SlideShare ist ein Scribd-Unternehmen logo
1 von 65
By
    Md gayasuddin
M.Pharm (pharmacology)
MNR college of pharmacy
Contents

 introduction
 ICH objectives
 ICH Organizational structure
 ICH Harmonisation process
 ICH Work Product
 ICH Guidelines
introduction
What does ICH stands for?
 The complete name of ICH is the "International
 Conference on Harmonisation of Technical
 Requirements for Registration of Pharmaceuticals for
 Human Use".
What is ICH ?
 ICH is a joint initiative involving both regulators and
  research-based industry representatives of the
  European Union, Japan and the USA in scientific and
  technical discussions of the testing procedures
  required to assess and ensure the safety, quality and
  efficacy of medicines.
What is the goal of ICH ?
 The goal of ICH is to promote international harmonization
  by bringing together representatives from the three ICH
  regions (EU, Japan and USA) to discuss and establish
  common guidelines.
 Another goal of ICH is to make information available on
  ICH, ICH activities and ICH guidelines to any country or
  company that requests the information, and to promote a
  mutual understanding of regional initiatives in order to
  facilitate harmonisation processes related to ICH
  guidelines regionally and globally, and to strengthen the
  capacity of drug regulatory authorities and industry to
  utilise them.
 The ICH Global Cooperation Group (GCG) was formed in
  1999 and is charged with this task.
ICH objectives
 The objective of ICH is to increase international
 harmonisation of technical requirements to ensure
 that safe, effective, and high quality medicines are
 developed and registered in the most efficient and
 cost-effective manner. These activities have been
 undertaken to promote public health, prevent
 unnecessary duplication of clinical trials in humans,
 and minimize the use of animal testing without
 compromising safety and effectiveness.
ICH Organisational Structure

The ICH structure consists of the
 ICH Steering Committee,
 ICH Coordinators,
 ICH Secretariat and
 ICH Working Groups.
 The ICH Global Cooperation Group (GCG) and the
  ICH MedDRA Management Board are sub-committees
  of the ICH Steering Committee.
Steering committee
 The Steering Committee is the body that governs the
 ICH, determines the policies and procedures for ICH,
 selects topics for harmonisation and monitors the
 progress of harmonisation initiatives. Each of the six
 ICH parties has two seats on the ICH Steering
 Committee. Each of the Observers nominates non-
 voting participants to attend the ICH Steering
 Committee Meetings. IFPMA also participates as a
 non-voting member.
The Coordinators
 The Coordinators are fundamental to the smooth
  running of the ICH and are nominated by each of the
  six parties.
 An ICH Coordinator acts as the main contact point
  with the ICH Secretariat. Due to structural differences
  within the EU and MHLW,
 ICH Technical Coordinators are also designated from
  the EMA and PMDA respectively.
The ICH Secretariat
The Secretariat operates from the IFPMA offices, in Geneva, and is
primarily concerned with preparations for, and documentation of,
meetings of the Steering Committee.


At the time of ICH Conferences, the Secretariat is responsible for the
technical documentation and for liaison with the speakers for the
Conference.
Working Groups
  SAFETY                  EFFICACY


  QUALITY               MULTIDISCIPLINARY




    STEERING COMMITTEE
  Endorses topics, guidelines and monitors
                 progress
Global Cooperation Group (GCG)
 The ICH Global Cooperation Group (GCG) was formed
  on March 11, 1999, as a subcommittee of the ICH
  Steering Committee.
 It is made up of one representative from each of the six
  parties on the ICH Steering Committee, plus the
  IFPMA.
 The ICH Observers, WHO, Canada and EFTA are also
  part of the GCG.
Members of ICH
 ICH is comprised of representatives from six parties that represent the
    regulatory bodies and research-based industry in the European Union,
    Japan and the USA.
   In Japan, the members are the Ministry of Health, Labour and Welfare
    (MHLW), and the Japan Pharmaceutical Manufacturers Association
    (JPMA).
   In Europe, the members are the European Union (EU), and the European
    Federation of Pharmaceutical Industries and Associations (EFPIA).
   In the USA, the members are the Food and Drug Administration (FDA),
    and the Pharmaceutical Research and Manufacturers of America
    (PhRMA).
   Additional members include Observers from the World Health
    Organization (WHO), European Free Trade Association (EFTA), and
    Canada. This important group of non-voting members represent non-ICH
    countries and regions.
   The International Federation of Pharmaceutical Manufacturers &
    Associations (IFPMA) has been closely involved with ICH since its
    inception and participates as a non-voting member.
ICH parties
 The ICH Parties are the founding members of ICH and
 represent the regulatory bodies and research-based
 industry in the EU, US and Japan.
ICH parties
6 parties
 EU
 EFPIA (European federation of pharmaceutical industries’ associations)
    MHLW (Ministry of health, Labor and welfare, Japan)
   JPMA (Japan Pharmaceuticals manufacturers Association)
   US FDA
   PhRMA
   Observers : WHO, TPP(canada)
   International federation of Pharmaceutical manufacturer’s
    association
The Observers
The ICH Observers include
 the European Free Trade Association (EFTA) -
  currently represented by Swiss medic (Swiss Agency
  for Therapeutic Products),
 Health Canada and World Health Organization
  (WHO).
 The Observers have been associated with the ICH
  process from the beginning to act as a link with non-
  ICH countries and regions.
ICH Harmonisation Process
 The ICH Steering Committee is responsible for the governance
    of ICH. This includes deciding on the adoption of every ICH
    project, whether a new topic, maintenance of an existing
    Guideline, or a specific implementation work.
   Each harmonisation activity is initiated by a Concept Paper
    which is a short summary of the proposal. Depending on the
    category of harmonisation activity a Business Plan may also be
    required.
   Any ICH Party or Observer is welcomed to submit a proposal for
    a new ICH activity.
   The ICH Steering Committee decides on the adoption of every
    ICH project and then endorses the creation of an EWG/IWG.
   ICH harmonisation activities fall into 4 categories: Formal ICH
    Procedure, Q&A Procedure, Revision Procedure and
    Maintenance Procedure.
Process of Harmonisation
Steps of ICH
5 Steps in the ICH process
 Consensus building
 Rapporteur prepares initial draft of a guideline/rcommendation for comment
 with fixed deadline for comment (fax, e-mail). Interim report made to SC
 meeting, if consensus is reached, sign-off - all members
 Start of regulatory action
 Wide ranging regulatory consultation
 EU: published as a draft CPMP guideline; US: published as a draft guidance in
 the Federal Register; Japan: translated & issued by MHLW for internal and
 external consultation. A Regulatory Rapporteur is dessignated to draw up the
 final document and sign-off
 Adoption of a tripartite harmonised text
 Both regulatory and industry parties of SC must be satisfied. Adoption takes
 place on the signatures from the 3 regulatory parties to ICH, affirming that the
 Guideline is recommended for adoption by the 3 regulatory bodies
 Implementation
ICH Products
    ICH has developed over 50 harmonised Guidelines aimed at
       eliminating duplication in the development and registration process,
       so that a single set of studies can be generated to demonstrate the
       quality, safety and efficacy of a new medicinal product.
      Quality-21 Guidelines
      Safety -14 Guidelines
      Efficacy -20 Guidelines
      Multidisciplinary -5 Guidelines

 Electronic Standards for the Transfer of Regulatory Information (ESTRI,
  E2B)
 Common Technical Document (CTD & eCTD)
 Medical dictionary for adverse event reporting and coding of clinical
  trial data (MedDRA)
 Considerationdocuments
ICH Guidelines
The ICH Topics are divided into four major categories and ICH Topic
Codes are assigned according to these categories.

         Q                      S                      E                     M
"Quality" Topics,      "Safety" Topics,       "Efficacy" Topics,     "Multidisciplinary"
i.e., those relating   i.e., those relating   i.e., those relating   Topics, i.e., cross-
to chemical and        to in vitro and in     to clinical studies    cutting Topics
pharmaceutical         vivo pre-clinical      in human subject       which do not fit
Quality                studies                (Dose Response         uniquely into one
Assurance              (Carcinogenicity       Studies, Good          of the above
(Stability Testing,    Testing,               Clinical Practices,    categories
Impurity Testing,      Genotoxicity           etc.)                  (MedDRA,
etc.)                  Testing, etc.)                                ESTRI, M3, CTD,
                                                                     M5)
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.
ICH GUIDELINES ON SAFETY OF ANIMALS
 S1A Guideline on the Need for Carcinogenicity Studies of
    Pharmaceuticals Nov. 1995
   S1B Testing for Carcinogenicity of Pharmaceuticals July 1997
   S1C(R2) Dose Selection for Carcinogenicity Studies of
    Pharmaceuticals Mar. 2008
   S2(R1) Guidance on Genotoxicity Testing and Data
    Interpretation for Pharmaceuticals Intended for
   Human Use Nov. 2011
   S3A Note for Guidance on Toxicokinetics: The Assessment of
    Systemic Exposure in Toxicity Studies Oct. 1994
   S3B Pharmacokinetics: Guidance for Repeated Dose Tissue
    Distribution Studies Oct. 1994
   S4 Duration of Chronic Toxicity Testing in Animals (Rodent
    and Non Rodent Toxicity Testing) Sept. 1998
 S5(R2) Detection of Toxicity to Reproduction for
    Medicinal Products and Toxicity to Male Fertility June
    1993
   S6(R1) Preclinical Safety Evaluation of Biotechnology-
    Derived Pharmaceuticals June 2011
   S7A Safety Pharmacology Studies for Human
    Pharmaceuticals Nov. 2000
   S7B The Non-clinical Evaluation of the Potential for
    Delayed Ventricular Repolarization (QT Interval
    Prolongation) by Human Pharmaceuticals May 2005
   S8 Immunotoxicity Studies for Human Pharmaceuticals
    Sept. 2005
   S9 Nonclinical Evaluation for Anticancer
    Pharmaceuticals Oct. 2009
   S10 Photo safety Evaluation Nov. 2012
S1A (Nov. 1995)
 Need for Carcinogenicity Studies of Pharmaceuticals

 This document provides a consistent definition of the
  circumstances under which it is necessary to undertake
  carcinogenicity studies on new drugs. These
  recommendations take into account the known risk factors
  as well as the intended indications and duration of
  exposure.

 Results from genotoxicity studies, toxicokinetics, and
  mechanistic studies can now be routinely applied in
  preclinical safety assessment.
S1B (July 1997)
 Testing for Carcinogenicity of Pharmaceuticals


 Guidance on the need to carry out carcinogenicity studies
  in both mice and rats, and guidance is also given on
  alternative testing procedures which may be applied
  without jeopardizing safety.
S1C-R2 (March 2008)
 Dose Selection for Carcinogenicity Studies of
  Pharmaceuticals

 addresses the criteria for the selection of the high dose to be
  used in carcinogenicity studies on new therapeutic agents to
  harmonise current practices and improve the design of studies.

 the pharmacokinetic endpoint of 25 is declared to be applicable
  also for pharmaceuticals with positive genotoxicity signals.

 This change has implications on reducing the pain or discomfort
  of the animals at the maximally tolerated dose (MTD).
S2-R1 (March 2008)
 Guidance On Genotoxicity Testing And Data Interpretation For
  Pharmaceuticals Intended For Human Use

 S2A: Guidance on Specific Aspects of Regulatory Genotoxicity
  Tests for Pharmaceuticals :specific guidance and recommendations
  for in vitro and in vivo tests and on the evaluation of test results. It
  includes a glossary of terms related to genotoxicity tests to improve
  consistency in applications.

 S2B: Genotoxicity: A Standard Battery for Genotoxicity Testing
  for Pharmaceuticals; the identification of a standard set of assays to
  be conducted for registration, and the extent of confirmatory
  experimentation in any particular genotoxicity assay in the standard
  battery.
S3A (Oct. 1994)
 Note For Guidance On Toxicokinetics:
  The Assessment Of Systemic Exposure In Toxicity
  Studies

 gives guidance on developing test strategies in
  toxicokinetics and the need to integrate pharmacokinetics
  into toxicity testing, in order to aid in the interpretation of
  the toxicology findings and promote rational study design
  development.
S3B (Oct. 1994)
 Pharmacokinetics: Guidance For Repeated Dose
  Tissue Distribution Studies

 This study is required when appropriate data cannot be
  derived from other sources

 A comprehensive knowledge of the absorption,
  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 (Sep. 1998)
 Duration Of Chronic Toxicity Testing In Animals
  (Rodent And Non Rodent Toxicity Testing)

 safety evaluation of a medicinal product


 for the development of medicinal products with the
  exception of those already covered by the ICH Guideline on
  Safety Studies for Biotechnological Products, e.g.,
  Monoclonal antibodies, recombinant DNA proteins.
S5-R2 (Nov. 2000)
 Detection Of Toxicity To Reproduction
  For Medicinal Products & Toxicity To Male Fertility

 This document provides guidance on tests for reproductive
  toxicity. It defines the periods of treatment to be used in
  animals to better reflect human exposure to medical
  products and allow more specific identification of stages at
  risk
S6 (Oct. 2009)
 Addendum To Ich S6: Preclinical Safety Evaluation Of
  Biotechnology-derived Pharmaceuticals

 It addresses the use of animal models of disease,
  determination of when genotoxicity assays and
  carcinogenicity studies should be performed, and the
  impact of antibody formation on duration of toxicology
  studies

 Clarification on species selection, study design,
  immunogenicity, reproductive and developmental toxicity
  and assessment of carcinogenic potential.
S7A (Nov. 2000)
 Safety Pharmacology Studies for Human
  Pharmaceuticals

 addresses the definition, objectives and scope of safety
  pharmacology studies

 also addresses which studies are needed before initiation of
  Phase 1 clinical studies as well as information needed for
  marketing.
S7B (May 2005)
 The Non-clinical Evaluation Of The Potential For
  Delayed Ventricular Repolarization (Qt Interval
  Prolongation) By Human Pharmaceuticals

 This Guideline describes a non-clinical testing strategy for
  assessing the potential of a test substance to delay
  ventricular repolarization.

 Includes non-clinical assays and integrated risk
  assessments
S8 (Sep. 2005)
 Immunotoxicity Studies for Human Pharmaceuticals
 addresses the recommendations on nonclinical testing for
  immunosuppression induced by low molecular weight
  drugs (non-biologicals and how each immunotoxicity
  study should be performed
 It applies to new pharmaceuticals intended for use in
  humans, as well as to marketed drug products proposed for
  different indications or other variations on the current
  product label in which the change could result in
  unaddressed and relevant toxicologic issues
 Also applicable during CT and following approval to
  market.
S9 (Oct. 2009)
 Nonclinical Evaluation for Anticancer
  Pharmaceuticals

 provides information for pharmaceuticals that are only
  intended to treat cancer in patients with late stage or
  advanced disease regardless of the route of administration,
  including both small molecule and biotechnology-derived
  pharmaceuticals. It describes the type and timing of
  nonclinical studies in relation to the development of
  anticancer pharmaceuticals and references other guidance
  as appropriate.
S10 (June 2010)
 Photosafety Evaluation of Pharmaceuticals


 This new Guideline on photosafety testing will be a
  valuable adjunct to the guidance provided in the M3(R2)
  Guideline.
ICH-E Guidelines
Efficacy Guidelines
 The work carried out by ICH under the Efficacy
 heading is concerned with the design, conduct, safety
 and reporting of clinical trials. It also covers novel
 types of medicines derived from biotechnological
 processes and the use of pharmacogenetics/
 pharmacogenomics techniques to produce better
 targeted medicines.
Efficacy Guidelines
 Clinical Safety E1-E2F
 Clinical Study Reports E3
 Dose Response Studies E4
 Ethnic Factors E5
 Good Clinical Practice E6
 Clinical Trials E7-E11
 Guidelines for Clinical Evaluation by Therapeutic Category
  E12
 Clinical Evaluation E14
 Pharmacogenomics E15-E16
 Joint Safety / Efficacy Topic M3
E1 (Oct. 1994)
 E1 – The Extent Of Population Exposure To Assess Clinical Safety
  For Drugs Intended For Long-term Treatment Of Non-life-
  threatening Conditions

 The tripartite harmonized ICH Guideline was finalised under Step 4 in
  October 1994. This document gives recommendations on the numbers
  of patients and duration of exposure for the safety evaluation of drugs
  intended for the long-term treatment of non-life-threatening
  conditions.
 Events where the rate of occurrence changes over a longer period of
  time may need to be characterized depending on their severity and
  importance to the risk-benefit assessment of the drug.
 The safety evaluation during clinical drug development is not expected
  to characterise rare adverse events, for example, those occurring in less
  than 1 in 1000 patients.
E2A (Oct. 1994)
 Clinical Safety Data Management: Definitions And
  Standards For Expedited Reporting E2A

 There are two issues within the broad subject of clinical safety data
  management that are appropriate for harmonization at this time:

 (1) the development of standard definitions and terminology for key
  aspects of clinical safety reporting, and

 (2) the appropriate mechanism for handling expedited (rapid)
  reporting, in the investigational (i.e., pre-approval) phase.
E2B-R2 (Feb. 2001)
 Maintenance Of The ICH Guideline On Clinical Safety
 Data Management :
 Data Elements For Transmission Of Individual Case
 Safety Reports E2B(R2)

 to standardize the data elements for transmission of
 individual case safety reports by identifying, and where
 necessary or advisable, by defining the data elements for
 the transmission of all types of individual case safety
 reports, regardless of source and destination.
E2C-R1(Nov. 1996)
 Clinical Safety Data Management: Periodic Safety Update
  Reports for Marketed Drugs E2C (R1)

 guidance on the format and content of safety updates, which need to
  be provided at intervals to regulatory authorities after products have
  been marketed. The Guideline is intended to ensure that the worldwide
  safety experience is provided to authorities at defined times after
  marketing with maximum efficiency and avoiding duplication of effort.

 E2C(R2) This Revision was endorsed by Steering Committee in
  Dec.2010. It will Evaluate the ICH Pharmacovigilance documentation,
  conduct a gap and potential improvement analysis of ECH E2C, E2E
  and E2F and draft a new ICH E2C R2 covering periodic benefic risk
  evaluation reporting.
E2D (Nov. 2003)
 Post Approval Safety Data Management: Definitions
  and Standards for Expedited Reporting

 provides a standardized procedure for post-approval safety
  data management including expedited reporting to
  relevant authority.

 The definitions of the terms and concept specific to post-
  approval phase are also provided.
E2E (Nov. 2004)
 Pharmacovigilance Planning
 planning of pharmacovigilance activities, especially in
  preparation for the early post marketing period of a new
  drug (chemical entities, biotech-derived products,
  vaccines)
 Main focus: Safety specification and PV plan that mighty
  be submitted at the time of license application
E2F (Aug. 2010)
 Development Safety Update Report
 The main focus of the DSUR is data from interventional
  clinical trials (referred to in this document as "clinical
  trials") of investigational drugs including biologicals, with
  or without a marketing approval, whether conducted by
  commercial or non-commercial sponsors.
 Intended for periodic reporting on drugs under
  development (including marketed drugs that are under
  further study) among the ICH regions.
E3 (Nov. 1995)
 Structure and Content of Clinical Study Reports

 is an "integrated" full report of an individual study of any therapeutic,
  prophylactic or diagnostic agent (referred to herein as drug or
  treatment) conducted in patients, in which the clinical and statistical
  description, presentations, and analyses are integrated into a single
  report, incorporating tables and figures into the main text of the
  report, or at the end of the text, and with appendices containing the
  protocol, sample case report forms, investigator related information,
  information related to the test drugs/investigational products
  including     active     control/comparators,       technical   statistical
  documentation, related publications, patient data listings, and
  technical statistical details such as derivations, computations, analyses,
  and computer output etc.
E4 (March, 1994)
 Dose-response      Information      To    Support     Drug
  Registration

 Knowledge of the relationships among dose, drug-
  concentration in blood, and clinical response (effectiveness
  and undesirable effects) is important for the safe and
  effective use of drugs in individual patients.
 concepts of minimum effective dose and maximum useful
  dose do not adequately account for individual differences
  and do not allow a comparison, at various doses, of both
  beneficial and undesirable effects.
 Any given dose provides a mixture of desirable and
  undesirable effects, with no single dose necessarily optimal
  for all patients.
E5 R1 (Feb. 1998)
 Ethnic Factors In The Acceptability Of Foreign Clinical
  Data

 This document addresses the intrinsic characteristics of the
  drug recipient and extrinsic characteristics associated with
  environment and culture that could affect the results of
  clinical studies carried out in regions and describes the
  concept of the "bridging study" that a new region may
  request to determine whether data from another region are
  applicable to its population.
E6-R1 (May 1996)
 Good Clinical Practice

 describes the responsibilities and expectations of all
  participants in the conduct of clinical trials, including
  investigators, monitors, sponsors and IRBs.
 GCPs cover aspects of monitoring, reporting and archiving
  of clinical trials and incorporating addenda on the
  Essential Documents and on the Investigator's Brochure
  which had been agreed earlier through the ICH process.
E7 (June 1993)
 Studies in Support of Special Populations: Geriatrics

 This document provides recommendations on the special
  considerations which apply in the design and conduct of
  clinical trials of medicines that are likely to have significant
  use in the elderly.
 It requires special consideration due to the frequent
  occurrence of underlying diseases, concomitant drug
  therapy and the consequent risk of drug interaction.
E8 (July 1997)
 General Considerations for Clinical Trials


 This document sets out the general scientific principles for
  the conduct, performance and control of clinical trials.
 The Guideline addresses a wide range of subjects in the
  design and execution of clinical trials.
E9 (Feb. 1998)
 Statistical Principles for Clinical Trials

 This      biostatistical   Guideline   describes    essential
  considerations on the design and analysis of clinical trials,
  especially the "confirmatory" (hypothesis-testing) trials
  that are the basis for demonstrating effectiveness.
 The document will also assist scientific experts charged
  with preparing application summaries or assessing
  evidence of efficacy and safety, principally from clinical
  trials in later phases of development.
E10 (July 2000)
 Choice of Control Group and Related Issues in
 Clinical Trials

 This document addresses the choice of control groups in
 clinical trials considering the ethical and inferential
 properties and limitations of different kinds of control
 groups. It points out the assay sensitivity problem in active
 control equivalence / non-inferiority trials that limits the
 usefulness of trial design in many circumstances.
E11 (July 2000)
 Clinical Investigation of Medicinal Products in the
  Pediatric Population

 This document addresses the conduct of clinical trials of
  medicines in pediatric populations. This document will
  facilitate the development of safe and effective use of
  medicinal product in pediatrics.
E12 (March 2000)
 Principles  for Clinical         Evaluation     of    New
  antihypertensive Drugs

 It provides a set of "Principles" on which there is general
  agreement among all three ICH regions covering endpoints
  and trial designs.
 It will not be subject to the usual procedures leading to a
  fully harmonized document.
E14 (May 2005)
 The Clinical Evaluation of QT/QTc Interval Prolongation and
  Proarrhythmic Potential for Non-Antiarrhythmic Drugs.

 This document provides recommendations to sponsors concerning the
  design, conduct, analysis, and interpretation of clinical studies to
  assess the potential of a drug to delay cardiac repolarisation.
 This assessment should include testing the effects of new agents on the
  QT/QTc interval as well as the collection of cardiovascular adverse
  events. The investigational approach used for a particular drug should
  be     individualised,    depending      on    the    pharmacodynamic,
  pharmacokinetic, and safety characteristics of the product, as well as
  on its proposed clinical use.
 The assessment of the effects of drugs on cardiac repolarisation is the
  subject of active investigation.
 When additional data (non-clinical and clinical) are accumulated in
  the future, this document may be reevaluated and revised.
E15 (Nov. 2007)
 Definitions For Genomic Biomarkers, Pharmaco-
 genomics, Pharmacogenetics, Genomic Data And
 Sample Coding Categories

 Pharmacogenomics     and pharmacogenetics have the
 potential to improve the discovery, development and use of
 medicines.
E16 (Aug. 2010)
 Biomarkers Related To Drug Or Biotechnology
 Product Development:
 Context, Structure And Format Of Qualification
 Submissions

 The Guideline describes recommendations regarding
 context, structure, and format of regulatory submissions
 for qualification of genomic biomarkers, as defined in ICH
 E15.
M3 R2 (June 2009)
 Guidance On Non-clinical Safety Studies For The
 Conduct Of Human Clinical Trials And Marketing
 Authorization For Pharmaceuticals

 Harmonisation of the guidance for non-clinical safety
 studies will help to define the current recommendations
 and reduce the likelihood that substantial differences will
 exist among regions.
Thank you…

Weitere ähnliche Inhalte

Was ist angesagt?

Investigational New Drug Application
Investigational New Drug ApplicationInvestigational New Drug Application
Investigational New Drug ApplicationSuhas Reddy C
 
NEW DRUG APPLICATION ( NDA)
NEW DRUG APPLICATION ( NDA)NEW DRUG APPLICATION ( NDA)
NEW DRUG APPLICATION ( NDA)Suvarta Maru
 
New Drug Application [NDA]
New Drug Application [NDA]New Drug Application [NDA]
New Drug Application [NDA]Sagar Savale
 
Regulatory authorities (US-FDA, WHO and ICH)
Regulatory authorities (US-FDA, WHO and ICH)Regulatory authorities (US-FDA, WHO and ICH)
Regulatory authorities (US-FDA, WHO and ICH)Sagar Savale
 
New drug application
New drug applicationNew drug application
New drug applicationVKEkbote
 
Regulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical IndustryRegulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical IndustryRama Shukla
 
TGA, MHRA, MCC, MCA
TGA, MHRA, MCC, MCATGA, MHRA, MCC, MCA
TGA, MHRA, MCC, MCASagar Savale
 
Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]Sagar Savale
 
Clinical trial protocol development
Clinical trial protocol developmentClinical trial protocol development
Clinical trial protocol developmentSACHIN C P
 
4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.Audumbar Mali
 
Investigational new drug application
Investigational new drug application Investigational new drug application
Investigational new drug application Kirsha K S
 
Drug regulatory authority
Drug regulatory authority Drug regulatory authority
Drug regulatory authority Rahul Gawande
 
Ich guidelines QSEM suraj seminar
Ich guidelines  QSEM suraj seminarIch guidelines  QSEM suraj seminar
Ich guidelines QSEM suraj seminarSuraj Pund
 
New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)
New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)
New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)Vamsikrishna Reddy
 
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
 
THERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRA
THERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRATHERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRA
THERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRAManikant Prasad Shah
 

Was ist angesagt? (20)

MHRA
MHRAMHRA
MHRA
 
Investigational New Drug Application
Investigational New Drug ApplicationInvestigational New Drug Application
Investigational New Drug Application
 
NEW DRUG APPLICATION ( NDA)
NEW DRUG APPLICATION ( NDA)NEW DRUG APPLICATION ( NDA)
NEW DRUG APPLICATION ( NDA)
 
New Drug Application [NDA]
New Drug Application [NDA]New Drug Application [NDA]
New Drug Application [NDA]
 
Regulatory authorities (US-FDA, WHO and ICH)
Regulatory authorities (US-FDA, WHO and ICH)Regulatory authorities (US-FDA, WHO and ICH)
Regulatory authorities (US-FDA, WHO and ICH)
 
New drug application
New drug applicationNew drug application
New drug application
 
Regulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical IndustryRegulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical Industry
 
TGA, MHRA, MCC, MCA
TGA, MHRA, MCC, MCATGA, MHRA, MCC, MCA
TGA, MHRA, MCC, MCA
 
Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]
 
INDA/NDA/ANDA
INDA/NDA/ANDAINDA/NDA/ANDA
INDA/NDA/ANDA
 
Clinical trial protocol development
Clinical trial protocol developmentClinical trial protocol development
Clinical trial protocol development
 
4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.
 
Code of Federal Regulations
Code of Federal RegulationsCode of Federal Regulations
Code of Federal Regulations
 
Investigational new drug application
Investigational new drug application Investigational new drug application
Investigational new drug application
 
Drug regulatory authority
Drug regulatory authority Drug regulatory authority
Drug regulatory authority
 
Ich guidelines QSEM suraj seminar
Ich guidelines  QSEM suraj seminarIch guidelines  QSEM suraj seminar
Ich guidelines QSEM suraj seminar
 
The introduction of an orange book
The introduction of an orange bookThe introduction of an orange book
The introduction of an orange book
 
New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)
New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)
New Drug Application(Nda) Vs Abbreviated New Drug Application (Anda)
 
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.
 
THERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRA
THERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRATHERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRA
THERAPEUTIC GOODS ADMINISTRATION (TGA) and MHRA
 

Ähnlich wie Ich guidelines seminar

1.5 international conference on harmonization
1.5 international conference on harmonization1.5 international conference on harmonization
1.5 international conference on harmonizationShital Patil
 
Nivedita ICH GUIDELINES
Nivedita  ICH GUIDELINESNivedita  ICH GUIDELINES
Nivedita ICH GUIDELINESNiveditaSingla
 
PROCESS OF ICH (International Council for Harmonisation)
PROCESS OF ICH (International Council for Harmonisation)PROCESS OF ICH (International Council for Harmonisation)
PROCESS OF ICH (International Council for Harmonisation)Swathi P
 
Jignesh ich
Jignesh ichJignesh ich
Jignesh ichjigs2163
 
International council for harmonisation (ich) guidelines
International council for harmonisation (ich) guidelinesInternational council for harmonisation (ich) guidelines
International council for harmonisation (ich) guidelinesMasarrat Khan
 
ICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIESICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIESHEALY LAD
 
Toxicity studies
Toxicity studiesToxicity studies
Toxicity studiessopi_1234
 
ICH GUIDELINES AND ITS IMPACT IN INDIA
ICH GUIDELINES AND ITS IMPACT IN INDIAICH GUIDELINES AND ITS IMPACT IN INDIA
ICH GUIDELINES AND ITS IMPACT IN INDIAasmitadey785
 
3 briefintrototheich guidelinesindia2007
3 briefintrototheich guidelinesindia20073 briefintrototheich guidelinesindia2007
3 briefintrototheich guidelinesindia2007abualinajdi
 

Ähnlich wie Ich guidelines seminar (20)

1.5 international conference on harmonization
1.5 international conference on harmonization1.5 international conference on harmonization
1.5 international conference on harmonization
 
Ich guideline
Ich guidelineIch guideline
Ich guideline
 
Nivedita ICH GUIDELINES
Nivedita  ICH GUIDELINESNivedita  ICH GUIDELINES
Nivedita ICH GUIDELINES
 
PROCESS OF ICH (International Council for Harmonisation)
PROCESS OF ICH (International Council for Harmonisation)PROCESS OF ICH (International Council for Harmonisation)
PROCESS OF ICH (International Council for Harmonisation)
 
Jignesh ich
Jignesh ichJignesh ich
Jignesh ich
 
International council for harmonisation (ich) guidelines
International council for harmonisation (ich) guidelinesInternational council for harmonisation (ich) guidelines
International council for harmonisation (ich) guidelines
 
ICH Introduction
ICH IntroductionICH Introduction
ICH Introduction
 
Ich introduction
Ich introductionIch introduction
Ich introduction
 
International council for harmonization by akshay trivedi
International council for harmonization by akshay trivediInternational council for harmonization by akshay trivedi
International council for harmonization by akshay trivedi
 
ICH
ICHICH
ICH
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
 
ICH guidelines
ICH  guidelinesICH  guidelines
ICH guidelines
 
Ich guidelines ppt
Ich guidelines pptIch guidelines ppt
Ich guidelines ppt
 
ICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIESICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIES
 
Toxicity studies
Toxicity studiesToxicity studies
Toxicity studies
 
ICH PPT
ICH PPTICH PPT
ICH PPT
 
ICH GUIDELINES AND ITS IMPACT IN INDIA
ICH GUIDELINES AND ITS IMPACT IN INDIAICH GUIDELINES AND ITS IMPACT IN INDIA
ICH GUIDELINES AND ITS IMPACT IN INDIA
 
ICH ....
ICH ....ICH ....
ICH ....
 
3 briefintrototheich guidelinesindia2007
3 briefintrototheich guidelinesindia20073 briefintrototheich guidelinesindia2007
3 briefintrototheich guidelinesindia2007
 
Ich
IchIch
Ich
 

Ich guidelines seminar

  • 1. By Md gayasuddin M.Pharm (pharmacology) MNR college of pharmacy
  • 2. Contents  introduction  ICH objectives  ICH Organizational structure  ICH Harmonisation process  ICH Work Product  ICH Guidelines
  • 4. What does ICH stands for?  The complete name of ICH is the "International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use".
  • 5. What is ICH ?  ICH is a joint initiative involving both regulators and research-based industry representatives of the European Union, Japan and the USA in scientific and technical discussions of the testing procedures required to assess and ensure the safety, quality and efficacy of medicines.
  • 6. What is the goal of ICH ?  The goal of ICH is to promote international harmonization by bringing together representatives from the three ICH regions (EU, Japan and USA) to discuss and establish common guidelines.  Another goal of ICH is to make information available on ICH, ICH activities and ICH guidelines to any country or company that requests the information, and to promote a mutual understanding of regional initiatives in order to facilitate harmonisation processes related to ICH guidelines regionally and globally, and to strengthen the capacity of drug regulatory authorities and industry to utilise them.  The ICH Global Cooperation Group (GCG) was formed in 1999 and is charged with this task.
  • 7. ICH objectives  The objective of ICH is to increase international harmonisation of technical requirements to ensure that safe, effective, and high quality medicines are developed and registered in the most efficient and cost-effective manner. These activities have been undertaken to promote public health, prevent unnecessary duplication of clinical trials in humans, and minimize the use of animal testing without compromising safety and effectiveness.
  • 8. ICH Organisational Structure The ICH structure consists of the  ICH Steering Committee,  ICH Coordinators,  ICH Secretariat and  ICH Working Groups.  The ICH Global Cooperation Group (GCG) and the ICH MedDRA Management Board are sub-committees of the ICH Steering Committee.
  • 9. Steering committee  The Steering Committee is the body that governs the ICH, determines the policies and procedures for ICH, selects topics for harmonisation and monitors the progress of harmonisation initiatives. Each of the six ICH parties has two seats on the ICH Steering Committee. Each of the Observers nominates non- voting participants to attend the ICH Steering Committee Meetings. IFPMA also participates as a non-voting member.
  • 10. The Coordinators  The Coordinators are fundamental to the smooth running of the ICH and are nominated by each of the six parties.  An ICH Coordinator acts as the main contact point with the ICH Secretariat. Due to structural differences within the EU and MHLW,  ICH Technical Coordinators are also designated from the EMA and PMDA respectively.
  • 11. The ICH Secretariat The Secretariat operates from the IFPMA offices, in Geneva, and is primarily concerned with preparations for, and documentation of, meetings of the Steering Committee. At the time of ICH Conferences, the Secretariat is responsible for the technical documentation and for liaison with the speakers for the Conference.
  • 12. Working Groups SAFETY EFFICACY QUALITY MULTIDISCIPLINARY STEERING COMMITTEE Endorses topics, guidelines and monitors progress
  • 13. Global Cooperation Group (GCG)  The ICH Global Cooperation Group (GCG) was formed on March 11, 1999, as a subcommittee of the ICH Steering Committee.  It is made up of one representative from each of the six parties on the ICH Steering Committee, plus the IFPMA.  The ICH Observers, WHO, Canada and EFTA are also part of the GCG.
  • 14. Members of ICH  ICH is comprised of representatives from six parties that represent the regulatory bodies and research-based industry in the European Union, Japan and the USA.  In Japan, the members are the Ministry of Health, Labour and Welfare (MHLW), and the Japan Pharmaceutical Manufacturers Association (JPMA).  In Europe, the members are the European Union (EU), and the European Federation of Pharmaceutical Industries and Associations (EFPIA).  In the USA, the members are the Food and Drug Administration (FDA), and the Pharmaceutical Research and Manufacturers of America (PhRMA).  Additional members include Observers from the World Health Organization (WHO), European Free Trade Association (EFTA), and Canada. This important group of non-voting members represent non-ICH countries and regions.  The International Federation of Pharmaceutical Manufacturers & Associations (IFPMA) has been closely involved with ICH since its inception and participates as a non-voting member.
  • 15. ICH parties  The ICH Parties are the founding members of ICH and represent the regulatory bodies and research-based industry in the EU, US and Japan.
  • 16. ICH parties 6 parties  EU  EFPIA (European federation of pharmaceutical industries’ associations)  MHLW (Ministry of health, Labor and welfare, Japan)  JPMA (Japan Pharmaceuticals manufacturers Association)  US FDA  PhRMA  Observers : WHO, TPP(canada)  International federation of Pharmaceutical manufacturer’s association
  • 17. The Observers The ICH Observers include  the European Free Trade Association (EFTA) - currently represented by Swiss medic (Swiss Agency for Therapeutic Products),  Health Canada and World Health Organization (WHO).  The Observers have been associated with the ICH process from the beginning to act as a link with non- ICH countries and regions.
  • 18. ICH Harmonisation Process  The ICH Steering Committee is responsible for the governance of ICH. This includes deciding on the adoption of every ICH project, whether a new topic, maintenance of an existing Guideline, or a specific implementation work.  Each harmonisation activity is initiated by a Concept Paper which is a short summary of the proposal. Depending on the category of harmonisation activity a Business Plan may also be required.  Any ICH Party or Observer is welcomed to submit a proposal for a new ICH activity.  The ICH Steering Committee decides on the adoption of every ICH project and then endorses the creation of an EWG/IWG.  ICH harmonisation activities fall into 4 categories: Formal ICH Procedure, Q&A Procedure, Revision Procedure and Maintenance Procedure.
  • 21. 5 Steps in the ICH process  Consensus building Rapporteur prepares initial draft of a guideline/rcommendation for comment with fixed deadline for comment (fax, e-mail). Interim report made to SC meeting, if consensus is reached, sign-off - all members  Start of regulatory action  Wide ranging regulatory consultation EU: published as a draft CPMP guideline; US: published as a draft guidance in the Federal Register; Japan: translated & issued by MHLW for internal and external consultation. A Regulatory Rapporteur is dessignated to draw up the final document and sign-off  Adoption of a tripartite harmonised text Both regulatory and industry parties of SC must be satisfied. Adoption takes place on the signatures from the 3 regulatory parties to ICH, affirming that the Guideline is recommended for adoption by the 3 regulatory bodies  Implementation
  • 22. ICH Products  ICH has developed over 50 harmonised Guidelines aimed at eliminating duplication in the development and registration process, so that a single set of studies can be generated to demonstrate the quality, safety and efficacy of a new medicinal product.  Quality-21 Guidelines  Safety -14 Guidelines  Efficacy -20 Guidelines  Multidisciplinary -5 Guidelines  Electronic Standards for the Transfer of Regulatory Information (ESTRI, E2B)  Common Technical Document (CTD & eCTD)  Medical dictionary for adverse event reporting and coding of clinical trial data (MedDRA)  Considerationdocuments
  • 23. ICH Guidelines The ICH Topics are divided into four major categories and ICH Topic Codes are assigned according to these categories. Q S E M "Quality" Topics, "Safety" Topics, "Efficacy" Topics, "Multidisciplinary" i.e., those relating i.e., those relating i.e., those relating Topics, i.e., cross- to chemical and to in vitro and in to clinical studies cutting Topics pharmaceutical vivo pre-clinical in human subject which do not fit Quality studies (Dose Response uniquely into one Assurance (Carcinogenicity Studies, Good of the above (Stability Testing, Testing, Clinical Practices, categories Impurity Testing, Genotoxicity etc.) (MedDRA, etc.) Testing, etc.) ESTRI, M3, CTD, M5)
  • 24. 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.
  • 25. ICH GUIDELINES ON SAFETY OF ANIMALS  S1A Guideline on the Need for Carcinogenicity Studies of Pharmaceuticals Nov. 1995  S1B Testing for Carcinogenicity of Pharmaceuticals July 1997  S1C(R2) Dose Selection for Carcinogenicity Studies of Pharmaceuticals Mar. 2008  S2(R1) Guidance on Genotoxicity Testing and Data Interpretation for Pharmaceuticals Intended for  Human Use Nov. 2011  S3A Note for Guidance on Toxicokinetics: The Assessment of Systemic Exposure in Toxicity Studies Oct. 1994  S3B Pharmacokinetics: Guidance for Repeated Dose Tissue Distribution Studies Oct. 1994  S4 Duration of Chronic Toxicity Testing in Animals (Rodent and Non Rodent Toxicity Testing) Sept. 1998
  • 26.  S5(R2) Detection of Toxicity to Reproduction for Medicinal Products and Toxicity to Male Fertility June 1993  S6(R1) Preclinical Safety Evaluation of Biotechnology- Derived Pharmaceuticals June 2011  S7A Safety Pharmacology Studies for Human Pharmaceuticals Nov. 2000  S7B The Non-clinical Evaluation of the Potential for Delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceuticals May 2005  S8 Immunotoxicity Studies for Human Pharmaceuticals Sept. 2005  S9 Nonclinical Evaluation for Anticancer Pharmaceuticals Oct. 2009  S10 Photo safety Evaluation Nov. 2012
  • 27. S1A (Nov. 1995)  Need for Carcinogenicity Studies of Pharmaceuticals  This document provides a consistent definition of the circumstances under which it is necessary to undertake carcinogenicity studies on new drugs. These recommendations take into account the known risk factors as well as the intended indications and duration of exposure.  Results from genotoxicity studies, toxicokinetics, and mechanistic studies can now be routinely applied in preclinical safety assessment.
  • 28. S1B (July 1997)  Testing for Carcinogenicity of Pharmaceuticals  Guidance on the need to carry out carcinogenicity studies in both mice and rats, and guidance is also given on alternative testing procedures which may be applied without jeopardizing safety.
  • 29. S1C-R2 (March 2008)  Dose Selection for Carcinogenicity Studies of Pharmaceuticals  addresses the criteria for the selection of the high dose to be used in carcinogenicity studies on new therapeutic agents to harmonise current practices and improve the design of studies.  the pharmacokinetic endpoint of 25 is declared to be applicable also for pharmaceuticals with positive genotoxicity signals.  This change has implications on reducing the pain or discomfort of the animals at the maximally tolerated dose (MTD).
  • 30. S2-R1 (March 2008)  Guidance On Genotoxicity Testing And Data Interpretation For Pharmaceuticals Intended For Human Use  S2A: Guidance on Specific Aspects of Regulatory Genotoxicity Tests for Pharmaceuticals :specific guidance and recommendations for in vitro and in vivo tests and on the evaluation of test results. It includes a glossary of terms related to genotoxicity tests to improve consistency in applications.  S2B: Genotoxicity: A Standard Battery for Genotoxicity Testing for Pharmaceuticals; the identification of a standard set of assays to be conducted for registration, and the extent of confirmatory experimentation in any particular genotoxicity assay in the standard battery.
  • 31. S3A (Oct. 1994)  Note For Guidance On Toxicokinetics: The Assessment Of Systemic Exposure In Toxicity Studies  gives guidance on developing test strategies in toxicokinetics and the need to integrate pharmacokinetics into toxicity testing, in order to aid in the interpretation of the toxicology findings and promote rational study design development.
  • 32. S3B (Oct. 1994)  Pharmacokinetics: Guidance For Repeated Dose Tissue Distribution Studies  This study is required when appropriate data cannot be derived from other sources  A comprehensive knowledge of the absorption, distribution, metabolism and elimination of a compound is important for the interpretation of pharmacology and toxicology studies.  useful for designing toxicology and pharmacology studies
  • 33. S4 (Sep. 1998)  Duration Of Chronic Toxicity Testing In Animals (Rodent And Non Rodent Toxicity Testing)  safety evaluation of a medicinal product  for the development of medicinal products with the exception of those already covered by the ICH Guideline on Safety Studies for Biotechnological Products, e.g., Monoclonal antibodies, recombinant DNA proteins.
  • 34. S5-R2 (Nov. 2000)  Detection Of Toxicity To Reproduction For Medicinal Products & Toxicity To Male Fertility  This document provides guidance on tests for reproductive toxicity. It defines the periods of treatment to be used in animals to better reflect human exposure to medical products and allow more specific identification of stages at risk
  • 35. S6 (Oct. 2009)  Addendum To Ich S6: Preclinical Safety Evaluation Of Biotechnology-derived Pharmaceuticals  It addresses the use of animal models of disease, determination of when genotoxicity assays and carcinogenicity studies should be performed, and the impact of antibody formation on duration of toxicology studies  Clarification on species selection, study design, immunogenicity, reproductive and developmental toxicity and assessment of carcinogenic potential.
  • 36. S7A (Nov. 2000)  Safety Pharmacology Studies for Human Pharmaceuticals  addresses the definition, objectives and scope of safety pharmacology studies  also addresses which studies are needed before initiation of Phase 1 clinical studies as well as information needed for marketing.
  • 37. S7B (May 2005)  The Non-clinical Evaluation Of The Potential For Delayed Ventricular Repolarization (Qt Interval Prolongation) By Human Pharmaceuticals  This Guideline describes a non-clinical testing strategy for assessing the potential of a test substance to delay ventricular repolarization.  Includes non-clinical assays and integrated risk assessments
  • 38. S8 (Sep. 2005)  Immunotoxicity Studies for Human Pharmaceuticals  addresses the recommendations on nonclinical testing for immunosuppression induced by low molecular weight drugs (non-biologicals and how each immunotoxicity study should be performed  It applies to new pharmaceuticals intended for use in humans, as well as to marketed drug products proposed for different indications or other variations on the current product label in which the change could result in unaddressed and relevant toxicologic issues  Also applicable during CT and following approval to market.
  • 39. S9 (Oct. 2009)  Nonclinical Evaluation for Anticancer Pharmaceuticals  provides information for pharmaceuticals that are only intended to treat cancer in patients with late stage or advanced disease regardless of the route of administration, including both small molecule and biotechnology-derived pharmaceuticals. It describes the type and timing of nonclinical studies in relation to the development of anticancer pharmaceuticals and references other guidance as appropriate.
  • 40. S10 (June 2010)  Photosafety Evaluation of Pharmaceuticals  This new Guideline on photosafety testing will be a valuable adjunct to the guidance provided in the M3(R2) Guideline.
  • 42. Efficacy Guidelines  The work carried out by ICH under the Efficacy heading is concerned with the design, conduct, safety and reporting of clinical trials. It also covers novel types of medicines derived from biotechnological processes and the use of pharmacogenetics/ pharmacogenomics techniques to produce better targeted medicines.
  • 43. Efficacy Guidelines  Clinical Safety E1-E2F  Clinical Study Reports E3  Dose Response Studies E4  Ethnic Factors E5  Good Clinical Practice E6  Clinical Trials E7-E11  Guidelines for Clinical Evaluation by Therapeutic Category E12  Clinical Evaluation E14  Pharmacogenomics E15-E16  Joint Safety / Efficacy Topic M3
  • 44. E1 (Oct. 1994)  E1 – The Extent Of Population Exposure To Assess Clinical Safety For Drugs Intended For Long-term Treatment Of Non-life- threatening Conditions  The tripartite harmonized ICH Guideline was finalised under Step 4 in October 1994. This document gives recommendations on the numbers of patients and duration of exposure for the safety evaluation of drugs intended for the long-term treatment of non-life-threatening conditions.  Events where the rate of occurrence changes over a longer period of time may need to be characterized depending on their severity and importance to the risk-benefit assessment of the drug.  The safety evaluation during clinical drug development is not expected to characterise rare adverse events, for example, those occurring in less than 1 in 1000 patients.
  • 45. E2A (Oct. 1994)  Clinical Safety Data Management: Definitions And Standards For Expedited Reporting E2A  There are two issues within the broad subject of clinical safety data management that are appropriate for harmonization at this time:  (1) the development of standard definitions and terminology for key aspects of clinical safety reporting, and  (2) the appropriate mechanism for handling expedited (rapid) reporting, in the investigational (i.e., pre-approval) phase.
  • 46. E2B-R2 (Feb. 2001)  Maintenance Of The ICH Guideline On Clinical Safety Data Management : Data Elements For Transmission Of Individual Case Safety Reports E2B(R2)  to standardize the data elements for transmission of individual case safety reports by identifying, and where necessary or advisable, by defining the data elements for the transmission of all types of individual case safety reports, regardless of source and destination.
  • 47. E2C-R1(Nov. 1996)  Clinical Safety Data Management: Periodic Safety Update Reports for Marketed Drugs E2C (R1)  guidance on the format and content of safety updates, which need to be provided at intervals to regulatory authorities after products have been marketed. The Guideline is intended to ensure that the worldwide safety experience is provided to authorities at defined times after marketing with maximum efficiency and avoiding duplication of effort.  E2C(R2) This Revision was endorsed by Steering Committee in Dec.2010. It will Evaluate the ICH Pharmacovigilance documentation, conduct a gap and potential improvement analysis of ECH E2C, E2E and E2F and draft a new ICH E2C R2 covering periodic benefic risk evaluation reporting.
  • 48. E2D (Nov. 2003)  Post Approval Safety Data Management: Definitions and Standards for Expedited Reporting  provides a standardized procedure for post-approval safety data management including expedited reporting to relevant authority.  The definitions of the terms and concept specific to post- approval phase are also provided.
  • 49. E2E (Nov. 2004)  Pharmacovigilance Planning  planning of pharmacovigilance activities, especially in preparation for the early post marketing period of a new drug (chemical entities, biotech-derived products, vaccines)  Main focus: Safety specification and PV plan that mighty be submitted at the time of license application
  • 50. E2F (Aug. 2010)  Development Safety Update Report  The main focus of the DSUR is data from interventional clinical trials (referred to in this document as "clinical trials") of investigational drugs including biologicals, with or without a marketing approval, whether conducted by commercial or non-commercial sponsors.  Intended for periodic reporting on drugs under development (including marketed drugs that are under further study) among the ICH regions.
  • 51. E3 (Nov. 1995)  Structure and Content of Clinical Study Reports  is an "integrated" full report of an individual study of any therapeutic, prophylactic or diagnostic agent (referred to herein as drug or treatment) conducted in patients, in which the clinical and statistical description, presentations, and analyses are integrated into a single report, incorporating tables and figures into the main text of the report, or at the end of the text, and with appendices containing the protocol, sample case report forms, investigator related information, information related to the test drugs/investigational products including active control/comparators, technical statistical documentation, related publications, patient data listings, and technical statistical details such as derivations, computations, analyses, and computer output etc.
  • 52. E4 (March, 1994)  Dose-response Information To Support Drug Registration  Knowledge of the relationships among dose, drug- concentration in blood, and clinical response (effectiveness and undesirable effects) is important for the safe and effective use of drugs in individual patients.  concepts of minimum effective dose and maximum useful dose do not adequately account for individual differences and do not allow a comparison, at various doses, of both beneficial and undesirable effects.  Any given dose provides a mixture of desirable and undesirable effects, with no single dose necessarily optimal for all patients.
  • 53. E5 R1 (Feb. 1998)  Ethnic Factors In The Acceptability Of Foreign Clinical Data  This document addresses the intrinsic characteristics of the drug recipient and extrinsic characteristics associated with environment and culture that could affect the results of clinical studies carried out in regions and describes the concept of the "bridging study" that a new region may request to determine whether data from another region are applicable to its population.
  • 54. E6-R1 (May 1996)  Good Clinical Practice  describes the responsibilities and expectations of all participants in the conduct of clinical trials, including investigators, monitors, sponsors and IRBs.  GCPs cover aspects of monitoring, reporting and archiving of clinical trials and incorporating addenda on the Essential Documents and on the Investigator's Brochure which had been agreed earlier through the ICH process.
  • 55. E7 (June 1993)  Studies in Support of Special Populations: Geriatrics  This document provides recommendations on the special considerations which apply in the design and conduct of clinical trials of medicines that are likely to have significant use in the elderly.  It requires special consideration due to the frequent occurrence of underlying diseases, concomitant drug therapy and the consequent risk of drug interaction.
  • 56. E8 (July 1997)  General Considerations for Clinical Trials  This document sets out the general scientific principles for the conduct, performance and control of clinical trials.  The Guideline addresses a wide range of subjects in the design and execution of clinical trials.
  • 57. E9 (Feb. 1998)  Statistical Principles for Clinical Trials  This biostatistical Guideline describes essential considerations on the design and analysis of clinical trials, especially the "confirmatory" (hypothesis-testing) trials that are the basis for demonstrating effectiveness.  The document will also assist scientific experts charged with preparing application summaries or assessing evidence of efficacy and safety, principally from clinical trials in later phases of development.
  • 58. E10 (July 2000)  Choice of Control Group and Related Issues in Clinical Trials  This document addresses the choice of control groups in clinical trials considering the ethical and inferential properties and limitations of different kinds of control groups. It points out the assay sensitivity problem in active control equivalence / non-inferiority trials that limits the usefulness of trial design in many circumstances.
  • 59. E11 (July 2000)  Clinical Investigation of Medicinal Products in the Pediatric Population  This document addresses the conduct of clinical trials of medicines in pediatric populations. This document will facilitate the development of safe and effective use of medicinal product in pediatrics.
  • 60. E12 (March 2000)  Principles for Clinical Evaluation of New antihypertensive Drugs  It provides a set of "Principles" on which there is general agreement among all three ICH regions covering endpoints and trial designs.  It will not be subject to the usual procedures leading to a fully harmonized document.
  • 61. E14 (May 2005)  The Clinical Evaluation of QT/QTc Interval Prolongation and Proarrhythmic Potential for Non-Antiarrhythmic Drugs.  This document provides recommendations to sponsors concerning the design, conduct, analysis, and interpretation of clinical studies to assess the potential of a drug to delay cardiac repolarisation.  This assessment should include testing the effects of new agents on the QT/QTc interval as well as the collection of cardiovascular adverse events. The investigational approach used for a particular drug should be individualised, depending on the pharmacodynamic, pharmacokinetic, and safety characteristics of the product, as well as on its proposed clinical use.  The assessment of the effects of drugs on cardiac repolarisation is the subject of active investigation.  When additional data (non-clinical and clinical) are accumulated in the future, this document may be reevaluated and revised.
  • 62. E15 (Nov. 2007)  Definitions For Genomic Biomarkers, Pharmaco- genomics, Pharmacogenetics, Genomic Data And Sample Coding Categories  Pharmacogenomics and pharmacogenetics have the potential to improve the discovery, development and use of medicines.
  • 63. E16 (Aug. 2010)  Biomarkers Related To Drug Or Biotechnology Product Development: Context, Structure And Format Of Qualification Submissions  The Guideline describes recommendations regarding context, structure, and format of regulatory submissions for qualification of genomic biomarkers, as defined in ICH E15.
  • 64. M3 R2 (June 2009)  Guidance On Non-clinical Safety Studies For The Conduct Of Human Clinical Trials And Marketing Authorization For Pharmaceuticals  Harmonisation of the guidance for non-clinical safety studies will help to define the current recommendations and reduce the likelihood that substantial differences will exist among regions.