Download as doc, pdf, or txt
Download as doc, pdf, or txt
You are on page 1of 8

Dr.

Stephanie Fay Cagayan


1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development

Outline: • Global economy


• Interdisciplinary
I. Introduction

II. Drug Discovery

III. Pre-clinical Research and Development DRUG DISCOVERY


IV. IND
Steps in Developing a New Drug
V. Clinical Trials
1. Preclinical

Transers’ Note: This six-page trans is equivalent to five points in


the exam. Haha. Focus on phases of clinical trials and rational
• Discovery or synthesis of a new drug
drug designs. molecule and correlating it with a biological
Dr. Cagayan: “yung table (phases of clinical trials) na lang ipaaral target
mo sa kanila para hindi toxic.”
• In vitro and animal studies to assess the
safety and efficacy of drugs (as required
by law)
INTRODUCTION
Objectives:
• Describe the drug discovery process
• Define key players in drug development 2. Clinical
• Discuss cost of drug development

Drug discovery is the process of by which drugs are


discovered and/or designed • Human testing

• Discovery includes: Concept, mechanism, assay, • Safety monitoring after approval for
screening, hit identification, lead demonstration, general use
lead optimization
• Discovery also includes in vivo proof of concept in
Drug Discovery Overview
animals and concomitant demonstration of a
therapeutic index
- A drug discovery effort addresses a biological
Drug Development is the process taking a new chemical target that has been shown to play a role in the
lead through the stages necessary to allow it to be tested development of the disease or starts from a
in human clinical trials. molecule with interesting biological activities

• Development begins when the decision is made to 1. Identification of a new potential drug target
put a molecule into phase I clinical trials 2. Rational drug design
3. Modification of an existing drug/potential drug
Aims of Drug Development 4. Screening of chemical/macromolecule/ organic
chemical libraries
5. Biotechnical approaches using the human
• Provide information on the optimal use of a new
genome.
drug in treatment or prevention of a disease
6. Combinations of known drugs to obtain added or
• To document the quality of the pharmaceutical synergistic effects.
product
• Provide for an economically sound investment

Drug Development and Regulation

• The cost estimate to bring a single drug to market


range from $150 million to $900 million for
research & development; this does not include the
costs of marketing.
• Only 3 of 10 marketed drugs return their R&D
costs.
• Literally hundreds of thousands of both synthetic
and naturally occurring compounds are tested for
each successful drug discovery.
• Compounds which show potential for therapeutic
benefit usually undergo further modification to
improve efficacy, potency and/or safety prior to
formal evaluation.

Profile of Today’s Pharmaceutical Business


Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 1 of 6
Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development
• begins with identifying the function of a
possible therapeutic target and its role in
disease

Note: The main difference of the between the two


paradigms lies in the time at which the drug
target is actually identified.

Target Identification

1. Disease Mechanism

• understanding the disease mechanism


directs research and formulates a possible
treatment to slow or reverse the disease
process
• predicts a change of the disease pattern
and its implications

2. Disease Genes and Functional Genomics

• specific gene defects or mutations that


bring about a hereditary disorder have
been identified for a number of diseases
Figure 1 | Drug Discovery
• functional genomics aims to determine
disease mechanisms and to identify
Types of Targets disease genes and disease markers
• guide the understanding of signal
transduction pathways that either lead to
disease or indicate therapeutic strategies
for the development of novel therapeutics.

3. Target Type and “Druggability”

• targets can be receptors, proteins,


enzymes, DNA, RNA or Ribosomal targets
• “druggability" of a given target is defined
either by how well a therapeutic, such as
small molecule drugs or antibodies, can
access the target, or by the efficacy a
Figure 2 | Therapeutic target classes. therapeutic can actually

Target Discovery Lead Discovery

1. Physiology-based Drug Discovery • identification of a compound that triggers specific


biological actions
• follows physiological readouts • approaches:

• E.g. the amelioration of a disease 1. Serendipity


phenotype in an animal model or cell-
based assay. - “Chance favors the prepared mind”

- Compounds are screened and - 1928 Fleming studied Staph, but


profiled based on this readout. contamination of plates with
airborne mold. Noticed bacteria
- A purely physiology-based were lysed in the area of mold. A
approach would initially forgo mold product inhibited the growth
target identification/validation and of bacteria: the antibiotic penicillin
instead jump right into screening.

- Identification of the drug target


and the mechanism of action 2. Screening of biologic activity of a wide
would follow in later stages of the array of natural products or chemical
process by deduction based on entities
the specific pharmacological
properties of lead compounds. - Testing a random and large
number of different molecules for
biological activity reveals leads.
Innovations have led to the
2. Target-based Drug Discovery automation of synthesis
Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 2 of 6
Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development
(combinatorial synthesis) and groups in the same 3D locations.
testing (high-throughput (“Mimic” the active groups)
screening). - Advantage: Don’t need to know the
biological target structure
- Example: Prontosil is derived from
a dye that exhibited antibacterial Receptor-based Drug Design
properties.
- Examine the 3D structure of
the biological target
- Look for specific chemical
3. Chemical modification of known molecules groups that could be part of an
attractive interaction between the
- Traditional method: an analog of a target protein and the drug.
known, active compound is
synthesized with a minor - Design a drug candidate that
modification, that will lead to will have multiple sites of
improved biological activity. complementary interactions with the
biological target.
- Advantage and Limitation: you
end up with something very similar - Advantage: Visualization
to what you start with allows direct design of molecules

Note: Typical projects are not purely receptor-


based or pharmacophore-based; they use
4. Rational drug design combination of information, hopefully
synergistically.
- Establishes structural relationships
between the biological properties Example of Rational Drug Design: Cimetadine
and the molecular structures. (Tegamet)
- Basis of drug-target interactions is
molecular recognition: the specific - Starts with a validated
attractions between the chemical biological target and ends up with a
groups of a biological target (large drug that optimally interacts with the
protein, usually) and a drug (small target and triggers the desired
molecule, usually). biological action.
- New molecules that can optimally - Problem: histamine triggers
interact with a biological target can release of stomach acid. Want a
be designed to block or trigger a histamine antagonist to prevent
specific biological activity. stomach acid release by histamine
= VALIDATED BIOLOGICAL
- Begins with the design of
TARGET.
compounds that conform to
- Histamine analogs were
specific requirements. The
synthesized with systematically
molecules are synthesized, tested.
varied structures (chemical
Then the molecule is redesigned,
modification), and SCREENED. N-
synthesized, tested.
guanyl-histamine showed some
antagonist properties = LEAD
compound.
- Two Sources:
a. Chemical modifications were
 3D structure of biological made of the lead = LEAD
target (receptor-based OPTIMIZATION.
drug design)
b. More potent and orally active,
but thiourea found to be toxic in
 Structure(s) of known active clinical trials.
small molecules
(pharmacophore-based
c. Replacement of the group led
drug design) to an effective and well-tolerated
product.
Pharmacophore-based Drug Design d. Eventually replaced by Zantac
with an improved safety profile.
- Examine features of inactive small
molecules (ligands) and the
features of active small molecules Lead optimization
(ligands).
- Generate a hypothesis about what • properties of the lead are tested with biological
chemical groups on the ligand are assays; new molecules are designed and
necessary for biological function; synthesized to obtain the desired properties
what chemical groups suppress
biological function.
- Generate new ligands which have PRE-CLINICAL RESEARCH AND DEVELOPMENT
the same necessary chemical
Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 3 of 6
Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development
• The goal of preclinical studies includes identifying
potential human toxicities and designing INVESTIGATIONAL EXEMPTION FOR A NEW DRUG (IND)
mechanisms to monitor for these toxicities in
clinical trials.
• The discovery of a significant toxicity may not
preclude further development but indicate the
need for some drug modification.
• At the preclinical stage, the FDA will generally
ask, at a minimum, that sponsors:
1. develop a pharmacological profile of the
drug;
2. determine the acute toxicity of the drug
in at least two species of animals, and
3. conduct short-term toxicity studies
ranging from two weeks to three
months, depending on the proposed
duration of use of the substance in the
proposed clinical studies.
• Specific testing is determined by the proposed
therapeutic use of an agent.

Figure 3 | IND Application

• Once a drug has been judged


ready to be studied in humans
a Notice of Claimed
Investigational Exemption for a
New Drug (IND) must be filed
with the FDA; this application
must include an extensive
profile of drug including the
mechanism of action,
formulation & composition of
the drug, the proposed clinical
plans and protocols, the names
and credentials of physicians
who will conduct the trials.

• All this data is made available


Table 1 | Safety Tests to investigators and their
institutional review boards.

Limitations of Preclinical Testing • The investigational new drug


(IND) application is the result of
1. Toxicity testing is time consuming and a successful preclinical
expensive. It may require 2-6 years to acquire development program. The IND
this data prior to human testing. is also the vehicle through
2. Large numbers of animal subjects are which a sponsor advances to
needed; Molecular, cell and tissue culture the next stage of drug
methods may be substituted but their predictive development known as clinical
value is severely limited. trials (human trials)
3. The extrapolation of animal data to humans may
not be accurate for all toxicities (antiemetics • Generally, this includes data
tested in a rat, cinnamyl anthranilate). and information in three broad
areas:
4. Rare adverse effects are unlikely to be detected
(AA with chloramphenicol). 1. Animal Pharmacology and Toxicology
Studies

Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 4 of 6


Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development
2. Manufacturing Information
Phase 4 Clinical Trials
3. Clinical Protocols and Investigator
Information • Is the treatment safe over time?
• Phase 4 trials are less common and serve to
answer questions after the FDA has already
CLINICAL TRIALS approved a drug for general use. These can
address questions such as long-term safety of a
Human Trials drug, or other circumstances in which the drug
may be helpful.
• Less than 1/3rd of drugs tested in clinical trials
reach the market place.
• Human clinical trials are replete with oversight NEW DRUG APPLICATION (NDA)
from a number of institutions.
• Federal law requires human drug trials are
conducted in accordance with strict rules.
• Sophisticated, elegant trial design is required to
successfully identify appropriate drugs

Confounding Factors in Clinical Trials

1. The variable natural history of most diseases


2. Most diseases have a variable clinical course,
exacerbations & remissions are common and
some get better with no intervention. Study
design must account for the natural history of the
disease. (cross over design).
3. The presence of other diseases and risk factors
4. Subject and observer bias
5. Study design must account for the placebo effect;
a review of any drug trial reveals the remarkable
power of a placebo to produce a response (both
beneficial & detrimental). Single-blind and
double-blind studies seek to remove this bias.

Phase 1 Clincal Trials

• Is the treatment safe?


• After an experimental drug or treatment has been
tested in the lab and/or on animals, it enters a
phase 1 trial. These trials involve a small number
of patients to test safety in humans and
determine the correct dose of a drug. These trials
also help determine the best way to give the
drug, whether oral or intravenously.

Phase 2 Clinical Trials


Figure 4 | NDA Chart
• Does the treatment work?
• In the primary review process, reviewers attempt
• After determining that a treatment is reasonably to confirm and validate the sponsor's conclusion
safe in people, it enters phase 2 trials. These are that a drug is safe and effective for its proposed
done to test for effectiveness – does the use
treatment work? Since a larger number of people • NDAs can consist of as many as 15 different
are studied, further information is gained on sections:
safety during phase 2 trials. - Index
- Summary
Phase 3 Clinical Trials - Chemistry, Manufacturing, and Control
(CMC)
• Does the new treatment work better than the - Samples, Methods Validation Package,
standard treatment? and Labeling
• Phase 3 trials test the new drug or treatment on - Nonclinical Pharmacology and
hundreds or thousands of individuals. These Toxicology
studies are often “double-blind” trials, which • Human Pharmacokinetics and Bioavailability
mean that neither the patient nor the investigator - Microbiology (for anti-microbial
knows which treatment is being used. They are drugs only)
designed to answer the question of whether or - Clinical Data
not the new treatment works better, or has fewer - Safety Update Report (typically
side effects, than the standard treatment. submitted 120 days after the NDA's
submission)
- Statistical
Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 5 of 6
Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development
- Case Report Tabulations • Given the massive cost for drug development and
- Case Report Forms approval drugs the development of drugs for rare
- Patent Information disease would be cost prohibitive (they are drug
- Patent Certification companies).
- Other Information • Frequently these diseases involve pediatric age
patients in whom testing justifiably is severely
restricted.
GUIDELINES OF DRUG DEVELOPMENT • Often promising treatments would languish due to
the lack of potential return, the Orphan Drug Act
Good Laboratory Practice (GLP) of 1983 provided incentives for the investigation
and development of drugs for rare diseases
• In vitro screening ( <200,000 patients in the US or if >200,000 the
• Animal studies expected return will not meet costs).
• The purpose of these Principles of Good • Since 1983 the FDA has approved 268 orphan
Laboratory Practice is to promote the drugs for 82 rare diseases.
development of quality test data
• Comparable quality of test data forms
the basis for the mutual acceptance of ADVERSE DRUG REACTIONS
test data among countries

Good Clinical Practice (GCP)


• Human clinical trials
• An international ethical and scientific quality
standard for designing, conducting, recording,
and reporting trials that involve the participation
of human subjects

• To provides public assurance that the rights,


safety, and wellbeing of trial subjects are
protected and that the clinical trial data are
credible

• To provide a unified standard for the European


Union (EU), Japan, and the United States to
facilitate the mutual acceptance of clinical data by
the regulatory authorities in these jurisdictions. • An ADR is a reaction to a drug that is harmful or
The guidance was developed with consideration an unintended response.
of the current good clinical practices of the • ADRs are claimed to be the 4th leading cause of
European Union, Japan, and the United States, death in the US (> AIDS, accidents & automobile
as well as those of Australia, Canada, the Nordic deaths).
countries, and the World Health Organization • During the IND and clinical phase (1-3) all
(WHO) adverse events related to use of the drug must be
reported to allow continued testing. This process
continues after market approval by the FDA but
Good Marketing Practice reporting is on a voluntary basis; drug recalls due
to this phase IV reporting are not uncommon
• a system for ensuring that products are (phen-phen, COX-2inhibitor Vioxx,).
consistently produced and controlled according to
quality standards.

• Designed to minimize the risks involved in any


pharmaceutical production that cannot be
eliminated through testing the final product. The
main risks are:

- unexpected contamination of products,


causing damage to health or even
death;

- incorrect labels on containers, which


could mean that patients receive the
wrong medicine;

- insufficient or too much active


ingredient, resulting in ineffective
treatment or adverse effects.

ORPHAN DRUGS AND RARE DISEASES

Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 6 of 6


Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development

Everly: thanks to Ed and Julie for this trans. To my researchmates, don’t ever let
go of me, please! Hello to bea and al, my beks girls anne and april. BILI KAYO NG
RSO SCRUBS AND PANTS. AVAILABLE IN GRAY, PINK AND APPLE GREEN.
Goodluck and Godbless 2014 in this yearead.
Edge: Lunchmates hello! Hi kay Jean at Default! Thanks sa pagpapainternet sakin.
Haha. Tags! Badminton ulit! Jesh, miss earth! Marcus, wag naiinggit. Jason,
musta? Haha. Ado, Aidz, Pats, train na tayo! Happy birthday Evs! Belated Sharms!
Julie: Ask me anything (about the trans). 09158507391 I love 35G Psych
Ward+Motch! Hello Psych! Hello S.Karla, S.Carla, S.Coy, S.Shayne, S.Maetrix,
S.Janna, S.Mindy and B.Ace, B.Apol, B.Karl, B.Monchi, B.Mark, B.Pangs,
B.Tonicci! Edge, magtrabaho ka naman h*y*p ka. Hi Lance! Bryan pengeng pasta!
Ikaw din, Austeon.Anna alam mo na. Pors I miss you. Hi Pito! Jean, Leeann, no.4!
Marvie JTama na gailt na si Edge.
Ado: MSSR Cereals Party. Wed, 5pm, MSU 2F. See you there!

Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 7 of 6


Dr. Stephanie Fay Cagayan
1
THER 201
Pharmacologic Basis of Therapeutics
Lecture 4: Drug Discovery and Development

Everly, Edge, Julie, Ado Mon, July 5, 2010 Page 8 of 6

You might also like