SlideShare una empresa de Scribd logo
1 de 82
What is ICH?"
• International Conference on
  Harmonization of Technical Requirements
  for Registration of Pharmaceuticals for
  Human Use" . ICH is a joint initiative
  involving both regulators and research-
  based industry representatives of the EU,
  Japan and the US in scientific and technical
  discussions of the testing procedures
  required to assess and ensure the safety,
  quality and efficacy of medicines.
What is the purpose of ICH?
• The objective of ICH is to increase
  international harmonization of technical
  requirements to ensure that safe,
  effective, and high quality medicines are
  developed and registered in the most
  efficient and cost- effective manner.
Safety Guidelines
Safety Guidelines
• ICH has produced a comprehensive set of
  safety guidelines to uncover potential risks
  like carcinogenicity, genotoxicity and
  reprotoxicity. A recent breakthrough has
  been a non-clinical testing strategy for
  assessing the QT interval prolongation
  liability: the single most important cause of
  drug withdrawals in recent years
S1 _guideline
         14 November 2012
•    Rodent Carcinogenicity Studies for Human
    Pharmaceuticals : A change to the current S1 harmonised
    Guidelines on rodent carcinogenicity testing is proposed
    to introduce a more comprehensive and integrated
    approach to addressing the risk of human carcinogenicity
    of pharmaceuticals, clarify and update, without
    compromising safety, the criteria for deciding whether
    the conduct of a two-year rodent carcinogenicity study of
    a given pharmaceutical would add value to this risk
    assessment.
November 2012
• Proposal: It is expected that proposal will
  modify the ICH S1 Guideline, by advancing
  an approach in which the need for 2-year
  bioassays are not automatically triggered,
  but evaluated instead based on a case-by-
  case approach taking both predicted
  positive carcinogenicity study outcomes as
  well as negative outcomes into
  consideration for a scientifically justifiable
  waiver.
S1A Guideline
Need For Carcinogenicity Studies Of
  Pharmaceuticals
 OBJECTIVES
      To identify a tumorigenic potential in animals
       To assess the relevant risk in humans.
      studies also preceded our current understanding of
       tumorigenesis with non-genotoxic agents .
      To avoid the unnecessary use of animals in testing .
HISTORICAL BACKGROUND
• IN JAPAN (1990)
        Guidelines for Toxicity Studies of Drugs Manual . carcinogenicity
         studies were needed if the clinical use was expected to be
         continuously for 6 months or longer.
• In the United States
        pharmaceuticals were tested in animals for their carcinogenic
         potential before widespread use in humans.
• IN EUROPE
        Rules Governing Medicinal Products in the European Community
         defined the circumstances when carcinogenicity studies were
         required.
FACTORS FOR
  CARCINOGENICITY TESTING
• DURATION & EXPOSURE: Carcinogenicity studies
  should be performed for any pharmaceutical
  whose expected clinical use is continuous for at
  least 6 months. Certain classes of compounds
  may not be used continuously over a minimum of
  6 months but may be expected to be used
  repeatedly.
• Cause for Concern: concern about their carcinogenic
  potential.
 Factors which could be considered may include:
        previous demonstration of carcinogenic potential in the product
         class that is considered relevant to humans.
        structure-activity relationship suggesting carcinogenic risk.
        evidence of pre neoplastic lesions in repeated dose toxicity studies.
        long-term tissue retention of parent compound or metabolite(s)
         resulting in local tissue reactions or other pathophysiological
         resposes.
• Genotoxicity:
      Unequivocally genotoxic compounds, in the absence of other
       data, are presumed to be trans-species carcinogens, implying
       a hazard to humans. Such compounds need not be subjected
       to long-term carcinogenicity studies.
      if such a drug is intended to be administered chronically to
       humans a chronic toxicity study (up to one year) may be
       necessary to detect early tumorigenic effects.
Indication and Patient Population:
o When carcinogenicity studies are required they usually need to be
   completed before application for marketing approval.
o Completed rodent carcinogenicity studies are not needed in advance
   of the conduct of large scale clinical trials, unless there is special
   concern for the patient population.
o To treat certain serious diseases, carcinogenicity testing need not be
   conducted before market approval although these studies should be
   conducted post-approval.
Route of Exposure:
 should be the same as the intended clinical route
  when feasible.
 carcinogenicity studies should only be conducted
  by a single route.
 it is important that relevant organs for the clinical
  route (e.g., lung for inhalational agents) be
  adequately exposed to the test material.
Extent of Systemic Exposure:
 showing poor systemic exposure from topical routes in
   humans.
 may not need studies by the oral route to assess the
   carcinogenic potential to internal organs.
 Pharmaceuticals administered by the ocular route may
   not require carcinogenicity studies.
 Evidence should be provided that there are no significant
   changes in pharmacokinetics, pharmacodynamics, or
   toxicity.
Endogenous Peptides and Protein Substances or Their
  Analogs:
 produced by chemical synthesis, by
  extraction/purification from an animal/human source or
  by biotechnological methods such as recombinant DNA
  technology.
 Carcinogenicity studies are not generally needed for
  endogenous substances.
 long-term carcinogenicity studies in rodent species
  should be considered for the other biotechnology
  products.
Summary of Types of Data Useful for Evaluation of Carcinogenicity Bioassay
                                     Protocols
Dose            General       Genotoxicity   Human and    Animal AUC   Human AUC
Selection       Toxicity                     Animal       a.           a.
Endpoint        Information                  Metabolism   (Unbound     (Unbound
Toxicity        √             A              m            ---          ---
(MTD)
Multiple of     √             √              √            √            √
Human
Exposure
(25 X)
Saturation of   √             A              m            √            ---
Absorption
of Drug
Related
Substances
MFD             √             A              m            ---          ---
Limit Dose      √             √              √            √            √
Pharmacodyn     √             A              m            ---          ---
amic
Effects
S1B GUIDELINE
1.OBJECTIVE:provides guidance on approaches for
  evaluating the carcinogenic potential of
  pharmaceuticals.
2.THE GUIDELINE:
      Preamble.
      Experimental approaches to testing for carcinogenic
       potential.
                                    CONTD…..
3.MECHANISTIC STUDIES: interpretation of tumor findings in a
    carcinogenicity study.
           Cellular changes.
           Biochemical measurements
           Additional genotoxicity testing
           Modified protocols
4.   CHOICE OF AN APPROPRIATE SPECIES FOR LONG TERM
     CARCINOGENICITY TESTING
           Information from surveys on pharmaceuticals.
           Potential to study mechanisms.
           Metabolic disposition.
           Practicality.
           Testing in more than one species.
           Exceptions.
5. EVALUATION OF CARCINOGENIC
  POTENTIAL :Evidence of tumorigenic
  effects of the drug in rodent models
  should be evaluated in light of the
  tumor incidence and latency
S1C_R2 GUIDELINE
• Introduction: Carcinogenicity studies for chemical
  agents
      • Dose selection based on Maximal Tolerated
        Dose(MTD)
      • MTD based on 3 months study
• For Pharmaceuticals
      • Dose selection based on several
        approaches including “high
        multiple”approach in case of low toxicity
        compounds in some regions
Dose selection in
        carcinogenicity studies
         Situation in the past
 Endpoints for high dose selection
       Default: Maximum Tolerated Dose
       With compounds of low toxicity:
• Europe/ Japan
       High dose selection > 100 times Maximum recommended
       human daily dose ( in mg/kg)
• USA
       Maximum Tolerated Dose
       All areas: maximum feasible dose (if applicable)
Dose selection in
       carcinogenicity studies
• High Doses selected should provide an exposure
       Allows an adequate margin of safety over the
        human therapeutic dose.
      . Is tolerated without significant chronic
        physiological dysfunction and is compatible with
        good survival
      Is guided by a comprehensive set of animal and
        human data.
      Permits data interpretation in the context of
        clinical use
Dose selection in
      carcinogenicity studies
        Rational approach
o General considerations
     1. Carcinogenicity studies in limited number of rat and mouse
        strains. Ideally the same metabolic profile
     2. Dose range studies for males and females
     3. Dose selection determined from >/= 90-day studies
     4. Appropriate dosing schedule based on clinical use and
        exposure patterns
     5. Ideally toxicity profile and dose-limiting toxicity to be defined
     6. Changes in metabolic profiles over time should be
        understood
Toxicity endpoints
  harmonized definition of MTD
Operational definition
• The top dose or maximum tolerated dose is that which is
  predicted to produce a minimum toxic effect over the
  course of the carcinogenicity study.
• Such an effect can be predicted from a 90-day dose
  range finding study in which minimal toxicity is observed.
• Factors to consider are alterations in physiological
  function which would be predicted to alter the animal's
  normal life span or interfere with interpretation of the
  study.
• Such factors include: no more than 10% decrease in body
  weight gain relative to controls; target organ
  toxicity;significant alterations in clinical pathological
  parameters.
Pharmacokinetic endpoints
     criteria based on AUC
           comparison
• Systemic exposure representing large multiple of
  human exposure at Maximum Recommended
  Human dose
• Unbound plasma concentration most relevant
  measure and AUC most comprehensive
  parameter
• Selection of a high dose representing a 25- to-1
  exposure ratio of rodent to human plasma AUC
  of parent compound and/or metabolites
  pragmatic proposal
Pharmacokinetic high dose
             endpoint
             (ratio 25)
• Old situation
       • Only to be used in case of nongenotoxic
         compounds i.e. strictly no positive signals in
         battery
• New situation after revision
       • To be used in case of all human pharmaceuticals,
         also
       • if positive genotoxic signals are present
• Note: Real genotoxic compounds are not expected to be
  tested
Saturation of absorption in high
              dose
           selection
• If saturation of absorption measured by
  systemic availability occurs
• Mid and low doses should take into
  accountmetabolic and elimination
  pathways(might be different from the high
  dose)
Pharmacodynamic endpoints in
           high
       dose selection
• Utility of pharmaceuticals depend on their
  pharmacodynamic selectivity.
• Pharmacodynamic response might lead to
  disturbances in physiology or homeostasis
  e.g. hypotension, inhibition of blood
  clotting, diabetis (?)
Maximum feasible dose
• Currently: 5% of the diet in case of oral
  administration via the diet.
• When other routes of administration (e.g.
  dermal) are appropriate the limit might be based
  on practicality and local tolerance.
• It is to be expected that because of the
  introduction of the pharmacokinetic endpoint,
  the maximum feasible dose will used less as an
  endpoint.
Limit Dose
As a general guidance the high dose should not be higher
   than 1500 mg/kg/day.The limit is applicable in all cases
   where the maximum human dose does not exceed
   500mg/day.
Exposure in animals should be compared to humans and
   should be an order of magnitude higher.
If the human dose is higher than 500 mg/day the dose
   might be increased to the maximum feasible dose.
• Additional endpoints in high dose selection:
   – Other endpoints might be used based on a scientific rationale
   – Such designs should be evaluatied based on their individual
     merits
Selection of middle and low
             doses in
     carcinogenicity studies
• Linearity of pharmacokinetics and saturation of
  metabolic pathways.
• Human exposure and therapeutic dose.
• Pharmacodynamic response in rodents.
• Alterations in normal rodent physiology.
• Mechanistic information and potential for
  threshold effects.
• The unpredictability of the progression of
  toxicity observed in short-term studies.
S2R1_STEP 4 GUIDELINES
GUIDANCE ON GENOTOXICITY TESTING AND DATA INTERPRETATION
  FOR PHARMACEUTICALS INTENDED FOR HUMAN USE
 OBJECTIVE:
     o purpose of the revision is to optimize the standard
       genetic toxicology battery for prediction of
       potential human risks
     o to provide guidance on interpretation of results,
       with the ultimate goal of improving risk
       characterization for carcinogenic effects that have
       their basis in changes in the genetic material.
     o The focus of this guidance is testing of new “small
       molecule” drug substances, and the guidance does
       not apply to biologics.
General Principles

• Genotoxicity tests can be defined as in vitro and
  in vivo tests designed to detect compounds that
  induce genetic damage by various mechanisms.
• Tests enable hazard identification with respect to
  damage to DNA and its fixation.
• Fixation of damage to DNA in the form of gene
  mutations, larger scale chromosomal
STANDARD TEST BATTERY FOR
     GENOTOXICITY
• Rationale:
      Extensive reviews have shown that many compounds that
       are mutagenic in the bacterial reverse mutation (Ames) test
       are rodent carcinogens.
      Addition of in vitro mammalian tests increases sensitivity for
       detection of rodent carcinogens.
      broadens the spectrum of genetic events detected, but also
       decreases the specificity of prediction
General features of standard
        test battery
 Assessment of mutagenicity in a bacterial reverse gene mutation test This test has
  been shown to detect relevant genetic changes and the majority of genotoxic rodent
  and human carcinogens.
 Genotoxicity should also be evaluated in mammalian cells in vitro and/or in vivo.
 In vitro metaphase chromosome aberration assay, the in vitro micronucleus assay.
 In vivo test(s) are included in the test battery because some agents are mutagenic in
  vivo but not in vitro.
 In vitro and in vivo tests that measure chromosomal aberrations in metaphase cells
  can detect a wide spectrum of changes in chromosomal integrity.
Description of the Two Options
  for the Standard Battery
• Option 1
       i. A test for gene mutation in bacteria.
       ii. A cytogenetic test for chromosomal damage (the in vitro metaphase
            chromosome aberration test or in vitro micronucleus test), or an in vitro
            mouse lymphoma Tk gene mutation assay.
       iii. An in vivo test for genotoxicity, generally a test for chromosomal damage
            using rodent hematopoietic cells, either for micronuclei or for chromosomal
            aberrations in metaphase cells.


• Option 2
       i. A test for gene mutation in bacteria.
       ii. An in vivo assessment of genotoxicity with two different tissues, usually an
           assay for micronuclei using rodent hematopoietic cells and a second in vivo
           assay. Typically this would be a DNA strand breakage assay in liver.
Modifications to the Test
              Battery
o Exploratory Clinical Studies
        For certain exploratory clinical studies, fewer genotoxicity
         assays or different criteria for justification of the maximum
         dose in vivo might apply (see ICH M3(R2) guidance).

o Testing Compounds that are Toxic to Bacteria
         In cases where compounds are highly toxic to bacteria (e.g.,
          some antibiotics), the bacterial reverse mutation (Ames) test
          should still be carried out, just as cytotoxic compounds are
          tested in mammalian cells, because mutagenicity can occur
          at lower, less toxic concentrations. In such cases, any one of
          the in vitro mammalian cell assays should also be done, i.e.,
          Option 1 should be followed.
                                                CONTD…
o Compounds Bearing Structural Alerts for Genotoxic Activity
       . Structurally alerting compounds (Note 5) are usually
        detectable in the standard testbattery since the majority of
        “structural alerts” are defined in relation to bacterial
        mutagenecity.

o Limitations to the Use of In Vivo Tests:
        compounds for which many in vivo tests (typically in bone
          marrow, blood or liver) do not provide additional useful
          information.
        compounds for which data on toxicokinetics or
          pharmacokinetics indicate that they they are not systemically
          absorbed and therefore are not available to the target
          tissues.
        Examples of such compounds are some radio imaging agents,
          aluminum based antacids, some compounds given by
          inhalation, and some dermally or other topically applied
          pharmaceuticals.
Detection of Germ Cell
          Mutagens
• Most germ cell mutagens are likely to be
  detected as genotoxic in somatic cell tests
  so that negative results of in vivo somatic
  cell genotoxicity tests generally indicate
  the absence of germ cell effects.
RECOMMENDATIONS FOR IN
       VITRO TESTS
 Test Repetition and Interpretation: routine testing of
  drugs with standard, widely used genotoxicity tests does
  not call for replication. Tests are sufficiently well
  characterized and have sufficient internal controls.
 Recommended Protocol for the Bacterial Mutation
  Assay:protocols is given in the OECD guideline (1997) and
  the IWGT report (Gatehouse et al., 1994).
        CONTD……
 SELECTION OF TOP DOSE LEVEL-
    Maximum dose level-maximum dose level
     recommended is 5000 μg/plate when not limited by
     solubility or cytotoxicity.
    Limit of solubility- For bacterial cultures, precipitating
     doses are scored provided precipitate does not
     interfere with scoring, toxicity is not limiting, and the
     top concentration does not exceed 5000 μg/plate.
    Limit of cytotoxicity-In the Ames test, the doses
     scored should show evidence of significant toxicity,
     but without exceeding a top dose of 5000 μg/plate.
     Toxicity might be detected by a reduction in the
     number of revertants, and/or clearing or diminution
     of the background lawn.
 Study Design/Test Protocol-recommended set of
  bacterial strains (OECD) includes those that detect base
  substitution and frame shift mutations
    Salmonella typhimurium TA98
    Salmonella typhimurium TA100
    Salmonella typhimurium TA1535
    either Salmonella typhimurium TA1537 or TA97 or
     TA97a
    either Salmonella typhimurium TA102 or Escherichia
     coli WP2 uvrA or Escherichia coli WP2 uvrA (pKM101).
 Recommended Protocols for the Mammalian Cell Assays
      Selection of Top Concentration
         Maximum concentration-maximum top
           concentration recommended is 1 mM or 0.5
           mg/ml
         Limit of solubility- solubility is limiting, the
           maximum concentration, if not limited by
           cytotoxicity, should be the lowest
           concentration at which minimal precipitate is
           visible in cultures
         Cytotoxicity-in vitro cytogenetic assays for
           metaphase chromosome aberrations or for
           micronuclei, cytotoxicity should not exceed a
           reduction of about 50% in cell growth
                                 CONTD….
 Study Design/Test Protocols:
        test protocol should include the conduct of tests
         with and without metabolic activation, with
         appropriate positive and negative controls.
       Metaphase aberration assay, information on the
         ploidy status should be obtained by recording the
         incidence of polyploid.
 Positive Controls:
           Invitro mammalian cell tests for genetic toxicity
             are sufficiently standardized.
            Use of positive controls can generally be
             confined to a positive control with metabolic
             activation.
RECOMMENDATIONS FOR IN
       VIVO TESTS
 Tests for the Detection of Chromosome Damage In Vivo:
          analysis of chromosomal aberrations or the
             measurement of micronucleated
             polychromatic erythrocytes in bone marrow
             cells
          in vivo is considered appropriate for the
             detection of clastogens. Both rats and mice are
             considered appropriate for use in the bone
             marrow micronucleus
Other In Vivo Genotoxicity Tests: Type of
 effect seen in vitro and any knowledge of
 the mechanism can help guide the choice
 of in vivo assay. Investigation of
 chromosomal aberrations or of gene
 mutations in endogenous genes is not
 feasible with standard methods in most
 tissues.
Dose Selection for In Vivo
              Assays
• Short-Term Studies:
        • Top dose recommended for genotoxicity assays is a
          limit dose of 2000 mg/kg.
        • if this is tolerated, or a maximum tolerated dose
          defined as the dose producing signs of toxicity such
          that higher dose levels, based on the same dosing
          regimen, would be expected to produce lethality.
• Multiple Administration Studies:In vivo genotoxicity test
  is integrated into a multiple administration toxicology
  study. When carrying out follow-up studies to address
  any indication of genotoxicity, or when using Option 2
  with no in vitro mammalian cell assay, several factors
  should be evaluated to determine whether the top dose
  is appropriate for genotoxicity evaluation
Testing Compounds that are
    Toxic for Blood or Bone
            Marrow
Compounds that induce aneuploidy, such as potent spindle
  poisons, are detectable in in vivo micronucleus assays in
  bone marrow or blood only within a narrow range of
  doses approaching toxic doses.
 Early blood sampling (at 3-4 days) is advisable when there
  are marked increases in toxicity with increasing
  treatment time.
 An in vitro mammalian cell micronucleus assay.
 An acute bone marrow micronucleus assay
GUIDANCE ON EVALUATION OF
FOLLOW-UP TEST STRATEGIES
 In vitro test system generates both false negative
  and positive results to predict rodent
  carcinogenicity.
 Genotoxicity test batteries detect carcinogens
  that are act primarily via a mechanism involving
  direct genetic damage.
      • ASSESSEMENT OF BIOLOGICAL RELEVANCE:Small increases in
        apparent genotoxicity in vitro or in vivo should first be
        assessed for reproducibility and biological significance
• Evaluation of Results Obtained in In Vitro Tests:
        in a Bacterial Mutation Assay.
        in Mammalian Cell Assays.
        Evaluation of In Vitro Negative Results.
• Evaluation of Results Obtained from In Vivo Tests: If the
  in vivo and in vitro results do not agree, then the
  difference should be considered/explained on a case-by-
  case basis. In vivo genotoxicity tests also have the
  potential to give misleading positive results that do not
  indicate true genotoxicity.
S3A_GUIDLINE
THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY
  STUDIES:
Guidance concerns toxicokinetics with respect to the
  development of pharmaceutical products intended for
  use in human subjects.
Toxicokinetics is an integral component in the conduct of
  non-clinical toxicity studies or in specially designed
  supportive studies.
TOXICOKINETICS
• provide a means of obtaining multiple dose
  pharmacokinetic data in the test species.
• integral part of the non-clinical testing
  programme.
• enhance the value of the toxicological data
  generated.
• involves continuous feed-back between non-
  clinical and clinical studies.
Objectives of Toxicokinetics
• Primary objective of toxicokinetics 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 (Note 1) 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.
General principles
• Introduction:Toxicokinetic studies retrospectively
  designed to generate specific sets of data under
  conditions which mimic those of the toxicity studies
  should also conform to GLP when they are necessary for
  the evaluation of safety.
• Quantification of exposure:Provides an assessment of
  the burden on the test species. Assists in the
  interpretation of similarities and differences in toxicity
  across species, dose groups and sexes exposure might be
  represented by plasma (serum or blood) concentrations
  or the AUCs of parent compound and/or metabolite(s) .
• Justification of time points for sampling : Time points for
  collecting body fluids in concomitant toxicokinetic studies
  should be as frequent, but not so frequent as to interfere
  with the normal conduct of the study or to cause undue
  physiological stress to the animals.
• Setting of dose levels in order to produce adequate
  exposure:The setting of dose levels in toxicity studies is
  largely governed by the toxicology findings and the
  pharmacodynamic responses of the test species.
       • Low dose levels:no-toxic-effect dose level
       • Intermediate dose levels:exposure at lower (or
          higher) dose levels dependent upon the objectives
          of the toxicity study
       • High dose levels : toxicity studies determined by
          toxicological considerations
• Extent of exposure assessment in toxicity
  studies:Systemic exposure is estimated in an appropriate
  number of animals and dose groups to provide a basis for
  risk assessment.Samples for the generation of
  toxicokinetic data may be collected from main study
  animals.
• Complicating factors in exposure interpretation: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
         .Pharmacological activity of metabolites, the
         toxicology of metabolites.
• Route of administration:Inhalation, topical or parenteral delivery,
  should be based on the pharmacokinetic properties of the substance
  administered by the intended route. oral formulation may
  subsequently be developed for intravenous administration.
• 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
• Statistical evaluation of data:Data allow
  representative assessment of the
  exposure.Calculation of mean or median values
  and estimates of variability.
• Analytical methods:
     • should be specific for the entity to be measured
       and of an adequate accuracy and precision.
     • choice of analyte and the matrix to be assayed
Toxicokinetics in areas of
    toxicity testing - specific
             aspects
• Introduction: In this the frequency of exposure
  monitoring or profiling may be extended or
  reduced where necessary.
• Single-dose toxicity studies:Studies are
  performed in early phase of development before
  a bioanalytical method has been developed and
  toxicokinetic monitoring of these studies is not
  normally possible.
• Repeated-dose toxicity studies: Treatment
  regimen and species should be selected
  whenever possible. 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.
• 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.
• Carcinogenicity ( oncogenicity ) studies1
     Sighting or dose-ranging studies.
      The main studies
• Reproductive toxicity studies2:
Introduction :Preferable to information on
  pharmacokinetics before initiating
  reproduction studies. Toxicokinetic
  monitoring in reproductive toxicity studies
  valuable in some instances, especially with
  compounds with low toxicity.
• Fertility studies : Need to monitor on
  studies depend on the dosing regimen
  used and the information already available
  from earlier studies.
• Studies in pregnant and lactating animals:
  Consideration should be given to the
  possibility that the kinetics will differ in
  pregnant and non-pregnant
  animalsToxicokinetics involve exposure
  assessment of dams, embryos, foetuses or
  newborn at specified days .
REPEATED DOSE TISSUE
        DISTRIBUTION
STUDIES(S3B_guideline oct1994)
 • This study is required when appropriate
   data cannot be derived from other sources
 • A comprehensive knowledge ofabsorption,
   distribution, metabolism and elimination
   of a compound is important for the
   interpretation of pharmacology and
   toxicology studies.
 • useful for designing toxicology and
   pharmacology studies
S4_guideline( DURATION OF
    CHRONIC TOXICITY TESTING IN
        ANIMALS Sep. 1998)
RODENT AND NON RODENT TOXICITY
 TESTING :
• OBJECTIVE:safety evaluation of a medicinal
  product .
• SCOPE: guidance has been prepared for the
  development of medicinal products with the
  exception of those already covered by the ICH
  Guideline on Safety Studies for Biotechnological
  Products
•
BACKGROUND
• chronic toxicity testing in non-rodents, there were
  different approaches to the duration of testing.
• The lack of harmonised duration led to the need for
  pharmaceutical companies to perform partially
  duplicative studies for both 6 and 12 months duration
  when developing new medicinal products.
• Studies of 12 months duration are usually not necessary
  and studies of shorter than 9 months duration may be
  sufficient
• In the EU, studies of 6 months duration in non-rodents
  are acceptable according to Council Directive
  75/318/EEC, as amended. To avoid duplication, where
  studies with a longer duration have been conducted, it
  would not be necessary to conduct a study of 6 months
GUIDANCE ON DURATION OF
 CHRONIC TOXICITY TESTING FOR
 TRIPARTITE DEVELOPMENT PLAN
• extensive analysis and review of the above mentioned
  data in non-rodents and based upon the achievements of
  ICH1 for testing in rodents, and so as to avoid duplication
  and follow a single development plan for chronic toxicity
  testing of new medicinal products, the following studies
  are considered acceptable for submission in the 3
  Regions:
• 1) Rodents : a study of 6 months duration;
• 2) Non-rodents : a study of nine months duration.
S5(R2)_guideline
DETECTION OF TOXICITY TO REPRODUCTION
  FOR MEDICINAL PRODUCTS & TOXICITY TO
  MALE FERTILITY: This guideline attempts to
  consolidate a strategy based on study designs currently in
  use for testing of medicinal products;
• It should encourage the full assessment on the safety of
  chemicals on the development of the offspring.
• It is perceived that tests in which animals are treated
  during defined stages of reproduction better reflect
  human exposure to medicinal products and allow more
  specific identification of stages at risk.
• While this approach may be useful for most medicines,
  long term exposure to low doses does occur and may be
  represented better by a one or two generation study
Aim of studies
• The aim of reproduction toxicity studies is
  to reveal any effect of one or more active
  substance(s) on mammalian reproduction.
  For this purpose both the investigations
  and the interpretation of the results should
  be related to all other pharmacological and
  toxicological data available to determine
  whether potential reproductive risks to
  humans are greater, lesser or equal to
  those posed by other toxicological
  manifestations.
2. ANIMAL CRITERIA
• Selection and number of species: Studies should be
  conducted in mammalian species. It is generally desirable
  to use the same species and strain as in other
  toxicological studies.
• Reasons for using rats as the predominant rodent species
  are practicality, comparability with other results obtained
  in this species and the large amount of background
  knowledge accumulated.
• In embryotoxicity studies only, a second mammalian
  species traditionally has been required, the rabbit being
  the preferred choice as a "non-rodent". Reasons for using
  rabbits in embryotoxicity studies include the extensive
  background knowledge that has accumulated, as well as
  availability and practicality.
Other test systems
• Other test systems have been developed and used in
  preliminary investigations ("pre-screening" or priority
  selection) and secondary testing.
• For preliminary investigation of a range of analogue
  series of substances it is essential that the potential
  outcome in whole animals is known for at least one
  member of the series to be studied (by inference, effects
  are expected). With this strategy substances can be
  selected for higher level testing.
• For secondary testing characterisation other test systems
  offer the possibility to study some of the observable
  developmental processes in detail, e.g. to reveal specific
  mechanisms of toxicity, to establish concentration-
  response relationships, to select 'sensitive periods', or to
  detect effects of defined metabolites
3. GENERAL RECOMMENDATIONS
       CONCERNING TREATMENT
• Dosages :Selection of dosages is one of the most
  critical issues in design of the reproductive
  toxicity study.
• The choice of the high dose should be based on
  data from all available studies (pharmacology,
  acute and chronic toxicity and kinetic studies,
  Note 7).
• A repeated dose toxicity study of about 2 - 4
  weeks duration provides a close approximation
  to the duration of treatment in segmental
  designs of reproductive studies
Route and frequency of
          administration.
• In general the route or routes of administration should be
  similar to those intended for human usage. One route of
  substance administration may be acceptable if it can be
  shown that a similar distribution (kinetic profile) results
  from different routes (Note 9).
• The usual frequency of administration is once daily but
  consideration should be given to use either more
  frequent or less frequent administration taking kinetic
  variables into account (see also Note 10).
STATISTICS
• Analysis of the statistics of a study is the means by which
  results are interpreted.
• The most important part of this analysis is to establish
  the relationship between the different variables and their
  distribution (descriptive statistics), since these determine
  how groups should be compared.
• The distributions of the endpoints observed in
  reproductive tests are usually non normal and extend
  from almost continuous.
DATA PRESENTATION

• The key to good reporting is the tabulation of
  individual values in a clear concise manner to
  account for every animal that was entered into
  the study.
• A reader should be able to follow the history of
  any individual animal from initiation to
  termination
• It should be able to deduce with ease the
  contribution that the individual has made to any
  group summary values.
PRECLINICAL SAFETY EVALUATION OF
    BIOTECHNOLOGY-DERIVED
PHARMACEUTICALS(S6_R1_guideline )
• Objectives:
  1) to identify an initial safe dose and subsequent
    dose escalation schemes in humans.
  2) to identify potential target organs for toxicity
    and for the study of whether such toxicity is
    reversible.
  3) to identify safety parameters for clinical
    monitoring.
SPECIFICATION OF THE TEST
        MATERIAL
• purification processes to remove
  impurities and contaminants
• to establish a preclinical testing program
  for their qualification
• Comparability can be evaluated on the
  basis of biochemical and biological
  characterisation
PRECLINICAL SAFETY
            TESTING
• OBJECTIVES: To define pharmacological and toxicological
  effects not only prior to initiation of human studies but
  throughout clinical development.
• Preclinical safety testing should consider:
   – 1) selection of the relevant animal species;
   – 2) age;
   – 3) physiological state;
• 4) the manner of delivery, including dose, route of
  administration, and treatment regimen; and
• 5) stability of the test material under the conditions of
  use
Biological
Activity/Pharmacodynamics
Evaluated using in vitro assays to determine which effects of
  the product may be related to clinical activity
The use of cell lines and/or primary cell cultures can be
  useful to examine the direct effects on cellular phenotype
  and proliferation.
• Animal Species/Model Selection: Safety evaluation
  programs should include the use of relevant species. A
  relevant species is one in which the test material is
  pharmacologically active species that express the desired
  epitope and demonstrate a similar tissue cross-reactivity
  profile as for human tissues.
• Number/Gender of Animals: No. of animals used per
  dose has a direct bearing on the ability to detect toxicity.
Administration/Dose
           Selection
Route and frequency of administration should be as close as
  possible to that proposed for clinical use.
• SPECIFIC CONSIDERATIONS:
   – Safety Pharmacology : measure functional indices of
     potential toxicity.The aim of the safety pharmacology
     studies should be to reveal any functional effects on
     the major physiological systems.
   – Pharmacokinetics and Toxicokinetics :
     Pharmacokinetic studies should, whenever possible,
     utilise preparations that are representative of that
     intended for toxicity testing and clinical use, and
     employ a route of administration
• Metabolism: degradation to small peptides
  and individual amino acids.
• Single Dose Toxicity Studies:useful data to
  describe the relationship of dose to
  systemic and/or local toxicity. These data
  can be used to select doses for repeated
  dose toxicity studies.
• Repeated Dose Toxicity Studies: The
  duration of repeated dose studies should
  be based on the intended duration of
  clinical exposure and disease indication.
Immunotoxicity Studies
• It includes assessment of potential
  immunogenicity.
• Biotechnology-derived pharmaceuticals
  are intended to stimulate or suppress the
  immune system and may affect not only
  humoral but also cell-mediated immunity

Más contenido relacionado

La actualidad más candente

ICH: Introduction, objectives & guidelines: A brief insight.
ICH: Introduction, objectives & guidelines: A brief insight.ICH: Introduction, objectives & guidelines: A brief insight.
ICH: Introduction, objectives & guidelines: A brief insight.RxVichuZ
 
Investigational New drug application [INDA]
Investigational New drug application [INDA]Investigational New drug application [INDA]
Investigational New drug application [INDA]Sagar Savale
 
institutional review board and independent ethics committee
institutional review board and independent ethics committeeinstitutional review board and independent ethics committee
institutional review board and independent ethics committeeMOHAMMAD ASIM
 
Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...
Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...
Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...Dinesh Gangoda
 
NEW DRUG APPLICATION
NEW DRUG APPLICATIONNEW DRUG APPLICATION
NEW DRUG APPLICATIONSACHIN C P
 
Pharmacovigilance program of India (PvPI)
Pharmacovigilance program of India (PvPI)Pharmacovigilance program of India (PvPI)
Pharmacovigilance program of India (PvPI)SnehaKhandale1
 
Institutional review board/Independent ethics Committee
Institutional review board/Independent ethics CommitteeInstitutional review board/Independent ethics Committee
Institutional review board/Independent ethics CommitteeChintamBaladattaSai
 
Drug regulatory authority
Drug regulatory authority Drug regulatory authority
Drug regulatory authority Rahul Gawande
 
Q1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS
Q1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTSQ1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS
Q1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTSBhaumik Bavishi
 
Regulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy IIRegulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy IIJafarali Masi
 
Investigational New Drug Application
Investigational New Drug ApplicationInvestigational New Drug Application
Investigational New Drug ApplicationSuhas Reddy C
 
7.Safety Monitoring in Clinical Trails.pptx
7.Safety Monitoring in Clinical Trails.pptx7.Safety Monitoring in Clinical Trails.pptx
7.Safety Monitoring in Clinical Trails.pptxbrahmaiahmph
 

La actualidad más candente (20)

Schedule y
Schedule ySchedule y
Schedule y
 
ICH: Introduction, objectives & guidelines: A brief insight.
ICH: Introduction, objectives & guidelines: A brief insight.ICH: Introduction, objectives & guidelines: A brief insight.
ICH: Introduction, objectives & guidelines: A brief insight.
 
Investigational New drug application [INDA]
Investigational New drug application [INDA]Investigational New drug application [INDA]
Investigational New drug application [INDA]
 
Ctd ppt
Ctd pptCtd ppt
Ctd ppt
 
institutional review board and independent ethics committee
institutional review board and independent ethics committeeinstitutional review board and independent ethics committee
institutional review board and independent ethics committee
 
CDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATION
CDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATIONCDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATION
CDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATION
 
CONTRACT RESEARCH ORGANIZATION
CONTRACT RESEARCH ORGANIZATIONCONTRACT RESEARCH ORGANIZATION
CONTRACT RESEARCH ORGANIZATION
 
ICH GUIDELINES
ICH GUIDELINESICH GUIDELINES
ICH GUIDELINES
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
 
ICH Guidelines Q1 - Q10
ICH Guidelines Q1 - Q10ICH Guidelines Q1 - Q10
ICH Guidelines Q1 - Q10
 
Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...
Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...
Guidelines for Preparation of Documents, Clinical Study Report Clinical Trial...
 
NEW DRUG APPLICATION
NEW DRUG APPLICATIONNEW DRUG APPLICATION
NEW DRUG APPLICATION
 
Pharmacovigilance program of India (PvPI)
Pharmacovigilance program of India (PvPI)Pharmacovigilance program of India (PvPI)
Pharmacovigilance program of India (PvPI)
 
Institutional review board/Independent ethics Committee
Institutional review board/Independent ethics CommitteeInstitutional review board/Independent ethics Committee
Institutional review board/Independent ethics Committee
 
Drug regulatory authority
Drug regulatory authority Drug regulatory authority
Drug regulatory authority
 
Schedule y
Schedule ySchedule y
Schedule y
 
Q1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS
Q1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTSQ1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS
Q1A(R2): STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS
 
Regulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy IIRegulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy II
 
Investigational New Drug Application
Investigational New Drug ApplicationInvestigational New Drug Application
Investigational New Drug Application
 
7.Safety Monitoring in Clinical Trails.pptx
7.Safety Monitoring in Clinical Trails.pptx7.Safety Monitoring in Clinical Trails.pptx
7.Safety Monitoring in Clinical Trails.pptx
 

Destacado

Ich guidelines seminar
Ich guidelines seminarIch guidelines seminar
Ich guidelines seminarMd Gayasuddin
 
Design And Conduct Safety Pharmacology And Toxicology Study For Pharmaceuticals
Design And Conduct Safety Pharmacology And Toxicology Study For PharmaceuticalsDesign And Conduct Safety Pharmacology And Toxicology Study For Pharmaceuticals
Design And Conduct Safety Pharmacology And Toxicology Study For PharmaceuticalsProf. Dr. Basavaraj Nanjwade
 
Pharmaceutical industrial safety
Pharmaceutical industrial safetyPharmaceutical industrial safety
Pharmaceutical industrial safetyNamdeo Shinde
 
Pharmaceutical manufacturing health safety 2-
Pharmaceutical manufacturing  health  safety  2-Pharmaceutical manufacturing  health  safety  2-
Pharmaceutical manufacturing health safety 2-aameerkahn
 
Industrial hazards and safety measures
Industrial hazards and safety measuresIndustrial hazards and safety measures
Industrial hazards and safety measuresBiocon ltd
 
Clinical research overview
Clinical research overviewClinical research overview
Clinical research overviewAcri India
 
Interpretation of Low-Incidence Findings in Developmental and Reproductive To...
Interpretation of Low-Incidence Findings in Developmental and Reproductive To...Interpretation of Low-Incidence Findings in Developmental and Reproductive To...
Interpretation of Low-Incidence Findings in Developmental and Reproductive To...Joseph Holson
 
Current Regulatory Requirements in Developmental and Reproductive Toxicity As...
Current Regulatory Requirements in Developmental and Reproductive Toxicity As...Current Regulatory Requirements in Developmental and Reproductive Toxicity As...
Current Regulatory Requirements in Developmental and Reproductive Toxicity As...Joseph Holson
 
Clinical response evaluation dr.varun
Clinical response evaluation dr.varunClinical response evaluation dr.varun
Clinical response evaluation dr.varunVarun Goel
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelinesAnshul2593
 
REGULATORY SIGNIFICANCE AND SALIENT FEATURES OF GOOD LABORATORY PRACTICE (GLP...
REGULATORY SIGNIFICANCE AND SALIENT FEATURES OFGOOD LABORATORY PRACTICE (GLP...REGULATORY SIGNIFICANCE AND SALIENT FEATURES OFGOOD LABORATORY PRACTICE (GLP...
REGULATORY SIGNIFICANCE AND SALIENT FEATURES OF GOOD LABORATORY PRACTICE (GLP...Dr.Dhanya Nair
 
Comparison of RECIST 1.0 and 1.1 - Impact on Data Management
Comparison of RECIST 1.0 and 1.1 - Impact on Data ManagementComparison of RECIST 1.0 and 1.1 - Impact on Data Management
Comparison of RECIST 1.0 and 1.1 - Impact on Data ManagementKevin Shea
 
OECD and genotoxicity guidelines
OECD and genotoxicity guidelinesOECD and genotoxicity guidelines
OECD and genotoxicity guidelinesSandhya Talla
 
International Guidelines and Regulatory Agencies for Toxicity Studies
International Guidelines and Regulatory Agencies for Toxicity StudiesInternational Guidelines and Regulatory Agencies for Toxicity Studies
International Guidelines and Regulatory Agencies for Toxicity StudiesSuneal Saini
 

Destacado (20)

Ich guidelines seminar
Ich guidelines seminarIch guidelines seminar
Ich guidelines seminar
 
Design And Conduct Safety Pharmacology And Toxicology Study For Pharmaceuticals
Design And Conduct Safety Pharmacology And Toxicology Study For PharmaceuticalsDesign And Conduct Safety Pharmacology And Toxicology Study For Pharmaceuticals
Design And Conduct Safety Pharmacology And Toxicology Study For Pharmaceuticals
 
ICH
ICHICH
ICH
 
Pharmaceutical industrial safety
Pharmaceutical industrial safetyPharmaceutical industrial safety
Pharmaceutical industrial safety
 
Pharmaceutical manufacturing health safety 2-
Pharmaceutical manufacturing  health  safety  2-Pharmaceutical manufacturing  health  safety  2-
Pharmaceutical manufacturing health safety 2-
 
Industrial hazard ppt
Industrial hazard pptIndustrial hazard ppt
Industrial hazard ppt
 
Carcinogenicity testing
Carcinogenicity testingCarcinogenicity testing
Carcinogenicity testing
 
Industrial hazards and safety measures
Industrial hazards and safety measuresIndustrial hazards and safety measures
Industrial hazards and safety measures
 
Clinical Trials - An Introduction
Clinical Trials - An IntroductionClinical Trials - An Introduction
Clinical Trials - An Introduction
 
Clinical research overview
Clinical research overviewClinical research overview
Clinical research overview
 
Interpretation of Low-Incidence Findings in Developmental and Reproductive To...
Interpretation of Low-Incidence Findings in Developmental and Reproductive To...Interpretation of Low-Incidence Findings in Developmental and Reproductive To...
Interpretation of Low-Incidence Findings in Developmental and Reproductive To...
 
Current Regulatory Requirements in Developmental and Reproductive Toxicity As...
Current Regulatory Requirements in Developmental and Reproductive Toxicity As...Current Regulatory Requirements in Developmental and Reproductive Toxicity As...
Current Regulatory Requirements in Developmental and Reproductive Toxicity As...
 
Full recist violet
Full recist violetFull recist violet
Full recist violet
 
Clinical response evaluation dr.varun
Clinical response evaluation dr.varunClinical response evaluation dr.varun
Clinical response evaluation dr.varun
 
Recist1.1
Recist1.1Recist1.1
Recist1.1
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
 
REGULATORY SIGNIFICANCE AND SALIENT FEATURES OF GOOD LABORATORY PRACTICE (GLP...
REGULATORY SIGNIFICANCE AND SALIENT FEATURES OFGOOD LABORATORY PRACTICE (GLP...REGULATORY SIGNIFICANCE AND SALIENT FEATURES OFGOOD LABORATORY PRACTICE (GLP...
REGULATORY SIGNIFICANCE AND SALIENT FEATURES OF GOOD LABORATORY PRACTICE (GLP...
 
Comparison of RECIST 1.0 and 1.1 - Impact on Data Management
Comparison of RECIST 1.0 and 1.1 - Impact on Data ManagementComparison of RECIST 1.0 and 1.1 - Impact on Data Management
Comparison of RECIST 1.0 and 1.1 - Impact on Data Management
 
OECD and genotoxicity guidelines
OECD and genotoxicity guidelinesOECD and genotoxicity guidelines
OECD and genotoxicity guidelines
 
International Guidelines and Regulatory Agencies for Toxicity Studies
International Guidelines and Regulatory Agencies for Toxicity StudiesInternational Guidelines and Regulatory Agencies for Toxicity Studies
International Guidelines and Regulatory Agencies for Toxicity Studies
 

Similar a What is ICH and its Safety Guidelines

Acute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsAcute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsSwaroopaNallabariki
 
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 PRECLINICALTMU
 
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 ichAnimatedWorld
 
toxicokinetics and saturation kinetics
toxicokinetics and saturation kineticstoxicokinetics and saturation kinetics
toxicokinetics and saturation kineticspharmacistnitish
 
Chronic Toxicity Study (OECD TG-452).pptx
Chronic Toxicity Study (OECD TG-452).pptxChronic Toxicity Study (OECD TG-452).pptx
Chronic Toxicity Study (OECD TG-452).pptxSIRAJUDDIN MOLLA
 
Approaches of drug discovery
Approaches of drug discoveryApproaches of drug discovery
Approaches of drug discoveryGaurav Sharma
 
Toxicological Approach to Drug Discovery
Toxicological Approach to Drug DiscoveryToxicological Approach to Drug Discovery
Toxicological Approach to Drug DiscoverySuhas Reddy C
 
Pre clinical studies
Pre clinical studiesPre clinical studies
Pre clinical studiesKirsha K S
 
Toxicological approach for drug discovery
Toxicological approach for drug discovery Toxicological approach for drug discovery
Toxicological approach for drug discovery Dr Duggirala Mahendra
 
Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...
Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...
Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...AMIT KUMAR
 
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and  Population PK and Its' ValueUPDATED-Early Phase Drug Developmetn and  Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and Population PK and Its' ValueE. Dennis Bashaw
 
Current Pharmacology & Toxicology Guidlines For Pharmaceutical Industry
Current Pharmacology & Toxicology Guidlines For Pharmaceutical IndustryCurrent Pharmacology & Toxicology Guidlines For Pharmaceutical Industry
Current Pharmacology & Toxicology Guidlines For Pharmaceutical IndustryProf. Dr. Basavaraj Nanjwade
 
Safety guidelines- S3A and S3B
Safety guidelines- S3A and S3BSafety guidelines- S3A and S3B
Safety guidelines- S3A and S3Banshagrawal2121
 
1 Phases of clinical trial.pptx
1 Phases of clinical trial.pptx1 Phases of clinical trial.pptx
1 Phases of clinical trial.pptxDr Abisha T
 

Similar a What is ICH and its Safety Guidelines (20)

Acute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsAcute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animals
 
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
 
toxicokinetics and saturation kinetics
toxicokinetics and saturation kineticstoxicokinetics and saturation kinetics
toxicokinetics and saturation kinetics
 
Extrapolation.pptx
Extrapolation.pptxExtrapolation.pptx
Extrapolation.pptx
 
Phase 1 protocol
Phase 1 protocolPhase 1 protocol
Phase 1 protocol
 
Chronic Toxicity Study (OECD TG-452).pptx
Chronic Toxicity Study (OECD TG-452).pptxChronic Toxicity Study (OECD TG-452).pptx
Chronic Toxicity Study (OECD TG-452).pptx
 
Approaches of drug discovery
Approaches of drug discoveryApproaches of drug discovery
Approaches of drug discovery
 
Toxicological Approach to Drug Discovery
Toxicological Approach to Drug DiscoveryToxicological Approach to Drug Discovery
Toxicological Approach to Drug Discovery
 
MMM PPT.pptx
MMM PPT.pptxMMM PPT.pptx
MMM PPT.pptx
 
Pre clinical studies
Pre clinical studiesPre clinical studies
Pre clinical studies
 
Toxicological approach for drug discovery
Toxicological approach for drug discovery Toxicological approach for drug discovery
Toxicological approach for drug discovery
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...
Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...
Ich (s5 r2) The International Council for Harmonisationof Technical Requireme...
 
Dose selection
Dose selectionDose selection
Dose selection
 
Ich guidlines
Ich guidlinesIch guidlines
Ich guidlines
 
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and  Population PK and Its' ValueUPDATED-Early Phase Drug Developmetn and  Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
 
Current Pharmacology & Toxicology Guidlines For Pharmaceutical Industry
Current Pharmacology & Toxicology Guidlines For Pharmaceutical IndustryCurrent Pharmacology & Toxicology Guidlines For Pharmaceutical Industry
Current Pharmacology & Toxicology Guidlines For Pharmaceutical Industry
 
Safety guidelines- S3A and S3B
Safety guidelines- S3A and S3BSafety guidelines- S3A and S3B
Safety guidelines- S3A and S3B
 
1 Phases of clinical trial.pptx
1 Phases of clinical trial.pptx1 Phases of clinical trial.pptx
1 Phases of clinical trial.pptx
 

Último

ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTiammrhaywood
 
Karra SKD Conference Presentation Revised.pptx
Karra SKD Conference Presentation Revised.pptxKarra SKD Conference Presentation Revised.pptx
Karra SKD Conference Presentation Revised.pptxAshokKarra1
 
USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...
USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...
USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...Postal Advocate Inc.
 
Grade 9 Q4-MELC1-Active and Passive Voice.pptx
Grade 9 Q4-MELC1-Active and Passive Voice.pptxGrade 9 Q4-MELC1-Active and Passive Voice.pptx
Grade 9 Q4-MELC1-Active and Passive Voice.pptxChelloAnnAsuncion2
 
Inclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdf
Inclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdfInclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdf
Inclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdfTechSoup
 
Q4 English4 Week3 PPT Melcnmg-based.pptx
Q4 English4 Week3 PPT Melcnmg-based.pptxQ4 English4 Week3 PPT Melcnmg-based.pptx
Q4 English4 Week3 PPT Melcnmg-based.pptxnelietumpap1
 
Roles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceRoles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceSamikshaHamane
 
What is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPWhat is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPCeline George
 
How to do quick user assign in kanban in Odoo 17 ERP
How to do quick user assign in kanban in Odoo 17 ERPHow to do quick user assign in kanban in Odoo 17 ERP
How to do quick user assign in kanban in Odoo 17 ERPCeline George
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Celine George
 
ACC 2024 Chronicles. Cardiology. Exam.pdf
ACC 2024 Chronicles. Cardiology. Exam.pdfACC 2024 Chronicles. Cardiology. Exam.pdf
ACC 2024 Chronicles. Cardiology. Exam.pdfSpandanaRallapalli
 
Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...Jisc
 
Gas measurement O2,Co2,& ph) 04/2024.pptx
Gas measurement O2,Co2,& ph) 04/2024.pptxGas measurement O2,Co2,& ph) 04/2024.pptx
Gas measurement O2,Co2,& ph) 04/2024.pptxDr.Ibrahim Hassaan
 
ENGLISH6-Q4-W3.pptxqurter our high choom
ENGLISH6-Q4-W3.pptxqurter our high choomENGLISH6-Q4-W3.pptxqurter our high choom
ENGLISH6-Q4-W3.pptxqurter our high choomnelietumpap1
 
Proudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxProudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxthorishapillay1
 
Field Attribute Index Feature in Odoo 17
Field Attribute Index Feature in Odoo 17Field Attribute Index Feature in Odoo 17
Field Attribute Index Feature in Odoo 17Celine George
 

Último (20)

ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
 
Karra SKD Conference Presentation Revised.pptx
Karra SKD Conference Presentation Revised.pptxKarra SKD Conference Presentation Revised.pptx
Karra SKD Conference Presentation Revised.pptx
 
USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...
USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...
USPS® Forced Meter Migration - How to Know if Your Postage Meter Will Soon be...
 
Grade 9 Q4-MELC1-Active and Passive Voice.pptx
Grade 9 Q4-MELC1-Active and Passive Voice.pptxGrade 9 Q4-MELC1-Active and Passive Voice.pptx
Grade 9 Q4-MELC1-Active and Passive Voice.pptx
 
Inclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdf
Inclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdfInclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdf
Inclusivity Essentials_ Creating Accessible Websites for Nonprofits .pdf
 
Q4 English4 Week3 PPT Melcnmg-based.pptx
Q4 English4 Week3 PPT Melcnmg-based.pptxQ4 English4 Week3 PPT Melcnmg-based.pptx
Q4 English4 Week3 PPT Melcnmg-based.pptx
 
Roles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceRoles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in Pharmacovigilance
 
What is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPWhat is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERP
 
Raw materials used in Herbal Cosmetics.pptx
Raw materials used in Herbal Cosmetics.pptxRaw materials used in Herbal Cosmetics.pptx
Raw materials used in Herbal Cosmetics.pptx
 
How to do quick user assign in kanban in Odoo 17 ERP
How to do quick user assign in kanban in Odoo 17 ERPHow to do quick user assign in kanban in Odoo 17 ERP
How to do quick user assign in kanban in Odoo 17 ERP
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
YOUVE_GOT_EMAIL_PRELIMS_EL_DORADO_2024.pptx
YOUVE_GOT_EMAIL_PRELIMS_EL_DORADO_2024.pptxYOUVE_GOT_EMAIL_PRELIMS_EL_DORADO_2024.pptx
YOUVE_GOT_EMAIL_PRELIMS_EL_DORADO_2024.pptx
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17
 
ACC 2024 Chronicles. Cardiology. Exam.pdf
ACC 2024 Chronicles. Cardiology. Exam.pdfACC 2024 Chronicles. Cardiology. Exam.pdf
ACC 2024 Chronicles. Cardiology. Exam.pdf
 
Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...Procuring digital preservation CAN be quick and painless with our new dynamic...
Procuring digital preservation CAN be quick and painless with our new dynamic...
 
Gas measurement O2,Co2,& ph) 04/2024.pptx
Gas measurement O2,Co2,& ph) 04/2024.pptxGas measurement O2,Co2,& ph) 04/2024.pptx
Gas measurement O2,Co2,& ph) 04/2024.pptx
 
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdfTataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
 
ENGLISH6-Q4-W3.pptxqurter our high choom
ENGLISH6-Q4-W3.pptxqurter our high choomENGLISH6-Q4-W3.pptxqurter our high choom
ENGLISH6-Q4-W3.pptxqurter our high choom
 
Proudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxProudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptx
 
Field Attribute Index Feature in Odoo 17
Field Attribute Index Feature in Odoo 17Field Attribute Index Feature in Odoo 17
Field Attribute Index Feature in Odoo 17
 

What is ICH and its Safety Guidelines

  • 1.
  • 2. What is ICH?" • International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use" . ICH is a joint initiative involving both regulators and research- based industry representatives of the EU, Japan and the US in scientific and technical discussions of the testing procedures required to assess and ensure the safety, quality and efficacy of medicines.
  • 3. What is the purpose of ICH? • The objective of ICH is to increase international harmonization of technical requirements to ensure that safe, effective, and high quality medicines are developed and registered in the most efficient and cost- effective manner.
  • 5. Safety Guidelines • ICH has produced a comprehensive set of safety guidelines to uncover potential risks like carcinogenicity, genotoxicity and reprotoxicity. A recent breakthrough has been a non-clinical testing strategy for assessing the QT interval prolongation liability: the single most important cause of drug withdrawals in recent years
  • 6. S1 _guideline 14 November 2012 • Rodent Carcinogenicity Studies for Human Pharmaceuticals : A change to the current S1 harmonised Guidelines on rodent carcinogenicity testing is proposed to introduce a more comprehensive and integrated approach to addressing the risk of human carcinogenicity of pharmaceuticals, clarify and update, without compromising safety, the criteria for deciding whether the conduct of a two-year rodent carcinogenicity study of a given pharmaceutical would add value to this risk assessment.
  • 7. November 2012 • Proposal: It is expected that proposal will modify the ICH S1 Guideline, by advancing an approach in which the need for 2-year bioassays are not automatically triggered, but evaluated instead based on a case-by- case approach taking both predicted positive carcinogenicity study outcomes as well as negative outcomes into consideration for a scientifically justifiable waiver.
  • 8. S1A Guideline Need For Carcinogenicity Studies Of Pharmaceuticals  OBJECTIVES To identify a tumorigenic potential in animals  To assess the relevant risk in humans. studies also preceded our current understanding of tumorigenesis with non-genotoxic agents . To avoid the unnecessary use of animals in testing .
  • 9. HISTORICAL BACKGROUND • IN JAPAN (1990)  Guidelines for Toxicity Studies of Drugs Manual . carcinogenicity studies were needed if the clinical use was expected to be continuously for 6 months or longer. • In the United States  pharmaceuticals were tested in animals for their carcinogenic potential before widespread use in humans. • IN EUROPE  Rules Governing Medicinal Products in the European Community defined the circumstances when carcinogenicity studies were required.
  • 10. FACTORS FOR CARCINOGENICITY TESTING • DURATION & EXPOSURE: Carcinogenicity studies should be performed for any pharmaceutical whose expected clinical use is continuous for at least 6 months. Certain classes of compounds may not be used continuously over a minimum of 6 months but may be expected to be used repeatedly.
  • 11. • Cause for Concern: concern about their carcinogenic potential. Factors which could be considered may include:  previous demonstration of carcinogenic potential in the product class that is considered relevant to humans.  structure-activity relationship suggesting carcinogenic risk.  evidence of pre neoplastic lesions in repeated dose toxicity studies.  long-term tissue retention of parent compound or metabolite(s) resulting in local tissue reactions or other pathophysiological resposes.
  • 12. • Genotoxicity:  Unequivocally genotoxic compounds, in the absence of other data, are presumed to be trans-species carcinogens, implying a hazard to humans. Such compounds need not be subjected to long-term carcinogenicity studies.  if such a drug is intended to be administered chronically to humans a chronic toxicity study (up to one year) may be necessary to detect early tumorigenic effects.
  • 13. Indication and Patient Population: o When carcinogenicity studies are required they usually need to be completed before application for marketing approval. o Completed rodent carcinogenicity studies are not needed in advance of the conduct of large scale clinical trials, unless there is special concern for the patient population. o To treat certain serious diseases, carcinogenicity testing need not be conducted before market approval although these studies should be conducted post-approval.
  • 14. Route of Exposure:  should be the same as the intended clinical route when feasible.  carcinogenicity studies should only be conducted by a single route.  it is important that relevant organs for the clinical route (e.g., lung for inhalational agents) be adequately exposed to the test material.
  • 15. Extent of Systemic Exposure:  showing poor systemic exposure from topical routes in humans.  may not need studies by the oral route to assess the carcinogenic potential to internal organs.  Pharmaceuticals administered by the ocular route may not require carcinogenicity studies.  Evidence should be provided that there are no significant changes in pharmacokinetics, pharmacodynamics, or toxicity.
  • 16. Endogenous Peptides and Protein Substances or Their Analogs:  produced by chemical synthesis, by extraction/purification from an animal/human source or by biotechnological methods such as recombinant DNA technology.  Carcinogenicity studies are not generally needed for endogenous substances.  long-term carcinogenicity studies in rodent species should be considered for the other biotechnology products.
  • 17. Summary of Types of Data Useful for Evaluation of Carcinogenicity Bioassay Protocols Dose General Genotoxicity Human and Animal AUC Human AUC Selection Toxicity Animal a. a. Endpoint Information Metabolism (Unbound (Unbound Toxicity √ A m --- --- (MTD) Multiple of √ √ √ √ √ Human Exposure (25 X) Saturation of √ A m √ --- Absorption of Drug Related Substances MFD √ A m --- --- Limit Dose √ √ √ √ √ Pharmacodyn √ A m --- --- amic Effects
  • 18. S1B GUIDELINE 1.OBJECTIVE:provides guidance on approaches for evaluating the carcinogenic potential of pharmaceuticals. 2.THE GUIDELINE:  Preamble.  Experimental approaches to testing for carcinogenic potential. CONTD…..
  • 19. 3.MECHANISTIC STUDIES: interpretation of tumor findings in a carcinogenicity study.  Cellular changes.  Biochemical measurements  Additional genotoxicity testing  Modified protocols 4. CHOICE OF AN APPROPRIATE SPECIES FOR LONG TERM CARCINOGENICITY TESTING  Information from surveys on pharmaceuticals.  Potential to study mechanisms.  Metabolic disposition.  Practicality.  Testing in more than one species.  Exceptions.
  • 20. 5. EVALUATION OF CARCINOGENIC POTENTIAL :Evidence of tumorigenic effects of the drug in rodent models should be evaluated in light of the tumor incidence and latency
  • 21. S1C_R2 GUIDELINE • Introduction: Carcinogenicity studies for chemical agents • Dose selection based on Maximal Tolerated Dose(MTD) • MTD based on 3 months study • For Pharmaceuticals • Dose selection based on several approaches including “high multiple”approach in case of low toxicity compounds in some regions
  • 22. Dose selection in carcinogenicity studies Situation in the past  Endpoints for high dose selection  Default: Maximum Tolerated Dose  With compounds of low toxicity: • Europe/ Japan  High dose selection > 100 times Maximum recommended  human daily dose ( in mg/kg) • USA  Maximum Tolerated Dose  All areas: maximum feasible dose (if applicable)
  • 23. Dose selection in carcinogenicity studies • High Doses selected should provide an exposure  Allows an adequate margin of safety over the human therapeutic dose. . Is tolerated without significant chronic physiological dysfunction and is compatible with good survival Is guided by a comprehensive set of animal and human data. Permits data interpretation in the context of clinical use
  • 24. Dose selection in carcinogenicity studies Rational approach o General considerations 1. Carcinogenicity studies in limited number of rat and mouse strains. Ideally the same metabolic profile 2. Dose range studies for males and females 3. Dose selection determined from >/= 90-day studies 4. Appropriate dosing schedule based on clinical use and exposure patterns 5. Ideally toxicity profile and dose-limiting toxicity to be defined 6. Changes in metabolic profiles over time should be understood
  • 25. Toxicity endpoints harmonized definition of MTD Operational definition • The top dose or maximum tolerated dose is that which is predicted to produce a minimum toxic effect over the course of the carcinogenicity study. • Such an effect can be predicted from a 90-day dose range finding study in which minimal toxicity is observed. • Factors to consider are alterations in physiological function which would be predicted to alter the animal's normal life span or interfere with interpretation of the study. • Such factors include: no more than 10% decrease in body weight gain relative to controls; target organ toxicity;significant alterations in clinical pathological parameters.
  • 26. Pharmacokinetic endpoints criteria based on AUC comparison • Systemic exposure representing large multiple of human exposure at Maximum Recommended Human dose • Unbound plasma concentration most relevant measure and AUC most comprehensive parameter • Selection of a high dose representing a 25- to-1 exposure ratio of rodent to human plasma AUC of parent compound and/or metabolites pragmatic proposal
  • 27. Pharmacokinetic high dose endpoint (ratio 25) • Old situation • Only to be used in case of nongenotoxic compounds i.e. strictly no positive signals in battery • New situation after revision • To be used in case of all human pharmaceuticals, also • if positive genotoxic signals are present • Note: Real genotoxic compounds are not expected to be tested
  • 28. Saturation of absorption in high dose selection • If saturation of absorption measured by systemic availability occurs • Mid and low doses should take into accountmetabolic and elimination pathways(might be different from the high dose)
  • 29. Pharmacodynamic endpoints in high dose selection • Utility of pharmaceuticals depend on their pharmacodynamic selectivity. • Pharmacodynamic response might lead to disturbances in physiology or homeostasis e.g. hypotension, inhibition of blood clotting, diabetis (?)
  • 30. Maximum feasible dose • Currently: 5% of the diet in case of oral administration via the diet. • When other routes of administration (e.g. dermal) are appropriate the limit might be based on practicality and local tolerance. • It is to be expected that because of the introduction of the pharmacokinetic endpoint, the maximum feasible dose will used less as an endpoint.
  • 31. Limit Dose As a general guidance the high dose should not be higher than 1500 mg/kg/day.The limit is applicable in all cases where the maximum human dose does not exceed 500mg/day. Exposure in animals should be compared to humans and should be an order of magnitude higher. If the human dose is higher than 500 mg/day the dose might be increased to the maximum feasible dose. • Additional endpoints in high dose selection: – Other endpoints might be used based on a scientific rationale – Such designs should be evaluatied based on their individual merits
  • 32. Selection of middle and low doses in carcinogenicity studies • Linearity of pharmacokinetics and saturation of metabolic pathways. • Human exposure and therapeutic dose. • Pharmacodynamic response in rodents. • Alterations in normal rodent physiology. • Mechanistic information and potential for threshold effects. • The unpredictability of the progression of toxicity observed in short-term studies.
  • 33. S2R1_STEP 4 GUIDELINES GUIDANCE ON GENOTOXICITY TESTING AND DATA INTERPRETATION FOR PHARMACEUTICALS INTENDED FOR HUMAN USE  OBJECTIVE: o purpose of the revision is to optimize the standard genetic toxicology battery for prediction of potential human risks o to provide guidance on interpretation of results, with the ultimate goal of improving risk characterization for carcinogenic effects that have their basis in changes in the genetic material. o The focus of this guidance is testing of new “small molecule” drug substances, and the guidance does not apply to biologics.
  • 34. General Principles • Genotoxicity tests can be defined as in vitro and in vivo tests designed to detect compounds that induce genetic damage by various mechanisms. • Tests enable hazard identification with respect to damage to DNA and its fixation. • Fixation of damage to DNA in the form of gene mutations, larger scale chromosomal
  • 35. STANDARD TEST BATTERY FOR GENOTOXICITY • Rationale:  Extensive reviews have shown that many compounds that are mutagenic in the bacterial reverse mutation (Ames) test are rodent carcinogens.  Addition of in vitro mammalian tests increases sensitivity for detection of rodent carcinogens.  broadens the spectrum of genetic events detected, but also decreases the specificity of prediction
  • 36. General features of standard test battery  Assessment of mutagenicity in a bacterial reverse gene mutation test This test has been shown to detect relevant genetic changes and the majority of genotoxic rodent and human carcinogens.  Genotoxicity should also be evaluated in mammalian cells in vitro and/or in vivo.  In vitro metaphase chromosome aberration assay, the in vitro micronucleus assay.  In vivo test(s) are included in the test battery because some agents are mutagenic in vivo but not in vitro.  In vitro and in vivo tests that measure chromosomal aberrations in metaphase cells can detect a wide spectrum of changes in chromosomal integrity.
  • 37. Description of the Two Options for the Standard Battery • Option 1 i. A test for gene mutation in bacteria. ii. A cytogenetic test for chromosomal damage (the in vitro metaphase chromosome aberration test or in vitro micronucleus test), or an in vitro mouse lymphoma Tk gene mutation assay. iii. An in vivo test for genotoxicity, generally a test for chromosomal damage using rodent hematopoietic cells, either for micronuclei or for chromosomal aberrations in metaphase cells. • Option 2 i. A test for gene mutation in bacteria. ii. An in vivo assessment of genotoxicity with two different tissues, usually an assay for micronuclei using rodent hematopoietic cells and a second in vivo assay. Typically this would be a DNA strand breakage assay in liver.
  • 38. Modifications to the Test Battery o Exploratory Clinical Studies  For certain exploratory clinical studies, fewer genotoxicity assays or different criteria for justification of the maximum dose in vivo might apply (see ICH M3(R2) guidance). o Testing Compounds that are Toxic to Bacteria  In cases where compounds are highly toxic to bacteria (e.g., some antibiotics), the bacterial reverse mutation (Ames) test should still be carried out, just as cytotoxic compounds are tested in mammalian cells, because mutagenicity can occur at lower, less toxic concentrations. In such cases, any one of the in vitro mammalian cell assays should also be done, i.e., Option 1 should be followed. CONTD…
  • 39. o Compounds Bearing Structural Alerts for Genotoxic Activity  . Structurally alerting compounds (Note 5) are usually detectable in the standard testbattery since the majority of “structural alerts” are defined in relation to bacterial mutagenecity. o Limitations to the Use of In Vivo Tests:  compounds for which many in vivo tests (typically in bone marrow, blood or liver) do not provide additional useful information.  compounds for which data on toxicokinetics or pharmacokinetics indicate that they they are not systemically absorbed and therefore are not available to the target tissues.  Examples of such compounds are some radio imaging agents, aluminum based antacids, some compounds given by inhalation, and some dermally or other topically applied pharmaceuticals.
  • 40. Detection of Germ Cell Mutagens • Most germ cell mutagens are likely to be detected as genotoxic in somatic cell tests so that negative results of in vivo somatic cell genotoxicity tests generally indicate the absence of germ cell effects.
  • 41. RECOMMENDATIONS FOR IN VITRO TESTS  Test Repetition and Interpretation: routine testing of drugs with standard, widely used genotoxicity tests does not call for replication. Tests are sufficiently well characterized and have sufficient internal controls.  Recommended Protocol for the Bacterial Mutation Assay:protocols is given in the OECD guideline (1997) and the IWGT report (Gatehouse et al., 1994).  CONTD……
  • 42.  SELECTION OF TOP DOSE LEVEL-  Maximum dose level-maximum dose level recommended is 5000 μg/plate when not limited by solubility or cytotoxicity.  Limit of solubility- For bacterial cultures, precipitating doses are scored provided precipitate does not interfere with scoring, toxicity is not limiting, and the top concentration does not exceed 5000 μg/plate.  Limit of cytotoxicity-In the Ames test, the doses scored should show evidence of significant toxicity, but without exceeding a top dose of 5000 μg/plate. Toxicity might be detected by a reduction in the number of revertants, and/or clearing or diminution of the background lawn.
  • 43.  Study Design/Test Protocol-recommended set of bacterial strains (OECD) includes those that detect base substitution and frame shift mutations  Salmonella typhimurium TA98  Salmonella typhimurium TA100  Salmonella typhimurium TA1535  either Salmonella typhimurium TA1537 or TA97 or TA97a  either Salmonella typhimurium TA102 or Escherichia coli WP2 uvrA or Escherichia coli WP2 uvrA (pKM101).
  • 44.  Recommended Protocols for the Mammalian Cell Assays Selection of Top Concentration  Maximum concentration-maximum top concentration recommended is 1 mM or 0.5 mg/ml  Limit of solubility- solubility is limiting, the maximum concentration, if not limited by cytotoxicity, should be the lowest concentration at which minimal precipitate is visible in cultures  Cytotoxicity-in vitro cytogenetic assays for metaphase chromosome aberrations or for micronuclei, cytotoxicity should not exceed a reduction of about 50% in cell growth CONTD….
  • 45.  Study Design/Test Protocols:  test protocol should include the conduct of tests with and without metabolic activation, with appropriate positive and negative controls. Metaphase aberration assay, information on the ploidy status should be obtained by recording the incidence of polyploid.  Positive Controls: Invitro mammalian cell tests for genetic toxicity are sufficiently standardized.  Use of positive controls can generally be confined to a positive control with metabolic activation.
  • 46. RECOMMENDATIONS FOR IN VIVO TESTS  Tests for the Detection of Chromosome Damage In Vivo: analysis of chromosomal aberrations or the measurement of micronucleated polychromatic erythrocytes in bone marrow cells in vivo is considered appropriate for the detection of clastogens. Both rats and mice are considered appropriate for use in the bone marrow micronucleus
  • 47. Other In Vivo Genotoxicity Tests: Type of effect seen in vitro and any knowledge of the mechanism can help guide the choice of in vivo assay. Investigation of chromosomal aberrations or of gene mutations in endogenous genes is not feasible with standard methods in most tissues.
  • 48. Dose Selection for In Vivo Assays • Short-Term Studies: • Top dose recommended for genotoxicity assays is a limit dose of 2000 mg/kg. • if this is tolerated, or a maximum tolerated dose defined as the dose producing signs of toxicity such that higher dose levels, based on the same dosing regimen, would be expected to produce lethality. • Multiple Administration Studies:In vivo genotoxicity test is integrated into a multiple administration toxicology study. When carrying out follow-up studies to address any indication of genotoxicity, or when using Option 2 with no in vitro mammalian cell assay, several factors should be evaluated to determine whether the top dose is appropriate for genotoxicity evaluation
  • 49. Testing Compounds that are Toxic for Blood or Bone Marrow Compounds that induce aneuploidy, such as potent spindle poisons, are detectable in in vivo micronucleus assays in bone marrow or blood only within a narrow range of doses approaching toxic doses.  Early blood sampling (at 3-4 days) is advisable when there are marked increases in toxicity with increasing treatment time.  An in vitro mammalian cell micronucleus assay.  An acute bone marrow micronucleus assay
  • 50. GUIDANCE ON EVALUATION OF FOLLOW-UP TEST STRATEGIES  In vitro test system generates both false negative and positive results to predict rodent carcinogenicity.  Genotoxicity test batteries detect carcinogens that are act primarily via a mechanism involving direct genetic damage. • ASSESSEMENT OF BIOLOGICAL RELEVANCE:Small increases in apparent genotoxicity in vitro or in vivo should first be assessed for reproducibility and biological significance
  • 51. • Evaluation of Results Obtained in In Vitro Tests:  in a Bacterial Mutation Assay.  in Mammalian Cell Assays.  Evaluation of In Vitro Negative Results. • Evaluation of Results Obtained from In Vivo Tests: If the in vivo and in vitro results do not agree, then the difference should be considered/explained on a case-by- case basis. In vivo genotoxicity tests also have the potential to give misleading positive results that do not indicate true genotoxicity.
  • 52. S3A_GUIDLINE THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES: Guidance concerns toxicokinetics with respect to the development of pharmaceutical products intended for use in human subjects. Toxicokinetics is an integral component in the conduct of non-clinical toxicity studies or in specially designed supportive studies.
  • 53. TOXICOKINETICS • provide a means of obtaining multiple dose pharmacokinetic data in the test species. • integral part of the non-clinical testing programme. • enhance the value of the toxicological data generated. • involves continuous feed-back between non- clinical and clinical studies.
  • 54. Objectives of Toxicokinetics • Primary objective of toxicokinetics 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 (Note 1) 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.
  • 55. General principles • Introduction:Toxicokinetic studies retrospectively designed to generate specific sets of data under conditions which mimic those of the toxicity studies should also conform to GLP when they are necessary for the evaluation of safety. • Quantification of exposure:Provides an assessment of the burden on the test species. Assists in the interpretation of similarities and differences in toxicity across species, dose groups and sexes exposure might be represented by plasma (serum or blood) concentrations or the AUCs of parent compound and/or metabolite(s) .
  • 56. • Justification of time points for sampling : Time points for collecting body fluids in concomitant toxicokinetic studies should be as frequent, but not so frequent as to interfere with the normal conduct of the study or to cause undue physiological stress to the animals. • Setting of dose levels in order to produce adequate exposure:The setting of dose levels in toxicity studies is largely governed by the toxicology findings and the pharmacodynamic responses of the test species. • Low dose levels:no-toxic-effect dose level • Intermediate dose levels:exposure at lower (or higher) dose levels dependent upon the objectives of the toxicity study • High dose levels : toxicity studies determined by toxicological considerations
  • 57. • Extent of exposure assessment in toxicity studies:Systemic exposure is estimated in an appropriate number of animals and dose groups to provide a basis for risk assessment.Samples for the generation of toxicokinetic data may be collected from main study animals. • Complicating factors in exposure interpretation: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 .Pharmacological activity of metabolites, the toxicology of metabolites.
  • 58. • Route of administration:Inhalation, topical or parenteral delivery, should be based on the pharmacokinetic properties of the substance administered by the intended route. oral formulation may subsequently be developed for intravenous administration. • 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
  • 59. • Statistical evaluation of data:Data allow representative assessment of the exposure.Calculation of mean or median values and estimates of variability. • Analytical methods: • should be specific for the entity to be measured and of an adequate accuracy and precision. • choice of analyte and the matrix to be assayed
  • 60. Toxicokinetics in areas of toxicity testing - specific aspects • Introduction: In this the frequency of exposure monitoring or profiling may be extended or reduced where necessary. • Single-dose toxicity studies:Studies are performed in early phase of development before a bioanalytical method has been developed and toxicokinetic monitoring of these studies is not normally possible.
  • 61. • Repeated-dose toxicity studies: Treatment regimen and species should be selected whenever possible. 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. • 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.
  • 62. • Carcinogenicity ( oncogenicity ) studies1 Sighting or dose-ranging studies.  The main studies • Reproductive toxicity studies2: Introduction :Preferable to information on pharmacokinetics before initiating reproduction studies. Toxicokinetic monitoring in reproductive toxicity studies valuable in some instances, especially with compounds with low toxicity.
  • 63. • Fertility studies : Need to monitor on studies depend on the dosing regimen used and the information already available from earlier studies. • Studies in pregnant and lactating animals: Consideration should be given to the possibility that the kinetics will differ in pregnant and non-pregnant animalsToxicokinetics involve exposure assessment of dams, embryos, foetuses or newborn at specified days .
  • 64. REPEATED DOSE TISSUE DISTRIBUTION STUDIES(S3B_guideline oct1994) • This study is required when appropriate data cannot be derived from other sources • A comprehensive knowledge ofabsorption, distribution, metabolism and elimination of a compound is important for the interpretation of pharmacology and toxicology studies. • useful for designing toxicology and pharmacology studies
  • 65. S4_guideline( DURATION OF CHRONIC TOXICITY TESTING IN ANIMALS Sep. 1998) RODENT AND NON RODENT TOXICITY TESTING : • OBJECTIVE:safety evaluation of a medicinal product . • SCOPE: guidance has been prepared for the development of medicinal products with the exception of those already covered by the ICH Guideline on Safety Studies for Biotechnological Products •
  • 66. BACKGROUND • chronic toxicity testing in non-rodents, there were different approaches to the duration of testing. • The lack of harmonised duration led to the need for pharmaceutical companies to perform partially duplicative studies for both 6 and 12 months duration when developing new medicinal products. • Studies of 12 months duration are usually not necessary and studies of shorter than 9 months duration may be sufficient • In the EU, studies of 6 months duration in non-rodents are acceptable according to Council Directive 75/318/EEC, as amended. To avoid duplication, where studies with a longer duration have been conducted, it would not be necessary to conduct a study of 6 months
  • 67. GUIDANCE ON DURATION OF CHRONIC TOXICITY TESTING FOR TRIPARTITE DEVELOPMENT PLAN • extensive analysis and review of the above mentioned data in non-rodents and based upon the achievements of ICH1 for testing in rodents, and so as to avoid duplication and follow a single development plan for chronic toxicity testing of new medicinal products, the following studies are considered acceptable for submission in the 3 Regions: • 1) Rodents : a study of 6 months duration; • 2) Non-rodents : a study of nine months duration.
  • 68. S5(R2)_guideline DETECTION OF TOXICITY TO REPRODUCTION FOR MEDICINAL PRODUCTS & TOXICITY TO MALE FERTILITY: This guideline attempts to consolidate a strategy based on study designs currently in use for testing of medicinal products; • It should encourage the full assessment on the safety of chemicals on the development of the offspring. • It is perceived that tests in which animals are treated during defined stages of reproduction better reflect human exposure to medicinal products and allow more specific identification of stages at risk. • While this approach may be useful for most medicines, long term exposure to low doses does occur and may be represented better by a one or two generation study
  • 69. Aim of studies • The aim of reproduction toxicity studies is to reveal any effect of one or more active substance(s) on mammalian reproduction. For this purpose both the investigations and the interpretation of the results should be related to all other pharmacological and toxicological data available to determine whether potential reproductive risks to humans are greater, lesser or equal to those posed by other toxicological manifestations.
  • 70. 2. ANIMAL CRITERIA • Selection and number of species: Studies should be conducted in mammalian species. It is generally desirable to use the same species and strain as in other toxicological studies. • Reasons for using rats as the predominant rodent species are practicality, comparability with other results obtained in this species and the large amount of background knowledge accumulated. • In embryotoxicity studies only, a second mammalian species traditionally has been required, the rabbit being the preferred choice as a "non-rodent". Reasons for using rabbits in embryotoxicity studies include the extensive background knowledge that has accumulated, as well as availability and practicality.
  • 71. Other test systems • Other test systems have been developed and used in preliminary investigations ("pre-screening" or priority selection) and secondary testing. • For preliminary investigation of a range of analogue series of substances it is essential that the potential outcome in whole animals is known for at least one member of the series to be studied (by inference, effects are expected). With this strategy substances can be selected for higher level testing. • For secondary testing characterisation other test systems offer the possibility to study some of the observable developmental processes in detail, e.g. to reveal specific mechanisms of toxicity, to establish concentration- response relationships, to select 'sensitive periods', or to detect effects of defined metabolites
  • 72. 3. GENERAL RECOMMENDATIONS CONCERNING TREATMENT • Dosages :Selection of dosages is one of the most critical issues in design of the reproductive toxicity study. • The choice of the high dose should be based on data from all available studies (pharmacology, acute and chronic toxicity and kinetic studies, Note 7). • A repeated dose toxicity study of about 2 - 4 weeks duration provides a close approximation to the duration of treatment in segmental designs of reproductive studies
  • 73. Route and frequency of administration. • In general the route or routes of administration should be similar to those intended for human usage. One route of substance administration may be acceptable if it can be shown that a similar distribution (kinetic profile) results from different routes (Note 9). • The usual frequency of administration is once daily but consideration should be given to use either more frequent or less frequent administration taking kinetic variables into account (see also Note 10).
  • 74. STATISTICS • Analysis of the statistics of a study is the means by which results are interpreted. • The most important part of this analysis is to establish the relationship between the different variables and their distribution (descriptive statistics), since these determine how groups should be compared. • The distributions of the endpoints observed in reproductive tests are usually non normal and extend from almost continuous.
  • 75. DATA PRESENTATION • The key to good reporting is the tabulation of individual values in a clear concise manner to account for every animal that was entered into the study. • A reader should be able to follow the history of any individual animal from initiation to termination • It should be able to deduce with ease the contribution that the individual has made to any group summary values.
  • 76. PRECLINICAL SAFETY EVALUATION OF BIOTECHNOLOGY-DERIVED PHARMACEUTICALS(S6_R1_guideline ) • Objectives: 1) to identify an initial safe dose and subsequent dose escalation schemes in humans. 2) to identify potential target organs for toxicity and for the study of whether such toxicity is reversible. 3) to identify safety parameters for clinical monitoring.
  • 77. SPECIFICATION OF THE TEST MATERIAL • purification processes to remove impurities and contaminants • to establish a preclinical testing program for their qualification • Comparability can be evaluated on the basis of biochemical and biological characterisation
  • 78. PRECLINICAL SAFETY TESTING • OBJECTIVES: To define pharmacological and toxicological effects not only prior to initiation of human studies but throughout clinical development. • Preclinical safety testing should consider: – 1) selection of the relevant animal species; – 2) age; – 3) physiological state; • 4) the manner of delivery, including dose, route of administration, and treatment regimen; and • 5) stability of the test material under the conditions of use
  • 79. Biological Activity/Pharmacodynamics Evaluated using in vitro assays to determine which effects of the product may be related to clinical activity The use of cell lines and/or primary cell cultures can be useful to examine the direct effects on cellular phenotype and proliferation. • Animal Species/Model Selection: Safety evaluation programs should include the use of relevant species. A relevant species is one in which the test material is pharmacologically active species that express the desired epitope and demonstrate a similar tissue cross-reactivity profile as for human tissues. • Number/Gender of Animals: No. of animals used per dose has a direct bearing on the ability to detect toxicity.
  • 80. Administration/Dose Selection Route and frequency of administration should be as close as possible to that proposed for clinical use. • SPECIFIC CONSIDERATIONS: – Safety Pharmacology : measure functional indices of potential toxicity.The aim of the safety pharmacology studies should be to reveal any functional effects on the major physiological systems. – Pharmacokinetics and Toxicokinetics : Pharmacokinetic studies should, whenever possible, utilise preparations that are representative of that intended for toxicity testing and clinical use, and employ a route of administration
  • 81. • Metabolism: degradation to small peptides and individual amino acids. • Single Dose Toxicity Studies:useful data to describe the relationship of dose to systemic and/or local toxicity. These data can be used to select doses for repeated dose toxicity studies. • Repeated Dose Toxicity Studies: The duration of repeated dose studies should be based on the intended duration of clinical exposure and disease indication.
  • 82. Immunotoxicity Studies • It includes assessment of potential immunogenicity. • Biotechnology-derived pharmaceuticals are intended to stimulate or suppress the immune system and may affect not only humoral but also cell-mediated immunity