www.fgks.org   »   [go: up one dir, main page]

SlideShare a Scribd company logo
SAFETY GUIDELINES
S3A: NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF
SYSTEMIC EXPOSURE IN TOXICITY STUDIES
S3B: PHARMACOKINETICS: GUIDANCE FOR REPEATED DOSE TISSUE
DISTRIBUTION STUDIES
PRESENTED BY:
ANSHUL AGRAWAL
PM/2017/403
The International Council for Harmonisation of Technical Requirements for
Pharmaceuticals for Human Use (ICH) brings together the regulatory authorities and
pharmaceutical industry to discuss scientific and technical aspects of drug registration.
Since its inception in 1990, ICH has gradually evolved, to respond to the increasingly
global face of drug development.
ICH's mission is to achieve greater harmonisation worldwide to ensure that safe,
effective, and high quality medicines are developed and registered in the most
resource-efficient manner.
S3A: NOTE FOR GUIDANCE ON
TOXICOKINETICS: THE ASSESSMENT
OF SYSTEMIC EXPOSURE IN TOXICITY
STUDIES
Introduction
The objectives of toxicokinetics and the parameters which may
be determined
General principles to be considered
Toxicokinetics in the various areas of toxicity testing - specific
aspects

Recommended for you

Q1E Evaluation of Stability Data by Rahim Khoja presented on December 22, 2014
Q1E Evaluation of Stability Data  by Rahim Khoja presented on December 22, 2014Q1E Evaluation of Stability Data  by Rahim Khoja presented on December 22, 2014
Q1E Evaluation of Stability Data by Rahim Khoja presented on December 22, 2014

This document provides guidelines for using stability data to propose a retest period or shelf life in product registration. It describes how extrapolation of data beyond the available long-term data can be considered. The guidelines recommend a systematic approach to presenting and evaluating stability data from physical, chemical, biological and microbiological tests. Extrapolation may be proposed if no significant changes are observed under accelerated conditions and it is assumed the same change pattern will continue. A retest period granted via extrapolation should be verified with additional long-term data. The guidelines also provide decision trees and examples of statistical analyses to determine if data from different batches can be pooled for evaluating a single proposed period.

CPCSEA GUIDLINE
CPCSEA GUIDLINECPCSEA GUIDLINE
CPCSEA GUIDLINE

The document provides guidelines from the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA) on the care and use of animals for research. It discusses veterinary care, procurement, quarantine, housing, transportation, experimentation, and euthanasia. The objectives are to avoid unnecessary pain for animals and provide guidelines for housing, care, and experimental procedures. Veterinary care, quarantine for 1-6 weeks, and separation by species are recommended. Physical facilities, environment, food, bedding, and record keeping are also addressed. Anesthesia should control pain during experiments, and euthanasia aims to end pain and provide tissue for research.

Overview of ICH Guidelines, CPCSEA
Overview of ICH Guidelines, CPCSEA Overview of ICH Guidelines, CPCSEA
Overview of ICH Guidelines, CPCSEA

The document provides an overview of ICH guidelines and CPCSEA guidelines for quality control and quality assurance in pharmaceutical research. It discusses the ICH guidelines for quality, safety, efficacy, and multidisciplinary topics. It then summarizes the various Q-series ICH guidelines covering stability testing, analytical validation, impurities, pharmacopoeias, biotechnological products, specifications, and more. It concludes with an overview of the objectives, functions and various aspects of animal husbandry and management covered by the CPCSEA guidelines.

Toxicokinetics only with respect to the development of pharmaceutical products intended for
use in human subjects.
In this context, toxicokinetics is defined as the generation of pharmacokinetic data, either as
an integral component in the conduct of non-clinical toxicity studies or in specially designed
supportive studies, in order to assess systemic exposure. These data may be used in the
interpretation of toxicology findings and their relevance to clinical safety issues.
Toxicokinetic procedures may provide a means of obtaining multiple dose pharmacokinetic
data in the test species, if appropriate parameters are monitored, thus avoiding duplication of
such studies; optimum design in gathering the data will reduce the number of animals required.
However, the toxicokinetic data focus on the kinetics of a new therapeutic agent under the
conditions of the toxicity studies themselves.
Toxicokinetics is thus an integral part of the non-clinical testing programme; it should enhance
the value of the toxicological data generated, both in terms of understanding the toxicity tests and
in comparison with clinical data as part of the assessment of risk and safety in humans.
The development of a pharmaceutical product is a dynamic process which involves continuous
feed-back between non-clinical and clinical studies, no rigid detailed procedures for the application
of toxicokinetics are recommended.
It may not be necessary for toxicokinetic data to be collected in all studies and scientific
judgement should dictate when such data may be useful.
The need for toxicokinetic data and the extent of exposure assessment in individual toxicity
studies should be based on a flexible step-by-step approach and a case-by-case decision making
process to provide sufficient information for a risk and safety assessment.
 to describe the systemic exposure
achieved in animals and its
relationship to dose level and the time
course of the toxicity study.
 to relate the exposure achieved in toxicity
studies to toxicological findings and
contribute to the assessment of the
relevance of these findings to clinical safety.
 to support the choice of species and
treatment regimen in nonclinical toxicity
studies.
 to provide information which, in conjunction
with the toxicity findings, contributes to the
design of subsequent non-clinical toxicity
studies.
Primary objective Secondary objectives
These objectives may be achieved by the derivation of one or more pharmacokinetic parameters from
measurements made at appropriate time points during the course of the individual studies.
These measurements usually consist of plasma (or whole blood or serum) concentrations for the parent
compound and/or metabolite(s) and should be selected on a case-by-case basis.
Plasma (or whole blood or serum) AUC, Cmax and C(time) are the most commonly used parameters in
assessing exposure in toxicokinetics studies.
For some compounds it will be more appropriate to calculate exposure based on the (plasma protein)
unbound concentration.
These data may be obtained from all animals on a toxicity study, in representative subgroups, in satellite
groups or in separate studies.
Toxicity studies which may be usefully supported by toxicokinetic information include single and repeated-
dose toxicity studies, reproductive, genotoxicity and carcinogenicity studies.
Toxicokinetic information may also be of value in assessing the implications of a proposed change in the
clinical route of administration.

Recommended for you

ICH Guidelines for Pharmacovigilance
ICH Guidelines for PharmacovigilanceICH Guidelines for Pharmacovigilance
ICH Guidelines for Pharmacovigilance

Organization and objectives of ICH, expedited reporting, ICSR, PSURs, post approval expedited reporting, pharmacovigilance Planning, good clinical practices

ichexpeditedreporting
Analytical method validation 2020
Analytical method validation 2020Analytical method validation 2020
Analytical method validation 2020

This document provides a summary of analytical method validation. It discusses the types of analytical procedures that should be validated, including identification tests, quantitative impurity tests, limit tests, and active moiety assays. It also summarizes key validation characteristics that should be considered, such as accuracy, precision, specificity, range, detection limit, quantitation limit, linearity, and robustness. The document provides definitions and methodology recommendations for validating analytical procedures. It emphasizes that the validation process verifies that an analytical method is suitable for its intended purpose.

quality assurance
Stability studies of drug ICH Q1
Stability studies of drug ICH Q1 Stability studies of drug ICH Q1
Stability studies of drug ICH Q1

The ICH Q1A guideline provides recommendations for conducting stability studies on drug substances and drug products to establish retest and shelf-life periods. Key points include: - Stability studies should be conducted on 3 primary batches under long-term, intermediate, and accelerated storage conditions specified in the guideline. - Testing frequency is typically every 3 months for the first year of long-term studies and specifications cover physical, chemical, biological, and microbiological attributes. - The purpose is to evaluate how quality varies over time under influence of factors like temperature and humidity and provide evidence to support recommended storage conditions.

Some general principles are set out which should be taken into consideration in the design of
individual studies.
It should be noted that for those toxicity studies whose performance is subject to Good Laboratory
Practice (GLP) the concomitant toxicokinetics must also conform to GLP.
Toxicokinetic studies retrospectively designed to generate specific sets of data under conditions which
closely mimic those of the toxicity studies should also conform to GLP when they are necessary for
the evaluation of safety.
Reporting
Analytical methods
Statistical evaluation of data
Determination of metabolites
Route of administration
Complicating factors in exposure interpretation
Extent of exposure assessment in toxicity studies
Contribution to the setting of dose levels in order to produce adequate exposure [Low, Medium, High dose]
Justification of time points for sampling
Quantification of exposure
The quantification of systemic exposure provides an assessment of the burden on the test species and assists in
the interpretation of similarities and differences in toxicity across species, dose groups and sexes. The exposure
might be represented by plasma (serum or blood) concentrations or the AUCs of parent compound and/or
metabolite(s).
When designing the toxicity studies, exposure and dose-dependence in humans at therapeutic dose levels
(either expected or established), should be considered in order to achieve relevant exposure at various dose levels
in the animal toxicity studies. The possibility that there may be species differences in the pharmacodynamics of the
substance (either qualitative or quantitative) should also be taken into consideration.
Pharmacodynamic effects or toxicity might also give supporting evidence of exposure.
Toxicokinetic monitoring or profiling of toxicity studies should establish what level of exposure has been achieved
during the course of the study and may also serve to alert the toxicologist to non-linear, dose-related changes in
exposure which may have occurred.
Toxicokinetic information may allow better interspecies comparisons than simple dose/body weight (or surface
area) comparisons.
1. Quantification of exposure:
The time points for collecting body fluids in concomitant toxicokinetic studies should be as frequent as is
necessary, but not as frequent as to interfere with the normal conduct of the study or to cause undue
physiological stress to the animals.
In each study, the number of time points should be justified on the basis that they are adequate to estimate
exposure.
The justification should be based on kinetic data gathered from earlier toxicity studies, from pilot or dose range-
finding studies, from separate studies in the same animal model or in other models allowing reliable extrapolation.
2. Justification of time points for sampling:

Recommended for you

ICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validation

The document provides guidelines on validation of analytical procedures from the International Conference on Harmonisation (ICH) and the World Health Organization (WHO). It discusses validation characteristics like accuracy, precision, specificity, linearity, range, detection limit and quantitation limit that should be considered when validating identification tests, assays, and tests for impurities. It provides definitions for key terms and recommendations on how validation of these characteristics should be performed.

ichwhovalidation
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...

Microsomal assays are used to evaluate the metabolic stability and identify metabolites of drug compounds. Liver microsomes containing drug-metabolizing enzymes are incubated with a drug, and the percentage of intact parent compound over time is measured. Toxicokinetics applies pharmacokinetic principles to safety studies, quantifying drug exposure and establishing interspecies differences in metabolism. Toxicokinetic evaluations in preclinical studies help support clinical trial dose selection by measuring absorption, distribution, biotransformation and excretion of drugs.

toxicokineticsmicrosomal assays
Ich guidlines q and s
Ich guidlines  q and sIch guidlines  q and s
Ich guidlines q and s

The document provides information on the International Conference on Harmonization (ICH), including: - ICH aims to harmonize technical requirements for pharmaceutical registration across regions to ensure safety and efficacy. - It involves regulators and industry from the EU, Japan, and USA. - The goals are to establish common guidelines and make information available globally. - ICH guidelines cover quality, safety, efficacy, and multidisciplinary topics for drug development and review. - The document then focuses on specific ICH guidelines related to quality, including stability testing, analytical method validation, and impurities.

The setting of dose levels in toxicity studies is largely governed by the toxicology findings and the
pharmacodynamic responses of the test species.
3. Contribution to the setting of dose levels in order to produce
adequate exposure:
Low dose levels: At the low dose, preferably a
no-toxic-effect dose level, the exposure in the
animals of any toxicity study should ideally
equal or just exceed the maximum expected
(or known to be attained) in patients. It is
recognised that this ideal is not always
achievable and that low doses will often need
to be determined by considerations of
toxicology; nevertheless, systemic exposure
should be determined.
Intermediate dose levels: Exposure at
intermediate dose levels should normally
represent an appropriate multiple (or fraction)
of the exposure at lower (or higher) dose
levels dependent upon the objectives of the
toxicity study.
High dose levels: The high dose levels in
toxicity studies will normally be determined by
toxicological considerations. However, the
exposure achieved at the dose levels used
should be assessed.
In toxicity studies, systemic exposure should be estimated in an appropriate number of animals and dose groups
to provide a basis for risk assessment.
Concomitant toxicokinetics may be performed either in all or a representative proportion of the animals used in
the main study or in special satellite groups.
Normally, samples for the generation of toxicokinetic data may be collected from main study animals, where
large animals are involved, but satellite groups may be required for the smaller (rodent) species.
The number of animals to be used should be the minimum consistent with generating adequate toxicokinetic
data.
Where both male and female animals are utilised in the main study it is normal to estimate exposure in animals
of both sexes unless some justification can be made for not so doing.
Toxicokinetic data are not necessarily required from studies of different duration if the dosing regimen is
essentially unchanged.
4. Extent of exposure assessment in
toxicity studies:
A few caveats should be noted.
Species differences in protein binding, tissue uptake, receptor properties and metabolic profile should be
considered.
For example, it may be more appropriate for highly protein bound compounds to have exposure expressed as
the free (unbound) concentrations.
 In addition, the pharmacological activity of metabolites, the toxicology of metabolites and antigenicity of
biotechnology products may be complicating factors.
Furthermore, it should be noted that even at relatively low plasma concentrations, high levels of the
administered compound and/or metabolite(s) may occur in specific organs or tissues.
5. Complicating factors in exposure
interpretation:
The toxicokinetic strategy to be adopted for the use of alternative routes of administration, for example by inhalation, topical or
parenteral delivery, should be based on the pharmacokinetic properties of the substance administered by the intended route.
It sometimes happens that a proposal is made to adopt a new clinical route of administration for a pharmaceutical product; for
example, a product initially developed as an oral formulation may subsequently be developed for intravenous administration.
In this context, it will be necessary to ascertain whether changing the clinical route will significantly reduce the safety margin.
This process may include a comparison of the systemic exposure to the compound and/or its relevant metabolite(s) (AUC and
Cmax) in humans generated by the existing and proposed routes of administration.
6. Route of administration:
If the new route results in
increased AUC and/or Cmax,
or a change in metabolic
route, the continuing
assurance of safety from
animal toxicology and kinetics
should be reconsidered.
If exposure is not substantially
greater, or different, by the
proposed new route compared to
that for the existing route(s) then
additional non-clinical toxicity
studies may focus on local toxicity.

Recommended for you

Drug interactions linked to transporters
Drug interactions linked to transportersDrug interactions linked to transporters
Drug interactions linked to transporters

This document discusses how drug transporters affect the absorption, distribution, and excretion of drugs. It describes uptake and efflux transporters expressed in the intestine, liver, kidney, and blood-brain barrier that determine drug concentrations in tissues. Inhibition or induction of these transporters by other drugs can impact the pharmacokinetics and pharmacodynamics of victim drugs. The document then classifies and provides examples of important uptake transporters like OATPs and OCTs and efflux transporters like P-gp, BCRP, MRP2, and BSEP, noting their tissue expression and roles in drug absorption, distribution, and excretion.

druginteraction
ICH Pharmaceutical development guideline-Q8
ICH Pharmaceutical development guideline-Q8ICH Pharmaceutical development guideline-Q8
ICH Pharmaceutical development guideline-Q8

What is ICH Q8 guidelines? Image result for ICH Pharmaceutical development guideline-Q8 The ICH Q8 guideline is intended to provide guidance on the contents of Section 3.2. P. 2 (Pharmaceutical Development) for drug products as defined in the scope of Module 3 of the Common Technical Document (ICH topic M4).

parent guideline: pharmaceutical developmentannex to the parent guideline: pharmaceutical deveaddition of annex to the parent guideline
REGULATORY AFFAIRS & MASTER FORMULA RECORD
REGULATORY AFFAIRS & MASTER FORMULA RECORDREGULATORY AFFAIRS & MASTER FORMULA RECORD
REGULATORY AFFAIRS & MASTER FORMULA RECORD

Regulatory affairs is a profession that developed to ensure public health by regulating product safety. Regulatory affairs professionals work with regulatory agencies and internal departments to register products, track legislation changes, and provide strategic advice. They must understand both internal manufacturing processes and requirements from agencies like CDSCO in India or the USFDA. A Master Formula Record (MFR) is a key document that contains all information about manufacturing a pharmaceutical product, including starting materials, packaging, and processing instructions. It is prepared by R&D as the standard reference for making Batch Manufacturing Records. The MFR must include product details, a procedure description, and all ingredients and their quantities. It ensures consistent manufacturing of batches according to the standard process.

A primary objective of toxicokinetics is to describe the systemic exposure to the administered compound achieved
in the toxicology species. However, there may be circumstances when measurement of metabolite concentrations
in plasma or other body fluids is especially important in the conduct of toxicokinetics:
7. Determination of metabolites:
- When the administered
compound acts as a
'pro-drug' and the
delivered metabolite is
acknowledged to be the
primary active entity.
- When the compound is
metabolised to one or
more
pharmacologically or
toxicologically active
metabolites which could
make a significant
contribution to
tissue/organ responses.
- When the administered
compound is very
extensively
metabolised and the
measurement of plasma
or tissue concentrations
of a major metabolite is
the only practical means
of estimating exposure
following administration
of the compound in
toxicity studies.
•The data should allow a representative assessment of the exposure.
•However, because large intra- and inter-individual variation of kinetic parameters may occur and small
numbers of animals are involved in generating toxicokinetic data, a high level of precision in terms of
statistics is not normally needed.
•Consideration should be given to the calculation of mean or median values and estimates of variability, but
in some cases the data of individual animals may be more important than a refined statistical analysis of
group data.
•If data transformation (e.g. logarithmic) is performed, a rationale should be provided.
8. Statistical evaluation of data:
Integration of pharmacokinetics into toxicity testing implies early development of analytical methods for which
the choice of analytes and matrices should be continually reviewed as information is gathered on metabolism and
species differences.
The analytical methods to be used in toxicokinetic studies should be specific for the entity to be measured and of
an adequate accuracy and precision.
The choice of analyte and the matrix to be assayed (biological fluids or tissue) should be stated and possible
interference by endogenous components in each type of sample (from each species) should be investigated.
Plasma, serum or whole blood are normally the matrices of choice for toxicokinetic studies.
If the drug substance is a racemate or some other mixture of enantiomers, additional justification should be made
for the choice of the analyte [racemate or enantiomer(s)].
The analyte and matrix assayed in non-clinical studies should ideally be the same as in clinical studies. If different
assay methods are used in non-clinical and clinical studies they should all be suitably validated.
9. Analytical methods:
A comprehensive account of the toxicokinetic data generated, together with an evaluation of the results
and of the implications for the interpretation of the toxicology findings should be given. An outline of the
analytical method should be reported or referenced.
10. Reporting:

Recommended for you

M10 bioanalytical method validation
M10 bioanalytical method validationM10 bioanalytical method validation
M10 bioanalytical method validation

This document discusses the proposed new ICH guideline on bioanalytical method validation. The guideline will provide recommendations for validating bioanalytical methods used in non-clinical and clinical drug development. It will harmonize current regional validation guidelines and support streamlined global drug development. The guideline will define validation characteristics like specificity, accuracy, and precision and provide acceptance criteria. It will also address validation requirements and cases where partial or cross-validation are necessary.

Validation of dissolution apparatus
Validation of dissolution apparatusValidation of dissolution apparatus
Validation of dissolution apparatus

This document discusses the validation of dissolution test apparatus. It begins with a brief history of validation and reasons for validating equipment. Validation ensures equipment operates consistently and accurately. The document then discusses various types of dissolution test apparatus and the qualification process, including design, installation, operational, and performance qualification. It also addresses sources of error and concludes that acceptable qualification demonstrates the apparatus is validated for use in dissolution testing.

Importance and application of toxicokinetics
Importance and application of toxicokineticsImportance and application of toxicokinetics
Importance and application of toxicokinetics

Toxicokinetics describes how the body handles toxicants over time through absorption, distribution, metabolism and excretion (ADME). It is important in drug development to generate kinetic data for toxicity assessment, check safety ratios, and set safe dose levels in clinical trials. Toxicokinetic evaluation helps reduce animal testing, understand inter-individual differences in responses, and has applications in screening anticancer drugs, cell-based assays, and other areas of research.

toxicokineticapplicationimportance
Based on the principles of toxicokinetics outlined above the following specific considerations refer to individual
areas of toxicity testing. The frequency of exposure monitoring or profiling may be extended or reduced where
necessary.
Single-dose toxicity
studies
Repeated-dose
toxicity studies
Genotoxicity studies
Carcinogenicity
(oncogenicity)
studies
Reproductive toxicity
studies
Single-dose toxicity studies:
These studies are often performed in a very early phase of development before a bioanalytical method has
been developed and toxicokinetic monitoring of these studies is therefore not normally possible.
Plasma samples may be taken in such studies and stored for later analysis, if necessary; appropriate stability
data for the analyte in the matrix sampled would then be required.
Alternatively, additional toxicokinetic studies may be carried out after completion of a single-dose toxicity
study in order to respond to specific questions which may arise from the study.
Results from single-dose kinetic studies may help in the choice of formulation and in the prediction of rate
and duration of exposure during a dosing interval.
This may assist in the selection of appropriate dose levels for use in later studies.
Repeated-dose toxicity studies:
The treatment regimen and species should be selected whenever possible with regard to pharmacodynamic
and pharmacokinetic principles.
Toxicokinetics should be incorporated appropriately into the design of the studies.
It may consist of exposure profiling or monitoring at appropriate dose levels at the start and towards the
end of the treatment period of the first repeat dose study.
The procedure adopted for later studies will depend on the results from the first study and on any changes in
the proposed treatment regimen.
Monitoring or profiling may be extended, reduced or modified for specific compounds where problems have
arisen in the interpretation of earlier toxicity studies.
Genotoxicity studies:
For negative results of in vivo genotoxicity studies, it may be appropriate to have demonstrated
systemic exposure in the species used or to have characterised exposure in the indicator tissue.

Recommended for you

Clinical trial protocol development
Clinical trial protocol developmentClinical trial protocol development
Clinical trial protocol development

This document outlines the key components and considerations for developing a clinical trial protocol. It discusses that a protocol is a complete written plan for a research study involving human subjects. It identifies important sections such as the title page, objectives, study design, safety reporting, statistical analysis, and informed consent. It emphasizes that the protocol language should be clear, concise, and understandable for diverse readers. It also provides guidance on properly writing eligibility criteria, adverse event definitions, and obtaining informed consent to protect human subjects.

Investigational new drug (IND)
Investigational new drug (IND)Investigational new drug (IND)
Investigational new drug (IND)

1. An Investigational New Drug (IND) application is required for testing an experimental drug in humans and must be submitted to regulatory agencies like the FDA for approval. 2. The IND application contains preclinical research data on animal and microbiological studies as well as clinical trial protocols, manufacturing information, and investigator details. 3. There are different types of INDs including commercial, non-commercial, emergency use, and treatment INDs which have varying requirements and purposes in the drug development process.

drug regulatory affairsscience
Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptx

1. Toxicokinetics is the study of how toxic substances are affected by the body in terms of absorption, distribution, metabolism, and excretion. It applies pharmacokinetic principles to doses used in toxicology testing. 2. The primary objective of toxicokinetic evaluation in preclinical studies is to describe systemic exposure levels in animals and relate this to toxicity findings to assess clinical safety. Secondary objectives include supporting species and dose selection for toxicity studies. 3. Toxicokinetic data is collected in various required preclinical safety studies, including repeat-dose toxicity studies, reproduction toxicity studies, and genotoxicity studies, to interpret results and demonstrate drug exposure.

pharmaceuticalpharmacologypharmacy
Carcinogenicity (oncogenicity) studies:
Appropriate monitoring or profiling of these studies should be undertaken in order to generate toxicokinetic
data which may assist in the design of the main studies.
Particular attention should be paid to species and strains which have not been included in earlier toxicity
studies and to the use of routes or methods of administration which are being used for the first time.
In principle, the ideal study design would ensure that dose levels in oncogenicity studies generate a range of
systemic exposure values that exceed the maximum therapeutic exposure for humans by varying multiples.
However, it is recognised that this idealised selection of dose levels may be confounded by unavoidable
species-specific problems.
Thus, the emphasis of this guidance is on the need to estimate systemic exposure, to parent compound and/or
metabolite(s) at appropriate dose levels and at various stages of an oncogenicity study, so that the findings of the
study may be considered in the perspective of comparative exposure for the animal model and humans.
A highest dose based on knowledge of probable systemic exposure in the test species and in humans may be
an acceptable end-point in testing for carcinogenic potential.
Historically, a toxicity end-point1 has been often used to select the top dose level.
Reproductive toxicity studies:
It is preferable to have some information on pharmacokinetics before initiating reproduction studies since this
may suggest the need to adjust the choice of species, study design and dosing schedules.
Fertility studies: The general principles for repeated-dose toxicity studies apply. The need to monitor these
studies will depend on the dosing regimen used and the information already available from earlier studies in the
selected species.
Studies in pregnant and lactating animals: The treatment regimen during the exposure period should be
selected on the basis of the toxicological findings and on pharmacokinetic and toxicokinetic principles.
Consideration should be given to the possibility that the kinetics will differ in pregnant and non-pregnant
animals.
S3B: PHARMACOKINETICS: GUIDANCE
FOR REPEATED DOSE TISSUE
DISTRIBUTION STUDIES
This Guideline has been developed by the appropriate ICH Expert
Working Group and has been subject to consultation by the
regulatory parties.
Introduction
Circumstances Under Which Repeated Dose Tissue
Distribution Studies Should be Considered
Design and Conduct of Repeated Dose Tissue Distribution
Studies
Conclusion

Recommended for you

Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines

ICH guidelines provide standards for toxicity studies to ensure safe, effective, and high quality pharmaceutical products. Guideline S3A deals with conducting toxicity studies and quantifying exposure. General principles include quantifying exposure levels in different species and sexes using plasma concentration or area under the curve. Toxicokinetic studies should be performed to determine metabolite levels and justify dose levels. Reporting should include detailed toxicokinetic data and evaluation. Toxicokinetics are assessed in various toxicity studies including single dose studies, repeated dose studies, genotoxicity studies, carcinogenicity studies, and reproductive toxicity studies.

ich
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology

This document provides guidelines for safety pharmacology studies for human pharmaceuticals from the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). The guidelines discuss the objectives, scope, general principles, test systems, experimental design, dose levels/concentrations, duration of studies, and studies on metabolites for safety pharmacology evaluations. The goal is to help protect clinical trial participants and patients by identifying potential adverse effects of pharmaceuticals early in development.

Toxicological Approach to Drug Discovery
Toxicological Approach to Drug DiscoveryToxicological Approach to Drug Discovery
Toxicological Approach to Drug Discovery

This document outlines the toxicological approach to drug development. It discusses the importance of conducting various toxicity studies at different stages of drug development to ensure safety. These include single dose, repeated dose, fertility, reproductive, developmental and genotoxicity studies in animals. It describes the typical safety program involving staged approach and discusses factors to consider in designing toxicity studies. The goal is to obtain sufficient non-clinical safety data to support clinical trials and assess safety for human use.

A comprehensive knowledge of the absorption, distribution, metabolism and elimination of a compound is
important for the interpretation of pharmacology and toxicology studies.
Tissue distribution studies are essential in providing information on distribution and accumulation of the
compound and/or metabolites, especially in relation to potential sites of action; this information may be useful
for designing toxicology and pharmacology studies and for interpreting the results of these experiments.

In the EC, US and Japan, there has been a general agreement on the need to conduct single dose tissue
distribution studies as part of the non-clinical programme. These studies often provide sufficient information
about tissue distribution.
There has been no consistent requirement for repeated dose tissue distribution studies. However, there may be
circumstances when assessments after repeated dosing may yield important information.
1.When single dose tissue distribution studies suggest that the apparent half-life of the test
compound (and/or metabolites) in organs or tissues significantly exceeds the apparent half life of the
elimination phase in plasma and is also more than twice the dosing interval in the toxicity studies,
repeated dose tissue distribution studies may be appropriate.
2. When steady-state levels of a compound/metabolite in the circulation, determined in repeated
dose pharmacokinetic or toxicokinetic studies, are markedly higher than those predicted from single
dose kinetic studies, then repeated dose tissue distribution studies should be considered.
3. When histopathological changes, critical for the safety evaluation of the test substances, are
observed that would not be predicted from short term toxicity studies, single dose tissue distribution
studies and pharmacological studies, repeated dose tissue distribution studies may aid in the
interpretation of these findings. Those organs or tissues which were the site of the lesions should be
the focus of such studies.
4. When the pharmaceutical is being developed for site-specific targeted delivery, repeated dose
tissue distribution studies may be appropriate.
The objectives of these studies may be achieved using radiolabelled compounds or alternative methods of
sufficient sensitivity and specificity.
Dose level(s) and species should be chosen to address the problem that led to the consideration of the repeated
dose tissue distribution study.
Information from previous pharmacokinetic and toxicokinetic studies should be used in selecting the duration of
dosing in repeated dose tissue distribution studies.
One week of dosing is normally considered to be a minimum period.
A longer duration should be selected when the blood/plasma concentration of the compound and/or its
metabolites does not reach steady state. It is normally considered unnecessary to dose for longer than three weeks.
Consideration should be given to measuring unchanged compound and/or metabolites in organs and tissues in
which extensive accumulation occurs or if it is believed that such data may clarify mechanisms of organ toxicity.
Tissue distribution studies are an important component in the non-clinical kinetics programme. For most
compounds, it is expected that single dose tissue distribution studies with sufficient sensitivity and
specificity will provide an adequate assessment of tissue distribution and the potential for accumulation.
Thus, repeated dose tissue distribution studies should not be required uniformly for all compounds and
should only be conducted when appropriate data cannot be derived from other sources. Repeated dose
studies may be appropriate under certain circumstances based on the data from single dose tissue
distribution studies, toxicity and toxicokinetic studies. The studies may be most appropriate for compounds
which have an apparently long half life, incomplete elimination or unanticipated organ toxicity. The design
and timing of repeated dose tissue distribution studies should be determined on a case-by case basis.

Recommended for you

Toxiocokinetic evaluation in preclinical studies.pptx
Toxiocokinetic evaluation in preclinical studies.pptxToxiocokinetic evaluation in preclinical studies.pptx
Toxiocokinetic evaluation in preclinical studies.pptx

its importance in education

toxicokinetic evalution
presentation.pptx
presentation.pptxpresentation.pptx
presentation.pptx

This document summarizes information about toxicokinetics and saturation kinetics studies presented by Shilajit Das. It discusses how toxicokinetics studies are used to evaluate systemic drug exposure in animals and relate it to dose levels and toxicity findings to assess human safety. It provides the objectives, goals and general principles of toxicokinetics studies including quantifying exposure, justifying sampling timepoints, and determining metabolites. It also discusses how saturation kinetics can cause non-linear pharmacokinetics when enzyme or carrier capacities are exceeded, and how this non-linearity can be detected by evaluating parameters like bioavailability and clearance at different doses.

PTSM 2.pptx
PTSM 2.pptxPTSM 2.pptx
PTSM 2.pptx

Toxicokinetics deals with absorption , distribution , biotransformation and excretion of chemicals . According to ICH S3A guidance on the assessment of systemic exposure in toxicity studies , toxicokinetics is defined as the generation of pharmacokinetics data , either as an integral component in the conduct of non-clinical toxicity studies or in specially designed supportive studies , in order to assess systemic exposure. Toxicokinetics is the science to understand what the body does with a drug when the drug is given at a relatively high dose . Toxicokinetics play a major role in interpreting the histopathological finding in a toxicological study. Toxicokinetic is essentially the study of "how a substance gets into the body and what happens to it in the body." The primary objective of toxicokinetic is: • To describe the systemic exposure achieved in animals and its relationship to dose level and the time course of the toxicity study. Secondary objectives are: • To relate the exposure achieved in toxicity studies to toxicological findings and contribute to the assessment of the relevance of these findings to clinical safety. • To support the choice of species and treatment regimen in non-clinical toxicity studies. • To provide information which, in conjunction with the toxicity findings, contributes to the design of subsequent non-clinical toxicity studies. The primary purpose of toxicokinetic studies is to determine the rate, extent and duration of systemic exposure of the test animal species to the test compound at the different dose levels employed during toxicity studies and to provide data for direct comparison with human exposure to the test compound. These data help to understand the relationship between observed toxicity and administered dose. They also play a role in the clinical setting, assisting in the setting of plasma limits for early human exposure and in the calculation of safety margins. A major challenge in the risk/benefit assessment of new chemical entities is the rational and reliable extrapolation of preclinical (nonclinical) safety evaluation data from animals to humans. Toxicity studies, core to the safety evaluation process, are designed to determine a no observed toxic effect level as well as an observed toxic effect level. Traditionally, these levels have been those of administered dose, and safety margins are derived as the ratio between the preclinical no observed toxic effect dose level in animals and the clinical (or environmental) dose level in humans. In reality, however, safety margins thus derived are often optimistic guesses because they do not take into account the respective systemic exposures to the test compound during the preclinical studies and eventual human clinical (or environmental) use. Toxicokinetics involves the generation of kinetic data to assess systemic exposure, either as an integral component of preclinical toxicity studies, or in specially designed supportive studies.

importance of toxicokineticapplications of toxicokineticobjective of toxicokinetic
References:
https://www.ich.org/about/mission.html
http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guide
lines/Safety/S3A/Step4/S3A_Guideline.pdf
http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guide
lines/Safety/S3B/Step4/S3B_Guideline.pdf
THANK YOU

More Related Content

What's hot

Pharmacovigilance safety Mon. in clinical trials.pptx
Pharmacovigilance safety Mon. in clinical trials.pptxPharmacovigilance safety Mon. in clinical trials.pptx
Pharmacovigilance safety Mon. in clinical trials.pptx
Roshan Yadav
 
UNIT 4 TOPIC Managing Monitoring Clinical Trials.pdf
UNIT 4 TOPIC Managing Monitoring Clinical Trials.pdfUNIT 4 TOPIC Managing Monitoring Clinical Trials.pdf
UNIT 4 TOPIC Managing Monitoring Clinical Trials.pdf
Dr Yogi Pandya
 
PRINCIPLES of FT-NMR & 13C NMR
PRINCIPLES of FT-NMR & 13C NMRPRINCIPLES of FT-NMR & 13C NMR
PRINCIPLES of FT-NMR & 13C NMR
Aditya Sharma
 
Q1E Evaluation of Stability Data by Rahim Khoja presented on December 22, 2014
Q1E Evaluation of Stability Data  by Rahim Khoja presented on December 22, 2014Q1E Evaluation of Stability Data  by Rahim Khoja presented on December 22, 2014
Q1E Evaluation of Stability Data by Rahim Khoja presented on December 22, 2014
Rahim Khoja
 
CPCSEA GUIDLINE
CPCSEA GUIDLINECPCSEA GUIDLINE
CPCSEA GUIDLINE
Ashvin Bhoge
 
Overview of ICH Guidelines, CPCSEA
Overview of ICH Guidelines, CPCSEA Overview of ICH Guidelines, CPCSEA
Overview of ICH Guidelines, CPCSEA
MANIKANDAN V
 
ICH Guidelines for Pharmacovigilance
ICH Guidelines for PharmacovigilanceICH Guidelines for Pharmacovigilance
ICH Guidelines for Pharmacovigilance
Dr. Ramesh Bhandari
 
Analytical method validation 2020
Analytical method validation 2020Analytical method validation 2020
Analytical method validation 2020
PRANJAY PATIL
 
Stability studies of drug ICH Q1
Stability studies of drug ICH Q1 Stability studies of drug ICH Q1
Stability studies of drug ICH Q1
Manali Parab
 
ICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validation
SACHIN C P
 
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
siva ganesh
 
Ich guidlines q and s
Ich guidlines  q and sIch guidlines  q and s
Ich guidlines q and s
Bashant Kumar sah
 
Drug interactions linked to transporters
Drug interactions linked to transportersDrug interactions linked to transporters
Drug interactions linked to transporters
gopinathannsriramachandraeduin
 
ICH Pharmaceutical development guideline-Q8
ICH Pharmaceutical development guideline-Q8ICH Pharmaceutical development guideline-Q8
ICH Pharmaceutical development guideline-Q8
vedanshu malviya
 
REGULATORY AFFAIRS & MASTER FORMULA RECORD
REGULATORY AFFAIRS & MASTER FORMULA RECORDREGULATORY AFFAIRS & MASTER FORMULA RECORD
REGULATORY AFFAIRS & MASTER FORMULA RECORD
sachin suryawanshi
 
M10 bioanalytical method validation
M10 bioanalytical method validationM10 bioanalytical method validation
M10 bioanalytical method validation
sawantanil
 
Validation of dissolution apparatus
Validation of dissolution apparatusValidation of dissolution apparatus
Validation of dissolution apparatus
Shraddha Kumbhar
 
Importance and application of toxicokinetics
Importance and application of toxicokineticsImportance and application of toxicokinetics
Importance and application of toxicokinetics
Prajjwal Rajput
 
Clinical trial protocol development
Clinical trial protocol developmentClinical trial protocol development
Clinical trial protocol development
SACHIN C P
 
Investigational new drug (IND)
Investigational new drug (IND)Investigational new drug (IND)
Investigational new drug (IND)
Manish Rajput
 

What's hot (20)

Pharmacovigilance safety Mon. in clinical trials.pptx
Pharmacovigilance safety Mon. in clinical trials.pptxPharmacovigilance safety Mon. in clinical trials.pptx
Pharmacovigilance safety Mon. in clinical trials.pptx
 
UNIT 4 TOPIC Managing Monitoring Clinical Trials.pdf
UNIT 4 TOPIC Managing Monitoring Clinical Trials.pdfUNIT 4 TOPIC Managing Monitoring Clinical Trials.pdf
UNIT 4 TOPIC Managing Monitoring Clinical Trials.pdf
 
PRINCIPLES of FT-NMR & 13C NMR
PRINCIPLES of FT-NMR & 13C NMRPRINCIPLES of FT-NMR & 13C NMR
PRINCIPLES of FT-NMR & 13C NMR
 
Q1E Evaluation of Stability Data by Rahim Khoja presented on December 22, 2014
Q1E Evaluation of Stability Data  by Rahim Khoja presented on December 22, 2014Q1E Evaluation of Stability Data  by Rahim Khoja presented on December 22, 2014
Q1E Evaluation of Stability Data by Rahim Khoja presented on December 22, 2014
 
CPCSEA GUIDLINE
CPCSEA GUIDLINECPCSEA GUIDLINE
CPCSEA GUIDLINE
 
Overview of ICH Guidelines, CPCSEA
Overview of ICH Guidelines, CPCSEA Overview of ICH Guidelines, CPCSEA
Overview of ICH Guidelines, CPCSEA
 
ICH Guidelines for Pharmacovigilance
ICH Guidelines for PharmacovigilanceICH Guidelines for Pharmacovigilance
ICH Guidelines for Pharmacovigilance
 
Analytical method validation 2020
Analytical method validation 2020Analytical method validation 2020
Analytical method validation 2020
 
Stability studies of drug ICH Q1
Stability studies of drug ICH Q1 Stability studies of drug ICH Q1
Stability studies of drug ICH Q1
 
ICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validation
 
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
 
Ich guidlines q and s
Ich guidlines  q and sIch guidlines  q and s
Ich guidlines q and s
 
Drug interactions linked to transporters
Drug interactions linked to transportersDrug interactions linked to transporters
Drug interactions linked to transporters
 
ICH Pharmaceutical development guideline-Q8
ICH Pharmaceutical development guideline-Q8ICH Pharmaceutical development guideline-Q8
ICH Pharmaceutical development guideline-Q8
 
REGULATORY AFFAIRS & MASTER FORMULA RECORD
REGULATORY AFFAIRS & MASTER FORMULA RECORDREGULATORY AFFAIRS & MASTER FORMULA RECORD
REGULATORY AFFAIRS & MASTER FORMULA RECORD
 
M10 bioanalytical method validation
M10 bioanalytical method validationM10 bioanalytical method validation
M10 bioanalytical method validation
 
Validation of dissolution apparatus
Validation of dissolution apparatusValidation of dissolution apparatus
Validation of dissolution apparatus
 
Importance and application of toxicokinetics
Importance and application of toxicokineticsImportance and application of toxicokinetics
Importance and application of toxicokinetics
 
Clinical trial protocol development
Clinical trial protocol developmentClinical trial protocol development
Clinical trial protocol development
 
Investigational new drug (IND)
Investigational new drug (IND)Investigational new drug (IND)
Investigational new drug (IND)
 

Similar to Safety guidelines- S3A and S3B

Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptx
ARSHIKHANAM4
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
R Nadaf
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
Ramavath Aruna
 
Toxicological Approach to Drug Discovery
Toxicological Approach to Drug DiscoveryToxicological Approach to Drug Discovery
Toxicological Approach to Drug Discovery
Suhas Reddy C
 
Toxiocokinetic evaluation in preclinical studies.pptx
Toxiocokinetic evaluation in preclinical studies.pptxToxiocokinetic evaluation in preclinical studies.pptx
Toxiocokinetic evaluation in preclinical studies.pptx
manishaJyala2
 
presentation.pptx
presentation.pptxpresentation.pptx
presentation.pptx
SauravChaudhary54
 
PTSM 2.pptx
PTSM 2.pptxPTSM 2.pptx
PTSM 2.pptx
VishnuPrabhakar26
 
Dose selection
Dose selectionDose selection
Dose selection
pp_shivgunde
 
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptx
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptxTOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptx
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptx
Anmolkanda06
 
Safety Pharmacology Studies ICH guideline S7A
Safety Pharmacology Studies ICH guideline S7ASafety Pharmacology Studies ICH guideline S7A
Safety Pharmacology Studies ICH guideline S7A
R C Patel Institute of Pharmaceutical Education and Research , Shirpur
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
SourabhAtak
 
Extrapolation of in vitro data to preclinical and.pptx
Extrapolation of in vitro data to preclinical and.pptxExtrapolation of in vitro data to preclinical and.pptx
Extrapolation of in vitro data to preclinical and.pptx
ARSHIKHANAM4
 
toxicokinetics and saturation kinetics
toxicokinetics and saturation kineticstoxicokinetics and saturation kinetics
toxicokinetics and saturation kinetics
pharmacistnitish
 
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICALEXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
TMU
 
Regulatory guidelines for conducting toxicity studies by ich
Regulatory guidelines for conducting toxicity studies by ichRegulatory guidelines for conducting toxicity studies by ich
Regulatory guidelines for conducting toxicity studies by ich
AnimatedWorld
 
Safety s7 a-seminar-ppt-pptx
Safety s7 a-seminar-ppt-pptxSafety s7 a-seminar-ppt-pptx
Safety s7 a-seminar-ppt-pptx
Santhanakumar21
 
Safety pharmacology by tasnim
Safety pharmacology by tasnimSafety pharmacology by tasnim
Safety pharmacology by tasnim
Tasnim Taher
 
Carcinogenicity testing
Carcinogenicity testingCarcinogenicity testing
Carcinogenicity testing
saravanan gunasekar
 
toxicokinetics-1.pptx
toxicokinetics-1.pptxtoxicokinetics-1.pptx
toxicokinetics-1.pptx
Unnatikhare1
 
CARCINOGENICITY, CARCINOGENES, CANCER AND FACTORS
CARCINOGENICITY, CARCINOGENES, CANCER AND FACTORSCARCINOGENICITY, CARCINOGENES, CANCER AND FACTORS
CARCINOGENICITY, CARCINOGENES, CANCER AND FACTORS
Dheeraj Saini
 

Similar to Safety guidelines- S3A and S3B (20)

Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptx
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
Toxicological Approach to Drug Discovery
Toxicological Approach to Drug DiscoveryToxicological Approach to Drug Discovery
Toxicological Approach to Drug Discovery
 
Toxiocokinetic evaluation in preclinical studies.pptx
Toxiocokinetic evaluation in preclinical studies.pptxToxiocokinetic evaluation in preclinical studies.pptx
Toxiocokinetic evaluation in preclinical studies.pptx
 
presentation.pptx
presentation.pptxpresentation.pptx
presentation.pptx
 
PTSM 2.pptx
PTSM 2.pptxPTSM 2.pptx
PTSM 2.pptx
 
Dose selection
Dose selectionDose selection
Dose selection
 
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptx
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptxTOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptx
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIES.pptx
 
Safety Pharmacology Studies ICH guideline S7A
Safety Pharmacology Studies ICH guideline S7ASafety Pharmacology Studies ICH guideline S7A
Safety Pharmacology Studies ICH guideline S7A
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
Extrapolation of in vitro data to preclinical and.pptx
Extrapolation of in vitro data to preclinical and.pptxExtrapolation of in vitro data to preclinical and.pptx
Extrapolation of in vitro data to preclinical and.pptx
 
toxicokinetics and saturation kinetics
toxicokinetics and saturation kineticstoxicokinetics and saturation kinetics
toxicokinetics and saturation kinetics
 
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICALEXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
 
Regulatory guidelines for conducting toxicity studies by ich
Regulatory guidelines for conducting toxicity studies by ichRegulatory guidelines for conducting toxicity studies by ich
Regulatory guidelines for conducting toxicity studies by ich
 
Safety s7 a-seminar-ppt-pptx
Safety s7 a-seminar-ppt-pptxSafety s7 a-seminar-ppt-pptx
Safety s7 a-seminar-ppt-pptx
 
Safety pharmacology by tasnim
Safety pharmacology by tasnimSafety pharmacology by tasnim
Safety pharmacology by tasnim
 
Carcinogenicity testing
Carcinogenicity testingCarcinogenicity testing
Carcinogenicity testing
 
toxicokinetics-1.pptx
toxicokinetics-1.pptxtoxicokinetics-1.pptx
toxicokinetics-1.pptx
 
CARCINOGENICITY, CARCINOGENES, CANCER AND FACTORS
CARCINOGENICITY, CARCINOGENES, CANCER AND FACTORSCARCINOGENICITY, CARCINOGENES, CANCER AND FACTORS
CARCINOGENICITY, CARCINOGENES, CANCER AND FACTORS
 

More from anshagrawal2121

Selecting and Prioritizing Healthcare Projects by HTA
Selecting and Prioritizing Healthcare Projects by HTASelecting and Prioritizing Healthcare Projects by HTA
Selecting and Prioritizing Healthcare Projects by HTA
anshagrawal2121
 
Measuring And Controlling Assets employed
Measuring And Controlling Assets employedMeasuring And Controlling Assets employed
Measuring And Controlling Assets employed
anshagrawal2121
 
Impact of big data analytics on banking sector
Impact of big data analytics on banking sector Impact of big data analytics on banking sector
Impact of big data analytics on banking sector
anshagrawal2121
 
Technical Analysis & Financial Analysis
Technical Analysis & Financial AnalysisTechnical Analysis & Financial Analysis
Technical Analysis & Financial Analysis
anshagrawal2121
 
Demand forecasting
Demand forecasting Demand forecasting
Demand forecasting
anshagrawal2121
 
Biological warfare
Biological warfareBiological warfare
Biological warfare
anshagrawal2121
 
Facility layout
Facility layoutFacility layout
Facility layout
anshagrawal2121
 
Vyapam scam
Vyapam scamVyapam scam
Vyapam scam
anshagrawal2121
 

More from anshagrawal2121 (8)

Selecting and Prioritizing Healthcare Projects by HTA
Selecting and Prioritizing Healthcare Projects by HTASelecting and Prioritizing Healthcare Projects by HTA
Selecting and Prioritizing Healthcare Projects by HTA
 
Measuring And Controlling Assets employed
Measuring And Controlling Assets employedMeasuring And Controlling Assets employed
Measuring And Controlling Assets employed
 
Impact of big data analytics on banking sector
Impact of big data analytics on banking sector Impact of big data analytics on banking sector
Impact of big data analytics on banking sector
 
Technical Analysis & Financial Analysis
Technical Analysis & Financial AnalysisTechnical Analysis & Financial Analysis
Technical Analysis & Financial Analysis
 
Demand forecasting
Demand forecasting Demand forecasting
Demand forecasting
 
Biological warfare
Biological warfareBiological warfare
Biological warfare
 
Facility layout
Facility layoutFacility layout
Facility layout
 
Vyapam scam
Vyapam scamVyapam scam
Vyapam scam
 

Recently uploaded

Comfort, Rest, Sleep and Pain.pptx
Comfort, Rest, Sleep  and Pain.pptxComfort, Rest, Sleep  and Pain.pptx
Comfort, Rest, Sleep and Pain.pptx
Nandish Sannaiah
 
Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)
Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)
Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)
HafsaAnwaar1
 
BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATIONBASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
JaiDivyaTella
 
Westgard's rules and LJ (Levey Jennings) Charts.
Westgard's rules and LJ (Levey Jennings) Charts.Westgard's rules and LJ (Levey Jennings) Charts.
Westgard's rules and LJ (Levey Jennings) Charts.
Reenaz Shaik
 
Veterinary Medicines Manufacturers in India
Veterinary Medicines Manufacturers in IndiaVeterinary Medicines Manufacturers in India
Veterinary Medicines Manufacturers in India
Heilsaa Care
 
Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)
MuhammedMNasser
 
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptxWhy Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
AmandaChou9
 
lessonplan JTR.pptx For medical education
lessonplan JTR.pptx For medical educationlessonplan JTR.pptx For medical education
lessonplan JTR.pptx For medical education
DrJALAGAMTHIRUPATHIR
 
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Mohamad محمد Al-Gailani الكيلاني
 
Prodrug design for Sustained Drug action -.KB.pptx
Prodrug design for Sustained Drug action -.KB.pptxProdrug design for Sustained Drug action -.KB.pptx
Prodrug design for Sustained Drug action -.KB.pptx
ANAGHA K B
 
Introduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptxIntroduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptx
Shamsuddin Mahmud
 
APPROACH TO DIAGNOSIS OF LIVER DISEASES.ppt
APPROACH TO DIAGNOSIS OF LIVER DISEASES.pptAPPROACH TO DIAGNOSIS OF LIVER DISEASES.ppt
APPROACH TO DIAGNOSIS OF LIVER DISEASES.ppt
KarthikeyanA87
 
How to build a successful medical tourism practice
How to build a successful medical tourism practiceHow to build a successful medical tourism practice
How to build a successful medical tourism practice
Arlen Meyers, MD, MBA
 
Nursing management of patient with peritoneal dialysis
Nursing management of patient with peritoneal dialysisNursing management of patient with peritoneal dialysis
Nursing management of patient with peritoneal dialysis
shivalingatalekar1
 
Ventilation Perfusion Ratio, Physiological dead space and physiological shunt
Ventilation Perfusion Ratio, Physiological dead space and physiological shuntVentilation Perfusion Ratio, Physiological dead space and physiological shunt
Ventilation Perfusion Ratio, Physiological dead space and physiological shunt
MedicoseAcademics
 
Top 10 Habits for Longevity [Biohacker Summit 2024]
Top 10 Habits for Longevity [Biohacker Summit 2024]Top 10 Habits for Longevity [Biohacker Summit 2024]
Top 10 Habits for Longevity [Biohacker Summit 2024]
Olli Sovijärvi
 
General Principles of Law, History and various Acts related to Drugs and Phar...
General Principles of Law, History and various Acts related to Drugs and Phar...General Principles of Law, History and various Acts related to Drugs and Phar...
General Principles of Law, History and various Acts related to Drugs and Phar...
PratibhaSonawane5
 
Perforation.pptx
Perforation.pptxPerforation.pptx
Perforation.pptx
Nandish Sannaiah
 
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.GawadHemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
NephroTube - Dr.Gawad
 
Jurnalul Fericirii Life Care - Iulie 2024
Jurnalul Fericirii Life Care - Iulie 2024Jurnalul Fericirii Life Care - Iulie 2024
Jurnalul Fericirii Life Care - Iulie 2024
Life Care Romania
 

Recently uploaded (20)

Comfort, Rest, Sleep and Pain.pptx
Comfort, Rest, Sleep  and Pain.pptxComfort, Rest, Sleep  and Pain.pptx
Comfort, Rest, Sleep and Pain.pptx
 
Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)
Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)
Obstructive sleep apnea (OSA) and Obesity hypoventilation syndrome (OHS)
 
BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATIONBASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
 
Westgard's rules and LJ (Levey Jennings) Charts.
Westgard's rules and LJ (Levey Jennings) Charts.Westgard's rules and LJ (Levey Jennings) Charts.
Westgard's rules and LJ (Levey Jennings) Charts.
 
Veterinary Medicines Manufacturers in India
Veterinary Medicines Manufacturers in IndiaVeterinary Medicines Manufacturers in India
Veterinary Medicines Manufacturers in India
 
Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)
 
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptxWhy Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
 
lessonplan JTR.pptx For medical education
lessonplan JTR.pptx For medical educationlessonplan JTR.pptx For medical education
lessonplan JTR.pptx For medical education
 
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
 
Prodrug design for Sustained Drug action -.KB.pptx
Prodrug design for Sustained Drug action -.KB.pptxProdrug design for Sustained Drug action -.KB.pptx
Prodrug design for Sustained Drug action -.KB.pptx
 
Introduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptxIntroduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptx
 
APPROACH TO DIAGNOSIS OF LIVER DISEASES.ppt
APPROACH TO DIAGNOSIS OF LIVER DISEASES.pptAPPROACH TO DIAGNOSIS OF LIVER DISEASES.ppt
APPROACH TO DIAGNOSIS OF LIVER DISEASES.ppt
 
How to build a successful medical tourism practice
How to build a successful medical tourism practiceHow to build a successful medical tourism practice
How to build a successful medical tourism practice
 
Nursing management of patient with peritoneal dialysis
Nursing management of patient with peritoneal dialysisNursing management of patient with peritoneal dialysis
Nursing management of patient with peritoneal dialysis
 
Ventilation Perfusion Ratio, Physiological dead space and physiological shunt
Ventilation Perfusion Ratio, Physiological dead space and physiological shuntVentilation Perfusion Ratio, Physiological dead space and physiological shunt
Ventilation Perfusion Ratio, Physiological dead space and physiological shunt
 
Top 10 Habits for Longevity [Biohacker Summit 2024]
Top 10 Habits for Longevity [Biohacker Summit 2024]Top 10 Habits for Longevity [Biohacker Summit 2024]
Top 10 Habits for Longevity [Biohacker Summit 2024]
 
General Principles of Law, History and various Acts related to Drugs and Phar...
General Principles of Law, History and various Acts related to Drugs and Phar...General Principles of Law, History and various Acts related to Drugs and Phar...
General Principles of Law, History and various Acts related to Drugs and Phar...
 
Perforation.pptx
Perforation.pptxPerforation.pptx
Perforation.pptx
 
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.GawadHemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
 
Jurnalul Fericirii Life Care - Iulie 2024
Jurnalul Fericirii Life Care - Iulie 2024Jurnalul Fericirii Life Care - Iulie 2024
Jurnalul Fericirii Life Care - Iulie 2024
 

Safety guidelines- S3A and S3B

  • 1. SAFETY GUIDELINES S3A: NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES S3B: PHARMACOKINETICS: GUIDANCE FOR REPEATED DOSE TISSUE DISTRIBUTION STUDIES PRESENTED BY: ANSHUL AGRAWAL PM/2017/403
  • 2. The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) brings together the regulatory authorities and pharmaceutical industry to discuss scientific and technical aspects of drug registration. Since its inception in 1990, ICH has gradually evolved, to respond to the increasingly global face of drug development. ICH's mission is to achieve greater harmonisation worldwide to ensure that safe, effective, and high quality medicines are developed and registered in the most resource-efficient manner.
  • 3. S3A: NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES
  • 4. Introduction The objectives of toxicokinetics and the parameters which may be determined General principles to be considered Toxicokinetics in the various areas of toxicity testing - specific aspects
  • 5. Toxicokinetics only with respect to the development of pharmaceutical products intended for use in human subjects. In this context, toxicokinetics is defined as the generation of pharmacokinetic data, either as an integral component in the conduct of non-clinical toxicity studies or in specially designed supportive studies, in order to assess systemic exposure. These data may be used in the interpretation of toxicology findings and their relevance to clinical safety issues. Toxicokinetic procedures may provide a means of obtaining multiple dose pharmacokinetic data in the test species, if appropriate parameters are monitored, thus avoiding duplication of such studies; optimum design in gathering the data will reduce the number of animals required. However, the toxicokinetic data focus on the kinetics of a new therapeutic agent under the conditions of the toxicity studies themselves.
  • 6. Toxicokinetics is thus an integral part of the non-clinical testing programme; it should enhance the value of the toxicological data generated, both in terms of understanding the toxicity tests and in comparison with clinical data as part of the assessment of risk and safety in humans. The development of a pharmaceutical product is a dynamic process which involves continuous feed-back between non-clinical and clinical studies, no rigid detailed procedures for the application of toxicokinetics are recommended. It may not be necessary for toxicokinetic data to be collected in all studies and scientific judgement should dictate when such data may be useful. The need for toxicokinetic data and the extent of exposure assessment in individual toxicity studies should be based on a flexible step-by-step approach and a case-by-case decision making process to provide sufficient information for a risk and safety assessment.
  • 7.  to describe the systemic exposure achieved in animals and its relationship to dose level and the time course of the toxicity study.  to relate the exposure achieved in toxicity studies to toxicological findings and contribute to the assessment of the relevance of these findings to clinical safety.  to support the choice of species and treatment regimen in nonclinical toxicity studies.  to provide information which, in conjunction with the toxicity findings, contributes to the design of subsequent non-clinical toxicity studies. Primary objective Secondary objectives
  • 8. These objectives may be achieved by the derivation of one or more pharmacokinetic parameters from measurements made at appropriate time points during the course of the individual studies. These measurements usually consist of plasma (or whole blood or serum) concentrations for the parent compound and/or metabolite(s) and should be selected on a case-by-case basis. Plasma (or whole blood or serum) AUC, Cmax and C(time) are the most commonly used parameters in assessing exposure in toxicokinetics studies. For some compounds it will be more appropriate to calculate exposure based on the (plasma protein) unbound concentration. These data may be obtained from all animals on a toxicity study, in representative subgroups, in satellite groups or in separate studies. Toxicity studies which may be usefully supported by toxicokinetic information include single and repeated- dose toxicity studies, reproductive, genotoxicity and carcinogenicity studies. Toxicokinetic information may also be of value in assessing the implications of a proposed change in the clinical route of administration.
  • 9. Some general principles are set out which should be taken into consideration in the design of individual studies. It should be noted that for those toxicity studies whose performance is subject to Good Laboratory Practice (GLP) the concomitant toxicokinetics must also conform to GLP. Toxicokinetic studies retrospectively designed to generate specific sets of data under conditions which closely mimic those of the toxicity studies should also conform to GLP when they are necessary for the evaluation of safety.
  • 10. Reporting Analytical methods Statistical evaluation of data Determination of metabolites Route of administration Complicating factors in exposure interpretation Extent of exposure assessment in toxicity studies Contribution to the setting of dose levels in order to produce adequate exposure [Low, Medium, High dose] Justification of time points for sampling Quantification of exposure
  • 11. The quantification of systemic exposure provides an assessment of the burden on the test species and assists in the interpretation of similarities and differences in toxicity across species, dose groups and sexes. The exposure might be represented by plasma (serum or blood) concentrations or the AUCs of parent compound and/or metabolite(s). When designing the toxicity studies, exposure and dose-dependence in humans at therapeutic dose levels (either expected or established), should be considered in order to achieve relevant exposure at various dose levels in the animal toxicity studies. The possibility that there may be species differences in the pharmacodynamics of the substance (either qualitative or quantitative) should also be taken into consideration. Pharmacodynamic effects or toxicity might also give supporting evidence of exposure. Toxicokinetic monitoring or profiling of toxicity studies should establish what level of exposure has been achieved during the course of the study and may also serve to alert the toxicologist to non-linear, dose-related changes in exposure which may have occurred. Toxicokinetic information may allow better interspecies comparisons than simple dose/body weight (or surface area) comparisons. 1. Quantification of exposure:
  • 12. The time points for collecting body fluids in concomitant toxicokinetic studies should be as frequent as is necessary, but not as frequent as to interfere with the normal conduct of the study or to cause undue physiological stress to the animals. In each study, the number of time points should be justified on the basis that they are adequate to estimate exposure. The justification should be based on kinetic data gathered from earlier toxicity studies, from pilot or dose range- finding studies, from separate studies in the same animal model or in other models allowing reliable extrapolation. 2. Justification of time points for sampling:
  • 13. The setting of dose levels in toxicity studies is largely governed by the toxicology findings and the pharmacodynamic responses of the test species. 3. Contribution to the setting of dose levels in order to produce adequate exposure: Low dose levels: At the low dose, preferably a no-toxic-effect dose level, the exposure in the animals of any toxicity study should ideally equal or just exceed the maximum expected (or known to be attained) in patients. It is recognised that this ideal is not always achievable and that low doses will often need to be determined by considerations of toxicology; nevertheless, systemic exposure should be determined. Intermediate dose levels: Exposure at intermediate dose levels should normally represent an appropriate multiple (or fraction) of the exposure at lower (or higher) dose levels dependent upon the objectives of the toxicity study. High dose levels: The high dose levels in toxicity studies will normally be determined by toxicological considerations. However, the exposure achieved at the dose levels used should be assessed.
  • 14. In toxicity studies, systemic exposure should be estimated in an appropriate number of animals and dose groups to provide a basis for risk assessment. Concomitant toxicokinetics may be performed either in all or a representative proportion of the animals used in the main study or in special satellite groups. Normally, samples for the generation of toxicokinetic data may be collected from main study animals, where large animals are involved, but satellite groups may be required for the smaller (rodent) species. The number of animals to be used should be the minimum consistent with generating adequate toxicokinetic data. Where both male and female animals are utilised in the main study it is normal to estimate exposure in animals of both sexes unless some justification can be made for not so doing. Toxicokinetic data are not necessarily required from studies of different duration if the dosing regimen is essentially unchanged. 4. Extent of exposure assessment in toxicity studies:
  • 15. A few caveats should be noted. Species differences in protein binding, tissue uptake, receptor properties and metabolic profile should be considered. For example, it may be more appropriate for highly protein bound compounds to have exposure expressed as the free (unbound) concentrations.  In addition, the pharmacological activity of metabolites, the toxicology of metabolites and antigenicity of biotechnology products may be complicating factors. Furthermore, it should be noted that even at relatively low plasma concentrations, high levels of the administered compound and/or metabolite(s) may occur in specific organs or tissues. 5. Complicating factors in exposure interpretation:
  • 16. The toxicokinetic strategy to be adopted for the use of alternative routes of administration, for example by inhalation, topical or parenteral delivery, should be based on the pharmacokinetic properties of the substance administered by the intended route. It sometimes happens that a proposal is made to adopt a new clinical route of administration for a pharmaceutical product; for example, a product initially developed as an oral formulation may subsequently be developed for intravenous administration. In this context, it will be necessary to ascertain whether changing the clinical route will significantly reduce the safety margin. This process may include a comparison of the systemic exposure to the compound and/or its relevant metabolite(s) (AUC and Cmax) in humans generated by the existing and proposed routes of administration. 6. Route of administration: If the new route results in increased AUC and/or Cmax, or a change in metabolic route, the continuing assurance of safety from animal toxicology and kinetics should be reconsidered. If exposure is not substantially greater, or different, by the proposed new route compared to that for the existing route(s) then additional non-clinical toxicity studies may focus on local toxicity.
  • 17. A primary objective of toxicokinetics is to describe the systemic exposure to the administered compound achieved in the toxicology species. However, there may be circumstances when measurement of metabolite concentrations in plasma or other body fluids is especially important in the conduct of toxicokinetics: 7. Determination of metabolites: - When the administered compound acts as a 'pro-drug' and the delivered metabolite is acknowledged to be the primary active entity. - When the compound is metabolised to one or more pharmacologically or toxicologically active metabolites which could make a significant contribution to tissue/organ responses. - When the administered compound is very extensively metabolised and the measurement of plasma or tissue concentrations of a major metabolite is the only practical means of estimating exposure following administration of the compound in toxicity studies.
  • 18. •The data should allow a representative assessment of the exposure. •However, because large intra- and inter-individual variation of kinetic parameters may occur and small numbers of animals are involved in generating toxicokinetic data, a high level of precision in terms of statistics is not normally needed. •Consideration should be given to the calculation of mean or median values and estimates of variability, but in some cases the data of individual animals may be more important than a refined statistical analysis of group data. •If data transformation (e.g. logarithmic) is performed, a rationale should be provided. 8. Statistical evaluation of data:
  • 19. Integration of pharmacokinetics into toxicity testing implies early development of analytical methods for which the choice of analytes and matrices should be continually reviewed as information is gathered on metabolism and species differences. The analytical methods to be used in toxicokinetic studies should be specific for the entity to be measured and of an adequate accuracy and precision. The choice of analyte and the matrix to be assayed (biological fluids or tissue) should be stated and possible interference by endogenous components in each type of sample (from each species) should be investigated. Plasma, serum or whole blood are normally the matrices of choice for toxicokinetic studies. If the drug substance is a racemate or some other mixture of enantiomers, additional justification should be made for the choice of the analyte [racemate or enantiomer(s)]. The analyte and matrix assayed in non-clinical studies should ideally be the same as in clinical studies. If different assay methods are used in non-clinical and clinical studies they should all be suitably validated. 9. Analytical methods:
  • 20. A comprehensive account of the toxicokinetic data generated, together with an evaluation of the results and of the implications for the interpretation of the toxicology findings should be given. An outline of the analytical method should be reported or referenced. 10. Reporting:
  • 21. Based on the principles of toxicokinetics outlined above the following specific considerations refer to individual areas of toxicity testing. The frequency of exposure monitoring or profiling may be extended or reduced where necessary. Single-dose toxicity studies Repeated-dose toxicity studies Genotoxicity studies Carcinogenicity (oncogenicity) studies Reproductive toxicity studies
  • 22. Single-dose toxicity studies: These studies are often performed in a very early phase of development before a bioanalytical method has been developed and toxicokinetic monitoring of these studies is therefore not normally possible. Plasma samples may be taken in such studies and stored for later analysis, if necessary; appropriate stability data for the analyte in the matrix sampled would then be required. Alternatively, additional toxicokinetic studies may be carried out after completion of a single-dose toxicity study in order to respond to specific questions which may arise from the study. Results from single-dose kinetic studies may help in the choice of formulation and in the prediction of rate and duration of exposure during a dosing interval. This may assist in the selection of appropriate dose levels for use in later studies.
  • 23. Repeated-dose toxicity studies: The treatment regimen and species should be selected whenever possible with regard to pharmacodynamic and pharmacokinetic principles. Toxicokinetics should be incorporated appropriately into the design of the studies. It may consist of exposure profiling or monitoring at appropriate dose levels at the start and towards the end of the treatment period of the first repeat dose study. The procedure adopted for later studies will depend on the results from the first study and on any changes in the proposed treatment regimen. Monitoring or profiling may be extended, reduced or modified for specific compounds where problems have arisen in the interpretation of earlier toxicity studies.
  • 24. Genotoxicity studies: For negative results of in vivo genotoxicity studies, it may be appropriate to have demonstrated systemic exposure in the species used or to have characterised exposure in the indicator tissue.
  • 25. Carcinogenicity (oncogenicity) studies: Appropriate monitoring or profiling of these studies should be undertaken in order to generate toxicokinetic data which may assist in the design of the main studies. Particular attention should be paid to species and strains which have not been included in earlier toxicity studies and to the use of routes or methods of administration which are being used for the first time. In principle, the ideal study design would ensure that dose levels in oncogenicity studies generate a range of systemic exposure values that exceed the maximum therapeutic exposure for humans by varying multiples. However, it is recognised that this idealised selection of dose levels may be confounded by unavoidable species-specific problems. Thus, the emphasis of this guidance is on the need to estimate systemic exposure, to parent compound and/or metabolite(s) at appropriate dose levels and at various stages of an oncogenicity study, so that the findings of the study may be considered in the perspective of comparative exposure for the animal model and humans. A highest dose based on knowledge of probable systemic exposure in the test species and in humans may be an acceptable end-point in testing for carcinogenic potential. Historically, a toxicity end-point1 has been often used to select the top dose level.
  • 26. Reproductive toxicity studies: It is preferable to have some information on pharmacokinetics before initiating reproduction studies since this may suggest the need to adjust the choice of species, study design and dosing schedules. Fertility studies: The general principles for repeated-dose toxicity studies apply. The need to monitor these studies will depend on the dosing regimen used and the information already available from earlier studies in the selected species. Studies in pregnant and lactating animals: The treatment regimen during the exposure period should be selected on the basis of the toxicological findings and on pharmacokinetic and toxicokinetic principles. Consideration should be given to the possibility that the kinetics will differ in pregnant and non-pregnant animals.
  • 27. S3B: PHARMACOKINETICS: GUIDANCE FOR REPEATED DOSE TISSUE DISTRIBUTION STUDIES This Guideline has been developed by the appropriate ICH Expert Working Group and has been subject to consultation by the regulatory parties.
  • 28. Introduction Circumstances Under Which Repeated Dose Tissue Distribution Studies Should be Considered Design and Conduct of Repeated Dose Tissue Distribution Studies Conclusion
  • 29. A comprehensive knowledge of the absorption, distribution, metabolism and elimination of a compound is important for the interpretation of pharmacology and toxicology studies. Tissue distribution studies are essential in providing information on distribution and accumulation of the compound and/or metabolites, especially in relation to potential sites of action; this information may be useful for designing toxicology and pharmacology studies and for interpreting the results of these experiments.  In the EC, US and Japan, there has been a general agreement on the need to conduct single dose tissue distribution studies as part of the non-clinical programme. These studies often provide sufficient information about tissue distribution. There has been no consistent requirement for repeated dose tissue distribution studies. However, there may be circumstances when assessments after repeated dosing may yield important information.
  • 30. 1.When single dose tissue distribution studies suggest that the apparent half-life of the test compound (and/or metabolites) in organs or tissues significantly exceeds the apparent half life of the elimination phase in plasma and is also more than twice the dosing interval in the toxicity studies, repeated dose tissue distribution studies may be appropriate. 2. When steady-state levels of a compound/metabolite in the circulation, determined in repeated dose pharmacokinetic or toxicokinetic studies, are markedly higher than those predicted from single dose kinetic studies, then repeated dose tissue distribution studies should be considered. 3. When histopathological changes, critical for the safety evaluation of the test substances, are observed that would not be predicted from short term toxicity studies, single dose tissue distribution studies and pharmacological studies, repeated dose tissue distribution studies may aid in the interpretation of these findings. Those organs or tissues which were the site of the lesions should be the focus of such studies. 4. When the pharmaceutical is being developed for site-specific targeted delivery, repeated dose tissue distribution studies may be appropriate.
  • 31. The objectives of these studies may be achieved using radiolabelled compounds or alternative methods of sufficient sensitivity and specificity. Dose level(s) and species should be chosen to address the problem that led to the consideration of the repeated dose tissue distribution study. Information from previous pharmacokinetic and toxicokinetic studies should be used in selecting the duration of dosing in repeated dose tissue distribution studies. One week of dosing is normally considered to be a minimum period. A longer duration should be selected when the blood/plasma concentration of the compound and/or its metabolites does not reach steady state. It is normally considered unnecessary to dose for longer than three weeks. Consideration should be given to measuring unchanged compound and/or metabolites in organs and tissues in which extensive accumulation occurs or if it is believed that such data may clarify mechanisms of organ toxicity.
  • 32. Tissue distribution studies are an important component in the non-clinical kinetics programme. For most compounds, it is expected that single dose tissue distribution studies with sufficient sensitivity and specificity will provide an adequate assessment of tissue distribution and the potential for accumulation. Thus, repeated dose tissue distribution studies should not be required uniformly for all compounds and should only be conducted when appropriate data cannot be derived from other sources. Repeated dose studies may be appropriate under certain circumstances based on the data from single dose tissue distribution studies, toxicity and toxicokinetic studies. The studies may be most appropriate for compounds which have an apparently long half life, incomplete elimination or unanticipated organ toxicity. The design and timing of repeated dose tissue distribution studies should be determined on a case-by case basis.

Editor's Notes

  1. .