97-12139. International Conference on Harmonisation; Draft Guideline on Statistical Principles for Clinical Trials; Availability  

  • [Federal Register Volume 62, Number 90 (Friday, May 9, 1997)]
    [Notices]
    [Pages 25712-25726]
    From the Federal Register Online via the Government Publishing Office [www.gpo.gov]
    [FR Doc No: 97-12139]
    
    
    
    [[Page 25711]]
    
    _______________________________________________________________________
    
    Part III
    
    
    
    
    
    Department of Health and Human Services
    
    
    
    
    
    _______________________________________________________________________
    
    
    
    Food and Drug Administration
    
    
    
    _______________________________________________________________________
    
    
    
    International Conference on Harmonisation; Draft Guideline on 
    Statistical Principles for Clinical Trials; Notice of Availability
    
    Federal Register / Vol. 62, No. 90 / Friday, May 9, 1997 / Notices
    
    [[Page 25712]]
    
    
    =======================================================================
    -----------------------------------------------------------------------
    
    
    DEPARTMENT OF HEALTH AND HUMAN SERVICES
    
    Food and Drug Administration
    [Docket No. 97D-0174]
    
    
    International Conference on Harmonisation; Draft Guideline on 
    Statistical Principles for Clinical Trials; Availability
    
    AGENCY: Food and Drug Administration, HHS.
    
    ACTION: Notice.
    
    -----------------------------------------------------------------------
    
    SUMMARY: The Food and Drug Administration (FDA) is publishing a draft 
    guideline entitled ``Statistical Principles for Clinical Trials.'' The 
    draft guideline was prepared under the auspices of the International 
    Conference on Harmonisation of Technical Requirements for Registration 
    of Pharmaceuticals for Human Use (ICH). The draft guideline is intended 
    to provide recommendations to sponsors and scientific experts regarding 
    statistical principles and methodology which, when applied to clinical 
    trials for marketing applications, will facilitate the general 
    acceptance of analyses and conclusions drawn from the trials.
    
    DATES: Written comments by June 23, 1997.
    
    ADDRESSES: Submit written comments on the draft guideline to the 
    Dockets Management Branch (HFA-305), Food and Drug Administration, 
    12420 Parklawn Dr., rm. 1-23, Rockville, MD 20857. Copies of the draft 
    guideline are available from the Drug Information Branch (HFD-210), 
    Center for Drug Evaluation and Research, Food and Drug Administration, 
    5600 Fishers Lane, Rockville, MD 20857, 301-827-4573. Single copies of 
    the draft guideline may be obtained by mail from the Office of 
    Communication, Training and Manufacturers Assistance (HFM-40), Center 
    for Biologics Evaluation and Research (CBER), 1401 Rockville Pike, 
    Rockville, MD 20852-1448 or by calling the CBER Voice Information 
    System at 1-800-835-4709 or 301-827-1800. Copies may be obtained from 
    CBER's FAX Information System at 1-888-CBER-FAX or 301-827-3844.
    
    FOR FURTHER INFORMATION CONTACT:
        Regarding the guideline: Robert T. O'Neill, Center for Drug 
    Evaluation and Research (HFD-700), Food and Drug Administration, 5600 
    Fishers Lane, Rockville, MD 20857, 301-827-3195.
        Regarding the ICH: Janet J. Showalter, Office of Health Affairs 
    (HFY-20), Food and Drug Administration, 5600 Fishers Lane, Rockville, 
    MD 20857, 301-827-0864.
    
    SUPPLEMENTARY INFORMATION: In recent years, many important initiatives 
    have been undertaken by regulatory authorities and industry 
    associations to promote international harmonization of regulatory 
    requirements. FDA has participated in many meetings designed to enhance 
    harmonization and is committed to seeking scientifically based 
    harmonized technical procedures for pharmaceutical development. One of 
    the goals of harmonization is to identify and then reduce differences 
    in technical requirements for drug development among regulatory 
    agencies.
        ICH was organized to provide an opportunity for tripartite 
    harmonization initiatives to be developed with input from both 
    regulatory and industry representatives. FDA also seeks input from 
    consumer representatives and others. ICH is concerned with 
    harmonization of technical requirements for the registration of 
    pharmaceutical products among three regions: The European Union, Japan, 
    and the United States. The six ICH sponsors are the European 
    Commission, the European Federation of Pharmaceutical Industries 
    Associations, the Japanese Ministry of Health and Welfare, the Japanese 
    Pharmaceutical Manufacturers Association, the Centers for Drug 
    Evaluation and Research and Biologics Evaluation and Research, FDA, and 
    the Pharmaceutical Research and Manufacturers of America. The ICH 
    Secretariat, which coordinates the preparation of documentation, is 
    provided by the International Federation of Pharmaceutical 
    Manufacturers Associations (IFPMA).
        The ICH Steering Committee includes representatives from each of 
    the ICH sponsors and the IFPMA, as well as observers from the World 
    Health Organization, the Canadian Health Protection Branch, and the 
    European Free Trade Area.
        On January 17, 1997, the ICH Steering Committee agreed that a draft 
    guideline entitled ``Statistical Principles for Clinical Trials'' 
    should be made available for public comment. The draft guideline is the 
    product of the Efficacy Expert Working Group of the ICH. Comments about 
    this draft will be considered by FDA and the other regulatory agency 
    members of the Efficacy Expert Working Group.
        The draft guideline addresses principles of statistical methodology 
    applied to clinical trials for marketing applications. The draft 
    guideline provides recommendations to sponsors in the design, conduct, 
    analysis, and evaluation of clinical trials of an investigational 
    product in the context of its overall clinical development. The draft 
    guideline also provides guidance to scientific experts in preparing 
    application summaries or assessing evidence of efficacy and safety, 
    principally from late Phase II and Phase III clinical trials. 
    Application of the principles of statistical methodology is intended to 
    facilitate the general acceptance of analyses and conclusions drawn 
    from clinical trials.
        This draft guideline represents the agency's current thinking on 
    statistical principles for clinical trials of drugs and biologics. It 
    does not create or confer any rights for or on any person and does not 
    operate to bind FDA or the public. An alternative approach may be used 
    if such approach satisfies the requirements of the applicable statute, 
    regulations, or both.
        Interested persons may, on or before June 23, 1997, submit to the 
    Dockets Management Branch (address above) written comments on the draft 
    guideline. Two copies of any comments are to be submitted, except that 
    individuals may submit one copy. Comments are to be identified with the 
    docket number found in brackets in the heading of this document. The 
    draft guideline and received comments may be seen in the office above 
    between 9 a.m. and 4 p.m., Monday through Friday.
        An electronic version of this draft guideline is available on the 
    Internet using the World Wide Web (WWW) (http://www.fda.gov/cder/
    guidance.htm) or through the CBER home page (http://www.fda.gov/cber/
    cberftp.html).
        The text of the draft guideline follows:
    
    Statistical Principles for Clinical Trials
    
        Note: A Glossary of terms and definitions is provided as an 
    annex to this guideline.
    
    Table of Contents
    
    I. Introduction
        1.1 Background and Purpose
        1.2 Scope and Direction
    II. Considerations for Overall Clinical Development
        2.1 Study Context
          2.1.1 Development Plan
          2.1.2 Confirmatory Trial
          2.1.3 Exploratory Trial
        2.2 Study Scope
          2.2.1 Population
          2.2.2 Primary and Secondary Variables
        2.3 Design Techniques to Avoid Bias
          2.3.1 Blinding
          2.3.2 Randomization
    III. Study Design Considerations
        3.1 Study Configuration
          3.1.1 Parallel Group Design
          3.1.2 Cross-Over Design
          3.1.3 Factorial Designs
    
    [[Page 25713]]
    
        3.2 Multicenter Trials
        3.3 Type of Comparison
          3.3.1 Trials to Show Superiority
          3.3.2 Trials to Show Equivalence or Non-inferiority
          3.3.3 Dose-Response Designs
        3.4 Group Sequential Designs
        3.5 Sample Size
        3.6 Data Capture and Processing
    IV. Study Conduct
        4.1 Trial Monitoring
        4.2 Changes in Inclusion and Exclusion Criteria
        4.3 Accrual Rates
        4.4 Sample Size Adjustment
        4.5 Interim Analysis and Early Stopping
        4.6 Role of Independent Data Monitoring Committee (IDMC)
    V. Data Analysis
        5.1 Prespecified Analysis Plan
        5.2 Analysis Sets
          5.2.1 All Randomized Subjects
          5.2.2 Per Protocol Subjects
          5.2.3 Roles of the All Randomized Subjects Analysis and the 
    Per Protocol Analysis
        5.3 Missing Values and Outliers
        5.4 Data Transformation/Modification
        5.5 Estimation, Confidence Intervals and Hypothesis Testing
        5.6 Adjustment of Type I Error and Confidence Levels
        5.7 Subgroups, Interactions and Covariates
        5.8 Integrity of Data and Computer Software
    VI. Evaluation of Safety and Tolerability
        6.1 Scope of Evaluation
        6.2 Choice of Variables and Data Collection
        6.3 Set of Subjects to be Evaluated and Presentation of Data
        6.4 Statistical Evaluation
        6.5 Single Study versus Integrated Summary
    VII. Reporting
        7.1 Evaluation and Reporting
        7.2 Summarizing the Clinical Database
          7.2.1 Efficacy Data
          7.2.2 Safety Data
    Annex 1 Glossary
    
    I. Introduction
    
    1.1 Background and Purpose
    
        The efficacy and safety of medicinal products should be 
    demonstrated by clinical trials that follow the guidance in ``Good 
    Clinical Practice: Consolidated Guideline (E6)'' adopted by the ICH, 
    May 1, 1996. The role of statistics in clinical trial design and 
    analysis is acknowledged as essential in that ICH guideline. The 
    proliferation of statistical research in the area of clinical trials 
    coupled with the critical role of clinical research in the drug 
    approval process and health care in general necessitate a succinct 
    document on statistical issues related to clinical trials. This 
    guideline is written primarily to attempt to harmonize the 
    principles of statistical methodology applied to clinical trials for 
    marketing applications submitted in Europe, Japan, and the United 
    States.
        As a starting point, this guideline utilized the CPMP (Committee 
    for Proprietary Medicinal Products) Note for Guidance entitled 
    ``Biostatistical Methodology in Clinical Trials in Applications for 
    Marketing Authorizations for Medicinal Products'' (December 1994). 
    It was also influenced by ``Guidelines on the Statistical Analysis 
    of Clinical Studies'' (March 1992) from the Japanese Ministry of 
    Health and Welfare and the U.S. FDA document entitled ``Guideline 
    for the Format and Content of the Clinical and Statistical Sections 
    of New Drug Applications'' (July 1988). Some topics related to 
    statistical principles and methodology are also embedded within 
    other ICH guidelines, particularly those listed below. The specific 
    guideline that contains related text will be identified in various 
    sections of this document.
        E1: The Extent of Population Exposure to Assess Clinical Safety
        E2A: Clinical Safety Data Management: Definitions and Standards 
    for Expedited Reporting
        E2B: Clinical Safety Data Management: Data Elements for 
    Transmission of Individual Case Safety Reports
        E2C: Clinical Safety Data Management: Periodic Safety Update 
    Reports for Marketed Drugs
        E3: Structure and Content of Clinical Study Reports
        E4: Dose-Response Information to Support Drug Registration
        E5: Ethnic Factors in the Acceptability of Foreign Clinical Data
        E6: Good Clinical Practice: Consolidated Guideline
        E7: Studies in Support of Special Populations: Geriatrics
        E8: General Considerations for Clinical Trials
        E10: Choice of Control Group in Clinical Trials
        M1: Standardization of Medical Terminology for Regulatory 
    Purposes
        M3: Nonclinical Safety Studies for the Conduct of Human Clinical 
    Trials for Pharmaceuticals
        This guideline is intended to give direction to sponsors in the 
    design, conduct, analysis, and evaluation of clinical trials of an 
    investigational product in the context of its overall clinical 
    development. The document will also assist scientific experts 
    charged with preparing application summaries or assessing evidence 
    of efficacy and safety, principally from late Phase II and Phase III 
    clinical trials.
    
    1.2 Scope and Direction
    
        The focus of this guideline is on statistical principles. It 
    does not address the use of specific statistical procedures or 
    methods. Specific procedural steps to ensure that principles are 
    implemented properly are the responsibility of the sponsor. 
    Integration of data across clinical trials is discussed, but is not 
    a primary focus of this guideline. Selected principles and 
    procedures related to data management or clinical trial monitoring 
    activities are covered in other ICH guidelines and are not addressed 
    here.
        This guideline should be of interest to individuals from a broad 
    range of scientific disciplines. However, it is assumed that the 
    actual responsibility for all statistical work associated with 
    clinical trials will lie with an appropriately qualified and 
    experienced statistician, as indicated in the ``ICH Guideline for 
    Good Clinical Practice.'' The involvement of the statistician, in 
    collaboration with other clinical trial professionals, is to ensure 
    that statistical principles are applied appropriately in clinical 
    trials supporting drug development. Thus, the statistician should 
    have a combination of education/training and experience sufficient 
    to implement the principles articulated in this guideline.
        All important details of the design, conduct, and proposed 
    analysis of each clinical trial contributing to a marketing 
    application should be clearly specified in a protocol written before 
    the trial begins. The extent to which the procedures in the protocol 
    are followed and the primary analysis is planned a priori will 
    contribute to the degree of confidence in the final results and 
    conclusions of the trial. The protocol and subsequent amendments 
    should be approved by the responsible personnel, including the trial 
    statistician. The trial statistician should ensure that the protocol 
    and any amendments cover all relevant statistical issues clearly and 
    accurately, using technical terminology as appropriate.
        The principles outlined in this guideline are primarily relevant 
    to clinical trials conducted in the later phases of development, 
    many of which are confirmatory trials of efficacy. In addition to 
    efficacy, confirmatory trials may have as their primary variable a 
    safety variable (e.g., an adverse event, a clinical laboratory 
    variable, or an electrocardiographic measure) or a pharmacodynamic 
    or pharmacokinetic variable (as in a confirmatory bioequivalence 
    trial). Furthermore, some confirmatory findings may be derived from 
    data integrated across studies, and selected principles in this 
    guideline are applicable in this situation. Finally, although the 
    early phases of drug development consist mainly of clinical trials 
    that are exploratory in nature, statistical principles are also 
    relevant to these clinical trials. Hence, the substance of this 
    document should be applied as far as possible to all phases of 
    clinical development.
        Many of the principles delineated in this guideline deal with 
    minimizing bias and maximizing precision. As used in this guideline, 
    the term ``bias'' describes the systematic tendency of any factors 
    associated with the design, conduct, analysis, and interpretation of 
    the results of clinical trials to make the estimate of a treatment 
    effect deviate from its true value. It is important to identify 
    potential sources of bias to the extent possible so that attempts to 
    limit such bias may be made. The presence of bias may seriously 
    compromise the ability to draw valid conclusions from clinical 
    studies.
        Some sources of bias arise from the design of the trial, for 
    example an assignment of treatments such that subjects at lower risk 
    are systematically assigned to one treatment. Other sources of bias 
    arise during the conduct and analysis of a clinical trial. For 
    example, protocol violations and exclusion of subjects from analysis 
    based upon knowledge of subject outcomes are possible sources of 
    bias that may affect the accurate assessment of treatment effect. 
    Because bias can occur in subtle or unknown ways and its effect is 
    not measurable directly, it is important to evaluate the robustness 
    of the results and
    
    [[Page 25714]]
    
    primary conclusions of the trial. Robustness is a concept that 
    refers to the sensitivity of the overall conclusions to various 
    limitations of the data, assumptions, and analytic approaches to 
    data analysis. Robustness implies that, if a variety of analyses of 
    the data that take into account changing assumptions were to be 
    performed, the treatment effect and primary conclusions of the trial 
    would be consistent. The interpretation of statistical measures of 
    uncertainty of the treatment effect and treatment comparisons should 
    involve consideration of the potential contribution of bias to the 
    p-value, confidence interval, or inference.
        This guideline largely refers to the use of frequentist methods 
    when discussing hypothesis testing and/or confidence intervals. 
    However, the use of Bayesian or other approaches may be considered 
    when the reasons for their use are clear and when the resulting 
    conclusions are sufficiently robust compared to alternative 
    assumptions.
    
    II. Considerations for Overall Clinical Development
    
    2.1 Study Context
    
    2.1.1 Development Plan
    
        The broad aim of the process of clinical development of a new 
    drug is to find out whether there is a dose range and schedule at 
    which the drug can be shown to be simultaneously safe and effective, 
    to the extent that the risk-benefit relationship is acceptable. The 
    particular subjects who may benefit from the drug and the specific 
    indications for its use also need to be defined.
        Satisfying these broad aims usually requires an ordered program 
    of clinical trials, each with its own specific objectives. This 
    should be specified in a clinical plan, or a series of plans, with 
    appropriate decision points and flexibility to allow modification as 
    knowledge accumulates. A marketing application should clearly 
    describe the main content of such plans, and the contribution made 
    by each trial. Interpretation and assessment of the evidence from 
    the total program of trials involves synthesis of the evidence from 
    the individual trials (see section 7.2). This is facilitated by 
    ensuring that common standards are adopted for a number of features 
    of the trials, such as dictionaries of medical terms, definition and 
    timing of the main measurements, handling of protocol deviations, 
    and so on. A statistical overview or meta-analysis may be 
    informative when medical questions are addressed in more than one 
    trial. Where possible, this should be envisaged in the plan so that 
    the relevant trials are clearly identified and any necessary common 
    features of their designs are specified in advance. Other major 
    statistical issues (if any) that are expected to affect a number of 
    trials in a common plan should be addressed in that plan.
    
    2.1.2 Confirmatory Trial
    
        A confirmatory trial is a controlled trial in which a hypothesis 
    is stated in advance and evaluated. As a rule, confirmatory trials 
    are necessary to provide firm evidence of efficacy or safety. In 
    such trials, the key hypothesis of interest follows directly from 
    the trial's primary objective, is always predefined, and is the 
    hypothesis that is subsequently tested when the trial is complete. 
    In a confirmatory trial, it is equally important to estimate with 
    due precision the size of the effects attributable to the treatment 
    of interest and to relate these effects to their clinical 
    significance.
        Confirmatory trials are intended to provide firm evidence in 
    support of claims. Therefore, adherence to their planned design and 
    procedures is particularly important; unavoidable changes should be 
    explained and documented, and their effect examined. A justification 
    of the design of each such trial and of all other statistical 
    aspects, such as the planned analysis, should be set out in the 
    protocol. Each trial should address only a limited number of 
    questions.
        Firm evidence in support of claims requires that the results of 
    the confirmatory trials demonstrate that the investigational product 
    under test has clinical benefits. The confirmatory trials should 
    therefore be sufficient to answer each key clinical question 
    relevant to the efficacy or safety claim clearly and definitively. 
    In addition, it is important that the basis for generalization to 
    the intended patient population is understood and explained; this 
    may also influence the number and type of centers and/or trials 
    needed. The results of the confirmatory trial(s) should be robust. 
    In some circumstances, the weight of evidence from a single 
    confirmatory trial may be sufficient.
    
    2.1.3 Exploratory Trial
    
        The rationale and design of confirmatory trials nearly always 
    rests on earlier clinical work carried out in a series of 
    exploratory studies. Like all clinical trials, these exploratory 
    studies should have clear and precise objectives. However, in 
    contrast to confirmatory trials, their objectives may not always 
    lead to simple tests of predefined hypotheses. In addition, 
    exploratory trials may sometimes require a more flexible approach to 
    design so that changes can be made in response to accumulating 
    results. Their analysis may entail data exploration; tests of 
    hypothesis may be carried out, but the choice of hypothesis may be 
    data dependent. Such trials cannot be the basis of the formal proof 
    of efficacy, although they may contribute to the total body of 
    relevant evidence.
        Any individual trial may have both confirmatory and exploratory 
    aspects. For example, in most confirmatory trials the data are also 
    subjected to exploratory analyses which serve as a basis for 
    explaining or supporting their findings and for suggesting further 
    hypotheses for later research. The protocol should make a clear 
    distinction between the aspects of a trial that will be used for 
    confirmatory proof and the aspects that will provide data for 
    exploratory analysis.
    
    2.2 Study Scope
    
    2.2.1 Population
    
        In the earlier phases of drug development, the choice of 
    subjects for a clinical trial may be heavily influenced by the wish 
    to maximize the chance of observing specific clinical effects of 
    interest. Hence, they may come from a very narrow subgroup of the 
    total patient population for which the drug may eventually be 
    indicated. However, by the time the confirmatory trials are 
    undertaken, the subjects in the trials should more closely mirror 
    the intended users. In these trials, it is generally helpful to 
    relax the inclusion and exclusion criteria as much as possible 
    within the target indication, while maintaining sufficient 
    homogeneity to permit a successful trial to be carried out. No 
    individual clinical trial can be expected to be totally 
    representative of future users because of the possible influences of 
    geographical location, the time when it is conducted, the medical 
    practices of the particular investigator(s) and clinics, and so on. 
    However, the influence of such factors should be reduced wherever 
    possible and subsequently discussed during the interpretation of the 
    trial results.
    
    2.2.2 Primary and Secondary Variables
    
        The primary variable (``target'' variable, primary endpoint) 
    should be the variable capable of providing the most clinically 
    relevant and convincing evidence directly related to the primary 
    objective of the trial. There should generally be only one primary 
    variable. This will usually be an efficacy variable, because the 
    primary objective of most confirmatory trials is to provide strong 
    scientific evidence regarding efficacy. Safety/tolerability may 
    sometimes be the primary variable, and will always be an important 
    consideration. Measurements relating to quality of life and health 
    economics are further potential primary variables. The selection of 
    the primary variable should reflect the accepted norms and standards 
    in the relevant field of research. The use of a reliable and 
    validated variable with which experience has been gained either in 
    earlier studies or in published literature is recommended. There 
    should be sufficient evidence that the primary variable can provide 
    a valid and reliable measure of some clinically relevant and 
    important treatment benefit in the subject population described by 
    the inclusion and exclusion criteria. The primary variable should 
    generally be the one used when estimating the sample size (see 
    section 3.5).
        In many cases, and especially when treatment is directed at a 
    chronic rather than an acute process, the approach to assessing 
    subject outcome may not be straightforward and should be carefully 
    defined. For example, it is inadequate to specify mortality as a 
    primary variable without further clarification; mortality may be 
    assessed by comparing proportions alive at fixed points in time, or 
    by comparing overall distributions of survival times over a 
    specified interval. Another common example is a recurring outcome. 
    The measure of treatment effect may again be a simple dichotomous 
    variable (any occurrence during a specified interval), time to first 
    occurrence, or rate of occurrence (events per time units of 
    observation), to give a few possibilities. The assessment of 
    functional status over time in studying treatment for chronic 
    disease presents other challenges in selection of the primary 
    variable. There are many possible
    
    [[Page 25715]]
    
    approaches, such as comparisons of the assessments done at the 
    beginning and end of the interval of observation, comparison of 
    slopes calculated from all assessments throughout the interval, or 
    comparisons of the proportions of subjects exceeding or declining 
    beyond a prespecified threshold. To avoid multiplicity concerns, it 
    is critical to specify in the protocol the precise definition of the 
    primary variable as it will be used in the statistical analysis. In 
    addition, the clinical relevance of the specific primary variable 
    selected and the validity of the associated measurement procedures 
    will generally need to be addressed and justified in the protocol.
        The primary variable should be specified in the protocol, along 
    with the rationale for its selection. Redefinition of the primary 
    variable after unblinding will almost always be unacceptable, since 
    the biases this introduces are difficult to assess. When relevant, 
    the validity and reliability of the primary variable should be 
    described. Secondary variables are either supportive measurements 
    related to the primary objective or measurements of effects related 
    to the secondary objectives. Their predefinition in the protocol is 
    also important, as well as an explanation of their relative 
    importance and roles in interpretation of trial results. When the 
    clinical effect defined by the primary objective is to be measured 
    in more than one way, the protocol should identify one of the 
    measurements as the primary variable on the basis of clinical 
    relevance, importance, objectivity, and/or other relevant 
    characteristics, whenever such selection is feasible. Another 
    strategy that may be useful in some situations is to integrate or 
    combine the multiple measurements into a single or ``composite'' 
    variable, using a predefined algorithm. Indeed, the primary variable 
    sometimes arises as a combination of multiple clinical measurements 
    (e.g., the rating scales used in arthritis, psychiatric disorders, 
    and elsewhere). This approach addresses the multiplicity problem 
    without requiring adjustment for multiple comparisons. The method of 
    combining the multiple measurements should be specified in the 
    protocol, and an interpretation of the resulting scale should be 
    provided in terms of the size of a clinically relevant benefit. When 
    composite variables are used as primary variables, the individual 
    components of these variables are often analyzed separately. When a 
    rating scale is used as a primary variable, it is especially 
    important to address factors such as content validity, inter- and 
    intrarater reliability, and sensitivity for discriminating different 
    medical conditions.
        In some cases, ``global assessment'' variables are developed to 
    measure the overall safety, overall efficacy, and/or overall 
    usefulness of a treatment. This type of variable integrates 
    objective variables and the investigator's overall impression about 
    the state or change in the state of the subject, and is usually a 
    scale of ordered categorical ratings. Global assessments of overall 
    effectiveness are well established in many therapeutic areas, 
    especially psychotropic drugs and nonsteroidal anti-inflammatory 
    drugs.
        Global assessment variables generally have a subjective 
    component. When a global assessment scale is used as a primary or 
    secondary variable, fuller details should be included in the 
    protocol with respect to:
        (1) The relevance of the global scale to the primary objective 
    of the trial;
        (2) The basis for the validity of the scale;
        (3) How to utilize the data collected on an individual subject 
    to assign him/her to a unique category of the global assessment 
    scale;
        (4) How to uniquely categorize subjects with missing data.If 
    objective variables are considered by the investigator when making a 
    global assessment, then those objective variables should be 
    considered additional primary or, at least, important secondary 
    variables.
        Overall usefulness integrates components of both benefit and 
    risk and reflects the decisionmaking process of the treating 
    physician, who must weigh benefit and risk in making product use 
    decisions. A problem with global usefulness scales is that their use 
    could in some cases lead to the result of two products being 
    declared equivalent despite having very different profiles of 
    beneficial and adverse effects. For example, judging the global 
    usefulness of a treatment as equivalent or superior to an 
    alternative may mask the fact that it has little or no efficacy but 
    fewer adverse effects. Therefore, if usefulness is used as a primary 
    variable, it is important to consider specific efficacy and safety 
    outcomes separately as additional primary variables.
        It may sometimes be desirable to use more than one primary 
    variable, each of which (or a subset of which) could be a sufficient 
    basis for marketing approval, to cover the range of effects of the 
    therapies. The planned manner of interpretation of this type of 
    evidence should be carefully spelled out. For example, it should be 
    clear whether an impact on any of the variables, some minimum number 
    of them, or all of them, would be considered necessary for approval. 
    The primary hypothesis or hypotheses should be clearly stated with 
    respect to the primary variables identified and the approach to 
    testing the hypotheses described. This should include specification 
    of the statistical parameters being tested (e.g., mean, percentage, 
    distribution). The effect on the Type I error should be explained 
    because of the potential for multiple comparison problems (see 
    section 5.6); the method of controlling Type I error should be given 
    in the protocol. The extent of intercorrelation among the proposed 
    primary variables may be considered in evaluating the impact on Type 
    I error. If the success of the trial depends upon demonstrating 
    effects on all of the designated primary variables, then there is no 
    need for adjustment of the Type I error, but the impact on Type II 
    error and sample size needs should be carefully considered.
        When direct assessment of the clinical benefit to the subject 
    through observing actual clinical efficacy is not practical, 
    indirect criteria (surrogate variables) may be considered. Commonly 
    accepted surrogate variables are used in a number of indications 
    where they are believed to be reliable predictors of clinical 
    benefit. There are two principal concerns with the introduction of 
    any proposed surrogate variable. First, it may not be a true 
    predictor of the clinical outcome of interest. For example, it may 
    measure treatment activity along one particular pathway, but may not 
    provide full information on the range of actions and ultimate 
    effects of the treatment, whether positive or negative. There have 
    been many instances where treatments showing a highly positive 
    effect on a proposed surrogate have ultimately been shown to be 
    detrimental to the subjects' clinical status; conversely, there are 
    cases of treatments conferring clinical benefit without measurable 
    impact on proposed surrogates. Additionally, proposed surrogate 
    variables may not yield a quantitative measure of clinical benefit 
    that can be weighed directly against adverse effects. Statistical 
    criteria for validating surrogate variables have been proposed, but 
    the experience with their use is relatively limited. In practice, 
    the strength of the evidence for surrogacy depends upon the 
    biological plausibility of the relationship, the demonstration in 
    epidemiological studies of the prognostic value of the surrogate for 
    the clinical outcome, and evidence from clinical trials that 
    treatment effects on the surrogate correspond to effects on the 
    clinical outcome. Relationships between clinical and surrogate 
    variables for one product do not necessarily apply to a product with 
    a different mode of action for treating the same disease.
        Dichotomization or other categorization of continuous or ordinal 
    variables may sometimes be desirable. Criteria of ``success'' and 
    ``response'' are common examples of dichotomies that should be 
    specified precisely in terms of, for example, a minimum percentage 
    improvement (relative to baseline) in a continuous variable or a 
    ranking categorized as at or above some threshold level (e.g., 
    ``good'') on an ordinal rating scale. The reduction of diastolic 
    blood pressure below 90 mmHg is a common dichotomization. 
    Categorizations are most useful when they have clear clinical 
    relevance. The criteria for categorization should be predefined and 
    specified in the protocol, as knowledge of trial results could 
    easily bias the choice of such criteria. Because categorization 
    normally implies a loss of information, a consequence will be a loss 
    of power in the analysis; this should be accounted for in the sample 
    size calculation.
    
    2.3 Design Techniques to Avoid Bias
    
        The two most important design techniques for avoiding bias in 
    clinical trials are blinding and randomization, and these should be 
    a normal feature of most controlled clinical trials intended to be 
    included in a marketing application. Most such trials follow a 
    double-blind approach in which treatments are prepacked in 
    accordance with a suitable randomization schedule and supplied to 
    the trial center(s) labeled only with the subject number and the 
    treatment period, so that no one involved in the conduct of the 
    trial is aware of the specific treatment allocated to any particular 
    subject, not even as a code letter. This approach will be assumed in 
    section 2.3.1 and most of section 2.3.2, exceptions being considered 
    at the end. The protocol should also specify
    
    [[Page 25716]]
    
    procedures aimed at minimizing any anticipated irregularities in 
    study conduct that might impair a satisfactory analysis, including 
    various types of protocol violations, withdrawals, and missing 
    values. The protocol should consider ways both to reduce frequency 
    of such problems and to handle the problems that do occur in the 
    analysis of data.
    
    2.3.1 Blinding
    
        Blinding is intended to limit the occurrence of conscious and 
    unconscious bias in the conduct and interpretation of a clinical 
    trial arising from the influence that knowledge of treatment may 
    have on the recruitment and allocation of subjects, their subsequent 
    care, the attitudes of subjects to the treatments, the assessment of 
    end points, the handling of withdrawals, the exclusion of data from 
    analysis, and so on. The essential aim is to prevent identification 
    of the treatments until all such opportunities for bias have passed.
        A double-blind trial is one in which neither the subject nor any 
    of the investigator or sponsor staff involved in the treatment or 
    clinical evaluation of the subjects is aware of the treatment 
    received. This includes anyone determining subject eligibility, 
    evaluating endpoints, or assessing compliance with the protocol. 
    This level of blinding is maintained throughout the conduct of the 
    trial; only when the data are cleaned to an acceptable level of 
    quality will appropriate personnel be unblinded. If any of the 
    sponsor staff who are not involved in the treatment or clinical 
    evaluation of the subjects are required to be unblinded to the 
    treatment code (e.g., bioanalytical scientists, auditors, those 
    involved in serious adverse event reporting), the sponsor should 
    have adequate standard operating procedures (SOP's) to guard against 
    inappropriate dissemination of treatment codes. In a single-blind 
    trial the investigator and/or his staff are aware of the treatment 
    but not the subject. In an open-label trial the identity of 
    treatment is known to all. The double-blind trial is the optimal 
    approach. This requires that the treatments to be applied during the 
    trial cannot be distinguished in any way (appearance, taste, etc.) 
    either before or during administration, and that the blind is 
    maintained appropriately during the whole trial.
        Difficulties in achieving the double-blind ideal can arise 
    because: (1) The treatments may be of a completely different nature, 
    for example, surgery and drug therapy; (2) two drugs may have 
    different formulations and, although they could be made 
    indistinguishable by the use of capsules, changing the formulation 
    might also change the pharmacokinetic and/or pharmacodynamic 
    properties, so that bioequivalence of the formulations may need to 
    be established; (3) the daily pattern of administration of two 
    treatments may differ. One way of achieving double-blind conditions 
    under these circumstances is to use a ``double dummy'' technique. 
    This technique may sometimes force an administration scheme that is 
    sufficiently unusual to influence adversely the motivation and 
    compliance of the subjects. Ethical difficulties may also interfere 
    with its use when, for example, it entails dummy operative 
    procedures. Nevertheless, extensive efforts should be made to 
    overcome these difficulties.
        In some clinical trials, although double blinding is planned, it 
    may be partially compromised by apparent treatment induced effects. 
    In such cases, blinding may be improved by blinding investigators to 
    certain test results (e.g., selected clinical laboratory measures). 
    Similar approaches (see below) to minimizing bias in open-label 
    trials should be considered in trials where unique or specific 
    treatment effects may lead to unblinding individual patients.
        If a double-blind trial is not feasible, then the single-blind 
    option should be considered. In some cases only an open-label trial 
    is practically or ethically possible. Single-blind and open-label 
    trials provide additional flexibility, but it is particularly 
    important that the investigator's knowledge of the next treatment 
    should not influence the decision to enter the subject; this 
    decision should precede knowledge of the randomized treatment. Also, 
    under either of these circumstances, clinical assessments should be 
    made by medical staff who are not involved in treating the subjects 
    and who remain blind to treatment. In single-blind or open-label 
    trials, every effort should be made to minimize the various known 
    sources of bias and primary variables should be as objective as 
    possible. The reasons for the degree of blinding adopted, as well as 
    steps taken to minimize bias by other means, should be explained in 
    the protocol.
        Breaking the blind (for a single subject) should be considered 
    only when knowledge of the treatment assignment is deemed essential 
    by the subject's physician for the subject's care. Any intentional 
    or unintentional breaking of the blind should be reported and 
    explained at the end of the trial, irrespective of the reason for 
    its occurrence. The procedure and timing for revealing the treatment 
    assignments should be documented.
        In this document, the blind review of data refers to the 
    checking of data during the period of time between trial completion 
    (the last observation on the last subject) and the breaking of the 
    blind. If specific sponsor staff need to be unblinded during this 
    period to ensure the integrity of the database or the suitability of 
    statistical assumptions, appropriate SOP's should be developed to 
    describe how the treatment code will be protected from broader 
    dissemination.
    
    2.3.2 Randomization
    
        Randomization introduces a deliberate element of chance into the 
    assignment of treatments to subjects in a clinical trial. During 
    subsequent analysis of the trial data, it provides a sound 
    statistical basis for the quantitative evaluation of the evidence 
    relating to treatment effects. It also tends to produce treatment 
    groups in which the distributions of prognostic factors (known and 
    unknown) are similar. In combination with blinding, randomization 
    helps to avoid possible bias in the selection and allocation of 
    subjects arising from the predictability of treatment assignments.
        The randomization schedule of a clinical trial documents the 
    random allocation of treatments to subjects. In the simplest 
    situation, it is a sequential list of treatments (or treatment 
    sequences in a crossover trial) or corresponding codes by subject 
    number. The logistics of some trials, such as those with a screening 
    phase, may make matters more complicated, but the unique preplanned 
    assignment of treatment, or treatment sequence, to subject should be 
    clear. Different trial designs should have different procedures for 
    generating randomization schedules. The randomization schedule 
    should be capable of being reproduced (if the need arises). Whenever 
    possible, this should be accomplished through the use of the same 
    random number table, or the same computer routine and seed for its 
    random number generator.
        Although unrestricted randomization is an acceptable approach, 
    some advantages can generally be gained by randomizing subjects in 
    blocks. This helps to increase the comparability of the treatment 
    groups particularly when subject characteristics may change over 
    time, as a result, for example, of changes in recruitment policy. It 
    also provides a better guarantee that the treatment groups will be 
    of nearly equal size. In cross-over trials, it provides the means of 
    obtaining balanced designs with their greater efficiency and easier 
    interpretation. Care should be taken to choose block lengths that 
    are sufficiently short to limit possible imbalance, but long enough 
    to avoid predictability towards the end of the sequence in a block. 
    Investigators should generally be blind to the block length; the use 
    of two or more block lengths, randomly selected for each block, can 
    achieve the same purpose. (Theoretically, in a double-blind trial 
    predictability does not matter, but the pharmacological effects of 
    drugs often provide the opportunity for intelligent guesswork.)
        In multicenter trials, the randomization procedures should be 
    organized centrally. It is advisable to have a separate random 
    scheme for each center, i.e., to stratify by center or to allocate 
    several whole blocks to each center. More generally, stratification 
    by important prognostic factors measured at baseline (e.g., severity 
    of disease, age, sex, etc.) may sometimes be valuable in order to 
    promote balanced allocation within strata; this has greater 
    potential benefit in small trials. The use of more than two or three 
    stratification factors is rarely necessary, is less successful at 
    achieving balance, and is logistically troublesome. Where it is 
    necessary, the use of a dynamic allocation procedure (see below) may 
    help to achieve balance across all factors simultaneously, provided 
    the rest of the trial procedures can be adjusted to accommodate an 
    approach of this type.
        The next subject to be randomized into a study should always 
    receive the treatment corresponding to the next free number in the 
    appropriate randomization schedule (in the respective stratum, if 
    randomization is stratified). The appropriate number and associated 
    treatment for the next subject should only be allocated when entry 
    of that subject to the randomized part of the trial has been 
    confirmed. These tasks will normally be carried out by staff at the 
    investigator's center, who will then dispense the relevant blinded 
    trial supplies. Details of the
    
    [[Page 25717]]
    
    randomization which facilitate predictability (e.g., block length) 
    should not be contained in the study protocol. The randomization 
    schedule itself should be filed securely by the sponsor or an 
    independent party in a manner that ensures that blindness is 
    properly maintained throughout the trial. Access to the 
    randomization schedule during the trial should take into account the 
    possibility that, in an emergency, the blind may have to be broken 
    for any subject, either partially or completely. The procedure to be 
    followed, the necessary documentation, and the subsequent treatment 
    and assessment of the subject should all be described in the 
    protocol.
        Dynamic allocation is an alternative randomization procedure in 
    which the allocation of treatment to a subject is influenced by the 
    current balance of allocated treatments and, in a stratified trial, 
    by the stratum to which the subject belongs and the balance within 
    that stratum. Every effort should be made to retain the double-blind 
    status of the trial. For example, knowledge of the treatment code 
    may be restricted to a central trial office from where the dynamic 
    allocation is controlled, generally through telephone contact. This 
    in turn permits additional checks of eligibility criteria and 
    establishes entry into the trial, features that can be valuable in 
    certain types of multicenter trials. The usual system of prepacking 
    and labeling drug supplies for double-blind trials can then be 
    followed, but the order of their use is no longer sequential. It is 
    desirable to use appropriate computer algorithms to keep personnel 
    at the central trial office blind to the treatment code. The 
    complexity of the logistics and potential impact on the analysis 
    should be carefully evaluated when considering dynamic allocation.
    
    III. Study Design Considerations
    
    3.1 Study Configuration
    
    3.1.1 Parallel Group Design
    
        The most common clinical trial design for confirmatory trials is 
    the parallel group design in which subjects are randomized to one of 
    two or more arms, each arm being allocated a different treatment. 
    These treatments will include the investigational product at one or 
    more doses, and one or more control treatments, such as placebo and/
    or an active comparator. The assumptions underlying this design are 
    less complex than for most other designs. However, there may be 
    additional features of the design which complicate the analysis and 
    interpretation (e.g., covariates, repeated measurements over time, 
    interactions between design factors, protocol violations, dropouts, 
    and withdrawals).
    
    3.1.2 Cross-Over Design
    
        In the cross-over design, each subject is randomized to a 
    sequence of two or more treatments and hence acts as his own control 
    for treatment comparisons. This simple maneuver is attractive 
    primarily because it reduces the number of subjects and, usually, 
    the number of assessments needed to achieve a specific power, 
    sometimes to a marked extent. In the simplest 2x2 cross-over design, 
    each subject receives each of two treatments in randomized order in 
    two successive treatment periods, often separated by a washout 
    period. The most common extension of this entails comparing n(>2) 
    treatments in n periods, each subject receiving all n treatments. 
    Numerous variations exist, such as designs in which each subject 
    receives a subset of n(>2) treatments, or designs in which 
    treatments are repeated within a subject.
        Cross-over designs have a number of problems which can 
    invalidate their results. The chief difficulty concerns carryover, 
    that is, the residual influence of treatments in subsequent 
    treatment periods. In an additive model, the effect of unequal 
    carryover will be to bias direct treatment comparisons. In the 2x2 
    design, the relevant contrast cannot be statistically distinguished 
    from the interaction between treatment and period, and the test for 
    either of these lacks power because it is a ``between subject'' 
    contrast. This problem is less acute in higher order designs, but 
    cannot be entirely dismissed.
        Therefore, when the cross-over design is used, it is important 
    to avoid carryover. This is best done by selective and careful use 
    of the design on the basis of adequate knowledge of both the disease 
    area and the new medication. The disease under study should be 
    chronic and stable. The relevant effects of the medication should 
    develop fully within the treatment period. The washout periods 
    should be sufficiently long for complete reversibility of drug 
    effect. The fact that these conditions are likely to be met should 
    be established in advance of the trial by means of prior information 
    and data.
        A common, and generally satisfactory, use of the 2x2 cross-over 
    design is to demonstrate the bioequivalence of two formulations of 
    the same medication. In this particular application in healthy 
    volunteers, carryover effects on the relevant pharmacokinetic 
    variable are rather unlikely to occur if the wash-out time between 
    the two periods is sufficiently long. However, it is still important 
    to check this assumption during analysis on the basis of the data 
    obtained, for example, by demonstrating that no drug is detectable 
    at the start of each period.
        There are additional problems that need careful attention in 
    cross-over trials. The most notable of these are the complications 
    of analysis and interpretation arising from the loss of subjects. 
    Also, the potential for carryover leads to difficulties in assigning 
    adverse events that occur in later treatment periods to the 
    appropriate treatment. These and other issues are described in the 
    ICH E4 topic on ``Dose-Response Information to Support Drug 
    Registration.'' The cross-over design should generally be restricted 
    to situations where losses of subjects from the trial are expected 
    to be small.
    
    3.1.3 Factorial Designs
    
        In a factorial design, two or more treatments are evaluated 
    simultaneously in the same set of subjects through the use of 
    varying combinations of the treatments. The simplest example is the 
    2x2 factorial design in which subjects are randomly allocated to one 
    of the four possible combinations of two treatments, A and B. These 
    are: A alone; B alone; both A and B; neither A nor B. In many cases 
    this design is used for the specific purpose of examining the 
    interaction of A and B. The statistical test of interaction is model 
    dependent and may lack power to detect an interaction if the sample 
    size was calculated based on the test for main effects. This 
    consideration is important when this design is used for examining 
    the joint effects of A and B, in particular, if the treatments are 
    likely to be used together.
        Another important use of the factorial design is to establish 
    the dose-response characteristics of a combination product, e.g., 
    one combining treatments C and D, especially when the efficacy of 
    each monotherapy has been established at some dose in prior studies. 
    A number, m, of doses of C is selected, usually including a zero 
    dose (placebo), and a similar number, n, of doses of D. The full 
    design then consists of mn treatment groups, each receiving a 
    different combination of doses of C and D. The resulting estimate of 
    the response surface may then be used to help identify an 
    appropriate combination of doses of C and D for clinical use.
        In some cases, the 2x2 design may be used to make efficient use 
    of clinical trial subjects by evaluating the efficacy of the two 
    treatments with the same number of subjects as would be required to 
    evaluate the efficacy of either one alone. This strategy has proved 
    to be particularly valuable for very large mortality studies. The 
    efficiency of this approach depends upon the absence of interaction 
    between treatments A and B so that the effects of A and B on the 
    primary efficacy variables follow an additive model, hence the 
    effect of A is virtually identical whether or not it is additional 
    to the effect of B. As for the cross-over trial, evidence that this 
    condition is likely to be met should be established in advance of 
    the trial by means of prior information and data.
    
    3.2 Multicenter Trials
    
        Multicenter trials are carried out for two main reasons. First, 
    a multicenter trial is an accepted way of evaluating a new 
    medication more efficiently; under some circumstances, it may 
    present the only practical means of accruing sufficient subjects to 
    satisfy the trial objective within a reasonable timeframe. 
    Multicenter trials of this nature may, in principle, be carried out 
    at any stage of clinical development. They may have several centers 
    with a large number of subjects per center or, in the case of a rare 
    disease, they may have a large number of centers with very few 
    subjects per center.
        Second, a trial may be designed as a multicenter (and multi-
    investigator) trial primarily to provide a better basis for the 
    subsequent generalization of its findings. This arises from the 
    possibility of recruiting the subjects from a wider population and 
    of administering the medication in a broader range of clinical 
    settings, thus presenting an experimental situation that is more 
    typical of future use. In this case, the involvement of a number of 
    investigators also gives the potential for a wider range of clinical 
    judgement concerning the value of the medication. Such a trial would 
    be a confirmatory trial in the later phases of drug development and 
    would be likely to involve a large number of investigators and 
    centers.
    
    [[Page 25718]]
    
     It might sometimes be conducted in a number of different countries 
    to facilitate generalizability even further.
        If a multicenter trial is to be meaningfully interpreted and 
    extrapolated, then the manner in which the protocol is implemented 
    should be clear and similar at all centers. Furthermore, the usual 
    sample size and power calculations depend upon the assumption that 
    the differences between the compared treatments in the centers are 
    unbiased estimates of the same quantity. It is important to design 
    the common protocol and to conduct the trial with this background in 
    mind. Procedures should be standardized as completely as possible. 
    Variation of evaluation criteria and schemes can be reduced by 
    investigator meetings, by the training of personnel in advance of 
    the study, and by careful monitoring during the study. Good design 
    should generally aim to achieve the same distribution of subjects to 
    treatments within each center and good management should maintain 
    this design objective. Trials which avoid excessive variation in the 
    numbers of subjects per center and trials which avoid a few very 
    small centers have advantages if it is later found necessary to 
    examine the heterogeneity of the treatment effect from center to 
    center, because they reduce the differences between different 
    weighted estimates of the treatment effect. (This point does not 
    apply to trials in which all centers are very small and in which 
    center does not feature in the analysis.) Failure to take these 
    precautions, combined with doubts about the homogeneity of the 
    results, may, in severe cases, reduce the value of a multicenter 
    trial to such a degree that it cannot be regarded as giving 
    convincing evidence for the sponsor's claims.
        In the simplest multicenter trial, each investigator will be 
    responsible for the subjects recruited at one hospital, so that 
    ``center'' is identified uniquely by either investigator or 
    hospital. In many trials, however, the situation is more complex. 
    One investigator may recruit subjects from several hospitals; one 
    investigator may represent a team of clinicians (subinvestigators) 
    who all recruit subjects from their own clinics at one hospital or 
    at several associated hospitals. Whenever there is room for doubt 
    about the definition of center in a statistical model, the 
    statistical section of the protocol (see section 5.1) should clearly 
    define the term (e.g., by investigator, location, or region) in the 
    context of the particular trial. In most instances, centers can be 
    satisfactorily defined through the investigators. (ICH Guideline E6 
    provides relevant guidance in this respect.) In cases of doubt, the 
    aim should be to define centers to achieve homogeneity in the 
    important factors affecting the measurements of the primary 
    variables and the influence of the treatments. Any rules for 
    combining centers in the analysis should be justified and specified 
    prospectively in the protocol where possible, but in any case 
    decisions concerning this approach should always be taken blind to 
    treatment, for example, at the time of the blind review. It is 
    sometimes possible to characterize the centers by historical 
    measures of response to the control treatment or to other standard 
    treatments, and this information may help to support decisions 
    concerning the combination of centers for analysis.
        The statistical model to be adopted for the comparison of 
    treatments should be described in the protocol. The main treatment 
    effect may be investigated first using a model that allows for 
    center differences, but does not include a term for center by 
    treatment interaction. In the absence of a true center by treatment 
    interaction, the routine inclusion of interaction terms in the model 
    reduces the efficiency of the test for the main effects. In the 
    presence of a true center by treatment interaction, the 
    interpretation of the main treatment effect is controversial.
        In some studies, for example, some large mortality studies with 
    very few subjects per center, there may be no reason to expect the 
    centers to have any influence on the primary or secondary variables 
    because they are unlikely to represent influences of clinical 
    importance. In other studies, it may be recognized from the start 
    that the limited numbers of subjects per center will make it 
    impracticable to include the center effects in the statistical 
    model. In these cases, it is not appropriate to include a term for 
    center in the model, because in this situation randomization is 
    rarely stratified by center.
        If positive treatment effects are found in a trial with 
    appreciable numbers of subjects per center, there should generally 
    be a subsequent exploration of treatment by center interaction, as 
    this may affect the generalizability of the conclusions. Marked 
    treatment by center interaction may be identified by graphical 
    display of the results of individual centers or by analytical 
    methods, such as a significance test of the interaction. When using 
    such a statistical significance test, it is important to recognize 
    that this generally has low power in a trial designed to detect the 
    main effect of treatment.
        If a treatment by center interaction is found, this should be 
    interpreted with care and vigorous attempts should be made to find 
    an explanation in terms of other features of trial management or 
    subject characteristics. Such an explanation will usually define the 
    appropriate further analysis and interpretation. In the absence of 
    an explanation, marked quantitative interactions imply that 
    alternative estimates of the treatment effect may be needed, giving 
    different weights to the centers, in order to substantiate the 
    robustness of the estimates of treatment effect. It is even more 
    important to understand the basis of any marked qualitative 
    interactions, and failure to find an explanation may necessitate 
    further clinical trials before the treatment effect can be reliably 
    predicted.
    
    3.3 Type of Comparison
    
    3.3.1 Trials to Show Superiority
    
        Scientifically, efficacy is most convincingly established by 
    demonstrating superiority to placebo in a placebo-controlled trial, 
    by showing superiority to an active control treatment, or by 
    demonstrating a dose-response relationship. This type of trial is 
    referred to as a ``superiority'' trial (see section 5.2.3). In this 
    guideline, superiority trials are generally assumed unless 
    explicitly stated otherwise.
        For serious illnesses, when a therapeutic treatment that has 
    been shown to be efficacious by superiority trial(s) exists, a 
    placebo-controlled trial may be considered unethical. In that case, 
    the scientifically sound use of the active control should be 
    considered. The appropriateness of placebo control versus active 
    control should be considered on a study-by-study basis.
    
    3.3.2 Trials to Show Equivalence or Noninferiority
    
        In some cases, an investigational product is compared to a 
    reference treatment without the objective of showing superiority. 
    This type of trial is divided into two major categories according to 
    its objective; one is an ``equivalence'' trial and the other is a 
    ``noninferiority'' trial.
        Bioequivalence trials fall into the former category. In some 
    situations, clinical equivalence trials are also undertaken for 
    other regulatory reasons, such as demonstrating the clinical 
    equivalence of a generic product to the marketed product when the 
    compound is not absorbed and therefore not present in the blood 
    stream.
        Many active control trials are designed to show that the 
    efficacy of an investigational product is no worse than that of the 
    active comparator, and hence fall into the latter category. Another 
    possibility is a ``relative potency assay,'' which is a study where 
    multiple doses of the investigational drug are compared with the 
    recommended dose or multiple doses of the standard drug.
        Active control equivalence or noninferiority trials may also 
    incorporate a placebo, thus pursuing multiple goals in one trial, 
    for example, establishing superiority to placebo, thereby validating 
    the study design and evaluating the degree of similarity of efficacy 
    and safety to the active comparator. There are well-known 
    limitations associated with the use of the active control 
    equivalence (or noninferiority) trials that do not incorporate a 
    placebo. These relate to the implicit lack of any measure of 
    internal validity (in contrast to superiority trials), thus making 
    external validation necessary. The equivalence (or noninferiority) 
    trial is not conservative in nature, so many flaws in the design or 
    conduct of the trial will tend to bias the results towards a 
    conclusion of equivalence. For these reasons, the design features of 
    such trials should receive special attention.
        Active comparators should be chosen with care. An example of a 
    suitable active comparator would be a widely used therapy whose 
    efficacy in the relevant indication has been clearly established and 
    quantified in well-designed and well-documented superiority trial(s) 
    and that can be reliably expected to exhibit similar efficacy in the 
    contemplated active control study. To this end, the new trial should 
    have the same important design features (primary variables, the dose 
    of the active comparator, eligibility criteria, etc.) as the 
    previously conducted superiority trials in which the active 
    comparator clearly demonstrated clinically relevant efficacy.
        It is vital that the protocol of a trial designed to demonstrate 
    equivalence or
    
    [[Page 25719]]
    
    noninferiority contain a clear statement that this is its explicit 
    intention. An equivalence margin should be specified in the 
    protocol; this margin is the largest difference which can be judged 
    as being clinically acceptable. For the active control equivalence 
    trial, both the upper and the lower equivalence margins are needed, 
    while for the active control non-inferiority trial, only the lower 
    margin is needed. There should be clinical justification for the 
    choice of equivalence margins.
        Statistical analysis is generally based on the use of confidence 
    intervals (see section 5.5). For equivalence trials, the two-sided 
    1-2 (alpha) confidence limits should be used. Equivalence 
    is inferred when the entire confidence interval falls within the 
    equivalence margins. This is equivalent to the method of using two 
    simultaneous one-sided tests to test the (composite) null hypothesis 
    that the treatment difference is outside of the equivalence margins 
    versus the (composite) alternative that the treatment difference is 
    within the limits. With this method, the Type I error is controlled 
    at a level of . For noninferiority trials, the one-sided 1-
     interval should be used. The confidence interval approach 
    has a one-sided hypothesis test counterpart testing the null 
    hypothesis that the treatment difference (investigational product 
    minus control) is equal to the lower equivalence margin versus the 
    alternative that the treatment difference is greater than the lower 
    equivalence margin. Sample size calculations should be based on 
    these methods (see section 3.5). The choice of  should be a 
    consideration separate from the choice of a one-sided or two-sided 
    test.
        It is inappropriate to conclude equivalence or noninferiority 
    based on observing a nonsignificant test result of the null 
    hypothesis that there is no difference between the investigational 
    product and the active comparator.
        There are also special issues in the choice of analysis sets. 
    Subjects who withdraw or drop out of the treatment group or the 
    comparator group will tend to have a lack of response, hence the 
    analysis of all randomized subjects may be biased toward 
    demonstrating equivalence (see section 5.2.3).
    
    3.3.3 Dose-Response Designs
    
        How response is related to the dose of a new investigational 
    product is a question to which answers may be obtained in all phases 
    of development and by a variety of approaches (see ICH E4). Dose-
    response studies may serve a number of objectives, among which the 
    following are of particular importance: The confirmation of 
    efficacy; the investigation of the shape and location of the dose-
    response curve; the estimation of an appropriate starting dose; the 
    identification of optimal strategies for individual dose 
    adjustments; the determination of a maximal dose beyond which 
    additional benefit would be unlikely to occur. These objectives 
    should be addressed using the data collected at a number of doses 
    under investigation, including a placebo (zero dose) wherever 
    appropriate. For this purpose, the application of estimation 
    procedures, including the construction of confidence intervals and 
    of graphical methods is as important as the use of statistical 
    tests. The hypothesis tests that are used may need to be tailored to 
    the natural ordering of doses or to particular questions regarding 
    the shape of the dose-response curve (e.g., monotonicity). The 
    details of the planned statistical procedures should be given in the 
    protocol.
    
    3.4 Group Sequential Designs
    
        Group sequential designs are used to facilitate the conduct of 
    interim analysis (see section 4.5). While group sequential designs 
    are not the only acceptable types of designs permitting interim 
    analysis, they are the most commonly applied because it is more 
    practicable to assess grouped subject outcomes at periodic intervals 
    during the trial than on a continuous basis as data from each 
    subject become available. The statistical methods should be fully 
    specified in advance of the availability of information on treatment 
    outcomes and subject treatment assignments (i.e., blind breaking, 
    see section 4.5). An independent data monitoring committee (IDMC) 
    may be used to conduct the interim analysis of data arising from a 
    group sequential design (see section 4.6). While the design has been 
    most widely and successfully used in large, long-term trials of 
    mortality or major nonfatal endpoints, its use is growing in other 
    circumstances. In particular, it is recognized that safety must be 
    monitored in all trials, therefore, the need for formal procedures 
    to cover early stopping for safety reasons should always be 
    considered.
    
    3.5 Sample Size
    
        The number of subjects in a clinical trial should always be 
    large enough to provide a reliable answer to the questions 
    addressed. This number is usually determined by the primary 
    objective of the trial. If the sample size is determined on some 
    other basis, this should be made clear and justified. For example, a 
    trial sized on the basis of safety questions or requirements may 
    need larger numbers of subjects than one sized on the basis of 
    efficacy questions. (See, for example, ICH E1A ``Population 
    Exposure: The Extent of Population Exposure to Assess Clinical 
    Safety.'')
        When determining the appropriate sample size, the following 
    items should be specified: A primary variable; the test statistic; 
    the null hypothesis; the alternative (``working'') hypothesis at the 
    chosen dose(s) (embodying consideration of the treatment difference 
    to be detected or rejected at the dose and in the subject population 
    selected); the probability of erroneously rejecting the null 
    hypothesis (the Type I error) and the probability of erroneously 
    failing to reject the null hypothesis (the Type II error); as well 
    as the approach to dealing with treatment withdrawals and protocol 
    violations. In some instances, the event rate is of primary interest 
    for evaluating power, and assumptions should be made to extrapolate 
    from the required number of events to the eventual sample size for 
    the trial.
        The method by which the sample size is calculated should be 
    given in the protocol, together with the estimates of any quantities 
    used in the calculations (such as variances, mean values, response 
    rates, event rates, difference to be detected). The basis of these 
    estimates should also be given. It is important to investigate the 
    sensitivity of the sample size estimate to a variety of deviations 
    from these assumptions and this may be facilitated by providing a 
    range of sample sizes appropriate for a reasonable range of 
    deviations from assumptions. In confirmatory studies, assumptions 
    should normally be based on published data or on the results of 
    earlier studies. The treatment difference to be detected may be 
    based on a judgement concerning the minimal effect that has clinical 
    relevance in the management of patients or on a judgement concerning 
    the anticipated effect of the new treatment, where this is larger. 
    Conventionally, the probability of Type I error is set at 5 percent 
    or less or as dictated by any adjustments made necessary for 
    multiplicity considerations; the precise choice is influenced by the 
    prior plausibility of the hypothesis under test and the desired 
    impact of the results. The probability of Type II error is 
    conventionally set at 20 percent or less; it is in the sponsor's 
    interest to keep this figure as low as feasible, especially in the 
    case of studies that are difficult or impossible to repeat.
        Sample size calculations should refer to the number of subjects 
    required for the primary analysis. If this is the ``all randomized 
    subjects'' set, estimates about the effect size may need to be 
    reduced compared to the per protocol set. This is due to the 
    diluting effect of the inclusion of treatment withdrawals. The 
    assumptions of variability may also need to be revised.
        The sample size of an equivalence trial or a noninferiority 
    trial (see section 3.3.2) should normally be based on the objective 
    of obtaining a confidence interval for the treatment difference that 
    shows that the treatments differ at most by a clinically acceptable 
    difference. For equivalence trials, the power is usually assessed at 
    a true difference of zero but can be underestimated if the true 
    difference is not zero. For noninferiority trials, the power is 
    usually assessed at an expected (nonzero) difference, but can be 
    underestimated if the true difference is less than expected. The 
    choice of a ``clinically acceptable'' difference needs 
    justification, and may be smaller than the ``clinically relevant'' 
    difference referred to above in the context of superiority trials 
    designed to establish that a difference exists.
        The sample size in a group sequential trial cannot be fixed in 
    advance because it depends upon the play of chance in combination 
    with the chosen stopping rule and the true treatment difference. The 
    design of the stopping rule should take into account the consequent 
    distribution of the sample size, usually embodied in the expected 
    and maximum sample sizes.
        When event rates are lower than anticipated or variability is 
    larger than expected, methods for sample size reestimation are 
    available without unblinding data or making treatment comparisons 
    (see section 4.4).
    
    3.6 Data Capture and Processing
    
        The collection of data and transfer of data from the 
    investigator to the sponsor can take place through a variety of 
    media, including paper case record forms, remote site
    
    [[Page 25720]]
    
    monitoring systems, medical computer systems, and electronic 
    transfer. Whatever data capture instrument is used, the form and 
    content of the information collected should be in full accordance 
    with the protocol and should be established in advance of the 
    conduct of the clinical trial. It should focus on the data necessary 
    to implement the analysis plan, including the context information 
    (such as timing assessments relative to dosing) necessary to confirm 
    protocol compliance or identify important protocol deviations. 
    ``Missing values'' should be distinguishable from the ``value zero'' 
    or ``characteristic absent.''
        The process of data capture, through to database finalization, 
    should be carried out in accordance with good clinical practice 
    (GCP) (see ICH E6, section 5). Specifically, timely and reliable 
    processes for recording data and rectifying errors and omissions are 
    necessary to ensure delivery of a quality database and the 
    achievement of the trial objectives through the implementation of 
    the analysis plan.
    
    IV. Study Conduct
    
    4.1 Trial Monitoring
    
        Careful conduct of a clinical trial according to the protocol 
    has a major impact on the credibility of the results. Careful 
    monitoring can ensure that difficulties are noticed early and their 
    occurrence or recurrence minimized.
        There are two distinct types of monitoring that generally 
    characterize confirmatory clinical trials sponsored by the 
    pharmaceutical industry. Both types of trial monitoring, in addition 
    to entailing different staff responsibilities, involve access to 
    different types of study data and information, thus different 
    principles apply for the control of potential statistical and 
    operational bias.
        One type of monitoring concerns the oversight of the quality of 
    the trial, including whether the protocol is being followed, 
    acceptability of data being accrued, success of planned accrual 
    targets, checking the design assumptions, etc. (see sections 4.2 to 
    4.4). This type of monitoring does not require access to information 
    on comparative treatment effects, nor unblinding of data, and 
    therefore has no impact on Type I error. The monitoring of a trial 
    for this purpose is the responsibility of the sponsor and can be 
    carried out by the sponsor or an independent group selected by the 
    sponsor. The period for this type of monitoring usually starts with 
    the selection of the study sites and ends with the collection and 
    cleaning of the last subject's data.
        The other type of trial monitoring involves breaking the blind 
    to make treatment comparisons. It therefore involves the accruing of 
    comparative treatment results, which requires that a protocol (or 
    appropriate amendments prior to a first analysis) contain 
    statistical plans to prevent certain types of bias. This type of 
    trial monitoring involves unblinded (i.e., key breaking) access to 
    treatment group assignment (actual treatment assignment or 
    identification of group assignment) and comparative treatment group 
    summary information. This type of monitoring is discussed in 
    sections 4.5 and 4.6.
    
    4.2 Changes in Inclusion and Exclusion Criteria
    
        Inclusion and exclusion criteria should remain constant, as 
    specified in the protocol, throughout the period of subject 
    recruitment. Occasionally, however, changes may be appropriate; in 
    long-term studies, for example, growing medical knowledge either 
    from outside the trial or from interim analyses may suggest a change 
    of entry criteria. Changes may also result from the discovery by 
    monitoring staff that regular violations of the entry criteria are 
    occurring, or that seriously low recruitment rates are due to over-
    restrictive criteria. Changes should be made without breaking the 
    blind and should always be described by a protocol amendment that 
    should cover any statistical consequences, such as sample size 
    adjustments arising from different event rates, or modifications to 
    the analysis plan, such as stratifying the analysis according to 
    modified inclusion/exclusion criteria.
    
    4.3 Accrual Rates
    
        In studies with a long time-scale for the accrual of subjects, 
    the rate of accrual should be monitored; if it falls appreciably 
    below the projected level, the reasons should be identified and 
    remedial actions taken to protect the power of the trial and allay 
    concerns about selective entry and other aspects of quality. In a 
    multicenter trial, these considerations apply to the individual 
    centers.
    
    4.4 Sample Size Adjustment
    
        In long-term trials, there will usually be an opportunity to 
    check the assumptions which underlie the original design and sample 
    size calculations. This may be particularly important if the trial 
    specifications have been made on preliminary and/or uncertain 
    information. An interim check conducted on the blinded data may 
    reveal that overall response variances, event rates, or survival 
    experience are not as anticipated. A revised sample size may then be 
    calculated using suitably modified assumptions, and should be 
    justified and documented in a protocol amendment and in the final 
    report. The steps taken to preserve blindness and the consequences, 
    if any, for the Type I error and the width of confidence intervals 
    should be explained. The potential need for reestimation of the 
    sample size should be envisaged in the protocol whenever possible 
    (see section 3.5).
    
    4.5 Interim Analysis and Early Stopping
    
        Any analysis intended to compare treatment arms with respect to 
    efficacy or safety at any time prior to formal completion of a trial 
    is an interim analysis. Because the number, methods, and 
    consequences of these comparisons affect the interpretation of the 
    trial, all interim analyses should be carefully planned in advance 
    and described in the protocol, or otherwise specified in amendments 
    prior to unblinded access to treatment comparison data. When an 
    interim analysis is planned with the intention of deciding whether 
    or not to terminate a trial, this is usually accomplished by the use 
    of a group sequential design that employs statistical monitoring 
    schemes as guidelines (see section 3.4). The goal of such an interim 
    analysis is to stop the trial early if the superiority of the 
    treatment under study is clearly established, if the demonstration 
    of a relevant treatment difference has become unlikely, or if 
    unacceptable adverse effects are apparent. Generally, boundaries for 
    monitoring efficacy require more evidence to terminate a trial early 
    (i.e., more conservative) than do boundaries to terminate a trial 
    for safety reasons. When the trial design and monitoring objective 
    involve multiple endpoints, then this aspect of multiplicity may 
    also need to be taken into account.
        The schedule of interim analyses, or at least the considerations 
    which will govern its generation, should be stated in the protocol 
    or a protocol amendment before the time of the first interim 
    analysis; as flexible statistical methods are available to conduct 
    interim analyses according to a variety of needs (early or late in a 
    trial), the stopping guidelines and their properties should be 
    clearly stated in the protocol or amendments. This material should 
    be written or approved by the data monitoring committee, when the 
    study has one (see section 4.6). Deviations from the planned 
    procedure always bear the potential of invalidating the study 
    results. If it becomes necessary to make changes to the trial, any 
    consequent changes to the statistical procedures should be specified 
    in an amendment to the protocol at the earliest opportunity, 
    especially discussing the impact on any analysis and inferences that 
    such changes may cause. The procedures selected should always ensure 
    that the overall probability of Type I error is controlled.
        The execution of an interim analysis should be a completely 
    confidential process because unblinded data and results are 
    potentially involved. All staff involved in the conduct of the trial 
    should remain blind to the results of such analyses because of the 
    possibility that their attitudes to the trial will be modified and 
    cause changes in recruitment patterns or biases in treatment 
    comparisons. This principle applies to the investigators and their 
    staff and to staff employed by the sponsor that come into contact 
    with clinic staff or subjects. Investigators should be informed only 
    about the decision to continue or to discontinue the trial, or to 
    implement modifications to trial procedures.
        Most clinical trials intended to support the efficacy and safety 
    of an investigational product should proceed to full completion of 
    planned sample size accrual; trials should be stopped early only for 
    ethical reasons or if the power is no longer acceptable. However, it 
    is recognized that drug development plans involve the need for 
    sponsor access to comparative treatment data for a variety of 
    reasons, such as planning other studies or when only a subset of 
    trials will involve the study of serious life-threatening outcomes 
    or mortality which may need sequential monitoring of accruing 
    comparative treatment effects for ethical reasons. In either of 
    these situations, plans for interim statistical analysis should be 
    in place in the protocol or in protocol amendments prior to the 
    unblinded access to comparative treatment data in order to deal with 
    the
    
    [[Page 25721]]
    
    potential statistical and operational bias that may be introduced.
        For many clinical trials of investigational products, especially 
    those that have major public health significance, the responsibility 
    for monitoring comparisons of efficacy and/or safety outcomes should 
    be assigned to an external, independent group, often called an 
    independent data monitoring committee (IDMC), a data and safety 
    monitoring board, or a data monitoring committee, whose 
    responsibilities should be clearly described.
        When a sponsor assumes the role of monitoring efficacy or safety 
    comparisons and therefore has access to unblinded comparative 
    information, particular care should be taken to protect the 
    integrity of the trial and the sharing of information. The sponsor 
    should ensure and document that the internal monitoring committee 
    has complied with written SOP's and that minutes of decisionmaking 
    meetings are maintained.
        Any interim analysis that is not planned in the protocol or 
    specified in an amendment to the protocol prior to unblinding the 
    data (with or without the consequences of stopping the trial early) 
    may flaw the results of a trial and possibly weaken confidence in 
    the conclusions drawn. Therefore, such analyses should be avoided. 
    If unplanned interim analysis is conducted, the study report should 
    explain why it was necessary and the degree to which blindness had 
    to be broken, and provide an assessment of the potential magnitude 
    of bias introduced and the impact on the interpretation of the 
    results.
    
    4.6 Role of Independent Data Monitoring Committee (IDMC)
    
    (see sections 1.25 and 5.5.2 of ICH Guideline E6)
        An IDMC may be established by the sponsor to assess at intervals 
    the progress of a clinical trial, safety data, and critical efficacy 
    variables and recommend to the sponsor whether to continue, modify, 
    or terminate a trial. The IDMC should have written operating 
    procedures and maintain records of its meetings. The independence of 
    the IDMC is intended to control the sharing of important comparative 
    information and to protect the integrity of the clinical trial from 
    adverse impact resulting from access to trial information. The IDMC 
    is a separate entity from an institutional review board (IRB) or an 
    ethics board, and its composition should include clinical trial 
    scientists knowledgeable in the appropriate disciplines, including 
    statistics.
        When there are sponsor representatives on the IDMC, their role 
    should be clearly defined in the operating procedures of the 
    committee (for example, covering whether or not they can vote on key 
    issues). Since these sponsor staff would have access to unblinded 
    information, the procedures should also address the control of 
    dissemination of interim trial results within the sponsor 
    organization.
    
    V. Data Analysis
    
    5.1 Prespecified Analysis Plan
    
        When designing a clinical trial, the principal features of the 
    eventual statistical analysis of the data should be described in the 
    statistical section of the protocol. This section should include all 
    features of the proposed confirmatory analysis of the primary 
    variable(s) and the way in which anticipated analysis problems will 
    be handled. In the case of exploratory trials, this section could 
    describe more general principles and directions.
        Subsequently, a statistical analysis plan may be written as a 
    separate document. In this document, a more technical and detailed 
    elaboration of the principal features stated in the protocol may be 
    included. The statistical analysis plan is usually an internal 
    document and may include detailed procedures for executing the 
    statistical analysis. The statistical analysis plan should be 
    reviewed and possibly updated as a result of the blind review of the 
    data (see section 7.1 for definition).
        If the blind review suggests changes to the principal features 
    stated in the protocol, these should be documented in a protocol 
    amendment. Otherwise, it will suffice to update the statistical 
    analysis plan with the considerations suggested from the blind 
    review. Only results from analyses envisaged in the protocol 
    (including amendments) can be regarded as confirmatory.
        The statistical methodology, including when in the clinical 
    trial process methodology decisions were made, should be clearly 
    described in the statistical section of the clinical study report 
    (see ICH E3).
    
    5.2 Analysis Sets
    
        The set of subjects whose data are to be included in the main 
    analyses should be defined in the statistical section of the 
    protocol. In addition, documentation for all subjects for whom study 
    procedures (e.g., run-in period) were initiated may be useful. The 
    content of this subject documentation depends on detailed features 
    of the particular trial, but at least demographic and baseline data 
    on disease status should be collected whenever possible.
        If all subjects randomized into a clinical trial satisfied all 
    entry criteria, followed all trial procedures perfectly with no 
    losses to followup, and provided complete data records, then the set 
    of subjects to be included in the analysis would be self-evident. 
    The design and conduct of a trial should aim to approach this ideal 
    as closely as possible, but, in practice, it is doubtful if it can 
    ever be fully achieved. Hence, the statistical section of the 
    protocol should address any anticipated problems prospectively in 
    terms of how these affect the subjects and data to be analyzed. The 
    protocol should also specify procedures aimed at minimizing any 
    anticipated irregularities in study conduct that might impair a 
    satisfactory analysis, including various types of protocol 
    violations, withdrawals, and missing values. The protocol should 
    consider ways both to reduce the frequency of such problems and to 
    handle the problems that occur in the analysis of data. The blind 
    review of data to identify possible amendments to the analysis plan 
    due to the protocol violations should be carried out before 
    unblinding. It is desirable to identify any important protocol 
    violation with respect to the time when it occurred, its cause, and 
    its influence on the trial result. The frequency and type of 
    protocol violations, missing values, and other problems should be 
    documented in the study report and their potential influence on the 
    trial results should be described (see ICH E3).
        Decisions concerning the analysis set should be guided by the 
    following principles: (1) To minimize bias and (2) to avoid 
    inflation of Type I error.
    
    5.2.1 All Randomized Subjects
    
        The intention-to-treat principle implies that the primary 
    analysis should include all randomized subjects. In practice, this 
    ideal may be difficult to achieve, for reasons to be described. 
    Hence, analysis sets referred to as ``all randomized subjects'' may 
    not, in fact, include every subject. For example, it is common 
    practice to exclude from the all randomized set any subject who 
    failed to take at least one dose of trial medication or any subject 
    without data post randomization. No analysis is complete unless the 
    potential biases arising from these exclusions are addressed and can 
    be reasonably dismissed.
        In many clinical trials, the ``all randomized subjects'' 
    approach is conservative and also gives estimates of treatment 
    effects that are more likely to mirror those observed in subsequent 
    practice. Randomization prevents biased allocation of subjects to 
    treatments and provides the foundation of statistical tests. The 
    problems associated with the analysis of all randomized subjects lie 
    in the handling of protocol violations and the subtleties that this 
    can involve.
        There are two types of major protocol violations. One is 
    violation of entry criteria. The second is violation of the protocol 
    after randomization. Subjects who fail to satisfy an objective entry 
    criterion measured prior to randomization, but who enter the trial, 
    may be excluded from analysis without introducing bias into the 
    treatment comparison, assuming all subjects receive equal scrutiny 
    for eligibility violations. (This may be difficult to ensure if the 
    data are unblinded.) Not all entry criteria are sufficiently 
    objective for this to be done satisfactorily. Reasons for excluding 
    subjects from the analysis of all randomized subjects should be 
    justified.
        Other problems occur after randomization (error in treatment 
    assignment, use of excluded medications, poor compliance, loss to 
    followup, missing data, and other protocol violations). These 
    problems are especially difficult when their occurrence is related 
    to treatment assignment. It is good practice to assess the pattern 
    of such problems with respect to frequency and time to occurrence 
    among treatment groups. Subjects withdrawn from treatment may 
    introduce serious bias and, if they have provided no data after 
    withdrawal, there is no obvious solution. Severe protocol violation, 
    such as use of excluded medication, may also introduce serious bias 
    into measurements after such a violation. The necessary inclusion of 
    such subjects in the analysis may require some redefinition of the 
    primary variable or some assumptions about the subjects' outcomes.
        Measurements of primary variables made at the time of the loss 
    to followup of a subject for any reason or at the time of a severe
    
    [[Page 25722]]
    
    protocol violation, or subsequently collected in accordance with the 
    protocol, are valuable in the context of all randomized subjects 
    analysis. Their use in analysis should be described and justified in 
    the statistical section of the protocol and their collection 
    described elsewhere in the protocol. However, the use of imputation 
    techniques can lead to biased estimates of treatment effects, 
    particularly when the likelihood of the loss of a subject is related 
    to treatment or response. Any other methods to be employed to ensure 
    the availability of measurements of primary variables for every 
    subject in the all randomized subjects analysis should be described.
        Because of the unpredictability of some problems, it may 
    sometimes be preferable to defer detailed consideration of the 
    manner of dealing with irregularities until the blind review of the 
    data at the end of the study and, if so, this should be stated in 
    the protocol.
    
    5.2.2 Per Protocol Subjects
    
        The ``per protocol'' set of subjects, sometimes described as the 
    ``valid cases,'' the ``efficacy'' sample, or the ``evaluable 
    subjects'' sample, defines a subset of the data used in the all 
    randomized subjects analysis and is characterized by the following 
    criteria:
        (i) The completion of a certain prespecified minimal exposure to 
    the treatment regimen;
        (ii) The availability of measurements of the primary 
    variable(s);
        (iii) The absence of any major protocol violations, including 
    the violation of entry criteria where the nature of and reasons for 
    these protocol violations should be defined and documented before 
    breaking the blind.
        This set may maximize the opportunity for a new treatment to 
    show additional efficacy in the analysis, and most closely reflects 
    the scientific model underlying the protocol. However, it may or may 
    not be conservative, depending on the study, and may be subject to 
    bias (possibly severe) because the subjects adhering most diligently 
    to the study protocol may not be representative of the entire study 
    population.
    
    5.2.3 Roles of the All Randomized Subjects Analysis and the Per 
    Protocol Analysis
    
        In general, it is advantageous to demonstrate a lack of 
    sensitivity of the principal trial results to alternative choices of 
    the set of subjects analyzed. In confirmatory trials, it is usually 
    appropriate to plan to conduct both all randomized subjects and per 
    protocol analyses, so that any differences between them can be the 
    subject of explicit discussion and interpretation. In some cases, it 
    may be desirable to plan further exploration of the sensitivity of 
    conclusions to the choice of the set of subjects analyzed. When the 
    all randomized subjects and the per protocol analyses come to 
    essentially the same conclusions, confidence in the study results is 
    increased, bearing in mind, however, that the need to exclude a 
    substantial proportion of subjects from the per protocol analysis 
    throws some doubt on the overall validity of the study.
        All randomized subjects and per protocol analyses play different 
    roles in superiority trials (which seek to show the investigational 
    product to be superior) and in equivalence or noninferiority trials 
    (which seek to show the investigational product to be comparable, 
    see section 3.3.2). In superiority studies, the all randomized 
    subjects analysis usually tends to avoid the optimistic estimate of 
    efficacy which may result from a per protocol analysis, since the 
    noncompliers included in an all randomized subjects analysis will 
    generally diminish the overall treatment effect. However, in an 
    equivalence or noninferiority trial, the all randomized subjects 
    analysis is no longer conservative and its role should be considered 
    very carefully.
    
    5.3 Missing Values and Outliers
    
        Missing values represent a potential source of bias in a 
    clinical trial. Hence, every effort should be undertaken to fulfill 
    all the requirements of the protocol concerning the collection and 
    management of data. However, in reality there will almost always be 
    some missing data. A study may be regarded as valid, nonetheless, 
    provided the methods of dealing with missing values are sensible, 
    particularly if those methods are predefined in the analysis plan of 
    the protocol. Predefinition of methods may be facilitated by 
    updating this aspect of the analysis plan during the blind review. 
    Unfortunately, no universally applicable methods of handling missing 
    values can be recommended. An investigation should be made 
    concerning the sensitivity of the results of analysis to the method 
    of handling missing values, especially if the number of missing 
    values is substantial.
        A similar approach should be adopted to exploring the influence 
    of outliers, the statistical definition of which is, to some extent, 
    arbitrary. Clear identification of a particular value as an outlier 
    is most convincing when justified medically as well as 
    statistically, and the medical context will then often define the 
    appropriate action. Any outlier procedure set out in the protocol 
    should not favor any treatment group a priori. Once again, this 
    aspect of the analysis plan can be usefully updated during blind 
    review. If no procedure for dealing with outliers was foreseen in 
    the study protocol, one analysis with the actual values and at least 
    one other analysis eliminating or reducing the outlier effect should 
    be performed and differences between their results discussed.
    
    5.4 Data Transformation/Modification
    
        The decision to transform key variables prior to analysis is 
    best made during the design of the trial on the basis of similar 
    data from earlier clinical trials. Transformations (e.g., square 
    root, logarithm) should be specified in the protocol and a rationale 
    provided, especially for the primary variable(s). The general 
    principles guiding the use of transformations to ensure that the 
    assumptions underlying the statistical methods are met are to be 
    found in standard texts; conventions for particular variables have 
    been developed in a number of specific clinical areas. The decision 
    on whether and how to transform a variable should be influenced by 
    the preference for a scale that facilitates clinical interpretation.
        Similar considerations apply to other data modifications 
    sometimes used to create a variable for analysis, such as the use of 
    change from baseline, percentage change from baseline, the ``area 
    under the curve'' of repeated measures, or the ratio of two 
    different variables. Subsequent clinical interpretation should be 
    carefully considered, and the modification should be justified in 
    the protocol. Closely related points are made in section 2.2.2.
    
    5.5 Estimation, Confidence Intervals, and Hypothesis Testing
    
        The statistical section of the protocol should specify the 
    hypotheses that are to be tested and/or the treatment effects that 
    are to be estimated to satisfy the objectives of the trial. The 
    statistical methods to be used to accomplish these tasks should be 
    described for the primary (and preferably the secondary) variables, 
    and the underlying statistical model should be made clear. Estimates 
    of treatment effects should be accompanied by confidence intervals, 
    whenever possible, and the way in which these will be calculated 
    should be identified. The plan should also describe any intentions 
    to use baseline data to improve precision and to adjust estimates 
    for potential baseline differences, for example, by means of 
    analysis of covariance. The reporting of precise p-values (e.g., 
    ``P=0.034'') should be envisaged in the plan, rather than exclusive 
    reference to critical values (e.g., ``P<0.05''). it="" is="" important="" to="" clarify="" whether="" one-="" or="" two-sided="" tests="" of="" statistical="" significance="" will="" be="" used="" and,="" in="" particular,="" to="" justify="" prospectively="" the="" use="" of="" one-sided="" tests.="" if="" formal="" hypothesis="" tests="" are="" not="" considered="" appropriate,="" then="" the="" alternative="" process="" for="" arriving="" at="" statistical="" conclusions="" should="" be="" given.="" the="" particular="" statistical="" model="" chosen="" should="" reflect="" the="" current="" state="" of="" medical="" and="" statistical="" knowledge="" about="" the="" variables="" to="" be="" analyzed.="" all="" effects="" to="" be="" fitted="" in="" the="" analysis="" (for="" example,="" in="" analysis="" of="" variance="" models)="" should="" be="" fully="" specified="" and="" the="" manner,="" if="" any,="" in="" which="" this="" set="" of="" effects="" might="" be="" modified="" in="" response="" to="" preliminary="" results="" should="" be="" explained.="" the="" same="" considerations="" apply="" to="" the="" set="" of="" covariates="" fitted="" in="" an="" analysis="" of="" covariance.="" (see="" also="" section="" 5.7.).="" in="" the="" choice="" of="" statistical="" methods,="" due="" attention="" should="" be="" paid="" to="" the="" statistical="" distribution="" of="" both="" primary="" and="" secondary="" variables.="" when="" making="" this="" choice,="" it="" is="" important="" to="" bear="" in="" mind="" the="" need="" to="" provide="" statistical="" estimates="" of="" the="" size="" of="" treatment="" effects="" together="" with="" confidence="" intervals="" (in="" addition="" to="" significance="" tests),="" as="" this="" may="" influence="" the="" choice="" when="" there="" is="" any="" doubt="" about="" the="" appropriateness="" of="" the="" method.="" the="" primary="" analysis="" of="" the="" primary="" variable="" should="" be="" clearly="" distinguished="" from="" supporting="" analyses="" of="" the="" primary="" or="" secondary="" variables.="" within="" the="" statistical="" section="" of="" the="" protocol="" there="" should="" also="" be="" an="" outline="" of="" the="" way="" in="" which="" data="" other="" than="" the="" primary="" and="" secondary="" variables="" will="" be="" summarized="" and="" reported.="" this="" should="" include="" a="" reference="" to="" any="" approaches="" adopted="" for="" the="" purpose="" of="" achieving="" consistency="" of="" analysis="" across="" a="" range="" of="" studies,="" for="" example,="" for="" safety="" data.="" [[page="" 25723]]="" 5.6="" adjustment="" of="" type="" i="" error="" and="" confidence="" levels="" when="" multiplicity="" is="" present,="" the="" usual="" frequentist="" approach="" to="" the="" analysis="" of="" clinical="" trial="" data="" may="" necessitate="" an="" adjustment="" to="" the="" type="" i="" error.="" multiplicity="" may="" arise,="" for="" example,="" from="" multiple="" primary="" variables="" (see="" section="" 2.2.2),="" multiple="" comparisons="" of="" treatments,="" repeated="" evaluation="" over="" time,="" and/or="" interim="" analyses="" (see="" section="" 4.6).="" methods="" to="" avoid="" or="" reduce="" multiplicity="" are="" sometimes="" preferable="" when="" available,="" such="" as="" the="" identification="" of="" the="" key="" primary="" variable="" (multiple="" variables),="" the="" choice="" of="" a="" critical="" treatment="" contrast="" (multiple="" comparisons),="" the="" use="" of="" a="" summary="" measure="" such="" as="" ``area="" under="" the="" curve''="" (repeated="" measures).="" in="" confirmatory="" analyses,="" any="" aspects="" of="" multiplicity="" that="" remain="" after="" steps="" of="" this="" kind="" have="" been="" taken="" should="" be="" identified="" in="" the="" protocol;="" adjustment="" should="" always="" be="" considered="" and="" the="" details="" of="" any="" adjustment="" procedure="" or="" an="" explanation="" of="" why="" adjustment="" is="" not="" thought="" to="" be="" necessary="" should="" be="" set="" out="" in="" the="" analysis="" plan.="" 5.7="" subgroups,="" interactions,="" and="" covariates="" the="" primary="" variable(s)="" is="" often="" systematically="" related="" to="" other="" influences="" apart="" from="" treatment.="" for="" example,="" there="" may="" be="" relationships="" to="" covariates="" such="" as="" age="" and="" sex,="" or="" there="" may="" be="" differences="" between="" specific="" subgroups="" of="" subjects,="" such="" as="" those="" treated="" at="" the="" different="" centers="" of="" a="" multicenter="" trial.="" in="" some="" instances,="" an="" adjustment="" for="" the="" influence="" of="" covariates="" or="" for="" subgroup="" effects="" is="" an="" integral="" part="" of="" the="" analysis="" plan="" and="" hence="" should="" be="" set="" out="" in="" the="" protocol.="" prestudy="" deliberations="" should="" identify="" those="" covariates="" and="" factors="" expected="" to="" have="" an="" important="" influence="" on="" the="" primary="" variable(s),="" and="" should="" consider="" how="" to="" account="" for="" these="" in="" the="" analysis="" to="" improve="" precision="" and="" to="" compensate="" for="" any="" lack="" of="" balance="" between="" treatment="" groups.="" when="" the="" potential="" value="" of="" an="" adjustment="" is="" in="" doubt,="" it="" is="" often="" advisable="" to="" nominate="" the="" unadjusted="" analysis="" as="" the="" one="" for="" primary="" attention,="" the="" adjusted="" analysis="" being="" supportive.="" special="" attention="" should="" be="" paid="" to="" center="" effects="" and="" to="" the="" role="" of="" baseline="" measurements="" of="" the="" primary="" variable.="" it="" is="" not="" advisable="" to="" adjust="" the="" main="" analyses="" for="" covariates="" measured="" after="" randomization="" because="" they="" may="" be="" affected="" by="" the="" treatments.="" the="" treatment="" effect="" itself="" may="" also="" vary="" with="" subgroup="" or="" covariate--for="" example,="" the="" effect="" may="" decrease="" with="" age="" or="" may="" be="" larger="" in="" a="" particular="" diagnostic="" category="" of="" subjects.="" in="" some="" cases="" such="" interactions="" are="" anticipated,="" hence="" a="" subgroup="" analysis="" or="" a="" statistical="" model="" including="" interactions="" is="" part="" of="" the="" confirmatory="" analysis="" plan.="" in="" most="" cases,="" however,="" subgroup="" or="" interaction="" analyses="" are="" exploratory="" and="" should="" be="" clearly="" identified="" as="" such;="" they="" should="" explore="" the="" uniformity="" of="" any="" treatment="" effects="" found="" overall.="" in="" general,="" such="" analyses="" should="" proceed="" first="" through="" the="" addition="" of="" interaction="" terms="" to="" the="" statistical="" model="" in="" question,="" complemented="" by="" additional="" exploratory="" analysis="" within="" relevant="" subgroups="" of="" subjects,="" or="" within="" strata="" defined="" by="" the="" covariates.="" when="" exploratory,="" these="" analyses="" should="" be="" interpreted="" cautiously;="" any="" conclusion="" of="" treatment="" efficacy="" (or="" lack="" thereof)="" or="" safety="" based="" solely="" on="" exploratory="" subgroup="" analyses="" are="" unlikely="" to="" be="" accepted.="" 5.8="" integrity="" of="" data="" and="" computer="" software="" the="" credibility="" of="" the="" numerical="" results="" of="" the="" analysis="" depends="" on="" the="" quality="" and="" validity="" of="" the="" methods="" and="" software="" used="" both="" for="" data="" management="" (data="" entry,="" storage,="" verification,="" correction,="" and="" retrieval)="" and="" for="" processing="" the="" data="" statistically.="" data="" management="" activities="" should="" therefore="" be="" based="" on="" thorough="" and="" effective="" sop's.="" the="" computer="" software="" used="" for="" data="" management="" and="" statistical="" analysis="" should="" be="" reliable,="" and="" documentation="" of="" appropriate="" software="" testing="" procedures="" should="" be="" available.="" vi.="" evaluation="" of="" safety="" and="" tolerability="" 6.1="" scope="" of="" evaluation="" in="" all="" clinical="" trials,="" evaluation="" of="" safety="" and="" tolerability="" constitutes="" an="" important="" element.="" in="" early="" phases,="" this="" evaluation="" is="" mostly="" of="" an="" exploratory="" nature="" and="" is="" only="" sensitive="" to="" frank="" expressions="" of="" toxicity,="" whereas="" in="" later="" phases,="" the="" establishment="" of="" the="" safety="" and="" tolerability="" profile="" of="" a="" drug="" can="" be="" characterized="" more="" fully="" in="" larger="" samples="" of="" subjects.="" later="" phase="" controlled="" trials="" represent="" an="" important="" means="" of="" exploring,="" in="" an="" unbiased="" manner,="" any="" new="" potential="" adverse="" effects,="" even="" if="" such="" trials="" generally="" lack="" power="" in="" this="" respect.="" certain="" studies="" may="" be="" designed="" with="" the="" purpose="" of="" making="" specific="" claims="" about="" superiority="" or="" equivalence="" with="" regard="" to="" safety="" and="" tolerability="" compared="" to="" another="" drug="" or="" to="" another="" dose="" of="" the="" investigational="" drug.="" such="" specific="" claims="" should="" be="" supported="" by="" relevant="" evidence="" from="" confirmatory="" studies,="" similar="" to="" that="" necessary="" for="" corresponding="" efficacy="" claims.="" 6.2="" choice="" of="" variables="" and="" data="" collection="" in="" any="" clinical="" trial,="" the="" methods="" and="" measurements="" chosen="" to="" evaluate="" the="" safety="" and="" tolerability="" of="" a="" drug="" will="" depend="" on="" a="" number="" of="" factors,="" including="" knowledge="" of="" the="" adverse="" effects="" of="" closely="" related="" drugs,="" information="" from="" nonclinical="" and="" earlier="" clinical="" studies,="" and="" possible="" consequences="" of="" the="" pharmacodynamic/="" pharmacokinetic="" properties="" of="" the="" particular="" drug,="" the="" mode="" of="" administration,="" the="" type="" of="" subjects="" to="" be="" studied,="" and="" the="" duration="" of="" the="" study.="" laboratory="" tests="" concerning="" clinical="" chemistry="" and="" hematology,="" vital="" signs,="" and="" clinical="" adverse="" events="" (diseases,="" signs,="" and="" symptoms)="" usually="" form="" the="" main="" body="" of="" the="" safety="" and="" tolerability="" data.="" the="" occurrence="" of="" serious="" adverse="" events="" and="" treatment="" discontinuations="" due="" to="" adverse="" events="" are="" particularly="" important="" to="" register="" (see="" ich="" e2a="" and="" ich="" e3).="" furthermore,="" it="" is="" recommended="" that="" a="" consistent="" methodology="" be="" used="" for="" the="" data="" collection="" and="" evaluation="" throughout="" a="" clinical="" trial="" program="" to="" facilitate="" the="" combining="" of="" data="" from="" different="" trials.="" the="" use="" of="" a="" common="" adverse="" event="" dictionary="" is="" particularly="" important.="" this="" dictionary="" has="" a="" structure="" that="" makes="" it="" possible="" to="" summarize="" the="" adverse="" event="" data="" on="" three="" different="" levels:="" system-="" organ="" class,="" preferred="" term,="" or="" included="" term.="" the="" preferred="" term="" is="" the="" level="" on="" which="" adverse="" events="" usually="" are="" summarized,="" and="" preferred="" terms="" belonging="" to="" the="" same="" system-organ="" class="" could="" then="" be="" brought="" together="" in="" the="" descriptive="" presentation="" of="" data="" (see="" ich="" e2b).="" 6.3="" set="" of="" subjects="" to="" be="" evaluated="" and="" presentation="" of="" data="" for="" the="" overall="" safety="" and="" tolerability="" assessment,="" the="" set="" of="" subjects="" to="" be="" summarized="" is="" usually="" defined="" as="" those="" subjects="" who="" received="" at="" least="" one="" dose="" of="" the="" investigational="" drug.="" safety="" and="" tolerability="" variables="" should="" be="" collected="" as="" comprehensively="" as="" possible="" from="" these="" subjects,="" including="" type="" of="" adverse="" event,="" severity,="" onset,="" and="" duration="" (see="" ich="" e2b).="" additional="" safety="" and="" tolerability="" evaluations="" may="" be="" needed="" in="" specific="" subpopulations,="" such="" as="" females,="" the="" elderly="" (see="" ich="" e7),="" the="" severely="" ill,="" or="" those="" who="" have="" a="" common="" concomitant="" treatment.="" these="" evaluations="" may="" need="" to="" address="" more="" specific="" issues="" (see="" ich="" e3).="" all="" safety="" and="" tolerability="" variables="" need="" attention="" during="" evaluation,="" and="" the="" broad="" approach="" should="" be="" indicated="" in="" the="" protocol.="" all="" adverse="" events="" should="" be="" reported,="" whether="" or="" not="" they="" are="" considered="" to="" be="" related="" to="" treatment.="" all="" available="" data="" in="" the="" study="" population="" should="" be="" accounted="" for="" in="" the="" evaluation.="" definitions="" of="" measurement="" units="" and="" reference="" ranges="" of="" laboratory="" variables="" should="" be="" made="" with="" care;="" if="" different="" units="" or="" different="" reference="" ranges="" appear="" in="" the="" same="" trial="" (e.g.,="" if="" more="" than="" one="" laboratory="" is="" involved),="" then="" measurements="" should="" be="" appropriately="" standardized="" to="" allow="" a="" unified="" evaluation.="" use="" of="" a="" toxicity="" grading="" scale="" should="" be="" prespecified="" and="" justified.="" the="" incidence="" of="" a="" certain="" adverse="" event="" is="" usually="" expressed="" in="" the="" form="" of="" a="" proportion="" relating="" number="" of="" subjects="" experiencing="" events="" to="" number="" of="" subjects="" at="" risk.="" however,="" it="" is="" not="" always="" self-evident="" how="" to="" assess="" incidence.="" for="" example,="" depending="" on="" the="" situation,="" the="" number="" of="" exposed="" subjects="" or="" the="" extent="" of="" exposure="" (in="" person-years)="" could="" be="" considered="" for="" the="" denominator.="" whether="" the="" purpose="" of="" the="" calculation="" is="" to="" estimate="" a="" risk="" or="" to="" make="" a="" comparison="" between="" treatment="" groups,="" it="" is="" important="" that="" the="" definition="" is="" given="" in="" the="" protocol.="" this="" is="" especially="" important="" if="" long-term="" treatment="" is="" planned="" and="" a="" substantial="" proportion="" of="" treatment="" withdrawals="" or="" deaths="" are="" expected.="" for="" such="" situations,="" survival="" analysis="" methods="" should="" be="" considered="" and="" cumulative="" adverse="" event="" rates="" calculated="" in="" order="" to="" avoid="" the="" risk="" of="" underestimation.="" methods="" to="" account="" for="" situations="" where="" there="" is="" a="" substantial="" background="" noise="" of="" signs="" and="" symptoms="" (e.g.,="" in="" psychiatric="" trials)="" should="" be="" considered="" in="" the="" estimation="" of="" risk="" for="" different="" adverse="" events.="" one="" such="" method="" is="" to="" make="" use="" of="" the="" ``treatment="" emergent''="" concept="" in="" which="" adverse="" events="" are="" recorded="" only="" if="" they="" emerge="" or="" worsen="" relative="" to="" pretreatment="" baseline.="" other="" methods="" to="" reduce="" the="" background="" noise="" may="" also="" be="" appropriate,="" such="" as="" ignoring="" adverse="" events="" of="" mild="" severity="" or="" requiring="" that="" an="" event="" should="" have="" been="" [[page="" 25724]]="" observed="" at="" repeated="" visits="" to="" qualify="" for="" inclusion="" in="" the="" numerator.="" such="" methods="" should="" be="" explained="" and="" justified="" in="" the="" protocol.="" 6.4="" statistical="" evaluation="" the="" investigation="" of="" safety="" and="" tolerability="" is="" a="" multidimensional="" problem.="" although="" some="" specific="" adverse="" effects="" can="" usually="" be="" anticipated="" and="" specifically="" monitored="" for="" any="" drug,="" the="" range="" of="" possible="" adverse="" effects="" is="" very="" large,="" and="" new="" and="" unforeseeable="" effects="" are="" always="" possible.="" further,="" an="" adverse="" event="" experienced="" after="" a="" protocol="" violation,="" such="" as="" use="" of="" an="" excluded="" medication,="" may="" introduce="" a="" bias.="" this="" background="" underlies="" the="" statistical="" difficulties="" associated="" with="" the="" analytical="" evaluation="" of="" safety="" and="" tolerability="" of="" drugs,="" and="" means="" that="" confirmatory="" information="" from="" phase="" iii="" clinical="" trials="" is="" the="" exception="" rather="" than="" the="" rule.="" in="" most="" trials,="" the="" safety="" and="" tolerability="" implications="" are="" best="" addressed="" by="" applying="" descriptive="" statistical="" methods="" to="" the="" data,="" supplemented="" by="" calculation="" of="" confidence="" intervals="" wherever="" this="" aids="" interpretation.="" it="" is="" also="" valuable="" to="" make="" use="" of="" graphical="" presentations="" in="" which="" patterns="" of="" adverse="" events="" are="" displayed="" both="" within="" treatment="" groups="" and="" within="" subjects.="" the="" calculation="" of="" p-values="" is="" sometimes="" useful,="" either="" as="" an="" aid="" to="" evaluating="" a="" specific="" difference="" of="" interest="" or="" as="" a="" ``flagging''="" device="" applied="" to="" a="" large="" number="" of="" safety="" and="" tolerability="" variables="" to="" highlight="" differences="" worthy="" of="" further="" attention.="" this="" is="" particularly="" useful="" for="" laboratory="" data,="" which="" otherwise="" can="" be="" difficult="" to="" summarize="" appropriately.="" it="" is="" recommended="" that="" laboratory="" data="" be="" subjected="" to="" both="" a="" quantitative="" analysis,="" e.g.,="" evaluation="" of="" treatment="" means,="" and="" a="" qualitative="" analysis,="" where="" counting="" of="" numbers="" above="" or="" below="" certain="" thresholds="" are="" calculated.="" if="" hypothesis="" tests="" are="" used,="" statistical="" adjustments="" for="" multiplicity="" to="" quantitate="" the="" type="" i="" error="" are="" appropriate,="" but="" the="" type="" ii="" error="" is="" usually="" of="" more="" concern.="" care="" should="" be="" taken="" when="" interpreting="" putative="" statistically="" significant="" findings="" when="" there="" is="" no="" multiplicity="" adjustment.="" in="" the="" majority="" of="" studies,="" investigators="" are="" seeking="" to="" establish="" that="" there="" are="" no="" clinically="" unacceptable="" differences="" in="" safety="" and="" tolerability="" compared="" with="" either="" a="" comparator="" drug="" or="" a="" placebo.="" as="" is="" the="" case="" for="" noninferiority="" or="" equivalence="" evaluation="" of="" efficacy,="" the="" use="" of="" confidence="" intervals="" is="" preferred="" to="" hypothesis="" testing="" in="" this="" situation.="" in="" this="" way,="" the="" considerable="" imprecision="" often="" arising="" from="" low="" frequencies="" of="" occurrence="" is="" clearly="" demonstrated.="" 6.5="" single="" study="" versus="" integrated="" summary="" the="" safety="" and="" tolerability="" properties="" of="" a="" drug="" are="" commonly="" summarized="" across="" studies="" continuously="" during="" an="" investigational="" product's="" development="" and,="" in="" particular,="" for="" the="" submission="" of="" a="" marketing="" application.="" the="" usefulness="" of="" this="" summary,="" however,="" is="" dependent="" on="" adequate="" and="" well-controlled="" individual="" studies="" with="" high="" data="" quality.="" the="" overall="" usefulness="" of="" a="" drug="" is="" always="" a="" question="" of="" balance="" between="" risk="" and="" benefit;="" in="" a="" single="" trial,="" such="" a="" perspective="" could="" also="" be="" considered="" even="" if="" the="" assessment="" of="" risk/benefit="" usually="" is="" performed="" in="" the="" summary="" of="" the="" entire="" clinical="" trial="" program.="" (see="" section="" 7.1.2.)="" for="" more="" details="" of="" safety="" and="" tolerability="" reports,="" see="" section="" 12="" of="" the="" ich="" guideline="" e3="" on="" ``clinical="" study="" reports:="" structure="" and="" content.''="" vii.="" reporting="" 7.1="" evaluation="" and="" reporting="" as="" stated="" in="" the="" introduction,="" the="" structure="" and="" content="" of="" clinical="" reports="" is="" the="" subject="" of="" ich="" guideline="" e3.="" that="" ich="" guideline="" fully="" covers="" the="" reporting="" of="" statistical="" work,="" appropriately="" integrated="" with="" clinical="" and="" other="" material.="" the="" current="" section="" is="" therefore="" relatively="" brief.="" during="" the="" planning="" phase="" of="" a="" trial,="" the="" principal="" features="" of="" the="" analysis="" should="" have="" been="" specified="" in="" the="" protocol="" as="" described="" in="" section="" 5.="" when="" the="" conduct="" of="" the="" trial="" is="" over="" and="" the="" data="" are="" assembled="" and="" available="" for="" preliminary="" inspection,="" it="" is="" valuable="" to="" carry="" out="" the="" blind="" review="" of="" the="" planned="" analysis="" also="" described="" in="" section="" 5.="" this="" preanalysis="" review,="" blinded="" to="" treatment,="" should:="" (1)="" cover="" decisions="" concerning="" the="" exclusion="" of="" subjects="" or="" data="" from="" the="" analysis="" sets;="" (2)="" check="" possible="" transformations="" and="" define="" outliers;="" (3)="" add="" to="" the="" model="" important="" covariates="" identified="" in="" other="" recent="" research;="" (4)="" reconsider="" the="" use="" of="" parametric="" or="" nonparametric="" methods.="" decisions="" made="" at="" this="" time="" should="" be="" described="" in="" the="" report="" and="" should="" be="" distinguished="" from="" those="" made="" after="" the="" statistician="" has="" had="" access="" to="" the="" treatment="" codes,="" as="" blind="" decisions="" will="" generally="" introduce="" less="" potential="" for="" bias.="" many="" of="" the="" more="" detailed="" aspects="" of="" presentation="" and="" tabulation="" should="" be="" finalized="" at="" or="" about="" the="" time="" of="" the="" blind="" review="" so="" that,="" by="" the="" time="" of="" the="" actual="" analysis,="" full="" plans="" exist="" for="" all="" its="" aspects="" including="" subject="" selection,="" data="" selection="" and="" modification,="" data="" summary="" and="" tabulation,="" estimation="" and="" hypothesis="" testing.="" once="" data="" validation="" is="" complete,="" the="" analysis="" should="" proceed="" according="" to="" the="" predefined="" plans;="" the="" more="" these="" plans="" are="" adhered="" to,="" the="" greater="" the="" credibility="" of="" the="" results.="" particular="" attention="" should="" be="" paid="" to="" any="" differences="" between="" the="" planned="" analysis="" and="" the="" actual="" analysis="" as="" described="" in="" the="" protocol,="" the="" protocol="" amendments,="" or="" the="" updated="" statistical="" analysis="" plan="" based="" on="" a="" blind="" review="" of="" data.="" a="" careful="" explanation="" should="" be="" provided="" for="" deviations="" from="" the="" planned="" analysis.="" all="" subjects="" who="" entered="" the="" trial="" should="" be="" accounted="" for="" in="" the="" report,="" whether="" or="" not="" they="" are="" included="" in="" the="" analysis.="" all="" reasons="" for="" exclusion="" from="" analysis="" should="" be="" documented;="" for="" any="" subject="" included="" in="" the="" set="" of="" all="" randomized="" subjects="" but="" not="" in="" the="" per-protocol="" set,="" the="" reasons="" for="" exclusion="" from="" the="" latter="" should="" also="" be="" documented.="" similarly,="" for="" all="" subjects="" included="" in="" an="" analysis="" set,="" the="" measurements="" of="" all="" important="" variables="" should="" be="" accounted="" for="" at="" all="" relevant="" time-points.="" the="" effect="" of="" all="" losses="" of="" subjects="" or="" data,="" withdrawals="" from="" treatment,="" and="" major="" protocol="" violations="" on="" the="" main="" analyses="" of="" the="" primary="" variable(s)="" should="" be="" considered="" carefully.="" subjects="" lost="" to="" followup,="" withdrawn="" from="" treatment,="" or="" with="" a="" severe="" protocol="" violation="" should="" be="" identified;="" a="" descriptive="" analysis="" of="" the="" subjects="" should="" be="" provided,="" including="" the="" reasons="" for="" their="" loss="" and="" the="" relationship="" of="" the="" loss="" to="" treatment="" and="" outcome.="" descriptive="" statistics="" form="" an="" indispensable="" part="" of="" reports.="" suitable="" tables="" and/or="" graphical="" presentations="" should="" illustrate="" clearly="" the="" important="" features="" of="" the="" primary="" and="" secondary="" variables="" and="" of="" key="" prognostic="" and="" demographic="" variables.="" the="" results="" of="" the="" main="" analyses="" relating="" to="" the="" objectives="" of="" the="" trial="" should="" be="" the="" subject="" of="" particularly="" careful="" descriptive="" presentation.="" although="" the="" primary="" goal="" of="" the="" analysis="" of="" a="" clinical="" trial="" should="" be="" to="" answer="" the="" questions="" posed="" by="" its="" main="" objectives,="" new="" questions="" based="" on="" the="" observed="" data="" may="" well="" emerge="" during="" the="" unblinded="" analysis.="" additional="" and="" perhaps="" complex="" statistical="" analysis="" may="" be="" the="" consequence.="" this="" additional="" work="" should="" be="" strictly="" distinguished="" in="" the="" report="" from="" work="" that="" was="" planned="" in="" the="" protocol.="" the="" play="" of="" chance="" may="" lead="" to="" unforeseen="" imbalances="" between="" the="" treatment="" groups="" in="" terms="" of="" baseline="" measurements="" not="" predefined="" as="" covariates="" in="" the="" analysis="" plan="" but="" having="" some="" prognostic="" importance="" nevertheless.="" this="" is="" best="" dealt="" with="" by="" showing="" that="" a="" subsidiary="" analysis="" that="" accounts="" for="" these="" imbalances="" reaches="" essentially="" the="" same="" conclusions="" as="" the="" planned="" analysis.="" if="" this="" is="" not="" the="" case,="" the="" effect="" of="" the="" imbalances="" on="" the="" conclusions="" should="" be="" discussed.="" in="" general,="" sparing="" use="" should="" be="" made="" of="" unplanned="" subsidiary="" analyses.="" subsidiary="" analyses="" are="" often="" carried="" out="" when="" it="" is="" thought="" that="" the="" treatment="" effect="" may="" vary="" according="" to="" some="" other="" factor="" or="" factors.="" an="" attempt="" may="" then="" be="" made="" to="" identify="" subgroups="" of="" subjects="" for="" whom="" the="" effect="" is="" particularly="" beneficial.="" the="" potential="" dangers="" of="" over-interpretation="" of="" unplanned="" subgroup="" analyses="" are="" well="" known="" (see="" also="" section="" 5.7)="" and="" should="" be="" carefully="" avoided.="" although="" similar="" problems="" of="" interpretation="" arise="" if="" a="" treatment="" appears="" to="" have="" no="" benefit,="" or="" an="" adverse="" effect,="" in="" a="" subgroup="" of="" subjects,="" such="" possibilities="" need="" to="" be="" properly="" assessed="" and="" should="" therefore="" be="" reported.="" finally,="" statistical="" judgement="" should="" be="" brought="" to="" bear="" on="" the="" analysis,="" interpretation,="" and="" presentation="" of="" the="" results="" of="" a="" clinical="" trial.="" to="" this="" end,="" the="" trial="" statistician="" should="" be="" a="" member="" of="" the="" team="" responsible="" for="" the="" study="" report="" and="" should="" approve="" the="" final="" report.="" 7.2="" summarizing="" the="" clinical="" database="" an="" overall="" summary="" and="" synthesis="" of="" the="" evidence="" on="" safety="" and="" efficacy="" from="" all="" the="" reported="" clinical="" trials="" is="" required="" for="" a="" marketing="" application.="" this="" may="" be="" accompanied,="" when="" appropriate,="" by="" a="" statistical="" combination="" of="" results.="" within="" the="" summary="" a="" number="" of="" areas="" of="" specific="" statistical="" interest="" arise:="" describing="" the="" demography="" and="" clinical="" features="" of="" the="" population="" treated="" during="" the="" course="" of="" the="" [[page="" 25725]]="" clinical="" trial="" program;="" addressing="" the="" key="" questions="" of="" efficacy="" by="" considering="" the="" results="" of="" the="" relevant="" (usually="" controlled)="" trials="" and="" highlighting="" the="" degree="" to="" which="" they="" reinforce="" or="" contradict="" each="" other;="" summarizing="" the="" safety="" information="" available="" from="" the="" combined="" database="" of="" all="" the="" studies="" whose="" results="" contribute="" to="" the="" marketing="" application="" and="" identifying="" potential="" safety="" issues.="" during="" the="" design="" of="" a="" clinical="" program,="" careful="" attention="" should="" be="" paid="" to="" the="" uniform="" definition="" and="" collection="" of="" measurements="" which="" will="" facilitate="" subsequent="" interpretation="" of="" the="" series="" of="" trials,="" particularly="" if="" they="" are="" likely="" to="" be="" combined="" across="" trials.="" a="" common="" dictionary="" for="" recording="" the="" details="" of="" medication,="" medical="" history,="" and="" adverse="" events="" should="" be="" selected="" and="" used.="" a="" common="" definition="" of="" the="" primary="" and="" secondary="" variables="" is="" nearly="" alway="" aworthwhile="" and="" is="" essential="" for="" meta-analysis.="" the="" manner="" of="" measuring="" key="" efficacy="" variables,="" the="" timing="" of="" assessments="" relative="" to="" randomization/entry,="" the="" handling="" of="" protocol="" violators="" and="" deviators,="" and="" perhaps="" the="" definition="" of="" prognostic="" factors,="" should="" all="" be="" kept="" compatible="" unless="" there="" are="" valid="" reasons="" not="" to="" do="" so.="" any="" statistical="" procedures="" used="" to="" combine="" data="" across="" trials="" should="" be="" described="" in="" detail.="" attention="" should="" be="" paid="" to="" the="" possibility="" of="" bias="" associated="" with="" the="" selection="" of="" trials,="" to="" the="" homogeneity="" of="" their="" results,="" and="" to="" the="" proper="" modeling="" of="" the="" various="" sources="" of="" variation.="" the="" sensitivity="" of="" conclusions="" to="" the="" assumptions="" and="" selections="" made="" should="" be="" explored.="" 7.2.1="" efficacy="" data="" individual="" clinical="" trials="" should="" always="" be="" large="" enough="" to="" satisfy="" their="" objectives.="" additional="" valuable="" information="" may="" also="" be="" gained="" by="" summarizing="" a="" series="" of="" clinical="" trials="" that="" address="" essentially="" identical="" key="" efficacy="" questions.="" the="" main="" results="" of="" such="" a="" set="" of="" studies="" should="" be="" presented="" in="" an="" identical="" form="" to="" permit="" comparison,="" usually="" in="" tables="" or="" graphs="" that="" focus="" on="" estimates="" plus="" confidence="" limits.="" the="" use="" of="" meta-analytic="" techniques="" to="" combine="" these="" estimates="" is="" often="" a="" useful="" addition="" because="" it="" allows="" a="" more="" precise="" overall="" estimate="" of="" the="" size="" of="" the="" treatment="" effects="" to="" be="" generated="" and="" provides="" a="" complete="" and="" concise="" summary="" of="" the="" results="" of="" the="" trials.="" under="" exceptional="" circumstances,="" a="" meta-analytic="" approach="" may="" also="" be="" the="" most="" appropriate="" way,="" or="" the="" only="" way,="" of="" providing="" sufficient="" overall="" evidence="" of="" efficacy="" via="" an="" overall="" hypothesis="" test.="" 7.2.2="" safety="" data="" in="" summarizing="" safety="" data,="" it="" is="" important="" to="" examine="" the="" safety="" database="" thoroughly="" for="" any="" indications="" of="" potential="" toxicity="" and="" to="" follow="" up="" any="" indications="" by="" looking="" for="" an="" associated="" supportive="" pattern="" of="" observations.="" the="" combination="" of="" the="" safety="" data="" from="" all="" human="" exposure="" to="" the="" drug="" provides="" an="" important="" source="" of="" information="" because="" its="" larger="" sample="" size="" provides="" the="" best="" chance="" of="" detecting="" the="" rarer="" adverse="" events="" and,="" perhaps,="" of="" estimating="" their="" approximate="" incidence.="" however,="" incidence="" data="" from="" this="" database="" are="" difficult="" to="" evaluate="" without="" a="" natural="" comparator="" group,="" and="" data="" from="" comparative="" studies="" are="" especially="" valuable="" in="" overcoming="" this="" difficulty.="" the="" results="" from="" studies="" that="" use="" a="" common="" comparator="" (placebo="" or="" specific="" active="" comparator)="" should="" be="" combined="" and="" presented="" separately="" for="" each="" comparator="" providing="" sufficient="" data.="" all="" indications="" of="" potential="" toxicity="" arising="" from="" exploration="" of="" the="" data="" should="" be="" reported.="" the="" evaluation="" of="" the="" reality="" of="" these="" potential="" adverse="" effects="" should="" take="" into="" account="" the="" issue="" of="" multiplicity="" arising="" from="" the="" numerous="" comparisons="" made.="" the="" evaluation="" should="" also="" make="" appropriate="" use="" of="" survival="" analysis="" methods="" to="" exploit="" the="" potential="" relationship="" of="" the="" incidence="" of="" adverse="" events="" to="" duration="" of="" exposure="" and/or="" followup.="" the="" risks="" associated="" with="" identified="" adverse="" effects="" should="" be="" appropriately="" quantified="" to="" allow="" a="" proper="" assessment="" of="" the="" risk/benefit="" relationship.="" annex="" 1="" glossary="" all="" randomized="" subjects--the="" analysis="" set="" that="" includes="" all="" subjects="" who="" were="" randomized="" to="" treatment,="" with="" these="" subjects="" assigned="" to="" the="" treatment="" group="" to="" which="" they="" were="" randomized.="" practical="" considerations,="" such="" as="" missing="" data,="" may="" lead="" to="" some="" subjects="" in="" this="" set="" not="" being="" included="" in="" the="" corresponding="" analysis.="" analysis="" plan--the="" strategy="" for="" analysis="" predefined="" in="" the="" statistical="" section="" of="" the="" protocol="" and/or="" protocol="" amendments.="" the="" plan="" may="" be="" elaborated="" in="" a="" separate="" document="" (internal="" to="" the="" sponsor)="" to="" cover="" technical="" details="" and="" procedures="" for="" implementing="" the="" statistical="" analyses.="" the="" plan="" should="" be="" reviewed="" and="" possibly="" updated="" as="" a="" result="" of="" the="" blind="" review="" of="" the="" data.="" bayesian="" approaches--approaches="" to="" data="" analysis="" that="" provide="" a="" posterior="" probability="" distribution="" for="" some="" parameter="" (e.g.,="" treatment="" effect),="" derived="" from="" the="" observed="" data="" and="" a="" prior="" probability="" distribution="" for="" the="" parameter.="" the="" posterior="" distribution="" is="" then="" used="" as="" the="" basis="" for="" statistical="" inference.="" bias="" (statistical="" and="" operational)--the="" systematic="" tendency="" of="" any="" factors="" associated="" with="" the="" design,="" conduct,="" analysis,="" and="" evaluation="" of="" the="" results="" of="" a="" clinical="" trial="" to="" make="" the="" estimate="" of="" a="" treatment="" effect="" deviate="" from="" its="" true="" value.="" bias="" introduced="" through="" deviations="" in="" conduct="" is="" referred="" to="" as="" ``operational''="" bias.="" the="" other="" sources="" of="" bias="" listed="" above="" are="" referred="" to="" as="" ``statistical.''="" blind="" review--the="" checking="" and="" assessment="" of="" data="" during="" the="" course="" of="" the="" study,="" but="" before="" the="" breaking="" of="" the="" blind,="" for="" the="" purpose="" of="" finalizing="" the="" analysis="" plan.="" content="" validity--the="" extent="" to="" which="" a="" variable="" (e.g.,="" a="" rating="" scale)="" measures="" what="" it="" is="" supposed="" to="" measure.="" double="" dummy--a="" technique="" for="" retaining="" the="" blind="" when="" administering="" supplies="" in="" a="" clinical="" trial,="" when="" the="" two="" treatments="" cannot="" be="" made="" identical.="" supplies="" are="" prepared="" for="" treatment="" a="" (active="" and="" indistinguishable="" placebo)="" and="" for="" treatment="" b="" (active="" and="" indistinguishable="" placebo).="" subjects="" then="" take="" two="" sets="" of="" treatment;="" either="" a="" (active)="" and="" b="" (placebo),="" or="" a="" (placebo)="" and="" b="" (active).="" dropout--a="" subject="" in="" a="" clinical="" trial="" who="" for="" any="" reason="" fails="" to="" continue="" in="" the="" trial="" until="" the="" last="" visit="" required="" of="" him/her="" by="" the="" study="" protocol.="" equivalence="" trial--a="" trial="" with="" the="" primary="" objective="" of="" showing="" that="" the="" response="" to="" two="" or="" more="" treatments="" differs="" by="" an="" amount="" which="" is="" clinically="" unimportant.="" this="" is="" usually="" demonstrated="" by="" showing="" that="" the="" true="" treatment="" difference="" is="" likely="" to="" lie="" between="" a="" lower="" and="" an="" upper="" equivalence="" margin="" of="" clinically="" acceptable="" differences.="" frequentist="" methods--statistical="" methods,="" such="" as="" significance="" tests="" and="" confidence="" intervals,="" which="" can="" be="" interpreted="" in="" terms="" of="" the="" frequency="" of="" certain="" outcomes="" occurring="" in="" hypothetical="" repeated="" realizations="" of="" the="" same="" experimental="" situation.="" generalizability,="" generalization--the="" extent="" to="" which="" the="" findings="" of="" a="" clinical="" trial="" can="" be="" reliably="" extrapolated="" from="" the="" subjects="" who="" participated="" in="" the="" trial="" to="" a="" broader="" patient="" population.="" global="" assessment="" variable--a="" single="" variable,="" usually="" a="" scale="" of="" ordered="" categorical="" ratings,="" that="" integrates="" objective="" variables="" and="" the="" investigator's="" overall="" impression="" about="" the="" state="" or="" change="" in="" state="" of="" a="" subject.="" independent="" data="" monitoring="" committee="" (idmc)="" (data="" and="" safety="" monitoring="" board,="" monitoring="" committee,="" data="" monitoring="" committee)--="" an="" independent="" data="" monitoring="" committee="" that="" may="" be="" established="" by="" the="" sponsor="" to="" assess="" at="" intervals="" the="" progress="" of="" a="" clinical="" trial,="" the="" safety="" data,="" and="" the="" critical="" efficacy="" endpoints,="" and="" to="" recommend="" to="" the="" sponsor="" whether="" to="" continue,="" modify,="" or="" stop="" a="" trial.="" intention-to-treat="" principle--the="" principle="" that="" asserts="" that="" the="" effect="" of="" a="" treatment="" policy="" can="" be="" best="" assessed="" by="" evaluating="" on="" the="" basis="" of="" the="" intention="" to="" treat="" a="" subject="" (i.e.,="" the="" planned="" treatment="" regimen)="" rather="" than="" the="" actual="" treatment="" given.="" it="" has="" the="" consequence="" that="" subjects="" allocated="" to="" a="" treatment="" group="" should="" be="" followed="" up,="" assessed,="" and="" analyzed="" as="" members="" of="" that="" group="" irrespective="" of="" their="" compliance="" to="" the="" planned="" course="" of="" treatment.="" interaction="" (qualitative="" and="" quantitative)--the="" situation="" in="" which="" a="" treatment="" contrast="" (e.g.,="" difference="" between="" investigational="" product="" and="" control)="" is="" dependent="" on="" another="" factor="" (e.g.,="" center).="" a="" quantitative="" interaction="" refers="" to="" the="" case="" where="" the="" magnitude="" of="" the="" contrast="" differs="" at="" the="" different="" levels="" of="" the="" factor,="" whereas="" for="" a="" qualitative="" interaction="" the="" direction="" of="" the="" contrast="" differs="" for="" at="" least="" one="" level="" of="" the="" factor.="" inter-="" and="" intrarater="" reliability--the="" level="" of="" consistency="" of="" a="" rater="" (intra)="" or="" a="" group="" of="" raters="" (inter)="" in="" making="" an="" assessment="" of="" treatment="" outcome.="" interim="" analysis--any="" analysis="" intended="" to="" compare="" treatment="" arms="" with="" respect="" to="" efficacy="" or="" safety="" at="" any="" time="" prior="" to="" the="" formal="" completion="" of="" a="" trial.="" meta-analysis--the="" formal="" evaluation="" of="" the="" quantitative="" evidence="" from="" two="" or="" more="" trials="" bearing="" on="" the="" same="" question.="" this="" most="" commonly="" involves="" the="" statistical="" combination="" of="" summary="" statistics="" from="" the="" various="" trials,="" but="" the="" term="" is="" sometimes="" used="" to="" refer="" to="" the="" combination="" of="" the="" raw="" data.="" [[page="" 25726]]="" multicenter="" trial--a="" trial="" involving="" two="" or="" more="" study="" centers,="" a="" common="" study="" protocol,="" and="" a="" single="" analysis="" plan="" pooling="" the="" data="" across="" all="" centers.="" noninferiority="" trial--a="" trial="" with="" the="" primary="" objective="" of="" showing="" that="" the="" response="" to="" the="" investigational="" product="" is="" not="" clinically="" inferior="" to="" a="" comparative="" agent="" (active="" or="" placebo="" control).="" preferred="" and="" included="" terms--in="" a="" hierarchical="" medical="" dictionary,="" for="" example,="" who-art,="" the="" included="" term="" is="" the="" lowest="" level="" of="" dictionary="" term="" to="" which="" the="" investigator="" description="" is="" coded.="" the="" preferred="" term="" is="" the="" level="" of="" grouping="" of="" included="" terms="" typically="" used="" in="" reporting="" frequency="" of="" occurrence.="" for="" example,="" the="" investigator="" text="" ``pain="" in="" the="" left="" arm''="" might="" be="" coded="" to="" the="" included="" term="" ``joint="" pain,''="" which="" is="" reported="" at="" the="" preferred="" term="" level="" as="" ``arthralgia.''="" per="" protocol="" set="" (valid="" cases,="" efficacy="" sample,="" evaluable="" subjects="" sample)--the="" set="" of="" data="" generated="" by="" the="" subset="" of="" subjects="" who="" complied="" with="" the="" protocol="" sufficiently="" to="" ensure="" that="" these="" data="" would="" be="" likely="" to="" exhibit="" the="" effects="" of="" treatment="" according="" to="" the="" underlying="" scientific="" model.="" compliance="" covers="" such="" considerations="" as="" exposure="" to="" treatment,="" availability="" of="" measurements,="" and="" absence="" of="" major="" protocol="" violations.="" safety="" and="" tolerability--the="" safety="" of="" a="" medical="" product="" concerns="" the="" medical="" risk="" to="" the="" subject,="" usually="" assessed="" in="" a="" clinical="" trial="" by="" laboratory="" tests="" (including="" clinical="" chemistry="" and="" hematology),="" vital="" signs,="" clinical="" adverse="" events="" (diseases,="" signs="" and="" symptoms),="" and="" other="" special="" safety="" tests="" (e.g.,="" electrocardiograms,="" ophthalmology).="" the="" tolerability="" of="" the="" medical="" product="" represents="" the="" degree="" to="" which="" overt="" adverse="" effects="" can="" be="" tolerated="" by="" the="" subject.="" superiority="" trial--a="" trial="" with="" the="" primary="" objective="" of="" showing="" that="" the="" response="" to="" the="" investigational="" product="" is="" superior="" to="" a="" comparative="" agent="" (active="" or="" placebo="" control).="" surrogate="" variable--a="" variable="" that="" provides="" an="" indirect="" measurement="" of="" effect="" in="" situations="" where="" direct="" measurement="" of="" clinical="" effect="" is="" not="" feasible="" or="" practical.="" treatment="" effect--an="" effect="" attributed="" to="" a="" treatment="" in="" a="" clinical="" trial.="" in="" most="" clinical="" trials,="" the="" treatment="" effect="" of="" interest="" is="" a="" comparison="" (or="" contrast)="" of="" two="" or="" more="" treatments.="" treatment="" emergent--an="" event="" that="" emerges="" during="" treatment,="" having="" been="" absent="" pretreatment,="" or="" worsens="" relative="" to="" the="" pretreatment="" state.="" dated:="" april="" 30,="" 1997.="" william="" k.="" hubbard,="" associate="" commissioner="" for="" policy="" coordination.="" [fr="" doc.="" 97-12139="" filed="" 5-8-97;="" 8:45="" am]="" billing="" code="" 4160-01-f="">

Document Information

Published:
05/09/1997
Department:
Food and Drug Administration
Entry Type:
Notice
Action:
Notice.
Document Number:
97-12139
Dates:
Written comments by June 23, 1997.
Pages:
25712-25726 (15 pages)
Docket Numbers:
Docket No. 97D-0174
PDF File:
97-12139.pdf