SlideShare ist ein Scribd-Unternehmen logo
1 von 169
Downloaden Sie, um offline zu lesen
Clinical Study and Safety Reporting
Activities
Writing a Clinical Study Report
&
Overview of safety reports :Periodic Safety Reports: DSUR,
PSUR, PBRER, ASR, IB, and Orphan Designation Annual
Report
Pr. Peivand Pirouzi
ICH E3 Structure and Content of
Clinical Study Reports
1. Title page
2. Synopsis
3. Table of contents
4. List of abbreviations
5. Ethics
6. Investigators and study administrative
structure
7. Introduction
8. Study objectives
9. Investigational plan
10. Study patients
11. Efficacy evaluation
12. Safety evaluation
13. Discussion and overall conclusions
14. Tables, figures and graphs referred
to but not included in the
text
15. Reference list
16. Appendices
* Details for Sections 9 – 12 on next slides
2
ICH E3 Structure and Content of
Clinical Study Reports (cont.)
9. Investigational plan
9.1 Overall study design and plan
description
9.2 Discussion of study design, including
the choice of control groups
9.3 Selection of study population
9.3.1 Inclusion Criteria
9.3.2 Exclusion Criteria
9.3.3 Removal of Patients from Therapy or
Assessment
9.4 Treatments
9.4.1 Treatments Administered
9.4.2 Identity of Investigational Product(s)
9.4.3 Method of Assigning Patients to
Treatment Groups
9.4.4 Selection of Doses in the Study
9.4 Treatments (cont.)
9.4.5 Selection and Timing of Dose for each
Patient
9.4.6 Blinding
9.4.7 Prior and Concomitant Therapy
9.4.8 Treatment Compliance
9.5 Efficacy and safety variables
9.5.1 Efficacy and Safety Measurements
Assessed and Flow Chart
9.5.2 Appropriateness of Measurements
9.5.3 Primary Efficacy Variable(s)
9.5.4 Drug Concentration Measurements
9.6 Data quality assurance
9.7 Statistical methods planned in the
protocol & determination of sample
size
9.8 Changes in the conduct of the study
or planned analyses
3
ICH E3 Structure and Content of
Clinical Study Reports (cont.)
10 Study patients
10.1 Disposition of patients
10.2 Protocol deviations
11. Efficacy evaluation
11.1 Data sets analyzed
11.2 Demographic and other baseline
characteristics
11.3 Measurements of treatment
compliance
11.4 Efficacy results and tabulations of
individual patient data
12. Safety evaluation
12.1 Extent of exposure
12.2 Adverse events (AEs)
12.3 Deaths, other SAEs, and other
significant adverse events
12.4 Clinical laboratory evaluation
12.5 Vital signs, physical findings and
other observations related
to safety
12.6 Safety conclusions
4
1. Title Page
• The title page should contain the following information:
• study title
• name of test drug/investigational product
• indication studied
• if not apparent from the title, a brief (1 to 2 sentences) description giving design (parallel,
cross-over, blinding, randomised) comparison (placebo, active, dose/response), duration,
dose, and patient population
• name of the sponsor
• protocol identification (code or number)
• development phase of study
• study initiation date (first patient enrolled, or any other verifiable
• date of early study termination, if any
• study completion date (last patient completed)
• name and affiliation of principal or coordinating investigator(s) or sponsor's responsible
medical officer
• name of company/sponsor signatory (the person responsible for the study report within
the company/sponsor. The name, telephone number and fax number of the
company/sponsor contact persons for questions arising during review of the study report
should be indicated an this page or in the letter of application.)
• statement indicating whether the study was performed in compliance with Good Clinical
Practices (GCP), including the archiving of essential documents
• date of the report (identify any earlier reports from the same study by title and date)
2. Synopsis
• A brief synopsis (usually limited to 3 pages)
that summarises the study should be provided
(see Annex I of the guidance document for an
example of a synopsis format used in Europe).
The synopsis should include numerical data to
illustrate results, not just text or p-values.
3. Table of Contents For The
Individual Clinical Study Report
The table of contents should include:
• the page number or other locating
information of each section, including
summary tables, figures and graphs,
• a list and the locations of appendices,
tabulations and any case report forms
provided.
4. List of Abbreviations and Definition
of Terms
• A list of the abbreviations, and lists and
definitions of specialised or unusual terms or
measurements units used in the report should
be provided. Abbreviated terms should be
spelled out and the abbreviation indicated in
parentheses at first appearance in the text.
5. Ethics
5.1 Independent Ethics Committee (IEC) or
Institutional Review Board (IRB)
• It should be confirmed that the study and any
amendments were reviewed by an
Independent Ethics Committee or Institutional
Review Board. A list of all IECs or IRBs
consulted should be given in (appendix 16.1.3)
and, if required by the regulatory authority,
the name of the committee Chair should be
provided.
5.2 Ethical Conduct of the Study
• It should be confirmed that the study was
conducted in accordance with the ethical
principles that have their origins in the
Declaration of Helsinki.
5.3 Patient Information and Consent
• How and when informed consent was
obtained in relation to patient enrollment,
(e.g., at allocation, pre-screening) should be
described.
• Representative written information for the
patient (if any) and a sample patient consent
form should be provided in (appendix 16.1.3).
6. Investigators and Study
Administrative Structure
• The administrative structure of the study (e.g.,
principal investigator, coordinating
investigator, steering committee,
administration, monitoring and evaluation
committees, institutions, statistician, central
laboratory facilities, contract research
organization (C.R.O.), clinical trial supply
management) should be described briefly in
the body of the report.
6. Investigators and Study
Administrative Structure
The listing should include:
• Investigators
• Any other person carrying out observations of primary
or other major efficacy variables, such as a nurse,
physician's assistant, clinical psychologist, clinical
pharmacist, or house staff physician. It is not necessary
to include in this list a person with only an occasional
role, e.g., an on-call physician who dealt with a
possible adverse effect or a temporary substitute for
any of the above.
• The author(s) of the report, including the responsible
biostatistician(s).
6. Investigators and Study
Administrative Structure
• Where signatures of the principal or
coordinating investigators are required by
regulatory authorities, these should be
included in (appendix 16.1.5 see Annex II for a
sample form). Where these are not required,
the signature of the sponsor's responsible
medical officer should be provided in
(appendix 16.1.5).
7. Introduction
• The introduction should contain a brief statement
(maximum: 1 page) placing the study in the
context of the development of the test
drug/investigational product, relating the critical
features of the study (e.g., rationale and aims,
target population. treatment, duration, primary
endpoints) to that development. Any guidelines
that were followed in the development of the
protocol or any other agreements/meetings
between the sponsor/company and regulatory
authorities that are relevant to the particular
study, should be identified or described.
8. Study Objectives
• A statement describing the overall purpose(s)
of the study should be provided
9. Investigational Plan
9.1 Overall Study Design and Plan - Description
• The overall study plan and design (configuration) of the
study (e.g., parallel, crossover) should be described
briefly but clearly, using charts and diagrams as
needed. If other studies used a very similar protocol, it
may be useful to note this and describe any important
differences. The actual protocol and any changes
should be included as (appendix 16.1.1) and a sample
case report form (unique pages only; i.e., it is not
necessary to include identical pages from forms for
different evaluations or visits) as (appendix 16.1.2). If
any of the information in this section comes from
sources other than the protocol, these should be
identified.
9. Investigational Plan
9.1 Overall Study Design and Plan - Description
The information provided should include:
• treatments studied (specific drugs, doses and procedures)
• patient population studied and the number of patients to be included.
• level and method of blinding/masking (e.g., open, double-blind, single-
blind, blinded evaluators and unblinded patients and/or investigators)
• kind of control(s) (e.g., placebo, no treatment, active drug, dose-response,
historical) and study configuration (parallel, cross-over)
• method of assignment to treatment (randomisation, stratification)
• sequence and duration of all study periods, including pre-randomisation
and post-treatment periods, therapy withdrawal periods and single- and
double-blind treatment periods. When patients are randomized should be
specified. It is usually helpful to display the design graphically with a flow
chart which includes timing of assessments (see Annexes IIIa and Illb for
an example).
• any safety, data monitoring or special steering or evaluation committees
• any interim analyses.
9.2 Discussion of Study Design,
Including the Choice of Control
Groups
• The specific control chosen and the study design
used should be discussed, as necessary.
• Known or potential problems associated with the
study design or control group chosen, should be
discussed in light of the specific disease and
therapies being studied. For a crossover design,
for example, there should be consideration,
among other things, of the likelihood of
spontaneous change in the disease and of carry-
over effects of treatment during the study.
9.3 Selection of Study Population
9.3.1 Inclusion Criteria
• The patient population and the selection criteria used
to enter the patients into the study should be
described, and the suitability of the population for the
purposes of the study discussed. Specific diagnostic
criteria used, as well as specific disease requirements
(e.g., disease of a particular severity or duration,
results of a particular test or rating scale(s) or physical
examination, particular features of clinical history, such
as failure or success on prior therapy, or other
potential prognostic factors and any age, sex or ethnic
factors) should be presented.
9.3.2 Exclusion Criteria
• The criteria for exclusion at entry into the
study should be specified and the rationale
(e.g., safety concerns, administrative reasons
or lack of suitability for the trial) provided. The
impact of exclusions on the generalisability of
the study should be discussed in (section 13)
of the study report, or in an overview of safety
and efficacy.
9.3.3 Removal of Patients from
Therapy or Assessment
• The predetermined reasons for removing
patients from therapy or assessment
observation, if any, should be described, as
should the nature and duration of any
planned follow-up observations in those
patients.
9.4 Treatments
9.4.1 Treatments Administered
• The precise treatments or diagnostic agents to
be administered in each arm of the study, and
for each period of the study, should be
described including route and mode of
administration, dose and dosage schedule.
9.4.2 Identity of Investigational
Product(s)
• In the text of the report, a brief description of
the test drug(s)/investigational product(s)
(formulation, strength, batch number(s))
should be given. If more than one batch of
test drug/investigational product was used,
patients receiving each batch should be
identified in (appendix 16.1.6).
9.4.3 Method of Assigning Patients to
Treatment Groups
• The specific methods used to assign patients
to treatment groups, e.g., centralised
allocation, allocation within sites, adaptive
allocation (that is, assignment on the basis of
earlier assignment or outcome) should be
described in the text of the report, including
any stratification or blocking procedures. Any
unusual features should be explained.
9.4.4 Selection of Doses in the Study
• The doses or dose ranges used in the study
should be given for all treatments and the
basis for choosing them described (e.g., prior
experience in humans, animal data).
9.4.5 Selection and Timing of Dose for
Each Patient
• Procedures for selecting each patient's dose of
test drug/investigational product and active
control/comparator should be described. These
procedures can vary from simple random
assignment to a selected fixed drug/dose
regimen, to some specified titration procedure,
to more elaborate response-determined selection
procedures, e.g. where dose is titrated upward at
intervals until intolerance or some specified
endpoint is achieved. Procedures for back-
titration, if any, should also be described.
9.4.6 Blinding
• A description of the specific procedures used
to carry out blinding should be provided (e.g.,
how bottles were labeled, labels that reveal
blind-breakage, sealed code list/envelopes,
double dummy techniques), including the
circumstances in which the blind would be
broken for an individual or for all patients,
e.g., for serious adverse events, the
procedures used and who had access to
patient codes.
9.4.7 Prior and Concomitant Therapy
• Which drugs or procedures were allowed before
and during the study, whether and how their use
was recorded, and any other specific rules and
procedures related to permitted or forbidden
concomitant therapy should be described. How
allowed concomitant therapy might affect the
outcome due either to drug-drug interaction or to
direct effects on the study endpoints should be
discussed, and how the independent effects of
concomitant and study therapies could be
ascertained should be explained.
9.4.8 Treatment Compliance
• The measures taken to ensure and document
treatment compliance should be described,
e.g., drug accountability, diary cards, blood,
urine or other body fluid drug level
measurements, or medication event
monitoring.
9.5 Efficacy and Safety Variables
9.5.1 Efficacy and Safety Measurements Assessed and
Flow Chart
• The specific efficacy and safety variables to be
assessed and laboratory tests to be conducted,
their schedule (days of study, time of day, relation
to meals, and the timing of critical measures in
relation to test drug administration, e.g., just
prior to next dose, two hours after dose), the
methods for measuring them, and the persons
responsible for the measurements should be
described. If there were changes in personnel
carrying out critical measurements, these should
be reported.
9.5.2 Appropriateness of
Measurements
• If any of the efficacy or safety assessments
was not standard, i.e., widely used and
generally recognised as reliable, accurate, and
relevant (able to discriminate between
effective and ineffective agents), its reliability,
accuracy and relevance should be
documented. It may be helpful to describe
alternatives considered but rejected.
9.5.2 Appropriateness of
Measurements
• If a surrogate endpoint (a laboratory
measurement or physical measurement or
sign that is not a direct measure of clinical
benefit) was used as a study endpoint, this
should be justified e.g., by reference to clinical
data, publications, guidelines or previous
actions by regulatory authorities.
9.5.3 Primary Efficacy Variable(s)
• The primary measurements and endpoints used
to determine efficacy should be clearly specified.
Although the critical efficacy measurements may
seem obvious, when there are multiple variables,
or when variables are measured repeatedly, the
protocol should identify the primary ones, with
an explanation of why they were chosen, or
designate the pattern of significant findings or
other method of combining information that
would be interpreted as supporting efficacy.
9.5.4 Drug Concentration
Measurements
• Any drug concentrations to be measured, and the
sample collection times and periods in relation to
the timing of drug administration, should be
described. Any relation of drug administration
and sampling to ingestion of food, posture and
the possible effects of concomitant
medication/alcohol/caffeine/nicotine should also
be addressed. The biological sample measured,
the handling of samples and the method of
measurement used should be described, referring
to published and/or internal assay validation
documentation for methodological details.
9.6 Data Quality Assurance
• The quality assurance and quality control
systems implemented to assure the quality of
the data should be described in brief. If none
were used, this should be stated.
Documentation of inter-laboratory
standardisation methods and quality
assurance procedures, if used, should be
provided under (appendix 16.1.10).
9.7 Statistical Methods Planned in the
Protocol and Determination of
Sample Size
9.7.1 Statistical and Analytical Plans
• The statistical analyses planned in the protocol and any
changes made before outcome results were available
should be described. In this section emphasis should
be on which analyses, comparisons and statistical tests
were planned, not on which ones were actually used. If
critical measurements were made more than once, the
particular measurements (e.g., average of several
measurements over the entire study, values at
particular times, values only from study completers, or
last on-therapy value) planned as the basis for
comparison of test drug/investigational product and
control should be specified.
9.7.2 Determination of Sample Size
• The planned sample size and the basis for it,
such as statistical considerations or practical
limitations, should be provided. Methods for
sample size calculation should be given
together with their derivations or source of
reference. Estimates used in the calculations
should be given and explanations provided as
to how they were obtained.
9.8 Changes in the Conduct of the
Study or Planned Analyses
• Any change in the conduct of the study or planned
analyses (e.g., dropping a treatment group, changing
the entry criteria or drug dosages, adjusting the sample
size, etc.) instituted after the start of the study should
be described. The time(s) and reason(s) for the
change(s), the procedure used to decide on the
change(s), the person(s) or group(s) responsible for the
change(s) and the nature and content of the data
available (and to whom they were available) when the
change was made should also be described, whether
the change was documented as a formal protocol
amendment or not (Personnel changes need not be
included.) A
10. Study Patients
10.1 Disposition of Patients
• There should be a clear accounting of all patients who
entered the study, using figures or tables in the text of
the report. The numbers of patients who were
randomised, and who entered and completed each
phase of the study, (or each week/month of the study)
should be provided, as well as the reasons for all post-
randomisation discontinuations, grouped by treatment
and by major reason (lost to follow-up, adverse event,
poor compliance, etc.). It may also be relevant to
provide the number of patients screened for inclusion
and a breakdown of the reasons for excluding patients
during screening, if this could help clarify the
appropriate patient population for eventual drug use.
10.2 Protocol Deviations
• All important deviations related to study inclusion or
exclusion criteria, conduct of the trial, patient management
or patient assessment should be described.
• In the body of the text, protocol deviations should be
appropriately summarized by centre and grouped into
different categories, such as:
• those who entered the study even though they did not
satisfy the entry criteria
• those who developed withdrawal criteria during the study
but were not withdrawn
• those who received the wrong treatment or incorrect dose
• those who received an excluded concomitant treatment
In (appendix 16.2.2), individual patients with these protocol deviations should be listed,
broken down by centre for multicentre studies.
11. Efficacy Evaluation
11.1 Data Sets Analysed
• xactly which patients were included in each efficacy
analysis should be precisely defined, e.g., all patients
receiving any test drugs/investigational products, all
patients with any efficacy observation or with a certain
minimum number of observations, only patients
completing the trial, all patients with an observation
during a particular time window, only patients with a
specified degree of compliance, etc. It should be clear,
if not defined in the study protocol, when, (relative to
study unblinding), and how inclusion/exclusion criteria
for the data sets analysed were developed.
11.2 Demographic and Other Baseline
Characteristics
• Group data for the critical demographic and baseline
characteristics of the patients, as well as other factors
arising during the study that could affect response, should
be presented in this section and comparability of the
treatment groups for all relevant characteristics should be
displayed by use of tables or graphs in (section 14.1).
• The data for the patient sample included in the "all patients
with data" analysis should be given first. This can then be
followed by data on other groups used in principal analyses,
such as the "per-protocol" analysis or other analyses, e.g.,
groups defined by compliance, concomitant
disease/therapy, or demographic/baseline characteristics.
When such groups are used, data for the complementary
excluded group should also be shown.
11.2 Demographic and Other Baseline
Characteristics
• The critical variables will depend on the specific nature of the disease and on the protocol
but will usually include:
• demographic variables
– age
– sex
– race
• disease factors
– specific entry criteria (if not uniform), duration, stage and severity of disease and other clinical
classifications and sub-groupings in common usage or of known prognostic significance.
– baseline values for critical clinical measurements carried out during the study or identified as important
indicators of prognosis or response to therapy.
– concomitant illness at trial initiation, such as renal disease, diabetes, heart failure
– relevant previous illness
– relevant previous treatment for illness treated in the study
– concomitant treatment maintained, even if the dose was changed during the study, including oral
contraceptive and hormone replacement therapy; treatments stopped at entry into the study period (or
changed at study initiation)
• other factors that might affect response to therapy (e.g., weight, renin status, antibody
levels, metabolic status)
• other possibly relevant variables (e.g., smoking, alcohol intake, special diets) and, for
women, menstrual status and date of last menstrual period, if pertinent for the study
11.3 Measurements of Treatment
Compliance
• Any measurements of compliance of
individual patients with the treatment
regimen under study and drug concentrations
in body fluids should be summarised, analysed
by treatment group and time interval, and
tabulated in (Appendix 16.2.5).
11.4 Efficacy Results and Tabulations
of Individual Patient Data
11.4.1 Analysis of Efficacy
• Treatment groups should be compared for all critical
measures of efficacy (primary and secondary
endpoints; any pharmacodynamic endpoints
studied), as well as benefit/risk assessment(s) in each
patient where these are utilized.
• In general, the results of all analyses contemplated in
the protocol and an analysis including all patients
with on-study data should be performed in studies
intended to establish efficacy. The analysis should
show the size (point estimate) of the difference
between the treatments, the associated confidence
interval, and where utilized, the results of hypothesis
testing.
11.4.2 Statistical/Analytical Issues
• The statistical analysis used should be
described for clinical and statistical reviewers
in the text of the report, with detailed
documentation of statistical methods (see
section Annex IX) presented in (appendix
16.1.9).
11.4.2.1 Adjustments for Covariates
• Selection of, and adjustments for,
demographic or baseline measurements,
concomitant therapy, or any other covariate or
prognostic factor should be explained in the
report, and methods of adjustment, results of
analyses, and supportive information (e.g.,
ANCOVA or Cox regression output) should be
included in the detailed documentation of
statistical methods.
11.4.2.2 Handling of Dropouts or
Missing Data
• There are several factors that may affect dropout rates.
These include the duration of the study, the nature of the
disease, the efficacy and toxicity of the drug under study,
and other factors that are not therapy related. Ignoring the
patients who dropped out of the study and drawing
conclusions based only on patients who completed the
study can be misleading.
• A large number of dropouts, however, even if included in an
analysis, may introduce bias, particularly if there are more
early dropouts in one treatment group or the reasons for
dropping out are treatment or outcome related. Although
the effects of early dropouts, and sometimes even the
direction of bias, can be difficult to determine, possible
effects should be explored as fully as possible.
11.4.2.3 Interim Analyses and Data
Monitoring
• The process of examining and analysing data
accumulating in a clinical trial, either formally
or informally, can introduce bias and/or
increase type I error. Therefore, all interim
analyses, formal or informal, pre-planned or
ad hoc, by any study participant, sponsor staff
member, or data monitoring group should be
described in full, even if the treatment groups
were not identified.
11.4.2.4 Multicentre Studies
• A multicentre study is a single study under a
common protocol, involving several centres (e.g.,
clinics, practices, hospitals) where the data
collected are intended to be analysed as a whole
(as opposed to a post-hoc decision to combine
data or results from separate studies). Individual
centre results should be presented, however,
where appropriate, e.g., when the centres have
sufficient numbers of patients to make such
analysis potentially valuable, the possibility of
qualitative or quantitative treatment-by-centre
interaction should be explored.
11.4.2.5 Multiple
Comparison/Multiplicity
• False positive findings increase in number as the
number of significance tests (number of
comparisons) performed increases. If there was
more than one primary endpoint (outcome
variable), more than one analysis of particular
endpoint, or if there were multiple treatment
groups, or subsets of the patient population
being examined, the statistical analysis should
reflect awareness of this and either explain the
statistical adjustment used for type I error criteria
or give reasons why it was considered
unnecessary.
11.4.2.6 Use of an "Efficacy Subset" of
Patients
• Particular attention should be devoted to the
effects of dropping patients with available
data from analyses because of poor
compliance, missed visits, ineligibility, or any
other reason. As noted above, an analysis
using all available data should be carried out
for all studies intended to establish efficacy,
even if it is not the analysis proposed as the
primary analysis by the applicant.
11.4.2.7 Active-Control Studies
Intended to Show Equivalence
• If an active control study is intended to show
equivalence (i.e., lack of a difference greater than
a specified size) between the test
drug/investigational product and the active
control/comparator, the analysis should show the
confidence interval for the comparison between
the two agents for critical end points and the
relation of that interval to the prespecified
degree of inferiority that would be considered
unacceptable. (See 9.2 for important
considerations when using the active control
equivalence design.)
11.4.2.8 Examination of Subgroups
• If the size of the study permits, important
demographic or baseline value-defined
subgroups should be examined for unusually
large or small responses and the results
presented, e.g., comparison of effects by age,
sex, or race, by severity or prognostic groups,
by history of prior treatment with a drug of
the same class, etc.
11.4.3 Tabulation of Individual
Response Data
• In addition to tables and graphs representing group
data, individual response data and other relevant study
information should be presented in tables. Some
regulatory authorities may require all individual data in
archival case report tabulations.
• What needs to be included in the report will vary from
study to study and from one drug class to another and
the applicant must decide, if possible after consultation
with the regulatory authority, what to include in
appendix to the study report.
11.4.4 Drug Dose, Drug
Concentration, and Relationships to
Response
• When the dose in each patient can vary, the actual
doses received by patients should be shown and
individual patient's doses should be tabulated.
• Although studies not designed as dose-response
studies may have limited ability to contribute dose-
response information, the available data should be
examined for whatever information they can yield. In
examining the dose response, it may be helpful to
calculate dose as mg/kg body weight or mg/m2 body
surface.
11.4.5 Drug-Drug and Drug-Disease
Interactions
• Any apparent relationship between response
and concomitant therapy and between
response and past and/or concurrent illness
should be described.
11.4.6 By-Patient Displays
• While individual patient data ordinarily can be
displayed in tabular listings, it has on occasion
been helpful to construct individual patient
profiles in other formats, such as graphic
displays. These might for example, show the
value of a particular parameter(s) over time,
the drug dose over the same period, and the
times of particular events (e.g., an adverse
event or change in concomitant therapy).
11.4.7 Efficacy Conclusions
• The important conclusions concerning efficacy
should be concisely described, considering
primary and secondary endpoints, pre-
specified and alternative statistical
approaches and results of exploratory
analyses.
12. Safety Evaluation
• Analysis of safety-related data can be considered at
three levels. First, the extent of exposure (dose,
duration, number of patients) should be examined to
determine the degree to which safety can be assessed
from the study. Second, the more common adverse
events, laboratory test changes, etc. should be
identified, classified in some reasonable way,
compared for treatment groups, and analysed, as
appropriate, for factors that may affect the frequency
of adverse reactions/events, such as time dependence,
relation to demographic characteristics, relation to
dose or drug concentration, etc.
12.1 Extent of Exposure
• The extent of exposure to test
drugs/investigational products (and to active
control and placebo) should be characterised
according to the number of patients exposed,
the duration of exposure, and the dose to
which they were exposed.
– Duration
– Dose
– Drug concentration
12.2 Adverse Events (AES)
12.2.1 Brief Summary of Adverse
Events
• The overall adverse event experience in the
study should be described in a brief narrative,
supported by the following more detailed
tabulations and analyses. In these tabulations
and analyses, events associated with both the
test drug and control treatment should be
displayed.
12.2.2 Display of Adverse Events
• All adverse events occurring after initiation of
study treatments (including events likely to be
related to the underlying disease or likely to
represent concomitant illness, unless there is a
prior agreement with the regulatory authority to
consider specified events as disease related)
should be displayed in summary tables (section
14.3.1). The tables should include changes in vital
signs and any laboratory changes that were
considered serious adverse events or other
significant adverse events.
Adverse Events: Number Observed
and Rate with Patient Identifications
Adverse Events: Number Observed and Rate with Patient Identifications
Treatment Group X N=50
Mild Moderate Severe Total Total
Related
*
NR Related NR Related NR Related NR R+NR
*
NR = not related; related could be expanded, e.g., as definite, probable,
possible
**
Patient identification number
Body
System
A
Event 1 6(12%) 2(4%) 3(6%) 1(2%) 3(6%) 1(2%) 12(24%
)
4(8%)
N11
**
N21 N31 N41 N51 N61
N12 N22 N32 N52
N13 N33 N53
N14
N15
N16
Event 2
12.2.2 Display of Adverse Events
• In addition to these complete tables provided in 14.3.1,
an additional summary table comparing treatment and
control groups, without the patient identifying
numbers limited to relatively common adverse events
(e.g., those in at least 1 % of the treated group), should
be provided in the body of the report.
• In presenting adverse events, it is important both to
display the original terms used by the investigator and
to attempt to group related events (i.e., events that
probably represent the same phenomena) so that the
true occurrence rate is not obscured. One way to do
this is with a standard adverse reaction/events
dictionary.
12.2.3 Analysis of Adverse Events
• The basic display of adverse event rates
described in(section 12.2.2 and located in
section 14.3.1) of the report, should be used
to compare rates in treatment and control
groups. For this analysis it may be helpful to
combine the event severity categories and the
causality categories, leading to a simpler side-
by-side comparison of treatment groups. I
12.2.3 Analysis of Adverse Events
• In addition, although this is usually best done in an
integrated analysis of safety, if study size and design
permit, it may be useful to examine the more common
adverse events that seem to be drug related for
relationship to dosage and to mg/kg or mg/m2dose, to
dose regimen, to duration of treatment, to total dose,
to demographic characteristics such as age, sex, race,
to other baseline features such as renal status, to
efficacy outcomes, and to drug concentration. It may
also be useful to examine time of onset and duration of
adverse events. A variety of additional analyses may be
suggested by the study results or by the pharmacology
of the test drug/investigational product.
12.2.4 Listing of Adverse Events by
Patient
• All adverse events for each patient, including
the same event on several occasions should
be listed in (appendix 16.2.7), giving both
preferred term and the original term used by
the investigator.
12.2.4 Listing of Adverse Events by
Patient
The listing should be by investigator and by treatment group and should include:
• Patient identifier
• Age, race, sex, weight (height, if relevant)
• Location of CRFs, if provided.
• The adverse event (preferred term, reported term)
• Duration of the adverse event
• Severity (e.g. mild, moderate, severe)
• Seriousness (serious/non-serious)
• Action taken (none, dose reduced, treatment stopped, specific treatment instituted, etc.)
• Outcome (e.g., CIOMS format)
• Causality assessment, (e.g., related/not related). How this was determined should be described in the
table or elsewhere.
• Date of onset or date of clinic visit at which the event was discovered
• Timing of onset of the adverse event in relation to last dose of test drug/investigational product (when
applicable)
• Study treatment at time of event or most recent study treatment taken
• Test drug/investigational product dose in absolute amount, mg/kg or mg/m2 at time of event
• Drug concentration (if known)
• Duration of test drug/investigational product treatment
• Concomitant treatment during study
12.3 Deaths, Other Serious Adverse
Events, and Other Significant Adverse
Events
• Deaths, other serious adverse events, and
other significant adverse events deserve
special attention.
12.3.1 Listing of Deaths, Other Serious
Adverse Events and Other Significant
Adverse Events
• Listings, containing the same information as
called for in (section 12.2.4) above, should be
provided for the following events.
12.3.1.1 Deaths
• All deaths during the study, including the post
treatment follow-up period, and deaths that
resulted from a process that began during the
study, should be listed by patient in (section
14.3.2).
12.3.1.2 Other Serious Adverse Events
• All serious adverse events (other than death
but including the serious adverse events
temporally associated with or preceding the
deaths) should be listed in (section 14.3.2) The
listing should include laboratory
abnormalities, abnormal vital signs and
abnormal physical observations that were
considered serious adverse events.
12.3.1.3 Other Significant Adverse
Events
• Marked hematological and other laboratory
abnormalities (other than those meeting the
definition of serious) and any events that led
to an intervention, including withdrawal of
test drug/investigational product treatment,
dose reduction, or significant additional
concomitant therapy, other than those
reported as serious adverse events, should be
listed in (section 14.3.2).
12.3.2 Narratives of Deaths, Other
Serious Adverse Events and Certain
Other Significant Adverse Events
• There should be brief narratives describing
each death, each other serious adverse event,
and those of the other significant adverse
events that are judged to be of special interest
because of clinical importance. These
narratives can be placed either in the text of
the report or in (section 14.3.3), depending on
their number.
12.3.2 Narratives of Deaths, Other
Serious Adverse Events and Certain
Other Significant Adverse Events
In addition, the following information should be included:
• Patient identifier
• Age and sex of patient; general clinical condition of patient,
if appropriate
• Disease being treated (if the same for all patients this is not
required) with duration (of current episode) of illness
• Relevant concomitant/previous illnesses with details of
occurrence/duration
• Relevant concomitant/previous medication with details of
dosage
• Test drug/investigational product administered, drug dose,
if this varied among patients, and length of time
administered
12.3.3 Analysis and Discussion of
Deaths, Other Serious Adverse Events
and Other Significant Adverse Events
• The significance of the deaths, other serious
adverse events and other significant adverse
events leading to withdrawal, dose reduction
or institution of concomitant therapy should
be assessed with respect to the safety of the
test drug/investigational product. Particular
attention should be paid to whether any of
these events may represent a previously
unsuspected important adverse effect of the
test drug/investigational product.
12.4 Clinical Laboratory Evaluation
12.4.1 Listing of Individual Laboratory
Measurements by Patient (16.2.8) and
Each Abnormal Laboratory Value (14.3.4)
• When required by regulatory authorities, the
results of all safety-related laboratory tests
should be available in tabular listings, using a
display similar to the following, where each
row represents a patient visit at which a
laboratory study was done, with patients
grouped by investigator (if more than one)
and treatment group, and columns include
critical demographic data, drug dose data, and
the results of the laboratory tests.
List Of Laboratory Measurements
List Of Laboratory Measurements
Laboratory Tests
Patien
t
Time Age Sex Race Weigh
t
Dose SGOT SGPT AP....
X
*
Vn = value of a particular test
#1 T0 70 M W 70kg 400mg V1
*
V5 V9
T1 V2 V6 V10
T2 V3 V4 V15
T3 V4 V8 V12
#2 T10 65 F B 50kg 300mg V13 V16 V19
T21 V14 V17 V20
T32 V15 V18 V21
12.4.2 Evaluation of Each Laboratory
Parameter
• The necessary evaluation of laboratory values
must in part be determined by the results
seen, but, in general, the following analyses
should be provided. For each analysis,
comparison of the treatment and control
groups should be carried out, as appropriate
and as compatible with study size. In addition,
normal laboratory ranges should be given for
each analysis.
12.4.2.1 Laboratory Values Over Time
• For each parameter at each time over the
course of the study (e.g., at each visit) the
following should be described: the group
mean or median values, the range of values,
and the number of patients with abnormal
values, or with abnormal values that are of a
certain size (e.g., twice the upper limit of
normal, 5 times the upper limit; choices
should be explained). Graphs may be used.
12.4.2.2 Individual Patient Changes
• An analysis of individual patient changes by treatment group should
be given. A variety of approaches may be used, including:
1. "Shift tables" - These tables show the number of patients who are
low, normal, or high at baseline and then at selected time intervals.
2. Tables showing the number or fraction of patients who had a change
in parameter of a predetermined size at selected time intervals. For
example, for BUN, it might be decided that a change of more than 10
mg/dL BUN should be noted.
3. A graph comparing the initial value and the on-treatment values of a
laboratory measurement for each patient
12.4.2.3 Individual Clinically
Significant Abnormalities
• Clinically significant changes (defined by the
applicant) should be discussed. A narrative of
each patient whose laboratory abnormality
was considered a serious adverse event and,
in certain cases, considered an other
significant adverse event, should be provided
under (sections 12.3.2 or 14.3.3).
12.5 Vital Signs, Physical Findings and
Other Observations Related to Safety
• Vital signs, other physical findings, and other
observations related to safety should be
analysed and presented in a way similar to
laboratory variables. If there is evidence of a
drug effect, any dose-response or drug
concentration-response relationship or
relationship to patient variables (e.g., disease,
demographics, concomitant therapy) should
be identified and the clinical relevance of the
observation described.
12.6 Safety Conclusions
• The overall safety evaluation of the test
drug(s)/investigational product(s) should be
reviewed, with particular attention to events
resulting in changes of dose or need for
concomitant medication, serious adverse
events, events resulting in withdrawal, and
deaths.
13. Discussion and Overall
Conclusions
• The efficacy and safety results of the study
and the relationship of risks and benefit
should be briefly summarised and discussed,
referring to the tables, figures, and sections
above as needed. The presentation should not
simply repeat the description of results nor
introduce new results.
14. Tables, Figures and Graphs
Referred to But Not Included in the
Text
• igures should be used to visually summarise the
important results, or to clarify results that are not
easily understood from tables.
• Important demographic, efficacy and safety data
should be presented in summary figures or tables
in the text of the report. However, if these
become obtrusive because of size or number they
should be presented here, cross-referenced to
the text, along with supportive, or additional,
figures, tables or listings.
The following information may be presented in this
section of the core clinical study report:
• 14.1 Demographic Data
• Summary figures and tables
• 14.2 Efficacy Data
• Summary figures and tables
• 14.3 Safety Data
• Summary figures and tables
• 14.3.1 Displays of Adverse Events
• 14.3.2 Listings of Deaths, Other Serious and Significant
Adverse Events
• 14.3.3 Narratives of Deaths, Other Serious and Certain
Other Significant Adverse Events
• 14.3.4 Abnormal Laboratory Value Listing (each patient)
15. Reference List
• A list of articles from the literature pertinent
to the evaluation of the study should be
provided. Copies of important publications
should be attached in an (appendix 16.1.11
and 16.1.12).
16. Appendices
• This section should be prefaced by a full list of
all appendices available for the study report.
Where permitted by the regulatory authority,
some of the following appendices need not be
submitted with the report but need to be
provided only on request.
• 16.1 Study Information
• 16.1.1 Protocol and protocol amendments
• 16.1.2 Sample case report form (unique pages only)
• 16.1.3 List of IECs or IRBs (plus the name of the committee Chair if
required by the regulatory authority) - Representative written
information for patient and sample consent forms
• 16.1.4 List and description of investigators and other important
participants in the study, including brief (1 page) CVs or equivalent
summaries of training and experience relevant to the performance of the
clinical study
• 16.1.5 Signatures of principal or coordinating investigator(s) or sponsor's
responsible medical officer, depending on the regulatory authority's
requirement.
• 16.1.6 Listing of patients receiving test drug(s)/investigational product(s)
from specific batches, where more than one batch was used.
• 16.1.7 Randomisation scheme and codes (patient
identification and treatment assigned)
• 16.1.8 Audit certificates (if available) (see Annex IVa and
IVb of the guidance)
• 16.1.9 Documentation of statistical methods
• 16.1.10 Documentation of inter-laboratory
standardisation methods and quality assurance
procedures if used.
• 16.1.11 Publications based on the study
• 16.1.12 Important publications referenced in the report
• 16.2 Patient Data Listings
• 16.2.2 Protocol deviations
• 16.2.3 Patients excluded from the efficacy analysis
• 16.2.4 Demographic data
• 16.2.5 Compliance and/or drug concentration data (if available)
• 16.2.6 Individual efficacy response data
• 16.2.7 Adverse event listings (each patient)
• 16.2.8 Listing of individual laboratory measurements by patient,
when required by regulatory authorities
• 16.3 Case Report Forms
• 16.3.1 CRFs for deaths, other serious adverse events and
withdrawals forAE
• 16.3.2 Other CRFs submitted
• 16.4 Individual Patient Data Listings (us Archival Listings)
Annex I: Synopsis
Annex II: Principal Or
Coordinating Investigator(s)
Signature(s) Or Sponsor's
Responsible Medical Officer
Annex IIIa: Study
Design And
Schedule Of
Assessments
Annex IIIb: Study
Design And
Schedule Of
Assessments
Annex IVa Disposition Of Patient
Annex IVb Disposition Of Patient
Annex V: Listing
Of Patients Who
Discontinued
Therapy
Annex VI: Listing Of
Patients And
Observations
Excluded From
Efficacy Analysis
Annex VII: Number Of
Patients Excluded
From Efficacy Analysis
Annex VIII: Guidance For Section
11.4.2 - Statistical/analytical Issues
And Appendix 16.1.9
• Details of the statistical analysis performed on
each primary efficacy variable should be
presented in an appendix.
• A. STATISTICAL CONSIDERATIONS
• B. Format And Specifications For Submission
Of Data Requested By Regulatory Authority's
Statistical Reviewers
Details reported should include at
least the following information:
a) The statistical model underlying the analysis. This should be presented precisely and completely, using
references if necessary.
b) A statement of the clinical claim tested in precise statistical terms, e.g., in terms of null and alternative
hypotheses.
c) The statistical methods applied to estimate effects, construct confidence intervals, etc. Literature
references should be included where appropriate.
d) The assumptions underlying the statistical methods. It should be shown, insofar as statistically
reasonable, that the data satisfy crucial assumptions, especially when necessary to confirm the validity
of an inference.
e) The test statistic, the sampling distribution of the test statistic under the null hypothesis, the value of
the test statistic, significance level (i.e. p-value), and intermediate summary data, in a format that
enables the regulatory authority's statistical reviewer to verify the results of the analysis quickly and
easily. The p-values should be designated as one- or two-tailed. The rationale for using a one-tailed
test should be provided.
Overview of current US regulations
regarding clinical monitoring and
safety reporting
Objectives
• Current context regarding safety in clinical trials
• The concept of safety and safety monitoring and how
it relates to clinical trials research
• Protocol requirements pertaining to areas relevant to
safety
• Key roles and responsibilities related to safety
• Safety and adverse event terminology
• Expedited reporting of adverse events
109
Objectives
• Ensuring safety in clinical trials
• The adverse event life cycle
• What makes a well-documented adverse event,
including a comprehensive narrative
• How to assess an adverse event case, including
causality assessment
• Periodic Safety Report: DSUR, PSUR, PBRER, ASR, IB,
Orphan Designation Annual Report
110
Regulations: Federally Supported Research Involving Human
Subjects
111
45 CFR 46: Protection of Human Research Subjects
• Applies to all research involving human subjects
• Institution must provide assurance of compliance, such as a Federal Wide Assurance
(FWA) on file with the Office for Human Research Protection (OHRP)
• FWA provides assurance that research is conducted in accordance with the regulations
 Research reviewed and approved by IRB
 Subject to continuing review by IRB
Regulations: Non-Federally Supported Studies Involving
Human Subjects
112
21 CFR 50: Protection of Human Subjects
• Clinical investigations regulated by FDA
• Requirements for informed consent
• Elements of informed consent
• Documentation of informed consent
• Form approved by IRB
21 CFR 56: Institutional Review Boards
• Clinical investigations regulated by FDA
• Requirements for IRB review
• Membership, functions, review procedures, etc
• Criteria for IRB approval
Current Safety Environment
113
 Increasing demands for safety data:
• All Serious Adverse Events (SAEs); Follow all AEs/SAEs till resolved
or stable
• Additional adverse events of interest (e.g. cancers, MIs, hepatic
events)
• Pregnancy outcomes
• Food and Drug Administration Amendments Act of 2007 (FDAAA):
Provides FDA with additional requirements, authorities, and resources
with regard to both pre- and postmarket drug safety
• Global reporting to EMEA and regulatory agencies of European
Union (EU) member states
– Use of CIOMS form
– Country of origin of AE
Clinical Trial Continuum: From Drug Development to
Optimal Regimens to Treatment Strategies
114
PHASE I PHASE II PHASE III PHASE IV POST PHASE III/IV
PRE-MARKET POSTMARKET
CONTROLLED SETTING REAL-WORLD SETTING
Safety Monitoring
115
Why is safety monitoring
required in all clinical trials?
To Ensure Subject Safety and
Study Integrity
Investigator assures
Subject Safety and
Study Integrity by:
116
Implementing the protocol “as written”
Strict adherence to inclusion
and exclusion criteria
Monitoring subject status, i.e. subject wellbeing,
minimization of risk, toxicity management, etc.
Continued adherence
throughout study duration
Monitoring safety data collection:
• Study database
• Safety database
Roles and Responsibilities – Site Investigator
Roles and Responsibilities – Research Staff
117
• Record AE and/or SAE per
protocol specifications
• Follow protocol toxicity
management section
• Follow site SOP for
emergencies
• Follow site SOP to notify
study clinician/physician
• Record the AE/SAE
Is immediate/emergency intervention needed?
Yes No
Roles and Responsibilities –
Study Clinician/Physician
118
Subject reports AE
Documentation
•Follow until AE resolution
or condition stabilizes
Study clinician/physician
will assess and manage
the AE Decide if SAE
Emergency intervention vs.
Non-emergency care
Research provisions
vs.
Clinical care
Assurance of Safety and Well-Being:
Research vs. Medical Roles
119
 Emergency intervention vs. Non-emergency care
• Acute on-site management, as necessary, and per site SOP
• Referral to care when stable
 Research provisions vs. Clinical care
• Provide interventions permitted by the protocol
• Follow protocol specifications for toxicity management
• Beyond protocol specifications, refer out for clinical care
Clinical Role vs. Research Role
120
Balancing Both Roles
Balancing Both Roles
Clinical Role: Subject OK Research Role: Study/Data OK
• Is subject in imminent jeopardy?
• Provide appropriate management
commensurate with clinical situation,
e.g. toxicity management
• Provide appropriate referral: emergent
care or back to regular care
• Follow up with subject status
Not Subject’s Primary Clinician
• Identification of adverse event
• Immediate notification necessary? To
whom? [per protocol and safety monitoring
plans]
• Complete documentation of adverse event.
Follow until resolution/stability including
updating records
• Determine if AE meets criteria for SAE
• Adhere to reporting requirements
• Adhere to toxicity management as specified
• Adhere to stopping rules as specified
Therapeutic Misconception
121
• Subjects think they are receiving proven interventions, per
their usual clinical care, despite participating in a research
study
• Informed Consent Process must not be trivialized or relegated to administrative
status
• Check for understanding
• Time for questions, making decision
• Physicians think they can provide interventions, per usual
practice
• Strict adherence to protocol provisions for care, toxicity management
• Decide if subject can continue in study
Roles and Responsibilities –
Study Clinician/Physician
122
Study product:
Per site, per study?
Study status: Safety
pause, clinical hold, early
termination?
Study product:
Dose held, changed,
or discontinued?
Study participation:
Continue, withdraw?
Action taken with Study product
after AE
Study Study
Roles and Responsibilities –
Study Team
123
Safety: Ensure safety and well being of subjects at all times
• Monitor safety across all study sites
• Review all safety data at specified intervals
• Discuss need for change(s) driven by safety
Data: Ensure data integrity to assess the risks/safety profile of the study
intervention
• Data capture; especially safety data
• Be cognizant of expedited reporting requirements for safety data
Roles and Responsibilities –
Study Team vs. Sponsor/sponsor
124
• Safety monitoring by study team
• Acute on-site management and discussion with study team
• Periodic review by study team and monitoring committees
• Data generated by Data Management Centers (DMC)
• Expedited reporting to sponsor
• SAE sent to sponsor
• Sponsor is not part of discussions that occur within study/safety monitoring teams
regarding the event
• The sponsor only has information about the event from the SAE Form; site should
include relevant information from study team discussions
• sponsor processes event and sends queries to site to obtain additional information
• All follow-up information should be provided to sponsor
Safety Monitoring Environment
125
CTA/IND Trials
Pre-market
Postmarket
45 CFR 46 45 CFR 46
FDA
21 CFR Part 312 - IND
21 CFR 312.32 (IND Safety Reports)
21 CFR 312.33 (Annual Reports)
21 CFR 812.150 (IDE Reports)
21 CFR Part 314 - NDA
21 CFR 314.80 (Postmarketing)
21 CFR 314.98 (Generics)
21 CFR 600.80 (Biologics)
21 CFR 803 (Medical Devices)
ICH E2A (Oct 1994) ICH E2D (Nov 2003)
NIH Policy NIH Policy
Country/State Regulations Country/State Regulations
IRBs/ECs IRBs/ECs
Sponsor Sponsor
ICH: E Documents on Safety
Clinical Safety
• ICH E1 – The Extent of Population Exposure to Assess Clinical Safety for Drugs Intended for Long-Term
Treatment of Non-Life Threatening Conditions
• ICH E2A – Clinical Safety Data Management: Definitions and Standards for Expedited Reporting
• ICH E2B – Clinical Safety Data Management: Data Elements for Transmission of Individual Case Safety
Reports
• ICH E2C – Clinical Safety Data Management: Periodic Safety Update Reports for Marketed Drugs
• ICH E2D – Post-Approval Safety Data Management: Definitions and Standards for Expedited Reporting
• ICH E2E – Pharmacovigilance Planning
• ICH E2F – Development Safety Update Report
Good Clinical Practice
• ICH E6 – Good Clinical Practice
126
http://www.ich.org/cache/compo/276-254-1.html
Subject Enrolled
AE Reported
Record AE*
Yes
SAE?
To Sponsor
Record
SAE**
To IRB
To FDA
To other
Follow until
Resolution
or Stability
Outcome:
Resolved/
Stable?
Update SAE
No
Adverse Event Flowchart
127
Subject Enrolled
AE Reported
Record AE*
Yes
SAE?
To Sponsor
Record
SAE**
To IRB
To FDA
To other
Follow until
Resolution
or Stability
Outcome:
Resolved/
Stable?
Update SAE
No
Adverse Event
128
* Protocol specifications for AE
• When to collect e.g., study visit
• Method of collection e.g., in person, telephone call
• What to collect e.g., all AEs, only certain AEs by body system, only certain AEs by
severity
• What forms to use e.g. AE CRF, study CRFs
** Protocol specifications for SAE
• Criteria
• Expedited time frames
• Reporting form (e.g. SAE)
Documentation Differences Between AE CRF and SAE Form
Record on SAE Form
(includes narrative)
129
Record in source
document
Record on AE case
report form
Yes
Attach additional
documentation
Does AE
meet SAE
criteria?
Documentation Differences Between AE CRF and SAE Form:
Data Elements
AE CRF
Data Elements
• AE
• Start Date
• Stop Date / Continuing
• Is it SAE?
• Severity
• Relatedness
• Action taken with Study Agent
• Outcome (study participation)
130
SAE Form
Data Elements
• Participant Identifiers
• Study Agent details
• Narrative
• Past medical history
• Relevant labs, tests, procedures
• Concomitant meds
• Outcome of SAE
• Other supporting information
Safety Data from Clinical Trials
Obligations to report safety data (IND or Non-IND studies):
• Data for non-expedited reporting:
• Recorded on AE CRF, goes to clinical trial database
• Data for expedited reporting:
• Recorded on AE CRF and linked to an SAE type form
• Goes to safety database
• IND timelines: 24o to sponsor, 7/15 days to FDA, EMEA
• Post-marketing timelines: depends on sponsor, 15 days to FDA, EMEA
• Annual/Periodic Reports :
• Need safety data from clinical and safety database
• Must be reconciled
131
Adverse Event
Any untoward medical occurrence in a patient or
clinical investigation subject administered a
pharmaceutical product and which does not
necessarily have to have a causal relationship
with this treatment.
(ICH E2A)
132
Adverse Event Term
• The AE should best describe what the subject says (i.e.
verbatim description)
• Can be extracted from medical records
• Can incorporate medical assessment (including a diagnosis if
available)
• The more accurate the AE term, the more accurate the safety
database. The AE terms will be coded using a standard
dictionary, e.g. Medical Dictionary for Regulatory Activities
(MedDRA); this is NOT site responsibility
133
AE Term - Examples
134
• If “anaphylactic reaction” is associated with “rash,
dyspnea, hypotension, and laryngospasm,” report
primary AE as “anaphylactic reaction.”
• If “myocardial infarction” is associated with “chest
pain, dyspnea, diaphoresis, ECG changes and
jaundice,” report “myocardial infarction” and
“jaundice” as separate primary AEs.
Serious Adverse Event (SAE)
A serious adverse event (experience) or reaction is any untoward
medical occurrence that at any dose:
• Results in death,
• Is life-threatening
• Requires inpatient hospitalization or prolongation of existing hospitalization
• Results in persistent or significant disability/incapacity
• Is a congenital anomaly/birth defect
• In addition, “…important medical events that may not be immediately life-
threatening or result in death or hospitalization but may jeopardize the patient or
may require intervention to prevent one of the other outcomes listed in the
definition above…should also usually be considered serious.”
(ICH E2A)
135
Adverse Event vs. Event Outcome
Hospitalization
• Hospitalization is a consequence and is not usually considered
an AE.
• Example: If the subject was hospitalized due to congestive heart failure,
“congestive heart failure” is the primary AE and hospitalization is the
outcome.
• If the only information available is that the study subject was
hospitalized, “hospitalization” can be reported.
136
Hospitalization
Hospitalization in the absence of a medical AE is not in
itself an AE and does not need to be reported in an
expedited time frame, such as:
• Admission for treatment of a pre-existing condition (can include target
disease) not associated with the development of a new AE or with a
worsening of the pre-existing condition
• Diagnostic admission (e.g. for work-up of persistent existing condition
such as pre-treatment lab abnormality)
• Protocol-specified admission (e.g. procedure required by study protocol)
• Administrative admission (e.g. for yearly physical exam)
• Social admission (e.g. study subject has no place to sleep)
• Elective admission (e.g. elective surgery)
137
Severity
• Describes the intensity of the event
• Events are graded on a severity scale
• Mild, Moderate, Severe
• Numeric Scale e.g. 1 to 5
• Severity grading must match the clinical picture
• Presenting AE is Grade 1
• AE progressed to SAE (hospitalization)
• The expedited report should have the grade of the
SAE, not the AE
138
Seriousness is NOT the same as Severity
139
Based on the intensity of the AE
and is not a factor in
determining reportability
(clinical descriptor)
Based on outcome of the AE
and is a factor in determining
reportability (regulatory
definition)
Seriousness Severity
Determined using the SAE
criteria
Determined using the sponsor
grading table
Action Taken with Drug
• Action Taken with Drug:
• Withdrawn
• Dose reduced
• Dose increased
• Dose not changed
• Unknown
• Not applicable > ICH E2B (R3)
• Refer to protocol
• Refer to DAERS
140
Outcome
• Outcome of reaction/event at the time of last observation
• Recovered/resolved
• Recovering/resolving
• Not recovered/not resolved
• Recovered/resolved with sequelae
• Fatal
• Unknown > ICH E2B (R3)
• Outcome of subject in study
• Remains in Study
• Withdrawn
• Lost to follow-up
• Death
141
Expectedness
• Pertains to whether an event is expected or
unexpected (on the basis of previous observation,
not what might be anticipated from the
pharmacological properties of the product)
• Unexpected: the nature or severity of the adverse
event is not consistent with the applicable
product information (e.g. Investigator’s Brochure
for unapproved product, Package Insert for
approved product)
142
Relatedness (Causality)
• No standard international nomenclature
• Conveys that a “causal relationship” between the study
product and the adverse event is “at least a reasonable
possibility” [ICH E2A]
• Facts (evidence) exist to suggest the relationship
• Information on SAEs generally incomplete when first received
• Follow-up information actively pursued
• Judged by:
• Reporting health professional
• Sponsor
143
Determination of Causality
• Standard determinations include:
• Is there [Drug Exposure] and [Temporal
Association]?
• Is there [Dechallenge/Rechallenge] or [Dose
Adjustments]?
• Any known association per [Investigator’s
Brochure] or [Package Insert]?
• Is there [Biological Plausibility]?
• Any other possible [Etiology]?
144
• ‘May be more art than science’
• NR: evidence for alternate etiology and/or low or no biologic plausibility
• R: reasonable possibility; facts or evidence to substantiate relationship,
and biologic plausibility
• Is there evidence to compel change in previous conclusions?
• Clear-cut case; easy to make a determination
• Not so clear-cut: use your best judgment based on available
information; assure adequacy of information
• Unless clear-cut case, there’s no absolute right or wrong; give
your best judgment; substantiate and follow-up
• Err on conservative side
145
Determination of Causality
146
• Comprehensive, stand-alone “medical story”
• Written in logical time sequence
• Include key information from supplementary records
• Include relevant autopsy or post-mortem findings
• Summarize all relevant clinical and related
information, including:
• Study subject characteristics
• Therapy details
• Medical history
• Clinical course of the event(s)
• Diagnosis (workup, relevant tests/procedures, lab results)
• Other information that supports or refutes an AE
• > ICH E2D
Narrative
Narrative Template
147
• This is a [Age] year old [Race] [Male/Female] in [Study] who reported
[Primary AE] on [Date of AE]. Enrolled into study on [Date Enrolled], Study
medication was started on [Date], which is [Study Day _/Week _], taken
for [Duration]. The event occurred during the [Treatment/Follow-up
Phase].
• If fetus: provide [Gestational Age], (or mother’s LMP), at time of event.
Also, [Gestational Age/Trimester] at first drug exposure and duration of
exposure. If birth, provide details of [Infant Status] at birth. If hospital stay
is complicated, provide details of hospital stay.
• Provide details of the [AE] in chronological order, along with other
[Signs/Symptoms]. Provide details of [Physical Exam], along with all
relevant [Procedures] and [Lab Results].
Narrative Template
148
• Provide details of [Treatment] and [Treatment Rationale] on basis of
[Findings/Test Result(s)]. Describe [Treatment Response].
• If hospitalization, provide [Dates Hospitalization], describe relevant [Hospital
Course], [Diagnostic Work-up], [Procedures/Tests and Results], [Treatment],
[Treatment Response].
• Provide [Discharge Diagnosis], and any [Follow-up Information]. List [Discharge
Meds].
• Provide pertinent [Past Medical Hx], [Family Hx], [Concomitant Meds],
[Alcohol/Tobacco/Substance Use] and any previous similar [AEs].
Review and Assessment of SAE
149
• Assemble all information available and use medical judgment
• Standard for each AE:
• Select [Seriousness Criteria]
• Grade [Severity] per sponsor Toxicity Table
• Specify [Actions Taken on Study Product]
• Specify [Outcome of SAE]. If Outcome is not resolved at time of evaluation, follow
until resolution or stability at each study visit
• Is it [Expected]?
• Is it [Related]?
150
• Sponsor role: (ICH E2D)
• Information about the case should be collected from the
healthcare professionals who are directly involved in the
study subject’s care
• Clearly identified evaluations by the sponsor are considered
appropriate and are required by some regulatory authorities
• Opportunity to render another opinion; may be in
disagreement with; and/or provide another alternative to the
diagnosis/assessment given by initial reporter
• Sponsor makes an assessment of causality (attribution) just
as the PI makes an assessment of causality (attribution)
• If causality (attribution) is different between the sponsor and
the investigator, both assessments are reported
Clinical Case Evaluation
Site vs. Sponsor Assessment
151
Balancing Both RolesSite Assessment Sponsor Assessment
• Site advantage: has access to
subject; may elicit further info,
perform PE, obtain tests, labs,
records
• Information from self-report
(may lack validation)
• Know subject best
• Judgment stands
• Open to dialog with sponsor
• Information limited to what was submitted from
site
• May initiate queries to site: incur time and
delay
• Constraint: Must adhere to reporting timelines
to FDA
• Serious? Unexpected? Related?
• sign-off, agree with Site PI, agree with sponsor.
Any critical flaw in reasoning?
• Open to dialog with Site PI, sponsor
Periodic Safety Report: DSUR, PBRER
(replacing PSUR), ASR, IB, Orphan
Designation Annual Report
• During the drug development and after the drug is on
the market, the sponsor or market authorization holder
has obligations to submit the safety information to
regulatory agencies periodically.
• These periodic reports have different requirements and
sometimes are confusing. While these reports may be
prepared by the regulatory affairs department or
pharmacovigilence department, biostatistics group
may often be asked to provide the information for
these periodic reports.
Periodic Safety Report: DSUR, PBRER
(replacing PSUR), ASR, IB, Orphan
Designation Annual Report
• These reports and their abbreviations could
be very confusing especially for those who are
not working in the pharmacovigilence
department: DSUR, IND Annual Report, ASR,
IB, PSUR, and PBRER
Types of Safety Reports
If we take a close look at these reports, they may be
considered as four types:
• PSUR (Old report replaced by PBRER)
• PBRER (replacing PSUR) for drugs already on the market
• DSUR (replacing IND Annual Report and Annual Safety
Report) for drugs under developments
• IB is required whenever there is a clinical trial
• Orphan Designation Annual Report is required for the
developing product with orphan designation - for rare
disease
PSUR: Periodic Safety Update Reports
• The term PSUR comes from the previous ICH
E2C (R1) “Clinical Safety Data Management:
Periodic Safety Update Reports for Marketed
Drugs”. ICH guideline E2C has been
subsequently revised and renames as Periodic
Benefit-Risk Evaluation Report (PBRER).
PBRER: Periodic Benefit-Risk
Evaluation Report
• According to the ICH E2C (R2) “PERIODIC
BENEFIT-RISK EVALUATION REPORT (PBRER)”,
• The Periodic Benefit-Risk Evaluation Report
(PBRER) described in this Guideline is
intended to be a common standard for
periodic benefit-risk evaluation reporting on
marketed products (including approved drugs
that are under further study) among the ICH
regions.
PBRER: Periodic Benefit-Risk
Evaluation Report
• Other supporting documents regarding PBRER
are:
• Concept Paper
• Business Plan
• Contribute to the E2C(R2) IWG
PBRER: Periodic Benefit-Risk
Evaluation Report
• E2C (R2) is finalized at November 2012 and is
supposed to replace the PSUR. However, EMA
has not fully adopted the PBRER and
continues to use the term PSUR as defined in
its recent guideline “Guideline on good
pharmacovigilance practices (GVP) 4 Module
VII – Periodic safety update report (Rev 1)”
PBRER: Periodic Benefit-Risk
Evaluation Report
• FDA fully endorsed E2C (R2) and PBRER and
issued its guidance “Providing Postmarket
Periodic Safety Reports in the ICH E2C(R2)
Format (Periodic Benefit-Risk Evaluation
Report”
DSUR: Development Safety Update
Report
• According to ICH E2F “Development Safety
Update Report”,
• The Development Safety Update Report
(DSUR) proposed in this guideline is intended
to be a common standard for periodic
reporting on drugs under development
(including marketed drugs that are under
further study) among the ICH regions.
• US and EU regulators consider that the DSUR,
submitted annually, would meet national and
regional requirements currently met by the US
IND Annual Report and the EU Annual Safety
Report, respectively, and can therefore take the
place of these existing reports
• ICH E2F Guideline is finalized in August 2010 and
is replacing the previous IND (investigational new
drug) Annual Report (in US) and Annual Safety
Report (in EU).
Documents regarding DSUR can be
found at ICH.org website
We will cover the following:
• Concept Paper
• Business Plan
• DSUR Examples Commercial Sponsors
• DSUR Examples non-Commercial Sponsors
• Presentation on E2F
IB:Investigator Brochure
• According to ICH E6 “Good Clinical Practice”,
• The Investigator's Brochure (IB) is a
compilation of the clinical and nonclinical data
on the investigational product(s) that are
relevant to the study of the product(s) in
human subjects.
IB:Investigator Brochure
• Its purpose is to provide the investigators and
others involved in the trial with the
information to facilitate their understanding of
the rationale for, and their compliance with,
many key features of the protocol, such as the
dose, dose frequency/interval, methods of
administration, and safety monitoring
procedures. The IB also provides insight to
support the clinical management of the study
subjects during the course of the clinical trial.
IB:Investigator Brochure
• For post marketing commitment clinical trials,
the product label (package insert) may be
used in place of the investigator brochure
since the package insert includes the contents
required in the investigator brochure.
Orphan Drug Designation Annual
Report
• 21CFR Part 316 (Orphan Drugs) contains the
specific section (section 316.30) requiring the
annual reports of holder of orphan drug
designation:
• § 316.30 Annual reports of holder of orphan-
drug designation.
Orphan Drug Designation Annual
Report
• Within 14 months after the date on which a
drug was designated as an orphan drug and
annually thereafter until marketing approval,
the sponsor of a designated drug shall submit
a brief progress report to the FDA Office of
Orphan Products Development on the drug.
Orphan Drug Designation Annual
Report includes:
• (a) A short account of the progress of drug development
including a review of preclinical and clinical studies
initiated, ongoing, and completed and a short summary of
the status or results of such studies.
(b) A description of the investigational plan for the coming
year, as well as any anticipated difficulties in development,
testing, and marketing; and
(c) A brief discussion of any changes that may affect the
orphan-drug status of the product. For example, for
products nearing the end of the approval process, sponsors
should discuss any disparity between the probable
marketing indication and the designated indication as
related to the need for an amendment to the orphan-drug
designation pursuant to § 316.26.
• EU has the similar requirement and sponsors
are required to submit to the European
Medicines Agency (EMA) every year after their
medicine has been granted orphan
designation. See EMA Orphan Designation
Annual Report.

Weitere ähnliche Inhalte

Was ist angesagt?

Cinical trial protocol writing
Cinical trial protocol writingCinical trial protocol writing
Cinical trial protocol writingUrmila Aswar
 
Clinical study and gcp
Clinical study and gcpClinical study and gcp
Clinical study and gcpGaurav Kr
 
Clinical Trial Protocol Review for Study Feasibility Analysis
Clinical Trial Protocol Review for Study Feasibility AnalysisClinical Trial Protocol Review for Study Feasibility Analysis
Clinical Trial Protocol Review for Study Feasibility AnalysisYing Lu
 
Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........Jobin Kunjumon Vilapurathuu
 
Clinical trial design
Clinical trial designClinical trial design
Clinical trial designSandhya Talla
 
Clinical research basic things
Clinical research basic thingsClinical research basic things
Clinical research basic thingsSRIJIL SREEDHARAN
 
Ich guidelines e9 to e12
Ich guidelines e9 to e12Ich guidelines e9 to e12
Ich guidelines e9 to e12Khushboo Bhatia
 
1-Halt-it Protocol International v1.0 26nov12
1-Halt-it Protocol International v1.0 26nov121-Halt-it Protocol International v1.0 26nov12
1-Halt-it Protocol International v1.0 26nov12HALT-IT Trial
 
R&D GCP and Effective Site Management 011910
R&D GCP and Effective Site Management 011910R&D GCP and Effective Site Management 011910
R&D GCP and Effective Site Management 011910Myron Pyzyk
 
clinical trials types and design
clinical trials types and designclinical trials types and design
clinical trials types and designUttara Joshi
 
Protocol Design & Development: What You Need to Know to Ensure a Successful S...
Protocol Design & Development: What You Need to Know to Ensure a Successful S...Protocol Design & Development: What You Need to Know to Ensure a Successful S...
Protocol Design & Development: What You Need to Know to Ensure a Successful S...Brook White, PMP
 
Introduction to clinical research
Introduction to clinical researchIntroduction to clinical research
Introduction to clinical researchPradeep H
 
Understanding Clinical Trials
Understanding Clinical TrialsUnderstanding Clinical Trials
Understanding Clinical TrialsBartsMSBlog
 
Protocol development
Protocol developmentProtocol development
Protocol developmentAlisha Bansal
 
Introduction to clinical trials
Introduction to clinical trialsIntroduction to clinical trials
Introduction to clinical trialsShriyaDeshpande5
 

Was ist angesagt? (20)

Cinical trial protocol writing
Cinical trial protocol writingCinical trial protocol writing
Cinical trial protocol writing
 
Clinical study and gcp
Clinical study and gcpClinical study and gcp
Clinical study and gcp
 
Pr. Peivand Pirouzi - Lung or Lung and Heart Transplants - Clinical trial pro...
Pr. Peivand Pirouzi - Lung or Lung and Heart Transplants - Clinical trial pro...Pr. Peivand Pirouzi - Lung or Lung and Heart Transplants - Clinical trial pro...
Pr. Peivand Pirouzi - Lung or Lung and Heart Transplants - Clinical trial pro...
 
Clinical Trial Protocol Review for Study Feasibility Analysis
Clinical Trial Protocol Review for Study Feasibility AnalysisClinical Trial Protocol Review for Study Feasibility Analysis
Clinical Trial Protocol Review for Study Feasibility Analysis
 
Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........
 
Clinical trial design
Clinical trial designClinical trial design
Clinical trial design
 
Clinical research protocol
Clinical research protocolClinical research protocol
Clinical research protocol
 
Clinical research basic things
Clinical research basic thingsClinical research basic things
Clinical research basic things
 
Ich guidelines e9 to e12
Ich guidelines e9 to e12Ich guidelines e9 to e12
Ich guidelines e9 to e12
 
Clincial trials and types
Clincial trials and typesClincial trials and types
Clincial trials and types
 
1-Halt-it Protocol International v1.0 26nov12
1-Halt-it Protocol International v1.0 26nov121-Halt-it Protocol International v1.0 26nov12
1-Halt-it Protocol International v1.0 26nov12
 
R&D GCP and Effective Site Management 011910
R&D GCP and Effective Site Management 011910R&D GCP and Effective Site Management 011910
R&D GCP and Effective Site Management 011910
 
clinical trials types and design
clinical trials types and designclinical trials types and design
clinical trials types and design
 
Components of a clinical study protocol
Components of a clinical study protocolComponents of a clinical study protocol
Components of a clinical study protocol
 
Protocol Design & Development: What You Need to Know to Ensure a Successful S...
Protocol Design & Development: What You Need to Know to Ensure a Successful S...Protocol Design & Development: What You Need to Know to Ensure a Successful S...
Protocol Design & Development: What You Need to Know to Ensure a Successful S...
 
Introduction to clinical research
Introduction to clinical researchIntroduction to clinical research
Introduction to clinical research
 
Understanding Clinical Trials
Understanding Clinical TrialsUnderstanding Clinical Trials
Understanding Clinical Trials
 
Protocol development
Protocol developmentProtocol development
Protocol development
 
Introduction to clinical trials
Introduction to clinical trialsIntroduction to clinical trials
Introduction to clinical trials
 
Designs of clinical trials
Designs of clinical trialsDesigns of clinical trials
Designs of clinical trials
 

Andere mochten auch

Eu module VII: PSUR released for public consultation
Eu module VII: PSUR released for public consultationEu module VII: PSUR released for public consultation
Eu module VII: PSUR released for public consultationSaravanan Subramanian (SAS)
 
Market Research Report : Clinical trials market in india 2014 - Sample
Market Research Report : Clinical trials market in india 2014 - SampleMarket Research Report : Clinical trials market in india 2014 - Sample
Market Research Report : Clinical trials market in india 2014 - SampleNetscribes, Inc.
 
Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...
Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...
Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...Epochresearch
 
Introduction to clinical sas programming
Introduction to clinical sas programmingIntroduction to clinical sas programming
Introduction to clinical sas programmingray4hz
 
Case Report Form (CRF) Design Tips
Case Report Form (CRF) Design TipsCase Report Form (CRF) Design Tips
Case Report Form (CRF) Design TipsPerficient
 
Periodic safety update reports – gvp guidelines and changes
Periodic safety update reports – gvp guidelines and changesPeriodic safety update reports – gvp guidelines and changes
Periodic safety update reports – gvp guidelines and changesTuracoz Healthcare Solutions
 

Andere mochten auch (11)

Eu module VII: PSUR released for public consultation
Eu module VII: PSUR released for public consultationEu module VII: PSUR released for public consultation
Eu module VII: PSUR released for public consultation
 
Market Research Report : Clinical trials market in india 2014 - Sample
Market Research Report : Clinical trials market in india 2014 - SampleMarket Research Report : Clinical trials market in india 2014 - Sample
Market Research Report : Clinical trials market in india 2014 - Sample
 
Drug Safety and Pharmacovigilance - Need for internationally harmonized pape...
Drug Safety and Pharmacovigilance  - Need for internationally harmonized pape...Drug Safety and Pharmacovigilance  - Need for internationally harmonized pape...
Drug Safety and Pharmacovigilance - Need for internationally harmonized pape...
 
Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...
Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...
Clinical SAS Programming | SAS Training | Big Data | Hadoop | Business Analyt...
 
The essential changes in the new pbrer format
The essential changes in the new pbrer formatThe essential changes in the new pbrer format
The essential changes in the new pbrer format
 
Introduction to clinical sas programming
Introduction to clinical sas programmingIntroduction to clinical sas programming
Introduction to clinical sas programming
 
PSUR Requirements
PSUR RequirementsPSUR Requirements
PSUR Requirements
 
Data management & statistics in clinical trials
Data management & statistics in clinical trialsData management & statistics in clinical trials
Data management & statistics in clinical trials
 
Case Report Form (CRF) Design Tips
Case Report Form (CRF) Design TipsCase Report Form (CRF) Design Tips
Case Report Form (CRF) Design Tips
 
Periodic safety update reports – gvp guidelines and changes
Periodic safety update reports – gvp guidelines and changesPeriodic safety update reports – gvp guidelines and changes
Periodic safety update reports – gvp guidelines and changes
 
Slideshare ppt
Slideshare pptSlideshare ppt
Slideshare ppt
 

Ähnlich wie Professor Peivand Pirouzi - International clinical study and safety reporting activities - Publication, Canada - All rights reserved

clinial trail documentation ppt M pharm pharmacology
clinial trail documentation ppt M pharm pharmacologyclinial trail documentation ppt M pharm pharmacology
clinial trail documentation ppt M pharm pharmacologyMansi Nikhade
 
4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.Audumbar Mali
 
Designing of clinical study documentation -protocol and crf
Designing of clinical study documentation -protocol and crfDesigning of clinical study documentation -protocol and crf
Designing of clinical study documentation -protocol and crfRumana Hameed
 
CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...
CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...
CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...amitsoni240
 
NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...
NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...
NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...CGC, LANDRAN
 
Designing of clinical study protocol rumana hameed
Designing of clinical study protocol rumana hameedDesigning of clinical study protocol rumana hameed
Designing of clinical study protocol rumana hameedRumana Hameed
 
planning and initiation.pptx
planning and initiation.pptxplanning and initiation.pptx
planning and initiation.pptxseeja2
 
Clinical Trial Protocol.pptx
Clinical Trial Protocol.pptxClinical Trial Protocol.pptx
Clinical Trial Protocol.pptxVenugopal N
 
Protocol Understanding_Katalyst HLS
Protocol Understanding_Katalyst HLSProtocol Understanding_Katalyst HLS
Protocol Understanding_Katalyst HLSKatalyst HLS
 
Protocol Understanding_ Clinical Data Management_KatalystHLS
Protocol Understanding_ Clinical Data Management_KatalystHLSProtocol Understanding_ Clinical Data Management_KatalystHLS
Protocol Understanding_ Clinical Data Management_KatalystHLSKatalyst HLS
 
DEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptx
DEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptxDEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptx
DEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptx36PranavLendhey
 
Guidelines for the preparation of protocol and documents in clnical trials
Guidelines for the preparation of protocol and documents in clnical trialsGuidelines for the preparation of protocol and documents in clnical trials
Guidelines for the preparation of protocol and documents in clnical trialsSachin Kumar
 
Clinical Trial Protocol
Clinical Trial ProtocolClinical Trial Protocol
Clinical Trial ProtocolClinosolIndia
 
Steps For Designing And Development Of Protocol
Steps For Designing And Development Of ProtocolSteps For Designing And Development Of Protocol
Steps For Designing And Development Of Protocolpviral24
 

Ähnlich wie Professor Peivand Pirouzi - International clinical study and safety reporting activities - Publication, Canada - All rights reserved (20)

DEVELOPING THE CLINICAL TRIAL PROTOCOL | PPT
DEVELOPING THE CLINICAL TRIAL PROTOCOL | PPTDEVELOPING THE CLINICAL TRIAL PROTOCOL | PPT
DEVELOPING THE CLINICAL TRIAL PROTOCOL | PPT
 
PROTOCOL
PROTOCOLPROTOCOL
PROTOCOL
 
clinial trail documentation ppt M pharm pharmacology
clinial trail documentation ppt M pharm pharmacologyclinial trail documentation ppt M pharm pharmacology
clinial trail documentation ppt M pharm pharmacology
 
4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.4. Unit-IV:- Clinical Trials.
4. Unit-IV:- Clinical Trials.
 
Protocol
ProtocolProtocol
Protocol
 
Designing of clinical study documentation -protocol and crf
Designing of clinical study documentation -protocol and crfDesigning of clinical study documentation -protocol and crf
Designing of clinical study documentation -protocol and crf
 
CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...
CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...
CLINICAL TRAIL (TRIAL PROTOCOL & INSTITUTIONAL REVIEW BOARD/ INDEPENDENT ETHI...
 
Preparation of protocol
Preparation of protocolPreparation of protocol
Preparation of protocol
 
NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...
NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...
NISHA YADAV ROLLNO. 2116102 PRESENTATION ON DEVELOPING CLINICAL TRIAL PROTOCO...
 
Designing of clinical study protocol rumana hameed
Designing of clinical study protocol rumana hameedDesigning of clinical study protocol rumana hameed
Designing of clinical study protocol rumana hameed
 
planning and initiation.pptx
planning and initiation.pptxplanning and initiation.pptx
planning and initiation.pptx
 
Clinical Trial Protocol.pptx
Clinical Trial Protocol.pptxClinical Trial Protocol.pptx
Clinical Trial Protocol.pptx
 
Protocol Understanding_Katalyst HLS
Protocol Understanding_Katalyst HLSProtocol Understanding_Katalyst HLS
Protocol Understanding_Katalyst HLS
 
Protocol Understanding_ Clinical Data Management_KatalystHLS
Protocol Understanding_ Clinical Data Management_KatalystHLSProtocol Understanding_ Clinical Data Management_KatalystHLS
Protocol Understanding_ Clinical Data Management_KatalystHLS
 
DEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptx
DEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptxDEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptx
DEVELOPING CLINICAL TRIAL PROTOCOL BY PRANAV LENDHEY.pptx
 
Guidelines for the preparation of protocol and documents in clnical trials
Guidelines for the preparation of protocol and documents in clnical trialsGuidelines for the preparation of protocol and documents in clnical trials
Guidelines for the preparation of protocol and documents in clnical trials
 
Clinical Trial Protocol
Clinical Trial ProtocolClinical Trial Protocol
Clinical Trial Protocol
 
Steps For Designing And Development Of Protocol
Steps For Designing And Development Of ProtocolSteps For Designing And Development Of Protocol
Steps For Designing And Development Of Protocol
 
Different Types of Regulatory Documents
Different Types of Regulatory DocumentsDifferent Types of Regulatory Documents
Different Types of Regulatory Documents
 
Spirit 2013
Spirit 2013Spirit 2013
Spirit 2013
 

Mehr von Pharmaceutical Compliance Inspection unit, Crown College of Canada

Mehr von Pharmaceutical Compliance Inspection unit, Crown College of Canada (20)

Career competencies in Canada - Availablity and Flexibility Peivand Pirouzi P...
Career competencies in Canada - Availablity and Flexibility Peivand Pirouzi P...Career competencies in Canada - Availablity and Flexibility Peivand Pirouzi P...
Career competencies in Canada - Availablity and Flexibility Peivand Pirouzi P...
 
Career competencies in Canada - Communication Skills - Peivand Pirouzi, Ph.D.pdf
Career competencies in Canada - Communication Skills - Peivand Pirouzi, Ph.D.pdfCareer competencies in Canada - Communication Skills - Peivand Pirouzi, Ph.D.pdf
Career competencies in Canada - Communication Skills - Peivand Pirouzi, Ph.D.pdf
 
Creating Products and Pricing Strategies to meet customers needs - Peivand Pi...
Creating Products and Pricing Strategies to meet customers needs - Peivand Pi...Creating Products and Pricing Strategies to meet customers needs - Peivand Pi...
Creating Products and Pricing Strategies to meet customers needs - Peivand Pi...
 
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
 
Immigration and Citizenship Funded Seminars: Professor Peivand Pirouzi - Care...
Immigration and Citizenship Funded Seminars: Professor Peivand Pirouzi - Care...Immigration and Citizenship Funded Seminars: Professor Peivand Pirouzi - Care...
Immigration and Citizenship Funded Seminars: Professor Peivand Pirouzi - Care...
 
Immigration and Citizenship - Prof. Peivand Pirouzi - career competencies in ...
Immigration and Citizenship - Prof. Peivand Pirouzi - career competencies in ...Immigration and Citizenship - Prof. Peivand Pirouzi - career competencies in ...
Immigration and Citizenship - Prof. Peivand Pirouzi - career competencies in ...
 
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Mental H...
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Mental H...Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Mental H...
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Mental H...
 
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Manageme...
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Manageme...Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Manageme...
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Manageme...
 
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Entrepre...
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Entrepre...Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Entrepre...
Immigration and citizenship funded seminar - Prof. Peivand Pirouzi - Entrepre...
 
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
 
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
 
Crown College of Canada - Your next step towards becoming faster qualified fo...
Crown College of Canada - Your next step towards becoming faster qualified fo...Crown College of Canada - Your next step towards becoming faster qualified fo...
Crown College of Canada - Your next step towards becoming faster qualified fo...
 
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
 
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
 
Health Canada - Prof. Peivand Pirouzi - Règlements de la Santé Canada pour l’...
Health Canada - Prof. Peivand Pirouzi - Règlements de la Santé Canada pour l’...Health Canada - Prof. Peivand Pirouzi - Règlements de la Santé Canada pour l’...
Health Canada - Prof. Peivand Pirouzi - Règlements de la Santé Canada pour l’...
 
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
Immigration and Citizenship Canada Funded Seminar on Education and Qualificat...
 
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
 
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
 
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
 
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
Immigration and Citizenship Canada - Professor Peivand Pirouzi - Funded Progr...
 

Kürzlich hochgeladen

PPT- Voluntary Liquidation (Under section 59).pptx
PPT- Voluntary Liquidation (Under section 59).pptxPPT- Voluntary Liquidation (Under section 59).pptx
PPT- Voluntary Liquidation (Under section 59).pptxRRR Chambers
 
Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881
Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881
Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881mayurchatre90
 
Debt Collection in India - General Procedure
Debt Collection in India  - General ProcedureDebt Collection in India  - General Procedure
Debt Collection in India - General ProcedureBridgeWest.eu
 
589308994-interpretation-of-statutes-notes-law-college.pdf
589308994-interpretation-of-statutes-notes-law-college.pdf589308994-interpretation-of-statutes-notes-law-college.pdf
589308994-interpretation-of-statutes-notes-law-college.pdfSUSHMITAPOTHAL
 
Divorce Procedure in India (Info) (1).pdf
Divorce Procedure in India (Info) (1).pdfDivorce Procedure in India (Info) (1).pdf
Divorce Procedure in India (Info) (1).pdfdigitalnikesh24
 
如何办理(Lincoln文凭证书)林肯大学毕业证学位证书
如何办理(Lincoln文凭证书)林肯大学毕业证学位证书如何办理(Lincoln文凭证书)林肯大学毕业证学位证书
如何办理(Lincoln文凭证书)林肯大学毕业证学位证书Fs Las
 
Indemnity Guarantee Section 124 125 and 126
Indemnity Guarantee Section 124 125 and 126Indemnity Guarantee Section 124 125 and 126
Indemnity Guarantee Section 124 125 and 126Oishi8
 
The Active Management Value Ratio: The New Science of Benchmarking Investment...
The Active Management Value Ratio: The New Science of Benchmarking Investment...The Active Management Value Ratio: The New Science of Benchmarking Investment...
The Active Management Value Ratio: The New Science of Benchmarking Investment...James Watkins, III JD CFP®
 
IBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptx
IBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptxIBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptx
IBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptxRRR Chambers
 
BPA GROUP 7 - DARIO VS. MISON REPORTING.pdf
BPA GROUP 7 - DARIO VS. MISON REPORTING.pdfBPA GROUP 7 - DARIO VS. MISON REPORTING.pdf
BPA GROUP 7 - DARIO VS. MISON REPORTING.pdflaysamaeguardiano
 
LITERAL RULE OF INTERPRETATION - PRIMARY RULE
LITERAL RULE OF INTERPRETATION - PRIMARY RULELITERAL RULE OF INTERPRETATION - PRIMARY RULE
LITERAL RULE OF INTERPRETATION - PRIMARY RULEsreeramsaipranitha
 
FULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | Delhi
FULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | DelhiFULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | Delhi
FULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | Delhisoniya singh
 
WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)
WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)
WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)Delhi Call girls
 
一比一原版利兹大学毕业证学位证书
一比一原版利兹大学毕业证学位证书一比一原版利兹大学毕业证学位证书
一比一原版利兹大学毕业证学位证书E LSS
 
FINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.ppt
FINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.pptFINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.ppt
FINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.pptjudeplata
 
Human Rights_FilippoLuciani diritti umani.pptx
Human Rights_FilippoLuciani diritti umani.pptxHuman Rights_FilippoLuciani diritti umani.pptx
Human Rights_FilippoLuciani diritti umani.pptxfilippoluciani9
 
KEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptx
KEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptxKEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptx
KEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptxRRR Chambers
 

Kürzlich hochgeladen (20)

PPT- Voluntary Liquidation (Under section 59).pptx
PPT- Voluntary Liquidation (Under section 59).pptxPPT- Voluntary Liquidation (Under section 59).pptx
PPT- Voluntary Liquidation (Under section 59).pptx
 
Vip Call Girls Greater Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS Live
Vip Call Girls Greater Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS LiveVip Call Girls Greater Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS Live
Vip Call Girls Greater Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS Live
 
Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881
Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881
Negotiable Instruments Act 1881.UNDERSTAND THE LAW OF 1881
 
Debt Collection in India - General Procedure
Debt Collection in India  - General ProcedureDebt Collection in India  - General Procedure
Debt Collection in India - General Procedure
 
589308994-interpretation-of-statutes-notes-law-college.pdf
589308994-interpretation-of-statutes-notes-law-college.pdf589308994-interpretation-of-statutes-notes-law-college.pdf
589308994-interpretation-of-statutes-notes-law-college.pdf
 
Old Income Tax Regime Vs New Income Tax Regime
Old  Income Tax Regime Vs  New Income Tax   RegimeOld  Income Tax Regime Vs  New Income Tax   Regime
Old Income Tax Regime Vs New Income Tax Regime
 
Divorce Procedure in India (Info) (1).pdf
Divorce Procedure in India (Info) (1).pdfDivorce Procedure in India (Info) (1).pdf
Divorce Procedure in India (Info) (1).pdf
 
如何办理(Lincoln文凭证书)林肯大学毕业证学位证书
如何办理(Lincoln文凭证书)林肯大学毕业证学位证书如何办理(Lincoln文凭证书)林肯大学毕业证学位证书
如何办理(Lincoln文凭证书)林肯大学毕业证学位证书
 
Indemnity Guarantee Section 124 125 and 126
Indemnity Guarantee Section 124 125 and 126Indemnity Guarantee Section 124 125 and 126
Indemnity Guarantee Section 124 125 and 126
 
The Active Management Value Ratio: The New Science of Benchmarking Investment...
The Active Management Value Ratio: The New Science of Benchmarking Investment...The Active Management Value Ratio: The New Science of Benchmarking Investment...
The Active Management Value Ratio: The New Science of Benchmarking Investment...
 
IBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptx
IBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptxIBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptx
IBC (Insolvency and Bankruptcy Code 2016)-IOD - PPT.pptx
 
BPA GROUP 7 - DARIO VS. MISON REPORTING.pdf
BPA GROUP 7 - DARIO VS. MISON REPORTING.pdfBPA GROUP 7 - DARIO VS. MISON REPORTING.pdf
BPA GROUP 7 - DARIO VS. MISON REPORTING.pdf
 
LITERAL RULE OF INTERPRETATION - PRIMARY RULE
LITERAL RULE OF INTERPRETATION - PRIMARY RULELITERAL RULE OF INTERPRETATION - PRIMARY RULE
LITERAL RULE OF INTERPRETATION - PRIMARY RULE
 
FULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | Delhi
FULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | DelhiFULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | Delhi
FULL ENJOY - 8264348440 Call Girls in Netaji Subhash Place | Delhi
 
WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)
WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)
WhatsApp 📞 8448380779 ✅Call Girls In Nangli Wazidpur Sector 135 ( Noida)
 
一比一原版利兹大学毕业证学位证书
一比一原版利兹大学毕业证学位证书一比一原版利兹大学毕业证学位证书
一比一原版利兹大学毕业证学位证书
 
Sensual Moments: +91 9999965857 Independent Call Girls Vasundhara Delhi {{ Mo...
Sensual Moments: +91 9999965857 Independent Call Girls Vasundhara Delhi {{ Mo...Sensual Moments: +91 9999965857 Independent Call Girls Vasundhara Delhi {{ Mo...
Sensual Moments: +91 9999965857 Independent Call Girls Vasundhara Delhi {{ Mo...
 
FINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.ppt
FINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.pptFINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.ppt
FINALTRUEENFORCEMENT OF BARANGAY SETTLEMENT.ppt
 
Human Rights_FilippoLuciani diritti umani.pptx
Human Rights_FilippoLuciani diritti umani.pptxHuman Rights_FilippoLuciani diritti umani.pptx
Human Rights_FilippoLuciani diritti umani.pptx
 
KEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptx
KEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptxKEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptx
KEY NOTE- IBC(INSOLVENCY & BANKRUPTCY CODE) DESIGN- PPT.pptx
 

Professor Peivand Pirouzi - International clinical study and safety reporting activities - Publication, Canada - All rights reserved

  • 1. Clinical Study and Safety Reporting Activities Writing a Clinical Study Report & Overview of safety reports :Periodic Safety Reports: DSUR, PSUR, PBRER, ASR, IB, and Orphan Designation Annual Report Pr. Peivand Pirouzi
  • 2. ICH E3 Structure and Content of Clinical Study Reports 1. Title page 2. Synopsis 3. Table of contents 4. List of abbreviations 5. Ethics 6. Investigators and study administrative structure 7. Introduction 8. Study objectives 9. Investigational plan 10. Study patients 11. Efficacy evaluation 12. Safety evaluation 13. Discussion and overall conclusions 14. Tables, figures and graphs referred to but not included in the text 15. Reference list 16. Appendices * Details for Sections 9 – 12 on next slides 2
  • 3. ICH E3 Structure and Content of Clinical Study Reports (cont.) 9. Investigational plan 9.1 Overall study design and plan description 9.2 Discussion of study design, including the choice of control groups 9.3 Selection of study population 9.3.1 Inclusion Criteria 9.3.2 Exclusion Criteria 9.3.3 Removal of Patients from Therapy or Assessment 9.4 Treatments 9.4.1 Treatments Administered 9.4.2 Identity of Investigational Product(s) 9.4.3 Method of Assigning Patients to Treatment Groups 9.4.4 Selection of Doses in the Study 9.4 Treatments (cont.) 9.4.5 Selection and Timing of Dose for each Patient 9.4.6 Blinding 9.4.7 Prior and Concomitant Therapy 9.4.8 Treatment Compliance 9.5 Efficacy and safety variables 9.5.1 Efficacy and Safety Measurements Assessed and Flow Chart 9.5.2 Appropriateness of Measurements 9.5.3 Primary Efficacy Variable(s) 9.5.4 Drug Concentration Measurements 9.6 Data quality assurance 9.7 Statistical methods planned in the protocol & determination of sample size 9.8 Changes in the conduct of the study or planned analyses 3
  • 4. ICH E3 Structure and Content of Clinical Study Reports (cont.) 10 Study patients 10.1 Disposition of patients 10.2 Protocol deviations 11. Efficacy evaluation 11.1 Data sets analyzed 11.2 Demographic and other baseline characteristics 11.3 Measurements of treatment compliance 11.4 Efficacy results and tabulations of individual patient data 12. Safety evaluation 12.1 Extent of exposure 12.2 Adverse events (AEs) 12.3 Deaths, other SAEs, and other significant adverse events 12.4 Clinical laboratory evaluation 12.5 Vital signs, physical findings and other observations related to safety 12.6 Safety conclusions 4
  • 5. 1. Title Page • The title page should contain the following information: • study title • name of test drug/investigational product • indication studied • if not apparent from the title, a brief (1 to 2 sentences) description giving design (parallel, cross-over, blinding, randomised) comparison (placebo, active, dose/response), duration, dose, and patient population • name of the sponsor • protocol identification (code or number) • development phase of study • study initiation date (first patient enrolled, or any other verifiable • date of early study termination, if any • study completion date (last patient completed) • name and affiliation of principal or coordinating investigator(s) or sponsor's responsible medical officer • name of company/sponsor signatory (the person responsible for the study report within the company/sponsor. The name, telephone number and fax number of the company/sponsor contact persons for questions arising during review of the study report should be indicated an this page or in the letter of application.) • statement indicating whether the study was performed in compliance with Good Clinical Practices (GCP), including the archiving of essential documents • date of the report (identify any earlier reports from the same study by title and date)
  • 6. 2. Synopsis • A brief synopsis (usually limited to 3 pages) that summarises the study should be provided (see Annex I of the guidance document for an example of a synopsis format used in Europe). The synopsis should include numerical data to illustrate results, not just text or p-values.
  • 7. 3. Table of Contents For The Individual Clinical Study Report The table of contents should include: • the page number or other locating information of each section, including summary tables, figures and graphs, • a list and the locations of appendices, tabulations and any case report forms provided.
  • 8. 4. List of Abbreviations and Definition of Terms • A list of the abbreviations, and lists and definitions of specialised or unusual terms or measurements units used in the report should be provided. Abbreviated terms should be spelled out and the abbreviation indicated in parentheses at first appearance in the text.
  • 9. 5. Ethics 5.1 Independent Ethics Committee (IEC) or Institutional Review Board (IRB) • It should be confirmed that the study and any amendments were reviewed by an Independent Ethics Committee or Institutional Review Board. A list of all IECs or IRBs consulted should be given in (appendix 16.1.3) and, if required by the regulatory authority, the name of the committee Chair should be provided.
  • 10. 5.2 Ethical Conduct of the Study • It should be confirmed that the study was conducted in accordance with the ethical principles that have their origins in the Declaration of Helsinki.
  • 11. 5.3 Patient Information and Consent • How and when informed consent was obtained in relation to patient enrollment, (e.g., at allocation, pre-screening) should be described. • Representative written information for the patient (if any) and a sample patient consent form should be provided in (appendix 16.1.3).
  • 12. 6. Investigators and Study Administrative Structure • The administrative structure of the study (e.g., principal investigator, coordinating investigator, steering committee, administration, monitoring and evaluation committees, institutions, statistician, central laboratory facilities, contract research organization (C.R.O.), clinical trial supply management) should be described briefly in the body of the report.
  • 13. 6. Investigators and Study Administrative Structure The listing should include: • Investigators • Any other person carrying out observations of primary or other major efficacy variables, such as a nurse, physician's assistant, clinical psychologist, clinical pharmacist, or house staff physician. It is not necessary to include in this list a person with only an occasional role, e.g., an on-call physician who dealt with a possible adverse effect or a temporary substitute for any of the above. • The author(s) of the report, including the responsible biostatistician(s).
  • 14. 6. Investigators and Study Administrative Structure • Where signatures of the principal or coordinating investigators are required by regulatory authorities, these should be included in (appendix 16.1.5 see Annex II for a sample form). Where these are not required, the signature of the sponsor's responsible medical officer should be provided in (appendix 16.1.5).
  • 15. 7. Introduction • The introduction should contain a brief statement (maximum: 1 page) placing the study in the context of the development of the test drug/investigational product, relating the critical features of the study (e.g., rationale and aims, target population. treatment, duration, primary endpoints) to that development. Any guidelines that were followed in the development of the protocol or any other agreements/meetings between the sponsor/company and regulatory authorities that are relevant to the particular study, should be identified or described.
  • 16. 8. Study Objectives • A statement describing the overall purpose(s) of the study should be provided
  • 17. 9. Investigational Plan 9.1 Overall Study Design and Plan - Description • The overall study plan and design (configuration) of the study (e.g., parallel, crossover) should be described briefly but clearly, using charts and diagrams as needed. If other studies used a very similar protocol, it may be useful to note this and describe any important differences. The actual protocol and any changes should be included as (appendix 16.1.1) and a sample case report form (unique pages only; i.e., it is not necessary to include identical pages from forms for different evaluations or visits) as (appendix 16.1.2). If any of the information in this section comes from sources other than the protocol, these should be identified.
  • 18. 9. Investigational Plan 9.1 Overall Study Design and Plan - Description The information provided should include: • treatments studied (specific drugs, doses and procedures) • patient population studied and the number of patients to be included. • level and method of blinding/masking (e.g., open, double-blind, single- blind, blinded evaluators and unblinded patients and/or investigators) • kind of control(s) (e.g., placebo, no treatment, active drug, dose-response, historical) and study configuration (parallel, cross-over) • method of assignment to treatment (randomisation, stratification) • sequence and duration of all study periods, including pre-randomisation and post-treatment periods, therapy withdrawal periods and single- and double-blind treatment periods. When patients are randomized should be specified. It is usually helpful to display the design graphically with a flow chart which includes timing of assessments (see Annexes IIIa and Illb for an example). • any safety, data monitoring or special steering or evaluation committees • any interim analyses.
  • 19. 9.2 Discussion of Study Design, Including the Choice of Control Groups • The specific control chosen and the study design used should be discussed, as necessary. • Known or potential problems associated with the study design or control group chosen, should be discussed in light of the specific disease and therapies being studied. For a crossover design, for example, there should be consideration, among other things, of the likelihood of spontaneous change in the disease and of carry- over effects of treatment during the study.
  • 20. 9.3 Selection of Study Population 9.3.1 Inclusion Criteria • The patient population and the selection criteria used to enter the patients into the study should be described, and the suitability of the population for the purposes of the study discussed. Specific diagnostic criteria used, as well as specific disease requirements (e.g., disease of a particular severity or duration, results of a particular test or rating scale(s) or physical examination, particular features of clinical history, such as failure or success on prior therapy, or other potential prognostic factors and any age, sex or ethnic factors) should be presented.
  • 21. 9.3.2 Exclusion Criteria • The criteria for exclusion at entry into the study should be specified and the rationale (e.g., safety concerns, administrative reasons or lack of suitability for the trial) provided. The impact of exclusions on the generalisability of the study should be discussed in (section 13) of the study report, or in an overview of safety and efficacy.
  • 22. 9.3.3 Removal of Patients from Therapy or Assessment • The predetermined reasons for removing patients from therapy or assessment observation, if any, should be described, as should the nature and duration of any planned follow-up observations in those patients.
  • 23. 9.4 Treatments 9.4.1 Treatments Administered • The precise treatments or diagnostic agents to be administered in each arm of the study, and for each period of the study, should be described including route and mode of administration, dose and dosage schedule.
  • 24. 9.4.2 Identity of Investigational Product(s) • In the text of the report, a brief description of the test drug(s)/investigational product(s) (formulation, strength, batch number(s)) should be given. If more than one batch of test drug/investigational product was used, patients receiving each batch should be identified in (appendix 16.1.6).
  • 25. 9.4.3 Method of Assigning Patients to Treatment Groups • The specific methods used to assign patients to treatment groups, e.g., centralised allocation, allocation within sites, adaptive allocation (that is, assignment on the basis of earlier assignment or outcome) should be described in the text of the report, including any stratification or blocking procedures. Any unusual features should be explained.
  • 26. 9.4.4 Selection of Doses in the Study • The doses or dose ranges used in the study should be given for all treatments and the basis for choosing them described (e.g., prior experience in humans, animal data).
  • 27. 9.4.5 Selection and Timing of Dose for Each Patient • Procedures for selecting each patient's dose of test drug/investigational product and active control/comparator should be described. These procedures can vary from simple random assignment to a selected fixed drug/dose regimen, to some specified titration procedure, to more elaborate response-determined selection procedures, e.g. where dose is titrated upward at intervals until intolerance or some specified endpoint is achieved. Procedures for back- titration, if any, should also be described.
  • 28. 9.4.6 Blinding • A description of the specific procedures used to carry out blinding should be provided (e.g., how bottles were labeled, labels that reveal blind-breakage, sealed code list/envelopes, double dummy techniques), including the circumstances in which the blind would be broken for an individual or for all patients, e.g., for serious adverse events, the procedures used and who had access to patient codes.
  • 29. 9.4.7 Prior and Concomitant Therapy • Which drugs or procedures were allowed before and during the study, whether and how their use was recorded, and any other specific rules and procedures related to permitted or forbidden concomitant therapy should be described. How allowed concomitant therapy might affect the outcome due either to drug-drug interaction or to direct effects on the study endpoints should be discussed, and how the independent effects of concomitant and study therapies could be ascertained should be explained.
  • 30. 9.4.8 Treatment Compliance • The measures taken to ensure and document treatment compliance should be described, e.g., drug accountability, diary cards, blood, urine or other body fluid drug level measurements, or medication event monitoring.
  • 31. 9.5 Efficacy and Safety Variables 9.5.1 Efficacy and Safety Measurements Assessed and Flow Chart • The specific efficacy and safety variables to be assessed and laboratory tests to be conducted, their schedule (days of study, time of day, relation to meals, and the timing of critical measures in relation to test drug administration, e.g., just prior to next dose, two hours after dose), the methods for measuring them, and the persons responsible for the measurements should be described. If there were changes in personnel carrying out critical measurements, these should be reported.
  • 32. 9.5.2 Appropriateness of Measurements • If any of the efficacy or safety assessments was not standard, i.e., widely used and generally recognised as reliable, accurate, and relevant (able to discriminate between effective and ineffective agents), its reliability, accuracy and relevance should be documented. It may be helpful to describe alternatives considered but rejected.
  • 33. 9.5.2 Appropriateness of Measurements • If a surrogate endpoint (a laboratory measurement or physical measurement or sign that is not a direct measure of clinical benefit) was used as a study endpoint, this should be justified e.g., by reference to clinical data, publications, guidelines or previous actions by regulatory authorities.
  • 34. 9.5.3 Primary Efficacy Variable(s) • The primary measurements and endpoints used to determine efficacy should be clearly specified. Although the critical efficacy measurements may seem obvious, when there are multiple variables, or when variables are measured repeatedly, the protocol should identify the primary ones, with an explanation of why they were chosen, or designate the pattern of significant findings or other method of combining information that would be interpreted as supporting efficacy.
  • 35. 9.5.4 Drug Concentration Measurements • Any drug concentrations to be measured, and the sample collection times and periods in relation to the timing of drug administration, should be described. Any relation of drug administration and sampling to ingestion of food, posture and the possible effects of concomitant medication/alcohol/caffeine/nicotine should also be addressed. The biological sample measured, the handling of samples and the method of measurement used should be described, referring to published and/or internal assay validation documentation for methodological details.
  • 36. 9.6 Data Quality Assurance • The quality assurance and quality control systems implemented to assure the quality of the data should be described in brief. If none were used, this should be stated. Documentation of inter-laboratory standardisation methods and quality assurance procedures, if used, should be provided under (appendix 16.1.10).
  • 37. 9.7 Statistical Methods Planned in the Protocol and Determination of Sample Size
  • 38. 9.7.1 Statistical and Analytical Plans • The statistical analyses planned in the protocol and any changes made before outcome results were available should be described. In this section emphasis should be on which analyses, comparisons and statistical tests were planned, not on which ones were actually used. If critical measurements were made more than once, the particular measurements (e.g., average of several measurements over the entire study, values at particular times, values only from study completers, or last on-therapy value) planned as the basis for comparison of test drug/investigational product and control should be specified.
  • 39. 9.7.2 Determination of Sample Size • The planned sample size and the basis for it, such as statistical considerations or practical limitations, should be provided. Methods for sample size calculation should be given together with their derivations or source of reference. Estimates used in the calculations should be given and explanations provided as to how they were obtained.
  • 40. 9.8 Changes in the Conduct of the Study or Planned Analyses • Any change in the conduct of the study or planned analyses (e.g., dropping a treatment group, changing the entry criteria or drug dosages, adjusting the sample size, etc.) instituted after the start of the study should be described. The time(s) and reason(s) for the change(s), the procedure used to decide on the change(s), the person(s) or group(s) responsible for the change(s) and the nature and content of the data available (and to whom they were available) when the change was made should also be described, whether the change was documented as a formal protocol amendment or not (Personnel changes need not be included.) A
  • 41. 10. Study Patients 10.1 Disposition of Patients • There should be a clear accounting of all patients who entered the study, using figures or tables in the text of the report. The numbers of patients who were randomised, and who entered and completed each phase of the study, (or each week/month of the study) should be provided, as well as the reasons for all post- randomisation discontinuations, grouped by treatment and by major reason (lost to follow-up, adverse event, poor compliance, etc.). It may also be relevant to provide the number of patients screened for inclusion and a breakdown of the reasons for excluding patients during screening, if this could help clarify the appropriate patient population for eventual drug use.
  • 42. 10.2 Protocol Deviations • All important deviations related to study inclusion or exclusion criteria, conduct of the trial, patient management or patient assessment should be described. • In the body of the text, protocol deviations should be appropriately summarized by centre and grouped into different categories, such as: • those who entered the study even though they did not satisfy the entry criteria • those who developed withdrawal criteria during the study but were not withdrawn • those who received the wrong treatment or incorrect dose • those who received an excluded concomitant treatment In (appendix 16.2.2), individual patients with these protocol deviations should be listed, broken down by centre for multicentre studies.
  • 43. 11. Efficacy Evaluation 11.1 Data Sets Analysed • xactly which patients were included in each efficacy analysis should be precisely defined, e.g., all patients receiving any test drugs/investigational products, all patients with any efficacy observation or with a certain minimum number of observations, only patients completing the trial, all patients with an observation during a particular time window, only patients with a specified degree of compliance, etc. It should be clear, if not defined in the study protocol, when, (relative to study unblinding), and how inclusion/exclusion criteria for the data sets analysed were developed.
  • 44. 11.2 Demographic and Other Baseline Characteristics • Group data for the critical demographic and baseline characteristics of the patients, as well as other factors arising during the study that could affect response, should be presented in this section and comparability of the treatment groups for all relevant characteristics should be displayed by use of tables or graphs in (section 14.1). • The data for the patient sample included in the "all patients with data" analysis should be given first. This can then be followed by data on other groups used in principal analyses, such as the "per-protocol" analysis or other analyses, e.g., groups defined by compliance, concomitant disease/therapy, or demographic/baseline characteristics. When such groups are used, data for the complementary excluded group should also be shown.
  • 45. 11.2 Demographic and Other Baseline Characteristics • The critical variables will depend on the specific nature of the disease and on the protocol but will usually include: • demographic variables – age – sex – race • disease factors – specific entry criteria (if not uniform), duration, stage and severity of disease and other clinical classifications and sub-groupings in common usage or of known prognostic significance. – baseline values for critical clinical measurements carried out during the study or identified as important indicators of prognosis or response to therapy. – concomitant illness at trial initiation, such as renal disease, diabetes, heart failure – relevant previous illness – relevant previous treatment for illness treated in the study – concomitant treatment maintained, even if the dose was changed during the study, including oral contraceptive and hormone replacement therapy; treatments stopped at entry into the study period (or changed at study initiation) • other factors that might affect response to therapy (e.g., weight, renin status, antibody levels, metabolic status) • other possibly relevant variables (e.g., smoking, alcohol intake, special diets) and, for women, menstrual status and date of last menstrual period, if pertinent for the study
  • 46. 11.3 Measurements of Treatment Compliance • Any measurements of compliance of individual patients with the treatment regimen under study and drug concentrations in body fluids should be summarised, analysed by treatment group and time interval, and tabulated in (Appendix 16.2.5).
  • 47. 11.4 Efficacy Results and Tabulations of Individual Patient Data 11.4.1 Analysis of Efficacy • Treatment groups should be compared for all critical measures of efficacy (primary and secondary endpoints; any pharmacodynamic endpoints studied), as well as benefit/risk assessment(s) in each patient where these are utilized. • In general, the results of all analyses contemplated in the protocol and an analysis including all patients with on-study data should be performed in studies intended to establish efficacy. The analysis should show the size (point estimate) of the difference between the treatments, the associated confidence interval, and where utilized, the results of hypothesis testing.
  • 48. 11.4.2 Statistical/Analytical Issues • The statistical analysis used should be described for clinical and statistical reviewers in the text of the report, with detailed documentation of statistical methods (see section Annex IX) presented in (appendix 16.1.9).
  • 49. 11.4.2.1 Adjustments for Covariates • Selection of, and adjustments for, demographic or baseline measurements, concomitant therapy, or any other covariate or prognostic factor should be explained in the report, and methods of adjustment, results of analyses, and supportive information (e.g., ANCOVA or Cox regression output) should be included in the detailed documentation of statistical methods.
  • 50. 11.4.2.2 Handling of Dropouts or Missing Data • There are several factors that may affect dropout rates. These include the duration of the study, the nature of the disease, the efficacy and toxicity of the drug under study, and other factors that are not therapy related. Ignoring the patients who dropped out of the study and drawing conclusions based only on patients who completed the study can be misleading. • A large number of dropouts, however, even if included in an analysis, may introduce bias, particularly if there are more early dropouts in one treatment group or the reasons for dropping out are treatment or outcome related. Although the effects of early dropouts, and sometimes even the direction of bias, can be difficult to determine, possible effects should be explored as fully as possible.
  • 51. 11.4.2.3 Interim Analyses and Data Monitoring • The process of examining and analysing data accumulating in a clinical trial, either formally or informally, can introduce bias and/or increase type I error. Therefore, all interim analyses, formal or informal, pre-planned or ad hoc, by any study participant, sponsor staff member, or data monitoring group should be described in full, even if the treatment groups were not identified.
  • 52. 11.4.2.4 Multicentre Studies • A multicentre study is a single study under a common protocol, involving several centres (e.g., clinics, practices, hospitals) where the data collected are intended to be analysed as a whole (as opposed to a post-hoc decision to combine data or results from separate studies). Individual centre results should be presented, however, where appropriate, e.g., when the centres have sufficient numbers of patients to make such analysis potentially valuable, the possibility of qualitative or quantitative treatment-by-centre interaction should be explored.
  • 53. 11.4.2.5 Multiple Comparison/Multiplicity • False positive findings increase in number as the number of significance tests (number of comparisons) performed increases. If there was more than one primary endpoint (outcome variable), more than one analysis of particular endpoint, or if there were multiple treatment groups, or subsets of the patient population being examined, the statistical analysis should reflect awareness of this and either explain the statistical adjustment used for type I error criteria or give reasons why it was considered unnecessary.
  • 54. 11.4.2.6 Use of an "Efficacy Subset" of Patients • Particular attention should be devoted to the effects of dropping patients with available data from analyses because of poor compliance, missed visits, ineligibility, or any other reason. As noted above, an analysis using all available data should be carried out for all studies intended to establish efficacy, even if it is not the analysis proposed as the primary analysis by the applicant.
  • 55. 11.4.2.7 Active-Control Studies Intended to Show Equivalence • If an active control study is intended to show equivalence (i.e., lack of a difference greater than a specified size) between the test drug/investigational product and the active control/comparator, the analysis should show the confidence interval for the comparison between the two agents for critical end points and the relation of that interval to the prespecified degree of inferiority that would be considered unacceptable. (See 9.2 for important considerations when using the active control equivalence design.)
  • 56. 11.4.2.8 Examination of Subgroups • If the size of the study permits, important demographic or baseline value-defined subgroups should be examined for unusually large or small responses and the results presented, e.g., comparison of effects by age, sex, or race, by severity or prognostic groups, by history of prior treatment with a drug of the same class, etc.
  • 57. 11.4.3 Tabulation of Individual Response Data • In addition to tables and graphs representing group data, individual response data and other relevant study information should be presented in tables. Some regulatory authorities may require all individual data in archival case report tabulations. • What needs to be included in the report will vary from study to study and from one drug class to another and the applicant must decide, if possible after consultation with the regulatory authority, what to include in appendix to the study report.
  • 58. 11.4.4 Drug Dose, Drug Concentration, and Relationships to Response • When the dose in each patient can vary, the actual doses received by patients should be shown and individual patient's doses should be tabulated. • Although studies not designed as dose-response studies may have limited ability to contribute dose- response information, the available data should be examined for whatever information they can yield. In examining the dose response, it may be helpful to calculate dose as mg/kg body weight or mg/m2 body surface.
  • 59. 11.4.5 Drug-Drug and Drug-Disease Interactions • Any apparent relationship between response and concomitant therapy and between response and past and/or concurrent illness should be described.
  • 60. 11.4.6 By-Patient Displays • While individual patient data ordinarily can be displayed in tabular listings, it has on occasion been helpful to construct individual patient profiles in other formats, such as graphic displays. These might for example, show the value of a particular parameter(s) over time, the drug dose over the same period, and the times of particular events (e.g., an adverse event or change in concomitant therapy).
  • 61. 11.4.7 Efficacy Conclusions • The important conclusions concerning efficacy should be concisely described, considering primary and secondary endpoints, pre- specified and alternative statistical approaches and results of exploratory analyses.
  • 62. 12. Safety Evaluation • Analysis of safety-related data can be considered at three levels. First, the extent of exposure (dose, duration, number of patients) should be examined to determine the degree to which safety can be assessed from the study. Second, the more common adverse events, laboratory test changes, etc. should be identified, classified in some reasonable way, compared for treatment groups, and analysed, as appropriate, for factors that may affect the frequency of adverse reactions/events, such as time dependence, relation to demographic characteristics, relation to dose or drug concentration, etc.
  • 63. 12.1 Extent of Exposure • The extent of exposure to test drugs/investigational products (and to active control and placebo) should be characterised according to the number of patients exposed, the duration of exposure, and the dose to which they were exposed. – Duration – Dose – Drug concentration
  • 64. 12.2 Adverse Events (AES) 12.2.1 Brief Summary of Adverse Events • The overall adverse event experience in the study should be described in a brief narrative, supported by the following more detailed tabulations and analyses. In these tabulations and analyses, events associated with both the test drug and control treatment should be displayed.
  • 65. 12.2.2 Display of Adverse Events • All adverse events occurring after initiation of study treatments (including events likely to be related to the underlying disease or likely to represent concomitant illness, unless there is a prior agreement with the regulatory authority to consider specified events as disease related) should be displayed in summary tables (section 14.3.1). The tables should include changes in vital signs and any laboratory changes that were considered serious adverse events or other significant adverse events.
  • 66. Adverse Events: Number Observed and Rate with Patient Identifications Adverse Events: Number Observed and Rate with Patient Identifications Treatment Group X N=50 Mild Moderate Severe Total Total Related * NR Related NR Related NR Related NR R+NR * NR = not related; related could be expanded, e.g., as definite, probable, possible ** Patient identification number Body System A Event 1 6(12%) 2(4%) 3(6%) 1(2%) 3(6%) 1(2%) 12(24% ) 4(8%) N11 ** N21 N31 N41 N51 N61 N12 N22 N32 N52 N13 N33 N53 N14 N15 N16 Event 2
  • 67. 12.2.2 Display of Adverse Events • In addition to these complete tables provided in 14.3.1, an additional summary table comparing treatment and control groups, without the patient identifying numbers limited to relatively common adverse events (e.g., those in at least 1 % of the treated group), should be provided in the body of the report. • In presenting adverse events, it is important both to display the original terms used by the investigator and to attempt to group related events (i.e., events that probably represent the same phenomena) so that the true occurrence rate is not obscured. One way to do this is with a standard adverse reaction/events dictionary.
  • 68. 12.2.3 Analysis of Adverse Events • The basic display of adverse event rates described in(section 12.2.2 and located in section 14.3.1) of the report, should be used to compare rates in treatment and control groups. For this analysis it may be helpful to combine the event severity categories and the causality categories, leading to a simpler side- by-side comparison of treatment groups. I
  • 69. 12.2.3 Analysis of Adverse Events • In addition, although this is usually best done in an integrated analysis of safety, if study size and design permit, it may be useful to examine the more common adverse events that seem to be drug related for relationship to dosage and to mg/kg or mg/m2dose, to dose regimen, to duration of treatment, to total dose, to demographic characteristics such as age, sex, race, to other baseline features such as renal status, to efficacy outcomes, and to drug concentration. It may also be useful to examine time of onset and duration of adverse events. A variety of additional analyses may be suggested by the study results or by the pharmacology of the test drug/investigational product.
  • 70. 12.2.4 Listing of Adverse Events by Patient • All adverse events for each patient, including the same event on several occasions should be listed in (appendix 16.2.7), giving both preferred term and the original term used by the investigator.
  • 71. 12.2.4 Listing of Adverse Events by Patient The listing should be by investigator and by treatment group and should include: • Patient identifier • Age, race, sex, weight (height, if relevant) • Location of CRFs, if provided. • The adverse event (preferred term, reported term) • Duration of the adverse event • Severity (e.g. mild, moderate, severe) • Seriousness (serious/non-serious) • Action taken (none, dose reduced, treatment stopped, specific treatment instituted, etc.) • Outcome (e.g., CIOMS format) • Causality assessment, (e.g., related/not related). How this was determined should be described in the table or elsewhere. • Date of onset or date of clinic visit at which the event was discovered • Timing of onset of the adverse event in relation to last dose of test drug/investigational product (when applicable) • Study treatment at time of event or most recent study treatment taken • Test drug/investigational product dose in absolute amount, mg/kg or mg/m2 at time of event • Drug concentration (if known) • Duration of test drug/investigational product treatment • Concomitant treatment during study
  • 72. 12.3 Deaths, Other Serious Adverse Events, and Other Significant Adverse Events • Deaths, other serious adverse events, and other significant adverse events deserve special attention.
  • 73. 12.3.1 Listing of Deaths, Other Serious Adverse Events and Other Significant Adverse Events • Listings, containing the same information as called for in (section 12.2.4) above, should be provided for the following events.
  • 74. 12.3.1.1 Deaths • All deaths during the study, including the post treatment follow-up period, and deaths that resulted from a process that began during the study, should be listed by patient in (section 14.3.2).
  • 75. 12.3.1.2 Other Serious Adverse Events • All serious adverse events (other than death but including the serious adverse events temporally associated with or preceding the deaths) should be listed in (section 14.3.2) The listing should include laboratory abnormalities, abnormal vital signs and abnormal physical observations that were considered serious adverse events.
  • 76. 12.3.1.3 Other Significant Adverse Events • Marked hematological and other laboratory abnormalities (other than those meeting the definition of serious) and any events that led to an intervention, including withdrawal of test drug/investigational product treatment, dose reduction, or significant additional concomitant therapy, other than those reported as serious adverse events, should be listed in (section 14.3.2).
  • 77. 12.3.2 Narratives of Deaths, Other Serious Adverse Events and Certain Other Significant Adverse Events • There should be brief narratives describing each death, each other serious adverse event, and those of the other significant adverse events that are judged to be of special interest because of clinical importance. These narratives can be placed either in the text of the report or in (section 14.3.3), depending on their number.
  • 78. 12.3.2 Narratives of Deaths, Other Serious Adverse Events and Certain Other Significant Adverse Events In addition, the following information should be included: • Patient identifier • Age and sex of patient; general clinical condition of patient, if appropriate • Disease being treated (if the same for all patients this is not required) with duration (of current episode) of illness • Relevant concomitant/previous illnesses with details of occurrence/duration • Relevant concomitant/previous medication with details of dosage • Test drug/investigational product administered, drug dose, if this varied among patients, and length of time administered
  • 79. 12.3.3 Analysis and Discussion of Deaths, Other Serious Adverse Events and Other Significant Adverse Events • The significance of the deaths, other serious adverse events and other significant adverse events leading to withdrawal, dose reduction or institution of concomitant therapy should be assessed with respect to the safety of the test drug/investigational product. Particular attention should be paid to whether any of these events may represent a previously unsuspected important adverse effect of the test drug/investigational product.
  • 81. 12.4.1 Listing of Individual Laboratory Measurements by Patient (16.2.8) and Each Abnormal Laboratory Value (14.3.4) • When required by regulatory authorities, the results of all safety-related laboratory tests should be available in tabular listings, using a display similar to the following, where each row represents a patient visit at which a laboratory study was done, with patients grouped by investigator (if more than one) and treatment group, and columns include critical demographic data, drug dose data, and the results of the laboratory tests.
  • 82. List Of Laboratory Measurements List Of Laboratory Measurements Laboratory Tests Patien t Time Age Sex Race Weigh t Dose SGOT SGPT AP.... X * Vn = value of a particular test #1 T0 70 M W 70kg 400mg V1 * V5 V9 T1 V2 V6 V10 T2 V3 V4 V15 T3 V4 V8 V12 #2 T10 65 F B 50kg 300mg V13 V16 V19 T21 V14 V17 V20 T32 V15 V18 V21
  • 83. 12.4.2 Evaluation of Each Laboratory Parameter • The necessary evaluation of laboratory values must in part be determined by the results seen, but, in general, the following analyses should be provided. For each analysis, comparison of the treatment and control groups should be carried out, as appropriate and as compatible with study size. In addition, normal laboratory ranges should be given for each analysis.
  • 84. 12.4.2.1 Laboratory Values Over Time • For each parameter at each time over the course of the study (e.g., at each visit) the following should be described: the group mean or median values, the range of values, and the number of patients with abnormal values, or with abnormal values that are of a certain size (e.g., twice the upper limit of normal, 5 times the upper limit; choices should be explained). Graphs may be used.
  • 85. 12.4.2.2 Individual Patient Changes • An analysis of individual patient changes by treatment group should be given. A variety of approaches may be used, including: 1. "Shift tables" - These tables show the number of patients who are low, normal, or high at baseline and then at selected time intervals. 2. Tables showing the number or fraction of patients who had a change in parameter of a predetermined size at selected time intervals. For example, for BUN, it might be decided that a change of more than 10 mg/dL BUN should be noted. 3. A graph comparing the initial value and the on-treatment values of a laboratory measurement for each patient
  • 86. 12.4.2.3 Individual Clinically Significant Abnormalities • Clinically significant changes (defined by the applicant) should be discussed. A narrative of each patient whose laboratory abnormality was considered a serious adverse event and, in certain cases, considered an other significant adverse event, should be provided under (sections 12.3.2 or 14.3.3).
  • 87. 12.5 Vital Signs, Physical Findings and Other Observations Related to Safety • Vital signs, other physical findings, and other observations related to safety should be analysed and presented in a way similar to laboratory variables. If there is evidence of a drug effect, any dose-response or drug concentration-response relationship or relationship to patient variables (e.g., disease, demographics, concomitant therapy) should be identified and the clinical relevance of the observation described.
  • 88. 12.6 Safety Conclusions • The overall safety evaluation of the test drug(s)/investigational product(s) should be reviewed, with particular attention to events resulting in changes of dose or need for concomitant medication, serious adverse events, events resulting in withdrawal, and deaths.
  • 89. 13. Discussion and Overall Conclusions • The efficacy and safety results of the study and the relationship of risks and benefit should be briefly summarised and discussed, referring to the tables, figures, and sections above as needed. The presentation should not simply repeat the description of results nor introduce new results.
  • 90. 14. Tables, Figures and Graphs Referred to But Not Included in the Text • igures should be used to visually summarise the important results, or to clarify results that are not easily understood from tables. • Important demographic, efficacy and safety data should be presented in summary figures or tables in the text of the report. However, if these become obtrusive because of size or number they should be presented here, cross-referenced to the text, along with supportive, or additional, figures, tables or listings.
  • 91. The following information may be presented in this section of the core clinical study report: • 14.1 Demographic Data • Summary figures and tables • 14.2 Efficacy Data • Summary figures and tables • 14.3 Safety Data • Summary figures and tables • 14.3.1 Displays of Adverse Events • 14.3.2 Listings of Deaths, Other Serious and Significant Adverse Events • 14.3.3 Narratives of Deaths, Other Serious and Certain Other Significant Adverse Events • 14.3.4 Abnormal Laboratory Value Listing (each patient)
  • 92. 15. Reference List • A list of articles from the literature pertinent to the evaluation of the study should be provided. Copies of important publications should be attached in an (appendix 16.1.11 and 16.1.12).
  • 93. 16. Appendices • This section should be prefaced by a full list of all appendices available for the study report. Where permitted by the regulatory authority, some of the following appendices need not be submitted with the report but need to be provided only on request.
  • 94. • 16.1 Study Information • 16.1.1 Protocol and protocol amendments • 16.1.2 Sample case report form (unique pages only) • 16.1.3 List of IECs or IRBs (plus the name of the committee Chair if required by the regulatory authority) - Representative written information for patient and sample consent forms • 16.1.4 List and description of investigators and other important participants in the study, including brief (1 page) CVs or equivalent summaries of training and experience relevant to the performance of the clinical study • 16.1.5 Signatures of principal or coordinating investigator(s) or sponsor's responsible medical officer, depending on the regulatory authority's requirement. • 16.1.6 Listing of patients receiving test drug(s)/investigational product(s) from specific batches, where more than one batch was used.
  • 95. • 16.1.7 Randomisation scheme and codes (patient identification and treatment assigned) • 16.1.8 Audit certificates (if available) (see Annex IVa and IVb of the guidance) • 16.1.9 Documentation of statistical methods • 16.1.10 Documentation of inter-laboratory standardisation methods and quality assurance procedures if used. • 16.1.11 Publications based on the study • 16.1.12 Important publications referenced in the report • 16.2 Patient Data Listings
  • 96. • 16.2.2 Protocol deviations • 16.2.3 Patients excluded from the efficacy analysis • 16.2.4 Demographic data • 16.2.5 Compliance and/or drug concentration data (if available) • 16.2.6 Individual efficacy response data • 16.2.7 Adverse event listings (each patient) • 16.2.8 Listing of individual laboratory measurements by patient, when required by regulatory authorities • 16.3 Case Report Forms • 16.3.1 CRFs for deaths, other serious adverse events and withdrawals forAE • 16.3.2 Other CRFs submitted • 16.4 Individual Patient Data Listings (us Archival Listings)
  • 98. Annex II: Principal Or Coordinating Investigator(s) Signature(s) Or Sponsor's Responsible Medical Officer
  • 99. Annex IIIa: Study Design And Schedule Of Assessments
  • 100. Annex IIIb: Study Design And Schedule Of Assessments
  • 101. Annex IVa Disposition Of Patient
  • 102. Annex IVb Disposition Of Patient
  • 103. Annex V: Listing Of Patients Who Discontinued Therapy
  • 104. Annex VI: Listing Of Patients And Observations Excluded From Efficacy Analysis
  • 105. Annex VII: Number Of Patients Excluded From Efficacy Analysis
  • 106. Annex VIII: Guidance For Section 11.4.2 - Statistical/analytical Issues And Appendix 16.1.9 • Details of the statistical analysis performed on each primary efficacy variable should be presented in an appendix. • A. STATISTICAL CONSIDERATIONS • B. Format And Specifications For Submission Of Data Requested By Regulatory Authority's Statistical Reviewers
  • 107. Details reported should include at least the following information: a) The statistical model underlying the analysis. This should be presented precisely and completely, using references if necessary. b) A statement of the clinical claim tested in precise statistical terms, e.g., in terms of null and alternative hypotheses. c) The statistical methods applied to estimate effects, construct confidence intervals, etc. Literature references should be included where appropriate. d) The assumptions underlying the statistical methods. It should be shown, insofar as statistically reasonable, that the data satisfy crucial assumptions, especially when necessary to confirm the validity of an inference. e) The test statistic, the sampling distribution of the test statistic under the null hypothesis, the value of the test statistic, significance level (i.e. p-value), and intermediate summary data, in a format that enables the regulatory authority's statistical reviewer to verify the results of the analysis quickly and easily. The p-values should be designated as one- or two-tailed. The rationale for using a one-tailed test should be provided.
  • 108. Overview of current US regulations regarding clinical monitoring and safety reporting
  • 109. Objectives • Current context regarding safety in clinical trials • The concept of safety and safety monitoring and how it relates to clinical trials research • Protocol requirements pertaining to areas relevant to safety • Key roles and responsibilities related to safety • Safety and adverse event terminology • Expedited reporting of adverse events 109
  • 110. Objectives • Ensuring safety in clinical trials • The adverse event life cycle • What makes a well-documented adverse event, including a comprehensive narrative • How to assess an adverse event case, including causality assessment • Periodic Safety Report: DSUR, PSUR, PBRER, ASR, IB, Orphan Designation Annual Report 110
  • 111. Regulations: Federally Supported Research Involving Human Subjects 111 45 CFR 46: Protection of Human Research Subjects • Applies to all research involving human subjects • Institution must provide assurance of compliance, such as a Federal Wide Assurance (FWA) on file with the Office for Human Research Protection (OHRP) • FWA provides assurance that research is conducted in accordance with the regulations  Research reviewed and approved by IRB  Subject to continuing review by IRB
  • 112. Regulations: Non-Federally Supported Studies Involving Human Subjects 112 21 CFR 50: Protection of Human Subjects • Clinical investigations regulated by FDA • Requirements for informed consent • Elements of informed consent • Documentation of informed consent • Form approved by IRB 21 CFR 56: Institutional Review Boards • Clinical investigations regulated by FDA • Requirements for IRB review • Membership, functions, review procedures, etc • Criteria for IRB approval
  • 113. Current Safety Environment 113  Increasing demands for safety data: • All Serious Adverse Events (SAEs); Follow all AEs/SAEs till resolved or stable • Additional adverse events of interest (e.g. cancers, MIs, hepatic events) • Pregnancy outcomes • Food and Drug Administration Amendments Act of 2007 (FDAAA): Provides FDA with additional requirements, authorities, and resources with regard to both pre- and postmarket drug safety • Global reporting to EMEA and regulatory agencies of European Union (EU) member states – Use of CIOMS form – Country of origin of AE
  • 114. Clinical Trial Continuum: From Drug Development to Optimal Regimens to Treatment Strategies 114 PHASE I PHASE II PHASE III PHASE IV POST PHASE III/IV PRE-MARKET POSTMARKET CONTROLLED SETTING REAL-WORLD SETTING
  • 115. Safety Monitoring 115 Why is safety monitoring required in all clinical trials? To Ensure Subject Safety and Study Integrity
  • 116. Investigator assures Subject Safety and Study Integrity by: 116 Implementing the protocol “as written” Strict adherence to inclusion and exclusion criteria Monitoring subject status, i.e. subject wellbeing, minimization of risk, toxicity management, etc. Continued adherence throughout study duration Monitoring safety data collection: • Study database • Safety database Roles and Responsibilities – Site Investigator
  • 117. Roles and Responsibilities – Research Staff 117 • Record AE and/or SAE per protocol specifications • Follow protocol toxicity management section • Follow site SOP for emergencies • Follow site SOP to notify study clinician/physician • Record the AE/SAE Is immediate/emergency intervention needed? Yes No
  • 118. Roles and Responsibilities – Study Clinician/Physician 118 Subject reports AE Documentation •Follow until AE resolution or condition stabilizes Study clinician/physician will assess and manage the AE Decide if SAE Emergency intervention vs. Non-emergency care Research provisions vs. Clinical care
  • 119. Assurance of Safety and Well-Being: Research vs. Medical Roles 119  Emergency intervention vs. Non-emergency care • Acute on-site management, as necessary, and per site SOP • Referral to care when stable  Research provisions vs. Clinical care • Provide interventions permitted by the protocol • Follow protocol specifications for toxicity management • Beyond protocol specifications, refer out for clinical care
  • 120. Clinical Role vs. Research Role 120 Balancing Both Roles Balancing Both Roles Clinical Role: Subject OK Research Role: Study/Data OK • Is subject in imminent jeopardy? • Provide appropriate management commensurate with clinical situation, e.g. toxicity management • Provide appropriate referral: emergent care or back to regular care • Follow up with subject status Not Subject’s Primary Clinician • Identification of adverse event • Immediate notification necessary? To whom? [per protocol and safety monitoring plans] • Complete documentation of adverse event. Follow until resolution/stability including updating records • Determine if AE meets criteria for SAE • Adhere to reporting requirements • Adhere to toxicity management as specified • Adhere to stopping rules as specified
  • 121. Therapeutic Misconception 121 • Subjects think they are receiving proven interventions, per their usual clinical care, despite participating in a research study • Informed Consent Process must not be trivialized or relegated to administrative status • Check for understanding • Time for questions, making decision • Physicians think they can provide interventions, per usual practice • Strict adherence to protocol provisions for care, toxicity management • Decide if subject can continue in study
  • 122. Roles and Responsibilities – Study Clinician/Physician 122 Study product: Per site, per study? Study status: Safety pause, clinical hold, early termination? Study product: Dose held, changed, or discontinued? Study participation: Continue, withdraw? Action taken with Study product after AE Study Study
  • 123. Roles and Responsibilities – Study Team 123 Safety: Ensure safety and well being of subjects at all times • Monitor safety across all study sites • Review all safety data at specified intervals • Discuss need for change(s) driven by safety Data: Ensure data integrity to assess the risks/safety profile of the study intervention • Data capture; especially safety data • Be cognizant of expedited reporting requirements for safety data
  • 124. Roles and Responsibilities – Study Team vs. Sponsor/sponsor 124 • Safety monitoring by study team • Acute on-site management and discussion with study team • Periodic review by study team and monitoring committees • Data generated by Data Management Centers (DMC) • Expedited reporting to sponsor • SAE sent to sponsor • Sponsor is not part of discussions that occur within study/safety monitoring teams regarding the event • The sponsor only has information about the event from the SAE Form; site should include relevant information from study team discussions • sponsor processes event and sends queries to site to obtain additional information • All follow-up information should be provided to sponsor
  • 125. Safety Monitoring Environment 125 CTA/IND Trials Pre-market Postmarket 45 CFR 46 45 CFR 46 FDA 21 CFR Part 312 - IND 21 CFR 312.32 (IND Safety Reports) 21 CFR 312.33 (Annual Reports) 21 CFR 812.150 (IDE Reports) 21 CFR Part 314 - NDA 21 CFR 314.80 (Postmarketing) 21 CFR 314.98 (Generics) 21 CFR 600.80 (Biologics) 21 CFR 803 (Medical Devices) ICH E2A (Oct 1994) ICH E2D (Nov 2003) NIH Policy NIH Policy Country/State Regulations Country/State Regulations IRBs/ECs IRBs/ECs Sponsor Sponsor
  • 126. ICH: E Documents on Safety Clinical Safety • ICH E1 – The Extent of Population Exposure to Assess Clinical Safety for Drugs Intended for Long-Term Treatment of Non-Life Threatening Conditions • ICH E2A – Clinical Safety Data Management: Definitions and Standards for Expedited Reporting • ICH E2B – Clinical Safety Data Management: Data Elements for Transmission of Individual Case Safety Reports • ICH E2C – Clinical Safety Data Management: Periodic Safety Update Reports for Marketed Drugs • ICH E2D – Post-Approval Safety Data Management: Definitions and Standards for Expedited Reporting • ICH E2E – Pharmacovigilance Planning • ICH E2F – Development Safety Update Report Good Clinical Practice • ICH E6 – Good Clinical Practice 126 http://www.ich.org/cache/compo/276-254-1.html
  • 127. Subject Enrolled AE Reported Record AE* Yes SAE? To Sponsor Record SAE** To IRB To FDA To other Follow until Resolution or Stability Outcome: Resolved/ Stable? Update SAE No Adverse Event Flowchart 127 Subject Enrolled AE Reported Record AE* Yes SAE? To Sponsor Record SAE** To IRB To FDA To other Follow until Resolution or Stability Outcome: Resolved/ Stable? Update SAE No
  • 128. Adverse Event 128 * Protocol specifications for AE • When to collect e.g., study visit • Method of collection e.g., in person, telephone call • What to collect e.g., all AEs, only certain AEs by body system, only certain AEs by severity • What forms to use e.g. AE CRF, study CRFs ** Protocol specifications for SAE • Criteria • Expedited time frames • Reporting form (e.g. SAE)
  • 129. Documentation Differences Between AE CRF and SAE Form Record on SAE Form (includes narrative) 129 Record in source document Record on AE case report form Yes Attach additional documentation Does AE meet SAE criteria?
  • 130. Documentation Differences Between AE CRF and SAE Form: Data Elements AE CRF Data Elements • AE • Start Date • Stop Date / Continuing • Is it SAE? • Severity • Relatedness • Action taken with Study Agent • Outcome (study participation) 130 SAE Form Data Elements • Participant Identifiers • Study Agent details • Narrative • Past medical history • Relevant labs, tests, procedures • Concomitant meds • Outcome of SAE • Other supporting information
  • 131. Safety Data from Clinical Trials Obligations to report safety data (IND or Non-IND studies): • Data for non-expedited reporting: • Recorded on AE CRF, goes to clinical trial database • Data for expedited reporting: • Recorded on AE CRF and linked to an SAE type form • Goes to safety database • IND timelines: 24o to sponsor, 7/15 days to FDA, EMEA • Post-marketing timelines: depends on sponsor, 15 days to FDA, EMEA • Annual/Periodic Reports : • Need safety data from clinical and safety database • Must be reconciled 131
  • 132. Adverse Event Any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. (ICH E2A) 132
  • 133. Adverse Event Term • The AE should best describe what the subject says (i.e. verbatim description) • Can be extracted from medical records • Can incorporate medical assessment (including a diagnosis if available) • The more accurate the AE term, the more accurate the safety database. The AE terms will be coded using a standard dictionary, e.g. Medical Dictionary for Regulatory Activities (MedDRA); this is NOT site responsibility 133
  • 134. AE Term - Examples 134 • If “anaphylactic reaction” is associated with “rash, dyspnea, hypotension, and laryngospasm,” report primary AE as “anaphylactic reaction.” • If “myocardial infarction” is associated with “chest pain, dyspnea, diaphoresis, ECG changes and jaundice,” report “myocardial infarction” and “jaundice” as separate primary AEs.
  • 135. Serious Adverse Event (SAE) A serious adverse event (experience) or reaction is any untoward medical occurrence that at any dose: • Results in death, • Is life-threatening • Requires inpatient hospitalization or prolongation of existing hospitalization • Results in persistent or significant disability/incapacity • Is a congenital anomaly/birth defect • In addition, “…important medical events that may not be immediately life- threatening or result in death or hospitalization but may jeopardize the patient or may require intervention to prevent one of the other outcomes listed in the definition above…should also usually be considered serious.” (ICH E2A) 135
  • 136. Adverse Event vs. Event Outcome Hospitalization • Hospitalization is a consequence and is not usually considered an AE. • Example: If the subject was hospitalized due to congestive heart failure, “congestive heart failure” is the primary AE and hospitalization is the outcome. • If the only information available is that the study subject was hospitalized, “hospitalization” can be reported. 136
  • 137. Hospitalization Hospitalization in the absence of a medical AE is not in itself an AE and does not need to be reported in an expedited time frame, such as: • Admission for treatment of a pre-existing condition (can include target disease) not associated with the development of a new AE or with a worsening of the pre-existing condition • Diagnostic admission (e.g. for work-up of persistent existing condition such as pre-treatment lab abnormality) • Protocol-specified admission (e.g. procedure required by study protocol) • Administrative admission (e.g. for yearly physical exam) • Social admission (e.g. study subject has no place to sleep) • Elective admission (e.g. elective surgery) 137
  • 138. Severity • Describes the intensity of the event • Events are graded on a severity scale • Mild, Moderate, Severe • Numeric Scale e.g. 1 to 5 • Severity grading must match the clinical picture • Presenting AE is Grade 1 • AE progressed to SAE (hospitalization) • The expedited report should have the grade of the SAE, not the AE 138
  • 139. Seriousness is NOT the same as Severity 139 Based on the intensity of the AE and is not a factor in determining reportability (clinical descriptor) Based on outcome of the AE and is a factor in determining reportability (regulatory definition) Seriousness Severity Determined using the SAE criteria Determined using the sponsor grading table
  • 140. Action Taken with Drug • Action Taken with Drug: • Withdrawn • Dose reduced • Dose increased • Dose not changed • Unknown • Not applicable > ICH E2B (R3) • Refer to protocol • Refer to DAERS 140
  • 141. Outcome • Outcome of reaction/event at the time of last observation • Recovered/resolved • Recovering/resolving • Not recovered/not resolved • Recovered/resolved with sequelae • Fatal • Unknown > ICH E2B (R3) • Outcome of subject in study • Remains in Study • Withdrawn • Lost to follow-up • Death 141
  • 142. Expectedness • Pertains to whether an event is expected or unexpected (on the basis of previous observation, not what might be anticipated from the pharmacological properties of the product) • Unexpected: the nature or severity of the adverse event is not consistent with the applicable product information (e.g. Investigator’s Brochure for unapproved product, Package Insert for approved product) 142
  • 143. Relatedness (Causality) • No standard international nomenclature • Conveys that a “causal relationship” between the study product and the adverse event is “at least a reasonable possibility” [ICH E2A] • Facts (evidence) exist to suggest the relationship • Information on SAEs generally incomplete when first received • Follow-up information actively pursued • Judged by: • Reporting health professional • Sponsor 143
  • 144. Determination of Causality • Standard determinations include: • Is there [Drug Exposure] and [Temporal Association]? • Is there [Dechallenge/Rechallenge] or [Dose Adjustments]? • Any known association per [Investigator’s Brochure] or [Package Insert]? • Is there [Biological Plausibility]? • Any other possible [Etiology]? 144
  • 145. • ‘May be more art than science’ • NR: evidence for alternate etiology and/or low or no biologic plausibility • R: reasonable possibility; facts or evidence to substantiate relationship, and biologic plausibility • Is there evidence to compel change in previous conclusions? • Clear-cut case; easy to make a determination • Not so clear-cut: use your best judgment based on available information; assure adequacy of information • Unless clear-cut case, there’s no absolute right or wrong; give your best judgment; substantiate and follow-up • Err on conservative side 145 Determination of Causality
  • 146. 146 • Comprehensive, stand-alone “medical story” • Written in logical time sequence • Include key information from supplementary records • Include relevant autopsy or post-mortem findings • Summarize all relevant clinical and related information, including: • Study subject characteristics • Therapy details • Medical history • Clinical course of the event(s) • Diagnosis (workup, relevant tests/procedures, lab results) • Other information that supports or refutes an AE • > ICH E2D Narrative
  • 147. Narrative Template 147 • This is a [Age] year old [Race] [Male/Female] in [Study] who reported [Primary AE] on [Date of AE]. Enrolled into study on [Date Enrolled], Study medication was started on [Date], which is [Study Day _/Week _], taken for [Duration]. The event occurred during the [Treatment/Follow-up Phase]. • If fetus: provide [Gestational Age], (or mother’s LMP), at time of event. Also, [Gestational Age/Trimester] at first drug exposure and duration of exposure. If birth, provide details of [Infant Status] at birth. If hospital stay is complicated, provide details of hospital stay. • Provide details of the [AE] in chronological order, along with other [Signs/Symptoms]. Provide details of [Physical Exam], along with all relevant [Procedures] and [Lab Results].
  • 148. Narrative Template 148 • Provide details of [Treatment] and [Treatment Rationale] on basis of [Findings/Test Result(s)]. Describe [Treatment Response]. • If hospitalization, provide [Dates Hospitalization], describe relevant [Hospital Course], [Diagnostic Work-up], [Procedures/Tests and Results], [Treatment], [Treatment Response]. • Provide [Discharge Diagnosis], and any [Follow-up Information]. List [Discharge Meds]. • Provide pertinent [Past Medical Hx], [Family Hx], [Concomitant Meds], [Alcohol/Tobacco/Substance Use] and any previous similar [AEs].
  • 149. Review and Assessment of SAE 149 • Assemble all information available and use medical judgment • Standard for each AE: • Select [Seriousness Criteria] • Grade [Severity] per sponsor Toxicity Table • Specify [Actions Taken on Study Product] • Specify [Outcome of SAE]. If Outcome is not resolved at time of evaluation, follow until resolution or stability at each study visit • Is it [Expected]? • Is it [Related]?
  • 150. 150 • Sponsor role: (ICH E2D) • Information about the case should be collected from the healthcare professionals who are directly involved in the study subject’s care • Clearly identified evaluations by the sponsor are considered appropriate and are required by some regulatory authorities • Opportunity to render another opinion; may be in disagreement with; and/or provide another alternative to the diagnosis/assessment given by initial reporter • Sponsor makes an assessment of causality (attribution) just as the PI makes an assessment of causality (attribution) • If causality (attribution) is different between the sponsor and the investigator, both assessments are reported Clinical Case Evaluation
  • 151. Site vs. Sponsor Assessment 151 Balancing Both RolesSite Assessment Sponsor Assessment • Site advantage: has access to subject; may elicit further info, perform PE, obtain tests, labs, records • Information from self-report (may lack validation) • Know subject best • Judgment stands • Open to dialog with sponsor • Information limited to what was submitted from site • May initiate queries to site: incur time and delay • Constraint: Must adhere to reporting timelines to FDA • Serious? Unexpected? Related? • sign-off, agree with Site PI, agree with sponsor. Any critical flaw in reasoning? • Open to dialog with Site PI, sponsor
  • 152. Periodic Safety Report: DSUR, PBRER (replacing PSUR), ASR, IB, Orphan Designation Annual Report • During the drug development and after the drug is on the market, the sponsor or market authorization holder has obligations to submit the safety information to regulatory agencies periodically. • These periodic reports have different requirements and sometimes are confusing. While these reports may be prepared by the regulatory affairs department or pharmacovigilence department, biostatistics group may often be asked to provide the information for these periodic reports.
  • 153. Periodic Safety Report: DSUR, PBRER (replacing PSUR), ASR, IB, Orphan Designation Annual Report • These reports and their abbreviations could be very confusing especially for those who are not working in the pharmacovigilence department: DSUR, IND Annual Report, ASR, IB, PSUR, and PBRER
  • 154. Types of Safety Reports If we take a close look at these reports, they may be considered as four types: • PSUR (Old report replaced by PBRER) • PBRER (replacing PSUR) for drugs already on the market • DSUR (replacing IND Annual Report and Annual Safety Report) for drugs under developments • IB is required whenever there is a clinical trial • Orphan Designation Annual Report is required for the developing product with orphan designation - for rare disease
  • 155. PSUR: Periodic Safety Update Reports • The term PSUR comes from the previous ICH E2C (R1) “Clinical Safety Data Management: Periodic Safety Update Reports for Marketed Drugs”. ICH guideline E2C has been subsequently revised and renames as Periodic Benefit-Risk Evaluation Report (PBRER).
  • 156. PBRER: Periodic Benefit-Risk Evaluation Report • According to the ICH E2C (R2) “PERIODIC BENEFIT-RISK EVALUATION REPORT (PBRER)”, • The Periodic Benefit-Risk Evaluation Report (PBRER) described in this Guideline is intended to be a common standard for periodic benefit-risk evaluation reporting on marketed products (including approved drugs that are under further study) among the ICH regions.
  • 157. PBRER: Periodic Benefit-Risk Evaluation Report • Other supporting documents regarding PBRER are: • Concept Paper • Business Plan • Contribute to the E2C(R2) IWG
  • 158. PBRER: Periodic Benefit-Risk Evaluation Report • E2C (R2) is finalized at November 2012 and is supposed to replace the PSUR. However, EMA has not fully adopted the PBRER and continues to use the term PSUR as defined in its recent guideline “Guideline on good pharmacovigilance practices (GVP) 4 Module VII – Periodic safety update report (Rev 1)”
  • 159. PBRER: Periodic Benefit-Risk Evaluation Report • FDA fully endorsed E2C (R2) and PBRER and issued its guidance “Providing Postmarket Periodic Safety Reports in the ICH E2C(R2) Format (Periodic Benefit-Risk Evaluation Report”
  • 160. DSUR: Development Safety Update Report • According to ICH E2F “Development Safety Update Report”, • The Development Safety Update Report (DSUR) proposed in this guideline is intended to be a common standard for periodic reporting on drugs under development (including marketed drugs that are under further study) among the ICH regions.
  • 161. • US and EU regulators consider that the DSUR, submitted annually, would meet national and regional requirements currently met by the US IND Annual Report and the EU Annual Safety Report, respectively, and can therefore take the place of these existing reports • ICH E2F Guideline is finalized in August 2010 and is replacing the previous IND (investigational new drug) Annual Report (in US) and Annual Safety Report (in EU).
  • 162. Documents regarding DSUR can be found at ICH.org website We will cover the following: • Concept Paper • Business Plan • DSUR Examples Commercial Sponsors • DSUR Examples non-Commercial Sponsors • Presentation on E2F
  • 163. IB:Investigator Brochure • According to ICH E6 “Good Clinical Practice”, • The Investigator's Brochure (IB) is a compilation of the clinical and nonclinical data on the investigational product(s) that are relevant to the study of the product(s) in human subjects.
  • 164. IB:Investigator Brochure • Its purpose is to provide the investigators and others involved in the trial with the information to facilitate their understanding of the rationale for, and their compliance with, many key features of the protocol, such as the dose, dose frequency/interval, methods of administration, and safety monitoring procedures. The IB also provides insight to support the clinical management of the study subjects during the course of the clinical trial.
  • 165. IB:Investigator Brochure • For post marketing commitment clinical trials, the product label (package insert) may be used in place of the investigator brochure since the package insert includes the contents required in the investigator brochure.
  • 166. Orphan Drug Designation Annual Report • 21CFR Part 316 (Orphan Drugs) contains the specific section (section 316.30) requiring the annual reports of holder of orphan drug designation: • § 316.30 Annual reports of holder of orphan- drug designation.
  • 167. Orphan Drug Designation Annual Report • Within 14 months after the date on which a drug was designated as an orphan drug and annually thereafter until marketing approval, the sponsor of a designated drug shall submit a brief progress report to the FDA Office of Orphan Products Development on the drug.
  • 168. Orphan Drug Designation Annual Report includes: • (a) A short account of the progress of drug development including a review of preclinical and clinical studies initiated, ongoing, and completed and a short summary of the status or results of such studies. (b) A description of the investigational plan for the coming year, as well as any anticipated difficulties in development, testing, and marketing; and (c) A brief discussion of any changes that may affect the orphan-drug status of the product. For example, for products nearing the end of the approval process, sponsors should discuss any disparity between the probable marketing indication and the designated indication as related to the need for an amendment to the orphan-drug designation pursuant to § 316.26.
  • 169. • EU has the similar requirement and sponsors are required to submit to the European Medicines Agency (EMA) every year after their medicine has been granted orphan designation. See EMA Orphan Designation Annual Report.