Drug Discovery and Development: Medicinal Chemistry
Drug Discovery and Development: Medicinal Chemistry
Drug Discovery and Development: Medicinal Chemistry
Drug discovery and development process aims to make available medications that are safe and
effective in improving the length and quality of life and relieving pain and suffering. However, the
process is very complex, time consuming, and resource intensive, requiring multi-disciplinary expertise
and innovative approaches.
Recent estimates suggest that it takes up to 13.5 years and 1.8 billion U.S. dollars to bring a new drug
to the market. There is a growing urgency to identify and develop more effective, efficient, and
expedient ways to bring safe and effective products to the market. The drug discovery and
developmental process relies on utilizing relevant and robust tools, methods, and models that are
predictive of clinical effects in terms of diagnosis, prevention, therapy, and prognosis. There is a
growing emphasis on translational research, a bidirectional bench to the bedside approach. The all-
important predictivity depends on having robust, relevant, validated and qualified biomarkers for
physiological and pathological effects of interest.
Medicinal chemistry and pharmaceutics play a crucial role from the beginning of the drug discovery
and development process, involving chemical synthesis (including compliance with current Good
Manufacturing Practice, cGMP), characterization, purification, chemical alteration, stability
determination, and formulation of the drug candidate
Medicinal Chemistry
- relates to the design and production of compounds that can be used in medicine for the prevention,
treatment or cure of human and animal diseases.
2. An optimization step: that deals mainly with the synthetic modification of the lead structure in order
to improve potency, selectivity and lessen toxicity. Its characteristics are the establishment and
analysis of structure' -activity relationships (SARs). 3. A development step consisting of the
optimization of the synthetic route for bulk production. Modification of the pharmacokinetic and
pharmaceutical properties of the active substance to render it suitable for clinical use.
Disease has been recognized as an enemy of humankind since civilization began, and plagues of
infectious diseases arrived as soon as humans began to congregate in settlements about 5000 years
ago. Early writings on papyrus and clay tablets describe many kinds of disease, and list a wide variety of
herbal and other remedies used to treat them. The earliest such document, the famous Ebers papyrus,
dating from around issoBC, describes more than 80o such remedies. Disease was in those times
regarded as an affliction sent by the gods; consequently, the remedies were aimed partly at
neutralizing or purging the affliction, and partly at appeasing the deities. Despite its essentially theistic
basis, early medicine nevertheless discovered, through empiricism and common sense, many plant
extracts whose pharmacological properties we recognize and still use today; their active principles
include opium alkaloids, ephedrine, emetine, cannabis, senna and many others
The present medicinal system is dominated by the Allopathy or western medicine which is prominently
taught and practiced in most of the countries world wide. This system is still evolving and during last
few decades focus was based on chemical origin of most of the medicines. Thus majority of drugs in
current practice are from synthetic origin.
Ayurveda, one of the oldest systems used by mankind for well being (Sharma 1995), originated in
ancient India many thousand years ago (about 4500 BC as agreed by most scientists).
Ayurveda literally means "science of life" in Sanskrit (Ayur: Life; Veda: Science). It is not only a medical
system but a way of life. Ayurveda aims at a holistic management of health and diseases. It is widely
practiced in the Indian subcontinent and is also one of the official systems of medicine in India. Its
concepts and approaches are considered to have been perfected between z5oo-5oo BC.
Charak Samhita and Sushrut Samhita (loo-soo BC) are the two main Ayurvedic classics, wherein more
than no plants along with their classification, pharmacological and therapeutic properties have been
described.
Damstra Chikitsa (Toxicology) Damstra Chikitsa or Agada Tantra is the branch of toxicology in
Ayurveda that deals with treatments and prevention of toxins in the body. It dealt with poisoning from
animals, plants, vegetables, or metals or man-made poison. But more importantly, this branch of
Ayurveda also considered air and water pollution as a form of poisoning that needed to be purified for
health and well-being of man, as contamination would often lead to mass scale epidemics. Ayurveda
accords as much importance to purity of air, water, earth, and space as much as to the food,
environment and quality of life.
In beginning, guidelines were restricted to local needs. In clinical trilas, critical efficacy endpoints are
identifies in advance & sample sizes are estimated for assessment of effectiveness.
INTRODUCTION
In biology, the term "in situ" means that the examination and observation of a rare occurrence take
place where it occurs. Subjects are examined on position and are not moved to another or channel. An
example is an observation of dolphins at sea. they observed where they are found and are not moved
to an aquarium or other container which is more convenient. In cell science, in situ can mean
something
in between in vivo and in vitro.
IN VIVO
"in vivo" is a latin word which means "within the living."
it is the experiment or observations done on the living tissue of the whole living organism in a
controlled environment.
in vivo experiments are done in the organism's natural environment or in the organism itself.
precise cellular conditions are presented in these studies.
one example is a clinical testing or a clinical trial which can be controlled testing of a new drug
or device on human subjects. the subjects are given the drugs and are observed for a certain
period of time
another is animal testing which is an experiment that is done in animals usually rats, birds, frogs,
and other animals where the drugs are directly injected into the body. so in vivo experimetnts,
conditions are not manipualted or controlled.
it is found to be more suited on experiments done on organisms that are alive
in medicine, clinical trials and animal testing are performed in vivo to analyze the overall effects of
the experiments.
in vivo experiments, living cells, or animal models are used. in vivo studies are crucial for the
development of medical devices, surgical instruments, rocedures and novel therapeutics.
IN VITRO
"in vivo" is a latin word that means "within the glass." therefore the studies which are done
outside the living organism,inside glass (test tubes or petrishishes) are known as in vitro studies.
it is the experiment or observations done on the tissue outside of the living organism in a
controlled environment, usually using petri dishes and test tubes.
the term in vitro is used in cell biology to explain the techniques which are performed on a
controlled environment outside a living cell or organism
most experiments in cellular biology are done through in vitro studies and are not conducted in
the organism's natural environment. this results in the limited success of the experiments in
simulating the actual conditions inside an organism and make its outcome less precise.
compared to in vivo experiments, it is less epensive and provides quicker results.
most of the cellular, biochemical experiments are carried out in vitro in the labs to test.
in vitro methods are widely used in the pharmaceutical industry to produce large scale
pharmaceuticals using microorganisms due to its ease of production and economic benefits.
Weil (1972) published the following set of guidelines, which reflected a consensus among toxicologists.
These should be considered before initiating a toxicity test:
1. Use, wherever practical or possible, one or more species that biologically handle the material
qualitatively and/or quantitatively as similarly as possible to man. For this, metabolism, absorption,
excretion, storage and other physiological effects might be considered.
2. Where practical, use several dose levels on the principle that all types of toxicologic and
pharmacologic actions in man and animals are dose-related. The only exception to this should be the
use of a single, maximum dosage level if the material is relatively nontoxic.
3. Effects produced at higher dose levels (within the practical limits discussed in 2) are useful for
delineating the mechanism of action, but for any material effect, some dose level exists for man or
animal below which this adverse effect will not appear.
4. Statistical tests for significance are valid only on the experimental units that have been
mathematically randomized among the dosed and concurrent control groups.
6. Effects obtained by one route of administration to test animals are not a priori applicable to effects
by another route of administration to man. The routes chosen for administration to test animals should,
therefore, be the same as those to which man will be exposed. Thus, for example, food additives for
man should be tested by admixture of the material in the diet of animals.
The emphasis of the carcinogenicity study is the detection of tumors in animals. For these studies,
both concurrent and historical control data are used to evaluate the relevance of tumors.
Historical control data should be derived from studies in the same species and strain and,
preferably, in the same laboratory as used in the study under consideration.
The EPA prefers that the rat and the rabbit be used in these studies; however, hamster and mouse
are also acceptable. Doses should be administered over the period of major organogenesis (major
visceral and skeletal formation) in the fetus. The maternal animals only are dosed in this study and
only for specified periods. When day 0 is the day that evidence of mating was observed, the rat
and mouse are dosed on days 6 through 15; the rabbit, days 6 through 18; and the hamster, days 6
through 14.
Drug Discovery and
Development as a
Process
ABELLA
ABING
ALIMODEN
AMAN
ARSENAL
Table
Table of
of Content
Content
Overview & Definition
Phases Stages
Step 1: Discovery & Development
Step 2: Preclinical Research
Step 3: Clinical Development
Step 4: FDA Review
Step 5: Post-market Monitoring
Other Relevant Drug Development
Concepts
Overview & Definition
The complexity in drug development has
increased manifolds over the past 40 years,
requiring preclinical testing, investigational new
drug (IND) applications, and completed clinical
testing before marketing approval from the FDA.
Generally, new drug applications (NDAs) or
biologics license applications (BLA) are
reviewed comprehensively before approval, and
then drug performance is resubmitted to
regulatory agencies for post-marketing studies.
The overarching goal is to bring more efficient
and safer treatments to the patients as quickly
as possible after a thorough medical
evaluation.
DRUG DISCOVERY & DEVELOPMENT
Drug discovery is a process which aims at identifying a compound therapeutically useful in
curing and treating disease. This process involves the identification of candidates, synthesis,
characterization, validation, optimization, screening and assays for therapeutic efficacy. Once a
compound has shown its significance in these investigations, it will initiate the process of drug
development earlier to clinical trials.
New drug development process must continue through several stages in order to make a
medicine that is safe, effective, and has approved all regulatory requirements. The process is
sufficiently long, complex, and expensive so that many biological targets must be considered for
every new medicine ultimately approved for clinical use and new research tools may be needed
to investigate each new target. From initial discovery to a marketable medicine is a long,
challenging task. It takes about 12 - 15 years from discovery to the approved medicine and
requires an investment of about US $1 billion. On an average, a million molecules screened but
only a single is explored in late stage clinical trials and is finally made obtainable for patients.
Phases Stages
Phases Stages
Step 1:
Discovery and Development
Step 2:
Preclinical Research
Step 3:
Clinical Development
Step 4:
FDA Review
There are five critical steps in the U.S. Step 5:
drug development process, including FDA Post-market Safety
many phases and stages within each of
Monitoring.
them.
Hit to Lead
In the Hit to Lead (H2L) process, small molecule hits from an HTS are evaluated
and optimized in a limited way into lead compounds. These compounds then
move on to the lead optimization process.
Lead Optimization
In the lead optimization (LO) process, the lead compounds discovered in the
H2L process are synthesized and modified to improve potency and reduce
side effects. Lead optimization conducts experimental testing using animal
efficacy models and ADMET tools, designing the drug candidate.
The drug discovery process ends when one lead compound is found for a drug
candidate, and the process of drug development starts.
Step 2: Preclinical Research
Once a lead compound is found, drug development begins with preclinical research
to determine the efficacy and safety of the drug. Researchers determine the
following about the drug:
These three types of studies are conducted on the whole, living organisms or cells,
including animals and humans; or using non-living organisms or tissue extract. In
vivo, preclinical research examples are the development of new drugs using mice,
rat, and dog models. In vitro is research conducted in a laboratory. Ex vivo uses
animal cells or tissues from a non-living animal. Examples of ex vivo research
assays are finding effective cancer treatment agents; measurements of tissue
properties (physical, thermal, electrical, and optical); and realistic modeling for
new surgical procedures. In an ex vivo assay, a cell is always used as the basis for
small explant cultures that provide a dynamic, controlled, and sterile environment.
In Silico Assays
In silico assays are test systems or biological experiments performed on a computer or
via computer simulation. These are expected to become increasingly popular with the
ongoing improvements in computational power, and behavioral understanding of
molecular dynamics and cell biology.
Drug Delivery
New drug delivery methods include oral, topical, membrane, intravenous, and inhalation. Drug
delivery systems are used for targeted delivery or controlled release of new drugs. Physiological
barriers in animal or human bodies may prevent drugs from reaching the targeted area or
releasing when they should. The goal is to prevent the drug from interacting with healthy tissues
while still being effective.
The complexity of clinical trial design and its associated costs and
implementation issues may affect trials carried out during this phase.
Trials must be safe and efficacious and be completed under the drug
development budget, using a methodology to ensure the drug works as
well as possible for its intended purpose. This rigorous process must be
set up correctly and enroll many volunteers to be effective.
Clinical Trials– Dose Escalation, Single Ascending & Multiple Dose Studies
Proper dosing determines medication effectiveness, and clinical trial examine
dose escalation, single ascending, and multiple dose studies to determine the
best patient dosage.
Phase I – Healthy Volunteer Study
This phase is the first time the drug is tested on humans; less than 100 volunteers
will help researchers assess the safety and pharmacokinetics, absorption,
metabolic, and elimination effects on the body, as well as any side effects for safe
dosage ranges.
Blood, Plasma, Urine & Feces Sample Analysis for Drug and Metabolites
Biological samples used in clinical trials include blood, plasma, urine, and feces to
determine and analyze various properties and effects of the drug and its
metabolites on humans.
Patient Protection – GCP, HIPAA, & Adverse Event Reporting
Accelerated Approval
New drugs may be granted accelerated approval if there is strong
evidence of positive impact on a surrogate endpoint instead of
evidence of impact on actual clinical benefits the drug provides.
Expedition of approval means the medication can help treat severe or
life-threatening conditions.
Reasons for Drug Failure
New drug applications may fail for a variety of reasons, including toxicity, efficacy, PH properties,
bioavailability, or inadequate drug performance.
Toxicity: If the toxicity of a new drug is too high in human or animal patients, the
drug may be rejected due to safety concerns about its use following manufacture.
Efficacy: If a new drug’s efficacy is not high enough or evidence is inconclusive, the
FDA may reject it.
PK Properties or Bioavailability: PK properties or poor bioavailability due to low
aqueous solubility, or high first-pass metabolism, may also cause a drug to fail FDA
review. PK causes of drug failure include inadequate action duration and
unanticipated human drug interactions.
Inadequate Drug Performance: If the new drug performs the desired function, but
only at a shallow level, the FDA may reject the application in favor of a formulation
that performs better.
A Drug Master File (DMF) is a submission to the FDA used to provide confidential,
detailed information about facilities, processes, or articles used in the
manufacturing, processing, packaging, and storing of a human drug.
Drugs for pediatric use are intended for use in children or youth, generally under the
age of 21. In some cases, the American Academy of Pediatrics (AMA) may make
exceptions if a pediatrician and family agree on an older age adult.
Drugs for
Veterinary Use
Drugs for veterinary use are intended for use in animals, pets, and livestock.
However, some veterinary drugs get their start in humans and then change to
human and animal drugs.
The goal of drug development is to prevent human and animal pain and suffering
whenever possible and find and provide new drugs that we can depend on to
improve our health and happiness.
Computer aided drug design
(CADD)
CADD represents
computational
methods and
resources that are
used to facilitate the
design and discovery
of new therapeutic
solutions
INTRODUCTION TO CADD
To change from:
random screening against disease assays
Natural products, synthetic chemicals
INTRODUCTION TO CADD
To:
rational drug design and testing
speed-up screening process
efficient screening (focused, target directed)
De novo design (target directed)
Integration of testing into design process
Fail drugs fast (Remove hopeless ones as early as
possible)
Types of drug design
ligand-based drug design
relies on knowledge of other molecules that bind to the biological target of
interest.
used to derive a pharmacophore model that defines the minimum necessary
structural characteristics a molecule must possess in order to bind to the target
a model of the biological target may be built based on the knowledge of what
binds to it and this model in turn may be used to design new molecular entities
that interact with the target
Alternatively, a quantitative structure-activity relationship in which a correlation
between calculated properties of molecules and their experimentally determined
biological activity, may be derived. These QSAR raltionships in turn may be used
to predict the activity of new analogs
structure-based drug design
relies knowledge of the 3 dimensional structure of the biological target
obtained through:
1. x-ray
2. nuclear magnetic resonance spectroscopy
If an experimental structure of a target is not available, it may be
possible to create a homology model of the target based on the
experimental structure of a related protein.
Homology modeling, also known as comparative modeling of protein,
refers to constructing an atomicresolution model of the "target" and an
experimental three-dimensional structure of a related homologous
protein (the "template").
structure-based drug design
> Using the structure of the biological target, candidate drugs that are
predicted to bind with high affinity and selectivity to the target may be
designed using:
interactive graphics
Intelligence of a medicinal chemist.
various automated computational procedures may be used to
suggest new drug candidates.
structure-based drug design
1) Virtual screening: . The first method is identification of new ligands for a
given receptor by searching large databases of 3D structures of small
molecules to find those fitting the binding pocket of the receptor using fast
approximate docking programs.
2) de novo design of new ligands: In this method, ligand molecules are built up
within the constraints of the binding pocket by assembling small pieces in a
stepwise manner. These pieces can be either individual atoms or molecular
fragments. The key advantage of such a method is that novel structures can
be suggested.
3) optimization of known ligands by evaluating proposed analogs within the
binding cavity.
binding site indentification
It is the first step in structure based design.
relies on identification of concave surfaces on the protein that
can accommodate drug sized molecules that also possess
appropriate "hot spots" (hydrophobic surfaces, hydrogen
bonding sites, etc.) that drive ligand binding.
Direction: Enumerate the animals commonly used in laboratory testing and write
the name of the experiments being used and state the reason.
Miscellaneous
11. Frog Studying embryonic development Oxygen can pass through their
New type antibiotic experiment highly permeable skin
Action of drugs on CNS, heart, Anatomically their heart
neuromuscular junction contains three chambers
which is different from the
other mammals used in the
experiment
12. Pigeon Screening models for anti-emetic Have excellent visual acuity,
drugs color vision, and visual
Bioassay of prolactin memory, all of which rival or
Screening of intravenous even surpass these abilities in
anaesthetics highly visual primates.
Standardization of cardiac glycosides
13. Zebra fish Vertebrate embryo development Excellent model for studying
experiment development in vertebrates
Transgenic model study The embryos develop
Produce new disease models externally to the mother and
Gene-regulation pathways study are transparent, hence easily
viewed and manipulated
14. Chicken Development of atrial septal defect Widely used due to ease of
experiment availability
biological research Good model in toxicology and
pharmacology behavior
studies
Their development process is
quite similar to that of humans
15. Bird embryology and development Chickens have been
experiment domesticated for such a long
influenza, infectious diseases, and time, there is a lot of
viral infections studies information available on their
Pharmaceutical and Genetic physiology
Engineering Chickens lay eggs, which
Study of diseases including cancer means their embryos develop
Vaccine development for chickenpox, outside the mother’s body.
smallpox, and yellow fever and other This allows researchers to
infectious diseases. monitor the development of
the chick at every stage
including development of the
nervous system, limbs
development and cell
migration.