INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE. Current Step 4 version



Similar documents
Guideline for Industry

GENERAL CONSIDERATIONS FOR CLINICAL TRIALS E8

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE S1A. Current Step 4 version

ICH Topic E 8 General Considerations for Clinical Trials. Step 5 NOTE FOR GUIDANCE ON GENERAL CONSIDERATIONS FOR CLINICAL TRIALS (CPMP/ICH/291/95)

Risk Management Plan (RMP) Guidance (Draft)

Not All Clinical Trials Are Created Equal Understanding the Different Phases

Adoption by CHMP for release for consultation November End of consultation (deadline for comments) 31 March 2011

Guidance for Industry

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON DATA MONITORING COMMITTEES

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS GUIDELINE FOR THE CONDUCT OF POST-MARKETING SURVEILLANCE STUDIES OF VETERINARY MEDICINAL PRODUCTS

First In Human Pediatric Trials and Safety Assessment for Rare and Orphan Diseases

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE E15

RAPID ALERT SYSTEM (RAS) IN PHARMACOVIGILANCE

NONCLINICAL EVALUATION FOR ANTICANCER PHARMACEUTICALS

Sheffield Kidney Institute. Planning a Clinical Trial

Guidance for Industry Diabetes Mellitus Evaluating Cardiovascular Risk in New Antidiabetic Therapies to Treat Type 2 Diabetes

IMPURITIES IN NEW DRUG PRODUCTS

CHOICE OF CONTROL GROUP AND RELATED ISSUES IN CLINICAL TRIALS E10

Guidance for Industry Migraine: Developing Drugs for Acute Treatment

The Product Review Life Cycle A Brief Overview

E2C(R2) Implementation Working Group ICH E2C(R2) Guideline: Periodic Benefit-Risk Evaluation Report Questions & Answers

U.S. Food and Drug Administration

Guideline on good pharmacovigilance practices (GVP)

Rare Diseases: Common Issues in Drug Development Guidance for Industry

POST-APPROVAL SAFETY DATA MANAGEMENT: DEFINITIONS AND STANDARDS FOR EXPEDITED REPORTING E2D

ICH guideline E2C (R2) Periodic benefit-risk evaluation report (PBRER)

Guidance for Industry

Regulatory Pathways for Licensure and Use of Ebola Virus Vaccines During the Current Outbreak FDA Perspective

RADIOPHARMACEUTICALS BASED ON MONOCLONAL ANTIBODIES

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE

Guidance for Industry Determining the Extent of Safety Data Collection Needed in Late Stage Premarket and Postapproval Clinical Investigations

ICH Topic S 1 A The Need for Carcinogenicity Studies of Pharmaceuticals. Step 5

4.1 Objectives of Clinical Trial Assessment

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) DRAFT. Guideline on Risk Management Systems for Medicinal Products for Human Use

M4E(R2): The CTD Efficacy

VALIDATION OF ANALYTICAL PROCEDURES: TEXT AND METHODOLOGY Q2(R1)

Guideline for Industry

Glossary of Clinical Trial Terms

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON RISK MANAGEMENT SYSTEMS FOR MEDICINAL PRODUCTS FOR HUMAN USE

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE

Clinical Trials and Safety Surveillance of Drugs in Development

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP)

Subject: No. Page PROTOCOL AND CASE REPORT FORM DEVELOPMENT AND REVIEW Standard Operating Procedure

Clinical trials: from European perspective to National implementation. CTFG / FAMHP / pharma.be. Brussels, 19 November 2010

Reflection paper on clinical aspects related to tissue engineered products

PHARMACEUTICAL QUALITY SYSTEM Q10

Clinical trials in haemophilia

Guidance on Investigational Medicinal Products (IMPs) and other medicinal products used in Clinical Trials

Risk Management in the Pharmaceutical Industry. Elena Apetri, Global Medical Safety Surveillance Schering AG

OECD Recommendation on the Governance of Clinical Trials

February 2006 Procedural

Guidance on adverse drug reactions

POLICY AND PROCEDURES OFFICE OF NEW DRUGS. Good Review Practice: Management of Breakthrough Therapy-Designated Drugs and Biologics.

EU DIRECTIVE ON GOOD CLINICAL PRACTICE IN CLINICAL TRIALS DH & MHRA BRIEFING NOTE

Guidance for Industry

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE E2C(R2)

Guidance for Industry

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON THE EVALUATION OF MEDICINAL PRODUCTS FOR CARDIOVASCULAR DISEASE PREVENTION

Introduction to Post marketing Drug Safety Surveillance: Pharmacovigilance in FDA/CDER

Guidance for Industry

TRANSATLANTIC TRADE AND INVESTMENT PARTNERSHIP

WHO guideline for abbreviated licensing pathways for certain biological therapeutic products

Medicine Safety Glossary

The use of the WHO-UMC system for standardised case causality assessment

Guidance for Industry and Investigators

STABILITY TESTING: PHOTOSTABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) NOTE FOR GUIDANCE ON CLINICAL INVESTIGATION OF MEDICINAL PRODUCTS IN THE TREATMENT OF DEPRESSION

EUROPEAN COMMISSION HEALTH AND CONSUMERS DIRECTORATE-GENERAL

Clinical Study Synopsis

Guidance for Industry Expedited Programs for Serious Conditions Drugs and Biologics

Guideline on good pharmacovigilance practices (GVP)

September 19, 1984 FOOD PRODUCTION AND DIRECTION GÉNÉRALE, SECTION INSPECTION BRANCH PRODUCTION ET INSPECTION PESTICIDES DES ALIMENTS TRADE MEMORANDUM

Guidance for Industry

SAFETY PHARMACOLOGY STUDIES FOR HUMAN PHARMACEUTICALS S7A

Communicating uncertainty about benefits and

Risk Assessment in the Pharmaceutical Industry. Dorothea Köppe Liability Insurance: The Never-ending Story September 9th, 2009

Ethics and Scientific Oversight for Phase 1 Clinical Trials in Hong Kong. Sydney TANG Chairman, HKU/HA HKW IRB November 21, 2015

Overview of Phase 1 Oncology Trials of Biologic Therapeutics

National Institute of Public Health

The Trans-Tasman Early Warning System. Processes in Australia and New Zealand

Guidance for Industry

FSH Society s 2014 Biennial FSHD Connect Meeting: Natural History Studies

Response of the German Medical Association

Safety Assessment for IND Safety Reporting Guidance for Industry

Classification for Drug related problems

Challenges in the Regulation of Pediatric Clinical Trials

PHARMACOVIGILANCE PLANNING E2E

Overview of CDISC Implementation at PMDA. Yuki Ando Senior Scientist for Biostatistics Pharmaceuticals and Medical Devices Agency (PMDA)

Avastin in Metastatic Breast Cancer

Clinical Research in Mauritius

NATIONAL HEALTH COUNCIL RESOLUTION Nº 251, DATED 7 AUGUST 1997

Goals & Objectives. Drug Development & the FDA Pharmacy 309. Outline. An History of Disasters. Be able to describe

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE M3(R2)

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE Q4B ANNEX 12

PUBLIC HEALTH PROGRAMME GUIDANCE DRAFT SCOPE

Basic Study Designs in Analytical Epidemiology For Observational Studies

GUIDELINES ON MEDICAL DEVICES

Selected Requirements of Prescribing Information

National Environment Protection (Ambient Air Quality) Measure. Appendix 6

Comparative analysis between the possible regulatory approaches to GMP compliance TITOLO PRESENTAZIONE

Transcription:

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE ICH HARMONISED TRIPARTITE GUIDELINE THE EXTENT OF POPULATION EXPOSURE TO ASSESS CLINICAL SAFETY FOR DRUGS INTENDED FOR LONG-TERM TREATMENT OF NON-LIFE-THREATENING CONDITIONS Current Step 4 version dated 27 October 1994 This Guideline has been developed by the appropriate ICH Expert Working Group and has been subject to consultation by the regulatory parties, in accordance with the ICH Process. At Step 4 of the Process the final draft is recommended for adoption to the regulatory bodies of the European Union, Japan and USA.

Document History First Codification History Date New Codification November 2005 Approval by the Steering Committee under Step 2 and release for public consultation. 27 October 1993 Current Step 4 version Approval by the Steering Committee under Step 4 and recommendation for adoption to the three ICH regulatory bodies. 26 October 1994

THE EXTENT OF POPULATION EXPOSURE TO ASSESS CLINICAL SAFETY FOR DRUGS INTENDED FOR LONG-TERM TREATMENT OF NON-LIFE-THREATENING CONDITIONS ICH Harmonised Tripartite Guideline Having reached Step 4 of the ICH Process at the ICH Steering Committee meeting on 27 October 1994, this guideline is recommended for adoption to the three regulatory parties to ICH The objective of this guideline is to present an accepted set of principles for the safety evaluation of drugs intended for the long-term treatment (chronic or repeated intermittent use for longer than 6 months) of non-life-threatening diseases. The safety evaluation during clinical drug development is expected to characterise and quantify the safety profile of a drug over a reasonable duration of time consistent with the intended long-term use of the drug. Thus, duration of drug exposure and its relationship to both time and magnitude of occurrence of adverse events are important considerations in determining the size of the data base necessary to achieve such goals. For the purpose of this guideline, it is useful to distinguish between clinical data on adverse drug events (ADEs) derived from studies of shorter duration of exposure and data from studies of longer duration, which frequently are non-concurrently controlled studies. It is expected that short-term event rates (cumulative 3-month incidence of about 1%) will be well characterised. Events where the rate of occurrence changes over a longer period of time may need to be characterised 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. The design of the clinical studies can significantly influence the ability to make causality judgements about the relationships between the drug and adverse events. A placebo-controlled trial allows the adverse event rate in the drug-treated group to be compared directly with the background event rate in the patient population being studied. Although a study with a positive or active control will allow a comparison of adverse event rates to be made between the test drug and the control drug, no direct assessment of the background event rate in the population studied can be made. A study that has no concurrent control group makes it more difficult to assess the causality relationship between adverse events observed and the test drug. There was general agreement on the following: 1. A harmonised regulatory standard is of value for the extent and duration of treatment needed to provide the safety data base for drugs intended for long-term treatment of non-life-threatening conditions. Although this standard covers many indications and drug classes, there are exceptions. 2. Regulatory standards for the safety evaluation of drugs should be based on previous experience with the occurrence and detection of adverse drug events (ADEs), statistical considerations of the probability of detecting specified frequencies of ADEs, and practical considerations. 1

Population Exposure 3. Information about the occurrence of ADEs in relation to duration of treatment for different drug classes is incomplete, and further investigations to obtain this information would be useful. 4. Available information suggests that most ADEs first occur, and are most frequent, within the first few months of drug treatment. The number of patients treated for 6 months at dosage levels intended for clinical use, should be adequate to characterise the pattern of ADEs over time. To achieve this objective the cohort of exposed subjects should be large enough to observe whether more frequently occurring events increase or decrease over time as well as to observe delayed events of reasonable frequency (e.g., in the general range of 0.5%-5%). Usually 300-600 patients should be adequate. 5. There is concern that, although they are likely to be uncommon, some ADEs may increase in frequency or severity with time or that some serious ADEs may occur only after drug treatment for more than 6 months. Therefore, some patients should be treated with the drug for 12 months. In the absence of more information about the relationship of ADEs to treatment duration, selection of a specific number of patients to be followed for 1 year is to a large extent a judgement based on the probability of detecting a given ADE frequency level and practical considerations. 100 patients exposed for a minimum of one-year is considered to be acceptable to include as part of the safety data base. The data should come from prospective studies appropriately designed to provide at least one year exposure at dosage levels intended for clinical use. When no serious ADE is observed in a one-year exposure period this number of patients can provide reasonable assurance that the true cumulative one year incidence is no greater than 3%. 6. It is anticipated that the total number of individuals treated with the investigational drug, including short-term exposure, will be about 1500. Japan currently accepts 500-1500 patients: the potential for a smaller number of patients is due to the post-marketing surveillance requirement, the actual number for a specific drug being determined by the information available on the drug and drug class. 7. There are a number of circumstances where the harmonised general standards for the clinical safety evaluation may not be applicable. Reasons for, and examples of, these exceptions are listed below. It is expected that additional examples may arise. It should also be recognised that the clinical data base required for efficacy testing may be occasionally larger or may require longer patient observation than that required by this guideline. Exceptions: a. Instances where there is concern that the drug will cause late developing ADEs, or cause ADEs that increase in severity or frequency over time, would require a larger and/or longer-term safety data base. The concern could arise from: (1) data from animal studies; (2) clinical information from other agents with related chemical structures or from a related pharmacologic class; (3) pharmacokinetic or pharmacodynamic properties known to be associated with such ADEs. 2

Population Exposure b. Situations in which there is a need to quantitate the occurrence rate of an expected specific low-frequency ADE will require a greater long-term data base. Examples would include situations where a specific serious ADE has been identified in similar drugs or where a serious event that could represent an alert event is observed in early clinical trials. c. Larger safety data bases may be needed to make risk/benefit decisions in situations where the benefit from the drug is either (1) small (e.g., symptomatic improvement in less serious medical conditions) or (2) will be experienced by only a fraction of the treated patients (e.g., certain preventive therapies administered to healthy populations) or (3) is of uncertain magnitude (e.g., efficacy determination on a surrogate endpoint). d. In situations where there is concern that a drug may add to an already significant background rate of morbidity or mortality, clinical trials may need to be designed with a sufficient number of patients to provide adequate statistical power to detect prespecified increases over the baseline morbidity or mortality. e. In some cases, a smaller number of patients may be acceptable, for example, where the intended treatment population is small. 8. Filing for approval will usually be possible based on the data from patients treated through 6 months. Data on patients treated through 12 months must be submitted as soon as available and prior to approval in the United States and Japan but may be submitted after approval in the E.C.. In the U.S. the initial submission for those drugs designated as priority drugs must include the 12- months patient data. 3