You are on page 1of 19

Presented By: Presented to :

Minakshi Rana Dr. Parminder


Nain
Toxicology in Drug Discovery

 The Primary objective of toxicology studies in the drug development


process is to evaluate the safety of potential drug candidates.
 This is accomplished using relevant animal models and
validated procedures.
 The ultimate goal is to translate the animal model responses into an
understanding of the risk for human subjects.
 Such studies typically assess general toxicology (as determined by in
vivo experiments), safety pharmacology (effects on major organ
systems, e.g. cardiovascular) and genetic toxicity test batteries.
 Many Pharmaceutical companies have now invested in
“Discovery- phase Toxicology” or “Discovery Safety” to identify
potential hazards and to take steps to design out or significantly
reduce undesirable properties at an earlier stage, with the ultimate
aim of enhancing the probability of success in non-clinical and
clinical drug development.
Toxicology in Drug Discovery
Discovery safety assessment of drug projects can be considered in two
broad areas: Target-related safety and chemical related safety.

Target-Related Safety –
 Unintended adverse effects can arise as a consequence of the intended
(primary) pharmacology in tissues other than tissues of efficacy.
 Therefore a key role for the Discovery Safety scientist is to identify
potential adverse effects that may result from primary pharmacology.
 Careful analysis of potential target-related safety issues, together with
an experimental plan to investigate the issues can provide important
discovery project risk assessment data.
 Critically, these data may inform required isoform selectivity profile,
dosing route selection, drug pharmacokinetic properties to mitigate
tissue specific effects (e.g. limit exposure to the central nervous system
for undesirable on-target neuronal pharmacology), or may indeed
guide an early project termination decision.
Toxicology in Drug Discovery

Chemistry-Related Safety –
 A critical pillar of discovery phase safety assessment is to identify
toxicities associated with a chemical series. There are a number
of approaches that can be used to support chemical risk
assessment.
 Ideally, predictive tools identify hazards to be avoided; for
example it may be possible to identify a compound property or
structural features that are associated with adverse effects.
 computational tools that can be used to discover potential
relationships between chemical space and toxicological response.
Special skills of a toxicologist in drug
discovery
 A Discovery Toxicologist or Discovery Safety scientist requires
a broad understanding of the scientific principals & other main
facts of drug discovery, namely biology, pharmacology,
chemistry and DMPK.
 Toxicological risk assessment during the discovery phase
requires interpretation and integration of information from all
these disciplines, and others, which helps to define and provide
context to the safety findings and risks.
 For example, toxicity of a compound is often triggered by the
route of metabolism, clearance or deposition profile of the
molecule.
The skills and understanding required of a discovery
toxicologist with respect to other drug discovery disciplines.
Broad Perspective on Biological Science: Molecular Biology,
Biochemistry and in vitro Pharmacology
 Effective discovery phase toxicology requires a working knowledge of
a broad range of biosciences, including biochemistry, molecular
biology, cellular biology, pharmacology as well as the anatomy and
physiology in health and disease.
 Understanding the biochemical nature of the interactions between
drug-candidates and their intended and unintended targets (usually
proteins).
 By reducing these unintended, or “off-target”, drug protein
interactions, the unintended biological activity of a compound can be
minimized.
Medicinal Chemistry
 Discovery Phase toxicologist is to engage with chemists responsible
for drug design.
 In the first instance a basic understanding of molecular complexity
involved in compound synthesis is required.
 Similarly, an understanding of the role of physicochemical
properties in drug binding and activity alongside impacts on
distribution and pharmacokinetics provides a firm foundation for
consideration of the toxicological properties of the compound.
 In addition, certain structural features of small molecules are
associated with adverse biological therefore
responses,safety scientist should have a knowledge of
discovery thethe alerting
structural features that might constitute hazards for further risk
assessment.
 Finally, an awareness of computational chemistry tools and their
strengths and limitations will help place such alerts into context.
DMPK and In-Vivo Pharmacology
 The Discovery Safety scientist will be responsible for designing in vivo
experiments to determine lead compound (or reference compound)
tolerability and toxicology.
 Specifically, exploration of effects on key organs is important: central
nervous system; cardiovascular system; gastrointestinal system;
respiratory system; immune/inflammatory system and endocrine
system).
 In addition the scientist needs to appreciate compound DMPK (Drug
Metabolism and Pharmaco-Kinetics) properties.
 These properties are broadly spread, from the structure and properties
of metabolites (i.e. products of metabolism conducted by enzymatic
machinery in the target species), through to the concentrations attained
over time in plasma, blood or other tissues following administration
and further to modelling human pharmacokinetics to predict
efficacious doses (PK-pharmacodynamic (PKPD) modelling) or to
model markers of potential toxicity and thereby predict potentially
unsafe doses (toxicokinetic-toxicodynamic TKTD modelling).
DMPK and In-Vivo Pharmacology Continue….
.
 When considering the of a drug, a numb
of consequences may follow, each with
metabolism possible
downstream
implications for the safety/efficacy profile of the product.
 For Example - a metabolite may be toxic in itself, either through
amplification of off-target toxicity through metabolism, enhanced
pharmacology against the desired target, or by showing adverse
events through nonreversible binding to proteins, cell membranes
or organelles such as mitochondria (via activation to a ‘reactive
metabolite’).
 Interactions between the drug in question and other co-
administered medicines may also occur via saturation of metabolic
or other pathways responsible for removing these compounds
from the body.
 Indeed combining these insights with relevant
pharmacodynamics (PD) or toxicodynamic (TD) data (PKPD or
TD modeling) provide a valuable platform for a comprehensive
safety risk assessment.
Different types of toxicities study
performed during drug discovery process
Genetic Toxicology
 Genetic studies are designed to detect whether
toxicity induce genetic by a a
compound can
mechanisms, the consequence
damage of which includes cancer.
variety of
 The Discovery Phase toxicologist should be aware of the battery
of assays used to predict and detect genetic toxicity hazard; as no
single test is capable of detecting all genotoxic mechanisms
relevant to tumourigenesis.
 A familiarity with computational analyses, assessment of
mutagenicity in a bacterial reverse gene mutation test (such as
the Ames test) and evaluation of genotoxicity in mammalian
cells (in vitro and/or in vivo such as the micronucleus test) is
required.
 Awareness of how to respond to genetic toxicity hazards (e.g.
clastogenicty, aneugenicity) and manage these risks in the
context of the project aims and patient population is key.
General Toxicology
 The development of a compound is a stepwise process involving
an evaluation of both the animal and human safety information.
 Traditional toxicology aims to assess a compound in non-clinical
safety studies required to support “first time in human” studies and
through to marketing authorization of a product.
 The goals of the non-clinical safety evaluation generally include a
characterization of toxic effects with respect to target organs, dose
dependence, relationship to exposure, and when appropriate,
potential reversibility.
 For pharmaceuticals, biopharmaceuticals and newer modalities,
this information is helpful for the estimation of an initial safe
starting dose and dose range for the human trials and the
identification of parameters for clinical monitoring for potential
adverse effects.
Animal Toxicology:
Acute toxicity :
 Acute toxicity studies should be carried out in at least two species,
usually mice and rats using the same route as intended for
humans.
 In addition, at least two more route should be used to ensure
systemic absorption of the drug, this route may depend on the
nature of the drug.
 Mortality should be looked for up to 72 hours after parenteral
administration and up to 7 days after oral administration.
 Symptoms, signs and mode of death should be reported, with
appropriate macroscopic and microscopic findings where
necessary.
 LD 50s should be reported preferably with 95 percent confidence
limits, if LD 50s cannot be determined, reasons for this should be
stated.
Long-term toxicity:
 Long-term toxicity studies should be carried out in at least two
mammalian species, of which one should be a non-rodent.
 The duration of study will depend on whether the application is for
marketing permission or for clinical trial, and in the later case, on the
phases of trials.
 If a species is known to metabolize the drug in the same way as humans,
it should be preferred.
 In long-term toxicity studies the drug should be administered 7 days
a week by the route intended for clinical use in humans.
 The number of animals required for these studies, i.e. the minimum
number on which data should be available
 A control group of animals, given the vehicle alone, should always be
included, and three other groups should be given graded doses of the
drug; the highest dose should produce observable toxicity, the lowest
dose should not cause observable toxicity, but should be comparable to
the intended therapeutic dose in humans or a multiple of it.
Reproduction studies
Reproduction studies need to be carried out only if the new drug is proposed to be studied or used in
women of childbearing age .
Two species should generally be used, one of them being non-rodent if possible.
Fertility studies:

The drug should be administered to both males and females, beginning a
sufficient number of days before mating.
In females the medicationshouldbe continued after matingand the pregnant one should be treated
throughout pregnancy.
The highest dose used should not affect general health or growth of the
animals.
The route of administration should be the same as for therapeutic use in humans.
The control and the treated group should be of similar size and large enough to give at least 20 pregnant
animals in the control group of rodents and at least 8 pregnant animals in the control group of non-rodents.
Observations should include total examination of the litters from both the groups, including spontaneous
abortions, if any.
(b) Teratogenicity studies –
 The drugs should be administered throughout the period
of organogenesis, using three dose levels.
 One of the doses should cause minimum maternal
toxicity and one should be the proposed dose for clinical
use in humans or multiple of it.
 The route of administration should be the same as for
human therapeutic use.
 The control and the treated groups should consist of at
least 20 pregnant females in case of non-rodents, on each
dose used.
 Observations should include the number of implantation
sites, restorations if any; and the number fetuses with
their sexes, weights and malformations if any.
(c) Prenatal studies
 The drug should be administered throughout the last third of
pregnancy and then through lactation and weaning. The control
of each treated group should have at least 12 pregnant females
and the dose which causes low foetal loss should be continued
throughout lactation weaning. Animals should be sacrificed and
observations should include macroscopic autopsy and where
necessary, histopathology.
(d) Local toxicity:
 These studies are required when the new drug is proposed to
be used topically in humans.
 The drug should be applied to an appropriate site to determine
local effects in a suitable species such as guinea pigs or rabbits,
if the drug is absorbed from the site of applications, appropriate
systemic toxicity studies will be required.
(e) Mutagenicity and Carcinogenicity:
 These studies are required to be carried out if the drug or its metabolite is
related to a known carcinogen or when the nature and action of the drug
is such as to suggest a carcinogenic/mutagenic potential.
 For carcinogenicity studies, at least two species should be used.
 These species should not have high incidence of spontaneous tumours
and should preferably be known to metabolize the drug in the same
manner as humans.
 At least three does levels should be used; the highest does should be sub-
lethal but cause observable toxicity; the lowest does should be comparable
to the intended human therapeutic does or a multiple of it.
 A control group should always be included.
 The drug should be administered 7 days a week or a fraction of the life
span comparable to the fraction of human life span over which the drug is
likely to be used therapeutically.
 Observations should include macroscopic changes observed at autopsy
and detailed histopathology

You might also like