300 daltons, if water soluble, are readily excreted by the
Inulin
kidneys. Drugs in the molecular weight range 300 to 500 daltons can be
> 130 > 1 Drug ·filtered as well Polar, ionic excreted both in urine and bile. Molecules of size greater than 500
as· secreted actively drugs daltons are excreted in urine to a lesser extent.
650' 5 Clearance equal to lodopyraceJ, The influence of drug pKa on excretion has already been discussed.
(Highest value) renal plasma flow rate PAH Urinary excr.etion of an unchanged drug is inversely related to its lipophilicity.
This is because, a lipophilic drug is passively reabsorbed to ·a large extent.
189
188 BIOPHARMACEUTICS AND PHARMCOKI"J'I ETICS EXCRETION OF DRUGS

Urine concentration
Plasma Concentration of the Drug ClR = x Urine flow rate (7 .8)
Glomerular filtration and reabsorption are directly affected by plasma Plasma concentration
drug concentration since both are passive processes. A drug that is not Since only free drug can be excreted in the urine, the fraction of drug
bou~d to .plasma proteins and excreted by filtration only, shows a linear
bound to plasma proteins is important and can be computed from equation:
relattonshtp between rate of excretion and plasma drug concentration. In
~ase ~f dru~s whi.ch are s:creted or reabsorbed actively, the rate process £ =
cu (7.13)
mcreases with an Increase In plasma concentration to a point when satura- u c
tion . ~f carrier occurs. In case of actively reabsorbed drugs, excretion is where, fu = fraction of unbound drug in plasma,
ne~hgible at low plasma concentrations. Such agents are excreted in
Cu = concentration of unbound drug in plasma, and
urine only when their concentration in the glomerular filtrate exceeds the
active reabsorption capacity, e.g. glucose. With drugs that are actively C = total plasma concentration of drug.
secreted, the rate of excretion increases with increase in plasma concentra- Thus, equation 7.8 can be written as:
tion upto a saturation level. These situations are depicted in Fig. 7.3.
CIR = fu . Urine Flow Rate (7 .14)
.
I - drug excreted by
filtration only
Drugs extensively bound to proteins have long half-lives because the
renal clearance is small and urine flow rate is just 1 to 2 mVmin. The
II - drug filtered and Ill renal clearance of oxytetracycline which is 66o/o unbound is 99 mVmin
actively reabsorbed I
~ c:
0
while that of doxycycline (7% unbound) is just i 6 ml/min.
c ·-
co ~
II
~
(U
Ill - drug_!iltered as well ~ Actively secreted drugs are much less affected by protein binding, e.g.
0
-
Q)
(.)
as actively secreted ><
w penicillins. The free fraction of such drugs are filtered as well as secreted
-ns
c
Q)
----------111 o Q)
actively and dissociation of drug-protein complex occurs rapidly.
t------------1 /}_
~

a: The influence of urine pH on renal clearance has already been dis-


I II
cussed.

Blood Flow to the Kidneys


The renal blood flow is important in case of drugs excreted by glo-
merular filtration only and those that are actively secreted. In the latter
____.) Plasma Drug Concentration case, increased perfusion increases the contact of drug with the secretory
sites and enhances their elimination. Renal clearance in such instances is
'Fig. 7.3 Renal clearance and rate of excretion of a drug in relation to its
plasma concentration as affected by the physiologic processes said to be perfusion rate-limited.
- filtration , active reabsorption and active secretion
Biological Factors
Distributi~n and Binding Characteristics of the Drug Age, sex, species and strain differences, differences in the genetic
Clearance is inversely related to apparent volume of distribution of make-up, circadian rhythm, e~c. alter drug excretion. Renal excretion is
drugs. A drug with large Vd is poorly excreted in urine. Drugs restricted approximately 10°/o lower in females than in males. The renal function of
to blood compartment have higher excretion rates. newborns is 30 to 40% less in comparison to adults and attains maturity
Drugs that are bound to plasma proteins behave as macromolecules between 2.5 to 5 months of age. In old ·age, the GFR is reduced and
and thus cannot be filtered through the glomerulus. Only unbound or free tubular function is altered, the excretion of drugs is thus slowed down and
drug appear in the glomerular filtrate. An earlier equation given for renal half-life is prolonged.
clearance is:

191
190 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS

Inulin clearance provides an accurate measure of ?~R but has t~e


Drug Interactions
disadvantage of being a tedious method. Clinically, creattnme clearance ts
Any drug . interaction that results in alteration of binding characteris-
widely used to assess renal ful)ction.
tics, r~nal flood flow, active secretion, urine pH and intrinsic clearance
and forced diuresis would alter renal clearance of a drug. The renal Creatinine is an endogenous amine produced as a result of mus~~e
clearance of a drug extensively bound to plasma proteins is increased after catabolism. It is excreted unchanged in_t~e_ uri!le by ~_merular f}ltratton
displacement with another drug. An interesting example of this is gentamicin only. An advantage of this test is that it can be correlated to the s~eady­
induced nephrotoxicity by furosemide. Furosemide does not precipitate state concentration of creatinine in plasma and needs no ~o~lectton °~
this effect by its diuretic effect .but by displacing gentamicin from binding · -The method involves determination of serum creattntne levels.
urtne. d d ·f[l t
sites. The increased free antibiotic concentration accelerates its renal Since creatinine production varies with age, weight and gen er, I ere~
clearance. Acidification of urine with ammonium chloride, methionine or founulae are used to calculate creatinine clear.ance from the serum c~eatt-
ascorbic acid enhances excretion of basic drugs. Alkalinization of urine nine values.
with citrates, tartarates, bicarbonates and carbonic anhydr_ase inhibitors For Children (between 1 to 20 years),
promote excretion of acidic drugs. Phenylbutazone competes with w 0.7
0.48 H (7 .15)
·hydroxyhexamide, the active metabolite of antidiabetic agent acetohexamide, Clcr=---X
for active secretion and thus prolongs its action. Urinary excretion of Scr 70
digoxin is reduced by diazepam. All diuretics increase elimination of For Adults (above 20 years):
drugs whose renal clearance gets affected by urine flow rate.
(140- Age) W
(7 .16)
Disease States Renal Impairment Males, Clcr ==
72 X Scr
Renal dysfunction greatly impairs the elimination of drugs especially
(140 - Age) W
those that are primarily excreted by the kidneys. Some of the causes of Clcr == - - - - - - - (7 .17)
Females,
renal failure are hypertension, diabetes mellitus, hypovolemia (decreased 85 X Scr
blood supply to the kidneys), pyelonephritis (inflammation of kidney due = 0.9 . Clcr of Male
to infections, etc.), nephroallergens (e.g. nephrotoxic serun1) and nephrotoxic
· agents such as aminoglycosides, phenacetin and heavy metals such as lead where, Clcr = creatinine clearance in mllmin.
and mercury. Scr = serum creatinine in mgo/o,
Uremia, characterized by impaired glomerular filtration and . accumula- •
H = height in ems, and
.' ,
tion of fluids and protein metabolites, also impairs renal clearance of W = weight in Kg.
drugs. In both these conditions, the half-lives of drugs are increased. As
I '-
Age . is measured in years.
a consequence, drug accumulation and toxicity may result. ' Detettnination A direct method for determ.J.!tin~ creatinin~ clearan~~ is determination
of renal function is therefore important in such conditions in order to of theamount ~f creatinine excreted in urine in 24 hours. (~o calculate the
monitor the dosage regimen. rate of creatinine excretion) and the mean of serum creattnme from blood
I .
Renal function can be detennined by measuring the GFR. Both satnples taken just before and immediately after the urine collection pe-
endogenous .
and exogenous
.
substances have been used as markers to riod. Following formula is used: .
I •

measure GFR. In order to be useful as a marker, the agent should Rate of creatinine excretion
entirely get excreted in unchanged form by glomerular filtration only and Clcr = - - - - - - - - - - :.:----o;- (7 .18)
should be physiologically and pharmacologically inert. The rate at .which Serum creatinine in mg 1 0
these markers are excreted in urine reflects the GFR and changes in GFR The normal creatinine clearance value is 120 to 130 ml/min. A value
reflects renal dysfunction. Inulin (the exogenous fructose polysaccharide) of 20 to 50 ml/min denotes moderate renal failure and values below 10
and serum_f!eatinine level have been used successfully for such purposes. mllmin indicate severe renal in:tpairment.

EXCRETION OF DRUGS 191


192 BIOPHARMACEUTICS AND PHARMCOKINETICS

The renal function, RF is calculated by equation 7.19. There are two procedures for dialysis:
1. Peritoneal dialysis, and
Clcr of patient ·
RF = ---------- (7.19) 2. Hemodialysis.
Clcr of a normal person
In the fQrn1er, the semipermeable membrane is the natural membrane
Dose Adjustment in Renal Failure . of the peritoneal cavity. The method involves introduction of the dialy-
Generally speaking, drugs in patients with renal impairtnent have al- sate fluid into the abdotnen by inserting the catheter and draining and ·
tered phannacokinetic profile. Their renal clearance and elimination rate discarding the same after a certain period of time. In hemodialys!s, lhe
are _reduced, the elimination half-life is i~~reased and th~~pp~rent volum(. semipermeable membrane is an artificial membrane. Since the system is
of distribution is altered. .Thus, dose must be altered depending upon th~ · outside the body, it is also called as extracorporeal dialysis. The equip-
renal function in such patients. However, except for drugs having low ment is referred to as artificial kidney or hemodialyzer. Apart from the
therapeutic indices, the therapeutic range of others is sufficiently large and rem ova] of toxic waste from the body, hemodialysis is also useful in the
dosage adjustment is not essential. Dosage regimen need not be changed treatment of overdose or poisoning situations where rapid removal of drug
when the fraction of drug excreted unchanged, fu is <0.3 and the renal becomes necessary to save the life of the patient. Patients of kidney
function RF is >0. 7 of nonnal. This generalization is based on the failure require dialysis of blood every 2 days. Each treatment period lasts
assumption that the metabolites are inactive and binding characteristics for 3 to 4 hours.
and drug availability are unaltered and so is the renal function in kidney Factors that govern the removal of substances by hemodialysis are:
failure conditions. When the fu value approaches unity and RF ap-
Water Solubility : Only water soluble substances are dialyzed; lipid
proaches z~ro, elimination is extremely slowed down and dosing should
soluble drugs such as glutethimide cannot be removed by dialysis.
be reduced drastically. The significance of nonrenal clearance increases
in such conditions. Molecular Weight : Molecules with size less than 500 daltons are
dialyzed easily, e.g. many unbound drugs; drugs having large molecular
The required dose in patients with renal impainnent can be calculated
weight such as vancomycin cannot be dialyzed.
·by the simple fonnula:
Protein Binding : Drugs bound to plasma proteins or blood cells
Nonnal dose x RF (7.20)
cannot be dialyzed since dialysis is a passive diffusion process.
The dosing interval in hours can be · computed from the . following Volume of Distribution : Drugs with large volume of distribution are
equation: - extensively distributed throughout the body and therefore ·less easily re-
Noonal interval (in hours) moved by dialysis, e.g. digoxin.
(7.21)
RF The Fig. 7.4 shows schematic representation of hemodialysis.

-
,
When the drug is eliminated both by renal and nonrenal mechanisms, -+.
"'
Blood •

the dose to be administered in patients with renal failure is obtained ·f rom


equation 7.22. -

••
Nornlal dose RF x Fra~tion .e xcreted + Fraction eliminated
• •
In urine nonrenally (7.22)
--••
••
'

Sem ipermeable
---

:

Membrane

Dialysis and Hem.operfusion -••


In severe renal failure, the patients are put on dialysis to remove toxic
.
waste products and drugs and their metabolites which accumulate in the ~~ . _ Dialysate
body.
I
• •
\... I

.
Dialysis _is a process in which easily diffusible substances are ·sepa- Fig. 7.4 Diagrammatic representation of a hemodialyzer.
The blood and the dialysate flow counter.":currently .
rated from poorly diffusible ones by the use of semipermeable membrane.


194 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS 195
Th·e dialyzing fluid contains sodium, potassium, calcium, chloride and Biliary Excretion of Drugs Enterohepatic Cycling
1

acetate ions, and dextrose and other constituents in the same concentration
The hepatic cells lining the bile canaliculi produce bile. The produc-
as· that in plasma. The unwanted metabolites in the patient's blood such
tion and secretion of bile are active processes. The bile secreted from
as ure~, uric acjd, creatinine, etc. diffuse into the dialysate until equilib-
liver, after storage in the gall bladder. is secreted i~ the ~uo_de!lum. In
rium. Since the volume of dialysate is n1uch greater than that of blood humans the bile tlow rate is a steady 0.5 to 1 mllmm. B1le 1s unportant
~d since it is -replenished with fresh fluid from time to time, almost in the d,ioestion and absorption of fats. Almost 90% of the secreted bile
0
complete removal of unwanted substance~ from the blood is possible. acids are reabsorbed from the intestine and transported back to the liver
Drugs which can be removed by hemodialysis are barbiturates, for resecretion. The rest is excreted in feces.
· aminoglycosides, chloral hydrate, lithium, etc.
Being an active process, bile secretion is capacity-limited and subject
The rate at which a drug is removed by the dialyzer depends upon the to saturation. The process is exactly analogous to active renal secretion.
flow rate of blood to the machine and its perfonnance. The tenn dialysance,
Different transport mechanisms exist for the secretion of organic anions,
also called as dialysis clearance, is used to express the ability of machine
cations and neutral polar compounds. A drug whose biliary concentration
to clear the drug from blood. It is defined in a manner similar to
is less than that in plasma, has a small biliary clearance and vice versa. In
clearance by equation:
some instances, the bile to plasma concentration ratio of drug can ap-
proach 1000 jp. which cases, the biliary clearance can be as high as 500
(7.23)
mllmin or more.
I

where, Cld = dialysance or dialysis clearance Compounds that are excreted in bile have been classified into 3 cat-
Q = blood flow rate to dialyzer egories on the basis of their bile/plasma concentration ratios:
. .
Cin = concentration of drug in blood entering .the dialyzer Group A compounds whose ratio is approximately I, e.g. sodium,
Cout = concentration of drug in blood leaving the dialyzer potassium and chloride ions and glucose.
Group B compounds whose ratio is > 1, usually from I 0 to 1OOQ, e.g.
In hemoperfusion, the blood is passed through a bed of adsorbent
bile salts, bilirubin glucuronide, creatinine, sulfobromophthalein conju-
such as charcoal or resin; as a result, drugs and other unwanted molecules '
gates, etc.
are adsorbed while plasma proteins are not. I~ is also useful in treating
severe drug intoxication. The limitation of hemoperfusion is that it also Group C compounds with ratio < I, e.g. sucrose, inulin, phosphates,
· removes the blood platelets, white cells and endogenous steroids. phospholipids and mucoproteins.
.'\
Drugs can fall in any of the· above three categortes.
NON-RENAL ROUTES OF DRUG EXCRETION Several factors influence secretion of drugs in bile:
Drugs and their metabolites may also be excreted by routes other than
1. Physicochemical Properties of the Drug
the renal route, called as the extrarenal or nonrenal routes of drug
···-excretion. The various such excretion processes are: The most important factor governing the excretion of drugs in bile is
their molecular weight. Its influence on biliary excretion is summarized
1. Biliary excretion
i
in the Table 7.3.
2. .Pulmonary excretion
Polarity is the other physicochemical property of drug influencin,g
·3··/r Sal,~vary excretion
1 •; ' i I' biliary excretion. Greater the polarity, better the excretion. Thus, me-
4.'' Mammary excretion tabolites are more excreted in bile than the parent drugs because of their
5. 's kin/dettnal excretion increased polarity. The molecular weight threshold for biliary excretion
1 6. Gastrointestinal excretion of drugs is also dependent upon its pnlarity. A threshold of 300 daltons
.-
7. Genital' excretion and greater than 300 daltons is necessary for organic -caHons (e.g. quater-
naries) and organic anions respectively. Non ionic compounds should also
be highly polar for biliary excretion, e.g. cardiac glycosides.
I

196 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRNGS 197


TABLE 7.3 such as phenobarbital stim.ulate biliary excretion of drugs, firstly, by
Influence of Molecular Weight on Excretion Behavior of Drugs enhancing the rate of glucuronidation, and secondly, by promoting bile
Molecular Weight of Excretion Pattern flow. The route of drug administration also influences biliary drug excre-
Drug/Metabolite tion. Orally administered drugs which during absorption process go to the
- '
liver, are excreted more in bile in comparison to parenterally administered
Below 300 daltons Excreted mainly in urine; less than 5% is drugs. Food also has a direct influence on biliary excretion of drugs.
excreted in bile
Protein and fat rich food increase bile flow.
Above 500 daltons Excreted mainly in bile; less than 5% is
The efficacy of drug excretion by the biliary system and hepatic
excreted in urine
function can be tested by an agent that is exclusively and completely
Between 300 to Excreted both in urine and in bile; a decrease
eliminated unchanged in the bile, e.g. sulfobromophthafein. This mar~er
500 daltons in one excretory route is compensated by
is excreted within half an hour in the intestine when the hepatic function
excretion through the other route
is nonnal. A delay in its excretion is indicative of hepatic and biliary
malfunction. The marker is also useful in detettnining hepatic blood flow
2. Nature of Biotransformation Process
rate.
A metabolic reaction that greatly increases the polarity as well as the
The ability of liver to excrete the drug in the bile is expressed by
molecular weight of drug favors biliary excretion of the metabolite. Thus,
biliary clearance (equation 7 .24).
phase II reactions, mainly glucuronidation and conjugation with glutathio-
ne, result in metabolites with increased tendency for biliary excretion Biliary excretion rate
Biliary clearance = - - -- - .---- - - - - (7.24a)
(increase the molecular weight by 176 and 300 dalto~s _respectively). Plasma drug concentration
Examples of drugs excreted in the bile as glucuronides are morphinf . ~
chh)ramphenicol and indomethacin. Stilbestrol glucuronide is almost en 1 Bile flow x Biliary drug concentration •
(7.24b)
tirely excreted in bile. Glutathione conjugates are exclusively excreted via Plasma drug concentration
bile and are not observable in the urine because of their large molecular
Just as the major portion of bile salts excreted in intestine is reab-
size. Conjugation with amino acids and acetylation and methylation
sorbed, several drugs which are excreted unchanged in bile are also
reactions do not result in metabolites with greatly increased molecular
absorbed back into the circulation. Some drugs which are excreted as
weight and therefore have little influence on biliary excretion of xenobiotics.
glucuronides or as glutathione conjugates are hydrolyzed by the intestinal
For a drug to be excreted unchanged in the bile, it must have a highly
or bacterial enzymes to the parent drugs which are then reabsorbed. The
polar functional group such as -COOH (cromoglycic acid), -S03H (ama-
reabsorbed drugs are again carried to the liver for resecretion via bile
ranth), -NH4+ (oxyphenonium), etc. Clomiphene citrate, an ovulation
inducer, is -almost completely removed from the body via biliary excretion. Oral
administration ~~-BILE ......................._ Urinary excretion
3. Other Factors Metabolites
Biliary and/or conjugates Distribution
Miscellaneous factors influencing biliary excretion of drugs include excretion to tissues
1
sex and species differences, protein-drug binding, disease states, drug
ENTEROHEPATIC
' interactions, etc. SMALL
CIRCULATION
-· INTESTINE . LIVER
OF DRUGS
Substances having high molecular weight show good excretion in bile
in case of rats, dogs, and hen and poor excretion in rabbits, guinea pigs Hydrolysis of •

conjugate and
and monkeys. The· route is more important for the excretion of drugs in
absorption
laboratory animals than in man. Protein bound drugs can also be excreted
Fecal
in the bile since the secretion is an active process. In cholestasis, the bile excretion
flow rate is reduced thereby decreasing biliary excretion of drugs . Agents
I
Fig. 7.5 Enterohepatic cycling of drugs
198 BIOPHARMACEUTICS AND PHARMCOKINETICS
199
EXCRETION OF DRUGS
into the intestine. This phenomenon of drug cycling between the intestine ,Salivary Excretion
and the liver is called as enterohepatic cycling or .e nterohepatic circula-
Excretion of drugs in saliva is also a passive diffusion process and
tiob. . of drugs (Fig. 7.5.).
'
therefore predictable on the basis of pH-partition hypothesis. The pH of
Such a recycling process continues until the drug is biotransformed in saliva varies from 5.8 to 8.4. The mean salivary pH in man is 6.4.
the liver or is excreted in the urine or both. The drugs which are secreted Unionized, lipid soluble drugs at this pH are ·excreted passively in the i
, via bile in the intestine but not reabsorbed, are fmally excreted in the saliva. Equations analogous to 7.3, 7.5, 7.6 and 7.7 can be written for (
feces. drugs with known pK3 at the salivary pH ·and percent ionization and
Enterohepatic circulation is important in the conservation of important saliva/plasma drug concentration ratio (SIP) can be computed.
endogenous substances such as vitamin B12, vitamin D3, folic acid, sev-
for weak acids,
eral steroid hottno~es and bile salts. The process results in prolongation
of half-lives of several drugs (e.g. carbenoxolone) which are extensively s 1 + 1Q(pHsaliva-pKa) fplasma
(7.25).
Ra=- --------X---
excreted in bile. The half-life of agents such as DDT, which are resistant p 1 + 1Q(pHplasma-pKa) fsaliva \

to biottansfonnation and are highly lipophilic, m~y increase to several


Jays due to such a recycling phenomenon. The prolonged therapeutic for weak bases,
activity of oral contraceptives (upto 12 hours) is also due to such a S 1 + 1Q(PKa-pHsaliva) £
Rb = __________ X plasma (7.26)
recirculation. .Other examples of drugs undergoing enterohepatic circula-
p 1 + 1Q(PKa-pHplast;na) fsaliva
tion are cardiac glycosides, rifampicin, chlorpromazine and indomethacin.
Drug interactions affecting enterohepatic cycling occur when agents such where, fplasma and fsaliva are ·free drug fractions in plasma and in saliv~
as antibiotics kill the intestinal microflora and thus retard hydrolysis of .-espectively.
drug conjugates and their subsequent reabsorption, or the unabsorbable The SIP ratios have been found to be less than 1 for weak acids arid
ion exchange r~sins such as cholestyramine which bind strongly to the. greater than 1 for weak bases i.e. basic drugs are excreted more in saliva
acidic and neutral drugs (e.g. digitoxin) and thus prevent their reabsorption. as compared to acidic drugs. The salivary concentration of some drugs
The principle of adsorption onto the resins in the GIT can however be reaches as high as 0.1 °/o. Since the SIP ratio is fairly constant for several
used to treat pesticide poisoning by promoting their fecal excretion. drugs, their blood concentration can be determined by detecting the amount
Biliary excretion of drugs can be assessed by giving the drugs paren- of dru~~ excreted in saliva, e.g. caffeine, theophylline, phenytoin;
terally and detecting their pre~ence in feces. This also rules out the doubt carbama.~epine, etc. Some drugs are actively secreted in saliva, e.g.
about the incomplete absorption of such drugs when given orally and lithittrrJ, the concentration of which is sometimes 2 to 3 times that _in
observed in feces. plasma. Penicillin and phenytoin are also actively secreted in saliva. . .
Oral -_--.,.............._
Pulmonary Excretion
administration •.

Gaseous and volatile .substances such as the general anesthetics (e.g.


halothane) are absorbed through the lungs by simple diffusion. Similarly,
Distribution
their excretion by diffusion into the expired air is possible. Factors · GIT ...-.. and
influencing pulmonary excretion of a drug include pulmonary blood flow, elimihation
rate of respiration, solubilitY of the volatile substance, etc. Gaseous
anesthetics such as nitrous oxide which are not very soluble in blood are
excreted rapidly. Generally intact gaseous drugs are excreted but metabo- Fecal BLOOD
lites are not. Compounds like a~.cohol which have high solubility in blood excretion
and tissues are excreted slowly by the lungs. The principle involved in Fig. 7.6 Salivary cycling of drugs -
the pulmonary excretion of benzene and halobenzenes is analogous to that
of steam distillation. The bitter after taste in the mouth of a patient on medication is an
-
indication of drug excretion in saliva. In few instances, the process is ··,
I

'
!
201
200 BIOPHARMACEUTICS AND PHARMCOKINETICS EXCRETION OF DRUGS

responsible for side effects such as black hairy tongue in patients receiv- hypersensitivity reactions. Compounds such as benzoic acid, salicylic
ing antibiotics, gingival hyperplasia due to phenytoin, etc. Some basic acid, alcohol and antipyrine and heavy metals like lead, mercury and
drugs inhibit saliva secretion and are responsible for dryn~ss of mouth. arsenic are excreted in sweat.
Drugs excreted in saliva can undergo cycling in a fashion similar to Gastrointestinal Excretion
enterohepatic ,c ycling, e.g. sulfonamides, antibiotics, clonidine, etc. (Fig. Excretion of drugs into the GIT usually occurs after parenteral admin-
7 .6.). istration when the concentration gradient for passive diffusion is favorable.
Mammary Ex~retion The process is reverse of GI absorption of drugs. Water soluble and
ionized form of weakly acidic and basic drugs are excreted in the G IT,
Excretion of a drug in milk is important since it can gain entry into
e.g. nicotine and quinine are excreted in stomach. Orally administered
the breas~ feeding infant.
drugs can also be absorbed and excreted in the GIT. Drugs excreted in
Milk consists of lactic secretions originating from the extracellular the GIT are reabsorbed into the systemic circulation and undergo recy-
fluid and is rich in fats and proteins. About 0.5 to 1 liter/day of milk is
cling.
secreted in lactating mothers
Excretion of drugs in milk is a passive process and is dependent upon Genital Excretion
pH-partition behavior, molecular weight, lipid solubility and degree of Reproductive tract and genital secretions may contain the excreted
ionization. The pH of milk varies from 6.4 to 7.6 with a mean pH of 7.0. drugs. Some drugs have been detected in semen.
Free, unionized, lipid soluble drugs diffuse into the mammary alveolar Drugs can also get excreted via the lacrymal fluid.
cells passively. The extent of drug excretion in milk can be detern1ined A summary of drugs excreted by various routes is given in Table 7 .4.
from milk/plasma drug concentration ratio (M/P). Since milk is acidic in
comparison to plasma, as in the case of saliva, weakly basic drugs con-
TABLE 7.4
centrate more in milk and have M/P ratio greater than 1. The opposite is
' Excretion Pathways, Transport Mechanisms and Drugs Excreted
true for weakly acidic drugs. It has been shown that for acidic drugs,
excretion in milk is inversely related to the molecular weight and partition Excretory Mechanism Drugs Excreted
coefficient and that for basic drugs, is inversely related to the degree of Route
ionization and partition coefficient. Drugs extensively bound to plasma
Urine glomerular filtration, free , hydrophilic~ unchanged
· proteins, e.g. diazepam, are less secreted in milk. Since milk contains •
active secretion, drugs/metabolites/conjugates
proteins, drugs excreted in milk can bind to .it. The amount of drug of MW < 500
active/passive reabsorption
excreted in milk is generally less than 1% and the fraction consumed by
Bile active secretion hydrophilic, unchanged drugs/
the infant is too less to reach therapeutic or toxic levels. But some potent
metabolites/conjugates of
drugs such as barbiturates, morphine and ergotamine may induce toxicity
MW ~ 500
in infants. Discoloration of teeth with tetracycline and jaundice due to
interaction of bilirubin with sulfonamides are examples of adverse effects Lung passive diffusion gaseous and volatile~ blood
precipitated due to drug excretion in the milk. Nicotine is also secreted in and tissue insoluble drugs
/

the milk of mothers who smoke. Thus, wherever possible, nursing moth- Saliva passive diffusion, free , unionized, lipophilic
ers should avoid drugs and smoking. If medication is unavoidable, the active transport drugs, some polar drug"
·- infant should be bottle fed.
Milk passive diffusion free, unionized, lipophilic drugs
(generally basic)
Skin Excretion
Sweat passive diffusion free, unionized. lipophilic drugs
Drugs excreted through the skin via sweat also follow pH-partition
hypothesis. Passive excretion of drugs and their metabolites through skin Intestine passive diffusion water soluble, ionized drugs
is resp_onsible to some extent for the urticaria and dennatitis and other
EXCRETION OF DRUGS
203
202 . BIOPHARMACEUTICS AND PHARMCOKINETICS
21. What factors determine the pulmonary excretion of drugs?
QUESTIONS
22. Why should a nursing mother refrain from smoking and taking medication?
1. What physicochemical properties of a drug/metabolite govern its excretion . '

in urine? 23. What is the reason for bitter after taste of medicaments in a ' patient on drug
2. Name the principal processes involved in urinary excretion of drugs. How therapy?
do the excretion rate values correlate with the contribution of each of these 24. What is the cause of hypersensitivity skin reactions with some drugs?
processes in drug excretion? Quote with examples the markers used to 25. The normal dose of a drug is 200 mg. If the fraction excreted unchanged
estimate these parameters. in - urine is 0.75, what would be the dose for a patient whose creatinine
3. Secretion of an exogenous compound in tubules is an active process but clearance is 13 ml/min? Calculate the new dosing interval if the normal
reabsorption is generally a passive phenomenon. Explain? dosing frequency is every two hours.
4. Unlike glomerular filtration, active secretion of a drug is unaffected by Answer : Dose = 65 mg; Dosing interval = 20 hours.
protein binding. Explain. . 26. Estimate the creatinine clearance of a 30 years old, 70 Kg man with serum
. 5. How can the principle of competitive inhibition of tubular secretion be put creatinine value 2.0 mg%. What is the renal function value of such a
1

to therapeutic use? • patient?


6. How is the driving force for passive reabsorption of drugs from tubules Answer : Clcr = 53.5 ml/min; RF = 0.4.
established? 27. The pKa of an acidic drug is 6.0. Determine the % drug ionized in plasma
7. Discuss the factors influencing passive reabsorption of drugs from tubules. pH 7.4 and urine pH 6.0. Calculate the UIP ratio and compare it with the
value obtained when urine pH is 7.4. At what pH more of such a drug is
8. Based on the extent of ionization or pKa of a drug, reabsorption of which
drugs is affected and which remain unaffected by a change in urine pH? expected to be excreted in urine?
Answer : % ionized at pH 7.4 = 96.2; % ionized at pH 6.0 = 50; U/P when
I
9. How can the principle of urine pH control and forced diuresis be utilized to
urine pH 6.0 = 0.076; U/P ratio when urine pH 7.4 = 1.0; the drug will be
treat drug intoxication? To which drugs is such an approach applicable?
excreted more when urine pH is 7 .4.
10. Define clearance and renal clearance ratio of a jrug. Based on the renal
28. Calculate the dialysis clear~nce if the blood flow rate to the dialyzer is 50
clearance ratio values, how can one estimate the mechanism of renal clear-
ml/min and concentration of drug entering and leaving the dialyzer is 100
ance of a drug?
and 20 mcg/ml respectively.
11. List the factors influencing renal excretion of drugs.
Answer : Dialysis clearance = 40 ml/min.
12. What criteria are necessary for an agent to be useful as a marker to measure
29. Given in the table below are four fluoroquinolone antibiotics alongwith their
kidney function? Why is creatinine clearance preferred over inulin clear-
fractions excreted unchanged in urine and the renal function of the patient to
ance in estimating renal function?
whom they are to be administered. Rank the situations from most important
13. Based on fraction of drug excreted unchanged and renal excretion values, in to least important for considering a change in dosage regimen and name the
what situations does dose adjustment becomes necessary in renal failure? situation for which you recommend a change in usual dosage regimen.
What assumptions are made when adjusting a dose in renal failure patient? Assume that all drugs are administered in same doses of 1000 mg/day, have
14. What factors govern removal of a substance by hemodialysis? similar therapeutic indices and inactive metabolites. (Hint: Use equation
15. What are the various nonrenal routes of drug excretion? 7.22).
16. Discuss the factors influencing biliary excretion of drugs. Situation Drug fu RF

17. How are drugs excreted in bile classified on the basis of bile/plasma ratio? A Pefloxacin 0.15 0.1
18. Metabolites and conjugated forms are excreted more in bile than the parent Ciprofloxacin 0.40 0.3
B
drugs. Why'! 0.70 0.5
. c Norfloxacin
19. How can the efficiency of biliary system be tested? 0.80 0.7
D Ofloxacin
20. Explain enterohepatic circulation of drugs. What is the significance of such
a cycling? How can interaction at the cycling level be used therapeutically Answer : B ":;:>: C > D > A.
to treat pesticide poisoning?
' '
205
PHARMACOKINETIC DRUG INTERACTIONS

1. Antagonism : The interacting drugs have opposing actions, e.g.


acetyl choline and noradrenaline have opposing effects on heart
rate.
2. Addition or Summation : The interacting drugs have similar
8 actions and the resultant effect is the sum of individual drug
responses, e.g. CNS depressants like sedatives, hypnotics, etc.

PharmaCokinetic 3. Synergism or Potentiation : It is enhancement of ~ction. o.f one


drug by another, e.g. alcohol enhances the analgesic activity of
Drug Interactions •
asprrtn.

Indirect pharmacodynamic interactions are situations in which bo~h


the object and the precipitant drugs have unrelated effects but the latter zn
Drug interactions are said to occur when the pharmacologic activity some way alters the effects of the former -for example, salicylates decr~as.e
of a drug is altered by the concomitant use of another drug or by the the ability of the platelets to aggregate thus impairing the hemostasts tf
presence of food , drink or environmental chemicals. The drug whose warfarin induced bleeding occurs.
activity is affected by such an interaction is called as the object drug and The resultant effect of all pharmacodynamic interactions is thus altered
the agent which precipitates such an interaction is referred to as the drug action without a change in plasma concentration.
precipitant. Drug interactions are generally quantitative whereby the
intensity of effect is changed (for example, an increase or a decrease) and
seldom qualitative. Most interactions are specific types of adverse reac- Drug/food alters
absorption of
tions with altered efficacy of the drug, for example an enhanced phannacologic GIT another drug
activity (e.g. hemorrhagic tendency of wa~farin when phenylbutazone is
given subsequently) or a decrease in the therapeutic activity resulting in
loss of efficacy like that of tetracycline when concomitantly administered
with food , antacids or mineral supplements containing heavy metal ions. EXCRETORY SYSTEM
BLOOD
In some instances, drug interactions may be beneficial for example,
enhancement of activity of penicillins when administered with probenecid. Protein-drug
• binding:
Mechanisms of Drug Interactions DRUG competition for
The two major mechanisms by which an interaction can develop are binding sites
INTERACTIONS
- pharmacokinetic and phannacodynamic.
Pharmacokinetic interactions are t!·ose in ~~hich the absorption, dis-
tribution, metabolism and/or excretion (i. e. ADM£) o.f the object drug are .Induction/inhibition
Modified drug of drug metabolizing
altered by the precipitant. The resultant effect is altered plasma concen- excretion pattern LIVER
tration of the object drug. enzymes

Pharmacodynamic interactions are those in M'hich the activity of the


object drug at its site of action is altered by the precipitant. Such
interactions may be ·direct or indirect. A direct pharmacodynamic inter- Fig. 8.1 Major sites of pharmacokinetic interactions
action is· the one in which drugs having similar or opposing pharn1acologic Apart from the phannacokinetic and phannacodynamic interactions,
effects are used c oncurrent~v. The three consequences of direct interac-
the third type of interaction is pharmaceutical interaction. It is a
tions are:

204
206 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETIC DRUG INTERACTIONS 207
physicochemical interaction that occurs when drugs are mixed in intrave- other. Competitive displacement which results when two drug~ a~e ca-
nous infusions causing precipitation or inactivation of the active principles. pable of binding to the same site on ~he prot.ein, causes the ~ost significa~t
Such interactions are better expressed by the tenn incompatibility e.g. interactions. Greater risk of interactions exists when the displaced drug Is
ampicillin, chlorpromazine and barbiturates interact with dextran in solu- highly protein bound (more than 95%), has a s~all volu~e of distribut~on
tions and are broken down or fortn chemical complexes. and has a narrow therapeutic index (e.g. tolbutanude, warfarm and phenytom),
Of the various types of interactions, the pharmacokinetic interactions and when the displacer drug has a higher degree of affinity than the drug
are most common and often result in differences in ·phannacologic effects. to be displaced. In such situations, displacement of even a small percent
Several examples of such interactions are known but few are clinically of drug results in a tremendous increase in the free fonn of the drug
significant. Clinically important effects are precipitated by drugs having which precipitates increased therapeutic or toxic effects.
low therapeutic indices, e.g. digoxin or those having poorly defined thera- Drugs may also be displaced from binding sites in tissues. An inter-
peutic end-points, e.g. antipsychotics. esting example of this are oral hypoglycemics such as the s.ulfonyl ure~s
All •phannacokinetic interactions result due to alteration in the rate of (tolbutamide, glibenclamide, etc.). ~ese. a~ents. exe~ therr therapeutic
absorption, distribution, metabolism or excretion of drugs (and therefore effects by displacing insulin from protem bmdmg sites In pancreas, plasma
also called as ADME interactions) (see Fig.8.1 ). Such a change is and other regions resulting in its elevated levels.
reflected in the altered duration and intensity of phannacologic action of Interactions Affecting Metabolism of Drugs
the drug due to variation in the plasma concentration precipitated by
The most important and the most common cause of phannacokine~ic
altered ADME. All factors which influence the ADME of a drug affect
interactions is alteration in the rate of biotransformation of drugs. MaJor
its phannacokinetics. The same has already been dealt with in sufficient
problems arise when one drug either induces .or inhibits ~he metabolism of
details in the respective chapters. A summary of some of the important
phannacokinetic interactions is given in Table 8.1. another drug. Even the environmental chemicals can brmg about such an
effect. The influence of enzyme inducers and inhibitors become more
Interactions Affecting Absorption of Drugs pronounced when drugs susceptible to first-pass hepatic meta~olism are
Altered absorption after oral administration is very common. The given concurrently. The metabolic pathway usually affec~e~ IS phase I
interaction may result in a change in the rate of absorption (an increase or oxidation. Enzyme inducers reduce the blood level and clmical efficacy
a decrease), a change in the amount of drug absorbed (an increase or a of co-administered drugs ·but may also enhance the toxicity of drugs
decrease) or both. Several mechanisms may be involved in the alteration having active metabolites. In contrast to enzyme induc~ion which is
usually not hazardous, enzyme inhibition leads to accumulation of drug to
of drug a~sorption from the GIT. In general, drugs that are not absorbed
completely/rapidly are more susceptible to changes in GI absorption. A toxic levels and serious adverse effects may be precipitated.
decrease in the rate of absorption is clinically significant in acute condi- _ Interactions Affecting Excretion of Drugs ,
tions such as pain where the drug is administered in a single dose but is
Cli~igtlly significant renal excretion interactions occur .w~en an ~f>pre­
of little importance for drugs used in chronic therapy.
ciable amount of drug or its active metabolite(s) are elnnmated m the
An alteration in parenteral drug absorption is rare but can occur when urine. Excretion pattern can be affected by alteration in GFR, renal blood
an adrenergic agent such as adrenaline or a cholinergic drug such as flow, passive tubular reabsorption, active tubular secretion and urine pH.
·methacholine is extravascularly injected concomitantly with another drug. An interesting pharmacokinetic interaction that results due ~o. the
'

The systemic absorption of the latter is altered due to vasoconstriction or pharmacodynamic drug effect is between thiazide diuretics and ht~um.
vasodilation by these agents.
Owing to the influence of former on the renal ~ub~lar. trans~o:t of sodium,
the lithium ions are retained in ~e body resulting m Its toxicity.
Interactions Affecting· Distribution of Drugs ·
Biliary excretion, the other major mechanism of drug excretion, is
Though several factors govern the distribution of drugs to various
altered by agents that inhibit biliary transport or modify bile flow rate.
tissues, clinically significant interactions result due to competition between
drugs for binding to proteins/tissues and displacement of one drug by the
208 BIOPHARMACEUTICS AND PHARMACOKINETICS
PHARMACOKINETIC DRUG INTERACTIONS 209
TABLE 8.1
List of Some of the Important Pharmacokinetic (ADME) Interactions Object Drug(s) Precipitant Drug(s) Influence on Object Drug(s)
Object Drug(s) Precipitant Drug(s) Influence on Object Drug(s) oral contra- antibiotics decreased reabsorption of
ceptives (ampicillin) drugs secreted as conjugates
ABSORPTION INTERACTIONS via bile in the intestine
1. Complexation and Adsorption 6. Malabsorption Syndrome
tetracycline, antacids, food and mineral formation of poorly soluble vitamin A, B12, neomycin

inhibition of absorption due
penicillamine supplements containing AI, and unabsorbable complex digoxin
'
(and colchicine) to malabsorption/steatorrhea
Mg, Fe, Zn, Bi and Ca ions with such heavy metal ions caused by neomycin
ciprofloxacin, antacids containing AI, Mg reduced absorption due to
norfloxacin and Ca and sucralfate complexation with metal ions DISTRIBUTION INTERACTIONS
cephalexin, sul- cho lestyram ine reduced absorption due to (resulting from altered P-D binding)
famethoxazole, adsorption and binding 1. Competitive Displacement Interactions
trimethoprim,
warfarin and Displaced drug(s) Displacer(s)
'
thyroxine anticoagulants phenylbutazone, increased clotting time;
(warfarin) chloral hydrate, increased risk of hemor-
2. Alteration of GI pH sal icy lates rhage '
sulfonamides, antacids enhanced dissolution and
tolbutamide sulfonamides increased hypoglycemic
asp1nn absorption rate
(long acting) effect
ferrous sulfate
. sodium bicarbonate, decreased dissolution and increased methotrexate
methotrexate sulfonamides,
calcium carbonate hence absorption toxicity
sal icy 1ic acid
ketoconazo le, antacids decreased dissolution and phenytoin toxicity
phenytoin valproic acid
tetracycline, bioavailability
atenolol
METABOLISM INTERACTIONS
3. Alteration of Gut ~~tility
1. Enzyme Induction
aspirin, diazepam. metoclopram ide rapid gastric emptying;
levodopa, lithium increased rate of absorption corticosteroids, barbiturates decreased plasma levels; ,
carbonate, para- oral contraceptives, decreased efficacy of
cetamol, mexiletine coumarins, pheny- object drugs
toin, tolbutamide,
levodopa, lithium anticholinergics delayed gastric emptying; tricyclics
carbonate, (atropine, homatropine) decreased rate of absorption
mexiletine corticosteroids, phenytoin -do-
oral contraceptives
4. Inhibition of GI Enzymes (see metabolism interactions) theophylline,
cyclosporin
5. Alteration of GI Microflora
oral contraceptives, rifampicin -do-
digoxin antibiotics increased bioavailability
' oral hypoglycemics,
(erythromycin, due to destruction of . /

coumanns
tetracycline) bacterial flora that inactivates
dtgoxin in lower intestine ... continued
... continued
210 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETIC DRUG INTERACTIONS
211 -· -

Object Drug(s) Precipitant Drug(s) Influence on Object Drug(s) Object Drug(s) Precipitant Drug(s) Influence on Object Drug(s).

2. Enzyme Inhibition procainamide, cimetidine (base) increased pl3;sma levels of


ranitidine basic object drugs; risk of
tyramine rich food MAO inhibitors enhanced absorption of unme-
toxicity
(cheese, lil"er, (phenelzine, tabolized tyramine; increased
yeast proqucts) pargyline, etc.) pressor _activity, potentially fatal acetohexamide phenylbutazone increased hypoglycemic effect
risk of hypertensive crisis
2. Changes in urine pH
folic acid phenytoin decreased absorption of folic
amphetamine, antacids, thiazides, increased passive reabsorp-
acid due to inhibition of an
tetracycline, acetazo Iami de tion of basic drugs; increased
enzyme responsible for its
quinidine toxicity
efficient absorption
tricyclic :anti- chlorpromazine, increased plasma half-life of 3. Changes in Renal Blood Flow
depressants haloperidol tricyclics; increased risk of lithium NSAIDs (inhibitors of decreased renal clearance of
sudden death from cardiac prostaglandin synthesis; lithium; risk of toxicity
disease in such patients the latter control renal blood
.
coumanns metronidazole, increased anticoagulant flow partially by
phenylbutazone activity; risk of hemorrhage v~soconstricti~n)

oral ~ypo­ phenylbutazone, hypoglycemia may be


glycemics sulfaphenazole, precipitated QUESTIONS
chloramphenicol
1. Define drug interactions. Explain how drug interactions are of great c.on-
alcohol disulfiram, disulfiram like reactions
metronidazole, due to increase in plasma cern in drug therapy.
tinidazole acetaldehyde levels 2. Quote examples of beneficial drug interactions.
azathioprine, xanthine oxidase incr~ased toxicity of 3. What are the two major n1echanisms by which drug-drug interactions can
mercaptopurine inhibitors (allopurinol) antineoplastics develop? Which one is the most common of the two?
alcohol, cimetidine increased blood levels of 4. Define pharmacodynamic interactions. What are the three consequences of
benzodiazepines, object drugs direct pharmacodynamic interactions?
warfarin, phenytoin, 5. What is the basic difference between pharmacodynamic and pharmacokinetic
theophylline, drug interactions?
phenobarbital 6. What type of interaction is called pharmaceutical incompatibility?
7. What are ADME interactions? Enumerate some of the major causes of each
EXCRETION INTERACTIONS
of these interactions.
1. Changes in Active Tubular Secretion
penicrllin, PAS, probenecid (acid) elevated plasma -levels of
cep~alosp'u'rin, acidic drugs; risk of toxic
nalidixic acid, reactions
methotrexate,
dapsone

__ .continued
\
'•
''

PHARMACOKINETICS : BASIC CON SIDERA TIONS 213
.
Plasma Drug Concentration-Time Profile _ .
A direct relationship exists between the concentration of drug at the
biophase (site of action) and the concentration of drug in plasma. ~
typical plasma drug concentration-time curve obtained after a single oral ·
·9 dose of a drug and showing various pharmacokinetic and pharmacodynamic
parameter-s is depicted in Fig. 9 .1 . Such a profile can be obtained by
measuring the concentration of drug in plasma samples taken at various
Pharmacokinetics : intervals of time after administration of a dosage fonn and plotting the·
••

· concentration of drug in plasma (f-axis) versus the corresponding time at


Basic Considerations which the plasma sample was collected (X-axis ).

Peak Plasma
• Concentration
\ The duration of drug therapy I1anges from a single dose of a drug - Cmax (absorption rate = elimination rate)
taken for relieving an acute condition such as headache to drugs taken --------
-1) . -
lifelong for chronic conditions such as hypertension, diabetes, asthma or ·
c: ~,..... Toxic Level
epilepsy. The freqztency of administration of a drug in a particular dose
·--....
0
c: c; ~
ts · called as dosage regimen. Depending upon the therapeutic obJective
to be attained, the duration of drug therapy and .the dosage regimen are·
-(1S

c:
~
-
__ ·-
0
0
,__
Q. ~
co - -
~
~. -
00
- - - - - 4- -
'I
--
MSC Maximum Safe
-- - -
Concentration .
c: (I) 't-
decided. 0 .0 0 ~ '
() <( '1)
Therapeutic
Ra~ional and optimal therapy with a drug depends upon: o onset en
c: ~ •
range
2 of ~ 'S, I

1. Choice of a suitable drug, and ~ action c:


- :
I
· -MEC -Minimum Effective
2. A balance between the therapeutic and the toxic effects. E
en -L---
" duration - -
of action~
- - - - -•- - - - - - - - Concentration
' -a..
(1S
I
Subtherapeutic
Both, the therapeutic and the toxic effects, depend upon the concentra- I '
€ . Level
! Area• Under the Curve 11tnin .
tion of drug at the site of action which is difficult to measure. However,
it corresponds to a specific concentration of drug in plasma which can
·t
I l
!
A' onset time
1
- /tmax
atton Phase
I 1/Y
be measured with accuracy. The drug fails to elicit a therapeutic response
. / when the concentration is below the effective level and precipitates i - -+) Time
Administration
I
' adverse rea~tions when above the toxic level. The plasma drug concentra-
of drug
. tion between these twp limits is called as the therapeutic concentration
range or therapeutic ! window (the ratio of maximum safe concentration Fig. 9.1 A typical plasma concentration-time profile showing /
to minimum effective concentration of the drug is called as the therapeu- phannacokinetic and pharmacodynamic parameters.
tic index). Thus, in order to achieve therapeutic success, plasma concen- obtained after oral administration
.
of single
'
doseof a drug.
tration of the drug ·should be maintained within the therapeutic window. The three important pharmacokinetic p3J1imeters that describe the
For this, knowledge is needed not only of the mechanisms of drug absorp- plasma le vel-time curve and useful in .-assessing the bioavailability of a
tion, distribution, metabolism and excretion, but also of the kinetics of drug from its formulation are:
these processes i.e. pharmacokinetics. Pharmacokinetics is defined as the
1. Peak Plasma Concentration (Cmax)
kinetics of drug abso!"ption, distribution, metabolism and excretion (KADME)
The point of maximum concentration of drug in plasma is called as the
and their relationship with the pharmacologic, therapeutic or toxicologic
peak and the concentration of drug at peak is kno'"n as peak plasma
response in man and animals. The applications of pharmacokinetic prin-
concentration. It is also called as peak height concentration and maxi-
ciples in the safe and effective management of individual patient is called
mum drug concentration. Cmax is expressed in mcg/mL. The peak
as clinical pharmacokinetics.

212 •

I

I

214 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETICS : BASIC CONSIDERATIONS 215

level depends upon the administered dose and rate of absorption and precipitated. Concentration of drug above MSC is said to be in the toxic
-:. elimination. The peak represents the point of time when absorption rate level.
equals elimination rate of drug. The portion of curve to the left of peak
3. Onset of Action
.~

represents absorption phase i.e. when the rate of absorption is greater The beginning of pharmacologic response is called as onset of action,
than the rate of elimination. The section of curve to the right of peak It occurs when the plasma drug concentration just exceeds the required-
generally represents elimination phase i.e. when the rate of .elimination
MEC.
exceeds rate of· absorption. Peak concentration is often related to the
intensity of pharmacologic response and should ideally be above mini- · 4. Onset Time
It is' the time required for the drug to start producing pharmacologiq
.. mum effective concentration (MEC) but less than the maximum safe
concentration (MSC) . response. It corresponds to the time for the plasma concentration to reach

MEC after administration of drug.
2. Time of Peak Concentration (tmaxl ..
The time for drug to reach peak concentration in plasma (after ex- 5. Duration of Action
travascular administration) is called as the time of peak concentration. The time period for which the plasma concentration of drug remains
It is expressed in hours and is useful in estimating the rate of absorption. above the MEC level is called as duration of drug action.
Onset time and onset of action are dependent upon tmax· The parameter is · 6. Intensity of Action
of particular importance in assessing the efficacy of drugs used to treat It is the maximum pharmacologic response produced by the peak
acute conditions like pain and insomnia which can be treated by a single plasma concentration of drug. It is also called as peak response.
dose.
7. Therapeutic Range
3. Area Under the Curve (AUC) The drug concentration between MEC and MSC represents the thera- ;
It represents the total integrated area under the plasma level-time peutic range.
profile and expresses the total amount of drug that comes into the sys-
temic circulation after its administration. AVC is expressed in mcg/mL X Rate, Rate Constants and Orders of Reactions
hours. It is the most important parameter in evaluating the bioavailability Pharmacokinetics is the mathematical analysis of processes of ADME.
of a drug from its dosage fortn as it represents the extent of absorption. The movement of drug molecules from the site of application to the
AUC is also important for drugs that are administered repetitively for the systemic circulation, through various barriers, their conversion into an-
treatment of chronic conditions like asthma or epilepsy. , other chemical form ~nd finally their exit out of the body can be expressed
mathematically by the rate at which they proceed, the order of such
The various pharmacodynamic parameters are:
processes and the rate constants.
1. Minimum Effective Concentration (MEC) The velocity with which a reaction or a process occurs is called as its
It is defined as the minimum concentration of drug in plasma required rate. Consider the following chemical reaction:
to produce the therapeutic effect. It reflects the minimum concentration
of drug at the receptor site to elicit the desired phartnacologic response. drug A ---+> drug B (9.1)
The concentration of drug below MEC is said to be in the subtherapeutic The rate of forward reaction is expressed as:
level.
-<iA
- In ~ase of antibiotics, the tettn minimum inhibitory concentration '' (9.2)
dt
(MIC) -is used. It describes the minimum concentration of antibiotic in
plasma required to kill o_r inhibit the growth of microorganisms. Neoative
b
sion
b
indicates that the concentration of drug A decreases with
time t. As the reaction proceeds, the concentration of drug B increases
2. Maximum Safe Concentration (MSC)
Also called as minimum toxic concentration (MTC), it is the con- and the rate of reaction can also be expressed as:
centration of drug in plasma above which adverse or unwanted effects are dB
(9.3)
dt
216 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETICS : BASIC CONSIDERATIONS 217
Experimentally, the rate of reaction is determined by measuring the
decrease in concentration of drug A with time t.
The manner in which the concentration of drug (or reactants) influ-
ence the rate of reaction or process is called as the order of reaction or steady drug loss
de
order of process. If C is the concentration of drug A, the rate of
decrease in C of drug A as it is changed to B can be described by a
-
dt
c
Slope =-Ko
general expression as a function of time t.
dC
i
-=-KC0 (9.4)
dt
where, K = rate constant •
~~Time
n = order of reaction
If n = 0, its a zero-order process, if n = 1, it is a frrst-order process Fig. 9.2 Graphs of zero-order kinetics (equations 9.5 and 9. 7)
and so on. The three commonly encountered rate processes in a physio-
logic system are zero-order process, frrst-order process and mixed-order Zero-Order Half-Life
process. The pharmacokinetics of most drugs can be adequately described Half-life (tYz) or half-time is defined as the time ~oeriod required for
by_zero- and frrst-order processes of which the latter are more important. the concentration of drug to decrease by one-half When t = tYi, C = C0 /2 .
and the equation 9.7 becomes:
Zero-order Kinetics (Constant Rate Processes)
Co
If n = 0, equation 9.4 becomes: - = Co - KotYi (9.8)
2
dC
- = - Ko co = -Ko (9.5) Solving 9 .8, we get:
dt
where Ko = zero-order rate constant (in mg/min)
0.5 C0
(9.9)
From equation 9.5, the zero-order process can be defi~ed as the one
whose rate is independent of the concentration of drug undergoing Equation 9. 9 shows that the ty2 of a zero-order process is not constant
reaction i.e. the rate of reac!ion cannot be increased further by increasing but proportional to the initial concentration · of. drug C 0 and invers~ly
the concentration of reactants. · •
proportional to the zero-order rate constant K 0 . Since the zero-order t y2
Rearrangement of equation 9.5 yields: changes with the decl~ne in drug concentration, it is of little practical
importance. Zero-order equations do not require ' logarithmic transfonna-
dC =-~
--.-
d! (9.6) tions.
Integration of equation 9.6 gives: Examples of zero-order processes are:
1. Metabolism/protein-drug binding/enzyme or carrier-mediated transport
C- Co= -Ko t
under saturated conditions. The rate of metabolism, binding or
or simply, C = C0 - K0 t (9.7) transport of drug remains constant as long as its concentration is
where C 0 = concentration of drug at t = 0, and in excess of saturating concentration.
C = concen~ation of drug yet to undergo rea~tion at time t. 2. Administration of a drug as a constant rate i.v. infusion.
Equation 9. 7 is that of a straight line and states that the concentration 3. Controlled drug delivery such as that from i.m. implants or os-
of reactant decreases linearly with time. .t\
plot of C versus t yi~lds such motic pumps.
a straight line having s!op~ -Ko and y-i~tei7cept C0 (Fig.9.2.). •
218
BIOPHARMACEUTICS AND PHARMACOKH TICS PHARMACOKINETICS : BASIC CONSIDERATIONS 219
First-order Kinetics (Linear Kinetics)
Kt
If n = 1, equation 9.4 becomes: log C = log C0 - --- (9 .14)
2.303
dC
- = -KC (9.1 0) or log C = log C 0 - 0.434 Kt (9.15)
dt
where K = first-order rate constant (in time-1 or per hour) A semilogarithmic plot of equation 9.14 yields a straight line with
slope = - K/2.303 and y-intercept = log C0 (Fig. 9.4.).
From equation · 9. 10, it is clear that a first-order process is the one
~hose rat~ is ~irectly proportional to the concentration of drug undergo- 100
Ing reaction z. e. greater the concentration, faster the reaction. It is Regular Plot Semilogarithmic Plot
because of such a proportionality between rate of reaction and the concen-
tration of drug that a first-order process is said to follow linear kinetics
(Fig. 9.3. ). log C K
%C Curvilinear Slope =- --
- ·--· decline in drug 2.303
concentration
---- .-L.--
I
- = -0.434 K
I I
dC ---- - +- -- - _,.__ I ) I
I t I
Yy2
- I . 1/2 .
i4
I
..
I I

dt Slope= -K I I '. I" I .-

QL---~--~--------------~------~
~)Time
T Fig. 9.4 Regular and semi log graphs of first-order kinetics

-... First-Order Half-Life


-~)c
Substituting the value of C = C 0 /2 at t y; in equation 9. 14 and solving it
Fig. 9.3 Graph of first-order kinetics show ing linear yields:
relationship between rate of reaction and
0.693
concentrati on of drug (equation 9. J 0). t l/ = - - (9.16)
/2 K
Rearrangement of equation 9 .I 0 yields:
dC Equation 9.16 shows that, in contrast to zero-order process, the half-.
= - K dt (9. I 1) life of a first-order process is a constant. and independent of initial drug
c
concentration i.e. irrespective of what the initial drug concentration is, the
Integration of equation 9. 11 gives: time required for the concentration to decrease by one-half remains the
same (see Fig. 9.4.). The t y; of a first-order process is an important
In C = In C0 - Kt (9 .12) phannacokinetic parameter.
Equation 9.12 can also be written in exponential fonn as:
Most phannacokinetic processes viz. absorption, distribution and elimi-
C = Co e- Kt (9 .13)
nation follow first-order kinetics.
where e = natural (Naperian) log base.
Mixed-Order Kinetics (Nonlinear Kinetics)
Since equation 9.13 has only one exponent, the first-order process is In some instances, the kinetics of a pharn:tacokinetic process change
~lso called ~s monoexponential rate process. Thus, a first-order process from predominantly first-order to predominantly zero-order with increas-
IS charactenzed by logarithmic or exponential kinetics i.e. a constant
ing dose or chronic medication. A mixture of both frrst-order and zero-order
fraction of drug undergoes reaction per unit time.
kinetics is observed in such cases and therefore the process is said to
Since In = 2.303 log, equation 9.12 can be written as: follow mixed-order kinetics. Since deviations from an originally linear
ph at tnacokinetic profile is observed, the rate process of such a drug is
.
221
220 BIOrHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETICS : BASIC CONSIDERATIONS

called as nonlinear kinetics. Mixed order kinetics is also termed as dose The three different approaches to phannacokinetic analysis of experi-
dependent kinetics as it is observed at increased or multiple doses of mental data are:
some drugs. Nonlinearities have been observed in absorption (e.g. vita- 1. Compatttnent modeling
min C), distribution (e.g. naproxen) and elimination (e.g. riboflavin)
2. Noncompartrnental analysis
characteristics of few drugs. The phenomena is seen when a particular
phannacokinetic process involves presence of carriers or enzymes which 3. Physiologic modeling
are substrate specific and have defmite capacities and can get saturated at
Compartment Models
high drug concentration (i.e. capacity-limited). The kinetics of such ca-
Compartmental analysis is the traditional and most commonly use.d
pacity-limited processes can be described by the Michaelis-Menten
approach to pharmacokinetic characterization of a drug. Here, the_ body ts
kinetics.
considered as composed of several compartments that communicate re-
versibly with each other. If one considers every organ, tissue o~ bo~y
PHARMACOKINETIC MODELS fluid that _can get equilibrated with the drug as a comparttn.ent, tnfm~te
.
Drug movement within the body is a complex process. The major number of compartments can exist in the body and math~matlcal ~escrtp­
objective is therefore to develop a generalized and simple approach to tion of such a model will be too complex. Hence, tissues whtch are
describe, analyze and interpret the data obtained during in vivo drug approximately similar in their drug distribution characteristics are pooled
disposition studies. This involves development of pharmacokinetic to form a kinetically homogeneous hypothetical com pat trnent. Such a
models which provide concise means of expressing mathematically or compartment, thus, is not a real physiologic ?r anatomic .region but a
quantitatively, the time course of drug(s) throughout the body and com- fictive or virtual one, considered as a reflection of the btosystem and
pute meaningful pharmacokinetic parameters. .assumed to comprise of tissues having similar blood flow and ~g affin-
ity. The kinetics of most drugs can be described by a hypothettcal model ,
Phannacokinetic models are useful in-
consisting of one, two or at the most, three functional compartrnents
1. Characterizing the behavior of drugs in patients arranged either in series or parallel to each other. It is also assumed th.at
2. Predicting the concentration .of drug in various body fluids with the rate of drug movement between compartments (i.e. entry and extt)
any dosage regimen follow first-order kinetics.
3. Predicting the multiple-dose concentration curves from single dose Depending upon whether the compartments are arran~ed parallel ?r in
experiments a series, compartn1ent models are divided into two categones mammillary
4. Calculating the optimum dosage regimen for individual patients model and catemary model.
5. Evaluating the risk of toxicity with certain dosage regimens
Mammillary Model
6. Correlating plasma drug concentration with pharmflcologic response This model is the most common comparttnent model used in pharma-
7. Evaluating the bioequivalence/bioinequiyalence between different cokinetics. It consists of one or more peripheral comparttnents connected
formulations of the same drug to the central comparttnent in a manner similar to ~connection of satellites
8. Estimating the possible drug and/or metabolite(s) accumulation in to a planet (i.e. they are joined parallel to the central comparttnent). The
the body central compartment (or compartment 1) comprises of p~asma ~nd
9. Determining the influence of altered physiology/disease state on highly perfused tissues such as lungs, liver, kidneys, e~c. wh~ch raptdly
drug ADME equilibrate with the drug. The drug is directly ab~orbed Into this c?mpm:-
ment (i.e. blood). Elimination too occurs from this com~at tn1ent s1nc~ the
10. Explaining drug interactions I chief organs involved in drug elimination are liver and kidn~ys, the htgh!y
Caution must however be exercised in ensuring that the model fits the perfused tissues and therefor~ presumed to be rapidly access!bl~ to drug tn
experimental data; otherwise, a new, more complex and suitable model the systemic circulation. The peripheral co~partments or tissue co~­
may be proposed and tested. partments (denoted by numbers 2, 3, etc.) are those with low vascularity
222 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETICS : BASIC CONSIDERATIONS 223
Model 1
I-----~--~J----K_to__~ and poor perfus_ion. Distribution of drugs to these compartments is
through blood. Movement of drug between compartments is defined by
characteristic frrst-order rate constants denoted by letter K (see Fig. 9 .5).
One-compartment open model, intravenous administration
The subscript indicates the direction of drug movement; thus, K 12 (K-
Model 2
____K_o_~--~
> 1_____~____~1----K_l_o__~ one-two) refers to drug movement from compartment 1 to compartment 2
and reverse for K21·
'

One-compartment open model, extravascular administration (oral, rectal, etc.) The number of rate constants which will appear in a particular com-
Model 3 partrnent model is given by R.
K 12 •
I '\.
2 For intravenous administration, R = 2n - 1 (9.17)
" K 21
Kto

For extravascular administration, R = 2n (9.18)
\I
where n = number of comparttnents.
Two-compartment open model, intravenous administration
Caternary Model
Model 4 Kor K 12 In this model, the compartments are joined to one another in a series
'
..... 1
. "
' '
2 like comparttnents of a train (Fig. 9 .6). This is however not observable
K 21
physiologically/anatomically as the various organs are directly linked to
K1o
\v the blood compartment. Hence this model is rarely used.
Two-compartment open model. extravascular administration

Model 5 2
/
Fig. 9.6 A caternary model
K1 2 K 21
l!
K1 3 The compartment modeling approach has several advantages-
l '\.
3
1'\ ] . It gives a visual representation of various rate processes involved
K31
in drug disposition.
K to
\ v 2. It shows how many rate constants are necessary to describe these
Three-compartment o pen model, intraveno us administration processes .
• 3. It enables the pharmacokineticist to write differential equations
Model 6 2 for each of the rate processes in order to describe drug-concentra-
/ tion changes in each comparttnent.
K1 2 K 21
KoJ I 4. It is useful in predicting drug concentration-time profile in both
_........ K1 3
-- 1
\.
'\.
3 normal physiologic and in pathologic conditions.
J

K31 5. It is important in the development of dosage regimens.


' Kt o
\I
Disadvantages of compartment modeling include-
Three-compartment open mod e'l, extravascular administration 1. The compartments and parameters bear no relationship with the
Fig. 9.5 Various mammillary compartment models. The rate constant physiologic functions or the anatomic structure of the species;
,.
K ot ~s ~as ically Ka. the first-order absorption rate constant and
• several assumptions have to be made to facilitate data interpreta-
K w IS KE, the first-ord er elimination rate const ant. tion .
224 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETICS :BASIC CONSIDERATIONS 225
--
Practically, the AUMC and AUC can be calculated from the respective
2. Extensive efforts '!re req_u~ed in the development of an exact
model that predicts and describes correctly the ADME of a certain graphs by the trapezoidal rule (the method involves dividing the curve
drug. by a series of vertical lines into a number of trapezoids, calculating
separately the area· of each trapezoid and adding them together).
3. The model is based on curve fitting of plasma concentration with •

complex multiexponential mathematical equations.


4. The model may vary withm a study population.
5. The approach can be applied only to a specific drug under study. cP
E
c:' (t2- t1)(C1 + C2) ·-
....
6. The drug behavior within the body may fit different com pat trnen- ....0C'O
·- AUC=
2
)(
tal models depending upon the route of administration. ....c:
~
c:

7. Difficulties generally arise when using models to interpret the ~


c:
0 AUC=
(t4-t3)C3
--
0
·-
!
c:
differences between results from human and animal experiments. (.) 2 B
c:
Noncompartmental Analysis i 0
(.)

Because of the several drawbacks of and difficulties with the classical


comparttnent modeling, newer approaches have been devised to study the
i
t.ime course of drugs in the body. They are . physiologic models and
~)> Time
noncom_partmental methods.
Fig. 9. 7 AUC and AUMC plots
The noncompartmental analysis, also called as the model indepen-
dent method, does not require the assumption of specific comparttnent MRT is defined as the average amount of time spent by the drug in
model. This method can however be applied to any comparttnent model the body before being eliminated. In this sense, it is the statistical
provided the drugs or metabolites follow linear kinetics. The approach, moment analogy of half-life, ty2 • In effect, MRT represents the time for
based on the statistical moments theory, involves collection of experi- 63.2% of the intravenous bolus dose to be eliminated. The values will
mental data following a single dose of drug. If one considers the time always be greater when the drug is administered in a fashion other than
course of drug concentration in plasma as a statistical distribution curve, i.v. bolus.
then:
Noncomparttnental technique is widely used to estimate the important
MRT= AUMC pharmacokinetic parameters like bioavailability, clearance and apparent
(9 .19)
AUC volume of distribution. The method is also useful in detennining half-
where MRT = mean residence time life, rate of absorption and first-order absorption rate constant of the drug.
AUMC = area under the first-moment curve Advantages of noncomparttnental method include-
AUC = area under the zero-moment curve }. Ease of derivation of phannacokinetic parameters by simple alge-
AUMC is obtained from a plot of product of plasma drug concentra- braic equations.
to
tion and time (i.e~C.tf versus time t from ieio-· infmity (Ftg. 9.7). 2. The same mathematical treatment can be applied to almost any
,-Mathematically, it is expressed by equation: drug or metabolite provided they follow first-order kinetics.
3. A detailed description of drug disposition ·characteristics is not
AUMC = I:c t dt (9.20)
required.

AUC is obtained from a plot of plasma drug concentration versus The disadvantage of this method is that it provides limited infonna- '
time from zero to infinity. Mathematically, it is expressed by equation: tion regarding the plasma drug concentration-time profile; more often, it
deals with averages.
(9 .21)


-~226
·.::
BIOPHARMACEUTICS AND PHARMACOKINETiCS PHARMACOKINETICS : BASIC CONSIDERATIONS 227

Physiologic Models only to the highly membrane penneable drugs i.e. low molecular weight,
These models are also tenned as blood flow rate-limited models and poorly ionized and highly lipophilic drugs. For highly polar, ionized and
perfusion rate-limited models. The model is drawn on the basis of charged drugs, the model is referred to as membrane permeation rate-
~nown anatomic and physiologic data and thus presents a ~more realistic limited.
· . picture of drug disposition in various organs and tissues. The· number of Physiologic modeling has several advantages over the conventional
c~mpartinents to be included in the model depends upon the disposition comparttnent modeling:
characteristics of the drug. Organs or tissues .such as bones that have no
1. Mathematical treatment is straight forward.
drug penetration are excluded. . Fig. 9.8 shows such a physiologic model
where the co~parttnents are arranged in a series in a flow diagram. 2. Data fitting is not required; drug concentration in various body
{
regions can be predicted on the basis of organ or tissue volume,
• ; • - IV
. .DOSE ,
perfusion rate and experimentally detennined tissue-to-plasma partition
QLUNG
- ,.
[~ . coefficient.
LUNG
. 3. The model gives exact description of drug concentration-time
~ ko . profile in· any organ or tissue and thus better picture of drug
.
~ '•

...
distribution characteristics in the body .
...,.
r
HEART
., 4. The influence of altered physiology or pathology on drug disposi-
QL -QG ,
tion can be easily predicted from changes in the var.i ous
1- ~ phannacokinetic parameters since the parameters correspond to
QG aL .

.
,.• GUT

• liVER I<M •..
,
-.. actual physiologic and anatomic measures .
0
.
0
5. Correlation of data in several animal species is possible and
0 elimination 0
0
..J
-
. 0
with some drugs, can be extrapolated to humans.
. m ..J
en
.-
..J ,
<{
·-a::
'
'
.
.... KIDNEY •
QK
.... CJ)
:::>
The only disadvantage of these comprehensive models is obtainin.
. .
g
. Ke experimental data which is very ·exhaustive. ,
...a::
w ) elimination 0
z
w
< . . >
.
. QHPT
. QUESTIONS
, r
... HPT ..
...
I

' 1. In addition to mechanisms of drug ADME, explain how the knowledge


.
about the kinetics of these processes is also important..
.. . '
Oppr
..... PPT
..
p
2. Define pharmacokinetics. N arne and define the three pharrriacokinetic pa-
rameters that describe a typical plasma level-time curve .
. .
'· 3. On what parameters/variables are Cmax, tmax and AUC dependent? What is
the importance of these parameters in expressing pharmacodynamic behavior
of a drug?
4. What are the various pharmacodynamic parameters?
5. Quote examples of zero-order rate processes.
6. In contrast to zero-order process, the half-life of a first-order process is
considered to be an important pharmacokinetic parameter. Why?·
7~ . Why are first-order processes said to follow
'
linear kinetics?
8. What are mixed-order processes? By what other names such processes are
identified? Quote examples of su\h proc~sses.
i I

228 BIOPHARMACEUTICS AND PHARMACOKINETICS PHARMACOKINETICS : BASIC CONSIDERATIONS 229
9. What are pharmacokinetic models? What is the importance and utility of b. What is the rate constant for decomposition?
developing such models? Answer : Drug A = 42 mg/hour, and Drug B = 0 .198/hour.
10. Name the different approaches to pharmacokinetic analysis of experimental c. What is their half-life?
data.
Answer : Drug A = 4.76 hours, and Drug B = 3.5 hours.
11. What assumptions are made in the development of compartment models?
d. What were the original amounts of drug before decomposition?
Discuss the advantages and disadvantages of such an approach.
12. In compartment modeling, elimination is presumed to occur from central
Answer : Drug A = 400 mg, and Drug B = 200 mg.

compartment only. Why? ·· e. If the original quantities of drug taken were 800 mg for A and 400
mg for B then what will be their new half-lives?
13. In comparison to a mammillary model, the catemary model is less useful.
Explain. Answer : Drug A = 9.52 hours, and Drug B = t~ will remain
unchanged i.e. 3.5 hours.
14. On what theory is the noncompartmental analysis of pharmacokinetic data
based? Discuss the merits and demerits of such an approach. . f. Write equations for the line that best fits the experimental data for
both drugs.
15. What are the advantages of physiologic models over compartment models?
On what assumptions are such models based? Answer : Drug A : C = 400 - 42t, and
. ,.
16. The half-life for first-order photolysis of cefotaxime solution containing 150 Drug B : log C = log 200 - 0.198t/2 .303.
mg drug is 50 minutes.
a. How long will it take for the drug to decompose to 20% of its
original amount?
,
Answer : 116 minutes.
b. If one mL aliquot taken after 90 min of exposure to light was found
to contain 0.43 mg of cefotaxime, what was the original volume of
the solution ?
Answer : 100 mi.
17. The following data for decomposition of two drugs, A and 8 are given in
the table below :
·.
Time (hours) Drug A (mg) Drug B (mg)

0.5 379 181.2


1.0 358 164.0
1.5 337 148.6
2.0 316 134.6
3.0 274 110.4
4.0 232 90.6
6.0 148 61.0
8.0 64 41.0

a. Determine (by plotting or otherwise) the rate of decomposition of


both drugs.

Answer : Drug A = zero-order, and Drug B = first-order.


COMPARTMENT MODELING 231

upon th~ rate of input, several one-comparttnent open models can be


defmed:
One-compartment open model, intravenous bolus administration
One-compartment open _model, continuous intravenous infusion
10 One-compartment open model, extravascular administration, zero-order
absorption, and '
Compartment Modeling . One-comparttnent open model, extravascular administration, first-o~~er
. .
absorption. .

One-Compartment Open Model-


The time course of drug concentration detettnined after its administra- Intravenous Bolus Administration
tion can be satisfactorily explained by assuming the body as a single, well When a drug that distributes rapidly in the body is given in the form
mixed compartment with frrst-order disposition processes. In case of of a rapid intravenous injection (i.e. i.v. bolus or slug), it takes about one
other drugs, two or more body compattrnents may be postulated to de- to three minutes for complete circulation and therefore the rate of absorp-
scribe mathematically the data collected. tion is neglected in calculations. The model can be depicted as follo~s:

ONE-COMPARTMENT OPEN MODEL Blood and other . KE . ...


t------~-

(Instantaneous Distribution Model) Body Tissues --


The one-compartrnent open model is the simplest model which depicts
The general expression for rate of drug presentation to the body is:
the body as a single, kinetically homogeneous unit that has no barriers to
the movement of drug and fmal distribution equilibrium between the drug •
dX =Rate in (availability) - Rate out (elimination) (10.1)
· in plasma and other body fluids is attained instantaneously and maintained dt
at all times. 'This model thus applies only to those drugs that distribute
, rapidly throughout the body. The anatomical reference compartment is Since rate in or absorption is absent, the equation becomes:
the plasma and concentration of drug in plasma is representative of drug dX
- = - Rate out ( 10.2)
concentration in all body tissues i.e. any change in plasma drug concentra- dt
tion reflects a proportional ·change in drug concentration throughout the
body. However, the model does not assume that the drug concentration in If the rate out or elimination follows first-order kinetics, then:
plasma is equal to that in other body tissues. The term open indicates dX
that the input (availability) and output (elimination) are unidirectional -=- KE X (10.3)
dt
and that the drug can be eliminated from the body. Fig. 10.1 shows such
a one-compartment modeL where, KE = first-order elimination rate constant, and
Metabolism X = amount of drug in the body at any time t remaining to be eliminated ..
I

Ka Blood and KE Negative sign indicates that the drug is being lost from the body. The ·
Drug----~ Other Body
Input Tissues Output various related pharmacokinetic parameters can now be estimated.
(Absorption) (Elimination) Excretion
Elimination Rate Constant : For a drug that follows one-compart- .
~

Fig. 10.1 Representation of one-compartment open model ment kinetics and administered as rapid i. v. injection, the decline in
showing input and output processes. plasma drug concentration is only due to elimination "'f drug from the
body (and not due to distribution)~ the phase being._called as eli~inalion
.One-comparttnent open model is generally used to describe plasma
phase. Elimination phase can becharacterized by 3 parameters elimination
levels following administration of a single dose of a drug. Depending
rate constant.. elimination half-life and clearance.
230
232 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING
233

Integration of equation 10.3 yields: logC 0

ln X = lh X 0 - KE t

(1 0.4) a. Regular Plot . -
~
~
b. Semilog Plot
where, X 0 = amount of drug at time t = zero i.e. the initial amount of f/)
C)
drug injected.
-
-0
c:
Equation 10.4 can also be written in the exponential fonn as: c
0
....,
·- --
0
·-
l!
Slope=--
-KE
X= X0 e-KEt (10.5) ...
C'G
...., c:
~ I
2.303
c: C: I
The above equation shows that disposition of a drug that follows one- ~ 0 ,lOg C'1. I log c1 - log c2
0
c: (.) ----·----+------
I
-
compartment kinetics is monoexponential. () 0
2. 303 (t1 - t2)
. -
0 I
I
I
Transfottning equation 10.4 into common logarithms (log base 10), we
get:
T I
I
~
t% .
I
I
~
tv72
- ..... ~
I

: t1 ~ t2_
'
I
I
KE t I

log X = log X0 - -- (10.6) -----...)! Time


2.303
' Fig. 10.2 (a) Cartesian plot of a drug that follows one-compartment
Since it is difficult to detertnine directly the amount of drug in the kinetics and given by rapid i. v. injection, and
body X, advantage is taken of the fact that a constant relationship exists (b) Semilogarithmic plot for the rate of elimination in
between drug concentration in plasma C (easily measurable) and X; thus: a one-compartment modeL

., (I 0.7) The fraction of drug eliminated by a particular route can be evaluated


if the number of rate constants involved and their values are known. For
where, Vd = proportionality constant popularly known as the apparent ·
example, if a drug is eliminated by urinary excretion . and .metabolism
volume of distribution. It is a phartnacokinetic parameter that pettnits the
only, then, the fraction of drug excreted unchanged In urtne Fe and
use of plasma drug concentration in place of amount of drug in the body.
fraction of drug metabolized Fm can be given as:
The equation 10.6 therefore becomes:
KE t F = Ke (10.10a)
\
log C = log C 0 - -- (10.8) e KE
2.303
where, C 0 = plasma drug concentration immediately after i.v. injection. Km (IO.IOb)
F =-
m KE
Equation 10.8 is that of a straight line and indicates that a semiloga-
rithmic plot of log C versus t will be linear with Y- intercept log C 0 . The Elimination Half-Life : Also called as biological half-life, it is the
elimination rate constant is directly obtained from the slope of the line oldest and the best known of all pharmacokinetic paramters and was once
(Fig. 10.2b). It has units ·of min- 1. Thus, a linear plot is easier to handle considered as the most important characteristic of a drug. It is defined .as
mathematically than a curve which in this case will be obtained from a the time taken for the amount of drug in the body as well as plasm.a
plot of C versus t OQ. regular (Cartesian) graph paper (Fig. 10.2a). concentration to decline by one-half or 50% its initial value. It ts
Thus, C 0, KE (and tYl) can be readily obtained from log C versus t expressed in hours or minutes. Half-life is related to elimination rate

graph. The elimination or removal of the drug from the body is the sum constant by the following equation:
of urinary excretion~ metabolism, biliary excretion, pulmonary excretion, 0.693 (10.11)
and other mechanisms involved therein. Thus, KE is an additive property
KE
of rate constants for each of these processes and better called as overall
elimination rate constant. · Elimination half-life can be readily obtained from the graph of log C
versus t as shown in Fig 10.2.
(10.9)
234 BIO,PHARMACEUTICS AND PHARMACOKINETICS 235
COMPARTMENT MODELING
Today, increased physiologic understanqing of pharmacokinetics shows context and as a measure of drug elimination mechanisms. A much more
that half-life is a secondary parameter that depends upon the primary valuable alternative approach for such applications is use of clearance
parameters clearance and apparent volume of distribution according to parameters to characterize drug disposition. Clearance is the mo~t impor-
following equation: tant parameter in clinical drug applications and is useful in evaluating the
0.693 vd mechanism by which a drug is el~minated by the whole organism or by a
'tYi = - - - - (10.12)
CIT particular organ.
~pparent Volume of Distribution : Clearance and apparent volume Just as V d is needed to relate plasma drug concentration with amount
of distribution are two separate and independent pharmacokinetic charac- of drug in the body, clearance is a parameter to relate plasma drug
teristics of a drug. Since they are closely related with the physiologic concentration with the rate of drug elimination ·according to following
mechanisms in the body, they are called as primary parameters. equation:
Rate of elimination
Modification of equation I 0. 7 defines apparent volume of distribution: Clearance = - - - - -- - -- - - (10.16)
Plasma drug concentration
Amount of drug in the body X
vd = - - - - - - - - - - - - - {10.13)
Plasma drug concentration c or Cl =
dX/dt
( 10.17)
Vd is a measure of the extent of distribution of drug and is expressed · c
in liters. The best and the ~imp lest way of estimating V d of a drug is Clearance is defined as the theoretical volume of body fluid contain-
administering it by rapid i.v. injection and using the following equation: ing drug (i.e. that fraction of apparent volume of distribution) from which
i.v. bolus dose the drug is completely removed in a _given period of time. It is expressed
(10.14) ·in ml/min or li.ters!hour. Clearance is usually further defined as blood
I
clearance (Clb), plasma clearance (Clp) or clearance based on un?ound
Equation I 0.14 can only be used for drugs that obey one-compartrnent or free drug concentration (Clu) depending upon the concentration C
ki,netics. This is because the Vd can only be estimated when distribution measured for the right side of the equation 10. 17.
equilibrium is · achieved between drug in plasma and that in tissues and Total Body Clearance : Elimination of a drug from the body involves
such an equilibrium is established instantaneously for a drug tha\ follows processes occurring in kidney, liver, lungs and other eliminating organs.
one-compartment kinetics. A more general, fTlOre useful noncompartmental Clearance at an individual organ level is called as organ clearance. It
method that can be applied to many compartment models for estimating can be estimated by dividing the rate of elimination by each organ with
the Vd is: the concentration of drug presented to it. Thus,
For drugs given as i.v. bolus, Rate of elimination by kidney
Renal Clearance ClR = - - - - - - - - -- - ( 10.18a)
Xo C
V d(area) = - - - - - + -- (10.15a)
KE.AUC
Rate of elimination by liver
For drugs administered extravascularly (e. v. ), Hepatic Clearance ClH = - - - -- - - - - - (10.18b)
F X0 . C
v - ---~
d(area) - KE AU(' (10.15b)
Other Organ Clearance
Rate of elimination by .other organs
where, X0 = dose administered, and F = fractfon of drug absorbed into the Clothers = - - - - - - -- - - - - - (10.18c)
systemic circulation. r· is equal to one i.e. complete availability when the c
drug is administered intravenously. The total body clearance, ClT, also called as total systemic clear-
Clearance
., : Difficulties arise when one applies elimination rate con- ance, is an additive property of individual organ clearances. Hence,
stant and half-life as phannacokinetic parameters in an anatomical/physiological Total Systemic Clearance, CIT = ClR + ClH + Clothers (10. 18d.)
236
BIOPHARMACEUTICS AND PHARMACOKI~rETICS COMPARTMENT MODELING 237
Be~ause. o~ th~ additivity of clearance, the relative contributior by any For a drug given by i.v. bolus, the renal clearance· CIR may be esti-
organ In ehmtnattng a drug can be easily calculated. Clearance by all
mated by determining the total amount of unchanged drug excreted in
organs other than kidney is sometimes known as nonrenal clearance
urine, xux and AUC.
CINR. It is the difference between total clearance and renal clearance.
According to an earlier definition (equation I 0 .17), (10.23)

CIT = dX/dt
Organ Clearance : The best way t. of understanding clearance is at
(10.17)
c individual organ level. Such a physiologic approach is advantageous in
Substituting dX/dt = KEX from equation I 0.3 in above equation, we predicting and evaluating the influence of pathology, blood flow, P-D
get: ,binding, enzyme activity, etc. on drug elimfuation. At an organ level, the
rate of elimination can be written as:
(IO.I9)
Rate of elimination = Rate of presentation -Rate of exit ( 10.24)
Since X/C - Vd (from equation I 0.13), the equation 10.19 can be by an organ to the organ from the organ
written as: Rate of presentation = Organ blood flow x Entering concentration
(1 0.20a) (input) = Q.Cin (1 0.25)
Parallel equations can be written for renal and hepatic clearances as: Rate of exit (output) = Organ blood flow x Exiting concentration
= Q.C0 ut (I 0.26)
CIR = Ke Vd (I0.20h)
Substitution of equations 10.25 and I 0.26 in equation 10.24 yields:
CIH = Km Vd (10.20c)
Rate of elimination = Q.Cin- Q.Cout
Since KE = 0.693/tY2 (from equation 10.1 I), clearance can be related to
half-life by the following equation: (also called as rate of extraction) = Q (Cin - Cout) (1 0.27)

0.693 vd Division of above equation by concentration of drug that enters the


Clr = (I 0.2 I ) organ of elimination Cin yields an expression for clearance of drug by the
tY2 organ under consideration. Thus:
Identical equations can be written for CIR and CIH in which cases the Rate of extraction
tY2 will be urinary excretion half-life for unchanged drug and metaboJism = Cl = Q (Cin - Cout) = Q ER (10.28)
organ C· ·
ha.lf-li~e respe~tively. Equation I 0.21. shows that as CIT decreases, as in Cin Ill

renal Insufficiency, tY2 of the drug increases. As the Clr takes into where, ER = (Cin - C0 u1)/Cin is called as extraction ratio. It has no units
account Yd, changes in Vd as in obesity or edematous condition will and its value ranges from zero (no elimination) to one (complete elimina-
reflect changes in CIT. tion). Based on ER values, drugs can be classified into 3 groups:
The noncompa!tmental method of computing total clearance for a drug - drugs with high ER (above 0.7),
that follows one-comparttnent kinetics is: - drugs with intermediate ER (between 0.7 to 0.3), and
For drugs given as i. v. bolus, -drugs with low ER (below 0.3).

Xo ER is an index of how efficiently the eliminating organ clears the


CJT=-- (I 0.22a) blood flowing through it of drug. For example, an ER of 0.6 tells that
AUC 60% of the blood flowing through the organ will be completely cleared of
••
For drugs ad1ninistered e. v., drug. The fraction of drug that escapes removal by the organ is expressed
F X0 as:
CIT= (I 0.22b)
AUC F = 1 - ER (10.29)
where, F = systemic availability when the eliminating organ is liver.
238 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING 239

Hepatic Clearance : For certain drugs, the nonrenal clearance can be On the contrary, hepatic · blood·1 flow has very little or no effect on .
assumed as equal to ·hepatic clearance ClH. It is given as: drugs with low ERH e.g. theophylline. For such drugs, whatever concen-
tration of drug present in the blood perfuses liver, is more than what the
(1 0.30)
liver can eliminate (low first-pass hepatic metabolism). Similar discussion
An equation parallel to equation I 0.28 can also be. writteQ for hepatic can be extended to the influence of blood flow on renal clearance of
clearance: drugs. This is illustrated in Table 10.1 . ·Hepatic clearance of a drug with
CIH = QH.ERH (10.31) high ER is independent of protein binding.

where, QH = hepatic blood flow (about 1.5 liters/min), and 2. Intrinsic Capacity Clearance : Denoted a~ Clinb it is defined as
ERH = hepatic extraction ratio. the inherent ability of an organ to irreversibly remove a dr_ug in the
absence of any flow limitation. It depen~s, in this case, upon the hepatic
The hepatic clearance of drugs can be divided into two groups: I enzyme activity. Drugs with low ERH and with elimination primarily b)'
- drugs with hepatic blood flow rate-limited clearance, and metabolism are greatly affected by changes in enzyme activitv. _ !-f~tic
~drugs with intrinsic capacity-limited clearance. clearance of such drugs is said to be capacity-limited, e.g. theopnyllirc.
The tY2 of such drugs show great intersubject variability. -Hepatic cle=-- - ~
1. Hepatic Blood Flow : When ERH is one, CIH approaches its
ance of drugs with low ER is independent of blood flow rate but sensitive·-
. maximum value i.e. hepatic blood flow. In such a situation, hepatic •

to changes in protein binding.


clearance is said to be perfusion rate-limited or flow-dependent. Alter-
ation in hepatic blood flow significantly affects the elimination of drugs Th~
hepatic and renal extraction ratios of some drugs ·and metabolites
with high ERH e.g. propranolol, lidocaine, etc. Such drugs are removed are given in Table I 0.2.
from the blood as rapidly as they are presented to the liver (high first-pass TABLE 10.2
hepatic metabolism). Indocyanine green is so rapidly eliminated by the Hepatic and Renal Extraf:tion Ratio of Some Drugs and Metabolites
human liver that its clearance is often used as an indicator of hepatic
blood flow rate. First-pass hepatic extraction is suspected when there is Extraction Ratio
lack of unchC:lnged drug in systemic circulation after oral administration.
' ' High Intermediate Low
Maximum oral availability F for such drugs can be computed from
equation 10.29. An extension of the same equation is the noncompartmental Propranolol Aspirin Diazepam
method of estimating F: Lidocaine Codeine Phenobarbital

AUCoral Hepatic . Nitroglycerine Nortriptyline Phenytoin


( 10.32) Quinidine Procainamide
AUC·I. v. Extraction Morphine
Isoprenaline Theophylline
_ _ _ __;___+---- - - - - - ---- - - - ------·- - -- - · -- -
TABLE I 0.1
Some Penicillins Some Penicillins Digoxin
Influence of Blood Flow Rate and_Protein Binding on Total Clearance
of Drugs with High and with Low ER Values. Renal Hippuric acid Procainamide Furosemide
Extraction Several Sulfates Cimetidine Atenolol
Drugs with Changes in Total Clearance due to
1---- - -· - - . ---~ ···-- ·· -- · --
Several Glucuronides Tetracycline
t Blood Flow i Blood Flow t Binding i Binding .

.
1.
'
HighER No Change No Change
. (above 0. 7) One-Com·partment Open Model

-lntravenous Infusion '
LowER No Change
· (below 0.3) ·
No Change
1 1 Rapid i. v. injection is unsuitable when the drug has potential to p~e~.
cipitate toxicity or when maintenance of·a stable concentration &- amount
where, t = increase~ and ~ = decrease.
:•
241
240 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING

of drug in the body is desiretl. In such a situation, the drug (for example,
c:
several antibiotics, theophylline, procainamide, etc.) is administered at a ....0as
constant rate (zero-order) by i.v. infusion. In contrast to the short dura-
·-
....c:
'- • Infusion
rate= 2Ro
tion of infusion of an i. v. bolus (few seconds), the duration of constant Q)
0
c:
rate infusion is usually much longer than the half-life of the drug. Advan- 0
(.) Css Steady-state . Infusion stopped
tages of such a zero-order infusion of drugs include:- C>
--- ----- f
:::J
0
'- Infusion I
1. Ease of c_ontrol of rate of infusion to fit individual patient needs. When plotted on a
cu rate= Ro 't semilog graph yields
2. Prevents fluctuating maxima and minima (peak and valley) plasma E I
(/)
a straight line with
level, desired especially when the drug }J.as a narrow therapeutic -a.as I
I slope = -KE I 2.303
I
index. . _.
3. ' Other drugs, electrolytes and nutrients can be conveniently admin- i '1 nfusion time T ---tl
I

istered simultaneously by the same infusion line in critically ill


~)Tim~
patients.
10 3 P lasma concentration-time profile for a drug g.ive_n
''
The model can be represented as follows: Fig. . d t
j

by constant rate i.v. infusion (the two curves tn tea e


different infusion rates Ra and 2R, for the same drug)
_· Ro Blood and KE
Drug ~ Other Body ----~Elimination At steady-state, the rate of change of amount of drug in the body is
zero-order Tissues
infusion rate zero, hence, the equation 10.33 becomes:
Zero = Ro - KE Xss
At any time during infusion, the rate of change in the amount of drug
(1 0.36)
in the body, dX/dt is the difference between the zero-order rate of drug or KE Xss = Ro
infusion Ro and first-order rate of elimination, -KEX: Transforming to concentration tenns and rearranging the equation:.
dX
(10.33) Ro Ro . Infusion rate (l0. 37)
dt C = - t.e.
ss KEy d CIT Clearance
Integration and rearrangement of abo.ve equation yields: and-C are amount of drug in the body and concentration of
X
where, ss ss 1 f K ( d hence
~ an
X . ~ (I _ e-KEt) (10.34)
drug in plasma at steady-state respectively. The va ue o
tY:) can be obtained from the slope of straight line obtamed. a~e~ a
s~milogarithmic plot (log c
versus t) of the plasma c~ncentrat1on time
Since X = Vd C, the equation I 0.34 can be transformed into concen- data gathered from the time when infusion is stopped (F1g: 10,_3). ~Iter­
tration terms as follows: natively, KE can be calculated from the data collected dunng mfuswn to
C = .
Ro· (I _ .
e-KEt) =
Ro (l _ e-KEt) steady-state as follows:
( 10.35)
T -- c ss from equation 10 .37 in equation 10.35 we get:
'
SUbStl·rut·tng D/Cl
'

KEY d Clr J.'-Q

At the s~a.rt of constant rate infusion, the amount of drug in the body is C == Css ( 1 _ e-KEt) ( 10.38)
zero and hence, there is no elimination. As tim~ passes, the amount of
drug in the body rise~ gradually (elimination rate less than the rate of Rearrangement yields:
infusion) until a point after which the rate of elimination equals the rate of Css· -C == e- Kt
E (I 0.39)
••
;j
infusion i.e. the concentration of drug in plasma approaches a constant Css
value called as steady-state, plateau or infusion equilibrium (Fig.- ~ 0.3. ).

\
242 BIOPHARMACEUTICS AND PHARMACOKINETICS
COMPARTMENT MODELING 243
-
Transfonning into log fotrn, the equation becomes:
For therapeutic purpose, more than 90% of the steady-state drug con-.
Css - C · -KEt centration in the blood is desired which is reached in 3.3 half-lives. It
Iog ---- ( 10.40)
Css 2.303 takes 6.6 half-lives for the concentration to reach 99% of the .steady-state.- .
., Thus, the shorter the half-life (e.g. penicillin G, 30 min), sooner is the
A semilog plot of (Css - C)/C55 versus t results in a straight line with steady-state reached.
slope -KE/2.303 (Fig. 10.4).
Infusion Plus Loading Dose : It takes a very long time for the drugs
having longer half-lives before the plateau concentration is reached (e.g . .
phenobarbital, 5 days). Thus, initially, such drugs have subtherapeutic
concentrations. This can be overcome by administering an i. v. loading

dose large enough to yield the desired steady-state immediately upon
Css-C injection prior to starting the infusion. It should then be followed imme-
log -KE
Css Slope=
diately by i. v. infusion at a rate enough to maintain this concentration
2.303 (Fig. 10.5).
r loading dose
~ ,/
resultant constant
. / plasma level
~
0
--- --
--
t
..0
Q)
..
' .. , ,-
--
----...)! 5 •.. ;~.. . asymptotic rise
.~ '.. ,, of infused drug
Fig. 10.4 Semilog plot to compute K'E from
infusion data upto steady-state
' ·\
...
..
,,, ##

The time to reach steady-state concentration ·is dependent upon the


elimination half-life and not infusion rate. An increase in infusion rate
-g
c
I
I
.•#
':<
,
~,
will merely increase the plasma concentration attained at steady-state (Fig. E 1 '·~ exponer.tial decline
<( I ·~+-4-
1 start of ·~ of loading dose
10.3). If n is the numl?er of half-lives passed since the start of infusion ·~.
~tlty2 ), equation 10.38 can be written as: 1 I
1~ In USIOn
.. f .
'-•-..
-·- ...... _
'

\
'
l .. 6
C = ,C 5 ~ [1 - (~) ]0
(10.41) 0
,
I _.....) Half lives
The percent of Css achieved at the end of each tY2 is the sum of Css at •
Fig. 10.5 Intravenous infusion with loading dose.
previous tY2 and the concentration of drug remaining after a given tY2 As the amount of bolus dose remaining
(Table 10.3). . in the body falls, there is a complemen-
TABLE 10.3 tary rise resulting from the infusion
Percent of Css attained at the end of a given tYz ••

Recalling once again the relationship X = Vde, the equation for -


Half-life %Remaining % Css Achieved
CQ.ljnput_ing the loading dose Xo,L can be given:
1 50 50
Xo,L = Css Vd ( 10.42)
2 25 50 + 25 = 75
Substitution of Css = Ro/KE Vd from equation 10.37 in above equation
3 12.5 75 + 12.5 = 87.5
yields another expression for loading dose in terms of infusion rate:
4 . 6.25 87.5 + 6.25 = 93.75
5 3.125 93 .75 + 3.125 = 96.875
Ro
Xo,L = , KE (l 0.43)
'6 1.562 96.875 + 1.562 = 98.437
7 0.781 98.437 + 0.781 = 99.218 The equatio~ describing the plasma concentration-time profile 'follow-
ing simultaneous i.v. loading dose (i.v. bolus) and constant rate i.v. infusion
- 244 BIOPHARMACEUTICS AND PHARMACOKINETICS 245
(.'OMPARTMENT MODELING
is the sum of two equations describing each process (i.e. modified equa-
by a decline in the rate with ARA i.e. absorption rate is de~end~nt up~n
tion 10.5 and 10.35):
ARA; its regular plot is curvilinear and semilog plot a straight hne With
C = Xo,L e-KEt (I 0.44) absorption rate constant as its slope.
vd
a. Cartesian Plot b. Semilog Plot
If we substitute CssV d for Xo,L (from equation 10.42) and CssKEV d for
~ (from equation 10.37) in above equation and simplify it, it reduces to --- ~,
,,
C- =_Css indicating that the concentration of drug in plasm~ remains ',' , zero-order
constant (steady) throughout the infusion time. ' ~
<(
- 't ARA · Cl \
Assessment of Pharmacokinetic Parameters -
0
first-order
\
\\
The first-order elimination rate constant and elimination half-life can T \
\
be computed from a semilog plot of post-infusion concentration-time data.
Equation 10.40 can also be used for the same purpose. Apparent volume ''
of distribution and total systemic clearance can be estimated from steady- ~)Time ---+)l Time
state concentration and infusion rate (equation 10.37). These two parameters
can also be computed from the total area under the curve (Fig. 10.3) till Fig. 10.6 Distinction between zero-order and first-order absorption processes.
Figure a is regular plot, and Figure b a semi log plot of amount of
the end of infu.sion:
drug remaining to be absorbed (ARA) versus time t.
AUC = Ro T = Ro T - ·c ss T (10.45)
KE Vd CIT After e. v. administration, the rate of change in the amount of drug in
the body dX/dt is the difference between the rate of input (absorption)
where, T = infusion time.
dXevldt and rate of output (elimination) dXE/dt.
The above equation is a general expression which can be applied to
dXJdt = Rate of absorption - Rate of elimination
several phatrnacokinetic models.
dX dXev dXE
One-Compartment Open Model ---- ( 10.46a)
· Extravascular Administration dt dt dt
'
When a drug is administered by extravascular route (e.g. oral, i.m., Absorption rate =Elimination rate
rectal, etc.), absorption is a prerequisite for its therapeutic activity. Fac- - Cmax
c: -----
tors that influence drug absorption have already been discussed in chapter ·-....C1S
0 I
I
2. The rate of absorption may be described mathematically as a zero- ....c:
~
I
I
order or frrst-order process. A large number of plasma concentration-time ~ I Post-absorption Phase
c:
I
-profiles can be described by a one-compartrnent model with first-order 0
(.) I
...
absorption and elimination. However, under certain conditions, the ab-· "
/'
~
Cl
::J I
sorption of some drugs may be better described by assuming zero-order 0 Absorption I
C1S
Phase :
(constant rate) kinetics. Differences between zero-order and first-order E
f/)
I
r kinelics are illustrated in Fig. 10.6. -C1S
a. I
I
I Elimination Phase
. Zero-order absorption is characterized by a constant rate of absorp- I
tion. It is independent of amount remaining to be absorbed (ARA), and T J

its regular ARA versus t plot is linear with slope equal to rate of absorp- _ _.~ Time
tion while the semilog plot is described by an ever-increasing gradient
Fig. 10.7 The absorption and elimination phases of the plasma
with time. In contrast, the frrst-order absorption process is distinguished concentration-time profile obtained after extravascular
administration of a single dose of a drug.
246 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING .. .. .. 247

For a drug that follows one-compartment kinetics, the plasma concen- The differential form of the equation 10.46a is:
tration-time profile is characterized by absorption phase, post-absorption I
dX
phase and elimination phase (Fig. 10.7. ). - = Ka Xa- KE X (10.47)
dt
During the absor11;tion phase, the rate of absorption is greater than the
rate of elimination where, Ka = first-order absorption rate constant, and
~a = amount of drug at the absorption site remaining to be
dXev dXE
-->-- (10.46b) absorbed i.e. ARA.
dt dt
Integration of.equation 10.47 yields:
At peak plasma concentration, the ~te of absorption equals the rate of
K FX . -
elimination and the change_in amount of drug in the body is zero. X= a o e-KEt_e-Kat (10.48)
(Ka- KE)
----- (10.46c) Transforrning into concentration tenns, the equation becomes:
dt dt
Ka F X 0
During the post-absorption phase, there is some drug at the extravas- C= e-KEt - e-Kat ( 10.49)
cular site still remaining to be absorbed and the rate of elimination at this Vd (Ka- KE)
stage is greater than the absorption rate. where F = fraction of drug absorbed systemically after e. v. administration.
'

dXE A typical plasma concentration-time profile of a drug administered e.v. is


\ dXev
___...__ < - - (10.46d) shown...... in Fig. '10.7.
dt dt ~

After completion of drug absorption, its rate becomes zero and the Assessment of Pharmacokinetic Parameters '

pl~ma level time curve is characterized only by the elimination phase. Cmax and tmax : At peak plasma concentration, the rate of absorption
equals rate of elimination i.e. KaXa = KEX and the rate of change in
Zero-Order Absorption Model plasma drug concentration dC/dt = zero. This rate can be obtained by
This model is similar to that for constant rate infusion. differentiating equation 10.49 .

Drug at
Ro dC
Blood and ---------.. elimination ( 10.50)
e.v. site Other Body dt
zero-order
absorption Tissues
On simplifying, the above equation becomes:
The rate of drug absorption, as in the case of several controlled drug KE e- KEt = Ka e- Kat (10.51)
delivery systems, is constant and continues until the amount of drug at the
. absorption site (e.g. GIT) is depleted. All equations that explain the Converting to logarithmic form,
plasma concentration-time profile for constant rate i. v. infusion are also KE t Kat
applicable to this model. log KE - - - - - log K - - -- ( 10.52) .
2.303 a 2.303

'•
First-Order Absorption Model where t is tmax· Rearrai,Jgement of above equation yields:
For a drug that enters the body by a first-order absorption process,
gets distributed in the body according to one-compat ttnent kinetics and is 2.303 log (KalKE)
tmax .= (10.53)
eliminated by a frrst-order process, the model can be depicted as follows:
'
Ka The above equation shows that as Ka becomes larger than KE, tmax
Drug at Blood and
----~ elimination becomes smaller since (Ka - KE) increases much faster than log KalKE.
e.v. site first-order Other Body
absorption Tissues
249
248 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING

Cmax can be obtained by substituting equation 10.53 in equation 10.49. If KaFX0 /V d(Ka-KE) = A, a ~ybrid constant, then:
However, a simpler expression for the same is: C = A e-KEt __ A e-Kat (1 0.58)

F Xo · ·ts amos
1 t over, Ka >>
Cmax -- e
-KEtmax ( 10.54) During the elimination phase, when absorption
vd KE and the value of second exponential _e-Kat approache~ ze~o whereas th~
first exponential e-KEt retains some fintte value. At thts time, the equa
It has been shown that at Cmax' when Ka = KE, tmax = 1/KE. Hence,
the above equation further reduces to: tion 10.58 reduces to:
~
C =A e-KEt· ( 10.59)
F Xo
--e --1
(10.55)
vd In log fonn, the above equation is:

~ KE t (1 0.60)
Since FX0 N d represents C0 following i.v. bolus, the maximum plasma log C = log A -
concentration that can be attained after e.v. administration is just 37o/o of 2.303
the maximum level attainable with i. v. bolus in the same dose. If
bioavailability is less than 100%, still lower concentration will be attained.
where C represents the back extrapolated plasma conc~ntration ~alue~. A
plot of log c versus t yields a biexponential curve wtth a te~mal hne~
Elimination Rate Constant : This parameter can be computed from phase having slope - KE/2_-303 (~ig. 10.8). Back extrapolation of thts
the elimination phase of the plasma level time profile. For most drugs straight line to time zero ytelds y-mtercept equal to log A.
administered e. v ., absorption rate is significantly greater than the elimina- log KaFX 0
tion rate i.e. Kat >> KEt. Hence, one can say that e-Kat approaches zero .__ _ _ _ _ _ _ log A= V (K - K )
• c1 a E
much faster than does e-KEt. At such a stage, when absorption is com- ••• Back extrapolated terminal
plete, the change in plasma concentration is dependent only on elimination ~;..~~~~~.~-----portion of curve (log C values)
rate and equation 10.49 reduces to:
• True ptasma concentration
I \
Ka F Xo K ...,___ _ _ values (log C values)
C=
Vd (Ka- KE)
e- Et (10.56)
I\ • _ _ Residual curve (log Cr values)
Transfonning into log fonn, the equation becomes:
(.)
0)

-
0
I \. __,-
( 10.57) I \ slope= -KE 1 2.303

slope= ~
A plot of log C versus t yields a straight line with slope -KE/2.303 \ -K I 2.303
(half-life can then be computed from KE). KE can also be estimated from to lag time ·, a

urinary excretion data (see the section on urinary excretion data).


~~ t
Absorption Rate Constant : It can be calculated by the method of
residuals. The technique is also known as feathering, peeling and Fig. 10.8 Plasma concentration-time profile after oral administration of a
single dose of a drug. The biexponential cu~v~ ha~ been re~olved
stripping. It is commonly used in pharrnacokinetics to resolve a into its two components- absorption and eltmtnatton.
multiexponential curve into its individual components. For a drug that
follows one-:ompartment kinetics and administered e. v., the concentration Subtraction of true plasma concentration values i.e. equation 10.58
of drug in plasma is expressed by a biexponential equation 10.49. from the extrapolated plasma concentration values i.e. equation I 0.59
yields a series of residual concentration values Cr :
Ka F X0
c = e-KEt - e- Kat (1 0.49) (t- C) = Cr = A e- Kat (10.61)
251
250 BIOPHARMACEUTICS AND PHARMACOKINETICS
COMPARTMENT MODELING
·
of amount o f d rug tn
· the body X and the amount of drug. eliminated from
In log fonn, , the equation i$:
the body XE· thus:
Kat (10.63)
log Cr = log A - - - -
2.303
The amount of drug in the body is X = V de· The amount of drug
A plot of log Cr versus t yields a straight line with slope -Ka/2.303 eliminated at any time t can be calculated. as follows:
and Y-intercept log A (Fig. 10.8). Absorption half-life can then be
XE = KE Vd [AUC]ot (10.64)
computed from Ka using the relation 0.693/Ka. Thus, the method of .
residuals enables resolution of the biexponential plasma level-time curve Substitution of values of X and XE in equation 10.63 yields:
int<;> its tWo exponential components. The technique works best when the
XA = V d C + KE V d [AUC]ot (10.65)
difference between Ka and KE is large (KalKE > 3 ). In some instances,
the KE obtained after i.v. bolus of the same drug is very large, much The total amount of drug absorbed into the systemic circulation from
' .
larger than the Ka obtained by the method of residuals (e.g. isoprenaline) time zero to infinity XA00 can be given as:
and if KE!Ka ~ 3, the terminal slope estimates Ka and not KE whereas the XAoo = Vd coo+ KE Vd [AUC]ooo (10.66)
slope of residual line gives KE and not Ka. This is called as flip-flop
phenomenon since the slopes of the two lines have. exchanged their mean-
'
Since at t = oo, coo = O, the above equation reduces to:

zngs. 00 ( 10.67)
XA 00
= KE V d [AUC]o
- Ideally, the e~trapolated and the residual lines intersect each other on '
'
y-axis i.e. at time t = zero and there is no lag in absorption. However, if The fraction of drug absorbed at any time t is given as:
such an intersection occurs at a time greater than zero; it indicates time XA Vd C + KE Vd [AUC]ot
-- -
lag. It is defined as the time difference between drug administration and 00
XA
00
KE V d [AUC]o
start of absorption. It is denoted by symbol to and represents the begin-
ning of absorption process. Lag time should not be confused with onset 1
C + KE [AUC] 0 (10.68)
time. 00
KE [AUC]o
The above method for the estimation of Ka is a curve~ fitting method.
The method is best suited for drugs which are rapidly and completely Percent drug unabsorbed at any time is therefore:
absorbed and follow one-compartment kinetics even when given i.v1• However, XA C + KE [AUC] 0 t
if the absorption of the drug is affected in some way such as GI motility % ARA = 1_ 100 = 1- ] oo 100 (10.69)
XAoo KE [AUC 0
or enzymatic degradation and if the drug shows multicompartment charac-
teristics after i.v. administration (which is true for virtually all drugs), then
Ka computed by curve-fitting method is incorrect even if the drug were
truly absorbed by first-order kinetics. The Ka so obtained is at best,
estimate of first-order disappearance of drug from the GIT rather than of
first-order appearance in the systemic circulation.
slope = -K8 I 2.303
Wagner-Nelson Method for Estimation of Ka -
One of the better alternatives to curve-fitting method in the estimation
of Ka is Wagner-Nelson method. The method involves determination of
Ka from . percent unabsorbed-time plots and does not require the assump-
~~ t
tion of zero- or first-order absorption.
After oral administration of a single dose of a drug, at any given time, Fig. 10.9 Semilog plot of percent ARA versus t
according to Wagner-Nelson method
the amount of drug absorbed into the systemic circulation XA, is the sum
COMPARTMENT MODELING 253
252 BIOPHARMACEUTICS AND PHARMACOKINETICS
URINARY EXCRETION DATA
The method .requires collection of blood samples after a single oral (Disposition Viewed from Urine only)
dose at regular Intervals of time till the entire amount of drug is elimi-
In the absence of plasma level-time data, useful infonnation can still
nated from the body. KE is obtained from log C versus t plot and
be obtained from urine data regarding elimination kinetics of a drug. The
[AUC]ot and [AUC]o00 are o~tained from plots of C versus t. A semilog
method has several advantages in the analysis of a pharmacokinetic system:
~lot of percent u.nabsorbed (1.e. percent ARA) versus t yields a straight
hne. who~e slope IS -Ka/2.303 (Fig.l 0.9). If a regular plot of the same is a 1. The method is useful when there is lack of sufficiently sensitive
straight hne, then absorption is zero-order. analytic techniques to measure concentration of drugs in plasma
with accuracy.
Ka can similarly be estimated from urinary excretion data (see the
2. The method is noninvasive and therefore better subject compli- ·
relevant section). The biggest disadvantage of Wagner-Nelson method is
that it applies only to drugs with one-compartment characteristics. Prob- ance is assured.
lem . a:ises . when a drug that obeys one-compartment model after e. v. 3. Convenience of collecting urine samples in comparison to draw-
admtnistratton shows multicompartment characteristics on i.v. injection. ing of blood periodically.
4. Often, a less sensitive analytic method is required for determining
Effect of Ka and KE on Cmax' tmax and AUC urine drug concentration as compared to plasma concentrations; if
A summary of the influence of changes in Ka at constant KE and of the urine drug concentrations are low, assaying of larger sample
KE at constant Ka on Cmax, tmax and AUC of a drug administered e.v. is volumes is relatively easy.
shown in Table I 0.4.
5. First-order elimination, excretion and absorption rate constants
TABLE 10.4 and fraction excreted unchanged can be computed from such data;
Influence of Ka and KE on Cmax' tmax and AUC first-order metabolism or extrarenal excretion rate constant can
also be calculated subsequently from the difference (KE - Ke) =
Parameters Influence when KE is Influence when Ka is
Affected Constant Constant Km·
6. Direct measurement of bioavailability, both absol~te and relative,
'
Smaller Ka Larger Ka Smaller KE Larger KE is possible without the necessity of fitting the data to a math-
Cmax ~ t t ~ ematical model.
Long Short Long Short 7. When coupled with plasma level-time data, it can also be used to
tmax
estimate renal clearance of unchanged drug according to follow-
AUC No Change No Change t ~
ing equation:
where, t = increase and ~ = decrease. Total amount of drug excreted unchanged
ClR = (10.70)
Apparent Volume of Distribution and Clearance : For a drug that Area under the plasma level-time curve
follows one-compartment kinetics after e. v. administration, V d and CIT If V d is known, total systemic clearance and nonrenal clearance can
~an be co~puted from equation 10.15b and I 0.22b respectively where F
also be calculated.
IS the fractton absorbed into the systemic circulation.
One cannot however compute V d and CIT from l
u ..ine data alone. One
F X0 must also remember that urinary excretion data is not an accurate substi-
Vd = - - - (10.15b)
KE AUC tute for the plasma level data. At best, the data can be employed as a
rough estimate of the pharmacoiGnetic parameters. Moreover, if the drug
F X0 • product provides a very slow drug release or if the drug has a very long
CIT = - - ( 10.22b) biological half-life, the resulting low urinary drug concentration may be
AUC
too dilute to be assessed with accuracy . . In the latter case, i.e. for drugs

COMPARTMENT MODELING 255
254 BIOPHARMACEUTICS AND PHARMACOKINETICS
7. During sampling, the exact time and volume of urine excreted
with long ty2 , urine may have to be collected for several days to account
fQr total drug excreted. should be noted.
I
I
8. An indi\ddual collection peribd should not exceed one biologic
Criteria for Obtaining Valid Urinary Excretion Data half-life of the drug and ideally should be considerably less.
I. A significant amount of drug must be excreted unchanged in the 9. Urine samples must be collected for at least 7 biologic~! half-lives
urine (at least l 0%). in order to ensure collection of more than 99% of excreted drug.
2. The analytical method must be specific for the unchanged drug; 10. Changes in urine pH and urine volume may alter the urinary
'
metabolites should not interfere.' excretion rate.
.;

3. Water-loading should be done by taking 400 ml of water after The urine data can be set as shown in the Table 10.5. Observations
fasting overnight, to promote diuresis and enable collection of include times of urine collection, volumes collected and concentration of
sufficient urine samples. unchanged drug in each sample. These data are treated to derive further
4. Before administration of drug, the bladder must be emptied com- infonnation.
pletely after I hour from ·w ater-loading and the urine sample
taken as blank; the drug should then be administered with 200 ml Determination of KE from Urinary Excretion Data
of water and should be followed by 200 ml given at hourly The first-order elimination (and excretion) rate constants can be com-
intervals for the next 4 hours. puted from urine data by two methods:
5. Volunteers must be instructed to completely empty their bladder I . Rate of excretion method, and
while collecting urine samples. 2. Sigma-minus method.
6. Frequent sampling should be done in order to obtain a good Rate of Excretion Method : The rate of urinary drug excretion
curve. dXuldt is proportional to the amount of drug in the body X and written as:
TABLE 10.5
Urinary Excretion Data following i.v. Bolus of 100 mg of a Drug dXu = Ke X (10.71)
. .
dt
Observation Treatment of Data
where K = first-order urinary excretion rate constant. According to frrst-
Time of Volume Concen- Urine

Mid- Amount Excre- Cumu- Amount

order dis~osition kinetics, X = X0 e- KEt (equation 10.5). Substituting it in
unne of urine tration collect- point of excreted tion rate lative rema1n-
Sam- collect- collected of unch- •
I On unne•
in time (mg/H) amount ing to be above equation yields:
pie •
I On anged interval collec- interval excreted excreted dX
_ u_ = Ke Xo e- KEt (10.72)
drug in tion
t •
unne dt :mg) dXu (mg) Xu00
dt
(hours) (ml) (mcg/ml) (or ~t) t* (or ~Xu) dXu/dt Xut - Xut
where X0 = dose administered (i.v. bolus). Transforming to log fonn the
0 0 .. - - - - - 0 66.7 equation becomes:
1 0-2 140 250 2 1 35.0 17.5 35.0 31.7 dXu KEt
log - - - log Ke X 0 - (10.73)
2 2-4 150 100 2 3 15.0 7.5 50.0 16.7 dt 2.303
3 4-6 90 80 2 5 . 7.2 3.6 57.2 9.5 The above equation states that a semilog plot of rate of excretion
4 6-8 200 I·
20 2 7 4.0 2.0 61.2 5.5 versus time yields a straight line with slope -KE/2.303 (Fig. I 0.1 0). It
5 8 -12 310 . 10 4 10 3.1 0.8 64.3 2.4 must therefore be remembered that the slope of such an excretion rate
- 6 12-24 600 04 12
<!
18 2.4 0.2 66.7 -
versus time plot is related to elimination rate constant KE and --~ot to
excretion rate constant Ke· The excretion rate constant can be obtained
- . ~ - from the Y-intercept (log Ke X 0 ). Elimination half-life and, nonrenal
..
. Xu00
. ..... elimination rate constant can then be computed
.
from KE and Ke·
256 BIOPHARMACEUTICS AND PHARMACOKINETICS
COMPARTMENT MODELING 257

Substitution of equation 10.75 in equation 10.74 and rearrangement


yields:
dXu Xuoo _ Xu = Xuoo e-KEt (10.76)
Iog-
dt
slope = -KE I 2. 303 Converting to logarithms, we get:
t
log (Xu00 - Xu) = log Xu00 - (1 0.77)

where (Xu00 - Xu) = amount remaining to be excreted i.e. ARE at any


--~ t* (midpoint time of urine collection) given time. A semilog plot of ARE versus t yields a straight line with
slope -KE/2.303. The method is, therefore, also called as ARE plot
Fig. l 0.10 Semilog plot of excretion rate versus mid-
point time of urine collection period for method. A disadvantage of this method is that total urine collection has
computing elimination rate constant to be carried out until no unchanged drug can be detected in the urine i.e
after i. v. bolus administration. upto 7 half-lives, which may be tedious .for drugs having long t~.
'
A~ adva~tage of excretion rate method is that for drugs having long The equations until now for computing KE from the urinary excretion
half-IJ:es, unne may be collected for only 3 to 4 half-lives. Moreover, data apply to a drug that fits one-compartment model and given as i.v.
th_ere IS ~o ne~d to collect all urine samples since collection of any two bolus. Similarly, data obtained during constant rate i. v. infusion can be ~
. consecutive unne samples yield points on the rate plot from which a used to evaluate the elimination rate constant. The equation that describes
' straight line can be constructed. the urinary excretion rate of unchanged drug when administered as i.v.
bolus also applies when it is administered as i. v. infusion. Thus:
Sigma-Minus Method : A disadvantage of rate of excretion method
in estimating KE is that fluctuations in the rate of drug elimination are dX
__ u=KeX (10.71)
observed to a high degree and in most instances, the data are so scattered dt
that an estimate of half-life is difficult. These problems can be minimized
by using the alternative approach called as sigma-minus method. For a drug given as i.v. infusion, the amount of drug in the body X is
given by equation (described earlier):
From an earlier equation:
dXu
= Ke Xo e- KEt
X= ~ (I - e-KEt) (10.34)
. (10.72) KE
dt
Substitution of equation 10.34 in equation 10.71 and integration of the
Integration of equation 10.72 yields:
same yields: .

Xu = Ke Xo (I - e- KEt) (10.74) Xu = J<e ~ t - J<e ~ (1 - e-KEt) (10.78)


KE KE KE2
where Xu = cumulative amount of drug excreted unchanged in urine at When the drug has been infused for a period long enough to attain
any tim~ :.;., As time approaches infinity i.e. after 6 to 7 half-lives, the steady-state in the plasma, the tenn e-KEt approaches zero and the above
value e becomes zero and therefore the cumulative amount excreted equation reduces to:
at infinite time Xu 00_ can be given by equation:
I X= Ke~t _Ke~ (10.79)
u KE KE2
{10.75)
A regular plot of cumulative amount of drug excreted Xu versus t
-. yields a curvilinear plot the linear portion of which has a slope KeRJKE-
258 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING 259

Extrapolation of linear segment to time axis yields x-intercept equal to defme KE of the drug. The absorption rate constant K 3 can be estimated·
liKE since when Xu = 0, t= liKE (Fig. 10.11). by the method of residuals using the same data i.e. equation I 0.82.
.
Urinary excretion data after oral administration can also be treated · ...
according to Wagner-Nelson method to calculate K 3 by construction of%
ARA plots. The method requires urine collection for sufficiently long '

time to ensure accurate estimation of KE but need not be collected to. time
infmity. The equation derived to relate o/o ARA with urinary excretion
rate is:
i dXuldt + KE Xu
• o/oARA = I - 1 - - - - - -- 100 (1 0.83) .
KE Xuoo
1/ Ke .

A semilog plot of % ARA versus t yields a straight line with slope


-K3 /2.303.
Fig. 10.11 Regular plot of Xu versus t during Accurate determination of K3 from urinary excretion data is possible
constant rate i. v. infusion only for drugs with slow rate of absorption since for drugs with rapid
Relationships for rate of excretion when the drug is administered e. v. absorption, collection of urine samples at very short intervals of time is
can also be given similarly. Thus: difficult. \
dX
__ u = KX (I0.71)
dt e MULTICOMPARTMENT MODELS
(Delayed Distribution Models)
For a drug given e.v. and absorbed by a frrst-order process, X is given
The one-compartrnent model adequately describes pharrnacokinetics of
as:
many drugs. Instantaneous distribution equilibrium is assumed in such
X= Ka F Xo e- KEt - e-Kat ( 10.48) cases and decline in the amount of drug in the body with time is ex-
(Ka - KE) pressed by an equation with a monoexponential term (i.e. elimination).
Substitution of equation 10.48 in equation 10.71 and integration of' the However, instantaneous distribution is not truly poss~ble for an even larger
same yields: number of drugs and drug disposition is not monoexponential but bi- or
multi-exponential. This is because the body is composed of a heteroge--
Ke Ka F Xo l e- KEt
X = +---- (1 0.80) neous group of tissues each with different degree of blood flow and.
u KE Ka (KE - K3 ) affmity for drug and therefore different rates of equilibration. Ideally, a
true pharmacokinetic model should be the one with a rate constant for
At time infmity, the equation 10.80 reduces to:
each tissue undergoing equilibrium, which is difficult mathematically.
X oo = KeFXo Th~ best approach is therefore to pool together tissues on the basis of
u (10.81)
KE similarity in their distribution characteristics. As for one-compat tlnent
models, drug disposition in multicompar ttnent systems is also assumed to
Substitution/Of equation I 0.81 in equation 10.80 and subsequent rear- occur by first-order. Multicompat ttnent characteristics of a drug- are best
rangement yields: understood by giving it as i.v. bolus and observing the manner in which .

I x oo the plasma concentration declines with time. The number of exponentials
ARE = (X oo - X ) = . u (K e- KEt - KE e- Kat) (10.82)
u u (Ka - KE) a required to describe such..
a plasma level-time profile detetrnines the num-
her of kinetically homogeneous compartments into which a drug will
A semilog plot of (Xu00 - Xu) versus t results in a biexponential curve distribute.
and if K8 > KE, the slope of the tenninal linear portion of the curve ~ill
<'OMPARTMENT MODELING 261
260 BIOPHARMACEUTICS AND PHARMACOKINETICS
cnce of two disposition processes viz. distribution and elimination. These
TWO-COMPARTMENT OPEN MODEL
two processes are not evident to the eyes in a regular arithmetic plot but
The commonest of multi~ompartment models is two compartment model. when a semilog plot of C versus t is made, they can be identified (Fig.
In such a model, the body tissues are broadly classified into 2 categories: 10.12). Initially, the concentration of drug in the central comparbnent
1. Central Compartment or Compartment 1 comprising of blood declines rapidly; this is due to the distribution of drug from the central
and highly perfused tissues like liver, lungs, kidneys, etc. that compartment to the peripheral compartment. The phase during whic~ ·this
equilibrate with the drug rapidly. Elimination usually occurs from occurs is therefore called as the distributive phase. After somet~e, a
this comparttnent. pseudo-distribution equilibrium is achieved between the two c9tn~art­
2. Peripheral or Tissue Compartment or Compartment 2 com- ments following which the subsequent loss of drug from the / central
prising of poorly perfused and slow equilibrating tissues such as compartment is slow and mainly due to elimination. This second, slower
muscles, skin, adipose, etc. and considered as a hybrid of several rate process, is called as the post-distributive or elimination phase. In
functional physiologic units. contrast to the central compartrnent, the drug concentration in the periph-
. . eral compartment frrst increases and reac~es a maximum. This corresponds
Classification of a particular tissue, for example brain, into central or with the distribution phase. Foilowing peak, the drug concentration de-
. -
peripheral compartment depends uQ_on the physicochemical properties of
the drug. A highly lipophilic drug can cross the BBB and brain would
clines which corresponds to the post-distributive phase (Fig. I 0.12).
'
· _then be included in the central compartntent. In contrast, a polar . drug
-- ------ __.-- .... ---- _._, -----
cannot penetr~te the BBB and brain in this case will be a part of periph-
eral comparttnent despite the fact that it is a highly p~rfused organ. ~--------Initial rapid decline
· Depending upon the comparttnent from which the drug is eliminated, distributive phase
c:
the two-compartment model can be categorized into 3 types: 0

1. Two-compatttnent model with elimination from central compart-


---------------
ment.
.,.__ _ _ Slow terminal decline
2. Two-compartment model with elimination from peripheral com- elimination I phase
"
parbnent.
'6,t
~"q/
3~ Two-compartment model with elimination &6m both the compart-
"o.fJ).
men~. ' 0~

In the absence of infortnation, elimination is_assumed to occur exclu- "'~


~,,

sively from central comparttnent.

- -Two~Compartment Open Model--Intravenous _.._. Time


Bolus -Administration
Fig. 10.12 Changes in drug concentration in the central (plasma) and the peripheral
The model can be depicted as shown below with elimination from the compartment after i.v. bolus of a drug that fits two-compartment model.
centr-al COill:pat ttnent.
Let K 12 and K2 1 be the first-order distribution rate constants depicting
1 K12 2 drug transfer between the central and the peripheral com par trnents and let · ·.
. \
Central
'\
Peripheral subscript c and p define central and peripheral compartment r~spectively .
Compart~ent K21 Compartn1ent . The rate of change in drug concentration in the central compartrnent is
Ke given by:

' ~
( 10.84)

After the i. v. bolus of a drug that follows two-comparbnent kinetics,
the decline in plasma concentration is biexponential ·indic~ting the pres-
263

COMPARTMENT MODELING
262 . BIOPHARMACEUTICS AND PHARMACOKINETICS
Xo K21 - a K2t- a ( 10.93)
Extending the relationship X = Vde to the above equation, we have A= =Co

Vc ~-a p-a
dec K21 xp K xc
12 KE xc
- (10.85) K21 - (3 K21 - (3
dt vp vc vc Xo (1 0.94)
B= =Co
Vc a-~ a-~
where Xc and Xp are the amounts of drug in the central and peripheral
compartments respectively and Vc and VP are the apparent volumes of the where C = plasma drug concentration ·immediately after i.v. injection.
0
central and the peripheral compartinent respectively. The rate of chang~ Method of Residuals : The biexponential disposition curve obtained
in drug concentration in the peripheral comparttnent is given by: after i.v. bolus of a drug that fits two compartment model can be ·resolved
dCp ' into its individual exponents by the method of residuals. Rewriting the
dt = K12 Cc- K21 Cp (1 0.86)'
equation 10.92:

C = A e~t + B e-~t (10.92)

(1 0.87) As apparent from the biexponential curve given in Fig. 10.12., the
Vc Vp initial decline due to distribution is more rapid than the tenninal decline
due to elimination i.e. the rate constant a >> B and · hence the term e-«t
Integration of e9uations 10.85 and 10.87 yields equations that describe
approaches zero much faster than does e-13t. Thus, equation 10.92 reduces
the concentration of , ~g in the central and peripheral compartments at
any given time t: · to:
(10.95)
Cc = ~o { K21 - a ) e-at + { K21 - f3 ) e-Pt (10.88) '
c P- a a - f3 In log fonn , the equation becomes:
~ ~t
(10.96)
Xo log C = log B - .
cp = -v ( Kl2 ) e-at + ( Kt2 ) e- Pt 2 303
(I 0.89)
p f3 - .a a- f3 C
where = back extrapolated plasma concentration values. A semilog plot
where X0 = i. v. bolus dose, a and P are hybrid first-order constants for of C versus t yields the tenninal linear phase of the curve having slope
the rapid distribution phase and the slow elimination phase respectively -~/2.303 and when back extrapolated to time zero, yields y-intercept log
which depend entirely upon .the first-order constants K 12 , K 21 and KE. B (Fig. 10.13 .).The t y, for the elimination phase can be obtained from
equation t1; 2 = 0.693/~.
The constants ~12 and K21 that depict reversible transfer of drug
between compartments are called as microconstants or transfer con- Subtraction of extrapolated plasma concentration values. of the elimina-
stants. The mathematical relationships between hybrid and microconstants tion phase (equation 10.95) from the corresponding true plasma concentration

are gtven as: values (equation I0. 92) yields a series of residual concentration values Cr.
~
a + f3 = K12 + K21 + KE (1 0.90) Cr =C - C = A e-at (10.97)
a f3 = K21 KE (10.91)
In log fonn" the equation becomes:
Equation I 0.88 can be written in simplified fonn as: at
( 10.98)
leg Cr = log A - - -
Cc = A e-at + B e-~t (1 0.92) 2.303

Distribution Elimination A semilog plot of Cr versus t yields a straight line with slope -a/2.303
cc = exponent + exponent · and }'-intercept log A (Fig. 10.13).
where A and B are also hybrid constants for the two exponents and can
be resolved graphically by the method of residuals.
264 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING 265

A B
4 - - - - - - - - - - l o g C0 AUC- + (10.103)
(l

The apparent volume of central comparttnent V c is given as:


14-t--------log A
Xo

\ v =
c Co
- (1 0.1 04)
L+-~-------· lOg B
Back extrapolated terminal portion
Apparent volume of peripheral compartment can be obtained from
~--------- ~ equation:
tog C of elimination phase (log C value.s )
~
~--True plasma concentration
values (log C values)
vp = (10.105)

Residual Line (log Cr values) '


The apparent volume of distribution at steady-state or equilibrium can
. slope = -~ I 2.303 now be defined as:
•\ slope = -a I 2.303 (10.106)

~~ t .It is also given as:


Fig. 10.13 Resolution of biexponential plasma concentration-time curve by V d,area = (I 0.1 07)
the method of residuals for a drug that follows two-compartment ~ AUC
kinetics on i.v. bolus administration.
Total systemic clearance is given as:
Assessment of Pharmacokinetic Parameters : All the parameters of ( 10.1 08)
equation 10.92 can be resolved by the method of residuals as described
above. Other parameters of the model viz. K12, K2 1, KE, etc. can now be The phannacokinetic parameters can also be calculated by using uri-
derived by proper substitution of these values. nary excretion data:
dX 0
C0 =A+ B (10.99) --=KeVc (I O.I 09)
dt
-
KE-
a BCo ( 10.1 00) An equation identical to equation 10.92 can be derived for rate of
AB+Ba excretion of unchanged drug in urine:
_ A B (B - a)2
K 12 - (10.101) dXu = Ke A e-at + Ke B .e-~ 1 (I 0.11 0)
C 0 (A B+ B a) dt
A~+Ba The above equation can be resolved into individual exponents by the
K21 = (10.102) method of residuals as described for plasma concentration-time data.
Co
Renal clearance is given as:
It must be noted that for two-compartment model, KE is the rate
constant for elimination of drug from the central compartment and B is the ClR - Ke Vc (10.111)
rate constant for elimin~tion from the entire body. Overall eli~ination tYl
should therefore be calculated from B. · Two-Compartment Open Model
~Intravenous Infusion
Area under the plasma concentration-time curve can be obtained by
The model can be depicted as shown below with elimination from the
the following equation: ·
central comparttnent.
266 BIOPHARMACEUTICS AND PHARMACOKINFTICS
COMPARTMENT MODELING 267
Ro 1 K12 2 The plasma concentration at any time t is given by
-
eq~ation:
...,.
Central \.
Peripheral
Compartment I' K21 Compartment C = N e-Kat + L e-at+ M e-Pt (10.116)

KE Absorption Distribution Elimination
.,. C = + +
' exponent exponent exponent
The plasma or central compartment concentration of a drug that fits
where K 3 ,- a and P have usual meanings. L, M and N are coefficients.
~wo~com~art~ent. model when administered as constant rate (zero-order)
1. v. Infusion, 1s given . by equation: I
The 3 exponents can be resolved by stepwise application of method of
residuals assuming K3 > a > P as shown in Fig. I 0.14. The various
Ro
C= - - - pharmacokinetic parameters can then be estimated.
Vc KE
(10.112)

At steady-state (i.e. at time infinity), the second and the third tenn in
the bracket becomes zero and the equation reduces to: log N

(10.113)
\• True plasma concentration
curve (log C values)
log L Back extrapolated distri~ution
+=
Now VeKE = Vdl3. Substituting this in equation 10 . 113 , we get : curve (log C - C) val• .. es
- ~ Back extra:;olated elimination
logM~
(10.114) ""_._,a.__~-~-~ curve (log C values)
~.....~~- _ _ _ _ First residual curve (log Cr1 values)
. The loading dose Xo.L to obtain C 55 immediately at the start of infu- \
sion can be calculated from equation: Second residual liAe

~•
~-
(absorption) log Cr 2 values

\ stope = -P I 2.303
Xo.L = Css Vc = - - (10.115) tog C •
KE
.slope=
Two-Compartment Open Model \ -a /2.303
-Extravascu Ia r . ~d ministration \stope= -Ka /2.303
First-Order Absorption
The model can be depicted as follows: ---.) t

Ka 1
Fig. 10.14 Seinilog plot of C versus t of a drug with two-compartment
..
r
Central
K12
~
2
characteristics when administered extravascularly. The various
I' Peripheral
Compartment K21 Compartment
exponents have been resolved by the method of residuals.

KE Besides the method of residuals, Ka can also be estimated by Loo-


.., ~ Riegelman method for a drug that follows two-compartntent characteristics .
. ~or a drug that enters the body by a first-order absorption process and This method is in contrast to the Wagner-Nelson method for determination
dtstnbuted according to two compartment model, the rate of change in of Ka of a drug with one-compartment characteristics. The Loo-Riegelman
drug concent~ation in the central compartment is described by 3 expo~ents method requires plasma drug concentration-time data both after oral and
. an ~b.sorptton exponent, and the two usual exponents that describe drug i. v. administration of the drug to the same subject at different times in
dtsposttton. .... order to obtain all the necessary kinetic constants. Despite its complexity,
268 BIOPHARMACEUTICS AND PHARMACOKINETICS 269
COMPARTMENT MODELING
the method can be applied to drugs that distribute in any number of 18. What are the applications and limitations of method of residuals?
comparttnents.
19. What is flip-flop phenomenon and when is it observed?
20. Under what circumstances is the value of Ka computed from method of
QUESTIONS
residuals incorrect?
I. In one-compartment open model, what do you infer from plasma being 21. What are the m~rits and demerits of Wagner-Nelson method in computing
called as reference compartment? Ka?
2. In compartment modeling, what does the term open mean? 22. What is th~ influence of Ka and KE on Cmax' tmax and AUC?
3. Disposition of a drug that follows one-compartment kinetics is a monoexponential 23. What criteria are necessary for obtaining a valid urinary excretion data?
process. Explain. What are the advantages and disadvantages of such a method in assessment
4. Why do first-order rate equations require logarithmic transformations? of pharmacokinetic parameters?
5. With examples, explain what you understand by primary and secondary 24. What are the two methods for calculating KE from urinary excretion data?
parameters. Compare their merits and demerits.
6. The expression Vd = XJC 0 can only be used to estimate the. apparent 25. Why are urine samples required to be collected for at least 7 biolog~cal h~lf­
volume of distribution of a drug that follows one-compartment kinetics lives when using sigma-minus method whereas only two consecuttve unne
when administered as i. v. bolus. Why? samples are sufficient in case of rate of excretion method?
7. Clearance is a more important parameter than half-life or elimination rate 26. Determination of Ka using urinary excretion data is not suitable for rapidly
constant in expressing elimination characteristics of a drug. Explain. absorbed drugs. Why?
8. Define clearance, total body clearance and organ clearance. What are the 27. How are body tissues classified in a two-compartment .model? Depending
advantages of expressing clearance at an individual organ level? upon the drug's lipophilicity, in which compartment brain .should be in-

9. Define and explain extraction ratio. How is it related to oral availability of cluded?
a drug? What is the influence of blood flow rate and protein binding on 28. What is the cause of initial rapid decline and of subsequent slower decline
total clearance of drugs with high and those with low ER values? in plasma levels after administration of a drug that follows two-compartment
kinetics?
10. What are the rate-limiting steps in the hepatic clearance of drugs? Based on
29. The pariuneter KE has different meanings for one- and two-compartment
ERH values, to which drugs do they apply?
models. Explain.
11. What are the advantages of administering a drug by constant rate i. v.
infusion? 30. If the plasma concentration of viomyc,in after i.v. bolus administration was
found to be 10.0 and 5.5 mcg/ml at 2 and 4 hours respectively, assuming
12. Compute mathematically the number of half-lives required to attain 90% of
one-compartment kinetics, calculate:
steady-state concentration after i. v. infusion (Hint : use equation I 0.41 ).
a. the half-life of the drug
13. Prove mathematically that when an i. v. loading dose is followed immedi-
ately by a constant rate infusion, the plasma concentration remains steady as Answ~r : 2.3 hours.
long as the infusion ·is continued. b. the concentration of drug in plasma at time zero
14. On what parameters does the time to reach steady-state depend? Answer : 18.2 meg/mi.
\
15. How can the steady-state be attained rapidly? When is it advisable? Give c. the V d if dose administered was 30_0 mg
expressions for calculating such doses for a drug that fits one-compartment
Answer : 16.5 liters.
model.
d. the total systemic clearance
16. Compare the absorption characteristics of drugs absorbed by zero-order with
a
those absorbed by first-order process after e~. administration. Answer : 82.2 ml/min.
17. Show that after e.v. administration. the Cmax that can be attained is no more e. the renal clearance if fraction excreted unchanged in urine is 0.8
than 37°/o of maximum level attainable with the same dose given as i. v. Answer : 65.8 ml/min. , ·
bolus. Why is this so?
270 BIOPHARMACEUTICS AND PHARMACOKINETICS COMPARTMENT MODELING 271
31. ~ 70 Kg patient is to be given oubain by i. v. infusion. The drug has a half- e. When should the next dose be administered if the drug becomes ineffective
life of 22 .hou:s, apparent Vd 15.7 liters and the desired steady-state plasma when the plas.ma level falls below 50 mcg/ml?
c~ncentrat1on Is 0.0002 meg/mi. Assuming one-compartment kinetics, cal-
Answer : after 1.2 hours.
culate:
f. What will be -the therapeutic index of the drug if the therapeutic range is 50
a. the time required to reach 90% of C 55
to 500 mcg/ml?
Answer : 73 hours.
Answer : TI = 10.
b. the infusion rate to achieve the desired C 55
g. How much dose should be administered to attain an instantaneous plasma
Answer : 0.1 meg/hour. concentration of 500 mcg/ml?
c. the loading dose to attain C 55 rapidly Answer : 7000 mg.
Answer : 3.14 meg. • h. For how long will the plasma level lie in the therapeutic window if the
d. the concentration of drug in plasma after 48 hours from the start of infusion above dose is given as i.v. bolus?

Answer : 0.00014 mcg/ml and when loading dose is given, 0.00018 meg/mi. Answer : 2.6 hours.

32. The equation that best fits the pharmacokinetics of paracetamol after onil 34. The amount of drug excreted in urine after an i. v. dose of 400 mg of
administration of 500 mg dose is: norfloxacin was as follows:
c = 1.18 (e- 0.24t _ e- l.6t) t (hours) Xu (mg) dX,jdt (mg/H) t*

Assuming one-compartment kinetics, determine- 0 0 \


'
a. peak time 2 49.05

Answer : 1.4 hours. 4 80.0

b. peak plasma concentration After completing the table and using rate of excretion method, determine-
Answer : 0. 717 meg/mi. a. the elimination rate constant and half-life of th~ drug.
c. elimination half-life of the drug Answer : KE = 0.2303/hour and ty, = 3 hours.
Answer : 2. 88 hours. b. urinary exr,retion rate constant
d. apparent V d if fraction bioavailable is 0.4 Answer : Kc = 0.077/hour.
Answer : 200 liters. c. fraction of drug excreted unchanged in urine
e. concentration of drug in plasma after 3 hours of administration Answer : 33.4% '

Answer : 0.565 meg/mi. d. whether the drug can be used for urinary tract infections?

33. The equation that best fits the plasma level time curve of azlocillin after an 35. The half-life of propranolol in a 60 Kg patient is 4 hours and vd is
i.v. bolus dose of 2000 mg (assuming one-compartment kinetics) is: 5.5· liter/Kg.
C = 143 e-0· 87t a. Determine the total systemic clearance of the drug
a. What is the apparent V d? Answer : CIT = 953 ml/min.
Answer : 14 liters. b. What will be its renal clearance if fraction excreted unchanged in urine is
0.047?
b. Whar is the elimination tih of the drug?
I Answer : CIR = 44.8 ml/min.
~:~~wer : 0.8 Hours. •
I

c. Comment on the mechanism of renal excretion of drug.


~. What will be the plasma drug concentration after 6 hours?
d. If the drug is eliminated only by hepatic and renal routes, what will be the
Answer : 0. 77 meg/mi.
'
hepatic extraction ratio if blood flow to the liver is 1.5 liter/min?
d. How much of the drug will be left in the body after 6 hours?
Answer : ERH = 0.6054.
Answer : I 0.8 mg .
....
272 BIOPHARMACEUTICS AND PHARMACOKINETICS

e. If the blood flow rate to the liver reduces to 0.8 liter/min in situations of
CCF, what will be the new hepatic and total systemic clearance values?
Answer : CJH = 484.4 mllmin and CIT= 529.2 ml/min.
f. What is the % decrease in the overall clearance of the drug? 11
Answer : 44.5%.
36. Following a 650 mg i.v. bolus dose of a drug to a 65 Kg subject, the
plasma drug concentration was found to decline biexponentially. The equa-
Nonlinear Pharmacokinetics
''

tion that best described the drug kinetics was:


C = 67 e- 14t + 33 e-3t
In most cases, at therapeutic doses, the change in the amount of drug
where t is in hours and C is in meg/mi. Calculate the following:
in the body or the change in its plasma concentration due to absorption,
a. The volume of the central compartment. distribution, binding, metabolism or excretion, is proportional to its dose,
Answer : Vc = 6.5 liters. whether administered as a single dose or as multiple doses. In such
b. The volume of the peripheral compartment. situations, the rate processes are ~aid to follow first-order or linear kinetics
Answer : VP = 3.95 liters. and all semilog plots of C versus t for different doses when corrected for
dose administered, a~e superimposable. This is called as principle .o f
c. The apparent Vd at steady-state.
superposition. The important phannacokinetic parameters viz. F, K8 ,
Answer : Vd ss = I 0.45 liters. J<E, Vd' CIR .and CIH which describe the time-course of a drug in the body
'
--- - - - '

d. Volume of distribution by area. remain unattected by the dose I.e. the phannacokinetics is dose independent.
Answer: Vdarea = 13.7 liters. In some instances, the rate process of a drug's ADME are dependent
'
e. The microconstants K12 and K21 upon carrier or enzymes that are substrate specific, have definite capaci-
Answer : K12 = 4.035/hour and K21 = 6.63/hour. ties, and susceptible to saturation at high drug concentration. In such
f. The elimination rate constant for the disposal of drug from the central
cases, an essentially first-order kinetics transform into a mixture of first-
compartment. order and zero-order rate processes and the pharmacokinetic parameters
change with the size of.
the administered
.
dose. The phatmacokinetics of
Answer : KE = 6.33/hour.
such drugs are said to be dose-dependent. Other terms synonymous with
g. The overall elimination half-life it are mixed-order, nonlinear and capacity-limited kinetics. D~ugs ex-
Answer : t'h = 0.231 hours. hibiting such a kinetic proftleare sources of variability in phannacoiogic ·
b. The total systemic clearance of the drug (use Vd area for calculation). response.
' - - ....... -·
-~ ~
.,., .. -- - ....
, ,_.

Answer : CIT = 686.2 ml/min. There are several tests to detect non/inearity in pnannacokinetics but
i. The plasma level of the drug after 30 minutes of i. v. dose. the simplest ones are:
Answer : 7.42 meg/mi. 1. Detennination of steady-state plasma concentration at differ~nt
j. The infusion rate if the drug is to be given as constant rate i. v. infusion doses. If the steady-state co.ncentrations are directly proportional -
(desired Css is 20 mcg/ml). to the dose, ·then linearity in the kinetics exist. Such a proportion-
ality is not observable when there is nonlinearity.

Answer : Ro = 823 mg/hour.
k. The loading dose. to attain the Css rapidly.
'' 2. Detettnination of some of the important phannacokinetic param-
i
eters such as fraction bioavailable, elimination half-life or total
Answer : X0 L = 130 mg.
' '

• systemic clearance at different doses of the drug. Any change in I


I
these parameters which are usually constant, is indicative of
nonlinearity.
273
276 BIOPHARMACEUTICS AND PHARMACOKINETICS
NONLINEAR PHARMACOKINETICS 277
Three situations can now be considered depending upon the values of
Km and C: The above equation is identical to the one that describes a zero-order
1. When Km = C : Under this situation, the equation 11.1. reduces process i.e. the rate process occurs at a constant rate V max and is indepen-
to: dent of drug concentration e.g. metabolism of ethanol.
dC Vmax
( 11.2) Estimation of Km and V max
dt 2 The parameters of capacity-limited processes like metabolism, renal
tubular secretion and biliary excretion can be easily defmed by assuming
i.e. the rate of process is equal to OQ.e-half its maximum rate (Fig. 11.1 ). one-compartment kinetics for the drug and that elimination involves only
- ---- -
a single capacity-limited process.
4 ---------------
The parameters Km and Vmax can be assessed from the plasma con-
Zero-order rate at high doses centration-time data collected after i.v. bolus administration of a drug with
nonlinear elimination characteristics.
_ _ Mixed-order rate at
intermediate doses Rewriting equation 11.1.
dC Ymax C
de
-
(11.1)
-dt dt Km + C
.__-+-_First-order rate at low doses
t Integration of above equation followed by conversion to log base 10
yields:
(C 0 - C) Vmax t
log C = log C0 + - ( 11.5)
2.303 Km 2.303 Km
Fig. 11.1 A plot ofMichaelis-Menten equation (elimination rate dC/dt versus
A semilog plot of C versus t yields a curve with a tenninal linear
concentration C). Initially, the rate increases linearly (first-order)
with concentration, becomes mixed-order at higher concentration portion having slope - V max12.303Km and when back extrapolated to time
and then reaches maximum (Vmax) beyond which it proceeds at a
constant rate (zero-order).
zero gives Y-intercept log Co (see Fig. 11.2). The equation that describes
this line is:
~ Ymax t
2. When Km >> C : Here, Km + C = Km and the equation 11.1. log C = log C0 - - --- ( }}r.6)
reduces to: 2.303 Km
dC Vmax C ~
---- (11.3) log C 0
dt Km
'
The above equation is identical to the one that describes frrst-order log 'c;; ,
elimination of a drug where Vma!Km = KE. This means that the drug
concentration in the body that results from usual dosage regimens of most ''
drugs is well below ,the Km of the elimination process with certain excep- log C
tions such as phenytoin and alcohol.

3. When Km << C : Under this condition, Km + C = C and the r Terminal linear portion of
the curve with slope= -Vmax/2.303 Km
equation 11.1 will become:
dC
- - vmax (11.4)
.. dt - -+• t

Fig. 11.2 Sen1ilog plot of a drug gi,·en as i.v. bolus \\·ith nonlinear
clirnination and that fits one-compartment kinetics.
279
278 BIOPHARMACEUTICS AND PHARMACOKINETICS NONLINEAR PHARMACOKINETICS

At low plasma concentrations, equations 11.5 and 11.6 are identical. At steady-state, the dosing rate equals rate. of decline i? plasma d~g
Equating the two and simplifying further, we get: concentration and if the decline (elimination) 1s due to a smgle capacity-
~ limited process (for e.g. metabolism), then;
(C0 - C) C0
---=log (11.7) DR= V max Css (11.12)
2.303 Km C0
Km + Css
Km can thus be obtained from above equation. V max can be computed
A plot of Css versus DR yields a typical hockey-stick shaped curve as
by substituting the value of Km in the slope value.
shown in Fig. 11.3.
An alternative approach of estimating Vmax and Km is detennining the l
rate of change of plasma drug concentration at different times and using I
the reciprocal of the equation 11.1. Thus: . I
I
l
1 Km 1
----+ (11.8) I
dC/dt Ymax Cm Ymax •
l
I
where Cm = plasma=concentration at midpoint of the sampling interval. A I
double reciprocal plot or the Lineweaver-Burk plot of 1/(dC/dt) versus 1/ T l
I
Cm of the above equation yields a straight line with slope = Km/Vmax and Km I
y-intercept = 1/Vmax·
-------- I
I
1
' : Vmax /2 I
Vmax
A disadvantage of Lineweaver-Burk plot is that the points are clus-
tered. More reliable plots in which the points are uniformly scattered are ___..... DR (irl' mg/hour or mg/day)
Hanes-Woolf plot (equation 11.9) and Woolf-Augustinsson-Hofstee plot
Fig. 11.3 Curve for a drug \Vith nonlinear kinetics obtained by plotting
(equation 11. IO).
the steady-state concentration versus dosing rates.
Cm Km Cm
--+-- ( 11. 9)
dC/dt V max Vmax To c.'efine the characteristics of the curve with a reasonable d~gree of
,
accuracy, several measurements must be made at steady-state dunng .dos-
dC dC/dt Km age with different doses.
- = Vmax - ( 11.1 0)
dt · Cm Practicaily, one can graphically compute Knl and Vmax in 3 ways:
The above equations are rearrangements of equation 11.8. Equation 1. Lineweaver-Burk Plot : Taking reciprocal of equation 11.12, we
11.9 is used to plot Cm/(dC/dt) versus Cm and equation 11.10 to plot get:
dC/dt versus (dC/dt)/Cm. The parameters Km and Vmax can be computed 1 Knl 1 (11.13)
- - ---~ +--
from the slopes and y-intercepts of the two plots. DR V max C ss V n1ax
Km and V max from Steady-State Concentration A plot ot 1/DR versus I /C 55 yields a straight line with slope KmN max
When a drug is administered as a constant rate i.v. infusion or in a and y-intercept 1/Vmax·
multiple dose regimen, the steady-st~te concentration C55 is given in terms 2. Direct Linear Plot : Here~ the graph is conside:ed as sh~wn 'in
of dosing rate DR as: Fig. 11.4. A pair of c ss viz. c ss.l and Css. 2 obtained wtth two dtfferent
(11.11) do~ing rates DR 1 and DR2 are plotted. The p.oints ~s~.l and D~ ~ ~re
joined to fonn a line and a second line ;s obtame~ s1m1larly by JOmm.g
where DR = Ro when the drug is administered as zero-order i.v. infusion
and it is equal to FX0 /-r when administered as multiple oral dosage Css.2 an d DR 2· The point \\'here these t\vo lines I!ltersect
.
each other ts
extrapolated on DR axis to obtain Vn1a" and on x-axts to get Kn1·
regimen (F is fraction bioavailable, X0 is oral dose and t is dosing
interval).
\
280
BIOPHARMACEUTICS AND PHARMACOKINETICS
NONLINEAR PHARMACOKINETICS 281

There are several limitations of Km and V max estimated by assuming
vmax one-compartment system and a single capacity-limited process. More
DR
complex equations will result and the computed Km and V max will usually

i 0~1
f

'
I
be larger when:
1. The drug is eliminated by more than one capacity-limited process.
I
f
f
2. The drug exhibits parallel capacity-limited and first-order elimina-
I tion processes.
I

'f
I 3. The drug follows multicompartment kinetics.
Css. 1 Css. 2 '
:Km • .
However, Km and V max obtained under such circumstances have little
- ·- . -- - ··-
practical applications in dosage calculations.
Css • 0 ~ Km
. Drugs that behave nonlinearly within the therapeutic range (for ex-
Fig. 11.4 Direct line~ plot for e: timation of Km and Vmax at steady-state ample, phenytoin shows saturable metabolism) yield less predict~ble results
concentrations of a drug given at different dosing rates.
in drug therapy and possess greater potential in precipitating toxic effects.
3. !he thir~ graphical method of estimating Km and Vmax involves
rearrangmg equation 11.12. to yield: .
QUESTIONS

DR = Vmax- Km DR l. Define dose-dependent kinetics. Quote simple tests by which it can be


(11.14)
Css· detected in a rate process.

. A plot of DR versus DRIC55 yields a straight line with slope -K and 2. Why are drugs that show nonlinearity in pharmacokinetics considered sources
Y-mtercept V max· m of variability in pharmacologic response?
3. What processes of drug ADME are known to show nonlinearity? G ive
Km and ':'max can also be calculated numerically by setting up simulta- examples.
neous equations as shown below:
4. When administered at high doses, how does the pharmacokinetic parameters-t~,
DR! = Vmax Css,J V d, Cmax, etc. change for drugs known to undergo capacity-limited elimina-
( 11.15) tion?
Km + Css 1
' 5. What are the limitations in calculating Km and V max by assuming one-
DR = v c
max ss,2 compartment model and a single capacity-limited process?
2 (II. 16)
1
Km + Css,2 6. Using the following data of a drug having Km of 0.2 mcg/ml and V max of
• 1.0 mcg/ml/hour, determine the concentrations at which the drug shows
Combination of the above two equations yields
first-order, zero-order and mixed-order metabolic rates.

Km = DR1- DR2 Concentration (mcg/ml) 0.02 0 .2 20 .0


( 11.17)
DR1 DR2 Metabolic Rate (mcg/ml/H) 0 .1 0 .5 1.0
Css,l Css,2
7 .. Theophylline was administered to a patient at dosing rates of 600 mg/day
Aft~r _having computed Km, its subsequent substitution in any one of and 1200 mg/day and the respective steady ..state concentrations were · found
the two simultaneous equations will yield V . to be 9.8 mg/L and 28.6 mg/1. Find Km and V max · Determine .the dosing
max·
. It has been observed ~hat Km is much less variable than Vmax· Hence, rate to achieve a C 55 of 15 mg/ 1. "'
tf mean. Km for a drug ts known from an earlier study, then instead of Answer : Km = 31 . 14 mg/1, V max = 2507 mg/day and DR = 815 mg/day.
tw~, a smgle measurement of Css at any given dosing rate is sufficient to
compute V max·
BIOA VAILABILITY AND BIOEQUIV ALENCE 283

the administered dose. The amount of drug that reaches the systemic
circulation (i.e. extent of absorption) is called as systemic availability or
simply availability. The term bioavailable fraction F, refers to the
12 fraction of administered dose. that enters the systemic circulation.
Bioavailable dose
F= - - - - - - - ( 12.1)
Bioavailability Administered dose

and Bioequivalence Objectives of Bioavailability Studies


Bioavailability studies are important in the·-
1. Primary stages of development of a suitable dosage form for a
The therapeutic effectiveness of a drug depends upon the ability of the new drug entity.
dosage form to deliver the medicament to its site of action at a rate and 2. Determination of influence of excipients, patient related factors
amount sufficient to elicit the desired phannacologic response. This and possible interaction with other drugs on the efficiency of
attribute of the dosage fonn is referred to as physiologic availability, absorption.
biologic availability or simply bioavailability. For most drugs, the 3. Development of new formulations of the existing drugs.
pharmacologic response car be related directly. to the \plasma levels. Thus,
4. Control of quality of a drug product during the early stages of
the term bioavailability is defined as the rate and extent (amount) of
marketing in order _to determine the influence of processing fac-
absorption of unchanged drug from its dosage form. It is an absolute
tors, storage and stability on drug absorption.
term. The rate or rapidity with which a drug is absorbed is an important
consideration when a rapid onset of action is desired as in the treatment of
acute conditions such as asthma attack, pain, etc. A slower absorption · CONSIDERATIONS IN BIOAVAILABILITY STUDY DESIGN
.~ate is however desired when the aim is to prolong the duration of action Bioav r~ ilability Absolute versus Relative
or to avoid the adverse effects. On the other hand, extent of absorption is When the systemic availability of a drug administered orally is deter-
of special significance in the treatment of chronic conditions like hyper- mined in comparison to its intravenous administration, it is called as
tension, epilepsy, etc.
absolute bioavailability. It is denoted by symbol F. Its detertnination is
If the size of the dose to be administered is same, theQ bioavailability used to characterize a drug's inherent absorption properties from the e. v.
of a drug from its dosage form depends upon 3 major factors: site. Intravenous dose is selected as a standard because the drug is
L Pham1aceutic factors related to physicochemical properties of the administered directly into the systemic circulation ( 100 % bioavailability)
drug and characteristics of the dosage form and avoids absorption step. Intramuscular dose can also be taken as a
2. Patient related factors
standard if the drug is poorly water soluble. An oral solution as reference
standard has also been used in certain cases. However, there are several
3. Route of administration drawbacks of using oral solution as a standard instead of an i.v. dose-
The former two factors have been dealt with comprehensively in 1. Limits the pharmacokinetic treatment to one-compartment model
chapter 2 on Absorption of Drugs. The influence of route of administra- only; . one cannot apply the most applicable two-compartment
tion on drug's bioavailability is generally in the following order : parenteral kinetics to the data and all pharmacokinetic parameters cannot be
.> oral > rectal :?> topical with few exceptions. Within the parenteral assessed.
route. intravenous injection of a drug results in 1OOo/o bioavailability as
2. Differentiation between the fraction of dose unabsorbed and that
the absorption process is bypassed. However, for reasons of stability and
metabolized is difficult. '
convenience, most drugs are administered orally. In such cases, the close
available to the patient. called as the bioavailable dose, is often less than 3. If the rate of oral absorption is not sufficiently greater than the
rate of elimination, the true elimination rate constant cannot be
282
computed.

-
BIOA VAILABILITY AND BIOEQUIV ALENCE 285
284 BIOPHARMACEUTICS AND PHARMACOKH' 'TICS
Human Volunteers Healthy Subjects versus Patients
At best, when oral solution is used in conjunction with i.v. ro:tte, one Ideally, the bioavailability study should be carried out in patients for
can distinguish the dissolution rate limitation in drug absorption from whom the drug is intended to be used because of the apparent advan-
solid dosage fot n1s. tages-
When the systemic availability of a drug after oral administration is ,1. The patient will be benefited from the study.
compared with that of ~n oral standard of the same drug (such as an 2. Reflects better the therapeutic efficacy of a drug.
aqueous or nonaqueous solution or a suspension), it is referred to· as
3. Drug absorption pattern in disease states can be evaluated.
relative bioavailability. It is denoted by symbol Fr. In contrast to
4. Avoids the ethical quandary of administering drugs to healthy
absolute bioavailability, it is used to characterize absorption of a drug
from its fonnulation. F and F r are generally expressed in percentages. subjects.
Patients are generally preferred in multiple dose bioavailability studies.
Single Dose versus Multiple Dose Studies The drawbacks of using patients as volunteers are equally large disease,
The dose to be administered for a bioavailability study is determined other drugs, physiologic changes, etc. may modify the drug absorption
from preliminary clinical experiments. Single dose bioavailability studies pattern. Stringent study conditions such as fasting stat~ ~equired to be
are very common. They are easy, offer less exposure to drugs and are followed by the subject is also difficult. In short, estabhshtng a stand~rd
less tedious. However, it is difficult to predict the steady-state characteris- set of conditions necessary for a bioavailability study is difficult with
tics of a drug and intersubject variability with such studies. On the other patients as volunteers. Such studies are therefore usually perfonned in
hand, multiple dose study is difficult to control (poor subject compliance), young (20 to 40 years), healthy, male adult volunteers (body w~i~ht
~~poses the subject to more drug and is highly tedious and time consum- within a narrow range; ± 10% ), under restricted dietary and fiXed activity
ing. Nevertheless, such a study has several advantages- conditions. Female volunteers are used only when drugs such as oral
!. More accurately reflects the manner in which the drug should be contraceptives are to be· tested. The number of subjects to be selected
used. depends upon the extent of intersubject variability but should be kept to a
2. Easy to predict the peak and valley characteristics of the drug minimum required to obtain a reliable data. The consent of volunteers
since the bioavailability is determined at steady-state. must be obtained and they must be informed about the importance of the
study, conditions to be followed during the study and possible hazards if
3. Requires collection of fewer blood samples.
any, prior to starting the study. Medical examination should be perfonned
4. The drug blood levels are higher due to cumulative effect which in order to exclude subjects with any kind of abnonnality or disease. The
I

makes its determination possible even by the less sensitive ana- volunteers must be instructed to abstain from any medication for at least a
lytic methods . week and to fast overnight prior to and for a minimum of 4 hours after
5. Can be ethically performed in patients because of the therapeutic dosing. The volume and type of fluid and the standard diet to be taken
benefit to the patient. must also be specified. Drug washout period for a minimum of ten
6. Small intersubject variability is observed. in such a study which biological half-lives must be allowed for between any two studies in the

allows use of fewer subjects. same subject.
7. Better evaluation of the performance of a controlled release for-
• Measurement of Bioavailability
mutation is possible. The methods useful in quantitative evaluation of bioavailability can be
.8. Nonlinearity in pharmacokinetics, if present, can be easily de- broadly divided into two categories pharmacokinetic methods and
tected. pharmacodynamic methods.
In multiple dose study, one must ensure that the steady-state has been
I. Pharmacokinetic Methods
reached . For this, the drug should be administered for 5 to 6 elimination
These are very widely used and based on the assumption that the
half-lives before collecting the blood samples.
pharmacokinetic profile reflects the therapeutic effectiveness of a drug.
286 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOA V AI LABILITY AND BIOEQUIV ALENCE 287
Thus, these are indirect methods.. The two major phattnacokinetic meth- The extent of bioavailability can be determined by following equa-
ods are: tions:
1. Plasma level-time studies. [AUC]oral Div
F= (12.2)
2. Urinary excretion studies. [AUC]iv Doral
II. Pharmacodynamic IVfethods [AUChest Dstd
Fr=--- - - · - ( 12.3)
These methods are complementary to phannacokinetic approaches and
[AUC]std Dtest
involve direct measurement of drug effect on a (patho)physiologic pro-
cess as a function of time. The two pharmacodynamic methods involve where D stands for dose administered and subscripts iv and oral indica~es
detennination of bioavailability from: the route of administration. Subscripts test and std indicate the test and
- the standard doses of the same drug to determine relative availability.
1. Acute phannacologic response. ..
The rate of absorption can be computed from one of the several methods
2. Therapeutic response. discussed in chapter 10.
Plasma Level Time Studies Dose Dose Dose Dose Dose Dose
Unless detennination of plasma drug concentration is difficult or im- ~ ~ ~ ~ ~- t ~
possible, it is the most reliable method and method of choice in comparison
to urine data. . The method is based on the assumption that two dosage
fonns that exhibit superimposable plasma level-time profiles in a group of
(.) Css. max
subjects should result in identical therapeutic activity.
c
With single dose study, the method requires collection of serial blood
samples for a period of 2 to 3 biological half-lives after drug administra-
--
·-0
~
Steady-state
level
~ _Css. min _
tion, their analysis for drug concentration and making a plot of concentration c:
0
versus corresponding time of sample collection to obtain the plasma level- (.)
C'O
time profile. With i. v. dose, sampling should start within 5 minutes of E
drug administration and subsequent samples taken at 15 minute intervals. -
f/)
C'O ~~ii-----+-- Area under the
-
a. ,,,,,, curve for one
To adequately describe the disposition phase, at least 3 sample. points
dosing interval
should be taken if the drug follows one-compartment kinetics and 5 to 6 T at steady-state
points if it fits two-compartment model. For oral dose~ at least 3 points
should be taken on the ascending part of the curve for accurate detennina-
--+) Dosing interval -r
tion of :Ka. The points for disposition or descending phase of the curve
must be taken in a manner similar to that for i.v. dose. Fig. 12.1 Determination of AUC and Css,max on multiple dosing upto steady-state
The 3 paraq1eters of plasn1a level-time studies which are considered With multiple dose· study, the method involves drug administration for
important for detem1ining bioavailability are: at least ~ biological half-lives with a dosing interval equal to or greater
'
1
1. Cmax : The peak plasma concentration that gives an · indication than the biological half-life (i.e. administration of at least 5 doses) to
whether the drug is sufficiently absorbed systemically to provide reach the steady-state. A blood sample should be taken at the end of
a therapeutic response. previous dosing interval and 8 to 10 samples after the administration of
2. tmax : The .peak time that" . gives an indication of the rate of next dose. The extent of bioavailability is given as:
absorption.
[AUCltest Dstd 'ttest
3. AUC : The area under ~tie plasma level-time curve that gives a Fr = - - - - -· (12.4)
[AUClstd .Dtest 'tstd
measure of the extent of ·: absorption or the amount of drug that
reaches the systemic circulation.


288 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOA V AILABILITY AND BIOEQUIV ALENCE 289

where [AUC] values are area under the plasma level-time curve of one 3. Xu ~ . The cumulative amount of drug excreted in the urine, it
dosing interval in a multiple dosage regimen, after reaching the steady- is related to the AUC of plasma level data and increases as the extent .o f
state (Fig. 12.1) and 't is the dosing interval. absorption increases. •.

Bioavailability can also be detennined from the peak plasma concen-


tration at steady-state Css max according to following equation: (dXu I dt)max •
'
Fr = (Css,max)test Dstd 'ttest (l .S)
2 I
(Css,max)std Dtest 'tstd c:::
I
0
·-
.., I
The rate of absorption is not important in the multiple dosing method. G)
.... l
0

Urinary Excretion Studies .


)(
G)
'+-
I
0 I
This method of assessing bioavailability is based on the principle that ..,
G)
I
<U
the urinary excretion of unchanged drug is directly proportional to the 0:: I
I
plasma concentration of drug. Thus, even if a drug is excreted to some
extent (at least 10 to 20o/o) in the urine, bioavailabi Iity can be detennined.
I I
The study is particularly useful for drugs , extensively excreted unchanged I (tu>max
in the urine - for example, certain thiazide diuretics and' sulfonamides and -~~ Midpoint time of urine collection peri!Jd
for drugs that have urine as the site of action - for example, urinary
Fig. 12.2 Plot of excretion rate versus time. Note that the curve is
antiseptics such as nitrofurantoin and hexamine. The method has several analogous to a typical plasma level_-time profile obtained
advantages and disadvantages as discussed in chapter 10. Concentration after oral administration of a single dose of drug.
of metabolites excreted in urine is never taken into account in calculations
since a drug may undergo presystemic metabolism at different stages The extent of bioavailability is calculated from equations given below:
before being absorbed. The method involves collection of urine at regular 00
(Xu )oral Div
intervals for a time span equal to 7 biological half-lives, analysis of F= (12.6)
00
unchanged drug in the collected sample and detennination of the amount (Xu )iv Doral '

of drug excreted in each interval and cumulative amount excreted. A~ 00


(Xu )test Dstd
each sample collection, total emptying of the bladder is necessary to avoid Fr = - - - - - - - (12.7)
00
(Xu )std Dtest
errors resulting from addition of residual amount to the next urine sample.
Frequent sampling is also essential in the beginning in order to compute With multiple dose study to steady-state, the equation for computing .
correctly the rate of absorption. bioavailability is:
The three major parameters examined in urinary excretion data ob- F = (Xu,ss)test Dstd 'ttest . (l2.8)
tained with a single dose study are: r · (Xu,ss)std Dtest 'tstd
1. (dX 0 /dt)max : The maximum urinary excretion rate, it is ob- where (X u,ss) is the amount of drug excreted unchanged during a single
tained from the peak of plot between rate of excretion versus midpoint dosing interval at steady-state.
time of urine collection period. It is analogous to the Cmax derived from
Bioavailability can also be detennined for a few drugs by assay . of
plasma level studies since the rate of appearance of drug in the urine is
biologic fluids other than plasma d urine. In case. of t~e?p?ylline, .
proportional to its concentration in systemic circulation. Its value in-
salivary excretion can be used whe eas for cephalosporm antibtottcs, ap- . ,
creases as the rate of and/or extent• of absorption increases (see Fig. 12.2).
pearance of drug in CSF and bile- can be detettnined. ~aution. mu~st ,
2. (t 0 )max : The time for maximum excretion rate, it is analogous however be exercised to account for salivary and ent~rohepattc cy~lmg ·of- -
.
to the tmax of plasma level data. Its value decreases as the absorption rate the drugs. \

.. mcreases .


~0 BIOPHARMACEUTICS AND PHARMACOKINETICS
BIOAV AILABILITY AND BIOEQUIVALENCE 291
, Acu,te Pharmacologic Response
Despite attempts to standardize the test performance, the in vitro dissolu-
. ' .. When bioavailability measurement by pharmacokinetic methods is dif- tion technique is still by no means a perfect approach. The efforts are
fic~lt,fuaccurate or nonreproducible, an acute pharmacologic effect such mainly aimed at mimicking the environment offered by the biological
as change in ECG or EEG readings, pupil diameter, etc. is related to the system.
time c'ourse of a given drug. Bioavailability can then be determined by
construction of pharmacologic effect-time curve as well as dose-resQ<>nse There are several factors that must be considered in the design of a
graphs. The method requires measurement of responses for at least 3 dissolution test. They are:
· biological half-lives of the drug in order to obtain a go-od estimate of' 1. Factors relating to the dissolution apparatus such as-the design,
AUC. the size of the container (several mL to several liters), the shape of the
A disadvantage of this method is that the pharmacologic response container (round bottomed or flat), nature of agitation (stirring, rotating or
tends i:o be more variable and accurate correlation between me~sured oscillating methods), speed of agitation, performance precision of the
response and drug available from the formulation is difficult. Moreover, apparatus, etc.
the observed response may be due to an active metabolite whose concen- 2. Factors relating to the dissolution fluid such as--composition
tration is not proportional to the concentration of parent drug responsible (water, O.IN HCl, phosphate buffer, simulated gastric fluid, simulated
for the pharmacologic effect. intestinal fluid, etc.), viscosity, volume (generally larger than that neede'd
to completely dissolve the drug under test), temperature (generally 37°C)
Therapeutic Response
and maintenance of sink (drug concentration in solution maintained con-
Theoretically the most defmite, thjs method is based on observing the stant at a low level) or nonsink conditions (gradual increase in the drug
clinical response to fl drug formula'i~n given to patients suffering· from concentration in the dissolution medium).
disease for which it lis intended to btl used. A major drawback of this
method is that quantitation of observed response is too improper to all<;>w 3. Process parameters such as method of introduction of dosage
for reasonable assessrpent of relative bioavailability between two dosage form, sampling techniques, changing the dissolution fluid, etc.
forms of the same dru'g. The dissolution apparatus has evolved gradually and considerably from
a simple beaker type to a highly versatile and fully automated instrument.
Drug Dissolution Rate and Bioavailability The devices can be classified in a number of ways. Based on the absence
The physicochemical property of most drugs that has greatest intlu- or presence of sink conditions, there are two principal types of dis~olution
ence on their absorption characteristics from the GIT is dissolution rate. apparatus:
The best way of assessini therapeutic efficacy of drugs with a ~low
1. Closed-compartment apparatus : It is basically a limited-vol-
dissolution rate is in vivo determination of bioavailability which is usually
ume apparatus operating under nonsink conditions. The dissolution fluid
done whenever a new formulation is to be introduced into the market.
is restrained to the size of the container, e.g. beaker type apparatus.
However, monitoring batch-to-batch consistency through use of such in
vivo tests is extremely costly, tedious and time consuming·besides expos- 2. Open-compartment apparatus : It is the one in which the dos-
ing the healthy subjects to hazards of drugs. It would therefore be always age form is contained in a column which is brought in continuous contact
desirable to substitute the in vivo bioavailability tests with inexpensive in with fresh, flowing dissolution medium (perfect sink condition).
v{tro methods. The simple in vitro disintegration test is unreliable. The A third type called as dialysis systems are used for very poorly
best aVa~lable tool .today which can at least quantitatively assure abo11t aqueous soluble drugs for which maintenance of sink conditions would
. ths biologic availability of a _drug from its formulation is its in vitro otherwise require large volume of dissolution fluid. Only the official
dissolution J~t. methods will be discussed here briefly.
, I~ Vitro Drug· Dissolution Testing M6d¢1s · Rotating Basket Apparatus (Apparatus 1)
For !ID iin Vllto test to be useful, it must predict the in vivo beh~yiot
to It is basically a closed-compartment, beaker type apparatus comprising
such an extent .that in vivp bioavailability test need not be performed. of a cylindrical glass vessel with hemispherical bottom of one liter capac-
292 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOA VAI LABILITY AND BIOEQUIV ALENCE 293
.
ity -partially immersed in a water bath to maintain the temperature at 2. To serve as a tool in the development of a new dosage fonn with
37°C. A cylindrical basket made of 22 mesh to hold the dosage form is desired in vivo perfot tnance. ·
located centrally in the vessel at a distance of 2 em from the bottom and There are two basic approaches by which a correlation between disso-
rotated by a variable speed motor through a shaft (Fig. 12.3a). The lution testing and bioavailability can be developed:
basket should remain in motion during drawing of samples. All metal
parts like basket and shaft are made of S.S. 316. 1. By establishing a relationship, usually linear, between the in vitro
dissolution and the in vivo bioavailability parameters, and
Rotating Paddle Apparatus (Apparatus 2) 2. By using the data from previous bioavailability studies to modify
The assembly is same as that for apparatus I except that the rotating the dissolution methodology in order to arrive at meaningful in vitro-in
baske~ is replaced with a paddle which acts as a stirrer (Fig. 12.3b). The vivo correlation.
d~sage fonn is allowed to sink to the bottom of the vessel. A small, Though the fot n1er approach is widely used, the latter still holds ·
loose, wire helix may be attached to the dosage form that would otherwise substance, since to date, there is no single dissolution rate test methodol-
float.
ogy that can be applied to all drugs.
Rotating shaft -
(25-150 rpm)
Some of the often used quantitative linear in vitro-in vivo correlations
are:
~~- Sampling point---=---
--------~--~~~ ~~~--~-------- 1. Correlations Based on the Plasma Level Data : Here linear
relationships between dissolution parameters such as percent drug dis-
solved, rate of dissolution, rate constant for dissolution, etc. and parameters /
.,....__one liter flask---~
obtained from plasma level data such as percent drug absorbed, rate of
absorption, Cmax' tmax; Ka, etc. are developed; for example, percent drug
Dissolution medium - - + - -
- - 4 - - -· dissolved versus percent drug absorbed plots.
at 37°C
2. Correlation Based on the Urinary Excretion Data : Here, dis-
_____,__Basket 22 mesh
solution parameters are correlated to the amount of drug excreted unchanged
Paddle --\.----t'--- L-...t in the urine, cumulative amount of drug excreted as a function of time,
tf.-~..,t«--- Distance of 2 em ---~---4 etc.
Dosage form 3. Correlation Based on the Pharmacologic Response : An acute
(a) (b)
pharmacologic effect such as LDso in animals is related to any of the .
Fig. 12.3 Schematic representation of official dissolution apparatus dissolution parameters.
-forced convection non sink type (a) rotating basket
Statistical moments theory can also be used to detennine the rela-
apparatus, and (b) rotating paddle apparatus.
tionship such as mean dissolution time (in vitro) versus mean residence
'

In Vitro In Vivo Correlation time (in vivo).


A simple in vitro dissolution test on the drug product will be insuffi- Though examples of good correlations are many, there are instances
cient to predict its therapeutic efficacy. Convincing correlation between when positive correlation is difficult or impossible~ for example, in case
in v11ro dis~lution behavior of a drug and its in vivo bioavailability must of corticosteroids, the systemic availability may not depend upon the
be experimentally demonstrated to guarantee reproducibility of biologic dissolution characteristics of the drug. Several factors that limit such a
response. The two major objectives of developing such a correlation correlation include variables pertaining to the drug such as dissolution
are- methodology, physicochemical properties of the drug such as particle size.
physiologic variables like presystemic metabolism, etc.
1. To ensure batch-to-batch consistency in the physiologic perfor-
mance of a drug product by use of such in vitro values.
294 BIOPHARMACEUTICS AND PHARMACOKINETICS
BIOA V AILABILITY AND BIOEQUIV ALENCE 295,
. .
BIOEQUIVALENCE STUDIES are used in complete, open-label cross-over fashion such as the Latin
It is commonly observed that there are several forn1ulations of the square cross-over design in which
same drug, in the same dose, in a similar dosage forn1 and meant to be
-
~each fonnulation is administered just once to each subject and once
· given by the same route. Substitution of one product for another can be in each study period, and ~
made provided they are equally effective therapeutically. as the standard
- unlike parallel design, all the subjects do not -receive tJie same
I•
· a~cept~d. In order to ensure clinical perfonnance of such drug products,
formulation at the same. time; in a given study period, 'they ~are
bioequivalence smdies should be perfo11ned. Some of the important tetms
. · relevant in this context will be defmed. administered different fonnulations.
Such a randomized, balanced, cross-over study has several advan~
Equivalence : It is a relative tenn that compares drug products with
tages·-
. _, re_spect to a specific characteristic or function or to a defmed set of
. ' t/·
· ·· standards. There are several types of equivalences.
..
,
1. 'It minimizes the intersubject variability in plasma drug levels. -
2. Minimizes tli¢ carry-over effects which could occur when a given
Chemical Equivalence : It indicates that two or more drug products
dosage form- influences the bioavatlability of a subsequently -ad-
contain the same labeled chemical substance as an active ingredient in the
ministered product (intra-subject variability).
same amount. '
I 3. Minimizes the variations due to time effect, and thus,
· Pharmaceutic Equivalence : T~is tenn implies that two or more drug
products a~e id~ntical in strength, quality, purity, content uniformity and
/·+. Makes it possible to focus more on the fonnulation variables
which is the key to succ-ess for any bioequivalence study.
disintegratioH and dissolution characteristics; they may however differ in
containing different excipients. . ..- An examp~e of Latin square design for a bioequivalence study in
human volunteers is given in Table 12.1.
Bioequivalence : It is a relative tenn which denotes that the drug
substance in /two or more identical dosage fottns, reaches the systemic TABLE 12.1
cir~ulation at the same relative rate and t<;> the same relative extent i.e. Latin Square Cross-over Design for 6 (or 12) Subjects to /
their plasma concentration-time profiles will be identical without signifi- Compare Three Different Formulations, X, Y and Z
cant statistical differences.
Subject Drug Formulation
When statistically significant differences are observed in the bioavailability
I '

of two or more drug products, bioineq uivalence is indicated. Nun1ber Study Period I Study Period 2 Study Period 3 ·
1 '
I
I
' .
Therapeutic Equivalence : This tertn indicates that two or more drug 1.7 X y z
products that contain the same therapeutically active ingredient, elicit 2.8 y z X
identical phannacologic effects and can control the disease to the same y
3.9 z X '
extent. I

4.10 X z y
The in vivo bioequivalence study requires detertnination of relative
bioavailabil.ity after a$linistration of a single dose of test and reference
5. 11 z y X

fortnulations by the'' same route, in equal doses, but at different times. 6.12 y X z
The. referenc~ product ·is generally ~ previously approved product, usually
A drawback of such a cross-over design is that the study takes a long
the Innovator s product or some suitable reference standard. The study· is
time since an appropriate washout period between two administrations is
. perfonned in fasting, . young; healthy, adult male volunteers to assure -- -· - ...

essential which may be very long if the drug ,has a long tYl. Moreover
\ homogeneity in the population and to spare the patients, eld~rly or preg-
when the number of fonnulations to be testeq are more, the study be·
nant women from rigors of such a clinical investigation. Homogeneity in
comes n1ore difficult and subject dropout rates..are also high. 'This cari be
the study population pennits focus on fonnulation factors. The volunteers
overcome by use of _a__balaoced incomplete block design in which a
subject receives no more than 2 fon11ulations.

'
297
296 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOA VAILABILITY AND BIOEQUIV ALENCE

The three major approaches in overcoming the bioavailability prob-


.J As f9r bioa~ailability studies, either plasma level or uriuary excretion
--~s~~ie~ may be performed to assess bloequivalence between ' drug products. lems due to such causes are:
In-vitro-in vivo correlation can also be established for the fonnulations. 1. The Pharmaceutic Approach which involves modification of
fot n1ulation, manufacturing process or the physicochemical prop-
It is . always easier to establish bioequivalence between existing drug
1
erties of the drug without changing the chemical structure.
p~ducts than detertnination of phartnacokinetics of a new drug or
bioavailability of a new dosage fortn since . 2. The· Pharmacokinetic Approach in which the pharmacokinetics
of the drug is altered by modifying ' its chemical structure.
1: The human volunteers used for the study of both products are
sam~ and all phattnacokinetic parameters can be assumed to be 3. The Biologic Approach whereby the route of drug administration
same for both drug fortnulations and there is no need to investi- may ,be changed such as changing from oral to parenteral route.
-- gate nonlinearity. The second approach of chemical structure modification has a number
2. The study protocol for all subjects is unifottn, the efficiency of of drawbacks of being very expensive and time consuming, requires
drug absorption from both fonnulations can be considered as repetition of clinical studies and a long time for regulatory approval.
same and thus differences in absorption pattern can be ascribed to Moreover, the new chemical entity may suffer from another phannacoki-
differences in drug release from the two dosage fortn. netic disorder or bear the risk of precipitating adverse effects. Only the
phannaceutic approach ·will be dealt herewith.
Statistical Interpretation of Bioequivalence Data The attempts, whether optimizing the fonnulation, manufacturing pro-
· After the data has been collected, statistical methods must be applied cess or physicochemical properties of the drug, are mainly aimed at
to detennine the level of significance of any observed difference in the enhancement of dissolution rate as it is the major rate-limiting step in the
r~te ~d/or extent of absorption in order to establish bioequivalence be- absorption of most drugs. There are several ways in which the dissolution
tween two or more drug products. Typically, an analysis of variance rate of a drug can be enhanced. Some of the widely used methods, most
(ANOV A) method is applied to detertnine statistical differences. If a of which are aimed at increasing the effective surface area of the drugs,
statistically significant difference is observed, it is important to determine
will be discussed briefly.
if it is clinically significant. Quite often, it is found that for e.g. a
1. Micronization : The process involves reducing the size of the
statistically significant difference of 10% in the extent of absorption be-
solid drug particles to 1 to 10 microns commonly by spray drying or by
tween the two fonnulations is insignificant clinically. Currently, a simple
use of air attrition methods (fluid energy mill). Examples of drugs whose
~cccptable rule is that if the relative bioavailability of the test fonnulation
bioavailability have been increased by micronization include griseofulvin
is in the range 80 to 120% of reference standard, it is considered .bioequivalent.
Another accepted opinion is that the difference between the bioavailabilities and several steroidal and sulfa drugs.
of the test fotnlulations should not be greater than ± 20% of the average 2. Use of Surfactants : The surface-active agents enhance dissolu-
of reference standard. , tion rate primarily by promoting wetting and penetration of dissolution
fluid into the solid drug particles. They are generally used in concentra-
METHODS FOR ENHANCEMENT OF BIOAVAILABILITY tion below their critical micelle concentration (CMC) values since above
As far as the definition of bioavailability is concerned, a drug with CMC, the drug entrapped in the micelle structure fails to partition in the
poor bioavailability is the one with - dissolution fluid. Nonionic surfactants like polysorbates are widely used.
Examples of drugs whose bioavailability have been increased by use of
1. Poor aqueous solubility and/or slow dissolution rate in the bio-
surfactants in the formulation include steroids like spironolactone.
logic fluids.
3. Use of Salt Forms : Salts have i!llproved solubility and dissolu-
·2. Poor stability of the dissolved drug at the physiologic pH.
tion characteristics in comparison to the original drug. Alkali metal salts
3. Inadequate partition coefficient and thus poor pettneation through of acidic drugs like penicillins and strong acid salts of basic drugs b~~
the biomembrane.
atropine are more water-soluble than the parent drug.
4. Extensive presystemic metabolism.

,
BIOA V AILABILITY AND BIOEQUIV ALENCE
299
298 BIOPHARMACEUTICS AND PHARMACOKINETICS
are also called as molecular dispersions or mixed crystals. Because of
4. Alteration of pH of the Drug Microenvironment : This can be
reduction in particle size to the molecular level, solid so~utions sho~
achieved in two ways in situ salt fortnation, and addition of buffers to
greater aqueous solubility and faster dissolution than eutecttcs and sohd
the forn1ulation e.g. buffered aspirin tablets.
dispersions. They are generally prepared by fusion method whereby a
5. Use of Metastable Polymorphs : A metastable polymorph is physical mixture of solute and solvent are melted together followed by
more soluble than the stable polymorph of a drug that exhibits rapid solidification. Such systems, prepared by fusion, are. often c~lled as
polymorphism for example, the B fonn of chloramphenicol palmitate is melts e.g. griseofulvin-succinic acid (Fig. 12.4). The grts~ofulvtn. from
more water soluble than the A and the C fonns. such solid solution dissolves 6 to 7 times faster than pure grtseofulvtn.
-6. Solute-Solvent Complexation : Solvates of drugs with organic
solvents (also called as pseudopolymorphs) generally have higher aqueous Melt
~olubility than their respective hydrates or the original drug. Much higher
solubility can be attained by freeze drying such a solute in solution with
an organic solvent with which it is known to forni a solvate. Such a ~

process results in a powder of particles of submicron size, e.g. I :2 griseofulvin- -::J


as
......
CD
. Melt
+
benzene solvate. However, one should take care that the solvent is a.
E Solid solution
CD
nontoxic. t-
7. Solvent Deposition : In this method, the poorly aqueous soluble
Solid solution ·
\

drug such as nifedipine is dissolved in an organic solvent like alcohol and


deposited on an inert, hydropl!ili£-,~ solid matrix such as starch or micro-
Pure A Mole 0/o B Pure B
crystalline cellulose by evaporation of solvent. -----+)

8. Selective Adsorption on Insoluble Carriers : A highly active Fig. 12.4 Binary phase diagram for continuous solid solution of A a~d B.
·adsorbent such as the inorganic clays like bentonite can enhance the T A and T 8 are melting points of pure A and pure B respectively .
dissolution rate of poorly water soluble drugs such as griseofulvin, If the diameter of solute molecules is less than 60o/o of diameter of
indomethacin and prednisone by maintaining the concentration gradient at solvent molecules or its volume less than 20% of volume of solvent
its maximum. The two reasons suggested for the rapid release of drugs molecule the solute molecule can be accommodated within the intermo-
from the surface of clays are the weak physical bonding between the lecular s;aces of solvent molecules e.g. digitoxin-PEG 6000 s~lid sol~tio~.
adsorbate and the adsorbent, and hydration and swelling of the clay in the Such systems show faster dissolution. When the resultant solzd solut1on IS
aqueous media. a homogeneous transparent and brittle system, it is called as glass solu-
9. Solid Solutions : The three means by which the particle size of a tion. Carriers that form glassy structure are citric acid, urea, PYP and
drug can be reduced to submicron level are- PEG and sugars such as dextrose, sucrose and galactose.
i. use of solid solutions, The two mechanisms suggested for enhanced solubility and rapid dis-
ii. use of eutectic mixtures, and solution of molecular dispersions are:
iii. use of solid dispersions. 1. When the binary mixture is exposed to water, the soluble ca.rrier
In all these cases, the solute r.i~ frequently a poorly water soluble drug\ dissolves rapidly leaving the insoluble drug in a state of mtcro-
acting . as the guest and the solve'tft is a highly water-sol6ble compound or crystall ine dispersion of very fine particles, and
I
polymer acting as a ho~t or cart-fer. •
2. When the solid solution, which is said to be in a state of ran-
domly arranged solute and solvent molecules in the c~stal. la~ice,
I
....
A solid solution is a binary system comprising of a solid solute
molecularly dispersed in a solid solvent. Since the two components is exposed to the dissolution fluid, the soluble carrter dtssolves
rapidly leaving the insoluble drug stranded at almost molecular
crystallize together in a homogeneous one phase system, solid solutions
level. I
300 BIOPHARMACEUTICS AND PHARMACOKIN!:TICS
BIOA VAILABILITY AND BIOEQUIVAL~NCE 301
Fig. 12.5 shows a comparison between the dissolution rates of Jiffer-
ent fonns of griseofulvin. Examples of eutectics include paracetamol-urea, griseofulvin-urea,
griseofulvin-succinic acid, etc. Solid solutions and eutectics, which are
basically melts, are easy to prepare and economical' with no solvents
c ___..------~--Solid solution
involved. The method however cannot be applied to:
...:l0
·-
-0 · drugs which fail to crystallize from the mixed melt,
(/), __________ Eutectic mixture
c '
·-c --thermolabile drugs, anct
...
0
·- --'Carriers such as succinic acid that decompose at their melting point.
...f!
c The eutectic product is often tacky, intractable or irregular crystals. ·
Q)
.o ~,...------~-Micronized drug
'c · 11. Solid Dispersions : These are generally prepared by solvent or
0
(.)
~____.~---------Coarse drug co-precipitation method whereby both the guest solute and the solid
T carrier solvent are dissolved in a common volatile liquid solvent such as
alcohol. The liquid solvent is removed by evaporation under reduced
-4) Time pressure or by freeze drying which results in amorphous precipitation ~f
Fig. 12.5 Dissolution rates of griseofulvin as coarse particles, as micronized guest in a crystalline carrier. Thus, the basic difference between sohd
particles and as eutectic and solid solution with succinic acid. dispersions and solid solutions/eutectics is that the ~g is precipitated out
in an amorphous fonn in the fotmer as opposed to crystalline fonn in the
10. Eutectic Mixtures : These systems are also prepared by fusion latter; e.g. amorphous sulfathiazole in crystalline urea. Such dispersio~s
method. Eutectic melts differ from solid solutions in that the fused melt are often called as co-evaporates or co-precipitates. The method IS
of solute-solvent show complete miscibility but negligible solid-solid solu- suitable for thertnolabile substances but has a number of disadvantages
bility i.e. such systems are basically intimately blended physical mixture of like higher cost of processing, use of large quantities of solvent, difficulty
two crystalline components. A phase diagram of two component system in complete removal of solvent, etc. The carriers used are same as for
is shown in Fig. 12.6. When the eutectic mixture is exposed to water, the eutectics or solid solutions. With glassy materials, the dispersions fonned
soluble carrier dissolves leaving the drug in a microcrystalline state which are called as glass dispersions or glass suspensions. Fig. 12.7. shows
solubilizes rapidly.
comparative dissolution rates of griseofulvin from PVP dispersions.

·->c .--------·- Griseofulvin- PVP ( 1:20)


-....: l
0
Melt Q)
UJ .--------Griseofulvin-PVP (1: 10)
Cl)
.....
........
:l
ra
__,.,,-.---- Griseofulvin-PVP (1 :5)
c
Q)
c. Melt Melt ........ca
·-0
E
Q)
..... Solid A-
+ +
Solid B
...
c
~ ~,......,...,.....-.-----Micronized griseofulvin
c
T E
·u
0

Solid A + Solid B

Pure A -4) Time in Minutes


--..• Mole% B Pure B
Fig. 12.6 Simple binary phase diagram showing eutectic point E. The cuiecti:t Fig. 12.7 Dissolution rate enhancement of griseofulvin
by ·sol~d dispersion technique.
composition at point E of substances A and 8 represents the one
having lowest melting point. TA and T 8 arc 1nelting points of pure A
and pure 8 respectively .
I
..
302 BIOPHARMACEUTICS AND PHARMACOKINETICS BIOA VAI LABILITY AND BIOEQUIV ALENCE 303

12. Molecular Encapsulation with Cyclodextrins : The beta- and 8. What should be the duration of washout period between any two bioavailability
studies in the same subject? Why?
gamma-cyclodextrins and several of their derivatives are unique in having
the ability to forn1 molecular inclusion complexes with hydrophobic drugs 9. Name the methods for determining bioavailability of a drug from its dosage
form.
having poor aqueous solubility. These cyclodextrin molecules ·are versa-
tile in having a hydrophobic cavity of size suitable enough to accommodate 10. Which is the method of choice in bioavailability determination? On what
the lipophilic drugs as guests; the outside of the host molecule is relatively principle is such a study based? ,
..

hydrophilic (Fig. 12.8). Thus, · the molecularly encapsulated drug has l1. Explain with significance the parameters used in bioavailability determina-
greatly improved aqueous solubility and dissolution rate. There are sev- tion by plasma level studies.
eral examples of drugs with improved bioavailability due to such a 12. In multiple dose study, ~io~vailability determination is done at steady-state
phenomena thiazide diuretics, barbiturates, benzodiazepines and a num- and one dosing interval. Explain.
ber of NSAIDs. 13. Why is determination of absorption rate not considered important in the
multiple dosing method?
______ Hydrophilic surface 14. What is the principl~ behind assessment of bioavailability using urinary
~~~---Hydrophobic lining excretion studies?
15. Determination of metabolites in urine is not used as a measure of bioavailability?
------Cavity for encapsulating Why?
hydrophobic drugs 1~. Why should the volunteers be; instructe~ to completely empty their bladders
while giving urine samples? ,
Entrapped salicylic
acid molecule 17. Why should frequent sampling of urine/plasma be done initially after drug
\,~----- ... ,I
'~- ____ .,} administration in bioavailability determination?
18~ Name the parameters examined in urinary excretion data to determine
\ '

bioavailability,. . What is 'their analogy : with parameters of plasma level ·


studies?
Fig. 12.8 Functional and structural feature of a cyclodextrin.
molecule showing an encapsulated drug. 19. What are the drawbacks of using acute pharmacologic response and thera-
peutic response as _measures of bioavailability?
20. What is the reliable alternative to in yivo bioavailability studies in monitor-
QUESTIONS ing batch-to-batch consistency in pharmaceutical 'manufacturing? .
1. Define bioavailability. What are the objectives of bioavailability· studies? 21. What factors should be considered in the design of dissolution testing
'
models?
2. Enumerate the factors affecting bioavailability of a drug from its dosage
l.

form. 22. What are the objectives and approaches in developing in vitro- in vivo
correlation?
3. In _a ~ioavailability study, explain how determination of both rate and extent
of absorption are important. 23. Discuss the various methods of developing quantitative linear in vitro-in
vivo relationships.
4. petine absolute and relative bioavailability.. What is the basic difference
between the two? 24. Define ~he following equivalenc~, chemical ·equivalence, pharmaceutic equiva-
.
lence, bioequivalence and clinical equivalence. .
5. .What are the l.i~itations of using oral solution as a standard for determining
·absolute bioavailability? · 25. Why are bioequivalency studies always performed in healthy human volun-
teers?
6. Compare single dose with multiple dose b.ioavailability studies.
26. What are the advantages and limitations of randomized, balanced, cross-over .
7._Discuss the merits· and demerits of using healthy subjects and patients as
study in bioequivalence determination? _/
- v~lunteers for bioavailability studies. .. " .
..
., . .
)
••
BIOAV AI LABILITY AND BIOEQUIVALENCE 305
304 BIOPHARMACEUTICS AND PHARMACOKINETICS
37. The three pharmacokinetic parameters from urinary excretion data of a drug·
27. W~~- is it easy to establish bioequivalence between dosage forms in com- given as 50 mg oral formulations of two different companies, of which A is
parison· "to _determination of pharmacokinetics or bioavailability of a new
the innovator's product, are as follows:
formulation?
Parameters Formulation
28. What are the generally accepted, statistical rules for establishing bioequivalence
between formulations? A B
29. Layout a Latin square cross-over diagram for bioequivalency study on three 8.0
(dXuldt)max (mg/hr) 6.0
formulations- A; B and C, in six volunteers.
}
(tu)max (hour) 2.0 1.0
30. What are the va~ious approaches . aimed at enhancing bioavailability of a I

drug from its dosage form? Xu ex> (mg) 39.1 35.9


'
31. Discuss the methods aimed at enhancing bioavailability through enhance-
a. What is the relative availability of formulation B against A?
ment nf drug dissolution rate.
' . Answer : 92.0%.
32. What are the various means by .which the particle size of a drug can be
reduced to submicron level? b. Are the two formulations bioequivalent?
33. What is. the basic difference between solid solutions and solid dispersions? c. If the drug is meant for the treatment of an acute condition, which one of
G·ive reasons for the faster dissolution of the former. the two formulations is better?
34. Solid solutions dissolve faster than eutectics. Why?
35. A new drug undergoes both degradation in gastric fluids as well as exten- -- -

sive presystemic hepatic metabolism. In order to obtain optimum oral


bioavailability, which of the following formulations will you prefer .
solution, suspension, tablet, enteric coated tablet, S.R. tablet or enteric coated
microspheres? Why? Rank them in order of decreasing bioavailability.
I

36. Following data is obtained for 4 formulations of pentoxifylline in volunteers


of average weight 50 Kg. ' ·
Drug Product Dose (mg/kg) AUC (mcg.hr/1)
i.v. solution 1.2 450
oral solution 4.0 822
oral capsule 4.0 736
oral S.R. tablet 8.0 1040
...
a. What is the absolute bioavailability of the drug from capsule and S.R.
tablet?
Answer : Capsule = 49% and S.R.tablet = 34.6%.
b. What is the relative bioavailability of capsule and S.R.tablet against oral
solution?
Answer : Capsule = 89.5% and S.R.tablet = 63.2%. .. .. ...
.. .. -:
c. Which solid formulation shows better bioavailability?
.• . •

. ..
d. Are the two solid formulations bioequivalent? • • • • •
.
•• 11
'

e. What could be the possible reasons for poor bioavailability from S.R.
tablets? •
• • l

'


APPLICATIONS OF PHARMACOKINETIC PRINCIPLES 307
'
and amount. Selection of a suitable drug is based on achieving optimal
therapy by balancing the sJesirable and undesirable effects. · Co-administra-·
I

tion of several drugs to a patient may lead to changes in the pharmacokinetic


13 profile of a drug which is indicative of interactions between drugs. S~ch
an understanding of interaction makes possible more rational use of drugs
that have to be co-administered.
' Applications of '
Clinically, the two most important applications of pharmacokinetic
-Phat·tnacokinetic Principles principles are:
1. Design of an optimal dosage regimen, and
2. Clinical management of individual patient and therapeutic drug
monitoring.
The time course of drug concentration in the body after its administra-
1
tion can be defmed by a number of pharmacokinetic parameters. Often, Such applications pennit the physician to use certain drugs more safely
the inforrnation gained about the phattnacokinetics of one drug helps in and sensibly.
anticipating the pharmacokinetics of another. The knowledge of phanna-
cokinetic behavior of a drug coupled with important phannacodynamic DESIGN OF DOSAGE REGI~NS
parameters like therapeutic index can be put to several applications:
Dosage regimen is defined as the manner in which a drug is taken.
I. Design and development of new drugs with greatly improved For some drugs like analgesics, hypnotics, antiemetics, etc., a single dose
therapeutic effectiveness and fewer or no toxic effects may provide effective treatment. However, the duration of most illnesses
2. Design and development of an optimum forrnulation for better use is longer than the therapeutic effect produced by a single dose. In such
of the drug cases, drugs are required to be taken on a repetitive basis over a period of
3. Design and development of controlled/targeted release for ntulation time depending upon the nature of illness. Thus, for successful therapy,
design of an optimal multiple dosage regimen is necessary. An optimal·
4. Select the appropriate route for drug administration
multiple dosage regimen is the one in which the drug is administered in
5. Select the right drug for a particular illness suitable doses (by a suitable route), with sufficient frequency that ensures
6. Predict and explain drug-food and drug-drug interactions maintenance ofplasma concentration within the therapeutic window (with-
7. Design an appropriate multiple dosage regimen out excessive fluctuations and drug accumulation) for the entire duration
of therapy. For some drugs like antibiotics, a minimum effective concen-
8. Therapeutic drug monitoring in individual patients
'
tration should be maintained at all times and for drugs with narr<;>w
9. Dosage adjustments in situations of altered physiology and drug therapeutic indices like phenytoin, attempt should be made not to ~xceed
interactions. toxic concentration. -
The applications of pharmacokinetic principles are mainly aimed at In designing a dosage regimen ,,
"
achieving the therapeutic objective. The therapeutic objective is often
I. It is assumed that all phannacokinetic parameters of the drug
contr(JI or cure of the condition in shortest possible time with minimum
remain constant during the course of therapy once a dosage regi- -
side effects by the use of least amount of drug. New drug development or
men is established; the same becomes invalid if any change is
chemical modification is frequently done to improve phannacokinetic prop- 1
observed.
erties and increase efficacy. Drug product design is aimed at optimizing
· bioavailability or better controVcure of illness through controlled or tar- 2. The calculations are based on open one-comp~ent model which
geted re~ase. Proper choice of route · of administration is necessary · to can also be applied to two compartment model if B is used instead
ensure that the d.J:ug moves to the site of action at a sufficiently rapid rate of KE, and V d ss instead of V d while calculating the regimen.
' .
'

306
APPLICATIONS OF PHARMACOKINETIC PRINCIPLES 309
308 BIOPHARMACEUTICS AND PHARMACOKINETICS
frequency increased. It also results in greater drug accumulation in the
~ Irrespective of the route of administration and complexity of pharma- body and toxicity (Fig. 13 .2).
tokinetic equations, the two major parameters that can be adjusted in
High dosing frequency
developing a dosage regimen are~-
( t < t 112), lesser fluctua-
. 1. The dose size. the quantity of drug administered each time, and tions but shows both
c.. therapeutic and toxic ·
2. The dosing frequency the time ·interval between doses. 0
responses
Both parameters govern the amount of drug in the body · at any given
Optimum dosing
.time. frequency (t = t 112),
therapeutically
,Dose Size successful
- MEC
. The magnitude_ of both therapeutic and toxic responses depends upon
Less dosing
dQse size. Dose size c.alculation also requires the knowledge of amount of frequency (t > t 112),
drug absorbed after administration of each dose. Greater the dose size,
greater the fluctuations between Css,max and Css,min during each dosing
i large fluctuations,
therapeutically
unsuccessful
interval and greater the chances of toxicity (Fig. 13.1 ).
--+)' Time in hours
DOSES
Fig. 13.2 Schematic representation of the influence of dosing frequency
t :1' • t l t l .t ~ on plasma concentration-time profile obtained after oral
administration of fixed doses of a drug.
Larger dose profile; A proper balance between both dose size and dosing frequency is
larger fluctuationsl
shows both therapeutic
often desired to attain steady-state concentration with minimum fluctua-
c
...as
0
·- ~nd toxic responses tions and to ensure therapeutic efficacy and safety. The same cannot be
......c ~----.-----+----- · MSC
obtained by giving larger -doses less frequently. However, administering
fjl------4-#-_...~-- smaller doses more frequently .results in smaller fluctuations. A drug with
c
0 Optimum dose profile; a narrow therapeutic index such as digoxin is best administered in small
0 therapeutically
•E doses at frequent intervals, usually less than the t y2 of the drug, to obtain a
successful
•-t'O
profile with least fluctuations which is similar to that observed with
~ -~~--~~--~~---- -----~----~----- MEC constant rate infusion or controlled release system.
Smaller dose profile,
minimum fluctuations
Drug Accumulation During Multiple Dosing '-
but ineffective Consider the amount of drug in the body-time profile shown in Fig.
'
therapeutically 13 .3. obtained after i.v. multiple dosing with dosing interval eql!al to one
~--~----~------L-----~----~----~
/

--+) Time in\•Hours \


t y2"

Fig. 13.1 Schematic representation of influence of dose size on plasma I


After the adn1 inistration of first dose X0 at t = 0, the amount of drug
concentration-time profile after oral administration of a drug- in the body \'fill be X = 1X0 . At the next dosing interval when X = 1;2X 0 ..
at fixed intervals of time. the amount of drug remaining in the body. administration of the next i.v.
dose raises the bo~y content to X = X0 + 1h X 0 i.e. drug accumulation
- :, Dosing Frequency occurs in the body. Thus . accun1ulation occurs because drug .fron1 pre,·i-
The dose interval (inverse of dosing frequency) is calculated on the ../
ous doses has not been rernoved con1plete~\·. As the an1ount of drug in
basis of half-life of the drug. If the interval is increased and the dose is the body rises gradually due to accumulation. the rate of elitnination also
l I

.unchanged, :7~~~ ~min and Cav decre~se ?ut the .ratio Cmax1Cmin _in- rises proportionally until a steady-state or plateau is reached \\·hen the rate
cre~es. Opp9'_s1te Is observed when dostng Interval ts reduced or dostng
.
' '


310 BIOPHARMACEUTICS AND PHARMACOKINETICS
. . APPLICATIONS OF PHARMACOKINETIC PRINCIPLES 311
of drug entry into the body equals the rate of exit. The maximum and .-
minimum values of X i.e. Xss,max and Xss,min approach respective asymp- means that the rate at which the multiple dose. steady-state is reached is
totes at plateau. It is interesting to note that at plateau, Xss,min = 1X0 and determined only by KE· . The time taken to reach steady-state is indepen-
Xss,max = 2Xo i.e. Xss,min equals the amount of drug in the body after the dent of dose size, dosing interval and number of doses.
frrst dose and Xss,max equals twice the frrst dose. Also (Xss,max - Xss,mirJ
Maximum and Minimum Concentration During Multiple Dosing
= Xo and Xss,max!X·ss,min = 2. All this applies only when t = tYl and drug
is administered intravenously. When t < tY2, the degree of accumulation If n is the number of doses administered, the Cmax and Cmin obtained ·.
is greater and vice-versa. Thus, the extent to which a drug accumulates in on multiple dosing after the nth dose is given as:
. . the body during multiple dosing is a function of dosing interval and
(13.2) .
elimination half-life and is independent of dose size. The extent to which
a drug will accumulate with any dosing interval in a patient can be
derived from infortnation obtained with a single dose and is given by
(13.3)
accumulation index Rae as:
-.
1
Rae -- (13.1)
1_ e-KEt
The maximum and minimum concentration of drug in plasma

at steady-
state are found by following equations:
- . .
Xo ·X xo xo x.
~0 4,0 (13.4)
~ ~ J; Css,max =
1 _ e-KEt
-KEt
C
_ oe _ C -KEt (13'.5)
Css,min - K - ss,max e
~ Upper
1 -e - Et
asymptote,
Xss. max where C 0 = concentration that would be attained from instantaneous ab-
sorption and distribution (obtained by extrapolation of elimination curv~
Steady-state to time zero). Equations 13.2 to 13.5 can also be written in teuns of
amount of drug in the body. Fraction of dose absorbed, F, should be _
~ Lower taken into account in such equations.
asymptote,
Fluctuation is defined as the ratio Cmax1Cmin· Greater the ratio,
Xss. min greater th~ fluctuation. Like accumulation, it depends upon dosing frtt-
quency and half-life of the drug. It also depends upon the rate of
.... _
0 -~----~~-----L------L---~-~-~~~~~
~-~--
-- -- absorption. The greatest fluctuation is observed when the drug is given as
i. v. bolus. Fluctuations are small when the drug is given extravascuh1rly
Ch 1t 2t 3t 4t 5t because of continuous absorption.
--+~ Dosing Interval in hours (t =t112)
Average Concentration and Body Content on
Fig.l3.3 Accumulation of drug in the body during multiple dose regimen
of i. v. bolus with dosing interval equal to one half-life of the drug. Multiple Dosing to Steady-State
Approximately 5 half-lives are required for attainment of steady-state. The aver.age drug concentration at steady-state Css.av is a function of
maintenance dose X0 , the fraction of dose absorbed F. the dosing interval
.Time to Reach Steady-State During Multiple Dosing ,· 1
t and clearance CIT (or V d and KE or ty2 ) of the drug.
The time required to reach steady-state depends primarily upon the
half-life of the drug. Provided Ka >> KE, the .plateau is reached in ( 13.6)
Css.av =
approximately 5 half-lives. This is called as plateau principle. It also ·
313
·-
0

- 312 BIOPHARMACEUTICS AND PHARMACOKINETICS APPLICATIONS OF PHARMACOKINETIC PRINCIPLES

AUC (single dose) The ratio of loading dose to maintenance dose Xo,L!Xo is called as
-
1.44 F X 0 tYl - -------- (13.7) dose ratio. As a rule, when 't = ty,, dose ratio should be equal to. 2.0 but
Vdt must be smaller than 2.0 when 't > tv2 and greater when 't < ty2. F1 ~· 13 .4 ·
where the coefficient 1.44 is the reciprocal of 0.693 in equation 13.7. shows that if loading dose is not optimum, either too lo:-v or .too high, the
AUC is the area under the curve following a single maintenance dose. steady-state is attained within ~ppr?ximately 5 half-hves m a manner
Equation 13.7 can be used to calculate maintenance dose of a drug to similar to when no loading dose IS gtven.
achieve a desired concentration. Since X = V d C, the body content at Maintenace Doses X 0
Loading dose X 0 L
steady-sta'e is given as: I

1.44 F Xo tYl
Xss,av = ------ (13.8) '
t
'[
_ dose ratio > 2
~hese
average values are not arithmetic mean of Css,max and Css,min dose ratio =l 2
0

since the plasma drug concentration declines exponentially. .--t- dose ratio < 2

Loading and Maintenance Doses


A drug does not show therapeutic activity unless it reaches the de-
0

sired steady-state. It takes ab~ut 5 half-lives to attain it and therefore the


r,
time taken will be too long if the drug has a long half-life. Plateau can be
/ I '
reached immediately by administering a dose that gives the desired steady- I
I '\
state instantaneously before the commencement of maintenance doses X 0 . I :_1 _ _~\,\----~r-----, MEC
I
Such an initial or first dose intended to be therapeutic is ·called as I
priming dose or loading dose Xo,L· A simple equation for calculating
loading dose is:

Xo , -LCss ,av Vd
---F-- (13.9) ____...)' Dosing Interval in Hours

After e. v. administration, Cmax is always smaller than that after i. v. 4 Sch ematic representation of plasma concentration-time
Fig. 13. h 20
profiles that result when dose ratio is greater t an ..
administration and hence loading dose is proportionally smaller. For
equal to 2.0 and smaller than 2.0.
drugs having low therapeutic indices, the loading dose may be divided
into smaller doses to be given at various intervals before the frrst mainte- The foregoing discussion and the equatio~s e~pres~ed until now ~ppl~
nance dose. When V d is not known, loading dose may be calculated by only to drugs that follow one-compartment kmet1cs w1th first-order d1spo
the following equation: sition. Equations will be complex for multicompartment models.
XoL - 1
' --------- (13.10) Maintenance of Drug within the Therapeutic Range
The ease or difficulty in maintaining drug concentration within the
The above equation applies when Ka >> KE and drug is distributed therapeutic window depends upon
rapidly. When the drug is given i. v. or when absorption is extremely 1. The therapeutic index of the drug.
rapid, the absorption phase is neglected and the above equation reduces 2. The half-life of the drug.
to:
oo
3. Convenience of dosing.
Xo,L 1
It is extremely difficult to maintain such a level. fo~ a drug with sh~rt

- - - -- =Rae (13.11)
(1 _ e- KE-r)
half-life (less than 3 hours) and narrow therapeutiC mdex e.g. hepann ,
315
APPLICATIONS OF PHARMACOKINETIC PRINCIPLES
'

314 BIOPHARMACEUTICS AND PHARMACOKI!'!ETICS (Cupper - Ctower) (13 .16)


Css,av = - - - - - - - - - -
since the dosing frequency has to be essentially less than t H 2.303 log (CupperiCtower)
drugs s h · ·u· Y2· c wever
b . ucl as pemci m (ty, = 0.9 hours) with high therapeutic ir·dex rna;
t e ;IV~n ess frequently (every 4 to 6 hours) but the maintenance-dose has INDIVIDUALIZATION
.o . e. arger so that the plasma concentration persists above the minimum Because of reasonable homogeneity in humans, the dosage regimens
Inh.tbttory level. A drug with intermediate t~ (3 to 8 hours) may be given are calculated on population basis. However, same dose of a dtug may
. t -< t.~ I·f therapeutic
at mtervals · Index
· · low and those with
Is · high indices produce large differences in pharmacologic response in different 'lriH'ividu-
can be given at mtervals· be~een I to 3 half-lives. Drugs with half-lives als. This is called as intersubject variability. In other words, it means
g~ea~e~ than 8 hours are more convenient to dose. Such drugs are usually
that the dose required to produce a certain response varies from indi-
a ~mistere~ once every half-life. Steady-state in such cases can be vidual to individual. Rational drug therapy requires individualization of
attained raptdly by administering a loading dose. For drugs with ve
long h~lf-hves
(above 24 hours) e.g. amlodipine, once daily dose is veryry
dosage regimen to fit a particular patient's needs. This requires knowl-
edge of pharmacokinetics Of drugs. The application of pharmacokinetic
convenient.
principles in the dosage regimen design for the safe and effective manage-
Design of Dosage Regimen from Pla_sma ·concentrations ment of illness in individual patient is called as clinical pharmacokinetics.
d If .th~ therapeutic range, apparent Vd and clearaitce or half-life of a The two sources of variability in drug responses are:
rug IS ~own,. th_en dosage regimen can be designed to. maintain dru 1. Pharmacokinetic variability which is due to differences in drug
~onlcentrat~on.withm the specified therapeutic range. The latter is define~ concentration at 'the site of action (as reflected from plasma drug
Y ower hmtt (Clower) and an upper limit (C ) Th · d . concentration) because of interindividual differences in drug ab-
inter 1 h · h ·d II upper · e maxtmum osmg
va ~ w tc . I ea y depends upon JQ,~ therapeutic index (can be defined sorption, distribution, metabqlism and excretion.
as a ratiO of C upper to C lower) an d e rlffimatton
· . half-hfe . of the drug can be 2. Pharmacodynamic variabi~ity which is attributed to differences
expressed by equation 13 .12, a modification of equation 13.5. ' in effect produced by a giver ·drug concentration.,.
t = 2.303 log (CupperiCtower) The major cause for variations is phatmacokinetic variability. Differ-
max · ( 13. 12)
KE ences in the plasma levels of a given drug in the same subject when given
on different occasions is called as intrasubject variability. It is rarely
Since KE = 0.693/t yl, the abov~ .equation can also be written as: encountered in comparison to interindividual variations. The differences
' '•

'tmax = 3 ·32 ty./log ·{CupperiCtower) (13.13) in variability differ for different drugs. Some drugs show greater variabil-
ity than the others. The major causes of intersubject phannacokinetic
Understanda~ly, the d?sing inte~al selected is always .smaller. than variability are genetics, disease, age, body w~ight and drug-drug interac-
tmax· The maximum maintenance dose X that b . tions. Less important causes are pharmaceutical fonnulation , route of
tmax is expressed as: o,max can e given every
administration., environmental factors and patient noncompliance.
X = Y_ d (Copper- Clower) The main objective of individualization is aimed at optimizing the
o.max ---=--~,--=------ (13.14)
F dosage regimen. An inadequate therapeutic response calls for a higher
dosage whereas drug related toxicity calls for a reduction in dosage.
After .a convenient dosing interval t smaller than tmax has been
Thus, in order to aid individualization, a drug must be made available in
selected, the maintenance dose is given as:'
dosage fmms of different dose strengths. The number of dose strengths
= . .~p,,:nlax
·1·
(13.15)
in which a drug should be made available depends upon 2 major factors-
X0 -~ 1 .' I
'tmax i. The therapeutic index of the drug, and
Ad . . · ' ii. The degree of intersubject variability. Smaller .the therapeutic
t . ~mistratJOn
of Xo every t produces an average steady-state concen-
ratton efined by ~quation 13 .7. The value of C ss.av can also be calculated index and greater the variability, more the number of dose strengths
from the -therapeutic range according to equation 13.16. required.
316
BIOPHARMACEUTICS AND PHARMACOKINETICS
317
~n the assumption that all patients require the same plasma dru
APPLICATIONS OF PHARMACOKINETIC PRINCIPLES
Based
~o~c~ntra~wn. range for therapeutic effectiveness, the steps involved in th: tissues, the dose should be lesser on per Kg, total body weight basis but
tndtvtduahzatton of dosage regimen are:
more than that on IBW basis.
I. Estimation .o~ phannacokinetic parameters in individual patient 3. In case of drugs such as caffeine, theophylline, lidocaine and
and determmmg their deviation from the population values to lorazepam which distribute to the same extent in both lean and adipose
eva.lu~te the extent of variability; greater the accountability of tissues, the Vd is larger in obese patients but same on per Kg total body
va~JatJ.ons, better the chances of attaining the desired therapeutic weight basis; hence, dose should be administered on total body weight
obJective.
basis.
2. Attributi.ng the variability to some measurable characteristic such 4. For drugs such a phenytoin, diazepam and thiopental which are ·
as hepatic or renal disease, age, weight, etc. lipid soluble and distribute more in adipose tissues, the Vd is· larger per
3. Designing the new dosage regimen from the collected data. Kg body weight in obese patients and hence they require larger doses,
The design of new dosage regimen involves: . more than that o~ total body weight basis
1. Adjustment of dosage, or Changes in dose based on alteration of Vd is also-·attributed to modifi-
cati9n of clearance arid half-life of the drug.
2. Adjustment of dosing interval, or
3. Adjustment of both dosage and dosing interval. Dosing of Drugs in Neonates, · Infants and Children
The usual dosage regimen calculated on population basis refers to that
Dosi,n g of Drugs in Obese Patients
for adu~~s. Neopates, infants and children require different dosages than
The ~pparent v~lume. of distribution of a drug is greatly affected by adults because I 6f differences in body surface area, TBW and ECF on per
¢ha.nges m b,ody .we1ght smce the latter is directly related to the volume of Kg body weight basis. The dose for such patients are calculated on the
vanous boey flUJds . The ideal body weight (IBW) for men and women basis of their body surface area and not on body weight basis because the
I

can be calculated from following formulae : body surface area correlates better with dosage requirement, cardiac out-
IBW (men) = 50 Kg± I Kg/2.5 em above or below 150 em in height put, renal blood flow and glomeru~ar filtration in children. A simple
forrnula in comparison to DuBois and DuBois for computing surface area
( 13. I 7) (SA) in square meters' is Mosteller's equation:
IBW (women) = 45 Kg ± I Kg/2.5 em above or below 150 em in height
. (height x weight)Y2
SA ( tn m2) = ( 13. 19)
(13.18) 60
Any person whose body weight is more than 25% above the fBW is I

constdered obese. In such patients, the lean to adipose tissue ratio is Infants and children require larger mg/K.g doses than adults because:
small because of greater proportion of body fat which alters the v of 1. their body surface ar.ea per Kg body weight is larger, and hence
drugs. T~e ECF of adipose tissue is small in comparison to lean tissu~ in 2. larger volume of distribution (particularly TBW and ECF).
obese patrents.
The child's maintenance dose can be calculated from adult dose by
Foll?wing ge~1era/i::ations can be made regarding drug distribution and using the following equation:
dose adJustment tn obese patients: ·
SA nf Child in m2
I. For drugs such as digoxin that do not significantly distribute in Child's Dose= x Adult Dose (13.20)
.~. 73
the ~~cess body space. the Vd do not change and hence dose to be '

adn11n rstered should be calcu fated on I8 W basis. · where 1.73 is surface area in m2 of an average 70 Kg •
adult. Since the
.. 2. For polar drugs such as antibiotics (gentamicin) which distribute surface area of a chil.d is in proportion to the body ""eight according to
: ~

m excess body space of obese patients to an extent less than that in lean equation 13 .21 ,
SA (in m2) = Body Weight (in Kg)0·7 ( 13.21)
. .
:

APPLICATIONS OF PHARMACOKINETIC PRINCIPLES 319


318 BIOPHARMACEUTICS AND PHARMACOKINETICS
Dosing of D'r ugs in Renal Disease
the following relationship can also be .w ritten for child's dose: In patients with renal failure, the half-life of a drug is increased and its
clearance drastically decreased if it is predominantly eliminated by way of
Weight of Child in Kg 0.7
excretion. Hence, dosage adjustment should take into account the renal
Child's Dose = x Adult Dose (13.22)
70 function. of .the patient and t~e fraction of unchanged drug excreted in
urine. One such method was discussed in chapter 7 on Excretion of
As the TBW -in neonates is 30% more than that in adults,
Drugs.
1. the V d for most water soluble drugs is larger in infants, and
' There are two additional methods for dose adjustment in renal insuffi-
2. the V d for most lipid soluble drugs is smaller. ciency if the V d change is assumed to be negligible. These m~thods ~re
Acco.rdingly, the dose should be adjusted. based on maintaining the same average steady-state concentration durtng
. renal dysfunction as that achieved with the usual multiple dosage. regimen
Dosing of Drugs in Elderly and nonnal renal function. The adjustments are based on equations 13.6
Drug dose should be reduced in elderly patients because of a general and 13.7.
decline in body function with age. The lean body mass decreases and
1. Dose adjustment based on total body clearance : Rewriting
body fat increases by almost 100% in elderly persons as compared to
equation 13 .6, the parameters to be adjusted in renal insufficiency are
adults. Because of smaller volume of body water, higher peak alcohol
levels are observed in elderly subjects than in young adults. V d of a shown below:
1 ( Xo)
water soluble drug may decrease and that of a lipid soluble drug like -- F X X (13.6)
Css,av
diazepam increases with age. Age related chaQ.ges in hepatic and renal_ CIT 1

function greatly alters the clearance of drugs. Because of progressive t t t t


decrease in renal function, the dosage regimen of drugs that are predomi- to be kept assumed decreased needs
nantly. excreted unchanged in urine should be reduced in elderly patients. normal constant due to disease adjustment
A general equation that allows calculation of maintenance dose for a
I If CIT', X 0 ' and 1' represent the values for the renal failure 'patient, then
patient of any age (except neonates and infants) when maintenance of
the equation for dose adjustment is given as:
same Css av is desired is:
' Xo Xo'
. (Weight in Kg) 0·7 ( 140 - Age in years) c ss,av =CIT
-- (13.24)
Patient's Dose= x Adult Dose (13.23) 1 Clr' 1'
1660
Rearranging in tenns of dose and dose . interval to be adjusted, the
Dosing of Drugs in Hepatic Disease equation is:
Disease is a major source of variations in drug response. Both phar-
(13.25)
qjt:tcokinetics and pharmacodynamics of many drugs are altered by diseases 1' CIT 't
other than the one which is being treated.
From above equation, the regimen may be adjusted by reduction in
The influence of hepatic disorder on drug availability and disposition
dosage or increase in dosing interval or a combination of both.
is unpredict~ble because of the multiple effects that liver disease
produces effects on drug metabolizing enzymes, on drug binding and on 2. Dose adjustment based on elimination .rate constant or half-
hepatic blood flow. Hence, a correlation between altered drug pharmaco- life : Rewriting equation 13.7, the parameters to be adjusted in renal
kinetics and hepatic function is oft~ difficult. For example, unlike insufficiency are:
excretion, there are numerous pathways by which a drug may be metabo-
lized and each is affected to a different extent in hepatic disease. Generally
speaking, drug dosage should be reduced in patients with hepatic dysfunc-
tion since clearance is reduced and availability is increased in such a
situation.

,
320 BIOPHARMACEUTICS AND PHARMACOKINETICS
~· APPLICATIONS OF PHARMACOKINETIC PRINCIPLES 321
. 'fi:
,
•'
1.44 F Xo i.e. dosage regimen is titrated to a toxic endpoint e.g. excessive dryness
Css,av
-- .. X .• -
~

t!h X ( ) ( 13. 7)
vd r·
~
? --~
~

. '
t' of mouth with atropine when used as an antispasmodic agent. 1
:•

t t " -.·~
,...
f.

.. - t t Pharmacodynamic Monitoring
- ..
assumed ; ---
~
~
to be kept increased
.... needs '
.... ¢'

In some instances, the phartnacologic actions of a drug can be mea-


normal constant
.. -·
- due to disease
~-
.... ~...-;.. ...
adjustment
sured and used as a guide to therapeutic process. The response· observed
If tY2', X0 ' and t' represent the values for the renal failure patient, then: may or may not correlate exactly with the therapeutic effect e.g. blood
t!h X0 t!h' X0 ' glucose lowering with insulin, lowering of blood pressure with
Css,av = - -- - - - - (13.26) antihypertensives, enhancement of hemoglobin levels with hematinics, etc.
t t' .
Pharmacokinetic Monitoring
Rearranging the above equation in rt:et n1s of dose and dose ®erval to
be adjusted, we get: This approach involves monitoring the plasma drug concentration within
a target concentration range (called as target concentration strategy) and
-- - --- ( 13.27) based on the principle that free drug at the site of action is in equilibrium
with the drug in plasma. The strategy is particularly useful when:
Because of prolongation of half-life of a drug due to reduction in renal I. Therapeutic endpoints are difficult to defme or are lacking e.e_.
function, the. time taken to achieve the desired plateau takes longer the control of seizures with phenytoin.
more severe the dysfunction. Hence, such patients sometimes need load- 2. The objective is to maintain the therapeutic effect in order to
ing dose. obtain optimum drug use.
3. The probability of therapeutic failure is high as with drugs having
MONITORING DRUG THERAPY •
low therapeutic indices, erratic absorption, pharmacokinetic vati-
Management of drug therapy in. individual patient often requires evalu- ability or when the drug is used in multiple drug therapy.
ation of response of the patient to the recommended dosage regimen. Successful application of this approach requires complete knowledge
This is known as monitoring.. of drug therapy. It is necessary to ensure of pharmacokinetic parameters of the drug, the situation in which these
that the ·therapeutic objective is being attained and failure to ~o so re- parameters are likely to be altered and the extent to which they could be
quires readjustment of dosage regimen. altered, and a sensitive, specific and accurate analytic method for detetnti-
Depending upon the drug and the disease to be treated, management of nation of drug concentration. Examples of drugs monitored by this
drug therapy in individual patient can be accomplished by: •
meth9d include digoxin, phenytoin, gentamicin, theophylline, etc. The
1. Monitoring therapeutic effects therapeutic monitoring strategy is very useful in individualizi~g therapy in patients with hepatic
or renal impair tnent when dose adjustments are necessary.
2. Monitoring pharmacologic actions pharmacodynamic monitoring
3. Monitoring plasma drug concentration pharmacokinetic monitoring.
QUESTIO~S
Therapeutic Monitoring
1. Enlist various applications of pharmacokinetic principles.
In this approach, the management plan involves monitoring the inci-
2. What do you understand by therapeutic objective? What are the various
dence and intensity of both the desired therapeutic effects and the undesired ways of attaining it?
adverse effects.. A direct measure of the desired effect is considered as a
. therapeutic endpoint e.g. prev~ntion of an anticipated attack of angina or 3. Define optimal dosage regimen. What does it mean for a drug with narrow
th.e rapeutic index such as phenytoin and for drugs like antibiotics?
shortening of duration. of pain when .attack occurs through the use of
sublingual glyceryl trinitrate. In certain cases, definition of therapeutic 4. What assumptions are made in the design of a dosage regimen?

endpoint may not be clear and onset of toxicity is used as a dosing guide 5. Name the two parameters that are generally adjusted in developing a dosage

reg1men.

'
322 BIOPHARMACEUTICS AND PHARMACOKINETICS
323
6. It is safer to administer a drug with a narrOw therapeutic index in small
doses at frequent intervals. Explain. ._
7. On what parameters does extent of drug accumulation on multiple dosing
depend? Does dose size have a bearing on it? F Clr Therapeutic Range

8. State the plateau principle. Which parameters govern attainment of steady- 0.4 1.23 l/Kg 118.4 mllmin 0.2 to 1.3 mcg/ml
state?
' Design a dosage regimen to attain and rnaintain the plasma concentration
9. Define fluctuation of plasma level. Why are fluctuations smaller when the within the therapeutic range. Assume rapid absorption.
drug is given e.v. whereas larger when administered as multiple i. v. boluses?
Answer : C85 av = 0.588 mcg/ml, 'tmax = 16.21 hours, Xo,max = 169.12mg. It
10. The C 55 av is not an arithmetic mean of C ss,max and Css,min· Why? is convenient to administer the drug once every · 12 hours. Therefore, Xo in
'
11. Define dose ratio. Why is it always smaller for extravascularly administered such a ·situation should be 125.3 mg. Loading dose is 90.4 mg.
drugs in comparison to those given intravenously? ·
30. Diltiazem is administered in a dose of 60 mg q.i.d. The oral availability of
12. On what factors do maintenance of drug concentration within the therapeutic the drug is 50o/o, Vd is 30 liters and elimination half-life is 4 hours.
range depend? a. Determine the maximum and minimum amounts of drug in the body after 4
_ 13. How is dosing interval determined on the basis of half-life and therapeutic doses.
index of the drug?
Answer : X 4 ,max = 45.7 mg and X4,min = 16. 16 mg.
14. What are the two major sources of variability in drug response?
~

b. Calculate the maximum and minimum amounts of drug in the body at


15. Quote some of the more important causes of intersubject variability in drug steady -state.
response.
Answer : X ss,max = 46.4 mg and Xss,min = 16.4 mg.
16. Enlist the steps involved in individualization of dosage regimen.
c. What is the accumulation index?
17. How does obesity influence Vd of a drug and hence its dose size?
Answer : Rae = 1.55 .
18. The dose for neonates, infants and children should be calculated on body
d. What is the average amount of drug in the body at steady-state?
surface area basis rather than_on the basis of body weight. Explain.
Answer : X ss,av = 28.8 mg.
19. Why do neonates, infants and children require larger mg/Kg body weight
doses than adults? e. If the desired C ss.av is 0.099 mcg/ml, is there any need to administer the
loading dose?
20. Give possible reasons for reduction in dose of a drug in elderly patients.
31. Netihnicin is to be administered to a 70 Kg patient at a rate of 2 mg/Kg
21. Unlike renal function, why is it difficult to establish a correlation between
every 12 hours by multiple i.m. injections. The drug has a half-life of 2.2
hepatic dysfunction and altered drug pharmacokinetics ?
hours and Vd of 0.2 l/Kg.
22. Why does it take longer to attain the steady-state in a renal failure patient in
a. Determine Css,max' C ss,min and Css,av ·
comparison to a patient with normal renal function?
Answer : C ss,max = 10.2 mcg/ml, Css,min = 0.23 rncg/ml and
23. What do you understand by monitoring of drug therapy? When does it
C 58 av = 2.64 meg/mi.
become necessary? '
b. If the t•;2 increases to 5 hours in renal insufficiency, what should be the new
24. What are the various ways of monitoring drug therapy in individual patient?
dose or the new dosing interval?
25. What parameters are monitored in therapeutic drug monitoring?
I
Answer : New dose ('! unchanged) = 0.88 mg/Kg, new dosing interval (dose
26. On what principle is pharmacokinetic drug monitoring based? When does unchanged) = 27.3 hours.
such a strategy becomes useful?
' '
c. If the drug is to be injected once every 24 hours, in renal insufficiency,
27. What criteria are necessary for successful pharmacokinetic drug monitoring? what should be the new dose?
28. The loading dose is calculated on the basis of appa'rent Vd of a drug Answer : 1. 76 rhg/Kg.
whereas maintenance dose is determined from its tv,. Explain.
3.24 BIOPHARMACEUTICS AND PHARMACOKINETICS

d. If netilmicin is to be given to a child weighing 12 Kg with normal renal


function, what should be the dose? I

Answer : 3.4 mg/Kg.


e. If Ute same drug is to be administered to an elderly patient of 85 years and
55 Kg body weight, determine the dose.
Answer : 1.4 mg/Kg.
I
14
32.. Procainamide is to be administered to a 65 Kg arrhythmic patient as 500 mg
tablet.s every 4 hours. The drug has a half-life of 3 hours, Vd of 2 liter/J(g Drug Concentration and
and oral availability of 0.85.
a. Calculate the steady-state concentration of procainamide. Pharmacologic Response
Answer : Css,av = 3.53 meg/mi. -
b. Determine whether the patient is adequately dosed (therapeutic range . is 4 to .
12 mcg/ml) The empirical approach in optimization of drug 'therapy was based on
c. What changes would you recommend in the dosage regimen? relating phar tnacologic response to the dose administered. There are
- several drawbacks of such an approach tedious, time consuming, costly,
Answer : Desired Css:av = 7.3 mcg/ml which can be attained by increasing
the maintenance dose to 1000 mg or 2 tablets every 4 hours.
etc. Moreover, poor correlation may be observed between drug dosage
and response since a given dose or a dosing rate can result in large
33. Two aminoglycoside antibiotics alongwith some of their parameters are
deviations in plasma drug concentration which in most cases are attribut-
listed in the table below:
able to formulation factors and drug's elimination characteristics. ~t is.
Drug t'h · Fe TI Route Normal Normal well understood that the response correlates better with the plasma drug
dose interval concentration or with the amount of drug in the body rather than with the
Streptomycin 3 Hr 0.5 1.25 •
t.m. 7.5 mg/Kg 12 Hrs administered dose. Thus, it is easy and more appropriate to design dosage
Gentamicin 2 Hr 0.9 20.00 •
t.m. 1.0 mg/Kg regimens by application of phannacokinetic principles. The approach is
8 Hrs

based on the principle that the response produced is proportional to the
These drugs are to be administered to a uremic--patient. Suggest the changes concentration of drug at the site of action which in tum is reflected in the
that should be made ' in the dosage regimen-keeping the dose constant and concentrat;on of drug in plasma. However, such relationships between
-prolo11ging the interval or decreasing the dose and maintaining the dosage
plasma drug concentration and phannacologic response are difficult to
interval or changing both. Discuss the advantages and disadvantages of
each approach. establish \n certain cases.
The factors that complicat~ development of concentration-response re-
lationships are:
1. Delay in Drug Distribution· : Most sites of action are located in
the extravascular tissues and equilibration with drug takes a long time.
This results in delay in observation of response. Long delays may occur·
when the therapeutic response is an indirect measure of drug effect for
example, the anticoagulant effect of dicoumarol is due !O indirect inhibi-
tion and depletion of clotting factors. Often, the therapeutic action in
several such cases outlasts the plasma drug concentration e.g. reserpine.
2. Protein Binding : Ideally, the pharmacologic response should be
correlated with' the concentration of unbound drug in plasma since its
activity is elicited onJy by the free form. H~wever, quantification of
unbound drug is difficult and thus. if the relationship is based on total
'

325
326 BIOPHARMACEUTICS AND PHARMACOKINETICS DRU~G CONCENTRATION AND PHARMACOLOGIC RESPONSE 327
.... J '

plasma drug concentration, any variation in the degree of binding will Some drugs can be used to treat several diseases and will have differ-
obscure it. ent ranges for different conditions e.g. salicylic acid is useful both in
..
3. Active Metabolites : The metabolites of some drugs such as common aches and pains as well as in rheumatoid arthritis. The upper.
imipramine and amitriptyline are active which obscure the concentration- . limit of the therapeutic range may express loss of effectiveness with no .
'
response relationship if it is based on the concentration of parent drug. toxicity· e.g. tricyclic antidepressants or reflect toxicity which may be
Some drugs such as propranolol fonn active metabolites on first-pass related to phannacologic ·effect of the drug e.g. hemorrhagic tendency
hepatic metabolism and thus, result in greater response when administered · with warfarin or may be totally unrelated e.g. ototoxicity with
orally than when given intravenously. aminoglycosides.
4. Tolerance : The effectiveness of some drugs decreases with chronic
Concentration-Response Relationships
use due to development of acquired tolerance. Tolerance may be phartna-
With most drugs, the response produced is reversible i.e. a reduction
cokinetic i.e. through enhanced metabolism e.g. carbamazepine . or
in concentration at the site of action reverses the effect. The response
pharmacodynamic i.e. through diminished response after some period of ·
produced by a drug can be classified as graded or quanta/. A majority of
chronic use e.g. nitroglycerine. ' '

drugs produce gr-aded response i.e. the intensity of effect increases with
5. Racemates : Several drugs are administered as racemic mixture the dose or concentration of drug. The response. can be measured on a .
of two optically active enantiomers of which usually one is active; for continual basis in such cases and establishing a linear relationship between
example, the S(+) isomer of ibuprofen. Hence, a difference ·i n the ratio of drug concentration and intensity of response is easy. However, for certain
active to inactive isomer can lead to large differences in pharmacologic other drugs, the responses are not observed on a continuous basis. Such'
response. drugs may either show their effect or not at all for example, prevention
Therapeutic Concentration Range of seizures by phenytoin. Such responses are called as quantal or ali-or-
' none. Thus, establishing a concentration-response relationship in such
The therapeutic effectiveness of a drug depends upon its plasma con-
circumstances is difficult but can be developed in teuns of the frequency
centration. There is an optimum concentration range in which therapeutic
with which a partic~.dar event occurs at a given drug concentration.
success is most likely and · concentrations both above and below it are
more hannful than useful. This concentration range is called as thera- Afathe.lnatical tnodels that relate pharmacologic effect to a measured
peutic window or therapeutic range. Such a range is thus based on the drug concentration in plasma or at the effector site can be used to ~
difference between pharmacoiogic effectiveness and toxicity. The wider i~ ' de,·elop quuntitati\'e relationships. Such models are often called as ·
is, the more is the ease in .establishing a safe and effective dosage regi- pharmacodynamic models. Some of the commonly used relationships or
men. The therapeutic range of several drugs have been developed. For tnodels are discussed below.
many, the range is narrow and for others, it is wide (see Table 14.1). 1. Linear Model : When the pharmacologic effect (E) is directly
' .
proportional to the drug concentration (C), the relationship may be written
TABLE 14.1 Therapeutic Range for Some Drugs
as:
Drug Disease Therapeutic Range E =PC+ E0 (14.1)

Digoxin · Congestive cardiac failure 0.5 to 2.0 ng/ml where P is the slope of the line obtained fron1 a plot of E versus C and E0
'

Gentamicin Infections 1.0 to 10.0 mcg/ml is the extrapolated y-intercept called as baseline effect in the absence of .
Lidocaine Arrhythmias 1.0 to 6.0 mcg/ml
drug.
.....

Phenytoin Epilepsy 10.0 to 20.0 mcg/ml 2. Logarithmic Model : If the concentration-effect relationship does
.
Propranolol · Angina 0.02 to 0.2 mcg/ml not confonn to a sitnple linear function , a logarithmic transfonnation of
Salicy Iic acid Aches and pains 20.0 to 100.0 mcg/ml the data is needed.
E = P loo::::- C +I •
( 14.2)
Rheumatoid arthritis 100.0 to 300.0 mcg/ml
· 'Theophylline Asthma 6-.0 to 20.0 mcg/ml
329
318 BIOPHARMACEUTICS AND PHARMACOKINETICS DRUG CONCENTRATION AND PHARMACOLOGIC RESPONSE

where I is empiric constant. This transfottnation is pop~lar because it maximum (asymptote) at high concentrations (Fig. 14.2). Such a plot is
expand~ the initial part of the curve where response is changing markedly characteristic of most concentration-response curves. None of the preced-
with a small change in concentration and contracts the latter part where a ing models can account for the maximal drug effect as the Emax models.
large change in concentration produces only a slight change in response. Michaelis-Menten equation for a saturable process (saturation of recep-
_An .important feature of this transfotntation is the linear relationships tor sites by the drug molecules) is used to describe such a model.
between drug concentration and response at concentrations producing ef-
fects of between 20 to 80% of the maximum effect (Fig. 14.1 ). Beyond E = .Emax C (14.3)
this range, a larger dose produces a larger concentration of drug in the Cso + c
body.
where Emax = maximum effect, and
100
·- ....- Cso = the concentration at which 50% of the effect is produced.
When c << c , the equation reduces to a linear relationship. In the -
80 50
range 20 to 80%, the Emax model approximates equation 14.2. .
0 In certain cases, the concentration-response relationship is steeper or
~ shallower than that predicted from equation 14.3. A better fit may
w 50 slope=P
?fl. otherwise be obtainedby considering the shape factor 'n' to account fo~
deviations from a perfect hyperbola .
T . Region 1. Region 2 Region 3
20
E = Emax en (14.4)
Cso" +en
If n = 1, a normal plot is obtained. Larger the value of n, ste~per the
0
linear portion of the curve and greater its slope; such a plot IS oft~n
'

0.1 0.3 3.0 10.0


--+• log Concentration sigmoidal and the model may be called as sigmoid-Emax model (Fig.
*ig. 14.1 A typical sigmoidal shape log drug concentration-effect relationship 14.3).

,,. ----
3. Emax Models : Unlike earlier models, these models describe nonlinear .,.
concentration-effect relationships i.e. the response increases with an in- ,, n>1

crease in drug concentration at low concentrations and tends to' approach I

... ____ ..... _


,
I n=1
0 I
100r-----------------------~ ~ ___ •

w , n< 1
,' I
I ' II
T I ,

,_ ~'
0 I
CD
:c:
w 50
?fl. I
I
T I
I
Fig. 14.3 Effect of shape factor n on the concentration-response curves

:cso •

. Equation 14.3 can be linearized in several ~ays. _One linearization is


--+• Concentration
obtained by taking the reciprocal and constructmg a Lmeweaver-Burk plot
Fig. 14.2 A hyperbolic concentration-response relationship based on Emax model
(equation 14.5 and Fig. 14.4).
I

I
330
BIOPHARMACElJTICS AND PHARMACOK~NETICS DRUG CONCENTRATION AND PHARMACOLOGIC RESPONSE 331

1 Cso 1 The factors which influence duration of action of a drug are:


- - -- + ( 14.5) 1. The dose size, and
2. The rate of drug removal from the site of action which in tum
depends upon the redistribution of drug to poorly perfused tissues
1
- and elimination processes.
E
An increase in dose promotes rapid onset of action by reducing the
slope=
c50 time required to reach the Cmin and prolongs the duration of effect. The
' Em ax influence of dose on duration of action can be explained as follows.
Consider a drug that distributes rapidly (one-compartment kinetics) and
.. administered as i.v. bolus dose. The plasma drug concentration is given
by the equation:
------~--~1--~-------- Dose) KE t
- 4)- log C = log ( - --- ( 14. 7)
c Vd 2.303
Fig. 14.4 Lineweaver-Burk plot of equation 14.5. The plasma concentration falls ev~ntually to a level Cmin below which
a linearized form of the hyperbolic plot the drug does not show any response. At this time, t = td, the duration of
Adnother lin~ar plot is similar to Scatchard or Eadie-Hofstee plots and effect of a drug. The above equation thus becomes:
base on equation 14.6.
Dose) KE td (14.8)
log Cmin = log ( - - --
_E = - ( I ) E + Emax Vd 2.303
c . Cso Cso . (14.6)
Rearranging to define duration of effect, the equation is:
A plot of EIC versu s E yields a line with slope - 1/C- d .
Emax (Fi b·
a 14 S)
. .
- ) 0 an x- Intercept 2 .303
td = log Dose - log ( Cmin V d) ( 14.9)
KE
where Cmin V d = Xmin ' the minimum amount of drug in the body required
to produce a response. A plot of td versus log dose yields a straight line
.·E with slope 2.303/KE and x-intercept at zero duration of effect of log Xmin
-
c 1 (Fig. 14.6).
slope=--
J Cso
......
0
..
. ~
• w
'+-
- 0
c:
• "'
E 2.303 ..
-
- -4-) 0
·-as slope=
..... KE
Fig. 14.5 A Scatchard or Eadie-llofstcc type of plot of equation 14.6 ::J
Cl
log xmin
Onset/Duration of Effect-Concentrat'ion Relationships
The o~set of action of a drug that produces quanta! res onse occur
,
I !
when a n11nin1un1 effective level of druo in the plasma C p · h ds - --..) log Dose
The duratio f · f b mm IS reac e
n o ~ction o such a drug will depend upon how lono th~ Fig. 14.6 Relationship between dose of drug and duration of action
plasrna concentration ren1ains above the c rnm
. 1eve 1. b
333
DRUG CONCENTRATION AND PHARMACOLOGIC RESPONSE
332 BIOPHARMACEUTICS AND PHARMACOKINETICS
14.1). If a drug with rapid distribution characteristics is given as i.v.
Equation 14.9 shows that the duration of effect is also a function of t~ bolus dose large enough to elicit a maximum response, the log con.centr~­
(0.693/KE). With each doubling of dose the duration of effect increases tion-response plo~ obtained will be as shown in Fig. 14.1: The relatto~shtp
by one half-life. This can be explained by considering that a dose X0 between dose intensity of effect and time can be estabhshed by constder-
' .
produces a duration of effect td; so when double the dose i.e. 2X0 is ing the plots depicted in Fig. 14.7. which also shows 3 regions.
administered, the dose remaining after one half-life will be X 0 which can Region 3 indicates 80 to 100% maxim~m ~espo~se. !he .initial con-
produce a duration of effect equal to td· Thus, the total duration of effect centration of drug after i.v. bolus dose hes m thts regton tf the d~se
produced by 2X0 will be t~ + td. However, the approach of extending the injected is sufficient to elicit maximal response. The ~g conc~ntrat10n
duration of action by increasing the dose is hattnful if the drug has a falls raPidly in this region but intensity of response remams maxtmal and
narrow therapeutic index. An alternative approach is to administer the
almost constant with time.
same dose when the drug level has fallen to Xmin· Thus, after the second
dose, the drug in the body will be (dose + Xmin). If X min is small in
Region 2 denotes 20 to 80% maximum response. In this region, the
relation to dose, very little increase in duration of action will be observed. intensity of response is proportional to log of drug concentration and
expressed by equation 14.2.
-
Intensity of Effect-Concentration Relationships Intensity of Effect= P log C +I (14.2)
In ca~e of a drug that produces quantal response, the pharmacodynamic
parameter that correlates better with its concentration is duration of Since the decline in drug concentration is a frrst-order process, log C
action. The parameter intensity of response is more useful for correla- can be expressed as:
tion with the concentration of a drug that shows graded effect. Like KE t
log C =log C 0 - (14.10)
duration of action, intensity of action also depends upon the dose and rate 0

2.303
of removal of drug from the site of action. The intensity of action also
depends upon the region of the concentration-response curve (refer Fig. Substituting 14 . 10 in equation 14.2. and rearranging we get:
' P KE t
I.V. Slug Intensity of Effect= (P log Co+ I)- (14.11)
J, 2303
--------~--------------~-------------100
If E is the intensity of response when concentration is Co, then:
0
c
g ~r-------~ ~--------------~------------~80 ~
P KE t
Intensity of Effect= E 0 - . (14.12)
~ - - Intensity of ~ 2.303
B Effect-Time g_
5 Curve ~ Equation 14.12 shows that the intensity of response falls linearly (at a
u -~
2~~~~---4------~ ~------+-------------~so
·-
e constant zero-order rate) with time in region 2. This is true for most of
·the drugs. The drug concentration however declines logarithmically or
0
tV
E
exponentially in region 2 as shown by equation .14.1 0. -· _
-Cl"'
· tV
Region 1 denotes 0 to 20% maximum response. In this regi~n, the
:---------20
REGION 1 ·
i intensity of effect is directly proportional to the .drug concentratto~ but
falls exponentially with time and parallels the fall In drug concentration.

-4• Time QUESTIONS


1. Why is it that the pharmacologic effect Qf a drug shows better correlation
Fig. 14.7 The fall in intensity of response with drug concentration and with
time follo\ving administration of a single i.v. bolus dose (Adapted with its plasma concentration than with "its dosage?
frotn Rovvland.M. and Tozer. T.N.: Clinical Pharmacokinetics: 2. What are some of the more important causes of poor correlation between
('oncepts and Applicatlons. 2nd edition. Philadelphia. Lea & plasma drug concentration and pharmacologic r~sponse?
Febiger. 1989).
334 !~ BIOPHARMACEUTICS AND PHARMACOKINETICS

3. Define therapeutic range. What does plasma drug concentration beyond this
range signify?
4. Define: (a) Quantal response, and (b) Graded response.
5. What are pharmacodynamic models? Discuss some of the more important
models used to quantify plasma concentration-response relationships.
15
6. When does onset of action occur for a drug that shows graded response and
for the one that elicits quantal response?
Controlled Release
7. On what factors do duration and intensity of drug action depend?
8. Explain how with each doubling of dose, the duration of effect increases by
Medication
one half-life for a drug that elicits graded response.
9. When and why is it not advisable to extend the duration of action by
increasing the dose? What is the alternative in such cases? An ideal dosage regimen in the drug therapy of any disease is the one
which immediately attains the desired therapeutic concentration of drug in
10. How does intensity of drug action change with its plasma concentration?
plasma (or at the site of action) and maintains it constant for the entire
11. The Cso of a drug showing graded response is 5 mcg/ml and has a shape
duration of treatment. This is possible through administration of a con-
factor n of 2.0. Calculate the concentration range if the drug is found to be
ventional dosage form in a particular dose and at a particular frequency.
effective when the response is between 20 to 80% of the maximal value.
(Hint: Take reciprocal of equation 14.4 and express intensity of effect as 0.2 The frequency of administration or the dosing interval of any drug de-
Emax and solve to obtain C20· Similarly, take it next as 0.8 Emax and solve · pends upon its half-life or mean residence time (MRT) and its therapeu~ic
to determine c 80 ) ... index. In most cases, the dosing interval is much shorter than the half-hfe
Answer : C2o = 2.5 mcg/ml and Cso = 10.0 meg/mi. of the drug resulting in a number of limitations associated with such a
12. ~ez.locillin has a MEC of 5 mcg/ml for a particular infection when given. conventional dosage fonn:
1. v. tn a bolus dose of 2.5 mg/Kg. What will be the duration of effect if a 1. Poor patient compliance; increased chances of missing the dose of
dose of 50 mg/Kg is administered? The drug has a half-life of 0.8 hours. a drug with short half-life for which frequent administration is
What will be the duration of effect if the dose is doubled? Is the increase in·· necessary.
duration of action equal to one half-life?
2. A typical peak-valley plasma concentration-time profile is ob-
Answer : 3.5 hours and 4.3 hours. tained which makes attainment of steady-state condition difficult
13. A newly developed antihypertensive drug shows linear relationship between (see Fig. 15.1).
intensity of effect and log drug concentration. After i. v. administration of
3. The unavoidable fluctuations in the drug concentration may lead
0. 15 mg/Kg dose, the observed reduction in blood pressure with time is
shown in the table below: to undermedication or overmedication as the C ss values fall or
Time (hours) 0.5 3.0 rise beyond the therapeutic range.
% Reduction in B.P. 45 .0 40.0 4. The fluctuating drug levels may lead to precipitation of adverse
effects especially of a drug with small therapeutic index whenever
Given : P = 20.0%
overmedication occurs.
a. ~hat .is the half-life of the drug? (Hint: Use equation 14.12 for a plot of
tntenstty of effect versus time and comput~ values from slope) There are two ways to overcome such a situation:
Answer : t~ = 3 hours. 1. Development of new, better and safer drugs with long half-lives
b. What will be the o/o intensity of response immediately after injection of and large therapeutic indices, and
drug? 2. Effective and safer use of existing drugs through concepts and
Answer : 46.0o/o. techniques of controlled and targeted delivery systems.
c. How long is the reduction in B.P. expected to remain above 20.0% of
maximum response?


337
336 BIOPHARMACEUTICS AND PHARMACOKINETICS CONTROLLED RELEASE MEDICATION

The first approach has many disadvantages which therefore resulted in The several advantages of a controlled drug delivery system over a
increased interest in the second approach. An ideal controlled drug conventional dosage fottn are ,-
delivery system is the one which delivers the drug at a predetermined 1. Improved patient convenience and compliance due to less fre-
rate, locally or systemically, for a specified period of time. Thus, unlike quent drug administration. ·
conventional immediate release systems, the rate of appearance of drug in
2 Reduction in fluctuation in steady-state levels and therefore better
the body with such a system is not controlled by absorption process. . control of disease condition and reduced intensity of local or
Following absorption of drug from such a system, there is no control over systemic side effects (Fig. 15.1 ). . ··
its fate. An ideal targeted drug delivery system is the one which
3. Increased safety margin of high potency drugs due to better con-
delivers the drug only to its site of action and not to the nontarget organs
trol of plasma levels.
or tissues. With this approach, control of kinetics of drug release is
difficult. This chapter will deal only with controlled drug delivery sys- 4. Maximum utilization of drug enabling reduction in total. amount
tems. of dose administered.
There are several tenus used interchangeably viz. controlled release, 5. Reduction in health care costs through improved thera?y, ~horter
programmed release, sustained release, prolonged release, timed release, treatment period, less frequency of dosing ~nd red~ct1on tn per-
slow release, extended release and other such dosage fonns. However, sonnel time to dispense, administer and monitor patients.
controlled release will imply as defined earlier. It differs from sustained Disadvantages of controlled release dosage fottns include
l. Decreased systemic availability in comparison dto i~ediate r~­
1
release systems which simply prolong the drug release and hence plasma
drug levels for an extended period of time (i.e. not necessarily at a lease conventional dosage fonns; this may be ue ~o tn~o.mp ~ e
predetettnined rate). Thus, the chief objective of most products should be release, increased first-pass metabolism, increased ~nstabiltty,. In-
controlled delivery to reduce dosing frequency to an extent that once daily sufficient residence time for complete release, site-specific absorplton,
dose is sufficient for therapeutic management through a unifortn plasma pH-dependent solubility, etc.
concentration at steady-state.
2. Poor in vitro-in vivo ·correlation.
P ·b·l·ty of dose dumping due to food, physiologic or fottnula-
3 . 0 SSl l l . · b th
Peak tion variables or chewing or grinding of oral fottnulations Y e
..- (Css. max> Toxic level patient and thus, increased risk of toxicity .
c 4. Retrieval of drug is difficult in case of toxicity, poisoning or
·-
0
......
---- ----- -- MSC
0
1!
...... :g - Zero-order hypersensitivity reactions.
c
B .---...,__.....,._...,__-+---+---+---t-... X ~ - - controlled 5. Reduced potential for dosage adjustment of drugs nottnally ad-
c f! ~ release
0
()
Q)
.r::.
ministered in varying strengths.
......
0)
:::J
~
---- . - - - MEC 6. Higher cost of fonnulation. -.
c
C'O +Valley Subtherapeutic level
E
(/) (Css, min) DESIGN OF CONTROLLED DRUG DELIVERY SYSTEMS
-a.C'O
~,, Multiple dosing of Sustained release The basic rationale of a controlled drug delivery syste~ is to opt~ize
conventional dosage .
the biophannaceutic, pharmacokinetic ~d ph~n~acodynanuc prope~1es ~f
-... ..
form·-.......... Single dose of a
. '
•• ············conventional tablet/capsule
a drug in such a way that its utility 1s max1m1zed through red~ctlo~- m
side effects and cure or control of condition in the shortest posstble. t1me ,
by using . sm~llest quantity of drug, administered by the most sUitable
--+)! T.ime
,
Fig. 15.1 A hypothetical plasma concentration-time profile from , route.
conventional multiple dosing and single doses of

sustained and controlled delivery formulations
CONTROLLED RELEASE MEDICATION 339
Biopharmaceutic Characteristics of the Drug
The performance of a drug presented as a controlled release system 2. Aqueous Solubility of the Drug : A drug with good aqueous
depends upon its: solubility, especially if pH-independent, serves as a good candidate for
1. Release from the formulation. controlled release dosage forms e.g. pentoxifylline. Drugs with pH-depen-
dent aqueous solubility e.g. phenytoin, or drugs with solubility in nonaqueous
2. Movement within the body during its passage to the site of action.
solvents e.g. steroids, are suitable for parenteral (e.g. i.rn depots) con-
The former depends upon the fabrication of the fonnulation and the trolled r~lease dosage fonns; the drug precipitates at the injection site and
physicochemical properties of the drug while the latter element is depen- thus, its release is slowed down due to change in pH or contact with
~ent upon phannacokinetics of drug. In comparison to conventional aqueous bod}' fluids. Absorption of poorly soluble drugs is dissolution
dosage fotm where the rate-limiting step in drug availability is usually rate-limited which means that the controlled release device does not con-
absorption through the biomembrane, the rate-detennining step in the trol the absorption process; hence, they are poor candidates for such
availability of a drug from controlled delivery system is the rate of release systems.
of drug from the dosage fonn which is much smaller than the intrinsic
3. Apparent Partition Coefficient of the Drug : Greater the appar-
absorption rate for the drug (Fig. 15.2).
ent partition coefficient of a drug, greater is its rate and extent of absorption.
. Such drugs have increased tendency to cross even the more selective
RELEASE Drug at the ABSORPTION Drug in barriers like BBB. The apparent volume of distribution of such drugs also
Drug in
Dosage Form Absorption Site the Body increases due to increased· partitioning into the fatty tissues and since the
blood flow rate to such tissues is always lower than that to an aqueous
tissue like liver, they may exhibit characteristics of models having two or
'
more compartments. The parameter is also important in detennining the
Rate-limiting step Rate-limiting step
of controlled drug release rate of a drug from lipophilic matrix or device.
of conventional
delivery systems dosage forms 4. Drug pK8 and Ionization at Physiologic pH : The pK3 range
..
Fig. 15.2 Scheme representing the rate-limiting step in the for acidic drugs whose ionization is pH-sensitive is 3.0 to 7.5 and that for
design of controlled drug delivery system basic drugs is 7.0 to 11.0. For optimum passive absorption, the drugs
should be nonionized at that site at least to an extent 0.1 to 5%. Drugs
The type of delivery system and the route of administration of the existing largely in ionized fonns are poor candidates for controlled deliv-
drug presented in controlled release dosage foun depends upon the physi- ery e.g. hexamethonium.
cochemical properties of the drug and its biophat n1aceutic characteristics.
5. Drug Stability : Drugs unstable in GI environment cannot be
The desired biophannaceutic properties of a drug to be used in a con-
administered as oral controlled release forrnulation because of bioavailability
trolled delivery system are discussed below.
problems e.g. nitroglycerine. A different route of administration should
1. Molecular Weight of the Drug : The lower the molecular weight, then be selected such as the transdet tnal route.
the faster and more complete the absorption. For drugs absorbed by pore
6. Mechanism and Site of Absorption : Drugs absorbed by carrier-
transport mechanism, the molecular size tl!reshold is 150 daltons for
mediated transport processes and those absorbed through a window are
spherical compounds and 400 daltons for lineai compounds. However,
poor candidates for controlled release systems e.g. several B vitamins.
more than 95% of drugs are absorbed by passive diffusion. Diffusivity,
defined as the ability of a drug to diffuse through the membranes, is 7. Biopharmaceutic Aspects of Route of Administration : Oral
inversely related to molecular size. The upper limit of drug molecular and parenteral (i.m.) routes are the most popular followed by transdennal
size for passive diffusion is 600 daltons. Drugs with large molecular size application. Routes of minor importance in controlled drug delivery are
are poor candidates for oral controlled release systems e.g. peptides arid buccal/sublingual, rectal, nasal, ocular, pulmonary, vaginal and in.trauterinal.
proteins. . The features desirable for a drug to be given by a particular route are
discussed below.
CONTROLLED RELEASE MEDICATION •
341

340 BIOPHARMACEUTICS AND PHARMACOKINETICS 2. Elimination Half-Life : Smaller the ty2 , larger the amount of drug
to be incorporated in the controlled release dosage fon·n. For drugs with
(a) Oral Route : For a drug to be successful ·)is oral controlled t~ less than 2 hours, a · very large dose may be required to maintain the
release fottnulatioJ1, it must get absorbed throtrgh the entire length high release rate. Drugs with half-life in the range 2 to ·4 hours make
of GIT. Since -the main limitation of this rotlte is ·the transit time good candidates for such a sy Item e.g. propranolol. Drugs with long half- ·
( a mean of 14 hours), the duration of action can be extended for life need not be presented i such a founulation e.g. amlodipine. For
12 to 24 hours. The route is suitable for drugs given in dose as some drugs e.g. MAO inhib · ors, the duration of ac~ion is longer than that
high as 1000 mg. A drug whose absorption is pH dependent, predicted by their half-live:s;- A candidate drug must have t~ that can be
destabilized by GI fluids/enzymes, undergoes extensive presystemic correlated with its phattn~cologic response. In tet nts of MRT, a drug
metabolism (e.g. nitr9glycerine ), influenced by gut motility, has administered as controlled release dosage form should have MRT signifi-
an absorption window and/or absorbed actively (e.g. riboflavin), cantly longer than that from conventional dosage fortns.
is a poor candidate for oral controlled release formulations.
3. Rate of Metabolism : A drug which is extensively metabolized is
(b) Intramuscular/Subcutaneous Routes : These routes are suitable
suitable for controlled release syste~ as long as the rate of metabolism is
when the duration of action is to be prolonged from 24 hours to
not too rapid. The extent of metabolism should be identical and predict-
12 months. Only a small amount of drug, about 2 ml or 2 grams,
able when the drug is administered by different routes. A drug capable of
can be administered by these routes. Factors important in drug inducing or inhibiting metabolism is a poor candidate for such a product
release by such routes are solubility of drug in the surrounding
since steady-state blood levels ·would be difficult to maintain.
tissues, molecular weight, partition coefficient and pKa of the
drug and contact surface between the drug and the surrounding 4. Dosage Form Index (DI) : It is defined as the ratio ofCss.max to
tissues. .
CSS.f11111· Since the oooal of controlled
- release formulation is to improve
therap) by reducing the dosage form index \\'hile maintaining the plasma
(c) Transdermal Route : Low dose.....trugs like nitroglycerine can be
drug levels within the therapeutic "·indo\\:. ideally its value should be as
administered by this route. The route is best· suited for drugs
close to one as possible.
showing extensive first-pass metabolism upon oral administration.
Important factors to be considered for percu~aneous drug absorp- Pharmacodynamic Characteristics of the Drug
tion are partition coefficient ~f drug, contact ~rea, skin condition,
t. Therapeutic Range : A candida_te drug for controlled delivery
skin permeabiuty of drug, skin perfusion rate, etc.
systen1 should have a therapeutic range \\'"ide enouih such that variations
In short, the main detenninants in deciding a route for administration in the release rate do not resu It in a concentration beyond this level.
of a controlled release system are physicochemical properties of the drug,
dose size, absorption efficiency and desired duration of action. 2. Therapeutic Index (TI) : The release rate of a drug "·ith narrO\\'
therapeutic index should be such that the plasn1a concentration attained is
Pharmacokinetic Characteristics of th~ Drug within the therapeutically safe and effective range. This is necessary
A detailed knowledge of the ADME characteristics of a drug is essen- because such drugs have toxic concentration nearer to their therapeutic
tial in the design of a controlled release proQtict. An optimum ran-ge of a range. Precise control of release rate of a poten-t drug \Vith narro-w· margin
"" .
·given pharmacokinetic parameter of a drug is necessary beyond which of safetv is difficult. A drug \\:ith · short half-life and narrow therapeutic
r '--

controlled delivery is difficult or impossible. index should be ---adrninistered n1ore frequently than twice a day. One
n1ust also consider the activity of drug n1etabolites since controlled deliv-
1. Absorption Rate : For a drug to be administered as controlled ery systen1 controls only the release of parent drug but not its metabolism . .
relea;~ for rnulation, its absorption must be ·efficient since the desired rate- .

limiting step is. rate · of drug release Kr i.e. Kr << Ka. A drug with slow 3. Plasma Concentration-Response Relationship : Drugs such as
absorption is a poor candidate for such dosage fonns since continuous reserpine v.-hose pharmacologic activity is independent of its concentration
release will result lin a pool of unabsorbed drug e.g. iron. Aqueous are poor candidates for controlled release systen1s.
soluble but poorly absorbed potent drugs like decamethonium are also
unsuitable
, · candidates since a slight variation in the absorption may pre-
cipitate potential to~icity.
342 BIOPHARMACEUTICS AND PHARMACOKINETICS
343
CONTROLLED RELEASE MEDICATION
PHARMACOKINETIC PRINCIPLES IN THE DESIGN AND
FABRICATION OF CONTROLLED DRUG DELIVERY SYSTEMS
'
also undergo presystemic metabolism. Hence, ifF is the fraction bioavailable,
The controlled release dosage fortns are so• designed that they release then:
the medicament over a prolonged period of time usually longer than the Css CIT (15.5)
Ro=--
typical· dosing interval for a conventional for.tnulation. The drug release F
rate should be so monitored that a steady plasma concentration is attained
- , by reducing the ratio Css max/Css min while maintaining the drug levels (15.6)
. ' ' and
within the therapeutic window. The rate-controlling step in the drug input
should be dete11nined not by the absorption rate but by the rate of release

Substituting equation 15.6 in equation 15.5 and rearranging, we get:
from the fonnulation which ideally should be slower than the rate of
absorption. In most cases, the release rate is so slow that if the drug Css CIT 1 (15.7)
DM=----
exhibits two-comparttnent kinetics with delayed distribution under nonnal
F
circumstances, it will be slower than the rate of distribution and one can,
· · . .
where 1 = dosing mterval. From above equatton, one· can calculate the
thus, collapse the plasma concentration-time profile in such instances into
dose of drug that must be release d m· a given
· peno · d of time in order to
a one-compartment model i.e. a one-compartment model is suitable and
· · ·
achIeve the desrred target steady-state concentration. It also shows . that
applicable for the design of controlled drug delivery systems. Assuming
· c1earance Is
tota1 systemtc · an 1n1po
· rtan t paramet.er m · such a computation.
t.hat the KADME of a drug are first-order processes, to achieve a steady,
nonfluctuating plasma concentration, the rate of release and hence rate of . . · h
Stnce attamment of steady-state 1eve1s wit a zer o-order controlled
. ·.
input of drug (rom the controlled release dosage fonn should be identical drug release system ~ould require . a tune. · d o f. about 5 btologteal
perto ,
to that from constant rate intravenous infusion. In other words, the rate of half-lives, an immediate release dose, DJ, called as 'loading dose ' may be
drug release from such a system should ideally be zero-order or near zero- .
tncorporated . such a system In
In . add'Itlon
. to th e con tro lied release compo-
. .
order. One can thus treat the desired release rate Ro of controlled drug · · t herapeut'I c concentration 1n
nents. The total dose, DT needed to matntain
delivery system according to constant rate i. v. infusion. In order to the body would then be:
maintain the desired steady-state concentration Css' the rate of drug input, (15.8)
which is zero~order release rate (Ro), must be equal to the rate of output
The 1mmediate release dose is meant to provide the desired steady-
(assumed to be frrst-order elimination process). Thus:
state rapidly and can be calculated by equation:
Ro -= Routput (15.1)
Css Vd (15.9)
The rate of drug output is given as the product of maintenance dose DI =---
F
DM and first-order elimination rate constant KE·
The above equation . .
Ignores the poss1·bl e a dd'tttve
· e ffect
· from
· the im-
(15.2)
mediate and controlled release components. For many controlled release
For a zero-order constant rate infusion, the rate of output is also given products, there is no built-in loading dose.
as: . . t ervaI &:.or a d rug c.ollowt·na one-compartment kinetics
Th e d osing In 11 ~ •
Routput = KE Css V d ( 15.3) with linear disposition is related to elimination half-life and therapeutic
Since CIT = KE Vd, .the above equation can also be written as: index TI according to equation:
(15.10)
Routput = Css C 1T (15.4a) t < tt , In (TI/2)

or Ro = Css CIT (15.4b) DRUG RELEASE PATTERNS OF CONTROLLED


The bioavailability of a drug fro·m controlled release dosage form DELIVERY DOSAGE FORMS
cannot be 100% as the total release may not be 100% and the drug may If one assumes that-
1. drug disposition foliO\\'S first-order kinetics.
344 BIOPHARMACEUTICS AND PHARMACOKINETICS CONTROLLED RELEASE MEDICATION 345
2. the rate-limiting step in the absorption is rate of drug release from constant rate i.v. infusion. The time to reach steady-state depends upon
the controlled release fonnulation (i.e. Kr < Ka), and the elimination half-life of the drug. It is usually not possible with a
3. the released drug is rapidly and completely absorbed, single oral controlled r~=;lease dose to attain the plateau even with a drug·
having short half-life sul:h as 3 hours since the mean G I residence time is
then, the four models for drug input based on the drug release pattern
can be defmed: · around 12 hours. It takes 4.3 half-Jives for attainment of 95o/o steady-state
values. Thus, if t~ is 3 hours, about 13 hours will be required for the
1. Slow zero-or9er release. · drug to reach plateau. Slower the elimination, (longer the t~), more the
..
2. Slow first-order release. I
time required to reach steady-state. Once the desired steady-state is
' '

3. Initial rapid release of loading dose followed by slow zero-order reached with repeated dosing of zero-order .controlled release formulation,
release. minimal fluctuations will be observed. Zero-order release systems are
thus ideal controlled delivery fonnulations.
4. Initial rapid release of loading dose followed by slow first-order
release.
Slow First-Order Release Systems
The resulting profiles are depicted in Fig. 15.3. Such systems are easier to design but are inferior to zero-order sys-
tems especially when they are meant for oral use. This is because with
I. Zero-order II. Slow first-order Ill. Initial rapid IV. Initial rapid first-order release characteristics,\ smaller and smaller amounts of drug are
dose then dose then slow
. G) zero-order first-order
released as time passes and secondly, as the formulation advances along
..· U)
cu the GIT, the absorption efficiency generally decreases due to a number of
.
.-
G)
G)
reasons like decreased intestinal surface area, increased viscosity and ·de-
a: .
';!!. creased mixing. Thus, larger amounts are needed to be released at a later
stage when in fact the opposite happens with first-order systems.
The concentration of drug in plasma following administration of a
~>Time controlled release formulation with slow first-order release is given by
Fig. 15.3 Schematic representation of four major types of drug release equation:
characteristics from controlled release formulations DM-Kr-F- ·(e- K Et - e-Krt )
C = -- (15 .12)
Vd (Kr - KE) .
..
Zero-Order Release Systems '

If the drug released from controlled release formulations is stable in When Kr < KE, flip-flop phenomena is observed which is a common
fluids ~t th~ absorption site, has similar absorption efficiency. fram all feature for such controlled release formulations. With repeated dosing of
absorption ,srtes and is absorbed rapidly and completely after its release slow first-order release formulations, one generally observes a lower Cmax'
then, its rate of appearance in plasma will be governed by its rate o'f higher Cmin and longer tmax in comparison to conventional release formu-
release from the controlled release fonnulatio~. Thus, when the drug lations.
release follows zero-order kinetics, absorption will also be a zero-order
Zero-Order Release Systems with a Rapid Release Component
process and concentration of drug in pJasma at any given time can be
given by equation: With such fonnulations, an initial dose is rapidly released (burst-
F Ko (1 - e-KEt) effect) for immediate first-order availability while the remaining amount
C = - - - - - -- (15.11) is released and absorbed at a slow zero-order rate . l 'he equation for
KE Vd concentration-time course of such a formulation contains two portions,
~here Kp = zero-order release rate constant, aJ.so written as Ro, the zero- one each to denote rapid first-order release and slow zero-order release.
order release rate.
C
Dl Ka F -K t
(e
- Kat
e
Ke .F
) + --- ( J
L' t
e·- " r: )'
· The above equation is similar to the one that expresses the concentra- = E - -- (1 5. 13)
Vd (Ka - KE) KE Vd
tion-time course of a drug that shows one-compartrnent kinetics foiJowing
,

CONTROLLED RELEASE MEDICATION 347


346 BIOPHARMACEUTICS AND PHARMACOKINETICS
C= DI Ka F (e-KEt- e-Kat) + DM Kr F (e- KEt- e-Krt) (15.14)
Such a formulation is ideally suited for drugs with long ty2 in which V d (Ka - KE) V d (Kr - KE)
cases attainment of plateau would otherwise take a long time. The slow
As in the previous case, to obtain the desired plateau, the slow release
release component should ideally begin releasing the drug when the drug
levels from the fast component are at a peak. However, the approach component, OM should start releasing the drug:
suffers from a big disadvantage when the formulation is meant for repeti- 1. When the peak has been attained with rapid release dose, Oy; this
tive dosing the blood level profile shows a peak-trough pattern (which requires OM >> 0 1 which results in wastage of drug since the
normally does not result when all of the drug is released at a slow zero- absorption efficiency reduces as time passes and dosage fonn
order rate); this may cause a momentous rise in peak concentration immediately descends down the GIT, or
after each dose triggering toxic reactions (see Fig. 15.4). It is for this 2. When all of the 0 1 has been released; this requires relatively
reason that such a design is unpopular. small DM and therefore less drug wastage and better sustained
The transient fluctuations in the peak concentration with these fot n1u- levels despite fluctuations in drug levels (Fig. 15.5).
lations can however be overcome by: The problems that result from repeated dosing of this type of fonnula-
1. Decreasing the loading dose in the subsequent dosage forms (which tion are similar to that described for the third type of release pattern and
appears to be impractical), can be handled in a similar manner.
2. Increasing the dosing interval (this also seems to be tedious), or
3. Administering an immediate release conventional dosage form
prior to repetitive dosing of zero-order controlled release formula- c
0
tion instead of incorporating it in the latter. ·-......
ns
......
"-
c .,__fast release component
~ completely released
4 ·Undesirable transient peaks c
0
'' and release from slow
(.)
Toxic range component initiated
c
0--------
......
- - --tMSC
co
......
"-
Therapeutic range
c
~ Time
c
0
(.)
------ ---- - - - - - !MEC -4)

second dose third dose Fig. 15.5 'Plasma concentration-time profile which results after a single oral
dose of controlled delivery system containing a rapid release dose
first-dose and a slow first-order release component
_;___., Time

Fig. 15.4 Plasma concentration'7time profile which results from repeated


ORAL CONTROLLED RELEASE SYSTEMS
I
dosing of a controlled release formulation with zero-order Oral route has been the most popular and successfully used for con-
release and an initial fast release component. trolled delivery of drugs because of convenience and ease of administration,

great~r flexibility in dosage fonn design (possible because of versatility of
Slow First-Order Release Systems with a Rapid Release Component G I anatomy and physiology) and ease of production and low cost of such
The equation describing the time course of plasma drug concentration a system.
with this type of formulation will also contain two portions one to The controlled release systems for oral use are mostly solids and based
describe rapid first-order absorption and the other for slow first-order on dissolution, diffusion or a combination of both .mechanisms in the
absorption from controlled release portion.
348 BIOPHARMACEUTICS AND PHARMACOKP\JETICS

CONTROLLED RELEASE MEDICATION 349


control of release rate of drug. Depending upon the manner ~ Jf drug
release, these systems are classified as follows: i. with inherently slow dissolution rate e.g. griseoful~in and
A. Continuous Release Systems : These systems release the drug for digoxin; such drugs act as natural prolonged release products
a prolonged period of time along t~e .entire length of G IT (especially upto ii. that produce slow dissolving fonns wh~n it com.es in contact
the term ina) region of small intestine) with normal transit of the dosag~ with GI fluids e.g. ferrous sulfate, or
'
form. T'he various systems under this category are: iii. having high aqueous solubility and dissolution rate e.g.
1. Dissolution controlled release systems pentoxifylline.
2. Diffusion controlled release systems Drugs belonging to the last category present challenge in controlling
.
3. Dissolution and diffusion controlled release systems their dissolution rate. The techniques employed are:
4. Ion-exchange resin-drug complexes
...
1. Embedment in slowly dissolving or erodible ma~rix, and
5. Slow dissolving salts and complexes 2. Encapsulation or coating with slowly dissolving or erodible sub-
6. pH-dependent formulations stances (Fig. 15 .6. ).
7. Osmotic pressure controlled systems
8. Hydrodynamic pressure controlled systems
B. Delayed Transit and Continuous Release Systems : These sys-
tems are designed to prolong their residence in the GIT along with their slowly dissolving _
or erodible coat
release. Often. the dosage fonn is fabricated to detain in the stomach and
hence the drug present therein should be stable to gastric pH. Systems ~slowly dissoiving
included in this category are: or erodible matrix
(b)
(a)
1. Altered density systems
2. Mucoadhesive systems Fig. 15.6 Schematic representation of dissolution controlled release systems
-(a) matrix system, and (b) coated/encapsulated system
3. Size-based systems
C. Delayed Release Systems : The design of such systems tnvolve Matrix (or Monolith) Dissolu~ion Controlle~ Systems
release of drug only at a specific site in the G IT. The drugs contained in Matrix systems are also called as monoliths 'since the drug is homoge-
such a system are those that are: · neously dispersed throughout a rate-controlling medium. They are very
i.• destroyed in the stomach or by intestinal enzymes common and employ waxes such as beeswax, carnauba wax, hydroge-
ii . known to cause gastric distress nated castor oil, etc. which control drug dissolution by controlling the rate
of dissolution fluid penetration into the matrix by altering the porosity of
iii. absorbed from a speci fie intestinal site, or
tablet decreasing its wettability or by itself getting dissolved at a slower
iv. meant to exert local effect at a specific G I site. rate. ' The wax embedded drug is generally prepared by dispersing the
The two types of delayed release systems are: drug in molten wax and congealing and granul~ting the same. The drug
1. Intestinal release systerns release is often first-order from such matrices.
2. Colonic release systen1s Encapsulation/Coating Dissolution Controlled Systems (Reservoir
Dissolution Controlled Release Systems Devices)
.
Such systen1s are e~siest to design. The drug present in such a systen1 Here the drug particles are coated or encapsulated by one of the
n1ay be the one: several ~icroencapsulation techniques with slowly dissolving materials
like cellulose, PEGs, polymethacrylates, waxes, etc. The resulting pellets
may be filled as such in hard gelatin capsules (popularly called as spansules)
or compressed into tablets. The dissolution rate of coat depends upon the

I
350 BIOPHARMACEUTICS AND PHARMACOKINETICS
diffuses out of it, the swollen mass, devoid of drug appears transparent or
solubility and thickness of the coating which may range from 1 to 200 glasslike and therefore the system is sometimes called as glassy hydrogel

microns. (Fig. 15.7 b). The drug release follows Fickian first-order diffusion under
equilibrium conditions. However, during the swelling process, such an
Diffusion Controlled Release Systems equilibrium may not exist and the diffusion may be non-Fickian or anomalous
In these types of systems, the rate-controlling step is not the dissolu- diffusion.
tion rate but the diffusion of dissolved drug through a polymeric barrier.
The drug release rate is never zero-order since the diffusional path length Reservoir Devices (or Laminated Matrix Devices)
increases with time as the insoluble matrix is gradually depleted of drug. These systems are hollow containing an inner core of drug surrounded
The two types of diffusion controlled systems are matrix systems and in a water insoluble polymer membrane. The polymer can be applied by
reservoir devices. coating or microencapsulation techniques. The drug release mechanism
across the membrane involves its partitioning into the membrane with
Matrix Diffusion Controlled Systems subsequent release into the surrounding fluid by diffusion (Fig. 15.8).
Here, the drug is dispersed in an insoluble matrix of rigid nonswellable The polymers commonly used in such devices are HPC, ethyl cellulose
hydrophobic materials or swellable hydrophilic substances. Materials used and polyvinyl acetate. A disadvantage of all such microencapsulated drug
for rigid matrix are insoluble plastics such as PVC and fatty materials release systems is a chance of sudden drug dumping which is not common
like stearic acid, beeswax, etc. With plastic materials, the drug is gener- with matrix devices.
ally kneaded with the solution of PVC in an organic solvent and granulated.
Waxy matrix is prepared by dispersing the drug in molten fat followed by Rate Controlling Factors :
Polymeric content in coating,
congealing. The granules are then compressed into tablets (Fig. 15.7.a).
Thickness of coating,
Swellable matrix systems are popular for sustaining the release of highly
Hardness of microcapsule
water-soluble drugs. The material for such matrices are generally hydro-
philic gwns and may be of natural origin (guar gum, tragacanth), semisynthetic ~---- Insoluble membrane
(HPMC, CMC, xanthan gum) or synthetic (polyacrylamides). The drug ~----Reservoir of drug
and the gum are granulated together with a solvent such as alcohol and ·.-. (alone or as matrix
compressed into tablets. The release of drug from such initially dehy- with inert diluent)
drated hydrogels involves simultaneous absorption of water (resulting in Diffusion of soluble
hydration, gelling and swelling of gum) and desorption of drug via a drug by partitioning
swelling controlled diffusion mechanism. As the gum swells and the drug

Rate Controlling Step : Fig. 15.8 Drug release by diffusion across the insoluble
Diffusion of dissolved drug membrane of reservoir device
through the matrix
Dissolution and Diffusion Controlled Release Systems
In such systems, the drug core is encased in a partially soluble mem-
~- insoluble rigid matrix brane. Pores are thus created due to dissolution of parts of the membrane
which:
- permit entry of aqueous medium into the core and hence drug
swellable gum matrix--.~~-.... dissolution, and
-allow diffusion of dissolved drug out of the system (Fig. 15.9).
swollen glassy -~~
hydrogel from An example of obtaining such a coating is using a mixture of ethyl
which drug has cellulose with PVP or methyl cellulose; the latter dissolves in- water and
(a) completely diffused creates peres in the insoluble ethyl cellulose membrane.·
(b)

Fig. 15.7 Diffusion controlled devices-{ a) rigid matrix, and (b) swellable matnx
352 BIOPHARMACEUTICS AND PHARMACOKINETICS CONTROLLED RELEASE MEDICATION 353
pH-Independent Formulations
Rate Controlling Factor :
Such systems are designed to eliminate the influence of changing GI
Fraction of soluble polymer
in the coat
pH .on dissolution and absorption of drugs by fonnulating them with
sufficient amount of buffering agents (salts of phosphoric, citric or tartaric
acids) that adjust the pH to the desired value as the dosage fot·tn passes
- - - - Insoluble membrane
along the GIT and pennit drug dissolution and release at a constant rate
- - - P oI r e created by dissolution
• • •
••• \. --4~
of soluble fraction of membrane
independent of GI pH. The dosage fortn containing drug and buffer is
coated with a penneable substance that allows entry of aqueous medium
e e e l Entry of dissolution but prevents dispersion of tablet.
• • fluid
Osmotic Pressure Controlled Systems
• • • • • Diffusion of dissolved
e • • drug Unlike the solution-diffusion mechanism for most systems, an oral
osmotic pump, popularly called as oros, works on the principle of os-
motic pressure to release the drug at a ·constant zero-order rate. A core
Fig. 15.9 Di~solution and diffusion controlled release system comprising of drug and an osmotically active substance (also called as ·
osmogen) such as potassium chloride or mannitol is surrounded by a rigid
Ion-Exchange Res~il-Drug Complexes semipettneable membrane coating such as cellulose est~r or cellulose ether
Controlled delivery of ionizable acidic and basic drugs can be obtained having an orifice of 0.4 mm diameter produced by laser beam for drug ·
by complexing them with insoluble /nontoxic anion exchange and cation exit. When exposed to GI fluids, water flows through the semipenneable
exchange resins respectively. The drug is re!eased slowly by diffusion membrane into the tablet due to osmotic pressure difference which dis-
thr0ugh the resin particle structure. The following equation represents the solves the drug and pumps it out through the orifice by the osmotic force
release of a basic drug, NH2R', from a cation exchange resin RS03H (Fig. 15.10). Such devices can be used to target specific areas of the GIT.
fohen in contact with G I' fluid containing ~n iqnic compound A+~- ( ei~her
gastric HCI or intestinal NaCl): Rate Controlling Factors:
I

Orifice diameter
RSO 3-NH3+R' + ' A+s·- ) RS03-A+ + NH3+R'B- Membrane area
A 11umber of basic drugs like noscapine, phenylpropanolamine and Membrane thickness
Membrane permeability
phentermine have been retarded by such an approach. The c-omplex can
Osmotic properties of the core
be prepared by incubating the drug-resin solution or passing the drug
Drug solubility
solution through a column containing ion-exchange resin. The drug-resin
complex can be coated with cellulose or hard paraffin and fonnulated as delivery orifice
ion free suspension for pediatric use.
-~~~~--rigid semipermeable
Slow Dissolving Salts and Complexes • • memb~ne

Salts or complexes of drugs which are slowly soluble in the GI fluids


••••••••
••••••• ~t----
• • • • • • • water
can be used for controlled release of the active principle. Amine drugs
can be reacted with tannic acid to form poorly soluble complexes. that can
•••••••
••• •• • • ~-osmotic core
be formulated as long acting tablets. Penicillin G has been complexed containing drug
with N ,N' -dibenzy.l ethylenediamine to give benzathine p~nicillin G that
can be fonnulated as oral suspension. Such complexes can be obtained Fig. 15.10 Oral osmotic pump (oros)
by simple acid-base reaction on mixing together solutions of individual
The oros principle can be used to design multiunit dosage forms
compounds. consisting of drug core particles coated with a water pel'ineable membrane
354 BIOPHARMACEUTICS AND PHARMACOKINETICS CONTROLLED RELEASE MEDICATION 355
in which the delivery orifice is made by using a channeling agent such as High Density Pellets
PVP and the coated particles filled in a capsule. The density of GI fluids is around 1.4 glee. Use of drug pellets
having density greater than this value, preferably above 1.6 glee, results in
Hydrodynamic Pressure Controlled Systems
. prolonged Gl residence that is unaffected by food. Iron oxide, titanium
The hydrodynamic pressure generated by swelling of a hydrophilic dioxide and barium sulfate have been used to increase the density of drug
gum can also be used to activate the delivery of drugs. The device pellets. The drug is coated on the heavy core and then covered -by a
comprises of a rigid, shape retaining housing enclosing a collapsible, diffusion controlled membrane (Fig. 15.12a).
impenneable compartment containing liquid drug (Fig. 15.11). The space -
between the external housing and the drug compartment contains a layer Low Density Pellets
of swellable, hydrophilic gum such as polyhydroxyalkyl methacrylate. In Also called as hydrodynamically balanced systems, such pellets,
the GIT, the gum imbibes water through the opening present at the lower having density less than that of GI fluids, float on the gastric juice for an
side of external housing and swells creating a hydrodynamic pressure. extended period of time while slowly releasing the drug. Globular shells
The pressure thus created squeezes the collapsible drug reservoir to re- such as that of poprice and celluloses have been used to lower the density
lease the medicament through the delivery orifice at a zero-order rate. of system (Fig. 15 .12b). A swellable gum like HPMC can be used for a
Such systems are also called as push-pull osmotic pumps. similar purpose (Fig. 15.12c).

Rate Controlling Factors : rate controlling membrane

Fluid permeability ~-----drug


Surface area of wall with openings ~......,., ert heavy core
Hydrodynamic pressure gradient
mixture of drug ___,.._
and heavy material
delivery orifice
(a) High Density Pellets
~-rigid, shape retaining housing
_ _ liquid drug formulation
~-collapsible drug reservoir rate controlling membrane

f-_swellable hydrophilic gum

L----openings for entry of Gl fluid ~--- drug - - - +


Ill ---~,.,__empty shell .

Fig. 15.11 Hydrodynamic pressure controlled system (push-pull osmotic pump) low density core

Altered Density Systems


(b) Low Density Pellets with Hollow Core
The transit time of G I contents is usually less than 24 hours. This is
the major limiting factor in the design of oral controlled release fonnula-
tions which can reduce the frequency of dosing to a time period little
~~~--- mixture of drug +
more than the residence time of drug. However, if the residence time of swellable polymer .__...swollen gum,
drug in the stomach and/or intestine is prolonged in some way, the . density
frequency of dosing can be further reduced. There are 3 ways by which less than 1
Gl fluids,
this can· be achieved altering the density of drug particles, use of
density more than 1
mucoadhesive polymers and altering the size of the dosage fortn . The
altered density approach involves use of either high or low density pellets. (c) low Density Pellets with Hydrogels

Fig. 15.12 Altered density systems


356 BIOPHARMACEUTICS AND PHARMACOKINETICS
CONTROLLED RELEASE MEDICATION 357
Floating or buoyant tablets/capsules can be founulated by granulat-
i. Local action as in the treatment of ulcerative colitis with
ing a drug with 20 to 80% of hydrogel such as HPMC, HEC and HPC.
mesalamine, and
On contact with GI fluids, the tablet swells and fonns a diffusible gel
barrier that lowers the density of system to less than 1 allowing it to float. ii. Systemic absorption of protein and peptide drugs like insulin
Lipophilic polymers such as silicone elastomer can also be modified to and vasopressin.
hav~ swelling properties. This is achieved by impregnating a water Advantage is t~ken of the fact that pH-sensitive bioerodible polymers
-inis6ible
\
liquid
..
such as glycerol or a water-soluble salt such as sodium like polymethacrylates release the medicament only at the alkaline pH of
chloqde .
in · the . lipophilic matrix. On contact with aqueous medium; the colon or use of divinylbenzene cross-linked polymers that can be cleaved
modified lipophilic polymer swells due to absorption of water by the only by the azoreductase of colonic bacteria to release free drug for local
hydrophilic additives in the matrix. Alternatively, a gas filled flotation effect or systemic absorption .
..-

J
chamber can be arxached to a membrane coated tablet for making it •

buoyant.
' PARENTERAL CONTROLLED RELEASE SYSTEMS
Mucoadhesive Systems One of the major advantages of parenteral controlled drug delivery
A bioadhesive polymer such as cross-linked polyacrylic acid, when systems is that the duration of action can be extended for days or months
incorporated in a tablet, allows it to adhere to the gastric mucosa or and sometimes upto a year. The prime drawback is that, once adminis-
· . epithelium. Such a system continuously releases a fraction of drug into tered, the drug cannot be easily removed if an undesirable action is
the intestine over prolonged periods of time. precipitated or if the drug is no longer needed. Most of such systems are
administered by subcutane~us and intramuscular routes and few by intra-
Size-Based _S ystems venous and intraperitoneal routes. Subcutaneous route is limited to well
Gastric :emptying of a dosage fonn can be delayed in the fed state if absorbed water-soluble drugs like insulin and dose volume is limited to ·
its size is greater than 2 mm. Dosage fonn of size 2.5 em or larger is 0.5 to 1.5 mi. Deep intramuscular route i·s suitable for polymeric systems ·
often required to delay emptying long enough to allow once daily dosing. or slightly soluble drugs, the volume size restricted to 2 mi. Intravenous
Such fonns are however difficult to swallow. route is useful for administration of liposomes, nanoparticles, erythrocytes
and polypeptides. An important criteria for this route is drug particle size.
Intestinal Release Systems A disadvantage of i.v. routb is that the ~ystem _may be taken. up by the
A drug may be enteric coated for intestinal release for several known reticuloendothelial system but the same can be put to use in targeting
reasons such as to prevent gastric irritation, prevent destabilization in drugs to ~;uch a system. Intraperitoneal route is important in targeting of
gastric pH, etc. ~e~ain.. .
drugs are delivered to the distal end of small antineoplastics into the lymphatic system.
intestine for absorption via Peyer' s patches or lymphatic system. Peyer's The vehicle, polymers and other substances used in the formulation of
patches are mucosal lymphoid tissues that are known to absorb macro- parenteral controlled release dosage forms should be sterile, pyrogen free,
molecules like proteins and peptides and antigens by endocytosis. Selective nonirritating. biocompatible and biodegradable into nontoxic compounds
release of such agents to Peyer' s patch region prevents them from getting within an appropriate tin1e. preferably close to the duration of drug action.
destroyed/digested by the intestinal enzymes. Such a site can be utilized
for o_ral delivery of insulin. Lymph a tic system on the other hand is There are several approaches to achieve controlled drug delivery via
parenteral route. the release being controlled by dissolution. diffusion.
known to absorb highly lipophilic agents directly into the systemic circu-
lation without their first-pass through liver. The drug is absorbed by two dissociation. partitioning or bioerosion. The systen1s can be broadly

meclranisms chylomicrons which are fatty vesicles that entrap hydropho- classified as:
i bic drugs, and· pinocytic uptake of macromolecules. . A. lnjectables:
1. Solutions
'Colonic Release Syste·m s
2. Dispersions
· ' Drugs are poorly absorbed through colon but may be delivered to such
a site for two reasons,- 3. M icrospheres and Microcapsules
4. Nanoparticles and N.ioson1es
. ..
... ,,

..
358 BIOPHARMACEUTICS AND PHARMACOKINETICS CONTROLLED RELEASE MEDICATION 359 .

5. Liposomes steps. Release of water-soluble drugs can be. retarded by presenting it. as
6. Resealed Erythrocytes oil suspension,. anp vice versa for oil soluble drugs. Factors to be consid-
ered in the fonnulation of such a system include -
B. Implants i. Solid content : should be ideally in the range 0.5 to 5.0%
C. Infusion Devices: ii. Particle size : this factor is very important since larger the
-J. Osmotic Pumps particle size, slowe~ the dissolution; however, larger particles
2·. Vapor Pressure Powered Pumps have their own disadvantages like causing irritation at the injec-
tion site (size should therefore be below 10 microns), poor .
3. Battery Powered Pumps
syringeability and injectability and rapid sedimentation. The
Solutions latter problem can be overcome by use of viscosity builder$_
Both aqueous as well as oil solutions may be used for controlled drug which also retard drug diffusion.
release. With aqueous solutions (given intramuscularly), the drug release
may be controlled in three ways:
' . .

i. By increasing the viscosity of vehicle by use of MC, CMC or ..
PVP and thus, decreasing molecular diffusion and localizing the ~~Aqueous
injected drug. oil-soluble drug
E Phase
(e.g. lipidol) .Oil Phase
ii. By forming a complex with macromolecules like MC, CMC or
PVP from which the drug dissociates at a controlled rate (only
w/o/w Type Emulsion o/w/o Type Emulsion
free drug will get absorbed).
iii. By fonning complexes that control drug release not by dissocia- Fig. 15.13 Multiple emulsions for parenteral controlled release systems
tion but by reducing the solubility of parent drug e.g. protamine
zinc insulin and cyanocobalamin zinc tannate. Aqueous suspensions can be given by i.m. or s.c. routes. Generally
crystalline and stable polymorphic fonns of the drug are chosen rather
Oil solutions control the release by partitioning the drug out of;the oil than amorphous fonns to delay release. Solubility can be further reduced
· in the surrounding aqueous biofluids. Vegetable oils like arachis oil, by salt or complex fonnation e.g. crystalline zinc insulin shows ~ore
cottonseed oil, etc. are used for such a purpose. The method is applicable prolonged action than amorphous zinc insulin compte~ . Oil su~pens1on~ .
only to those drugs which ·are oil soluble and have optimum partition generally given i.m. ~ prolong drug action much more 1~ comparison .to otl __
coefficient. solution and aqueous suspension since drug release tnvolyes-two rate-
limiting steps viz. dissolution of drug particles, and partitioning of the
Dispersions
dissolved drug from oil to the aqueous biofluids.
Dispersed systems like emulsions and suspensions can be administered
by i.m., s.c. or i.v. routes. Among emulsions, the o/w systems have not Microspheres and Microcapsules
been used successfully since absorption of drug incorporated in the oil Microspheres are free flowing powders consisting of spherical ?ar-
phase is rapid due to large interfacial area and rapid partitioning. Simi- ticles of size ideally less than 125 microns that can be suspen,ded In a
larly, few w/o emulsions of water-soluble drugs have been tried for controlled suitable aqueous vehic,le and injected by an 18 or 20 number needle.
release. Multiple emulsions of w/o/w and o/w/o types (more correctly, ·Each particle is basically a l)latrix of drug dispersed in a polymer from
double emulsions) are becoming popular since an additional reservoir is which release occurs bv a first-order process. The polymers used are
presented to the drug for partitioning which can effectively retard its biocompatible and biodegradable e.g. polylactic acid. polylact~d,t coglycoli~e.
release rate (Fig. 15.13). etc. Drug release is controlled by dissolution/degradation of matrix.
Control of drug ' release from suspensions is easier and predictable. Sn1 all mat;ices release drug at a faster rate and thus. by using particles of
.
Drug dissolution and subsequent diffusion are the main rate controlling different sizes . various degrees of controlled release can be achieved. The

..
'
CONTROLLED RELEASE MEDICATION 361
360 BIOPHARMACEUTICS AND PHARMACOKINETICS

system is ideally suited for controlled release of peptide/protein drugs i. MLV (multilamellar vesicles) : These liposomes are made of
such as LHRH which have short half-lives and otherwise need to be series of concentric bilayers of lipids enclosing a small internal
injected once or more, daily, as conventional parenteral forrnulations. In volume.
comparison to peptides, proteins are difficult to formulate because of their ii. OLV (oligolamellar vesicles) : These are made of 2 to 10
higher molecular weight, lower solubility and the need to preserve their bilayers of lipids surrounding a large internal volume.
confottnational structure during manufacture. iii. UL V ( unilamellar vesicles) : These are made of single bilayer
In order to overcome uptake of intravenously administered microspheres of lipids. They may be SUV (small unilamellar vesicles) of
by the reticuloendothelial system and promote drug targeting to tumors size 20 to 40 run, MUV (medium unilamellar vesicles) of size
·w ith _good perfusion, magnetic microspheres were developed. They are 40 to 80 nm, LUV (large unilamellar vesicles) of size 100 to
prepared from albumin and magnetite (Fe20 3) and have a size of I.O 1000 nm or GUY (giant unilamellar vesicles) of size greater
micron to· pennit intravascular injection. The system is infused into an than I 000 nm. I

artery that perfuses the target site and a magnet is placed over the area to A large variety of drugs ( antineoplastics, antibiotics), peptides/proteins
localize it in that region. A I 00 times higher concentration of doxorubi- (including antibodies) and viruses and bacteria can be incorporated into
cin was attained at the target site by such an approach with just ·half the liposomes. Water-soluble drugs are trapped in the aqueous compartrnent
i.v. dose. while lipophilic ones are incorporated in the lipid phase of liposomes.
Microcapsules differ from microspheres in that the drug is centrally Because of their availability in various sizes, ability to incorporate both
located within the polymeric shell of finite thickness and release may be water as well as oil soluble drugs, their inertness and th~ir ability to
controlled by dissol~tion, diffusion or both. Quality microcapsules with protect labile drugs, liposomes are versatile carriers for parenteral drug
thick walls generally release their medicaments at a zero-order rate. Ste- delivery systems. Intramuscularly and subcutaneously injected liposomes
roids, peptides and antineoplastics have been successfully administered deliver drug at a controlled rate while intravenous administration selec-
parenterally by use of controlled release microcapsules. tively targets them to reticuloendothelial system and phagocytic cells. A
simple method by which liposomes can be produced involves •
drying an
Nanoparticles and Niosomes organic solvent solution of lipids onto the wall of a flask/beaker followed
Nanoparticles are also called as nanospheres or nanocapsules de- by hydration and dispersion of lipid by addition of buffer and mixing
pending upon ~·hether the drug is in a polymer matrix or encapsulated in (Fig. 15.14 ).
a shell. They differ from microspheres in having submicron particles in
the nanometer size range 10 to 1000 nm. The polymers used are the
usual biodegradable ones. The main advantage of this system is that it evaporation addition of buffer + drug
-------------.·~~----~
can be stored for upto I year and can be used for selective targeting via
reticuloendothelial system to liver and to cells that are active phagocytically.

• •









and agitation/sonication CD a~<!>
0
°
• • • • • ,tD(!)($)Q f!) 0 ...
• • •
Like nanoparticles, niosomes are inexpensive alternatives to liposomes. •

Solution of Lipid Dried Film of Lipid Liposomes


They are closed vesicles fonned in aqueous media from nonionic surfac-
in Organic Solvent on Vessel Wall Formed
tants with or without the presence of cholesterol or other lipids.
Fig. 15.14 Production procedure for liposomes.
Liposomes
The tenn liposomes (meaning lipid body) was derived on the basis of Resealed Erythrocytes
nanies of subcellular particles like lysosome and ribosome. It is defined Drug loading in body's own erythrocytes when used to serve as con-
as a spherule/vesicle of lipid bilayers enclosing an aqueous co~partment. trolled delivery systems have several advantages. They are full y
The lipid most commonly used is phospholipid. Sphingolipids, glycolip- biodegradable and biocompatible, nonimmunogenic, can circulate intrava s..
ids and sterols have also been used io prepare liposomes. .Their size cularly for days (act as circulatory drug depots) and allow large anto unt s
ranges from 25 to 5000 nm. Depending upon their structure, liposomes of drug to be carried. The drug need not be chemically n1od if u·d ""cl ,..
are classified as:
362 BIOPHARMACEUTICS AND PHARMACOKINeTICS CONTROLLED RELEASE MEDICATION 363

prot~cted from immunological detection and enzymatic inactivatiO!! Drug A major disadvantage of such systems is that a microsurgery is re-
loading can be done by immersing the cells in buffered hypotonic solution quired for implantation of device. Some devices can be easily implanted
of d~g which causes them to rupture and release hemoglobin and trap the by use of a specially designed implanter syringe. The devices are gener-
medicament. On restoration of isotonicity and incubation at 37° C, the ally implanted subcutaneously or intramuscularly. Subcutaneous tissue is
cells reseal and are ready for use (Fig. 15.15). an ideal location because of its easy access to implantation, poor perfu-
sion·, slow drug absorption and low reactivity towards foreign materials.
i//@~ .. -. ..•. ._-
-· . _...,
· ·.--· ••
~ -- •
----. __ _.,-~·.
.
• • •-~-
• - • --
·-- --• ·-
The drug may be dissolved, dispersed or embedded in a matrix of
.. . . ....
- • -
.
·-'-•
polymers that control release by dissolution, diffusion or both, bioerosion,
. ..;-
• • • ..!....·~ ·

. . . .. . . . .
••
~
_. -== ~·. ·.:.)_-
" . .
~-
- ___,..___
...,
biodegradation or an activation process such as osmosis or hydrolysis.
The system is generally prepared as implantable flexible/rigid moulded or
Red Cells placed Lysis of Cells and Resealed Red Cells extruded rods, spherical pellets or compressed tablets. Polymers used are
in Hypotonic Entry of Drug Loaded with Drug
Drug Solution silicone elastomers, polymethacrylates, polycaprolactone, polylactide/glycolide,
etc. Drugs generally presented in such systems are steroids like contra-
Fig. 15.15 Drug loading in erythrocytes. ceptives (megestrol acetate, norgestrone, etc.), morphine antagonists like
Damaged erythrocytes are removed by the liver and spleen. These naltrexone for opiod-dependent addicts, etc.
organs can thus be specifically targeted by drug loaded erythrocytes. Infusion Devices
Implants These are also implantable devices but are versatile in the sense that
. An ideal implantable parenteral system should possess following prop- they are intrinsicalJy powered to release the medicament at a zero-order
erties.- rate and the drug reservoir can be replenished from time to time. De-
pending upon the mechanism by which these implantable pumps are
I. Environmental(v stable : should not breakdown under the influ-
powered to release the contents, they are classified into following types:
ence of heat, light air and moisture.
I. Osmotic pressure activated drug delivery systems -..
2. Biostab/e : should not undergo physicochemical degradation when ..
in contact with bioflu ids (or drugs). 2. Vapor pressure activated drug delivery systems
3. Biocon1patible : should neither stimulate immune responses (oth- 3. Battery powered drug delivery systems
erwise the in1plant will be rejected) nor thrombosis and fibrosis Osmotic Pumps (Alzet)
formation .
These pumps are capsular in shape and made in a variety of sizes.
4. f\./ontoxic and noncarcinogenic : its degradation products or leached The device is shown in Fig. 15.16.
additives must be completely safe.
The pump is made of three concentric layers the innermost drug
5. Should have a n1inirnum surface area. smooth texture and struc-
reservoir contained in a collapsible impermeable polyester bag (which is
tural characteristics sin1 ilar to the tissue in which it is to be
open to the exterior via a single portal) followed by a sleeve of dry
in1planted to avoid irritation.
osmotic energy source (sodium chloride) and the outermost rigid, rate-
6. Should be ren1ovahle when required. controlling semipenneable membrane fabricated from substituted cellulosic
7. Should release the rnedican1ent at a constant predetennined rate for polymers. A rigid polymeric plug is used to form a leakproof seal
a prcdctennined period of tirne. between the drug reservoir and the semipermeable housing. An additional
component, the flow modulator, comprising of a cap and a tube made of
Sornc of the in1portailt ad\'antages of in1plants over injectable con-
trolled release fonnulations are-- stainless steel is inserted into the body of osmotic pump after filling.
After implantation, water from the surrounding tissue fluids is imbibed
1. More effective and rnore prolonged action (for over a year). through the semipermeable membrane at a controlled rate that dissolves
:2. A significantly srnall dose is sufficient. the osmogen creating an osmotic pressure differential across the rnem -


CONTROLLED RELEASE MEDICATION 365
364 BIOPHARMACEUTICS AND PHARMACOKINETICS
vaporizes at the body temperature and creates a vapor pressure that com-
brane. The osmotic sleeve thus expands and since the outer wall is rigid,
presses the bellows and expels the infusate through a series of flow
it squeezes the inner flexible drug reservoir and drug solution is expelled
regulators at a constant rate. Insulin for diabetics and- morphine for
in a constant volume per unit time fashion. The drug delivery continues
tenninally ill cancer patients have been successfully delivered by such a
until the reservoir is completely collapsed. Ionized drugs, macromol-
device.
ecules, steroids and peptides (insulin) can be delivered by such a device. Inlet port

Rate Controlling Factors : -+ Orug outlet


Porosity of semipermeable membrane ,------
Osmotic pressure difference
across -the membrane Housing
--+--t--urug infusate
chamber
~ Drug solution leaving
via delivery portal
~~~~~~~~~~~~~~~~~~~~---~exiblefre~y
movable bellows
.;,.......-~--Flow modulator
~~~~~ . .~... w.M.IIill ~~MAll~. . . . . . . . ....._.._ Vaporizable fluid
Removable cap ....~ chamber

Q)•
c:::
Fig. 15.17 Cross section of vapor pressure driven device
C'O
:+-+--- Flexible, imperrneabre
"-
.0 reservoir wall
E-..
Q)
Battery Powered Pumps
E Two types of battery powered implantable programmable pumps used
0)

·--c Saturated solution successfully to deliver insulin are peristaltic pump and solenoid driven
·~-- of osmotic agent
- 0
"-
c.:
0
u
(osmogen sleeve)
reciprocating pump, both with electronic controls. The systems can be
programmed to deliver drug at desired rates. Their design is s.u ch that the
-<1,)

C'O
"-
Rate controlling rigid
drug moves towards the exit and there is no backflow of the infusate.
0>
,.... semipermeable membrane
-
"-
TRANSDERMAL DRUG DELIVERY SYSTEMS
-c:::
(1) •

Q)
Water entering
._ • rate controlling Transdermal delivery systems are topically administered medicaments
-~m.
<U
membrane in the fonn of patches that deliver drugs for systemic effects at a predeter-
.:·.~~~-- Drug reservoir mined and controlled rate. Some of the advantages of these systems over
other controlled release forntulations are:
1. Drugs with very short half-lives e.g. nitroglycerine when adminis-
Fig. 15.16 Cross section of osmotic pump tered as transdennal patches, release medicaments at a constant
rate for _a time .period more than that obtainable with oral fonnu-
Vapor Pressure Powered Pump (lnfusaid) lations. •
This device is based on the principle that at a given temperature, a 2. Drugs ·w ith narrow therapeutic indices can be safely administered
liquid in equilibrium with its vapor phase exerts a constant pressure that is since better control of release is possible.
independent of enclosing volume . The device is shown in Fig._15.17. 3. The noninvasive nature of these systems penn its easy removal
• I

The d!sc shaped device consists of two chambers an infusate chan1- and termination of drug action in situations of toxicity.
ber containing the drug solution which is separated by a freely movable 4. Problems encountered with oral administration like degradation,
flexible bellow from the vapor chamber containing inexhaustible vaporiz- gastric irritation, frrst-pass effect, etc. are avoided.
able fluid such as fluorocarbons. After implantation. the volatile liquid

CONTROLLED RELEASE MEDICATION 367


366 BIOPHARMACEUTICS AND PHARMACOKINETICS

The route is unsuitable when -- in the polymer matrix and the other in which the drug is dispersed
(generally much above saturation levels). The polymers employed for
1. the drug dose is large.
matrix systems may be hydrophilic or lipophilic and includes PVC, PVP,
2. the drug has large molecular size (makes absorption difficult; polysaccharides, polyesters, microporous polypropylene and ethylene vinyl
should ideally be below 800-1 000). acetate copolymers. The drug release rate from matrix systems is rapid
3. the drug i~ skin sensitizing and irritating.
4. the drug is metabolized in skin. Rate Controlling Factors ~

5. the drug undergoes protein binding in skin. Drug concentration in


'
polymer matrix
6. the drug is highly lipophilic or hydrophilic (should be moderately Chemical nature of
soluble in both oil and water). polymer matrix
Geometry of device
Other disadvantages of such systems include variation in absorption
efficiency at different sites of skin, difficulty of adhesion to certain skin
types and length of time for which a patch can be left on any area due to ---II-- Impermeable backing
penneability changes (usually not more than 7 to 10 days). ........ Adhesive layer
Several types of transdermal drug delivery devices are available but
~----Drug-polymer matrix
they can be basically divided into two broad categories based on the
mechanism by which drug release is controlled:
Fig. 15.18 Schematic representation of a monolith (matrix)
1. Monolithic (or matrix) systems. transdermal drug delivery system
2. Reservoir (or membrane) systems.
'
initially and falls as the matrix gets depleted of drug. The rate is thus
All such devices are fabricated as multilayer laminate structures in
proportional to the square root of time. Fig. 15 .18. shows a typical matrix
which the drug-polymer matrix or a drug reservoir is sandwiched between
tvvo polymeric layers. 'The outer layer, called as backing layer, is imper- system.
meable and meant to prevent drug loss through it. It is generally composed
Reservoir (or Membrane) Devices
of metallized plastic. The other layer which contacts the device with the
skin is adhesive layer. It is permeable and in some cases, may act as rate- These devices are used when drug permeation rate is rapid and absorp-
limiting membrane. It is generally made of pressure sensitive adhesive tion should therefore be controlled by controlling drug release (Rl < R2).
materials like acrylic copolymers, poiyisobutylene and polysiloxane or It is also suitable for potent drugs with low therapeutic indices where
contact adhesives. monitoring drug levels in a narrow range is essential. The drug is usually

The choice of mo~olithic or reservoir type of system for controlling


Rate Controlling Factors :
drug release depends upon the major rate-limiting step in the absorption
Membrane thickness
of drug from such de vt ~es . The two rate-limiting steps are· Membrane permeability
..
1. Rate of drug diffusion from the device, R 1, and ·
~------------- _ _ i,mpermeable backing
2. Rate of drug pestneation through the stratum corneum, R2.
~

The overall rate of drug transport is proportional to the sum (R 1 + R2). • • • ••••
• ~---+--Drug reservoir
~Rate con~rolling
membrane
Monolithic (or Matrix) Devices
L . . . -_ _ Adhesive layer
These devices are used when R 2 is the rate-controlling step (R2 <
Rt) and the drug has a large therapeutic index so that overdosing does Fig. 15.19 Schematic representation of reservoir (membrane)
not precipitate toxic reactions. The two categories of matrix devices type of transdermal drug delivery system
are one in which the drug is dissolved (usually below saturation levels)
368 BIOPHARMACEUTICS AND PHARMACOKINETICS CONTROLLED RELEASE MEDICATION . ~69 .
I

contained within the reservoir as a suspension in a liquid (such as sili- oped for providing zero-order input. The. best ~o~n system is ocular
cone) or gel carrier. The rate-controlling thin polymeric membrane is insert or ocusert developed to deliver pilocarpme tn the treatment of
made of olefinic polymers and copolymers, cellulosic esters, polyamides glaucoma. Available in two release fon_ns _20 ~d ~0 meg/hour, the
or PVC. When applied on skin, the device shows a rapid release at frrst system provides relief for 7 days (following tnsert~on m the c~l-de-sac,
(initial burst effect) followed by a constant zero-order release as long as just below the cornea) in contrast to ey~drop~ which ~e requ_Ired to be
the solution inside the reservoir is saturated. Fig. 15.19 shows such a instilled 3 to 4 times daily. The sy .;tern 1s basically a t~tn, tlextble wafer, .
· device. composed of a drug reservoir core surrounded on eithet si_de by rate-
controlling membranes of ethylene-vinyl acetp··~ copolymer (Ftg. 15.20).
Mixed Monolithic-Reservoir Devices
A third type of system, it is basically a device having drug release
kinetics intennediate between monolithic and reservoir systems. Here the

drug-polymer matrix is layered by a rate-controlling membrane. Release


is controlled initially by_the membrane but as the drug gets depleted, the
rate is controlled by diffusion of drug through a thicker layer of polymer
matrix.

A major limitation of transdennal therapy is poor skin penetrability of Drug reservoir
several drugs. This problem can be overcome by use of penetration '

enhancers such as glycerol, propylene glycol, DMSO, SLS, etc.


Drugs commonly presented in such systems are nitroglycerine, clonidine,
scopolamine and estradiol.
Fig. 15.20 ~_chematic representation of ocusert

OPHTHALMIC DRUG DELIVERY SYSTEMS


INTRAVAGINAL & INTRAUTERINE DRUG DELIVERY SYST~MS
Absorption of ophthalmic drugs across the corneal membrane is a
diffusion process and depends to a large extent on: Controlled release intravaginal systems are used for delivery of contra-
ceptive steroid hormones. The _advantage of, this route i~cludes n~
I. Physicochemical properties of the pem1eating molecule, and
first-pass effect, improved bioavailability and lesser drug d?se In comp~rt­
2. Drainage and output of tears. son to that required by oral route. Two types of devtces have been
Most drugs for ophthalmic use like pilocarpine, epinephrine, local developed a matrix diffusion c,ontrolled device e.g. medroxy ~rogest_er­
anesthetics, atropine, etc. are weak bases which are generally fortnulated one acetate dispersed in viscous silicone elastomer, and the other, dtsso~utton
at acidic pH to enhance stability. But due to their highly ionized form, controlled device e.g. dispersion of a progestin and an estrogen tn an
ocular diffusion is poor. This, coupled with tear drainage, further reduces
- - - - - - - - D r u g reservoir
the- rate and extent of absorption. Moreover, if the drug has short half-
life, the problems become more complicated. Frequent dosing of large
doses of such drugs becomes necessary to .achieve the therapeutic objec-
tive which often results in corresponding increase in local (e.g. irritation)
and systemic side effects. One of the approaches to improve drug effec-
_ ....._Silastic polymer
tiveness is to prolong its contact with corneal surface. Highly viscous
coating
preparations like · suspensions and ointments are intended to achieve this
purpose but do not offer ·the amount of control desired. Continuous
. '
delivery of drugs in a controlled manner can overcome most of these
'
problems. A number of ocular drug delivery systems have been devel- Fig. 15.21 Schematic representation of a vaginal ring


\

370 BIOPHARMACEUTICS AND PHARMACOKINETICS


CONTROLLED RELEASE MEDICATION . 371
aqueous solution of PEG 400 to form microscopic drug reservoir in a
BIOAV AILABILITY TESTING OF CONTROLLED
mixture of silicone elastomers. The device is generally prepared by
RELEASE FORMULATIONS
. extrusion of the resultant composition into a doughnut shaped vaginal ring
-(Fig. 15.21). The system_releases the medicament for 21 days to achieve The purpose of in vivo bioavailability study on a controlled release
a cyclic intravaginal contraception. fottnulation is to detennine:-
1. the fraction of drug absorbed (should ideally be ~ 80% of con-
Intrauterine route is also used for fertility control. Two types of ventional release dosage fortn ).
·medicated intrauterine devices (IUDs) have been developed to produce
2. occurrence of dose dumping.
effective contraception for 12 months or more.
3. influence of food as well as circadian effect on drug a)?sorption.
1. .Copper Medicated IUD 4. the time period for which the plasma concentration stays within

- It .consists of a polypropylene or polyethylene plastic support of num- the therapeutic range i.e. therapeutic occupancy time.
ber-7 or letter T with certain amount of copper wire wound around them.
5. Cmax1Cmin ratios at steady-state.
T shape IUD is popular since its shape confonns to the uterine cavity and
resis~s displacement and rotation within the cavity as well as expulsion 6. percent fluctuation calculated from equation:
'
from the cavity (see Fig. 15.22a). The· copper wire of surface area. 200 1OO(Cmax - Cmin)/Css (15.15)
mm2 shows maximum con_traceptive activity. Oxidation of copper in the '

body fluids releases its. ion slowly a~d exert its effect locally. The device A single dose study is sufficient to assess the frrst thre·e objectives but
is effective for more than 3 years. the subsequent ones can only be evaluated from a multiple dose study.
The reference standard is a solution· or a suspension of the drug or a
2. Progesterone Releasing IUD (P.rogestasert) currently marketed controlled release fonnulation.
It is also aT shaped polyethylene device with a progesterone reservoir For in vitro bioequivalence testing, the dissolution test should be
dispersed in a silicone polymer placed in the vertical arm which is en- designed to account for the major variables to which a controlled release
closed in a sleeve of rate-controlling membrane of ethylene-vinyl acetate
~
fonnulation is exposed in vivo before getting absorbed e.g. pHs from 1 to
copolymer (Fig. 15 ·tj~). The device releases progesterone at a rate of 8, influence of food/fasti~g state, solubilizing influence of bile and pan-
65 meg/day for a penod of one year. creatic secretions, etc. when considering oral controlled release fotntulations.
Establishing in vivo-in vitro correlation for controlled drug delivery is
often difficult due to complexity of variables involved.

QUESTIONS

Rate controlling 1. Define ideal dosage regimen. How is attainment of therapeutic objective
membrane possible by use of conventional formulations?
Progesterone-~
.. reservoir 2. What are the limitations of multiple dosing of conventional immediate
. release dosage forms? What are the various approaches by which they can
Copper
'
wi.ne wound be overcome?
around plastic T
3. Define an ideal controlled drug delivery system. How does it differ from
Threads for insertion or _.,._.----;n targeted and sustained release systems?
removal of device
4. What is the major objective of controlled release fonnulations? List the
(a) (b)
advantages and disadvantages of such a systent.
5. What factors should be considered in the design of a controlled drug deliv-
Fig. 15.22 Contraceptive IUDs--(a) Copper T located ery system?
in the uterus, _and (b) Progestasert

I
CONTROLLED RELEASE MEDICATION 373
372 BIOPI-It\RMACEUTICS AND PHARMACOKINETICS
What would be the dosing frequency if the drug can be formulated as oral
6. Unlike conventional immediate release dosage torrns, what is the rate-limit- controlled release system?
.· ing step in the availability of a drug from controlled release formulation? 25. Which .principle is utilized in the oral delivery of protein and .peptide drugs?
-'-
7. List the ~physicochemical and biopharmaceutic properties of the drug and 26. What are the various ways by which controlled drug release through inject-
state wh~t pharma~okinetic and pharmacodynamic parameters are important . able solutions can be attained?
in the design of controlled release formulation.
27. Among injectable dispersed systems for con.trolled drug release, multiple
8. What criteria are necessary for the selection of a drug as candidate for emulsions and suspensions are superior in comparison to tlie simple emul-
formulation of a controlled release dosage form? Explain giving optimum sions and aqueous suspensions respectively. Explain.
ranges for the biopharmaceutic and phannacokinetic properties/parameters of
the drug·. 28. How are liposomes classified? , Why are they considered versatile. carriers .
for parenteral drug delivery?
9. What are the main determinants in deciding a route for administration
.
of a
controlled release system? 29. What are the advantages and disadvantages of transdermal drug delivery
systems? What criteria are necessary for a drug to be given by such a
10. Zero-order release systems are considered ideal controlled delivery formula- route?
tions. Why?
30. Enumerate the properties of an ideal implantabl.e parenteral system. Why is
11. Even if a drug follows two-compartment kinetics, one-compartment model is subcutaneous tissue considered an ideal site for implants?
considered
'
suitable for the. design of controlled release dosage forms. Why?
31. Ho\\. are infusion devices for controlled drug delivery classified? Give the
12. What assumptions are made in order to define the release pattern of a drug principle behind activation of such devices to effect zero-order drug release.
from controlled release formulation by a suitable model?
32. What are the advantages of using resealed RBCs as drug delivery systemS'? .
13. Name the four models commonly used to express drug input rate from
controlled release formulations. 33. What are the t\VO rate-limiting steps in the absorption of a drug fro!ll
transdermal device? Ho\v do they govern the selection of a particular
14. Give reasons for the observance of flip-flop phenomenon in the pharmacoki- device for controlled drug delivery?
netics of a slow drug release formulation?
34. What are the objectives of zn vivo bioavailability studies. on controlled
15. When is it advisable to incorporate a loading dose in the controlled release release formulations? Why is correlation of such studies \\'ith in vitro
formulation? What limitations are encountered in the multiple dosing of release difficult?
such systems? How can such drawbacks be circumvented?
35. The pharmacokinetic parameters of 1\vo NSAIDs are given in the table
16. What are the causes of poor drug availability from oral controlled release belo\v:
formulations in comparison to immediate release systems?
'
Paranf'eters Diclofenac Ibuprofen
17. Controlled drug delivery systems with slow first-order release are inferior in
comparis9n to zero-order systems for oral use. Explain. t• •~ (hours) 2.0 2.0
18. Classify the oral controlled release formulations. vd (liters) 7.0 8.4
19. Why is it easy to design a dissolution controlled release system? F 0.6 0.9
20. The drug release from diffusion controlled matrix can never be a zero-order Desired C55 (n1cg/ml) 2.0 10.0
process. Explain.
Determine the follov.-ing for each drug:
21. Why are reservoir devices susceptible to dose dumping?
a. The dose needed to maintain the therapeutic concentration for 12 hours.
22. Name and distinguish the· two categories of intrinsically powered oral con- '

trolled delivery formulations for zero-order drug release. Answer : Diclofenac 97 mg and Ibuprofen 388 mg.
23. What are the various approaches by which the gastric transi.t of a dosage b. The zero-order release rate to n1aintain the desired concentration
form can be delayed? To what category of drugs are such approaches Answer : f)iclofenac 8.085 mg/H and Ibuprofen 32.34 mg/H.
'
applicable for controlled delivery?
c. The tin1c in hours to reach 90° o of C55 Yalues.
24. If the rriean residence time of a dosage form in· the GIT is 14 hours and the
A n..,·wer : 6.64 hours for both drugs.
tYl of a drug is 6 hours, for how long can the drug effect be prolonged?
374 BIOPHARMACEUTICS AND PHARMACOKINETICS 375
CONTROLLED RELEASE MEDICATION
d. The loading dose if C85 is to be attained rapidly. d. What should be the zero-order release rate for both drugs?
Answer : Diclofenac 23.3 mg

and Ibuprofen 93.3 mg . Answer : Propranolol 9.52 mg!H and Sotalol 13.58 mg/H. . ..
e. The plasma drug concentration at the end of 5 hours if the release is zero- . · the desired level for 24
e. How much drug should be incorporated to matntatn
order.
hours?
· Answer : Diclofenac 1.64 mcg/ml and Ibuprofen 8.23 meg/mi. Answer : Propranolol 228.5 mg and Sotalol 326 mg.
. I ?
f. The possibility to formulate once daily formulation for both drugs . f. How much time will be required to attain 90% of desired Css va ue ·

36. An oral controlled release tablet of pentoxifylline having a loading dose of Answer : Propranolol 13.3 hours and Sotalol 40 bours. .
170 mg to attain the desired C 55 of 0.3 mcg/ml is to be formulated. If the · . . C · to be attained raptdly?
1
V d is 170 ! liters and tYz and F after rapid release are 1 hour and 0.35
g. What should be the loading dose tf the destred ss ts .
respectively, calculate: - Answer : Propranolol 55 mg and Sotalol 235.2 mg. .
• · d release systems wtth
. ·a. The amount of drug to be placed in the zero-order controlled release core to h. If both the drugs are formulated as slow first-or er O!H and 0 _5/H
maintain the desired level for 12 hours. dose/unit 240 mg and 320 mg and release rate constant, Kr 1·
. . d . the plasma drug concen-
for propranolol and sotalol respectively, etenntne ·
Answer : 1212 mg.
tration after 5 hours from administration.
b. If it is observed that oral availability from CR drug reduces to 0.3 and tYz
Answer : Propranolol 0.22 mcg/ml .and Sotalol 2.3 meg/mi.
increases to 3.5 hours, what should be the 12 hour maintenance dose?
Answer : 404 mg.
c. What will be the plasma drug concentration after 4 hours of administration
of such a formulation if Ka is 2.2/H and F and tYz for zero-order doses are
0.3 and 3.5 hours respectively?
Answer : 0.192 meg/mi.
37. The following information is available about two beta-blockers:
I

Propranolol Sotalol

Molecular weight 259.3 277.4


pKa (base) 9.5 9.8


Aqueous solubility . Soluble Freely soluble
Octanol/water partition coefficient 3.65 0.8
Oral availability 0.5 1.0
tmax (hours) 1.0 2.0
Therapeutic index 10.0 8.0
Elimination half-life (hours) 4.0 12.0
Apparent volume of distribution (L) 275.0 105.0
Minimum effective concentration (mcg/ml) 0.02 0.56

a. · Evaluate from the data the suitability of both drugs as candidates for oral
controlled release system.
b~
·, .
Out of the two, which drug is a better candidate? Why?
c. Determine the desired Css for both drugs (Hint: C = MEC X TI/2)
55

An.Swer : Propranolol 0.1 mcg/ml and Sotalol 2.24 meg/mi.


BIBLIOGRAPHY 377

Kydoneus, A.: Treatise on Controlled Drug Delivery, New York, Marcel


Dekker, Inc., 1992.
La Du, B.N., Mandel, H.G. and Way, E.L.: Fundamentals of Drug Me-
BIBLIOGRAPHY tabolism and Drug Disposition, Baltimore, Williams and Wilkins,
Abdou, H.M.: Dissolution, Bioavailability and Bioequivalence, Pennsylva- 1971.
nia, Mack Publishing Company, 1989. Loper, A.E. and Gardner, C.R.: Gastrointestinal Absorption of Drugs. In:
Banakar, U.V.: Pharmaceutical Dissolution Testing, New York, Marcel Encyclopedia of Pharmaceutical Technology, vol. 6 (Swarbrick, J.
--Dekker, Inc., 1992. and Boylan, J.C., eds.), New York, Marcel Dekker, Inc., 1992.
Blanchard, J., Sawchuk, R.J. and Brodie, B.B.: Principles and Perspec- Low, L.K. and Castagnoli, N.Jr.: Metabolic Changes of Drugs and Related
tives in Drug Bioavailability, New York, S. Karger, 1979. Organic Compounds. In: Wilson and Gisvold's Textbook of Organic,
Medicinal and Pharmaceutical Chemistry, 9th edition (Delgado, J.N. '
Bourne, D.W.A. and Dittert, L.W.: Phartnacokinetics. In: Modern
and Remers, W.A., eds.), Philadelphia, J.B. Lippincott Company, 1991.
Pharmaceutics, 2nd edition (Banker, G.S. and Rhodes, C.T;, eds.),
New York, Marcel Dekker, Inc., 1990. Mayersohn, M.: Principles of Drug Absorption. In: Modern Pharmaceutics,
2nd edition (Banker, G.S. and Rhodes, C.T., eds.), New York, Marcel
Chasseand, L.F. and Hawkins, D.R.: Biotransfottnation of Drugs. In:
Dekker, Inc., 1990.
Encyclopedia of Pharmaceutical Technology, vol. 2 (Swarbrick, J.
and Boylan, J.C., eds.), New York, Marcel Dekker, Inc., 1990. \
Me Elnay, J.C.: Buccal Absorption of Drugs. In: Encyclopedia of Phar-
maceutical Technology, vol. 2 (Swarbrick, J. and Boylan, J.C., eds.),
Chien, Y.W.: Novel Drug Delivery Systems, 2nd edition, New York,
New York, Marcel Dekker, Inc., 1990.
Marcel Dekker, Inc., 1992.
Meyer, M.C.: Bioavailability of Drugs and Bioequivalence. In: Encyclo-
Florence, A.T. and Attwood, D.: Physicochemical Principles of Pharmacy,
.. pedia of Pharmaceutical Technology, vol. 1 (Swarbrick, J. and Boylan,
London, Macmillan, 1981 .
J.C., eds.), New York, Marcel Dekker, Inc., 1988.
Ford, J.L.: Dissolution and Dissolution Testing. In: Encyclopedia of
Niazi, S.: Textbook of Biopharmaceutics and C'/inica/ Pharmacokinetics,
Pharmaceutical Technology, vol. 4 (Swarbrick, J. and Boylan, J.C.,
New York, Appleton Century Crofts, 1979.
eds.), New York, Marcel Dekker, Inc., 1991.
Notari, R.E.: Biopharmaceutics and Clinical Pharn1acol<if1eiics: An Intro-
Gibaldi, M.: Biopharmaceutics and Clinical Pharmacokinetics 4th edi-
duction, 4th edition, New York, Marcel Dekker, Inc., 1987.
. tion, Philadelphia, Lea and Febiger, 1991. '
- - . .. .. - ·+-
Reidenberg, M.M. and EriiJ, S.: Drug-Protein Binding, New York, Praeger,
Gibaldi, M. and Perrier, D: Pharmacokinetics, 2nd edition, New York, ,
1986.
Marcel Dekker, Inc., 1982.
Ritschel, W.A.: Handbook of Clinical Pharmacokinetics, ~rd edition, Illi-
Gibson, C.G. and Skett, P.: Introduction to Drug Metabolism London
' ' nois, Drug Intelligence Publications, Inc., 1986.
Chapman and Hall, 1986.
Robinson, J.R. and Lee, V.H.: Controlled Drug Delivery: Fundamentals
G~iffm, ~.P., D' Arcy, P.F. and Speirs, C.J.:' A Manual of Drug Interac-
and Applications, 2nd edition, New York, Marcel Dekker, Inc., - 1987.
tions, 4th edition, Bombay, K.M.Varghese Company, 1988. /

Rowland, M. and Tozer, T.N.: Clinical Pharmacokinetics: Concepts and


Hoener, B.A. and Benet, L.Z.: Factors Influencing Drug Absorption and
Applications, 2nd edition, Philadelphia, Lea and Febiger, 1989.
Drug Availability. In: Modern Pharmaceutics, 2nd edition (Banker,
G.S. and Rhodes,. C.T., eds.), New York, Marcel Dekker, Inc., 1990. Sharjel, L. and Yu, A.B.C.: Applied Biopharmaceutics and Pharmacoki-
netics, 2nd edition, Connecticut, Appleton Century Crofts, 1985.
Jolles, J. and Woolridge, K.R.H.: Drug Design-Fact or Fantasy?, Lon-
don, Academic Press, 1984. Sloan, ~.B.: Prodrugs: Topical and Ocular Drug Delivery, New York,
Marcel Dekker, Inc., 1992 .
..
376
.
378 BIOPHARMACEUTICS AND PHARMACOKINETICS

Smith, H.J.: Smith and William's Introduction to the Principles of Drug SUBJECT INDEX
Design, 2nd edition, London, Wright, 1988.
Stockley, I.H.: Drug Interactions: A Sourcebook of Drug Interactions, Page numbers followed by f refer to figures; by t indicate tables.
Their Mechanisms, Clinical Importance and Management, 2nd edi-
tion, London, BlackweJI Scientific Publications, 1991. Absolute bioavailability, 283 sublingual, 64
Absolute solubility, defined, 19 vaginal, 70
Swarbrick, J.: Current Concepts in Pharmaceutical Sciences: Absolute surface area, ·25 Absorption half-life, 250
Biopharmaceutics, Philadelphia, Lea and Febiger, 1970. Absorption, (See also Availability; Absorption interactions, 208t
Swarbrick, J.: Current Concepts in Pharmaceutical Sciences: Dosage Form Bioavailability) Absorption rate constant, 248
Design and Bioavailability, Philadelphia, Lea and Febiger, 1973. active transport in, 13 determination of,
• adsorption and, 61 by method of residuals, 248
Taylor, J.B.: Comprehensive Medicinal Chemistry-val. 5: Biopharmaceutics, barrier, 7 by Wagner-Nelson method, 250
England, Pergamon Press, 1990. buccal, 64 from urinary excretion data, 259
Testa, B. and Jenner, P.: Drug Metabolism: Chemical and Biochemical capacity-limited, 12 Absorption window, defined, 12
Aspects, New York, Marcel Dekker, Inc., 1976. carrier-mediated transport processes Accumulation during multiple dosing, 309
in, 10 Accumulation index, 3 10
Tyle, P: Drug Delivery Devices: Fundamentals and Applications, New cell membrane - structure and physi- Acetylation, 144
York, Marcel Dekker, Inc., 1988. ology in, 6, 7/ polymorphism, 145
defined, 1, 5 a 1-Acid glycoprotein (AAG, AGP), 92t
Vadnere, M.K.: Coprecipitates and Melts. In: Encyclopedia of Pharma-
dissolution rate-limited, 19 drug binding to, 94
ceutical Technology, vol. 3 (Swarbrick, J.and Boylan, J.C., eds.) New dosage form characteristics and phar- influence of disease on levels of, and
York, Marcel Dekker, Inc., 1990. maceutical ingredients affecting, binding to, 102t
Wagner, J.G.: Biopharmaceutics and Relevant Pharmacokinetics, Wash- 18t, 39 Acidification of urine, 184
ington, DC, Drug Intelligence Publications, 1971. endocytosis and, 15 Acidosis and urine pH, 182
facilitated diffusion in, 12 Active metabolites, · 113t
Welling, P.G.: Absorption of Drugs. In: Encyclopedia of Pharmaceutical factors influencing, 16, 18t forniation of, and concentration-re-
Technology, voi. 1 (Swarbrick, J. and Boylan, J.C., eds.), New York, from noninvasive routes, 64f sponse relationship, 326
Marcel Dekker, Inc., 1988. from non per os routes, 63, 71 t Active transport,
gastrointe~tinal, 6 in biliary excretion of drugs, 195
Welling, P.G., Tse, F.L.S. and Dighe, S.V.: Pharmaceutical Bioequivalence,
New York, Marcel Dekker, Inc., 1991. interactions affecting, 206, 208t in drug absorption, 13, 16/
intramuscular, 67 Active tubular reabsorption of nutrients,
Wolff, M.E.: Burger's Medicinal Chemistry, Part I: The Basis of Medici- mechanisms of, 7, 71t, 16/ 180
nal Chemistry, 4th edition, John Wiley & Sons, 1980. nature and type of dosage form af- Active tubular secretion, 178, 180
fecting, 18!, 45 mechanism of, 180
nonlinearities in, 274 Acute pharmacologic response,
passive. See Passive diffusion/tran$- bioavailability determination from,
port 290
patient related factors affecting, l8t, 50 Addition interactions, defined, 205
percutaneous, 66 ADEPT technique in cancer chemo-
permeability rate-limited, 19 therapy, 175/
physicochemical factors affecting, 18t, Adipose tissue,

I
• 19 drug localization in. 97
pulmonary, 69 Administration. (See also particular routes
rate-determining processes in, 19 of administration)
rectal, 65 routes of, 6
subcutaneous, 68 transmucosal noninvasive routes oC 64/

379
380 BIOPHARMACEUTICS AND PHARMACOKI~ '~TICS
INDEX 381
Age Aqueous suspensions
differences in metabolism due to, 152 for parenteral controlled release, 359 apparent volume of distribution and, Bioprecursors, defined (See also
influence on Aqueous unstirred diffusion layer, 38 103 Prodrugs ), 161
drug absorption, 53 Area under the curve (AUC), 214 drug interactions involving. See Dis- Bioreductions. See Reductive reactions
drug distribution, 85 as a measure of bioavailability, 214, placement interactions Bioreversible derivatives. See Prodrugs
drug metabolism, 152 286 factors affecting, 97 Biotransformation. See also Metabolism
protein-drug binding, I 0 I assessment of, 225 irreversible, 92 chemical pathways of, I16t
renal function and drug excretion, Area under the first moment curve kinetics of, 106 defined, 2, Ill
I

189 ' significance of, 103 factors affecting, 147


(AUMC), 224
of product and drug dissolution, 50 Area under the zero moment curve to blood components, 92 Blood~ binding of drugs-to components
of skin and drug absorption, 67 (AUC), 224 Bioactivation, defined, 154 of, 92
Agglomerative phase of communition Aromatic hydroxylation. See Oxidation (See also Toxicological activation) Blood-brain barrier (BBB ), 80/
(APOC), effect on dissolution, 40 of aromatic C-atoms Bioavailable ·. dose, defined, 282 altered permeability of, in disease
Albumin Artificial kidney (See also Hemodialyzer, Bioavailable fraction, defined, _283 states, 86
drug binding to, 92t, 93, 94[ 193 Bioavailability approaches to promote crossing of, 81
in disease states, I 02t Asynthetic reactions. See Phase I reactions absolute, 283 Blood cells, drug binding to, 95
Alicyclic hydroxylation. See Oxidation Autoinduction, 148 defined, 283 Blood-~erebrospinal fluid barrier, 81/
of alicyclic C-atoms Availability (See also Abs~rption; considerations in study design, 283 Blood flow
Aliphatic hydroxylation. See ()xidation Bioavai lability) correlation with in vitro dissolution, hepatic, determination of, 238
of aliphatic C-atoms defined. 283 290, 292 to GIT and absorption, 59
Alkalinization of urine. 182 relative. 284 defined, 1, 17, 282 to kidneys, perfusion rate-limited ex-
Alkalosis and urine pH. 182 Average concentration on multiple dos- factors influencing 16, 18t cretion, 189
Ali-or-none response(s). 327 ing to steady-state. 311 methods for enhancement of, 296 Blood flow rate-limited models. See
(See also Quanta I response) Azapropazone binding site methods for measurement of, 284 Physiologic models
Allosteric changes in protein n1olecule. \varfarin and. ,,93 multiple dose studies, 284 Blood proteins
I 0I objectives of, 283 and drug binding to, 92t
Allosteric effector. I 0 I Barrier. See Physiologic barriers problems, and approaches to over- Blood-testis barrier, 83
Altered density systems Bile salts. absorption interactions "·ith come, 297 Body surface area
.
for prolonged Gl residence. 351 drugs. 61 rel~ive, defined, 284 dose estimation for children USing,
Amount remaining to be absorbed BiliarY, clearance. defined. 197 determination of bioequivalence 317
(ARA). 251 BiliarY, excretion. 195 from, 294 ideal. See Ideal body weight
Amount remaining to be excreted (ARE). assessment of. 1.98 single-dose studies, 284 Body water, compartments of, 87t
257 drug interactions involving. {)2. Bioequivalence Body weight, relationship with surface
'
Amorphism. 27 198. 207 defined, 294 area, 317
Antagonism. defined. 205 factors influencing. 195 Latin square study design for, 295 Bolus dose
Antibody directed enzyme prodrug Biliary, excretion rate. 197 statistical interpretation of data, 296 drug concentration after multiple i.v.
therapy. See ADEPT Bilirubin Bioinequivalence, defined, 294 injections, 309
Antifrictional agents. See Lubricants impaired binding to HSA by drugs, Biological approach to overcome in one-compartment open model, 23 I
Apparent partition coefficient of drug 105 bioavailability problems, 297 in two-compartment open model, 260
in· design of controlled release sys- precipitation of kernicterus in neo- Biological availability. See Bioavailability Bones, drug binding to, 97
tems. 339 nates bY.
, I 01. I 05 Biopharmaceutics, defined, 1
/
Brain
Apparent volume of distribution. 86. 234 Binders Biopharmaceutic characteristics of drug redox system for d~g delivery , to,
(See also Volume of distribution) granulating agents and. 42 in design of controlled release sys- 81, 172 .
assessment of. 234. 244. 252 influence on dissolution and absorp- tems, 338 Brodie et a/ hypothesis. See pli-parti-
at steady-state. 265 tion. 42 Biopharmaceutic considerations in dos- tion hypothesis
defined. 86 Binding (See also Protein binding of age form design. See Factors Buccal administration, 64
protein binding of drugs and. I03 drugs) 91 influencing absorption and absorption mechanism after, 71 t
bioavailability factors in absorption after. 65
INDEX 383
382 BIOPHARMACEUTICS AND PHARMACOKINETICS

Buffers Chemical equivalence, defined, 294 enteric, Concentration-time profile


influence on drug dissolution, 44 Chemical factors, affecting biotransfor- effect on drug absorption, 49 absorption rate and, 245/
Buoyant ta~let/capsule. See Floating tab- mation of drugs, 148 factors influencing drug release from, after constant rate i. v. infusion,
let/capsule Chemical formulation, defined, 162 49 with one-compartment open model,
Burst effect, in controlled release for- Chemical pathways of biotransforma- Coatings,

effect on drug dissolution, 43 241/
mulations, 345 tion, 114, 116t Coevaporation, 301 after e. v. administration, 245[, 249[,
Chemical stability, enhancement of, by Colonic release systems, 356 267
Calcium, complexation with tetracycline, prodrug design, 165 Colorants, effect on dissolution rate, 45 after i. v. bolus administration,
42, 61, 208t Children, dosing of drugs in, 31 7 Compartment(s) with one-compartment open model,
Cancer Child's dose central, 221 233
ADEPT technique in chemotherapy equation for calculating, 317, 318 defined, 221 with two-compartment open model,
of, 175 Cholestasis models, (See also particular models}, 261
site-specific delivery in, 174 influ~nce on biliary excretion, 196 221 area under the (AUC), 214
Capacity-limited kinetics. See Nonlinear Chronokinetics, defined, 154 peripheral, 221 first moment of, 224
pharmacokinetics Chronophannacology, defined, 154 reference, 230 post .distribution phase of,
Capillary endothelial barrier, 79 Chylomicrons, 94, 356 tissue, 221 zero moment of, 224
Capsule(s) Circadian rhythm Compartmental analysis, 221 Conjugation
factors in absorption of drug from influence on biotransformation, 154 Compartmental modeling, 23 0 of cyanide, 146
hard gelatin, 41, 4 7 Clearance advantages and disadvantages of, 223 reactions (See also Phase II reac-
soft elastic, 48 apparent Vd and, 236, 252 Competition for active transport tions), 116t
Carbon glucuronides, 141 biliary. See Biliary clearance therapeutic use of, 180 with alpha-amino acids, 142
Carbon-nitrogen systems, oxidation of, 122 creatinine. See Creatinine clearance Competitive displacement interactions, with glucuronic acid, 13 9
Carbon-oxygen systems, oxidation of, 128 defined, 186, 23 5 209t with glutathione (GSH), 143
Carbon-sulfur systems, oxidation of, 127 dialysis. See Dialysis clearance Competitive inhibition, 149 with ribose, 147
Carbonic anhydrase, drug binding to, 95 estimation of, Complexation with sulfate moieties, 141
Cardiovascular disease after constant rate i. v. infusion, 244 influence on absorption, 61 with taurine, 147
influence on bioavailability, 58 after e. v. administration, 236, 252 to enhance bioavailability, 298 Constant rate processes. See Zero-order
Carrier after i. v. bolus do,se, 236 Complexes " kinetics/processes
of drug mol~cules, 11 half-life and, 236 poorly soluble, Contraceptive(s)
of prodrugs, 161 hepatic. See Hepatic clearance in controlled drug release, 352 controlled release intrauterine devices
Carrier-linked prodrugs, 161 in renal impairment, 191 resin-drug, (IUDs), 370
Carrier-mediated transport, I 0-12 intrinsic. See Intrinsic capacity clear- in controlled release, 3 52 controlled release intravaginal sys-
Cascade latentiation. See Double prodrugs ance Complexing agents, 45 tems, 369
Catemary model, 223 nonrenal, 236 Compression force implants, 363
Cation radical, 155 organ, 23 5, 23 7 influence on tablet dissolution rate, oral, enterohepatic circulation of, 62
Celiac disease, 58 plasma, 235 41/ I Controlled release medication, 335
Cell membrane renal. See Renal clearance Concentration. (See also Plasma con- advantages of, 33 5
of RBC, drug binding to, 95 total body, 185 centration) bioavailability testing of, 371
structure and physiology of, 6, 7/ total systemic, 18 5 and pharmacologic response, 325 drug· release patterns of, 343
Cell membrane barrier, 79/ unbound, 235 Concentration-effect relationship(s) 327 Controlled release/delivery systems
Central compartment Clinical equivalence. See therapeutic Concentration gradient, defined, 8 biopharmaceutic characteristics in the
apparent volume of, 262 Concentrati~n-response curve( s) design of, 338
equivalence
defined, 221, 260 Clinical pharmacokinetics, defined, 2, effect of shape factor on, 329 classification of, 348
drug concentration in, 262 212, 315 Concentration-response relationship(s}, defined, 336
Cerebrospinal fluid Coated tablets 327 ·design of, 337
drug entry into, 82 •
bioavailability from, 49 factors complicating, 325 ideal. See Ideal controlled drug deliv-
Ceruloplasmin, 95 quantitative models for, 327 ery system
384 BIOPHARMACEUTICS AND PHARMACOKINETICS INDEX 385

intravaginal and intrauterine, 369 Delayed transit and continuous release Disintegration time, rotating basket apparatus, 291
ophthalmic, 368 systems, 348 influence on dissolution and absorp- rotating paddle apparatus, 292
oral.. 347 .~ . Dermal excretion. See Skin excretion tion, 39 Distribution
~

parenteral, 3 57 l- Desulfuration, 128 Dispersions, solid. See Solid dispersions apparent volume of. See Apparent
pharmacodynamic·/ characteristics in Detoxication, 115 Dispersions for parenteral controlled re- volume of distribution
. "'
the destgn of, 341 Dialysance. See Dialysis clearance lease, 358 . defi~ed, 1, 75, 76

pharmacokinetic characteristics in the Dialysate, 194 Displaced drug, defined, 99 exponent, 262
design of, 340 Dialysis, 192 Displacement interaction(s), 99, 209t factors affecting, 76
pharmacokinetic principles in the de- defined, 192. (See also Hemodialysis) and toxicity, 105 binding of drugs to tissue compo-
sign and fabrication of, 342 extracorporeal, 193 clinically significant, 99 nents, 85
transdennal, 365 hemoperfusion and, 192 defined, 99 blood flow to the organ/tissue, 83
Conventional dosage fonn(s), limitations peritoneal, 193 Displacer, defined, 99
~-
miscellaneous, 85
of, 335 systems for drug dissolution, 291 Disposition. (See also Distribution; Elimi- tissue permeability of drugs, 76
Coprecipitation, 301 Dialysis clearance, 194 nation) - interactions affecting, 206, 209t
Corpuscular transport. See Endocytosis Dialyzing fluid, composition of, 194 defined, 75 nonlinearity in, 274
Correlations, in vitro-in vivo, 292 Diazepam binding site, 93, 94t monoexponential, rate-limiting steps in, 76
Corticosteroid binding globulin (CBG), 95 Diet, influence on metabolism, 152 in one-comparttnent model, 232 rate of penetrability, 76
Creatinine Diffusion Disposition curve, birxponential, rate of perfusion, 83
serum, 191 facilitated, 12 for two-compartment kinetics, 261 Distribution half-life, 8Li
age related changes in, 191 passive. See Passive diffusion/trans- Dissociation constant of drug Distribution rate constant, 84
determination of renal function port and pH-partition hypothesis, 18t, 32 Distributiv·e phase, 261
from, 192 Diffusion coefficient, 21 and urinary excretion, 183 Diuresis, forced, to treat overdose, 185
use in the determination of GFR, Diffusion controlled release systems, 350 Dissolution, defined, 20 Dizygotic twins, 151
179 Diffusion layer, 20 rate-determining step in absorption, 19 Dosage form(s)
Creatinine clearance, aqueous unstirred, 38f theories of, 20 characteristics affecting absorption,
formula( e) to calculate, 191 pH of, Danckwert's model, 23 18t' 39 --- --
methods( s) for determining, 192 drug dissolution and, 38 diffusion layer model, 20 nature and type of,
renal function and, 192 model, 20 interfacial barrier model, 24 affecting bioavailability, l8t, 45
Crohn's disease, effect on absorption, 58 Diffusivity. See Diffusion coefficient Dissolution and diffusion controlled re- Dosage form index, defined, 341
Crystal growth inhibitors Digitoxin binding site, 93, 94f , lease systems, 351 Dosage regimen( s)
influence on drug in suspension, 45 Diluents, influence on dissolution and Dissolution controlled release systems, 348 defined, 4, 307
· Crystalline form( s) absorption, 42 Dissolution rate design of, 307
polymorphism and, 27 Direct linear plot and bioavailability, 290 from plasma concentration, 314
Cytochrome bs-reductase. See Cyto- for estimation of Km and Vmax' 279 apparatus. See Dissolution testing mod- ideal. See Ideal dosage regimen
chrome P-450 reductase Direct pharmacodynamic interactions, els individualization of, 315
Cytochrome P-450 defined, 204 defined, 19 optimal multiple, defined, 307

oxidation-reduction cycle, 117, 118/ Discontinuous variation factors affecting, 25 Dose


Cytochrome P-450 reductase, 117 in drug metabo Iism, 151 methods for enhancement of, 297 loading. See Loading dose
Cubic root law of dissolution Disease states. (See also specific dis- ' in vitro, maintenance. See Maintenance dose
Hixson and Crowell's, 23 eases) correlation with in vivo absorption, Dose adjustment
affecting drug absorption, 58 292 in renal di~ease, 192
Danckwert's model . affecting drug distribution, 86 Dissolution test based on elimination rate constant
for drug dissolution, . .20, 23 affecting drug metabolism, 153 factors in the design of, 291 and-half-life, 319
I -

Dehydrogenation. See Oxidative aroma- affecting protein-drug bi~ding, 102t Dissolution testing models, 290 based on total body clearance, 3 19
tization Disinte~ts
4
closed-compartment apparatus, 291 Dose-dependent kinetics. See Nonlinear
Delayed distribution models. See influence on absorption, 43 dialysis system, 291 phannacokinetics ·
Multicompartment moQels Disintegration test, 39 open-compartment apparatus, 291 Dose ratio, defined, 313
'
.
-
BIOPHARMACEUTICS AND PHARMACOKINETICS INDEX 387 .
-Dose size- Electrophiles Enzyme induction, defined, 148 False nutrient~-
inthe design of dosage regimen, 308 tissue toxicity due to, 154 Enzyme induction interactions, 209t carrier-mediated transport of, ll
influence of, Elimination, defined, 2, 75, 111 Enzyme inhibition, defined, 149 Fats, drug accumulation in, 97
on plasma concentration-time pro- (See also Metabolism; Excretion) Enzyme inhibition interactions, 21 Ot Feathering technique. See Method of
r file, 308 Elimination half-life, .. Epidermis, barrier to percutaneous ab- residuals
Dosing frequency, 308 as a secondary parameter, 234 sorption, 66 Fetus
·. Dosing interval, 308 clearance and, 234 Equivalence, defined, 294 drug danger to, 83
-, Dosing of drugs defined, 233 Erythrocytes, drug loading in, 362/ drug transport to, 82
in elderly, 318 dose adjustment based on, 319 Esophageal transit, defined, 56 (See also Placental barrier)
-_.- in hepatic diseases; 318 estimation of, Ethnic variations in metabolism, defined, Fick's first law of diffusion, 8, 21
· in neonates, infants and children, 317 from plasma data, 232 157 Fick 's second law of diffusion, 21
in obese patients, 316 from urinary excretion data, 253 Eutectic mixtures, 300 Fillers. See Diluents
in renal disease, 319 Elimination phase, 231, 261 Excipients, influence on absorption/ Film theory for dissolution. See Diffu-
Double barrier model. See Interfacial
~
of plasma concentration-time profile, bioavailability, I8t, 41 sion layer model
- barrier model 214, 245/ Excretion, 178 First moment curve, 224
Double prod~g, 161 Elimination rate constant, (See also Clearance; Elimination) First-order absorption, 246
.. . Doublt( reciprocal plot. (See also estimation of, biliary. See Biliary excretion, First-order absorption model, 246
Line':"~aver Bud( plot) • from plasma data, 232, 244, 248, defined, 2, 178 First-order half-life, 219
for ptotein-drug binding, I 09 · 265 gastrointestinal. See Gastrointestinal First-order kinetics. (See also Linear
·pownhill transport, 9, 12, 14 from urinary excretion data, 255, excretion kinetics), 218
· Drug administration and therapy.. 256 genital. See Genital excretion First-order process(es), 218
four phases of, . 2 Emulsions interactions, 21 Ot First-order rate constants, 218
Drug-drug interactions in the GIT, 61 factors in the absorption of drug from, mammary. See Mammary excretion First pass effect. See Presystemic me-
Drug interactions, 204 47 nonlinearity in, 275 tabolism
ADME, 206 multiple. See Multiple emulsions pulmonary. See Pulmonary excretion Flip-flop phenomena, defined, 250
defined, 204 Enantiotropic polymorphs, 27 ratio. See Renal clearance ratio in controlled release formulations, 345
mechanisms of, 204 Encephalitis renal. See Renal excretion Floating tablets/capsules, 356
Drug latentiation, defmed, 160 permeability of BBB in, 86 salivary. See Salivary excretion Fluctuation, defined, 311
Drug release patterns Endocytosis, 15, 16/,· 71t skin. See Skin excretion Fluid compartments, 87t
of CO!ltrolled delivery systems, 343 Endogenous agents Exogenous compounds. See Xenobiotics Fluid volume
,~Drug transport, defined, 7 as natural soft drugs, 113, 160 Exponential kinetics, 218 influence on GI absorption, 60
.'
' ''{_S~e also Mechanisms of absorption) Enteral route, 6 Extracellular fluid volume, 87t Food. (See also Diet)
Duration of action, Enteric coated tablets. See coated tablets Extraction ratio, defined, 23 7 interactions due to, 59
defined, 215 Enterohepatic cycling. (See also Biliary hepatic, 238 Food-drug interactions
prodrugs to prolong, 167 excretion), 195, 197/, 198 Extrahepatic metabolism, defined, 113 effect on absorption, 59t
Environmental chemicals Extrarenal routes of excretion. See Formulation factors, absorption and, 41
Eadie-Hofstee plot, 330/ influence on metabolism, 150 Nonrenal routes of excretion Fraction excreted unchanged, 23 3
Effect(s). See Response Enzyme(s) Extravascular administration, Fraction metabolized, 233
Effective surface area of particles, 25 bacterial, effect on orally adminis- one-compartment kinetics after, 245 Fraction unbound, I 04

Elder~¥, dosing of drugs in, 3 18 . tered drugs, {?3 two-compartment kinetics after, 266 Fraternal twins. See Dizygotic twins
Electrical gradient, 14 drug metabolizing, 113 Eye(s). (See also Intraocular adminis- Free radicals, and tissue toxicity, 154
Electrochemical diffusion, defined, 14 gut wall, effect on orally adminis- tration) Functionalization reactions. See Phase I
(See also Ionic diffusion) tered drugs, 63 controlled release medication for, 268 reactions
Electrochemical grad~ent. See Concen- hepatic, effect on orally administered drug binding to melanin of, 96
. tration gradient · drugs, 63 prodrugs for selective targeting to, Gastric emptying
Electron transfer chain lumenal, effect on orally adminis- 166, 170 as a rate-limiting step in absorption,
components of, 117 tered drugs, 63 topical application to, 70 53
388 BIOPHARMACEUTICS AND PHARMACOKINETICS INDEX 389
defined, 53 Hairs, drug binding to, 97 Hydration of skin, 67 Inducers, of drug metabolism, 1481
delayed, 54 Half-life Hydrodynamic pressure controlled sys- categories of, 148
factors influencing, 54 absorption. See Absorption half-life tems Infant(s)
Gastric emptying rate, 54 biological. See Elimination half-life for oral controlled release, 354 differences in P-D binding in, 101
Gastric emptying t~, 54 distribution. See Distribution half-life Hydrolysis dosing of drugs in, 3 17
Gastric emptying time, 54 elimination half-life. See Elimination of amides, 136 Infusion
Gastrointestinal contents half-life of ethers and esters, 134 advantages of, 240
influence on absorption, 59 Hanes-Woolf plot, 278 Hydrolytic cleavage of nonaromatic het- constant rate, 241
Gastrointestinal diseases Hard drugs, defined, 159 -et_:ocycles, 137 equilibrium, 2~0
influence on absorption, 58 Hemodialysis, factors governing removal Hydrolytic dehalogenation, 138 plus loading dose, 243-
Gastrointestinal enzymes of substances by, 193 Hydrolytic reactions, 1161, 134 , plasma concentration-time profile
1 presystemic metabolism by, 62 Hemodialyzer, 193/ miscellaneous, 138 after, 243/
Gastrointestinal excretion, 201 Hemoglobin, drug binding to, 95 Hydrophilic diluents Infusion devices, as parenteral controlled
Gastrointestinal pH Hemoperfusion, 194- to reduce hydrophobicity, 42 release systems, 363
influence on absorption, 57 Henderson-Hasselbach equation(s) Hydrophilic-lipophilic balance (HLB) battery powered systems, 365
Gastrointestinal tract (GIT), 50 to determine degree of ionization, 33 for optimum bioavailability, 35 osmotic pressure activated systems, 363
blood flow to the, Hepatic and biliary malfunction Bydroxylases. See Mixed function vapor pressure activated systems, 364
influence on absorption, 59 marker used to determine, 197 oxidases Inhalation route. See Pulmonary admin-
presystemic metabolism in, 62 Hepatic blood flow, 238 Hyperlipoproteinemia istration
Genital excretion, 201 Hepatic clearance, 238 effect on binding, 102 Inhibition of enzymes, 149
Glass dispersion, 301 flow dependent, 238 Intensity of action, defined, 215
Hypoalbuminemia
Glass solution, 299 intrinsic capacity-limited, 239 · -effect on binding, 102 Intensity of effect/response
Glass suspension, 301 Hepatic diseases concentration relationships, 332/
Glassy hydrogels, defined, 3 51 dosing of drugs in, 318 Ideal body weight, 3 16 Interactions. See Drug interactions
Globulins, binding of drugs to, 95 · influence on bioavailability, 59 Ideal controlled drug delivery system,· 336 Interfacial barrier model for dissolution,
Glomerular filtration, 178, 179/ Hepatic enzymes Ideal dosage regimen, 33 5 24
Glomerular filtration rate (GFR), 179 in first-pass metabolism, 63 Ideal implantable parenteral system, Intersubject variability, 315
creatinine clearance

and, Hepatic extraction ratio of drugs, 2391 properties of, 362 in drug binding, 102
det~rmination of, 179 Hepatic function, determination of, 143 Ideal prodrug, properties of, 162 Intestinal release systems, to bypass
Glucopyranosiduronic acid conjugate,·· Hepatotoxicity of paracetamol, 126, 15 5 Ideal targeted drug delivery system, de- presystemic metabolism, 356
140 . High density lipoproteins (HDL ), 94 fmed, 336 Intestinal transit, 56,
Glucosiduronic acid conjugate, 140 High density pellets Identical twins, drug metabolism in, 151 Intracellular fluid volume
Glucuronic acid, 139 for prolonged GI residence, 355 Immediate release dose. See Loading determination of, 881
Glucuronidation. (See also Conjugation Highly perfused tissues, 84t, 221, 260 dose Intramuscular administration, 67
with glucuronic acid), 139 Hixson and Crowell's Implants, as parenteral controlled re- · factors in absorption after, 68
reasons for importance of, 139 cubic root law of dissolution, 23 lease systems, 362 Intranasal administration/absorption
steps involved in, 140 Homing device, in ADEPT system, 175 Incompatibility. See Pharmaceutical molecular weight and, 69
Glucuronide(s) Hormonal imbalance interactions Intraocular administration/absorption, 70
carbon, 141 influence on metabolism, 153 Indirect pharmacodynamic interactions, Intraocular penetration
formatjon of. See Glucuronidation Human serum albumin (HSA), 92t defined, 205 !) prodrugs for, 166, 170
nitrogen, 141 (See also Albumin) Individualization Intrasubject variability, defmed, 315
oxygen, 140 binding sites on, 93 of dosage regimen, 3 15 Intrauterine devices (IUDs), 369
sulfur, 140 drug binding to, 93 Indocyanine green copper medicated, 370
Graded respon·se, defined, 327 Hybrid first-order constants, 262 as marker for deterrriination of plasma
I

progesterone releasing (progestasert),


Granulating agents. See Binders Hydrates, 29 volume, 87, 881 370
Gut motility. See Gastric emptying (See also Pseudopolymor-phism) clearance of, as indicator of hepatic Intravaginal controlled release systems,
' blood flow rate, 238 369
390 BIOPHARMACEUTICS AND PHARMACOKINETICS INDEX 391
vaginal ring, 369 binding of drugs to, 94 Maintenance of drug nonmicrosomal. See Nonmicrosomal
Intravenous bolus administration, classes of, 94 within the therapeutic range, 312 enzymes
one-compartment kinetics after, 231 Liposomes Malabsorption Metabolizing organs, 113
two-compartment kinetics after, 260 classification of, 361 influence on absorption, 58 Metallothi onin
Intravenous infusion, defined, 360 Mammary excretion, 200 heayy metals binding to, 96
constant rate, 239 production of, 362f Mammillary model, 221, 222! ~etastable polymorph(s)
Intrinsic capacity clearance, 239 Liver Manufacturing processes aqueous solubility of, 28
Intrinsic ,dissolution rate constant, de- as major drug metabolizing organ, influence on dissolution, 40 to enhance bioavailability, 298
fined, 21 113 Mathematical models, for concentration- Method of granulation, influence on dis- ·
Inulin clearance, 191 binding of drugs to, 96 response relationships, 327 solution and absorption, 40
Ion-exchange resin-drug complex blood flow to. See Hepatic blood Matrix Method of residuals
for oral controlled release, 352 flow diffusion controlled, 3 50 for calculation of K3 , 248
Ion-pair transport, 15, 16/ clearance by. See Hepatic clearance dissolution controlled, 349 Methylation reactions, 145 \

Ionic diffusion, 14 diseases. See Hepatic diseases transdennal devices, 360 Michaelis constant (Km)
Ionization and drug absorption, 33 Loading dose Maximum dosing interval, 314 estimation of, 277
Ionized drugs, absorption of, 37 ' and maintenance dose, 312 Maximum maintenance dose, 314 Michaelis-Menten equation, 275
calculation of, 3 I 2 Maximum plasma concentration. See Michaelis-Menten kinetics. (See also
Jejunum, 50, 561 attainment of desired steady-state with, Peak plasma concentration Nonlinear pharmacokinetics), 12,
'
243, 312 Maximum safe concentration (MSC), 220
KADME. See Pharmacokinetics defined, 2 I 4
defined, 312 Microcapsules, microspheres and,
Kernicterus, in neonates, I 0 I, I 05 Maximum urinary excretion rate, 288
jncorporation in controlled release for parenteral controlled release, 359
Kidney(s) time for, 288 Microcli~ate pH, 3 7
systems, 343
blood flow to, 179, 189 Mean residence time (MRT) Microconstants, 262-
infusion plus, 243
drug binding to, 96 in noncompartmental analysis, 224 Micronization
Logarithmic kinetics. See Exponential
failure. See Renal failure, 190 of controlled release dosage forms, 341
kinetics a method of enhancing bioavailability,
selective drug delivery to, 169 Mechanisms of renal clearance, 186t 297 :
Logarithmic model, for concentration-
response relationships, 327 Medium unilamellar vesicles, 361 decrease in effective surface area due
Lag time. See Time lag
Loo-Riegelman method Melts, dl~fined, 299 to, 26
Latentiated drugs. See Prodrugs
for estimation of Kl\) 267 Membran~ cell, See Cell membrane reduction of parti~le size by! 26
Latin square design
Loop of Henle. See Nephron Meni~gitis, penneability of BBB in, 86 Microsomal enzyme(s)
for bioequivalence study, 29.5t
Lubricants, influence on dissolution and Mercapturic acid fonnation important characteristics o( 1: 4
Limited solvation theory. See Interfa-
absorption, 43 conjugation \vith glutathione and, 143 Microspheres ~d microcapsules.
- cial barrier model
Lung(s) Met~bolic precursors. See Bioprecursors for parenteral controlled release.• 359
Linear kinetics, 218. (See also First-
dnig accumulation in, 96 Metabolic. - stabilization. 159 Milk, drug excretion in.~ 20 It
order kinetics/processes)
drug delivery to. See Pulmonary ad- • Metabolic S\\:itching! 160 (See also Mammary excretion)
defined, 218
ministration Metabolism. Ill Minimum effective concentration (l\1EC).
Linear model, for concentration-response
excretion by. See Pulmonary excre- • (See also Biotransfonnation) defined. 214
relationships, 327
tion nonlinearitv.. in. 274 Minimum inhibitory concentration
Lineweaver-Burk plot, for
Lung cancer defined. Ill (MIC). defined. 214
concentration-response relationships, •

Michaelis-Menten' kinetics, 278, 279 by benzo( a)pyrene, I 56! species differences in. 150

Minimum toxic concentration (MT()~
strain differences in. 151 defined. 214
P-D binding, 109
Magnetic microspheres Metabolism interactions. 209t Mixed cr)'stals. See Molecular dispersions
Lipophilicity. (See also :pH-partition
for drug targeting to tumors, 360 'Metabolites Mixed function oxidases. I t 7
hypothesis) '
Maintenance dose active. See Active metabolites Mixed-order kinetics. defined. 12~ 219
and drug absorption, 3 5
Metabolizing en~mes. 113 .
• I

tn controlled release systems, 342 (See also Nonlinear phannacokinetics)


prodrugs for enhancement of, 166
loading dose and, 312 · microsomal. See Microsomal enzymes. Molec,ular dispersions. 299
Lipoproteins, 921 •


l


392 BIOPHARMACEUTICS AND PHARMACOKINETICS INDEX 393

Molecular encapsulation N-acetyl cysteine Obese patients, dosing of drugs in, 316 of carbon-heteroatom systems, 122
with cyclodextrins, 302 to treat poisoning by drugs, 156 Obesity, 85 of carbon-nitrogen systems, 122
· Molecular inclusion complexes, 302 N-dealkylation, 122 Object drug, in drug interactions, de- of carbon-oxygen systems, 128
Molecular weight N-hydroxylation, 126 fined, 204 of carbon-sulfur ~ystems, 127
in the design of controlled delivery N-oxide formation, I 25 Ocular insert. See Ocusert of olefins,
\
120
system, 339 Nanocapsu.les. See Nanoparticles Ocusert, ocular controlled delivery sys- steps in~ of xenobiotics, 118
for passive absorption, 9 Nanoparticles tems, 368 Oxidative aromatization, 130
. for pore transport, I 0 for parenteral controlled release, 360 Odor, improvement of Oxidative deamination, 124
. . .
Increase In, Nanospheres. See Nanop~icles by prodrug design, 163 Oxidative reactions~ 116!, 117
-- after conjugation, 13 9 N.asal mucosa, drug transport across, 69 Oil suspensions o/w/o emulsions. See multiple emulsions
influence of, Nasal route of administration. See Intra- for parenteral controlled release, 359
on excretion pattern, 139, 196t nasal administration · ~ O·t1Set of action/effect \ Pain on injection, reduction of,
,influence on biliary excretion, 195 Nature of biotransformation process concentration relationships, 330 by prodrug design,. 164
Monitoring influence on biliary excretion, 196 defit:ted, 215 Parameters of plasma level data
drug therapy, defined, · 320 Neonates Onset time, defined, 215 for determining bioavailability, 286
. pharmacodynamic, 321 ·: dosing of drugs in, 3 17 Ophthalmic drug delivery systems, 368 Parameters of urinary excretion data
phannacokinetic, 321 drug binding in, 10 I Optimal dosage regimen, defined, 307 for determining bioavailability, 288
. therapeutic, 320 · Nephron Oral administration, 6. · (See also Ex- Parenteral controlled release systems, 357
Monolithic devices. See Matrix devices/ functional unit of kidneys, 178 travascular administration) infusion devices, 363
systems Niosomes typical plasma concentration-time pro- injectables, 357
Monomorphism for parenteral controlled release, 360 file after, 213 implants, 362
. · in drug metabolism, 151 Non compartmental analysis, 224 Oral controlled release systems, 347 routes for administration of, 357
· · · Mondoxygenases. See Mixed function Noncompetitive inhibition classification of, 348 Parenteral route. (See also particular
oxidases . of metabolizing enzymes, 149 , Oral mucosal delivery parenteral rout~s ), 6
.· Monotropic polymorph(s), defined, 27 Nonionic diffusion. See Passive diffu- factors to be considered in, 65 Particle size
. '
Monozygotic twins. See Identical twins •
SlOO sites for, 64 and effective surface area,
Mosteller's equation, 3I 7 Nonlinear kinetics. See Nonlinear phar- Oral osmotic pump, 353 1 influence on dissolution and ab-
Mucin
.. macokinetics Order(s) of reaction, 215 sorption, 25
endogenous ion, ion-pair transport, 15 Nonlinear pharmacokinetics, 12, 273 defined, 216 techniques for reduction to micro level,
interaction with drugs, 60 Nonlinearities, causes of, 274 Organ(s), volume of, blood flow and 27
Mucoadhesive systems Nonlinearity, tests to detect, 273 perfusion rate· to, 84t Passive diffusion/transport, 7. s~ I 0,
for oral controlled release, 356 N onmicrosomal enzymes, 114 Oros. · See Oral osmotic pump 16/, 711
Multi compartment models, 259 N onrenal clearance, 9efined, Orosomucoid. See a 1-Acid glycoprotein characteristics of, 9
Multilamell(U" vesicles, 361 • Nonrenal excretion, defined, 178 Osmogen, 353 Passive tubular reabsorption, 181
.
Multiple dose study, for determining Nonrenal routes of excretion, 194 Osmotic pressure controlled systems, Patient related variables, affecting absor-
bioavailability, 284, 289 - (See also specific routes) 353 ption and bioavailability, 18t, 50
Multiple dosing Nonstoichiometric complex, 28/ Osmotic pump( s) Peak, defined, 213
average concentration and body con- Noyes-Whitney's equation, 21, 25 for parenteral controlled release, 363 Peak plasma concentration, defined, 213
tent on, 311 modified, 21 Oxidation Peak response. See Intensity of action
drug accumulation during, 309 Nucleic acids, drug binding to, 97 of alcohol, carbonyl and acid func- Peeling technique. See Method of
maximum and minimum concentra- Nucleophiles tions, 129 residuals
tion during, 3 11 affinity for electrophiles, 1_43 of alicyclic ·carbon atoms, 122 Percutaneous absorption
time to reach steady-state during, 310 Nucleophilic addition, a mechanism of of aliphatic carbon atoms, 121 factors influencing, 67
Multiple emulsions •
'
GSH conjugation, 144 of aromatic carbon atoms, I 19 Percutaneous delivery, defined, 66
for parenteral controlled release, ~ 359 Nucleophilic .substitution, a· mechanism of benzylic, allylic C-atoms and C- Perfusion rate. (See also Blood flow)
Mutual prodrugs, 161 of GSH conjugation, 144 atoms alpha to carbonyl and imines, a rate-limiting step in distribution, 83
120.. 121 defined, 83


394 BIOPHARMACEUTICS AND PHARMACOKINETICS INDEX 395
of various tissues, 84t Pharmacodynamic characteristics, in con- Pharmacokinetics Plateau principle, 31 0
tissues classified on the basis of, 84t trolled delivery system design, 341 basic considerations~ 212 Polarity of drug
Perfusion rate-limited models. See Physi- Pharmacodynamic methods clinical. See Clinic.al pl\.armacokinet- influence on biliary excretion, 195
ologic models for evaluating bioavailability, 286 •
ICS Polymorphism, 27
Peripheral compartment acute pharmacologic response defined, 2, 212, 215 defined, 27
apparent volume of, 265 method, 290 Pharmacologic activation, 113! in acetyJation of isoniazid, 151
perfusion of, 221 ... therapeutic response method, 290 Pharmacologic inactivation, 112! variations in metabolism, 151
Permeability Pharmacodynamic interactions, 204 Phannacophore, defined, 134 Polymorph(s)
defined, 10 Pharmacodynamic models Phase I reaction(s), 114, 116t, 117 defined, 27
of tissue membranes. See Physiologic E-max model, 328 Phase II reactions, 115, 116!, 138 types of, 27, 28/
barriers to distribution through the linear model, 327 Phase III reactions, 115 Poorly perfused tissues,
biomembrane, logarithmic model, 327 Phosphatidyl choline, 117 Pore transport, I 0, 16[, 71 t
a rate-limiting step in absorption, Pharmacodynamic monitoring, 321 Physical form of the drug, change of, Post absorptive phase, 246
19 Pharmacodynamic process, of drug by prodrug design, 163
/

Pctentiation. See Synergism


Permeation enhancers therapy, 2, 3f Physicochemical factors in drug absorp- Powders, factors in absorption from, 47
in topical formulations, 67 Pharmacodynamic variability, 315 tion, 18t, 19 Precipitant drug, defined, 204
to promote crossing of BBB, 81 Pharmacodynamics, defined, 2 Physiologic availability. See Pregnancy
pH ~ Pharmacogenetics, defined, 151 danger of drugs during, 83
Bioavailability
gastrointestinal. See Gastrointestinal Pharmacokinetic analysis Physiologic barriers to distribution of influence on distribution, 85
pH ! three approaches to, 221 influence on metabolism, 152
drugs, 79
efrect on gastric emptying) 55 I
Phannacokinetic applications
-
of prodrug (See also individual barriers) Presystemic metabolism, 6, 62
influence on absorption, 32 design, 162 Physiologic fluid compartments, 87t effect on absorptionlbioavailability, 62
urine. See Urin.e pH Pharmacokinetic approach, in overcom- Physiologic models, 226/ prodrugs to prevent, · 16 7
pH-absorption curve, 37/ ing bioavailability problems, 297 Pinocytosis, 15 systems which affect, 62
pH-independent formulations Phannacokinetic interactions, 204, 205[, Primary parameters. See Pharmacoki-
pKa •
for oral controlled release, 353 . 208! netic parameters
and drug absorption. See pH-parti-
pH of microenvironment Pharmacokinetic methods tion hypothesis Priming dose. See Loading dose
alteration of, for bioavailability measurement, 285 and tubular reabsorption 182, 183!, Principle of superposition, 273
to enhance bioavailability, 298 plasma level-time studies, 286 184/ Proagents. See Prodrugs
pH-partition hypothesis, 18t, 32 . . urinary e?(cretion studies, 288 for candidates of controlled delivery Processing variables
assumptions of, 32 Pharmacokinetic models, 220 systems, 339 influence on dissolution. See Manu-
generalizations based on, •
Pharmacokinetic monitoring, 321 Plac~ntal barrier, 82/ facturing processes
for absorption of drugs, 33 Pharmacokinetic parameters. (See also Plasma clearance. See Clearance Prodrug( s),
limitations of, 36 particular parameters) Plasma concentration-time curve/profile bioprecursors, defined, 161
statement of, 32 estimation/assessment of, 213/ carrier-linked, defined, 161
pH-partition theory. See pH-partition for one-compartment open model, phases of, 214 defined, 160
hypothesis 23 1' 244' 24 7 Plasma level-time studies mixed type, 161
pH-shift, 37, 38 for two-compartment open model, for bioavailability determination, 286 properties \ of an ideal. See Ideal
Phagocytosis. 15 264. 267 . Plasma protein(s), 92 prod rug
Pharmaceutic applications of prod rug primary. defined, 234 (See also particular proteins ) Prodrug design
design, 162 secondary. 234 binding of drugs to. See Plasma pro- applications of, 162
Pharmaceutic approach. in overcoming Phannacokinetic principles · tein-drug binding limitations of, 176
bioavailability problems. 297 applications of. 306 Plasma protein-drug binding, 92 Prodrug-soft drugs, 168
Pharmaceutic equivalence. defined. 294 Pharmacokinctic process. of drug therapy. Plasma volume, 87t Product inhibition, defined, __) 49
Pharmaceutical interactions. 205 2. 3/ determination of, 87, 88t Progestasert. See Progesterone releasing
Pharmaceutic process, of drug therapy. Pharmacokinetic variability. 315 Plateau. See Steady-state IUD
2, 3f
396 . BIOPHARMACEUTICS AND PHARMACOKI1'T£TICS INDEX 397

Progesterone releasing IUD, 370 Rate of filtration, 179 Renal clearance ratio for P-D binding, 108
Pro-prodrugs. See Double prodrugs Rate of input (availability}, 231 defined, 187 Secretion
Protein. (See also particular proteins) Rate of penetrability values of, 186t active tubular. See Active tubular se-
binding to. See Protein-drug binding rate-limiting step in distribution, 76 Renal disease, dosing of drugs in, 3 19 cretion
Protein-drug binding, 91/ Rate of presentation, 231, 237 Renal dysfunction. See Renal impairment bile. See Biliary excretion
defined, 91 Rate of reabsorption, 179 Renal excretion, 178 Selective adsorptionon, on insoluble
factors affecting, 97 Rate of reaction, defined, 215 (See also Renal clearance; Urinary carriers, to enhance dissolution/
significance of, I 03 Rate of release excretion of drugs) bioavailability, 298
Pseudodistribution equilibrium' 261
. of prodrug, from site of application, factors affecting, 187 Selective uptake systems, 169
Pseudopolymorphism, defined, 29 167 •
Renal extraction ratio, 239t Self induction. See Autoinduction
Pseudopolymorphs, define·a: 29. limiting step of controlled delivery Renal failure. (See also Renal impair- Serum creatinine, for determining cre-
Pulmonary administration, 69 systems, 338[ ment; Renal disease) atinine clearance, 191
Pulmonary excretion, 198 Rate of secretion, 179 causes of, 190 Shape factor, effect of,
Reabsorption dose adjustment in, 192 on concentration-response curves,
Quantal response, 327 Renal function (RF), 190 /~329!
in renal tubules. See Tubular
Quaternary ammonium compounds calculation of dose on the basis of, ~snore et a/ equations, for GIT/plasma
reabsorption
absorption of, 15 192 concentration ratios, 33
forced diuresis and, 185
influence of urine pH and drug pK determination of, 190 Sigma minus method, to determine KE
R. T. Willia1p's chemical pathways of . equation for calculating, 192 from urinary excretion data, 256
metabolism, 114, 116t and lipid solubility on, 183 a
Real detoxication pathways, 138 Renal impairment, 190 Sigmoid-E-max model, 329
Racetnates
Real volume of distribution Renal plasma flow rate Single dose study
differences in pharmacologic response determination of, 180 for bioavailability determination, 284
due to, 326 relationship with body water, 87t
Rectal administration, 65 Repression, defined, 149 Sink condition(s)
Radicals, free. See Free radicals Resealed erythrocytes attainment of, in vitro, 22
Rate, defined, 215 Redox system
for drug delivery to brain, 172 for parenteral controlled release, 361 defined, 291
Rate constant(s), . 215 Residence time, 38 dissolution rate under, 23/
Reduction
(See also particular rate c~nstants) Response Site-specific delivery, 169
Rate-controlling step(s), in enhancement of alcohols and carbon-carbon double
bonds, 132 ali-or-none. See Quantal response in cancer therapy, 174
of duration of action, 167 concentration and relationships, 327 Size of counter ion ·
P. ate-determining step (RDS). See also of carbonyls (aldehydes and ketones),
131 factors that complicate correlation, 325 influence on solubili.ty of salts, 31
Rate-limiting steps graded. See Graded response Skin excretion, 200
defined, 17 of N-compounds, 132
of sulfur containing functional groups, Small intestine, components of, 52[
in absorption, 19 S-dealkylation, 127 Soft drug( s), defined, 113, 160
133
in availability of drug from controlled S-oxidation, 128 Solid dispersions
deli very 'systems, 338 . of toxicity,
Saliva/plasma concentration ratio, 199 to enhance bioavailability, 27, 30 I
Rate~limiting step(s)
prodrugs for, 168
Salivary excretion, 199 Solid solutions. (See also Molecular dis-
in distribution, 76 Reductive dehalogenation, 133
Salivary cycling, 199/ persions)
in hepatic metabolism, 238 Reductive reactions, 116t, 130
Salt form(s) defined, 298
in oxidation of xenobiotics, 118 miscellaneous, 13 3
factor influencing solubility, 18t, 29 to enhance bioavailability, 298
Rate of conversion Regimen(s). See Dosage regimen(s)
use of, to enhance bioavailability, 297 Solubility
of prodrug, into active drug, 167/ Relative bioavailability, defined. (See
Salt formation, in situ, 31 absolute, defined, 19
Rate of creatinine excretion, 191 also Bioavailability), 284
Saturation kinetics. See Nonlinear phar- and dissolution rate, 19
Rate of elimination, 23 7 · Renal clearance
macokinetics enhancement -of, by prodrug de-
Rate of excretion method defined, 186 '
Scatchard plot sign, 164
to determine K£, 25 5 factors affecting, 187
for concentration-response relation- Solute-solvent complexation, to enhance
Rate of exit, 237 perfusion rate-limited, 189
ship, 330/ solubility/bioavailability, 298
Rate of extraction, 237 values, range of, J86t

BIOPHARMACEUTICS AND PHARMACOKINETICS 399
398 INDEX
Subcutaneous tissue Targeting drug. See Site-specific ~rug Tissue-drug binding
Solution(s) fl. cing bioavailability ideal location for implants, 363
factors in uen delivery I examples of, 96
Sublingual administration, 64 Taste, improvement of, Tissue localization of drugs. See Tissue
from, 47 .
solid. See Solid soluttonss (See also Buccal administration) by prodrug design, 162 binding of drugs
defined. ( ee also Sulfation, 141. Temporal factors · Tissue permeability -·
Solvates, hism) 29 (See also Conjugation with sulfate •
PseudopolymofP ' affecting biotransformation, 154 rate-limiting step in distribution, 77
osition moieties) . Teratogenesis Tolerance
l t
So ven ed P bility/bioavailability, steps in, 141 caused by rea9~ive metabolites, 156t concentration-response correlation and,
to enhance solu
Sulfoconjugates, tissue reactive, 142 Termination of drug action · . 326
.298. transport mech antsms.
. see Sulfur glucuronides, 141 1
processes in, 11 1 . Topical administration, 66
Spectahzed . d transport Summation. See Addition interactions
Carrier-medtate Therapeutic concentration range, 326 Topical route, 6
h 28 Surface area (See also· Therapeutic range) Total body clearance. See Clearance
Stable polymorp s, .
absolute. See Absolute surface area defined, 212 Total body water (TBW)
b 1·rty
1 roblems
Sta ' P · vailability, 39 body, for some drugs, 3 26t composition of, 87t
Influence on btoa
Stagnant layer, defined, 20 equation for calculating, 3 17 Therapeutic efficacy . determination of, 88t
effective. See Effective surface area
(see a lso Diffusion. layer) approaches to improve, 159 Total systemic clearance. (See also Clear-
Surface renewal theory. See Danckwert's ance) ' --
Statistical in_terpretatl~:t Therapeutic· endpoint, 320
model
of bioequtvalence a, 296 Therapeutic equivalence, defined, 294 defined, 185
. . I ments theory Surfactants Therapeutic index (TI) equations for calculating, 23 5
5t~ttsttca mo ntal analysis, 224 influence on absorption, 44
tn noncompartme . . a characteristic in the design of con- Toxic endpoint, 321 <:!

to determine in vitro-zn vzvo corre1a- Suspending agents trolled delivery systems, 341 Toxicologicf acti~ati~~1 112t, 1?4 ,..
influence on absorption, 43 defined, 212 Transcortin. See Corticosteroid binding
tion, 293
Suspension(s) dosing frequency and, 3 13 globulin
steady-state . . ~ . 240 factors in bioavailability from, 47
t nt rate 1. v. tn1uston, Therapeutic monitoring, 320 Transcytosis, defined, 16
~::v~~~~~ity studies at, 287 solid. See Solid suspensions Therapeuti~ objective, deWted,. 306 Transdermal drug delivery systems, 365
Sustained release products Therapeutic occupancy time, defined, · advantages of, 365
defined, 240
bioavailability from, 49 371 types of, 366
desired 342
.' f controlled release sys- Sustained release systems Therapeutic process, of ·drug ther~py, mixed monolithic-reservoir devices,
destgn o .
th e basts of, 342 distinction from controlled release sys- . 2, 3[ 368
terns on · h tems, 336/
. f dosage regtmen on t e Therapeutic range monolithic devices, 366
destgn o
Synergism, defined, 205
basis of, 31 4. d defined, 215 rate-limiting steps in drug release
242t Systemic availability. See Availability; ·in the design of controlled delivery from, 366
percent of, attatne ' .
. for calculattng, 242 Bioavailability systems, 341 reservoir devices, 367
equatton t1
. . . complexes, 28~ Systemic clearance. See Clearance maintenance of drug within the, 313 Transfer constants. See Microconstants
StOtChJometrtC . fl 50 51/ total . See Clearance '
ctomach, drug absorption rom, d~ Therapeutic window. See Therapeutic r ·ransferrin, binding to, 95
:J d.t.10ns influence on tsso- range .-. Transmembrane rate-limited. See Per-
storage con 1 ' . Tablet(s)
. and absorption, 50
lutton Therapeutics, optimization of, meability rate-limited ·
coated. See Coated tablets
approaches for, 159 Transport moiety. See Carrier
Stratum. corneum · ·
try of xenobtottcs,
66 enteric coated. See coated tablets

Time lag, defined, 250 Trapezoidal rule
barrter to en f factors in bioavailability from, 48/,
. . h ique See Method o re- Time of peak concentration (tmax), de- for calculating AUC and AUMC. 27~
Strtpptng tee n · 49
siduals t'ton fined, 214 Tubular reabsorption, 18 1
· · tra Tamoxifen binding site, 93, 94f ·
ubcutaneous admtnts . Tissue(s), perfusion rate of, 84t active. See Active tubular t'L·ahsotl''"'''
S hance absorption after, Target concentration strategy, defined,
ways to en . d
68 Tissue binding of drugs, 96 · passive. See l)a s:~ t v \· \ 1ul11l '
oute for contro e 11 re- 321
Tissue compartment. See Peripheral com- reabsorption
subcutaneous ~ . ' 357 363 Targeted drug delivery system . ..
lease med!catton, ' partrnent . Tubular: seer ·1 in n t\;•. '
ideal. See Ideal targeted drug deliv- '
S ubcutaneous site . ery system
Tissue distribution half-life. See Distri-

ser l t•l "l t'
. ease absorption from, 68 bution half-life
ways to tncr
oC
O'
400 . BIOPHARMACEUTICS AND PHARMACOKINETICS (

Uphill transport. See Active transport
,)
Volume of central compartment, 265
Uremia, l9.Ql,o
·~ .
· Volume of distribution. (See also Ap-
Urinary exeretion. (See Renal excretion) parent volume of distribution), 86
principal processes of, 178 real. See Real volume of distribution
Urinary excretion data, 253 Volume of peripheral compartment, 265
advantages of, 253
criteria for obtaining valid, 254 Wagner-Nelson method
determination of Ka from, 259
1 for estimation of Ka 250
'
determination of KE from, 255 Warfarin 0

rate of excretion method, 255 and azapropazone binding site, 93


sigm.a-minus method, 256 Water, total body. See Total body water
Urinary excretion studies, assessment of Water compartments. See Body water,

bioavailability from, 288 . ~. compartments of
Urine flow rate, Water flux, 10
influence on tubular reabsorption, 184 Water loading, to promote diuresis, 2,54
l!rine pH Weight. See Body weight
influence on tubular reabsorption, 182 Wet granulation
Urine pH control tablet manufactured by,
to treat toxicity due to overdose, 185 limitations of, 40
Urine/plasma concentration ratio Woolf-Augustinsson-Hofstee plot, 278
equations for calculating, 182
Xenobiotics
Vaginal administration, 70 defined, 111
Vaginal ring, 369/ enzymes that biotransform. See Me-
Vapor pressure powered pump, 364 tabolizing enzymes
Variability site for metabolism of. See Metabo-
in drug response, lizing organs
sources of, 3 15
Zero-moment curve, 224
intersubject. See Intersubject variability
Zero-order absorption model, 246
pharmacodynamic.
Zero-order half-life, 217
See Pharmacodynamic variability
Zero-order kinetics (See also Constant
pharmacokinetic. See Pharmacokinetic
rate proce~ses/kinetics), f 16
·• · variability
Zero-order release
Vascular fluid volume, 87t
from controlled release; Iormulation,
Vesicular transport. See Endocytosis
344
Virtual membrane pH, influence on ion-
with a rapid release component,
ization and absorption, 37
345
Viscosity imparters. See Suspending
agents ,

. "'" ·"' .

·~ 0

··. l
...
, . ~ .·· ~
~

, -
-
..

0 Essentlats of Physical Pharmact~utlcs -CVS Subrahmanyam
0 Textbook of Physical Pharmaceutics -cvs Subrahmanvam

I Cl Blopharmaceutlcs and Pharmacokinetics-A Treatise


-OM Brahma·nkar and SB Jalswal
CJ A T /B of Pharmaceutical Formulation -BM Mlthal
CJ Controlled Drug Delivery-ConceptS and Advances ·Vya5 and Oar

0 Pharmaceutical Engineering -cvs Subrahmanyam et.al.
'
0 Pharmaceutical Microbiology -NK Jain
(
0 Pharmaceutical Biotechnology -ss Korl and MA Halkat
0 A Handbook of Cosmetlca -BM Mlthal 1nd RN Saha
o Dispensing Pharmacy - RM Mehta
0 A T•wtbook ·of Professional Pharmacy -NK Jain and SN Sharma
0 Lab. ~e~nual of Physical Pharmacy·! ·Subrahmanyam and vasantharaJU
..
o Lab Manual of Physical Pharmaceutics -Subrahmany•m and SettY
0 Pharmaceutics·! -RM Mehta
0 Pharmaceutlcs•ll ..RM Mehta
o Pharmaceutical Jndustrl•t Manag•ment -RM Mehta
o Drug Store and auslnesa Management -RM Mehta
o A Textbook of Forensic Pharmacy -BM Mlthal
0 A Textbook of Forensic Pharmacy -NK Jain
0 Pharmaceutical lurlspudence -NK 'lain
0 Medicinal ~nd Pharmaceutical Chemistry -H Slngh·and VK Kapoor
0 Organic Pharmaceutical Chemistry (PC-II) -H Singh and VK Kapoor
0 Inorganic ~harmaceutlcal Chemistry (PC·I) -P Gundu Rao
0 Pharmaceutical Chemistry·! -NC Chaudhry and NK Gurbanl
0 Practical Pharmaceutical Chemistry (PC-I • II) -Slngh and KapOOt
Cl Pharmaceutical Analysis·! -PC KamboJ
0 Laboratory Handbook for Instrumental Drug Analysis -BG NagaVI

0 Biochemistry -P Gundu Rao


0 Biochemistry and Clinical Pathology -PC Oandlya and PK Sharma
a Ph~rmaceutlcal Biochemistry -PK Sharma and PC Oandlya
0 MCQs In Medical Biochemistry -PK Sharma

0 Introduction to Pharmacology -PC Dandtya and SK Kulk~rnt


0 Handbook of Experimental Pharmacology -SK Kulkarni
0 Textbook of Hospital and Clinical Pharmacy -Oandlya and Mathur
· 0 Health Education and Community Pharmacy -Dandtya, Zafer 8t Zafer
0 Elementary Human Anatomy and Physiology
-Reghunandanan, Goplnathan and Reghunandanan
0 Textbook of Pharmacognosy -55 Handa and VK Kapoor
0 Practical Pharmacognosy -CK Kokate

a History of Pharmacy In India, Vol-1: Pharmacopoeias and Formul•rl••


-Harklshan Stngh Vol-2: Pharmaceutical Education ;-
Vol,-3: Pharmacy Practice :1
0 A Profuaor Rememben ·PC Oandlya

AP-53A P1tampura 1
DELHI 110088
Phone : 27317755
• e·mail : ve11abh@vsot .com
' -- .. .,

You might also like