Investigational New Drug Safety Reporting Requirements for Human Drug and Biological Products and Safety Reporting Requirements for Bioavailability and Bioequivalence Studies in Humans, 59935-59963 [2010-24296]
Download as PDF
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
Subtitle VII, Aviation Programs,
describes in more detail the scope of the
agency’s authority.
This rulemaking is promulgated
under the authority described in
Subtitle VII, Part A. Subpart I, Section
40103. Under that section, the FAA is
charged with prescribing regulations to
assign the use of airspace necessary to
ensure the safety of aircraft and the
efficient use of airspace. This regulation
is within the scope of that authority as
it amends controlled airspace at
Smithfield, NC.
Issued in College Park, Georgia, on
September 17, 2010.
Myron A. Jenkins,
Acting Manager, Operations Support Group,
Eastern Service Center, Air Traffic
Organization.
Lists of Subjects in 14 CFR Part 71
21 CFR Parts 312 and 320
Airspace, Incorporation by reference,
Navigation (Air).
Adoption of the Amendment
[FR Doc. 2010–24113 Filed 9–28–10; 8:45 am]
BILLING CODE 4910–13–P
Food and Drug Administration
[Docket No. FDA–2000–N–0108] (formerly
Docket No. 00N–1484)
RIN 0910–AG13
PART 71—DESIGNATION OF CLASS A,
B, C, D, AND E AIRSPACE AREAS; AIR
TRAFFIC SERVICE ROUTES; AND
REPORTING POINTS
Investigational New Drug Safety
Reporting Requirements for Human
Drug and Biological Products and
Safety Reporting Requirements for
Bioavailability and Bioequivalence
Studies in Humans
AGENCY:
1. The authority citation for Part 71
continues to read as follows:
■
§ 71.1
[Amended]
2. The incorporation by reference in
14 CFR 71.1 of Federal Aviation
Administration Order 7400.9U,
Airspace Designations and Reporting
Points, dated August 18, 2010, and
effective September 15, 2010, is
amended as follows:
■
Paragraph 6005 Class E Airspace Extending
Upward From 700 feet or More Above the
Surface of the Earth
*
*
*
erowe on DSK5CLS3C1PROD with RULES
ASO NC E5
*
*
Smithfield, NC [Amended]
Johnston County Airport, NC
(Lat. 35°32′27″ N., long 78°23′25″ W.)
Johnston Memorial Hospital
Point In Space Coordinates
(Lat. 35°31′23″ N., long 78°20′35″ W.)
That airspace extending upward from 700
feet above the surface within a 6.5-mile
radius of the Johnston County Airport and
within 2 miles each side of the 023° bearing
from the airport extending from the 6.5-mile
radius to 10.2 miles northeast of the Johnston
County Airport and within a 6-mile radius of
the point in space (lat.35°31′23″ N., long.
78°20′35″ W.) serving Johnston Memorial
Hospital.
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
Food and Drug Administration,
HHS.
ACTION:
Final rule.
The Food and Drug
Administration (FDA) is amending its
regulations governing safety reporting
requirements for human drug and
biological products subject to an
investigational new drug application
(IND). The final rule codifies the
agency’s expectations for timely review,
evaluation, and submission of relevant
and useful safety information and
implements internationally harmonized
definitions and reporting standards. The
revisions will improve the utility of IND
safety reports, reduce the number of
reports that do not contribute in a
meaningful way to the developing safety
profile of the drug, expedite FDA’s
review of critical safety information,
better protect human subjects enrolled
in clinical trials, subject bioavailability
and bioequivalence studies to safety
reporting requirements, promote a
consistent approach to safety reporting
internationally, and enable the agency
to better protect and promote public
health.
DATES: This rule is effective March 28,
2011.
FOR FURTHER INFORMATION CONTACT:
For information on IND safety
reporting for human drug products:
Janet Norden, Center for Drug
Evaluation and Research, Food and
Drug Administration, 10903 New
Hampshire Ave., Bldg. 51, rm. 6324,
Silver Spring, MD 20993–0002, 301–
796–2500.
SUMMARY:
Authority: 49 U.S.C. 106(g); 40103, 40113,
40120; E.O. 10854, 24 FR 9565, 3 CFR,
1959–1963 Comp., p. 389.
For information on IND safety
reporting for human biological products:
Laura Rich, Center for Biologics
Evaluation and Research, Food and
Drug Administration,1401 Rockville
Pike, suite 200N, Rockville, MD 20852–
1448, 301–827–6210.
SUPPLEMENTARY INFORMATION:
Table of Contents
DEPARTMENT OF HEALTH AND
HUMAN SERVICES
In consideration of the foregoing, the
Federal Aviation Administration
amends 14 CFR Part 71 as follows:
■
59935
PO 00000
Frm 00003
Fmt 4700
Sfmt 4700
I. Background
A. Rationale for Rulemaking
B. The Proposed Rule
II. Overview of the Final Rule
A. Definitions
B. Review of Safety Information
C. Reporting Requirements
III. Comments on the Proposed Rule
A. Definitions—Proposed § 312.32(a)
B. Review of Safety Information—
Proposed § 312.32(b)
C. IND Safety Reports (Requirement
for Minimum Data Set)—Proposed
§ 312.32(c)
D. Serious and Unexpected SADR—
Proposed § 312.32(c)(1)(i)
E. Alternative Reporting
Arrangements
F. Unblinding
G. Information Sufficient to Consider
Product Administration Changes—
Proposed § 312.32(c)(1)(ii)
H. Submission of Written Reports—
Proposed § 312.32(c)(1)(iii)
I. Telephone and Facsimile
Transmission Safety Reports—
Proposed § 312.32(c)(2)
J. Investigations of Marketed Drugs—
Proposed § 312.32(c)(4)
K. Followup—Proposed § 312.32(d)
L. Disclaimer—Proposed § 312.32(e)
M. Annual Reports
N. Investigator Reports—Proposed
§ 312.64(b)
O. Bioavailability and Bioequivalence
Requirements—Proposed
§ 320.31(d)
P. Reports to Investigators and IRBs
Q. Miscellaneous Comments
R. Initial Analysis of Impacts and
Paperwork Burden Estimates
IV. Legal Authority
V. Environmental Impact
VI. Analysis of Impacts
A. Need for the Regulation
B. Costs of the Regulation (to Prepare
and Submit Safety Reports)
C. Benefits of the Regulation
D. Final Regulatory Flexibility
Analysis
VII. Paperwork Reduction Act of 1995
VIII. Executive Order 13132: Federalism
IX. References
I. Background
In the Federal Register of March 14,
2003 (68 FR 12406), FDA issued a
proposed rule to revise its regulations
governing pre- and postmarketing safety
E:\FR\FM\29SER1.SGM
29SER1
59936
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
reporting for human drug and biological
products1, which appear in parts 310,
312, 314, 320, 600, 601, and 606 (21
CFR parts 310, 312, 314, 320, 600, 601,
and 606). The proposed revisions
represented a major effort to clarify and
integrate several safety reporting rules
and guidance documents that had been
issued by international organizations
and by FDA dating back to the 1990s.
The background for and description of
these regulations and guidance
documents are described in the
preamble of the proposed rule (68 FR
12406 at 12407 to 12410, Figure 1). The
proposal called for the submission of
comments by July 14, 2003. At the
request of industry, and to provide all
interested persons additional time to
comment, the comment period was
extended until October 14, 2003 (68 FR
36527, June 18, 2003).
FDA received numerous comments in
response to the proposed rule, many of
which stated that the proposal would
not meet its stated goals and requested
that the agency reevaluate specific
aspects of the proposal. FDA agreed
with some of these comments and has
reevaluated and revised aspects of the
proposal. To make the rulemaking
process more manageable, FDA has
decided to issue revisions to the
premarketing and postmarketing safety
reporting regulations in two separate
rulemakings. By separating these rules,
the agency has been able to reevaluate
and refine each requirement in the
premarketing and postmarketing
settings to better ensure that the rules
will achieve their goals.
This rule finalizes revisions to the
IND safety reporting regulations found
in part 312 and the safety reporting
requirements for bioavailability and
bioequivalence studies found in part
320. The agency is working on revisions
to the postmarketing safety reporting
regulations found in parts 310, 314, 600,
601, and 606 separately, and will
address these sections in a future rule.
Therefore, revisions to and comments
about postmarketing safety reporting
requirements found in parts 310, 314,
600, 601, and 606 are not addressed in
this rulemaking. This document
discusses information relevant to and
comments about the proposed revisions
found in parts 312 and 320.
erowe on DSK5CLS3C1PROD with RULES
A. Rationale for Rulemaking
In the proposed rule (68 FR 12406 at
12412 to 12415), FDA described its
goals for the proposed rulemaking.
1 For the purposes of this document, unless
otherwise specified, all references to ‘‘drugs’’ or
‘‘drug products’’ include human drug products and
biological products that are also drugs.
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
Many of the stated goals were primarily
applicable to postmarketing safety
reporting, but revising and clarifying the
IND safety reporting requirements was
also a critical component of FDA’s
stated efforts to: (1) Improve the overall
quality of safety reporting, thereby
strengthening the agency’s ability to
review critical safety information, (2)
monitor the safety of human drug and
biological products, and (3) harmonize
safety reporting internationally. Each of
these is discussed in turn in this
document.
First, the revisions to the IND safety
reporting requirements will improve the
overall quality of safety reporting and
the agency’s ability to review critical
safety information by ensuring that the
information that FDA receives in an IND
safety report is relevant and useful.
Under former regulations, there may
have been over-reporting of serious
adverse events for which there was little
reason to believe that the drug had
caused the event, complicating or
delaying FDA’s ability to detect a safety
signal. In this final rule, FDA clarifies
definitions, provides examples of the
types of evidence that suggest a causal
relationship for purposes of reporting a
suspected adverse reaction to the IND
and participating investigators, and
revises the requirements for expedited
reporting of serious and unexpected
suspected adverse reactions to the IND.
The final rule also allows sponsors to
arrange alternative formats and/or
frequencies for reporting and provides
that study endpoints must not be
submitted as IND safety reports except
in unusual cases. These revisions not
only have an impact on which reports
are sent to FDA and participating
investigators, but also affect the reports
that are sent by investigators to
Institutional Review Boards (IRBs).
These revisions and clarifications will
minimize reports that do not contribute
to FDA’s understanding of the
developing safety profile of the drug and
decrease the number of uninterpretable
reports (so-called ‘‘noise’’) in the system.
In addition, the revisions and
clarifications will help to make clear
under what circumstances the study
blind should be broken and when
unblinding is unnecessary. Ultimately,
these revisions and clarifications should
contribute toward more useful adverse
reaction information and more effective
monitoring of clinical trials.
Second, by requiring expedited
reports of certain safety information that
was not reported expeditiously under
former IND safety reporting
requirements or bioavailability or
bioequivalence requirements, the final
rule will help FDA monitor the safety of
PO 00000
Frm 00004
Fmt 4700
Sfmt 4700
human drug and biological products
and better protect human subjects
enrolled in clinical trials. Under the
final rule, FDA will receive expedited
reports of:
• Findings from clinical studies,
epidemiological studies or pooled
analyses of multiple studies that suggest
a significant risk in humans exposed to
the drug,
• Serious suspected adverse reactions
that occur at an increased rate than
listed in the protocol or investigator
brochure, and
• Serious adverse events from
bioavailability and bioequivalence
studies.
By receiving these reports
expeditiously, FDA will be better able to
monitor and evaluate the drug’s safety.
Finally, FDA had proposed certain
revisions to its IND safety reporting
requirements to harmonize the
regulations with recommendations by
the International Conference on
Harmonisation of Technical
Requirements for Registration of
Pharmaceuticals for Human Use (ICH)
and by the World Health Organization’s
Council for International Organizations
of Medical Sciences (CIOMS), and
which have been adopted by the
European Union (EU) (Ref. 1). In the
preamble to the proposed rule (68 FR
12406 at 12415, table 4), FDA detailed
the specific proposed revisions to the
definitions and reporting standards
based on international
recommendations in the ICH guidance
‘‘E2A Clinical Safety Data Management:
Definitions and Standards for Expedited
Reporting’’ (60 FR 11284, March 1, 1995)
(ICH E2A guidance). FDA received
numerous comments, described in more
detail in section III of this document,
stating that certain of FDA’s proposed
revisions were inconsistent with how
the provisions are interpreted and
implemented in other member ICH
nations. After reviewing the comments
and after discussions with our ICH
partners, FDA has revised the
definitions and reporting standards to
be as consistent as possible with
international definitions and standards,
recognizing that there may be
inconsistencies within ICH documents
and among the other member ICH
nations’ interpretations of these
definitions and standards.
B. The Proposed Rule
The following describes the proposed
revisions to the requirements in parts
312 and 320. FDA proposed the
following revisions to § 312.32 on IND
safety reports:
• Replace the defined phrase
‘‘associated with the use of the drug’’
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
with the term ‘‘suspected adverse drug
reaction (SADR),’’
• Require submission of expedited
reports of ‘‘information sufficient to
consider product administration
changes,’’
• Make it clear that safety reports of
overall findings or data in the aggregate
must be submitted in a narrative format,
• Permit the determination that an
SADR is life-threatening to be based on
the opinion of either the investigator or
sponsor (as opposed to only the
investigator),
• Require that the sponsor notify FDA
and all participating investigators of
each SADR that is both serious and
unexpected, based on the opinion of
either the investigator or sponsor (as
opposed to only the sponsor),
• Require a ‘‘‘‘minimum data set’’ for
each report of an SADR submitted to
FDA, and
• Clarify the sources of information
that sponsors must review for safety
surveillance and reporting purposes.
FDA proposed the following revision
to § 312.64(b):
• Make it clear that the investigator
must report to the sponsor any serious
SADR immediately and any other SADR
promptly, unless otherwise specified in
the protocol or investigator’s brochure.
FDA proposed the following revision
to § 320.31(d):
• Make bioavailability and
bioequivalence studies subject to IND
safety reporting requirements.
II. Overview of the Final Rule
This final rule amends parts 312 and
320 of FDA regulations by revising the
requirements for IND safety reporting
and for bioavailability and
bioequivalence studies. This final rule
reflects revisions the agency made in
response to comments on the March
2003 proposal (addressed in detail in
section III of this document) and other
revisions, including editorial changes to
clarify provisions and support the
agency’s plain language initiative
(addressed in this section).
erowe on DSK5CLS3C1PROD with RULES
A. Definitions
The definitions section for the IND
safety reporting regulations (§ 312.32(a))
now includes the following five terms:
• Adverse event,
• Life-threatening adverse event or
life-threatening suspected adverse
reaction,
• Serious adverse event or serious
suspected adverse reaction,
• Suspected adverse reaction, and
• Unexpected adverse event or
unexpected suspected adverse reaction.
FDA has revised and clarified terms
and definitions that were in the
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
proposed rule. First, as discussed in
detail in section III of this document,
the two terms ‘‘adverse event’’ and
‘‘suspected adverse reaction’’ replace the
proposed definition of ‘‘suspected
adverse drug reaction (SADR).’’ The
definitions ‘‘adverse event’’ and
‘‘suspected adverse reaction’’ also
replace the phrase ‘‘associated with the
use of the drug’’ defined in former
§ 312.32(a). The definitions of the terms
‘‘adverse event’’ and ‘‘suspected adverse
reaction’’ make clear a distinction in the
degree of evidence of a causal
relationship between the drug and the
adverse event within these terms.
Second, the final rule requires that the
determination for reporting purposes
about whether an adverse event or
suspected adverse reaction is ‘‘lifethreatening’’ or ‘‘serious’’ be based on the
opinion of either the investigator or
sponsor. FDA had proposed this
revision for the definition of ‘‘lifethreatening SADRs,’’ and the agency
decided that the determination about
whether an adverse event or suspected
adverse reaction is ‘‘serious’’ is
comparable to the determination of
whether it is life-threatening. Therefore,
FDA revised the definition ‘‘serious
adverse event or serious suspected
adverse reaction’’ to specify that the
determination of seriousness be based
on the opinion of either the investigator
or sponsor. In addition, FDA eliminated
the definition of ‘‘disability’’ as a
separate term and includes the meaning
of the term in the definition of ‘‘serious
adverse event or serious suspected
adverse reaction.’’
Third, the final rule makes clear what
adverse events or suspected adverse
reactions are considered unexpected.
The proposed definition of ‘‘unexpected
SADR’’ included the following sentence
from the then-current definition for
‘‘unexpected adverse drug experience’’
(with minor clarification): ‘‘‘Unexpected’
as used in this definition, refers to an
SADR that has not been previously
observed (e.g., in the investigator
brochure); it does not refer to an SADR
that might be anticipated from the
pharmacological properties of the drug
product.’’ To this clarification, FDA
proposed to add the following new
sentence: ‘‘SADRs that are mentioned in
the investigator’s brochure as occurring
with a class of drugs but not specifically
mentioned as occurring with the
particular drug are considered
unexpected.’’ In this final rule, FDA
combined these proposed sentences to
read as follows: ‘‘‘Unexpected,’ as used
in this definition, also refers to adverse
events or suspected adverse reactions
that are mentioned in the investigator
brochure as occurring with a class of
PO 00000
Frm 00005
Fmt 4700
Sfmt 4700
59937
drugs or as anticipated from the
pharmacological properties of the drug,
but are not specifically mentioned as
occurring with the particular drug under
investigation.’’ This revision makes clear
that adverse events that have not been
previously observed with the drug
under investigation, but are predicted to
occur based on the class of the drug or
pharmacological properties of the drug
are considered ‘‘unexpected’’ for
reporting purposes.
B. Review of Safety Information
The final rule clarifies what safety
information must be reviewed under
§ 312.32(b). The proposal would have
required sponsors to review ‘‘reports
from foreign regulatory authorities that
have not been previously reported to
FDA by the sponsor.’’ FDA has deleted
the phrase ‘‘that have not been
previously reported to FDA by the
sponsor,’’ because it confuses the review
with the reporting requirements. FDA
expects sponsors to review all
information, but to avoid duplicate
reporting to the agency. In addition, the
final rule clarifies the agency’s
expectations for analysis of previous,
similar reports (§ 312.32(c)(1)).
C. Reporting Requirements
In § 312.32(c), the final rule clarifies
how and when to submit IND safety
reports to FDA and participating
investigators, including the requirement
in § 312.32(c)(1)(v) that certain reports
be submitted in a narrative format
(proposed § 312.32(c)(1)(iii)). It provides
examples of the kinds of evidence that
suggest a causal relationship between
the drug and the adverse event when
determining whether a serious and
unexpected adverse event qualifies for
expedited reporting (§ 312.32(c)(1)(i)).
The final rule also requires that
sponsors submit expedited reports of
findings from clinical studies,
epidemiological studies, or pooled
analyses of multiple studies that suggest
a significant risk in humans
(§ 312.32(c)(1)(ii)); findings from animal
or in vitro testing that suggests a
significant risk in humans
(§ 312.32(c)(1)(iii)); and reports of an
increased rate of occurrence of serious
suspected adverse reactions over that
listed in the protocol or investigator
brochure (§ 312.32(c)(1))(iv)). The final
rule also provides for alternative
reporting arrangements (§ 312.32(c)(3))
and provides that study endpoints not
be reported except in unusual cases
(§ 312.32(c)(5)).
Furthermore, FDA has made it clear
in § 312.32(c)(1)(v) that the period of
time for submitting additional data
requested by the agency is 15 calendar
E:\FR\FM\29SER1.SGM
29SER1
59938
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
days (i.e., the same period of time that
is allowed for submitting followup
information under § 312.32(d)(3)). In
addition, the agency revised several
provisions to allow for electronic
submission of reports. First, in
§ 312.32(c)(1)(v) ‘‘Submission of IND
safety reports,’’ FDA renamed and
revised proposed § 312.32(c)(1)(iii)
‘‘Submission of written reports.’’
Second, FDA revised proposed
§ 312.32(c)(2) ‘‘Telephone and facsimile
transmission safety reports’’ to eliminate
the specificity that unexpected fatal or
life-threatening reports be submitted
only by telephone or facsimile
transmission so that other means of
rapid communication (e.g., e-mail) may
be accepted in the future. FDA also
renamed the provision to ‘‘Unexpected
fatal or life-threatening suspected
adverse reaction reports.’’ Last, in
§ 320.31(d)(3), FDA revised the
proposed requirement for submission of
IND safety reports and unexpected fatal
or life-threatening reports from
bioavailability and bioequivalence
studies to mirror these revisions.
The final rule allows for alternative
reporting arrangements, as provided in
former § 312.32(c)(3). However, the
agency revised the statement, ‘‘FDA may
request a sponsor to submit IND safety
reports in a format or at a frequency
different than that required under this
paragraph’’ by replacing the word
‘‘request’’ with ‘‘require’’ to reflect the
existing process. In addition, the final
rule clarifies the reporting requirements
for clinical investigations of drug
products that are marketed in the
United States (§ 312.32(c)(4)).
The final rule makes minor editorial
changes to § 312.32(d)(2) to clarify the
followup reporting requirements. In
addition, the agency eliminated the
redundant submission requirements for
information amendments and annual
reports under § 312.32(d)(4) because
they are already contained in §§ 312.31
and 312.33.
The final rule clarifies the
requirements for investigators to submit
reports of serious adverse events to the
sponsor and clarifies the requirement
for reporting study endpoints that are
serious adverse events (§ 312.64(b)).
Finally, the final rule requires that
applicants submit to FDA reports of
serious adverse events from
bioavailability and bioequivalence
studies. Proposed § 320.31(d) would
have required that these studies be
subject to the proposed IND safety
reporting requirements, thereby
requiring all reports under proposed
§ 312.32 (e.g., reports of serious and
unexpected SADRs, reports of
information sufficient to consider
product administration changes). FDA
has tailored the rule to require only
those reports that FDA believes would
be most informative (i.e., reports of all
serious adverse events). FDA also
revised this provision to make it
consistent with the final revisions for
submission of IND safety reports and
reports of any fatal or life-threatening
adverse event. The final rule requires
that reports must be submitted to the
Office of Generic Drugs.
Table 1 of this document identifies
the changes from the proposed rule in
the IND safety reporting requirements
that the agency made in this final rule.
TABLE 1—CHANGES MADE BY THE FINAL RULE FROM THE PROPOSED RULE
Description of Change See comment or section of this document (identified in parentheses)
for more detailed information regarding the change.
21 CFR Section in Final Rule
• Added definition for ‘‘adverse event’’ (1)
312.32(a) Life-threatening adverse event or lifethreatening suspected adverse reaction
• Made minor editorial revisions for clarity, including language changes to accommodate deletion of ‘‘SADR’’ definition and use of alternative terminology (2)
312.32(a) Serious adverse event or serious suspected adverse reaction
• Changed language to accommodate deletion of ‘‘SADR’’ definition and use of alternative
terminology (6)
• Incorporated the definition from former § 312.32(a) of ‘‘disability’’ within the definition of
‘‘serious’’ (III.A.2)
• Revised so that the seriousness determination is based on the opinion of either the sponsor or investigator (6)
312.32(a) Suspected adverse reaction
• Replaced the term ‘‘SADR’’ with the term ‘‘suspected adverse reaction,’’ clarifying the
meaning of ‘‘reasonable possibility’’ within the definition (1)
312.32(a) Unexpected adverse event or unexpected suspected adverse reaction
• Revised to make clear that ‘‘unexpected’’ adverse events or suspected adverse reactions
include those that may be anticipated from the pharmacological properties of the drug, or
that occur with members of the drug class, but that have not previously been observed
with the drug under investigation (8)
312.32(b) Review of safety information
• Made minor editorial changes for clarity and deleted the phrase ‘‘that have not been previously reported to FDA by the sponsor’’ (II)
312.32(c)(1) IND safety reports
• Withdrew the proposed requirement for each report of an SADR to contain a minimum
data set and to maintain records of efforts to obtain a minimum data set (5, 13, and 14)
312.32(c)(1)(i) Serious and unexpected suspected adverse reactions
erowe on DSK5CLS3C1PROD with RULES
312.32(a) Adverse event
• Clarified agency’s expectation for analysis of previous, similar reports or any other relevant
information (16)
• Withdrew the requirement that the causality assessment be based on the opinion of the investigator or the sponsor (15)
• Provided examples of the types of evidence that suggest a causal relationship between
the drug and the adverse event (18 to 21)
312.32(c)(1)(ii) Findings from other studies
• Revised proposed reports of ‘‘Information sufficient to consider product administration
changes’’ to clarify agency expectations of reports from clinical studies, epidemiological
studies or pooled analyses of multiple studies that suggest a significant risk in humans (23
to 25)
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
PO 00000
Frm 00006
Fmt 4700
Sfmt 4700
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
59939
TABLE 1—CHANGES MADE BY THE FINAL RULE FROM THE PROPOSED RULE—Continued
Description of Change See comment or section of this document (identified in parentheses)
for more detailed information regarding the change.
21 CFR Section in Final Rule
• Revised proposed reports of ‘‘Information sufficient to consider product administration
changes’’ to clarify agency expectations of reports from animal or in vitro testing that suggests a significant risk in humans (26 to 29)
312.32(c)(1)(iv) Increased rate of occurrence of
serious suspected adverse reactions
• Added the requirement for reports of any clinically important increase in the rate of a serious suspected adverse reaction over that listed in the protocol or investigator brochure
(32)
312.32(c)(1)(v) Submission of IND safety reports
• Revised to allow for electronic submission of IND safety reports and clarified time period
for reporting additional data or information requested by FDA (II)
312.32(c)(2) Unexpected fatal or life-threatening
suspected adverse reaction reports
• Revised to eliminate the specificity that unexpected fatal or life-threatening suspected adverse reaction reports be submitted only by telephone or facsimile transmission and renamed the requirement (II)
312.32(c)(3) Reporting format or frequency
• Replaced ‘‘request’’ with ‘‘require’’ (20)
312.32(c)(4) Investigations of marketed drugs
• Clarified requirements for investigations of marketed drugs (31)
312.32(c)(5) Reporting study endpoints
• Added requirement that study endpoints (e.g., mortality or major morbidity) must be reported according to the protocol instead of as IND safety reports except when there is evidence suggesting a causal relationship between the drug and the event (19 and 21)
312.32(d) Followup
• Deleted provision that required safety information to be submitted in an information
amendment or annual report and made minor editorial changes for clarity (III.K)
312.64(b) Investigator reports
• Clarified requirements for investigator reports (35 and 36)
320.31(d) Applicability of requirements regarding
an ‘‘Investigational New Drug Application’’
erowe on DSK5CLS3C1PROD with RULES
312.32(c)(1)(iii) Findings from animal or in vitro
testing
• Revised to require that persons conducting bioavailability and bioequivalence studies report all serious adverse events (II)
• Revised to make consistent with requirements for submission of IND safety reports and reports of any fatal or life-threatening adverse event (II)
III. Comments on the Proposed Rule
The agency received 110 comments in
the docket for the March 14, 2003,
proposed rule on premarket and
postmarket safety reporting revisions.
Comments were received from
prescription and nonprescription drug
manufacturers and related companies;
trade organizations representing drug
manufacturers and other interested
parties; blood banks and transfusion
facilities; international organizations
and non-U.S. agencies; professional
associations and organizations;
consultants; contract research
organizations; academic institutions;
health care and consumer advocacy
organizations, individual physicians,
pharmacists, and consumers; and
others.
To make it easier to identify
comments and our responses, the word
‘‘Comment,’’ in parentheses, appears
before the comment’s description, and
the word ‘‘Response,’’ in parentheses,
appears before our response. We have
numbered each comment to help
distinguish between different
comments. Similar comments are
grouped together under the same
number. The number assigned to each
comment is purely for organizational
purposes and does not signify the
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
comment’s value or importance or the
order in which it was received.
Comments addressing the proposed
requirements for IND safety reporting
and bioavailability and bioequivalence
studies and the agency’s responses
follow:
A. Definitions—Proposed § 312.32(a)
1. Suspected Adverse Drug Reaction
(SADR)
FDA proposed to add the term
‘‘suspected adverse drug reaction
(SADR)’’ and define the term as follows:
‘‘A noxious and unintended response to
any dose of a drug product for which
there is a reasonable possibility that the
product caused the response. In this
definition, the phrase ‘a reasonable
possibility’ means that the relationship
cannot be ruled out.’’
(Comment 1) Nearly all of the
comments overwhelmingly opposed the
agency adopting the proposed definition
of SADR and strongly encouraged the
agency to abandon the proposed
definition for many reasons, including
the following:
• Many comments did not agree that
‘‘reasonable possibility’’ should be
defined as ‘‘the relationship cannot be
ruled out.’’ Most comments stated that
this interpretation makes the definition
PO 00000
Frm 00007
Fmt 4700
Sfmt 4700
overly broad and will lead to reporting
almost every serious, unexpected
adverse event because no event could
ever be completely ruled out.
• Many comments stated that
although the proposed definition was
similar to the definition contained in
the ICH E2A guidance, the agency’s
interpretation was inconsistent with the
guidance. The ICH E2A guidance makes
clear that a causality assessment is
required for clinical investigations and
that a ‘‘reasonable causal relationship’’ is
meant to convey in general that there
are facts (evidence) or arguments to
suggest a causal relationship. The
comments expressed concern that the
agency’s interpretation of ‘‘reasonable
possibility’’ would lead to
inconsistencies in globally conducted
studies and reports.
• Many comments asserted that the
significantly increased numbers of
expedited reports that could result from
the proposed definition might dilute
real safety signals, making them harder
to detect. The lengthy in depth
investigations needed to rule out the
increased number of false positive
associations would take away resources
from other safety surveillance efforts
and potentially lead to a delay in
identification of real signals.
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59940
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
• Several comments expressed
concern that the proposed definition
would have a negative impact on the
conduct of clinical trials. In addition to
sharply increasing the number of reports
of cases from clinical trials that would
need to be sent to FDA in an expedited
manner, sponsors and investigators
would have to break the blind for nearly
all subjects with serious, unexpected
SADRs because the relationship
between drug and the event could not
definitively be ruled out. Increased
unblinding would compromise the
integrity of well-regulated clinical
investigations, lead to fewer patients
completing a trial, necessitate larger
patient enrollment, and lengthen the
timeline for new product development,
possibly leading to higher costs for
marketed drugs. One comment
expressed concern that, to minimize
unblinding, studies would be designed
to exclude patients with serious medical
conditions who are likely to experience
serious adverse events during the study
period, thereby limiting the
applicability of study results.
Many comments also stated that the
proposed definition would result in
significant increases in meaningless
individual expedited reports being sent
to already overburdened IRBs and
investigators. The comments pointed
out that an unintended effect of the
increase in volume of reports may be to
reduce an investigator’s and IRB’s
vigilance in detecting adverse events.
• Several comments expressed
concern that the proposed definition
would dilute the utility of drug product
labeling because many more events
would be regarded as ‘‘drug related’’
even though the likelihood of a true
causal relationship is minimal.
• Several comments stated that the
‘‘S’’ abbreviation for ‘‘suspected’’ in
SADR could be confused with the ‘‘S’’
abbreviation for ‘‘serious’’ in SAE
(serious adverse event).
The majority of the comments
recommended that reporting adverse
events from clinical trials should be
based on a scientific or medical
judgment that there is a possible causal
relationship between the drug and the
event, rather than simply being unable
to unequivocally exclude a drug’s role.
The comments suggested several
alternatives to the agency’s proposed
definition, including the following:
• Several comments recommended
that the definition of an adverse reaction
encompass all of the concepts presented
within the ICH E2A guidance, which are
supported by CIOMS and presented in
the European Union Clinical Trial
Directive. Comments recommended that
the definition of reasonable possibility
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
be technically consistent with the ICH
E2A guidance definition and clearly
delineate the concept of ‘‘reasonable
causal relationship’’ as conveying in
general that there are facts (evidence) or
arguments to suggest a causal
relationship.
• Some comments supported
retaining FDA’s former definition of
‘‘associated with the use of the drug’’ as
‘‘there is a reasonable possibility that the
experience may have been caused by the
drug.’’
Three comments supported adopting
the proposed definition because they
considered it an inclusive, conservative
approach to adverse event reporting.
(Response) Based on the comments,
and on review of definitions and
terminology used in the ICH E2A
guidance and in former § 312.32, the
agency has decided not to adopt the
proposed definition for ‘‘suspected
adverse drug reaction (SADR).’’ The
agency agrees with the comments
stating that there should be a causality
assessment applied and that the
threshold for reporting should be that
there is a ‘‘reasonable possibility’’ that
the drug caused the adverse event. The
agency also believes that it is important
to use definitions that are clear and
consistent, and in harmony with those
used internationally.
The agency believes that the
comments raised legitimate concerns
that the proposed definition was too
broad and could have a negative impact
on clinical trials, IRBs, investigators,
signal detection, and drug labeling.
Instead of adopting the proposed
definition, the agency has adopted the
terms for ‘‘adverse event’’ and
‘‘suspected adverse reaction’’ in the
definition section of this final rule,
which addresses these concerns. The
definitions of these terms should
contribute to harmonization of safety
reporting to regulatory authorities
worldwide because they are consistent
with the concepts and definitions
adopted by the ICH E2A guidance and
CIOMS. The terms are defined as
follows:
• ‘‘Adverse event’’ means any
untoward medical occurrence
associated with the use of a drug in
humans, whether or not considered
drug related. (For the purposes of this
definition, ‘‘untoward’’ means
unfavorable, negative, or harmful).
‘‘Suspected adverse reaction’’ means
any adverse event for which there is a
reasonable possibility that the drug
caused the adverse event. For the
purposes of IND safety reporting,
‘‘reasonable possibility’’ means there is
evidence to suggest a causal relationship
between the drug and the adverse event.
PO 00000
Frm 00008
Fmt 4700
Sfmt 4700
Suspected adverse reaction implies a
lesser degree of certainty about causality
than adverse reaction, which means any
adverse event caused by a drug.
These definitions reflect the varying
degrees of certainty that are part of a
causality assessment. For example:
• An adverse event (also referred to as
an ‘‘adverse experience’’) is any event
observed or reported that is associated
with the use of the drug, without regard
to causality.
• A suspected adverse reaction is a
subset of all adverse events in which
there is a reasonable possibility that the
drug caused the event.
• An adverse reaction, described
within the definition, is a subset of all
suspected adverse reactions for which
there is reason to conclude that the drug
caused the event.
With this change from the proposed
definition, the basis that the agency has
established for assessing the degree of
certainty about causality between a drug
and an adverse event for the purposes
of expedited IND safety reporting has
not changed from former § 312.32(c).
The sponsor must continue to evaluate
the evidence and use its judgment to
determine whether an adverse event
meets the definition of suspected
adverse reaction and qualifies for
expedited reporting under § 312.32(c).
The agency has also clarified the
requirements for reporting a serious and
unexpected suspected adverse reaction
under § 312.32(c)(1)(i) to assist sponsors
with making this determination (see
Comment 18 of this document).
Finally, the agency has concluded
that abbreviations are potentially
confusing (e.g., the ‘‘S’’ abbreviation for
‘‘suspected’’ in SADR could be mistaken
for an abbreviation of the term
‘‘serious’’). Although the agency has
retained the term ‘‘suspected’’ in
‘‘suspected adverse reaction,’’ our
preferred approach is to avoid use of
any abbreviation (e.g., ‘‘SAR’’ for
‘‘suspected adverse reaction’’). The
agency believes that sponsors are
familiar with the term ‘‘suspected’’ and
its use by the European Commission
and CIOMS (e.g., the acronym ‘‘SUSAR’’
means ‘‘suspected, unexpected, serious
adverse reaction’’ in guidance
documents and working group reports
(for example, see Ref. 1)).
Because the agency is not adopting
the proposed definition of ‘‘suspected
adverse drug reaction (SADR),’’ other
proposed definitions (e.g., ‘‘serious
SADR,’’ ‘‘life-threatening SADR’’) and
requirements that used this terminology
have been revised in this final rule to
use the terms ‘‘adverse event’’ or
‘‘suspected adverse reaction’’ as
appropriate.
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
2. Disability
The proposed rule included a
definition of the term ‘‘disability’’ to
mean a substantial disruption of a
person’s ability to conduct normal life
functions. Because the term ‘‘disability’’
appeared only within the definition of
‘‘serious SADR’’ in the proposed rule,
the agency eliminated the definition of
‘‘disability’’ as a separate term in this
final rule. Instead, the agency revised
the definition of ‘‘serious adverse event
or serious suspected adverse reaction’’
in this final rule to incorporate the
definition of ‘‘disability’’ by replacing
the phrase ‘‘a persistent or significant
disability/incapacity’’ with ‘‘a persistent
or significant incapacity or substantial
disruption of the ability to conduct
normal life functions.’’ Thus, in the final
rule, the term disability is replaced by
the proposed definition in the one place
where it appeared, and the definition
itself has been deleted.
erowe on DSK5CLS3C1PROD with RULES
3. Life-Threatening Suspected Adverse
Drug Reaction (SADR)
FDA proposed the term ‘‘lifethreatening suspected adverse drug
reaction (SADR)’’ to mean any SADR
that, in the view of the investigator or
sponsor, places the patient or subject at
immediate risk of death from the SADR
as it occurred. It does not include an
SADR that, had it occurred in a more
severe form, might have caused death.
(Comment 2) Several comments
agreed with FDA’s proposal to add the
term ‘‘or sponsor’’ to the definition of
life-threatening SADR. SADRs would be
reported as life-threatening if either the
investigator or sponsor considered them
to be life-threatening. However, several
comments expressed concern with
FDA’s proposal. The comments stated
that a trained investigator is most
qualified to make the sometimes
subjective assessment of whether an
event is life-threatening and that this
determination often is best made by the
health-care professional or the reporter
who is in direct contact with the
patient. These comments also stated that
sponsors may exercise medical and
scientific judgment in deciding whether
expedited reporting is appropriate. One
comment stated that allowing a sponsor
to determine severity would change the
nature of the assessment and result in
increased reporting of events assessed
by those with often incomplete
information. One comment pointed out
that FDA’s rationale for expanding the
role of the sponsor is not supported by
the quote from the ICH E2A guidance in
the preamble to the proposed rule (68
FR 12406 at 12419) because the ICH
E2A guidance quote refers to causality
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
assessment, not assessment of
seriousness.
(Response) The agency agrees with
the comments that support expanding
this definition to include reporting of an
adverse event as life-threatening if
either the investigator or the sponsor
considers it to be life-threatening. The
agency believes that, in some cases, the
sponsor may not agree with the
investigator’s assessment that an
adverse event does not qualify as lifethreatening. In such cases, because these
events are critically important for the
identification of significant safety
problems, the agency believes that
broadening the definition to allow
sponsors to also make this assessment is
prudent and appropriate. While the
agency agrees with the comment that
pointed out that the preamble to the
proposed rule misinterpreted the quote
from the ICH E2A guidance, we
nonetheless believe that the revision to
the definition is consistent with the
overall intent of the ICH E2A guidance.
(Comment 3) Several comments
disagreed with the agency’s position
articulated in the preamble to the
proposed rule that reasons for any
differences of opinion between the
investigator and sponsor regarding a
determination that an SADR is lifethreatening would be included in the
IND safety report (68 FR 12406 at
12419). The comments argued that this
adds no value and is not appropriate or
necessary in all cases. In addition,
comments stated that obtaining the
investigator’s view when he or she
deems the event non-life-threatening
would be difficult.
(Response) The agency agrees that
reasons for differences of opinion
between the sponsor and investigator
are not always important and, therefore,
not necessary to include in the IND
safety report in all cases. Therefore, in
this final rule, the agency does not
require including the reasons for
differences of opinion in the IND safety
report. However, it is important that any
adverse event or suspected adverse
reaction considered life-threatening by
either the sponsor or the investigator be
reported as such.
(Comment 4) Some comments
suggested that FDA clarify the definition
of life-threatening to take into account
the role of other study staff making
safety observations. The comments
suggested that the definition be clarified
to state that investigators or sponsors
must evaluate information
communicated to them or recorded by
their qualified staff or agents and
transmit reportable information to the
sponsor or FDA. One comment
recommended that the definition be
PO 00000
Frm 00009
Fmt 4700
Sfmt 4700
59941
modified to include contractors as well
as sponsors.
(Response) The agency does not agree
that the recommended revisions to the
definition are necessary because taking
the observations of staff into account is
inherent in the obligations of the
investigator. Any qualified study staff
could make pertinent safety
observations, and it is the investigator’s
responsibility in supervising the
conduct of the clinical investigation (see
§§ 312.53 and 312.60) to report adverse
experiences to the sponsor in
accordance with § 312.64. Further
information on the supervisory
responsibilities of investigators can be
obtained in the agency’s guidance for
industry entitled ‘‘Investigator
Responsibilities: Protecting the Rights,
Safety, and Welfare of Study Subjects’’
(74 FR 55052, October 26, 2009).2 The
agency does not believe that it is
necessary to change the definition to
include contractors because, under
§ 312.52, a contract research
organization that assumes any
obligation of a sponsor must comply
with the applicable regulation.
4. Minimum Data Set
Under § 312.32(a), FDA proposed the
term ‘‘minimum data set’’ to mean that
‘‘the report includes an identifiable
patient, an identifiable reporter, a
suspect drug product, and an SADR.’’
(Comment 5) Two comments
requested further clarification regarding
the meaning of ‘‘identifiable’’ with
respect to the kind and amount of
information needed to meet the criteria
for an ‘‘identifiable patient’’ and
‘‘identifiable reporter.’’ One comment
questioned whether patient
characteristics, such as age or gender,
would be adequate, or if the ability to
contact the patient is necessary.
(Response) As discussed in comments
13 and 14 of this document, because the
four elements of the minimum data set
are generally readily available in the
clinical trial setting, the agency has
determined that the definition and the
requirement are unnecessary and has
decided not to require a minimum data
set for IND safety reports as proposed in
§ 312.32(c). Because the agency is not
adopting this definition in the IND
safety reporting requirements, the
comments requesting clarification about
2 Draft and final guidances for the Center for Drug
Evaluation and Research (CDER)-related
information are posted on the Internet at https://
www.fda.gov/Drugs/GuidanceCompliance
RegulatoryInformation/Guidances/default.htm. The
Center for Biologics Evaluation and Research
(CBER)-related information is posted at https://
www.fda.gov/BiologicsBloodVaccines/Guidance
ComplianceRegulatoryInformation/Guidances/
default.htm (21 U.S.C. 371(h), 21 CFR 10.115).
E:\FR\FM\29SER1.SGM
29SER1
59942
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
the elements of the definition are no
longer relevant.
erowe on DSK5CLS3C1PROD with RULES
5. Serious SADR
FDA proposed to define ‘‘serious
SADR’’ in the same way as the thencurrent definition of ‘‘serious adverse
drug experience’’ under § 312.32(a) as
follows: ‘‘Serious SADR means any
SADR that results in any of the
following outcomes: Death, a lifethreatening SADR, inpatient
hospitalization or prolongation of
existing hospitalization, a persistent or
significant disability/incapacity, or a
congenital anomaly/birth defect.
Important medical events that may not
result in death, be life-threatening, or
require hospitalization may be
considered a serious SADR when, based
upon appropriate medical judgment,
they may jeopardize the patient or
subject and may require medical or
surgical intervention to prevent one of
the outcomes listed in this definition.
Examples of such medical events
include allergic bronchospasm requiring
intensive treatment in an emergency
room or at home, blood dyscrasias or
convulsions that do not result in
hospitalization, or the development of
drug dependency or drug abuse.’’
(Comment 6) One comment suggested
that the definition of ‘‘serious SADR’’ be
revised to expressly allow the sponsor
to determine if an adverse event is
serious, in the absence of a reporter’s
assessment of seriousness.
(Response) For reasons similar to
those stated in Comment 2 of this
document (definition of lifethreatening), the agency agrees that the
definition of ‘‘serious adverse event or
serious suspected adverse reaction’’
should be revised to allow the
determination that an adverse event or
suspected adverse reaction is ‘‘serious’’
if either the investigator or sponsor
considers it serious. Therefore, the
agency has revised this definition to add
the phrase ‘‘in the view of either the
investigator or sponsor.’’
6. Unexpected SADR
FDA proposed that the definition of
‘‘unexpected SADR’’ be the same as the
then-current definition for ‘‘unexpected
adverse drug experience’’ under
§ 312.32(a), except that the following
sentence was added to make clear
which SADRs are considered
unexpected: ‘‘SADRs that are mentioned
in the investigator’s brochure as
occurring with a class of drugs but not
specifically mentioned as occurring
with the particular drug are considered
unexpected.’’
(Comment 7) One comment stated
that in the proposed definition, the
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
‘‘severity’’ standard is vague, leaving the
determination of ‘‘expectedness’’ to the
investigator’s judgment.
(Response) Unless a sponsorinvestigator is responsible for the
clinical trial, the sponsor, rather than
the investigator, generally determines if
a suspected adverse reaction is
unexpected for reporting purposes.
However, the agency acknowledges that
judgment is needed to decide if the
severity of a suspected adverse reaction
is greater than described in the
investigator brochure. The definition of
‘‘unexpected adverse event or
unexpected suspected adverse reaction’’
in the final rule includes an example of
a suspected adverse reaction that would
be considered unexpected by virtue of
its greater severity than other suspected
adverse reactions mentioned in the
investigator brochure (i.e., hepatic
necrosis would be considered
unexpected where the investigator
brochure includes elevated hepatic
enzymes or hepatitis).
(Comment 8) Another comment
recommended that FDA provide
guidance on what should be considered
‘‘expected’’ for regulatory reporting
purposes, in particular, what safety
information to include in the
investigator brochure and what subset of
such information would be considered
‘‘expected’’ (i.e., only those for which a
causal relationship is suspected,
reasonably established, or inferred
based on evidence). Some comments
stated that if the basis for evaluating
expectedness is that an event is listed in
the investigator’s brochure, sponsors
may add long lists of adverse events,
thereby delaying important safety
reports from being submitted to FDA.
One comment recommended that FDA
require that, until the applicable
reference safety information document
is officially updated (e.g., reprinted and
distributed) to include a new serious,
suspected adverse reaction (thereby
making it expected), all subsequent
reports of similar serious adverse drug
reactions be submitted expeditiously as
an IND safety report. Another comment
suggested adopting use of the
Developmental Core Safety Information
(DCSI) document, proposed by a CIOMS
Working Group, as the reference for
‘‘expectedness’’ instead of the
investigator brochure because the DCSI
document contains only those adverse
events that, after careful analysis are
believed by the company to be likely
related to the drug (Refs. 2 and 3).
(Response) The purpose of the
investigator brochure is to provide the
investigator with information (clinical
and nonclinical) about the
investigational drug that is relevant to
PO 00000
Frm 00010
Fmt 4700
Sfmt 4700
study of the drug in human subjects.
The investigator brochure should
include the information that is
important for the investigator, who is
administering the drug to human
subjects, to know and understand. The
investigator brochure is required to
include information about the drug
substance and formulation,
pharmacological and toxicological
effects of the drug in animals (and in
humans, if known), pharmacokinetics
and biological disposition of the drug in
animals (and in humans, if known),
information relating to safety and
effectiveness in humans obtained from
prior clinical studies, and information
about possible risks and side effects to
be anticipated on the basis of prior
experience with the drug under
investigation or with related drugs, and
precautions or special monitoring to be
done as part of the investigational use
of the drug (see § 312.23(a)(5)).
In general, the investigator brochure
lists those adverse events that have been
observed with the investigational drug
and for which a causal relationship with
the drug is suspected or confirmed. It is
not appropriate for sponsors to add long
lists of adverse events that are unlikely
to have been caused by the drug to the
investigator brochure because such lists
could dilute the importance of clinically
meaningful risk information and as a
result, may put subjects at risk. The
sponsor needs to exercise judgment
when deciding if the threshold has been
reached for adding a newly observed
adverse event to the investigator
brochure. This decision usually
depends on the strength of the evidence
from individual or multiple cases and
previous knowledge about the drug or
drug class. In some cases, the threshold
for including an adverse event may be
lower if it could result in a significant
adverse outcome for trial participants.
The investigator brochure describes
adverse events that may be predicted to
occur based on the pharmacological
properties of the drug. For reporting
purposes, if an adverse event occurs that
has not previously been observed with
the drug under investigation, the event
is considered ‘‘unexpected.’’ To make
clear that such predicted adverse events
are considered ‘‘unexpected,’’ the final
rule revises the proposed definition of
‘‘unexpected’’ to state explicitly that the
term also refers to adverse events or
suspected adverse reactions that are
mentioned in the investigator brochure
as occurring with a class of drugs or as
anticipated from the pharmacological
properties of the drug, but are not
specifically mentioned as occurring
with the particular drug under
investigation.
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
erowe on DSK5CLS3C1PROD with RULES
The agency expects the sponsor to
update the investigator brochure on an
ongoing basis with new important safety
information. However, the agency agrees
with the comment that, until the
investigator brochure and other
applicable reference safety information
are updated to include a new serious,
suspected adverse reaction, subsequent
reports of similar serious, suspected
adverse reactions must be submitted
expeditiously in IND safety reports.
Finally, sponsors submit and the
agency accepts a variety of formats for
the investigator brochure. For this
reason, we are not formally adopting use
of the DCSI document in this final rule.
However, we agree that a sponsor could
incorporate a document such as the
DCSI into the investigator brochure for
use as the reference for ‘‘expectedness’’
for reporting purposes if the DCSI
contains the required safety information
about the investigational drug.
B. Review of Safety Information—
Proposed § 312.32(b)
IND safety reporting regulations in
former § 312.32(b) required that
sponsors promptly review all
information relevant to the safety of the
drug obtained or otherwise received by
the sponsor from any source, foreign or
domestic. Examples of potential sources
of information in the former regulation
included information derived from any
clinical or epidemiological
investigations, animal investigations,
commercial marketing experience,
reports in the scientific literature, as
well as unpublished scientific papers,
and reports from foreign regulatory
authorities that had not been previously
reported to FDA by the sponsor.
Proposed § 312.32(b) would have
amended this requirement to include in
vitro studies as another example of a
potential source of information and to
clarify that ‘‘reports from commercial
marketing experience’’ is intended to
apply only to reports from foreign
commercial marketing experience for
drugs that are not marketed in the
United States. As proposed, reports
from IND studies of drugs that are
marketed in the United States would be
required to be reported as described
under § 312.32(c)(4), if applicable.
(Comment 9) One comment stated
that reportable information can come
from a wider variety of media or sources
than those listed in the proposed rule.
The comment maintained that
investigators or sponsors participating
in public or private meetings or
conferences can learn of reportable
events from colleagues or other
professionals. The comment
recommended that the list of potential
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
sources of reportable information
include such alternative sources.
(Response) The sponsor is required to
‘‘promptly review all information
relevant to the safety of the drug
obtained or otherwise received by the
sponsor from foreign or domestic
sources, including information derived
from any clinical or epidemiological
investigations, animal or in vitro studies
* * *’’ (emphasis added). The sources
listed in the requirement are not all
inclusive, but represent examples of the
variety of sources that may yield safety
information. Therefore, the agency
agrees that reportable information can
come from sources other than those
listed in § 312.32(b) and that one such
source could be from public or private
meetings. However, the agency does not
believe that it is necessary to amend the
requirement to provide additional
examples.
(Comment 10) One comment agreed
with the clarification that reporting from
commercial marketing experience
applies only to foreign commercial
marketing experience for drugs that are
not marketed in the United States. The
comment requested that FDA further
make it clear that expedited reporting
under § 312.32 is not required for
reports from foreign commercial
marketing experience for a different
formulation of the same active moiety as
a drug product that is lawfully marketed
in the United States and that those
reports should be submitted to the most
appropriate new drug application (NDA)
for the active moiety.
(Response) As described further in
Comment 31 of this document, IND
safety reports are required under
§ 312.32(c)(4) for suspected adverse
reactions observed in clinical studies
that are being conducted under an IND
for a drug marketed or approved in the
United States. In general, an expedited
report from domestic or foreign
commercial marketing experience for a
drug lawfully marketed in the United
States would not be submitted to the
IND, but instead, must be submitted in
accordance with the relevant
postmarketing reporting requirements
(e.g., §§ 310.305, 314.80, and 600.80).
Similarly, a report of a suspected
adverse reaction from foreign marketing
experience for a different formulation of
the drug product (same active moiety)
that is lawfully marketed in the United
States must be submitted in accordance
with the relevant postmarketing
reporting requirements.
(Comment 11) One comment agreed
with the proposal to add in vitro studies
to the list of information that should be
reviewed by the sponsor in its ongoing
assessment of the safety of an
PO 00000
Frm 00011
Fmt 4700
Sfmt 4700
59943
investigational drug. Some comments
stated that it would be helpful if FDA
could provide examples, in addition to
carcinogenicity, mutagenicity and
teratogenicity, of when safety data from
in vitro studies would yield relevant,
important information that should be
reviewed for IND reporting purposes.
(Response) Data from in vitro
microsusceptibility, drug interaction, or
genotoxicity studies are examples of
other data from in vitro studies that may
yield important safety information.
(Comment 12) One comment
expressed concern that once a sponsor
provides FDA with the animal and in
vitro studies, emails, and reports from
foreign regulatory authorities and any
other information it reviewed in
determining whether to report safety
information, FDA may have to make the
information publicly available under the
Freedom of Information Act (FOIA). The
comment stated that, before
implementing the requirement, FDA
should explain why these additional
data are needed and how they will be
handled for FOIA purposes. The
comment requested that the requirement
be withdrawn.
(Response) The agency uses the safety
information submitted by the sponsor,
from any source, to continually monitor
and evaluate the safety of the drug. Data
and information in an IND are disclosed
consistent with applicable statutes and
regulations. The requirements under
§ 312.130 describe the availability for
public disclosure of data and
information in an IND. The minor
clarifications made to these
requirements do not change these
protections against public disclosure.
Therefore, the agency declines to
withdraw the requirement as requested
by the comment.
C. IND Safety Reports (Requirement for
Minimum Data Set)—Proposed
§ 312.32(c)
FDA proposed to amend § 312.32(c) to
require that sponsors must not submit
an individual case safety report for an
SADR if the report does not contain a
minimum data set, but instead must
maintain records of any information
received or otherwise obtained for the
SADR along with a record of its efforts
to obtain a minimum data set. In the
preamble to the proposed rule, the
agency stated that sponsors should
include in any written IND safety
reports subsequently filed with FDA a
chronological history of their efforts to
acquire the minimum data set if there is
a delay in obtaining the information, but
that it was not necessary to include the
chronological history in IND safety
reports sent to investigators (68 FR
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59944
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
12406 at 12424). In addition, FDA
proposed in § 312.32(c)(1)(i) that a
sponsor must submit an IND safety
report within 15 calendar days after
receipt by the sponsor of the minimum
data set for the SADR.
As noted in Comment 5 of this
document, the agency has reconsidered
the proposed requirement under
§ 312.32(c) that would have required
sponsors to only submit an individual
case safety report for an SADR if the
report contained a minimum data set.
Most IND safety reports are derived
from observations from clinical trials. In
the setting of a clinical trial, information
is collected in a controlled environment
where the four elements in the
definition of minimum data set, as well
as other information needed to evaluate
the suspected adverse reaction (e.g.,
information that would be contained in
a narrative report or on FDA Form
3500A), are generally readily available.
Accordingly, the agency has revised
§ 312.32(c)(1) to eliminate the minimum
data set language and to require instead
that the sponsor submit an IND safety
report after it determines that the
information qualifies for reporting
under § 312.32(c)(1)(i), (c)(1)(ii),
(c)(1)(iii), or (c)(1)(iv).
(Comment 13) One comment stated
that waiting for collection of all the
elements of the minimum data set,
especially for determination of
causality, could result in a significant
delay in reporting to FDA. The comment
requested clarification on when the
reporting timeclock would start.
Another comment requested
clarification on whether the date of
receipt of the minimum data set for the
SADR represents day zero or day one.
(Response) The reporting timeclock
starts (i.e., day zero) as soon as the
sponsor determines that the information
qualifies for reporting under
§ 312.32(c)(1)(i), (c)(1)(ii), (c)(1)(iii), or
(c)(1)(iv). For a serious and unexpected
suspected adverse reaction from a
clinical trial, this would be the day the
sponsor receives information from the
clinical investigator. If any information
necessary to evaluate and report the
suspected adverse reaction is missing or
unknown, the sponsor should actively
seek such information.
(Comment 14) Several comments
stated that including in an IND safety
report a chronological history of their
efforts to acquire the minimum data set
is inconsistent with standards for nonU.S. regulators and the ICH E2A
guidance, adds no value, may lead to
potential legal risk in the event of
litigation, may impede electronic
transmission of individual case safety
reports, and will become an
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
administrative burden. Some comments
suggested that records of efforts to
obtain the minimum data set should be
maintained within the case record in the
sponsor’s files, available upon request
or during agency inspections. One
comment suggested FDA require
manufacturers to have procedures in
place to acquire a minimum data set.
One comment stated that the agency
needs to define the minimum
requirements for conducting due
diligence to avoid variation from
sponsor to sponsor. Another comment
recommended reinforcing the need for
sponsors to conduct followup activities
and for FDA to audit industry for
compliance. One comment requested
clarification on the sponsor’s timeframe
for maintaining records of its efforts to
obtain the minimum data set. One
comment pointed out that although FDA
stated in the preamble that the
chronological history included in the
IND safety report would not need to be
sent to investigators, this statement
creates conflict because sponsors must
tell investigators the same information
that is reported to FDA.
(Response) The agency agrees with
comments that including a
chronological history in an IND safety
report of efforts to acquire information
is not necessary and could be an
administrative burden without added
value. Accordingly, the proposed
requirement for a chronological history
has been deleted from § 312.32(c).
Under § 312.32(d)(1), sponsors are
required to promptly investigate all
safety information received, so it is
inherent in that requirement that
sponsors promptly and diligently
attempt to obtain the information
necessary for evaluating a suspected
adverse reaction. If critical information
is missing or unknown, the sponsor
should actively seek the information.
The regulations do not include specific
procedures for conducting or
documenting due diligence activities
because the agency recognizes that there
is more than one approach that would
be appropriate, depending on the
situation.
Similarly, because the minimum data
set requirement is no longer included,
the agency is not adopting the proposed
requirement in § 312.32(c) to maintain
records of any information received or
otherwise obtained for the SADR when
the sponsor does not have a reportable
minimum data set. The agency notes
that sponsors are required under
§ 312.57(c) to retain records and reports
required under part 312 (including
safety information received by the
sponsor) for 2 years after a marketing
application is approved for the drug or,
PO 00000
Frm 00012
Fmt 4700
Sfmt 4700
if an application is not approved for the
drug, until 2 years after shipment and
delivery of the drug for an
investigational use is discontinued and
FDA has been so notified. The agency
may audit these records as part of its
inspection process.
D. Serious and Unexpected SADR—
Proposed § 312.32(c)(1)(i)
In proposed § 312.32(c)(1)(i), FDA
proposed that the sponsor must notify
FDA and all participating investigators
in a written IND safety report of any
SADR that, based on the opinion of the
investigator or sponsor, is both serious
and unexpected, as soon as possible, but
in no case later than 15 calendar days
after receipt by the sponsor of the
minimum data set for the serious,
unexpected SADR. In addition, FDA
proposed that the sponsor must identify
all safety reports previously filed with
the IND concerning a similar SADR, and
must analyze the significance of the
SADR in light of the previous, similar
reports.
(Comment 15) One comment agreed
with the proposal that the assessment of
whether the event is serious or
unexpected be based on the opinion of
the ‘‘investigator or sponsor,’’ while
other comments expressed concern.
Several comments indicated that
investigators should not be required to
assess ‘‘expectedness.’’ One comment
stated that ‘‘expectednessx’’ is a
regulatory definition that would be
difficult for an investigator to apply in
a consistent manner. Another comment
suggested replacing the proposed
language with ‘‘any SADR that is serious
based on the opinion of the investigator
or sponsor and unexpected.’’
(Response) The agency agrees that, in
contrast to the assessments of whether
an adverse event or suspected adverse
reaction is ‘‘serious’’ and ‘‘lifethreatening,’’ which require medical
judgment by the investigator or sponsor,
the assessment of whether an adverse
event or suspected adverse reaction is
‘‘unexpected’’ in this context refers to a
regulatory definition (i.e., not listed in
the investigator brochure) that is more
appropriately applied by the sponsor.
The sponsor is usually in a better
position to assess the adverse event
information and determine whether the
adverse event is ‘‘unexpected’’ for
reporting purposes because the sponsor
has access to more information (e.g.,
from all the investigative sites in a
multi-center study). Therefore, the
agency has revised this proposed
requirement by deleting the phrase
‘‘based on the opinion of the investigator
or sponsor,’’ which leaves this
determination to the sponsor.
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
(Comment 16) Several comments
asked for clarification on various
aspects of the requirement to identify all
safety reports previously filed with the
IND concerning a similar SADR and to
analyze the significance of the SADR in
the context of the previous, similar
reports. One comment requested
clarification on the meaning of
‘‘previously filed with the IND’’ and
whether this should include an analysis
of previous similar reports across
multiple open INDs or only a single
IND. The comment noted that there
could be company-sponsored IND
studies and investigator-sponsored IND
studies ongoing simultaneously, with
safety data stored in different places.
One comment requested clarification on
what constitutes a ‘‘similar’’ SADR and
on the meaning of ‘‘analyze the
significance.’’ This comment noted that
companies should already have
processes and procedures in place to
periodically review and analyze safety
data to detect ‘‘signals,’’ and asked
whether FDA expects an ‘‘analysis’’ for
postmarketing study reports filed to the
IND or all reports for the product,
including postmarketing spontaneous
reports. The comment suggested that
FDA remove this requirement for both
IND and postmarketing studies, since
for IND studies, companies should
already be performing these analyses
and updating their investigator
brochures with significant new safety
information, and for postmarketing
studies, analyses of all adverse events
are being performed in the periodic
safety update report (PSUR).
(Response) The agency expects the
analysis of the significance of the
suspected adverse reaction in the
context of similar reports to include all
INDs held by the sponsor and any other
relevant information of which the
sponsor is aware. To make this clear, the
agency revised the provision in final
§ 312.32(c)(1) to require that in each IND
safety report, the sponsor must identify
all IND safety reports previously
submitted to FDA concerning a similar
suspected adverse reaction, and must
analyze the significance of the
suspected adverse reaction in light of
previous, similar reports or any other
relevant information.
The agency declines to withdraw the
requirement as suggested by the
comment because we consider this
information to be critical for the ongoing
evaluation of the investigational drug’s
safety. Because this is not a new
requirement (see former
§ 312.32(c)(1)(ii)), the agency agrees that
companies should have processes in
place to periodically review and analyze
their safety data and update their
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
investigator brochures with significant
new safety information. This analysis
should include an evaluation of the
suspected adverse reaction in the
context of other related reports or
adverse events, including those that
may have occurred in postmarketing
studies.
(Comment 17) One comment asked
whether the IND safety report should be
sent only to investigators participating
in company-sponsored studies or to
studies conducted under all open INDs
for the product. One comment requested
that FDA clarify its expectations for
cross-reporting to investigators
participating in different trials under the
same IND or different INDs with the
same active moiety. One comment asked
if followup IND safety reports
containing only minor refinements are
to be sent to FDA and all investigators
who received the initial safety report or
only to FDA.
(Response) The sponsor must report
to any participating investigators under
all open INDs, including those held by
the sponsor and those to which the
sponsor provides the investigational
drug (investigator-sponsored). To make
this clear, the agency revised the
provision in § 312.32(c)(1) to require
that a sponsor notify FDA and all
participating investigators (i.e., all
investigators to whom the sponsor is
providing drug under its INDs or under
any investigator’s IND) in an IND safety
report of potential serious risks, from
clinical trials or any other source, as
soon as possible, but in no case later
than 15 calendar days after the sponsor
determines that the information
qualifies for reporting under
§ 312.32(c)(1)(i), (c)(1)(ii), (c)(1)(iii), or
(c)(1)(iv).
Followup reports should be sent to
investigators to inform and update them
about an important suspected adverse
reaction if it significantly affects the
care of the subjects or conduct of the
study. Minor refinements that do not
significantly affect care of subjects or
conduct of the study need to be sent to
FDA but need not be sent to
investigators. Such information may be
communicated to investigators in a
routine update of the investigator
brochure.
(Comment 18) As stated in Comment
1 of this document, there were many
comments opposed to FDA’s proposed
SADR definition, some of which
recommended against adopting the
proposed SADR definition, and instead,
urged FDA to clarify the types of
evidence that suggest there is a
reasonable possibility that a drug
product caused the adverse event.
PO 00000
Frm 00013
Fmt 4700
Sfmt 4700
59945
(Response) Before submitting an IND
safety report under § 312.32(c)(1)(i), the
sponsor must determine that the event:
(1) Is serious, (2) is unexpected, and (3)
meets the definition of ‘‘suspected
adverse reaction’’ in § 312.32(a) (i.e., that
there is a ‘‘reasonable possibility’’ that
the drug caused the event). These
criteria have not changed from former
§ 312.32(c)(1)(i)(A). Making this
determination will always require
judgment based on the best available
information.
Currently, sponsors often report in an
expedited manner serious adverse
events that may be due to the
underlying disease or that occur
commonly in the study population,
even when there is little reason to
believe that the drug caused the event.
Such reports are generally
uninformative and, therefore, do not
meaningfully contribute to the
developing safety profile of the drug.
The agency believes that clarifying what
evidence suggests a causal relationship
will increase the likelihood that
information reported to FDA will
meaningfully contribute to the
developing safety profile of the product
and improve the overall quality of safety
reporting.
Therefore, to assist sponsors with
determining whether an adverse event
meets the definition of suspected
adverse reaction, the agency revised the
proposed requirement under
§ 312.32(c)(1)(i) to make it clear that
sponsors are to report to FDA and all
participating investigators only if there
is evidence to suggest a causal
relationship between the drug and the
adverse event. Final § 312.32(c)(1)(i)
also provides the following examples:
• A single occurrence of an event that
is uncommon and known to be strongly
associated with drug exposure (e.g.,
angioedema, hepatic injury, StevensJohnson Syndrome).
• One or more occurrences of an
event that is not commonly associated
with drug exposure, but is otherwise
uncommon in the population exposed
to the drug (e.g., tendon rupture).
• An aggregate analysis of specific
events observed in a clinical trial (such
as known consequences of the
underlying disease or condition under
investigation or other events that
commonly occur in the study
population independent of drug
therapy) that indicates those events
occur more frequently in the drug
treatment group than in a concurrent or
historical control group.
E. Alternative Reporting Arrangements
In the preamble to the proposed rule,
FDA acknowledged that the proposed
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59946
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
definition of SADR (which defined
‘‘reasonable possibility’’ to mean that the
causal relationship between a product
and a response to the product cannot be
ruled out) may result in submission of
numerous safety reports to the agency
for which the reported SADR is not
informative as a single report because it
is very likely to have been a
consequence of the patient’s disease.
FDA invited comment on use of
alternative reporting methods that
would minimize overreporting of
uninformative events and assure
submission of meaningful reports of
unexpected events. For example, one
such alternative would be to include in
study protocols or other documentation
a list of known consequences of the
disease that would not be submitted to
FDA in an expedited manner as
individual case safety reports (e.g.,
events that are endpoints of the study)
(68 FR 12406 at 12418).
(Comment 19) Some comments agreed
with the agency’s suggestion that
protocols could be written to exclude
specific disease-related events from
expedited reporting if these events are
study endpoints. Other comments
expressed concern that alternative
reporting methods would not have the
intended effect of reducing
overreporting and could exacerbate
problems with the proposed SADR
definition of reasonable possibility in
which the causal relationship ‘‘cannot
be ruled out.’’ They argued that
effectively eliminating clinical judgment
in reporting coupled with an ad hoc
exemption mechanism would lead to
different standards across clinical
programs, between different sponsors of
studies, and across FDA review
divisions. These comments further
pointed out that negotiating and
managing exemptions to expedited
reporting would place a significant
burden on FDA and companies and
would necessitate the creation of an
FDA structure and process to ensure
consistency across products. While
many of these comments recommended
against finalizing the proposed
definition, others suggested alternatives
(e.g., waiver provisions) to alleviate
overreporting caused by the proposed
definition. One comment recommended
that approaches to minimize
overreporting only be considered for
late stage development (i.e., Phase 3 and
4 studies). One comment recommended
that FDA mandate expanded reporting
for clinical trials only for those
companies that have had documented
poor performance in the past or for
clinical trials once a study or design has
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
been identified as posing a potential or
unforeseen risk to participants.
(Response) As previously described in
the response to Comment 1 of this
document, the agency is not adopting
the proposed SADR definition and,
instead, is adopting a definition of
‘‘suspected adverse reaction’’ that relies
on clinical judgment to determine if
there is a reasonable possibility that the
drug caused the event. While FDA
believes this definition addresses many
of the concerns about overreporting, the
agency agrees with the comments that
stated that protocols could be written to
exclude from expedited reporting
specific disease-related events that are
study endpoints. The agency does not
believe that it is appropriate to report
study endpoints as IND safety reports
for trials that are designed to evaluate
the effect of the drug on disease-related
mortality or morbidity. Therefore, the
agency added the requirement at
§ 312.32(c)(5) that study endpoints (e.g.,
mortality or major morbidity) must be
reported to FDA by the sponsor as
described in the protocol and ordinarily
would not be reported under
§ 312.32(c). However, if a serious and
unexpected adverse event occurs for
which there is evidence suggesting a
causal relationship between the drug
and the event (e.g., death from
anaphylaxis), the event must be
reported under § 312.32(c)(1)(i) as a
serious and unexpected suspected
adverse reaction even if it is a
component of the study endpoint (e.g.,
all-cause mortality). FDA does not
believe that this requirement will pose
an additional burden on sponsors or the
agency because sponsors of large
outcome trials are accustomed to
describing in the protocol how mortality
or major morbidity endpoints will be
measured and analyzed, and FDA
review divisions are accustomed to
reviewing such protocols.
The agency does not agree that the
safety reporting requirements should be
revised, as suggested by the comment, to
address specific study or design risks or
company compliance. The agency is
authorized to require additional
reporting or inspection, or to take
action, on a case-by-case basis if, for
example, such problems expose human
subjects to unreasonable and significant
risk of illness or injury, or if the sponsor
does not comply with the requirements
under § 312.32 (see e.g., § 312.42
clinical holds and requests for
modifications, § 312.44 termination).
(Comment 20) Several comments
supported the use of alternative
reporting arrangements for serious
adverse events that are not the study
endpoints (e.g., known consequences of
PO 00000
Frm 00014
Fmt 4700
Sfmt 4700
the underlying disease or condition).
These comments recommended that
these events not be reported to FDA in
an expedited manner as individual case
safety reports, but be identified in the
study protocol with clear instructions
for handling, be monitored by the
sponsor, and be reported to the agency
if, in aggregate, it appears that the
product may be causing an increase in
these adverse events. One comment
endorsed this type of arrangement
because it offers the potential for
improvements in protocol design by
providing expanded opportunity for
sponsors to discuss the ‘‘ground rules’’
for SADR reporting for specific studies
with the agency during the protocol
design phase. Two comments
recommended that FDA make clear to
investigators, sponsors, manufacturers,
and IRBs that such arrangements are
acceptable. One comment stated that
allowing this type of alternative
reporting arrangement will provide a
loophole for industry to underreport
adverse events.
(Response) Under former
§ 312.32(c)(3), sponsors were permitted
to propose alternative reporting formats
or frequencies for submitting IND safety
reports; this requirement has not
changed in this final rule. The agency
agrees with the comments
recommending that at the time of
protocol development the sponsor
identify the serious adverse events (i.e.,
known consequences of the disease or
those otherwise common in the study
population) that it plans not to report
individually in an expedited manner
but that it will monitor during the
course of the trial. FDA encourages use
of this process. Should an aggregate
analysis indicate that those events occur
more frequently in the drug treatment
group, the sponsor must then report that
information in an IND safety report
under § 312.32(c)(1)(i). However, the
agency recognizes that it is not possible,
nor desirable, to list in the protocol
every adverse event that may be
anticipated to occur in the study
population; the protocol should
therefore limit such a list to those events
that are common, even in the absence of
drug exposure. For example, in a longterm osteoporosis trial in an elderly
population, it would be reasonable to
list myocardial infarction, but
unreasonable to list acute narrow angle
glaucoma—an event that can occur in
this elderly population, but is relatively
rare. In addition, the agency believes
that there may be other situations for
which alternative reporting
arrangements are appropriate based on
the clinical circumstances. For example,
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
erowe on DSK5CLS3C1PROD with RULES
the agency may require a sponsor to
continue to report expeditiously a
medically significant suspected adverse
reaction that is listed in the investigator
brochure as observed with the drug (i.e.,
expected) so that its rate can be
carefully monitored. The agency may
also require an alternative reporting
format or frequency for clinical trials
once a study or design has been
identified as posing a potential or
unforeseen risk to participants. In other
instances, a sponsor may request that a
certain adverse event be submitted in a
different format or at a different
frequency than required. Section
312.32(c)(3) permits such arrangements.
The agency does not agree that allowing
alternative reporting formats or
frequencies creates loopholes for
sponsors to underreport, but believes
that such arrangements will lead to
greater vigilance since particular
adverse events of interest have been
identified in advance. The agency is
clarifying the language in former
§ 312.32(c)(3) that stated ‘‘FDA may
request a sponsor to submit IND safety
reports in a format or at a frequency
different than that required under this
paragraph’’ by replacing the word
‘‘request’’ with ‘‘require’’ to better reflect
the existing process.
F. Unblinding
In the preamble to the proposed rule,
FDA noted that reports from blinded
clinical studies should have the blind
broken to identify the drug product, but
that alternative arrangements could be
made with FDA for exceptions to
breaking the blind for a clinical study in
which mortality or serious morbidities
are the clinical endpoint of the study.
FDA invited comment on whether the
blind should also be broken for other
serious SADRs that are not the clinical
endpoint of the study, but occur at a rate
high enough that the overall study blind
would be threatened if each such case
were individually unblinded (68 FR
12406 at 12420).
(Comment 21) Several comments
expressed concern that breaking the
blind to identify the suspect drug could
potentially bias both the sponsor and
investigator, and suggested alternatives
to unblinding so that sponsors and
investigators could remain blinded. In
addition, several comments responded
to FDA’s request for comment on
whether the blind should be broken for
serious SADRs that are not the clinical
endpoint of the study. One comment
stated that for other serious SADRs (e.g.,
expected), if a safety signal is observed,
sponsors are obligated to unblind
studies for individual subject cases, but
other comments stated that medical
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
management of the subject who
experiences the serious SADR does not
always require unblinding. One
comment stated that the sponsor and
FDA should define in advance the
nature of such serious SADRs that
would not be subject to routine
expedited reporting and unblinding.
One comment stated that for studies in
which alternative arrangements have
been made to maintain the blind, FDA
should receive interim analyses,
disaggregated by group, which might
suggest increased overall dangers to
those getting the drug.
(Response) The agency believes that
the concerns expressed about breaking
the blind have been addressed by
clarifying the reporting requirements for
serious and unexpected suspected
adverse reactions (§ 312.32(c)(1)(i)) and
for study endpoints (§ 312.32(c)(5)), and
the provision permitting alternative
reporting arrangements (§ 312.32(c)(3)).
In particular, because there should
generally be no need to report study
endpoints in an IND safety report,
unblinding due to such endpoints
should typically not occur. In other
cases, however, where the serious,
unexpected, suspected adverse reaction
must be reported expeditiously, the
agency expects the blind to be broken.
Knowledge of the treatment received
may be essential for the medical
management of the subject and may
provide critical safety information about
the drug that could have implications
for the ongoing conduct of the trial (e.g.,
monitoring, informed consent). The
agency does not believe that unblinding
single or small numbers of informative
cases will compromise the integrity of
the study. However, if patient safety can
be assured without breaking the blind,
the agency encourages the sponsor to
discuss alternative reporting
arrangements with the appropriate FDA
review division. Any anticipated
alternative arrangements to maintain the
blind would need to be described in the
protocol, including identification of the
serious adverse events that will not be
reported on an individual basis and the
plan for monitoring and reporting
results to FDA.
(Comment 22) Several comments
made recommendations on the need for,
and role of independent data safety
monitoring boards (DSMBs), called Data
Monitoring Committees (DMCs) in
FDA’s guidance for industry entitled
‘‘Guidance for Clinical Trial Sponsors:
Establishment and Operation of Clinical
Trial Data Monitoring Committees’’ (71
FR 15421, March 28, 2006) (DMC
guidance). One comment stated that an
obligation to have an independent
DSMB would prevent routine
PO 00000
Frm 00015
Fmt 4700
Sfmt 4700
59947
unblinding. Other comments
recommended the use of DSMBs that
have processes for vetting and reporting
adverse reactions to the agency,
including monitoring for increases in
disease-related complications. One
comment recommended that the agency
concurrently amend the IRB regulations
and guidelines to incorporate a mandate
of more frequent review of overall safety
data, including a requirement for an
independent safety monitoring
committee, under predefined
circumstances. Another comment urged
the agency to require a DSMB for all
Phase 3 studies and to also require that
sponsors provide DSMB reports to IRBs.
One comment said that clarity on the
role of the DSMB for Phase 3 and 4
studies when reviewing SADRs could
help reduce redundancy of SADR
reporting evaluations by IRBs, and allow
IRBs to more efficiently focus their
attention on local SADRs.
(Response) The agency agrees that
DMCs can be useful for monitoring
adverse events and preventing routine
unblinding in certain trials. A DMC is
not required and is not necessary for
most studies, particularly those
evaluating symptomatic treatments.
DMCs are generally associated with a
large, randomized multisite trial that is
designed to evaluate treatments
intended to improve survival or reduce
the risk of major morbidity. In that case,
the independent DMC would be
expected to monitor serious events that
are study endpoints and also may assess
the rate of other known consequences of
the underlying disease or other events
that are common in the study
population. FDA’s DMC guidance also
notes another potential use for a DMC.
Some sponsors have used a DMC to
monitor the overall event rates as the
safety database accumulates in ongoing
studies (DMC guidance at p. 23). A DMC
could periodically analyze and evaluate
the aggregated, unblinded events in the
entire IND safety database to determine
if the drug is the suspected cause.
During these analyses, investigators and
study participants would remain
blinded. FDA’s DMC guidance also
provides more information on
determining the need for and the role of
a DMC. In addition, the agency’s
guidance for industry entitled
‘‘Guidance for Clinical Investigators,
Sponsors, and IRBs: Adverse Event
Reporting—Improving Human Subject
Protection’’ provides recommendations
on efficient approaches to meeting the
requirements for reporting
unanticipated problems to IRBs (74 FR
2599, January 15, 2009).
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59948
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
G. Information Sufficient to Consider
Product Administration Changes—
Proposed § 312.32(c)(1)(ii)
In addition to requiring sponsors to
provide written IND safety reports to
FDA and investigators for any serious
and unexpected adverse experience,
former § 312.32(c)(1)(i) required a
written IND safety report for ‘‘[a]ny
finding from tests in laboratory animals
that suggests a significant risk for
human subjects including reports of
mutagenicity, teratogenicity, or
carcinogenicity.’’ FDA proposed to
revise this requirement to require
sponsors to submit a written IND safety
report if the sponsor receives
information sufficient to consider
product administration changes. The
proposed rule described information
sufficient to consider product
administration changes as ‘‘information
that, based on appropriate medical
judgment, might materially influence
the benefit-risk assessment of an
investigational drug or that would be
sufficient to consider changes in either
product administration or in the overall
conduct of a clinical investigation’’ (68
FR 12406 at 12476). Examples of the
types of information that might give rise
to such a report were described as ‘‘any
significant unanticipated safety finding
or data in the aggregate from an in vitro,
animal, epidemiological, or clinical
study, whether or not conducted under
an IND, that suggests a significant
human risk, such as reports of
mutagenicity, teratogenicity, or
carcinogenicity or reports of a lack of
efficacy with a drug product used in
treating a life-threatening or serious
disease’’ (68 FR 12406 at 12476).
(Comment 23) Several comments
maintained that the threshold for
submission of this category of IND
safety report—information sufficient to
consider product administration
changes—needs clarification. Some
comments stated the ‘‘information
sufficient to consider product
administration changes’’ is too vague a
criterion on which to base a reporting
requirement and that ‘‘product
administration’’ may have different
interpretations in the context of safety.
Some comments pointed out that there
is ongoing ‘‘consideration’’ of the
implications, for product
administration, of information that
emerges during the conduct of a trial
and often, upon consideration, it will be
concluded that no changes are needed.
Some comments recommended that
there be an IND safety report only in the
event of a product administration
change or other change in the conduct
of the investigation. One comment
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
recommended that FDA consider the
implications (e.g., potential confusion)
of informing investigators about
information sufficient to consider
product administration changes before a
decision has been made about whether
to make a change. That comment
recommended that only FDA receive the
information sufficient to consider
product administration changes and that
the investigator be notified only in the
event of an actual product
administration change. Some comments
pointed out that the proposed language
does not differentiate among the range
of possible product administration
changes and thus would seem to require
an expedited report for minor changes
that do not warrant expedited reporting.
The comments suggested that there be
expedited reporting only in the event of
significant product administration
changes. One comment stated that
information sufficient to consider
product administration changes is a
reasonable category for an IND safety
report. The comment asked that FDA
clarify that significant risk to humans is
intended to include instances of
significant impairment or dysfunction.
(Response) The agency concurs that,
as proposed, the requirement may be
confusing. In response to comments, the
agency has revised the proposed
requirement for reporting data or
findings from clinical or
epidemiological studies to address the
concerns about vagueness of terms and
criteria that could lead to differences in
interpretation. The revised requirement
eliminates the association with ‘‘product
administration changes’’ and makes
clear the types of findings that would
trigger the requirement to report under
this provision. In addition, the revised
requirement also makes clear that the
findings from clinical studies that are
subject to this requirement are other
than those reported under
§ 312.32(c)(1)(i) (e.g., findings from a
drug interaction study). The agency has
revised § 312.32(c)(1)(ii) to require the
sponsor to report any findings from
epidemiological studies, pooled analysis
of multiple studies, or clinical studies
(other than those reported under
§ 312.32(c)(1)(i)),whether or not
conducted under an IND and whether or
not conducted by the sponsor, that
suggest a significant risk in humans
exposed to the drug. The provision goes
on to state that, ordinarily, such a
finding would result in a safety-related
change in the protocol, informed
consent, investigator brochure
(excluding routine updates of these
documents), or other aspects of the
PO 00000
Frm 00016
Fmt 4700
Sfmt 4700
overall conduct of the clinical
investigation.
These changes to the proposed
requirement also address the comments
concerned about potentially
prematurely notifying all investigators
prior to conclusively determining
whether a finding might change the
product administration or conduct of
the investigation because the sponsor
would report to FDA and notify all
participating investigators, as required
by § 312.32(c)(1), after that
determination has been made by the
sponsor.
In addition, FDA agrees with the
comment that ‘‘significant risk in
humans’’ would include instances of
significant impairment or dysfunction.
(Comment 24) One comment asked
that FDA clarify what is meant by
‘‘might materially influence the benefitrisk assessment’’ (68 FR 12406 at 12476).
The comment pointed out that a literal
interpretation would require an IND
safety report for a finding that is
favorable to the benefit-risk assessment
as well as a finding that is unfavorable
to the benefit-risk assessment, but
would have no effect on the clinical use
of the drug. Another comment
maintained that the term benefit-risk
has no clear meaning in the premarket
context because efficacy has not been
proven, i.e., there is no established
benefit for the product being studied.
(Response) The agency agrees that the
proposed requirement may be
confusing. Therefore, the agency has not
included the phrase ‘‘might materially
influence the benefit-risk assessment’’ in
§ 312.32(c)(1)(ii).
(Comment 25) Some comments
questioned FDA’s intent and otherwise
expressed concern about requiring IND
safety reports of lack of efficacy for a
drug intended to treat a life-threatening
or serious disease. One comment
pointed out that ‘‘lack of efficacy’’ is
rarely used in the clinical trial setting to
refer to cases of disease progression or
nonresponders. The comment
maintained that because of the difficulty
in judging lack of efficacy, such reports
should be limited to cases in which the
investigator has specifically determined
that there was lack of efficacy. One
comment maintained that the term is
incongruous in the clinical trial setting
because efficacy of the drug has not
been demonstrated. One comment
pointed out that the term ‘‘lack of
efficacy’’ is not used consistently
throughout the proposed rule (i.e.,
premarket compared to postmarket
setting).
(Response) The agency agrees with
the comment stating that the term ‘‘lack
of efficacy’’ is incongruous in the
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
clinical trial setting because the
effectiveness of the drug has generally
not been established. Therefore, the
final rule does not include this
proposed provision.
(Comment 26) One comment stated
that in vitro and animal findings should
not be lumped together with clinical
findings for purposes of the information
sufficient to consider product
administration changes IND safety
reports because in vitro and animal
findings typically are assessed
differently than clinical findings. The
comment also argued that there is
significant variation in the
interpretation of the current reporting
requirements for nonclinical findings
and recommended establishing distinct,
well-defined criteria for reporting of
nonclinical findings. The comment
recommended a separate safety report
for animal and in vitro findings with the
following criteria: (1) A drug-related
finding, (2) an unanticipated finding,
and (3) a finding that suggests a serious
risk to humans. The comment further
maintained that the company’s core
safety information about the drug
should be the basis for determining
whether the finding is unanticipated
and the term ‘‘serious’’ should be
defined, in this context, as suggesting a
significant human risk, including, but
not limited to, reports of
carcinogenicity, mutagenicity, or
teratogenicity.
(Response) The agency agrees that the
way in which in vitro and animal
findings are assessed differs from
clinical findings. To make this
distinction clear, the agency has revised
the proposed requirement to separate
reports of findings from nonclinical and
clinical studies. Under
§ 312.32(c)(1)(iii), the sponsor must
report any findings from any animal or
in vitro testing, whether or not
conducted by the sponsor, that suggest
a significant risk in humans exposed to
the drug, such as reports of
mutagenicity, teratogenicity,
carcinogenicity, or reports of significant
organ toxicity at or near the expected
human exposure. The provision states
that, ordinarily, any such findings
would result in a safety-related change
in the protocol, informed consent,
investigator brochure (excluding routine
updates of these documents), or other
aspects of the overall conduct of the
clinical investigation.
The revised requirement also
eliminates the terms ‘‘unanticipated’’
and ‘‘serious.’’ The agency agrees with
the comment that an unanticipated,
drug-related finding that suggests a
significant risk to humans would meet
the requirement for reporting.
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
(Comment 27) Two comments asked
FDA to clarify the scope of what is
meant by ‘‘an animal finding suggestive
of significant human safety risk.’’ One
comment asked whether there are any
animal findings other than
carcinogenicity, mutagenicity, or
teratogenicity that would be considered
a significant human safety risk and
whether a finding needs to originate
from a reproducible validated controlled
model. One comment stated that the
final rule should state explicitly that
only those findings of carcinogenicity,
mutagenicity, or teratogenicity that the
sponsor considers suggestive of
significant risk to humans should be
reported. The comment pointed out that
some carcinogenicity, mutagenicity, and
teratogenicity findings are known to be
species-specific or for other reasons
known not to suggest significant
potential human risk and thus should
not be subject to expedited reporting.
Another comment suggested a
distinction be made between a
nonclinical finding that requires
‘‘changes in either product
administration or in the overall conduct
of a clinical investigation’’ as opposed to
a nonclinical finding that requires
information only (e.g., action is limited
to a nonurgent update of the investigator
brochure and informed consent).
(Response) The requirement has been
revised to make it clear that, ordinarily,
a finding would be considered
suggestive of a significant risk in
humans if it results in a safety-related
change in the protocol, informed
consent, investigator brochure, or other
aspects of the overall conduct of the
clinical investigation. Nonurgent,
routine updates to the investigator
brochure and informed consent would
not meet the criteria for reporting under
this provision and should not be
reported in an expedited IND safety
report.
The sponsor must determine whether
a finding suggests a significant risk in
humans in order for the finding to be
reportable. Animal findings such as
carcinogenicity, mutagenicity or
teratogenicity are meant to be examples
of the types of findings that could
suggest a significant human risk, but
there are others that could meet the
criteria for reporting. For clarity, the
agency added another example in
§ 312.32(c)(1)(iii) (i.e., reports of
significant organ toxicity at or near the
expected human exposure). Findings
from animal studies do not necessarily
need to be replicated to meet the criteria
for expedited reporting to FDA. For
example, the agency would not expect
a long-term carcinogenicity study to be
replicated if findings from the original
PO 00000
Frm 00017
Fmt 4700
Sfmt 4700
59949
study suggested a significant risk to
humans. The validity of the model
would be a factor taken into account in
evaluating the strength of the evidence
of significant risk.
(Comment 28) Many comments
expressed concern about in vitro testing
alone as a basis for an IND safety report.
One comment pointed out that certain
types of in vitro findings that are known
to be associated with an increased risk
of carcinogenicity or mutagenicity are
always reported, but other findings are
not obviously worthy of reporting. Some
comments argued that expanding the
scope of expedited reporting to include
in vitro testing is not warranted or
useful. Some comments maintained that
in vitro testing is often exploratory and
not validated and thus lends itself to
unanticipated findings, but the clinical
implications of in vitro testing are often
not understood until later when the data
can be assessed in light of animal or
clinical findings. Given this delay in the
interpretability of in vitro findings, the
comments asked FDA to clarify when an
in vitro finding becomes reportable for
purposes of an IND safety report. Some
comments argued that the increased
reporting burden for in vitro findings
would result in large numbers of
uninformative reports that would
burden FDA and dilute the impact of
truly informative safety reports. Some
comments also maintained that
expanded reporting requirements may
deter sponsors from conducting the
kinds of in vitro testing that could
reduce the number of animal studies
needed.
(Response) In response to comments
and as stated in Comments 26 and 27,
the agency has revised the proposed
requirement § 312.32(c)(1)(iii) to make it
clear that an in vitro or animal finding
is reportable for the purposes of an IND
safety report if it suggests a significant
risk in humans exposed to the drug. The
sponsor would not report an in vitro
finding in an expedited report unless it
determined that the finding suggests a
significant risk in humans.
(Comment 29) Some comments asked
FDA to clarify the timeframe for
reporting under this requirement,
including when in vitro and animal
studies become reportable sources of
safety information by explaining how
‘‘the determination by the sponsor that
the information qualifies for reporting
under this paragraph’’ applies to
nonclinical findings. One comment
suggested that the reporting clock for in
vitro and animal findings start on the
date the final study report is completed.
One comment asked that FDA clarify
that the day that the 15-day period
begins is day zero and not day one.
E:\FR\FM\29SER1.SGM
29SER1
59950
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
(Response) The agency believes that
the revisions to this requirement have
sufficiently detailed how information
qualifies for reporting by providing
examples of the outcome of such a
finding (i.e., the finding would
ordinarily result in a safety-related
change in the protocol, informed
consent, investigator brochure, or in
other aspects of the overall conduct of
the clinical investigation). The 15–day
reporting clock begins (i.e., day zero) on
the day that the sponsor determines that
a finding suggests a significant risk in
humans. In general, it is not necessary
for a final study report to be completed
before a sponsor is able to make this
determination.
erowe on DSK5CLS3C1PROD with RULES
H. Submission of Written Reports—
Proposed § 312.32(c)(1)(iii)
Under proposed § 312.32(c)(1)(iii),
FDA proposed that each written report
may be submitted on an FDA Form
3500A or in a narrative format. Foreign
SADRs may be submitted on an FDA
Form 3500A or on a CIOMS I form. FDA
also proposed that reports of overall
findings or data in the aggregate from
published and unpublished in vitro,
animal, epidemiological, or clinical
studies must be submitted in a narrative
format. In addition, FDA proposed to
require that each written notice bear
prominent identification of its contents
and be transmitted to the FDA review
division that has responsibility for the
review of the IND. FDA also proposed
to require that if the agency determines
that additional data are needed, FDA
may require further data to be
submitted.
The agency has also revised the
requirement (final § 312.32(c)(1)(v)) to
allow for electronic submission of these
reports because the agency anticipates
that these reports will be submitted by
means other than paper in the future. In
addition, the agency has revised the
requirement to make clear that the
period of time for submitting additional
data requested by the agency is 15
calendar days, the same as required
under § 312.32(d) for submitting
followup information. The time for
submission of this additional
information was not specified in the
proposed rule.
(Comment 30) Two comments asked if
the agency would accept the CIOMS I
form for reporting domestic SADRs. One
comment strongly recommended that
the CIOMS I form be acceptable for
reporting domestic SADRs because it
would decrease workload burden,
enhance timeliness compliance with
reporting timeframes, and integrate
globally accepted formats.
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
(Response) FDA will continue, as
proposed, the current practice of
permitting submission of IND safety
reports on FDA Form 3500A or in a
narrative format for reports of domestic
suspected adverse reactions and on FDA
Form 3500A, in a narrative format or on
a CIOMS I form for reports of foreign
suspected adverse reactions. FDA
declines to permit submission of
domestic suspected adverse reactions on
the CIOMS I form because the CIOMS I
form has fewer data elements than FDA
Form 3500A (see 60 FR 11284 at 11287,
March 1, 1995; 62 FR 52237 at 52246,
October 7, 1997) and FDA believes the
additional data elements are useful for
evaluating the report. FDA is continuing
to accept the CIOMS I form for foreign
reports because we believe that
harmonization facilitates compliance
with the reporting requirements, thereby
expediting FDA’s receipt of foreign
suspected adverse reaction reports. In
the future, the agency anticipates that
electronic reporting of suspected
adverse reactions will replace the use of
paper forms.
I. Telephone and Facsimile
Transmission Safety Reports—Proposed
§ 312.32(c)(2)
FDA proposed to require that the
sponsor notify FDA by telephone or by
facsimile transmission of any
unexpected fatal or life-threatening
SADR based on the opinion of the
investigator or sponsor as soon as
possible but in no case later than 7
calendar days after receipt by the
sponsor of the minimum data set.
Because the agency anticipates that
these reports will be submitted by
means other than telephone or facsimile
in the future (e.g., electronically), the
agency has revised the requirement to
eliminate the specificity that these
reports be submitted only by telephone
or facsimile. The agency also changed
the paragraph heading to ‘‘Unexpected
fatal or life-threatening suspected
adverse reaction reports.’’ For
consistency with the agency’s decision
that assessment of whether the event is
serious and unexpected should be based
on the opinion of the sponsor (not the
investigator), the agency eliminated the
phrase ‘‘based on the opinion of the
investigator or sponsor’’ (see comment
15 of this document and
§ 312.32(c)(1)(i)). For consistency with
the agency’s decision to eliminate the
definition of ‘‘minimum data set,’’ the
agency replaced the phrase ‘‘after receipt
by the sponsor of the minimum data set’’
in the proposed codified with ‘‘after the
sponsor’s initial receipt of the
information’’ (see section III.C of this
document).
PO 00000
Frm 00018
Fmt 4700
Sfmt 4700
J. Investigations of Marketed Drugs—
Proposed § 312.32(c)(4)
FDA proposed that ‘‘a sponsor of a
clinical study under an IND for a drug
marketed in the United States is only
required to submit IND safety reports to
FDA (review division that has
responsibility for the IND) for SADRs
from the clinical study itself, whether
from domestic or foreign study sites of
the IND.’’ As proposed, the sponsor
would also be required to submit to
FDA safety information from these
clinical studies as prescribed by the
postmarketing safety reporting
requirements under §§ 310.305, 314.80,
and 600.80.
(Comment 31) One comment
supported the clarification of this
requirement. Other comments requested
further clarification. One comment
asked what should be submitted to the
IND from foreign studies not conducted
under an IND (e.g., Phase 1–3 studies,
Phase 4 postmarketing studies), both
before and after a U.S. NDA is approved.
One comment recommended that FDA
finalize a provision to require that
serious, unexpected SADRs that occur
in studies not being conducted under an
IND be submitted as expedited reports
to an IND, if one exists. This comment
also requested that FDA clarify whether
serious, unexpected SADRs observed in
IND-exempt studies of marketed drugs
are required to be submitted to both an
IND if one exists and the NDA. The
comment recommended submitting
these cases only to the NDA. One
comment stated that although the
requirement references the
postmarketing safety reporting
requirements, the postmarketing
requirements do not mention foreign
studies. This comment requested that
FDA clarify the postmarketing
requirements. Another comment stated
that for products marketed and being
studied globally, it is confusing to
decide on the appropriate route of
reporting given the different licensed
status of products in different countries
and different indications being
investigated. This comment
recommended that FDA provide a
centralized reporting location so that
FDA could route and file the report to
the appropriate application.
(Response) The only reports that must
be submitted to an IND for a drug
marketed or approved in the United
States are those arising from a study
conducted under the IND (at domestic
or foreign sites). All other reports (e.g.,
marketing experience, studies not under
an IND), must be reported in accordance
with the relevant postmarketing safety
reporting requirements. In response to
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
the comments, the agency clarified
§ 312.32(c)(4) to state that a sponsor of
a clinical study of a drug marketed or
approved in the United States that is
conducted under an IND is required to
submit IND safety reports for suspected
adverse reactions that are observed in
the clinical study at domestic or foreign
study sites. The sponsor must also
submit safety information from the
clinical study as prescribed by the
postmarketing safety reporting
requirements (e.g., §§ 310.305, 314.80,
and 600.80).
59951
Table 2 of this document summarizes
the reporting requirements for the
various scenarios identified in the
comments about submitting safety
reports from a clinical study.
TABLE 2.—SAFETY REPORTING REQUIREMENTS FROM CLINICAL STUDIES1
Drug marketed or approved2 in
the United States?
Under U.S. IND?
Trial site
Must report per postmarketing requirements?
Must report to IND?
Yes
Yes
U.S. or Foreign
Yes
Yes
Yes
No
U.S. or Foreign
No
Yes
No
Yes
U.S.or Foreign
Yes
No
No
Foreign
1 Areas
erowe on DSK5CLS3C1PROD with RULES
in the table are left blank when an IND or marketing application would not exist.
2 If a drug is approved in the United States, but is not currently being marketed in the United States, the postmarketing requirements would still
apply.
The agency does not agree with the
comment that stated that the
postmarketing requirements do not
mention foreign studies. The
postmarketing reporting requirements
do apply to postmarketing studies
conducted at foreign sites if the drug is
marketed in the United States. For
example, §§ 314.80(b) and 600.80(b)
require applicants to review all adverse
drug experience information from any
source, ‘‘foreign or domestic,’’ and
§§ 314.80(e) and 600.80(b) require
expedited reporting from a
postmarketing study, whether or not
conducted under an IND, if there is a
reasonable possibility that the drug
caused the adverse experience.
In addition, the agency revised the
proposed language listing the
postmarketing safety reporting
requirements by including the
parenthetical ‘‘(e.g., §§ 310.305, 314.80,
and 600.80),’’ thereby clarifying that the
listed postmarketing regulations are
examples and other postmarketing
safety reporting requirements may apply
(e.g., reports related to certain over-thecounter (OTC) products under the
Dietary Supplement and
Nonprescription Drug Consumer
Protection Act (Public Law 109–462);
records regarding blood or blood
products under § 606.170).
With respect to submitting reports to
FDA to one central location, currently,
postmarketing safety reports are entered
into the Adverse Event Reporting
System (AERS) database, whereas IND
safety reports are sent directly to the
review division that has responsibility
for the review of the IND. Current
capabilities do not permit direct
electronic submission through a Webbased system. However, FDA is
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
committed to adapting its business
practices to evolving technology,
including using the significant
advancements in Web-based, electronic
systems. We anticipate that, in future
rulemakings, Web-based filing of most
submissions will eventually be required.
We anticipate that when such a change
to an electronic submission system is
implemented, future guidance will
address any technical questions related
to such submissions. Until such time
that FDA develops a system to route and
manage IND safety reports within the
AERS database, or another database, the
sponsor must submit them in the
manner described in the regulations and
to the appropriate FDA location
identified in the regulations.
K. Followup—Proposed § 312.32(d)
Section 312.32(d) provides the
requirements for investigating and
submitting followup information to an
IND safety report, making minor
revisions in § 312.32(d)(2) to clarify how
relevant followup information
submitted under this paragraph must be
identified (i.e., ‘‘Followup IND Safety
Report’’). The agency proposed revising
the terminology in § 312.32(d)(3) to be
consistent with the proposed use of the
term SADR. The terminology in
§ 312.32(d)(3) is consistent with terms
used in the final rule. Former
§ 312.32(d)(4) required that results of a
sponsor’s investigation of other safety
information must be submitted, as
appropriate, in an information
amendment or annual report. The
agency has eliminated this requirement
because it is redundant—§§ 312.31 and
312.33 contain the submission
requirements for information
amendments and annual reports.
PO 00000
Frm 00019
Fmt 4700
Sfmt 4700
L. Disclaimer—Proposed § 312.32(e)
The agency proposed revising the
terminology in § 312.32(e) to be
consistent with the proposed use of the
term SADR. The terminology in
§ 312.32(e) is consistent with terms used
in the final rule.
M. Annual Reports
Although the agency did not propose
any changes to the IND annual reporting
requirements, FDA stated in the
preamble to the proposed rule that it
would not require reports of an increase
in the rate of occurrence of expected,
serious SADRs to be submitted to the
agency in an expedited manner. The
agency stated that instead, sponsors
should report this information to FDA
in their IND annual reports under
§ 312.33(b)(1) (68 FR at 12406 at 12425).
(Comment 32) One comment
disagreed with FDA’s proposal to
deviate from the ICH E2A guidance,
which recommends rapid
communication to regulatory authorities
for an increase in the rate of occurrence
of an ‘‘expected,’’ serious ADR that is
judged to be clinically important (60 FR
11284 at 11286), because expedited
reporting of this information may alert
FDA to situations of more widespread
and serious risks than were previously
known or of use in populations that had
not been previously identified as at risk.
One comment agreed with the agency’s
departure from the ICH E2A guidance
recommendation for expedited reporting
of increased frequency of serious,
expected SADRs. However, it
questioned the utility of including this
information in the IND annual report, as
proposed by FDA. The comment stated
that including this information may be
difficult, given the timing of various
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59952
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
clinical trials relative to the IND annual
reporting cycle. The comment suggested
that rather than requiring increased
frequency analysis of serious SADRs in
IND annual reports, sponsors should
routinely review incidence rates of all
serious and nonserious adverse events
within their clinical program, and report
any significant changes in the IND
annual report, when detected. Another
comment recommended that the agency
provide guidance on what would be
deemed a ‘‘clinically important’’
increased rate of reports. The comment
asked that FDA explain what the value
added of such reporting is, given the
agency’s statements that such reports
have limited reliability and have proven
to be of little value in identifying
increased incidences of serious, labeled
events in the postmarketing setting (see
62 FR 34166, June 25, 1997).
(Response) To be consistent with the
recommendations in the ICH E2A
guidance and in response to comments
about reporting serious )‘‘expected’’
SADRs, the agency is adding a
requirement under § 312.32(c)(1)(iv) that
the sponsor expeditiously report any
clinically important increase in the rate
of a serious suspected adverse reaction
over that listed in the protocol or
investigator brochure. The agency
acknowledges that baseline incidence
rates from clinical trial data as a basis
for comparison may not be available in
all cases, and as explained in the
preamble to the proposed rule (68 FR
12406 at 12425), for this reason, FDA
did not explicitly propose to require
these reports in the proposed rule.
However, the agency believes that when
rates are available, a clinically
important increase provides important
safety information and warrants
expedited, rather than annual, reporting.
Deciding if an increase in the rate of a
serious suspected adverse reaction over
that listed in the protocol or investigator
brochure is ) ‘‘clinically important’’ is a
matter of judgment based on a variety of
factors including the study population,
the nature and seriousness of the
reaction, and the magnitude of the
observed increase in rate.
The agency also agrees with the
comment that sponsors should routinely
review incidence rates of all serious and
nonserious adverse events within their
clinical program and expects that this is
current practice within the industry. If
a clinically important increase in a
serious suspected adverse reaction is
identified when compared to the rate
described in the protocol or investigator
brochure, the sponsor must report it to
FDA expeditiously. Changes in
incidence rates for the most frequent
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
nonserious adverse events would be
reported in the IND annual report.
In response to the comment that
requested clarification on the utility of
these reports in the premarket setting
when they have proven to be of little
value in the postmarketing setting, the
agency believes that there are
differences between the premarket
setting (where these reports would
usually be based on incidence rates
from clinical trials) and the
postmarketing setting (where estimation
of incidence rates from spontaneous
reports is more difficult because, for
example, the size of the exposed
population is unknown). The agency
believes that these reports contribute
information important for
understanding and updating the safety
profile of the investigational drug
product.
(Comment 33) Another comment
noted that although FDA’s proposed
rule did not address the U.S. IND
annual reporting requirements, it
recommended that they be modified to
be consistent with the ICH and EU
annual reports in light of the
finalization of the EU Clinical Trial
Directive 2001/20/EC and the
publication of their final detailed
guidance.
(Response) The agency has been
participating in the development of the
ICH draft guidance, entitled ‘‘E2F
Developmental Safety Update Report’’
(DSUR draft guidance), that describes
the format, content, and timing for
periodic reporting for an investigational
drug. As stated in the notice announcing
the availability of the DSUR draft
guidance, the DSUR would serve as an
internationally harmonized, annual
clinical trial safety report that could be
submitted in the United States in place
of an annual report for an IND (73 FR
45462, August 5, 2008). After the DSUR
draft guidance is finalized, the agency
will evaluate the need to revise our IND
annual reporting requirements to take
into account international standards and
recommendations.
(Comment 34) One comment
requested clarification of IND annual
reporting after an NDA has been
approved and clinical studies continue
under the IND, particularly in light of
adoption of the PSUR, which includes
clinical study data. The comment asked
if safety sections in the IND annual
report would be required after the NDA
has been approved and the PSUR format
is then being followed. The comment
also requested clarification on whether
the data cutoff date would be the IND
effective date, the NDA approval date,
or the international birth date.
PO 00000
Frm 00020
Fmt 4700
Sfmt 4700
(Response) Clinical development of a
drug frequently continues even after it
has been approved for marketing (e.g.,
for new indications, new dosage
strengths, different populations).
Therefore, the IND annual report
continues to be important for evaluating
and monitoring the safety of the drug. In
addition, the DSUR draft guidance
discusses the relationship between the
DSUR and PSUR when clinical studies
continue after a drug is approved for
marketing, and when to initiate a DSUR
for a marketed product. The guidance
recommends that once a drug has
received marketing approval in any
country or region, and clinical trials
continue or are initiated, both a PSUR
and a DSUR should be prepared in
accordance with directions from local
authorities (DSUR draft guidance at p.
7). After the DSUR draft guidance is
finalized, the agency will consider
whether to revise our IND annual
reporting requirements to take into
account its current thinking on the
issue, including adopting an
international birthdate. Until that time,
the data cutoff date for the IND annual
report is the IND effective date because
the annual report must be submitted to
FDA within 60 days of the anniversary
of the date that the IND went into effect
(see § 312.33).
N. Investigator Reports—Proposed
§ 312.64(b)
FDA proposed to require that an
investigator report to the sponsor any
serious SADR immediately and any
other SADR promptly unless the
protocol or investigator’s brochure
specifies a different timetable for
reporting the SADR.
(Comment 35) One comment
suggested that FDA require investigators
to report all protocol-defined treatmentemergent adverse events (TEAEs)
expeditiously regardless of their causal
attribution, but record their causality
assessment when reporting such events.
The comment defined a TEAE as an
event that emerges during treatment
having been absent pretreatment, or
worsens relative to the pretreatment
state. The comment stated that if the
agency’s SADR definition is
implemented as proposed, it is
conceivable that investigators will not
report certain TEAEs if they feel a
causal relationship can be ruled out.
Because there are no standard
guidelines for ruling out a possible
causal relationship, there could be
inconsistent causality assessments and
adverse event reporting across study
sites. Another comment stated that
applying the SADR definition to
investigator reporting could result in
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
underreporting of serious adverse
events. The comment maintained that
the investigator should report all serious
adverse events to the sponsor, without
making a causality assessment. The
comment further stated that the
proposed approach would not be in
harmony with ICH standards and
European regulatory requirements,
which require that all serious adverse
events be immediately reported to the
sponsor. One comment stated that
investigators provide an important and
informed medical review of causality,
especially in the presence of complex
disease states where many adverse
events occur as a result of the
underlying disease process. The
comment suggested that FDA provide
clear guidance on reportable causality.
(Response) As noted in Comment 1 of
this document, the agency has decided
not to adopt the proposed SADR
definition. FDA believes that there is
more uncertainty when assessment of
causality is based on an individual
event rather than on aggregate data. The
agency also believes that the sponsor is
better positioned than the individual
investigator to assess the overall safety
of the investigational drug because the
sponsor has access to serious adverse
event reports from multiple study sites
and is able to aggregate and analyze
these reports. Therefore, the agency has
determined that the sponsors should
immediately receive reports from
investigators of any serious adverse
events, without regard to causality.
However, the agency agrees that,
because the investigator is
knowledgeable about the human subject
(e.g., medical history, concomitant
medications), administers the
investigational drug, and monitors the
subject’s response to the drug, the
investigator’s view on the causal
relationship between an adverse event
and the investigational drug is
important, especially in the presence of
complex disease states where many
adverse events occur as a result of the
underlying disease process. Because the
insight from the investigator is
important for the sponsor to consider in
assessing the safety of the drug and
determining whether to report
expeditiously to FDA, the agency has
revised the requirement to require that
the investigator include an assessment
of causality in the report to the sponsor.
Revised § 312.64(b) requires
investigators to immediately report to
the sponsor any serious adverse event,
whether or not considered drug related,
including those listed in the protocol or
investigator brochure and the report
must include an assessment of whether
there is a reasonable possibility that the
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
drug caused the event. Study endpoints
that are serious adverse events (e.g., allcause mortality) must be reported in
accordance with the protocol unless
there is evidence suggesting a causal
relationship between the drug and the
event (e.g., death from anaphylaxis). In
that case, the investigator must
immediately report the event to the
sponsor.
(Comment 36) Several comments
requested clarification of the terms
‘‘immediately’’ and ‘‘promptly’’ in the
proposed requirement. The comments
disagreed with the requirement to report
other SADRs (i.e., nonserious) promptly
to the sponsor, as the term ‘‘promptly’’
implies ‘‘quickly.’’ The comments stated
that nonserious SADRs are traditionally
recorded on case report forms during
the study, then verified and collected by
the sponsor during scheduled
monitoring visits. One comment
recommended that the requirement be
revised to require investigators to
record, rather than report, other SADRs
promptly.
(Response) The agency expects that,
for serious adverse events, the
investigator would notify the sponsor
immediately. The agency recognizes
that it may take a day to collect
adequate information to confirm the
occurrence of the adverse event but
expects that as soon as the investigator
has confirmed that the event occurred,
the investigator will report it to the
sponsor without delay.
The agency agrees with the comments
that the term ‘‘promptly’’ does not
appropriately describe the best process
for documenting and notifying the
sponsor about nonserious adverse
events. Therefore, the agency has
revised § 312.64(b) to state that the
investigator must record nonserious
adverse events and report them to the
sponsor according to the timetable for
reporting specified in the protocol. The
sponsor would need to determine the
appropriate interval for collecting and
analyzing nonserious adverse event
information based on the drug under
investigation and other study
considerations, and delineate the
timetable in the protocol.
O. Bioavailability and Bioequivalence
Requirements—Proposed § 320.31(d)
FDA proposed to require that persons
conducting human bioavailability or
bioequivalence studies that are not
subject to an IND submit expedited
safety reports to FDA in accordance
with § 312.32. In the preamble to the
proposed rule (68 FR 12406 at 12415),
the agency stated that, in general,
bioavailability and bioequivalence
studies that are not being conducted
PO 00000
Frm 00021
Fmt 4700
Sfmt 4700
59953
under an IND are safe. However, the
agency is occasionally made aware of
safety-related information associated
with these types of studies, which could
reflect either a problem with the drug
product being evaluated or with the
study design being used. Timely review
of this safety information is critical to
ensuring the safety of study subjects.
FDA proposed to require that these
safety reports be transmitted to all
participating investigators and to the
appropriate FDA division in CDER (i.e.,
safety reports for the reference listed
drug would be sent to the new drug
review division that has responsibility
for that drug, safety reports for the
investigational drug product would be
sent to the Director, Division of
Bioequivalence, Office of Generic Drugs)
and each report bear prominent
identification of its contents. For
reporting purposes under § 320.31(d)(3),
an unexpected SADR would be any
SADR the specificity or severity of
which is not consistent with the U.S.
labeling for the reference listed drug.
In general, the occurrence of a serious
adverse event is very unusual in a
bioavailability or bioequivalence study
because the number of subjects enrolled
in the study is small, the subjects are
usually healthy volunteers, and drug
exposure is typically brief. For these
reasons, the occurrence of any serious
adverse event is of interest. The agency
reviewed the numbers and types of
serious adverse events that we have
received from these trials (i.e., in study
reports submitted in abbreviated new
drug applications (ANDAs)), and
determined that they are typically listed
in the labeling of the reference listed
drug and, therefore, would not be
subject to reporting under
§ 312.32(c)(1)(i) as serious and
unexpected suspected adverse reactions
because they would not meet the
regulatory definition of ‘‘unexpected.’’ In
addition, because serious adverse events
are so unusual in these studies, FDA
believes that the causality assessment is
unnecessary under these circumstances
and that it is important to review all
serious ‘‘adverse events.’’ Thus, the
proposed requirement to report serious
and unexpected SADRs would not have
served its intended purpose of alerting
the agency to serious adverse events
occurring in these trials, so the agency
has revised the requirement. The agency
continues to believe that receiving
reports from these trials is important for
human subject protection and, therefore,
has revised § 320.31(d)(3) to require that
any serious adverse event must be
reported, instead of any serious and
unexpected SADR. The person
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59954
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
conducting the study, including any
contract research organization, must
notify FDA and all participating
investigators of any serious adverse
event, as defined in § 312.32(a), from the
study as soon as possible but in no case
later than 15 calendar days after
becoming aware of its occurrence. Each
report must be submitted on FDA Form
3500A or in an electronic format that
FDA can process, review, and archive.
FDA will periodically issue guidance on
how to provide the electronic
submission (e.g., method of
transmission, media, file formats,
preparation, and organization of files).
As proposed, each report must bear
prominent identification of its contents,
i.e., ‘‘bioavailability/bioequivalence
safety report.’’ The person conducting
the study, including any contract
research organization, must also notify
FDA of any fatal or life-threatening
adverse event from the study as soon as
possible but in no case later than 7
calendar days after becoming aware of
its occurrence. Each notification under
§ 320.31(d)(3) must be submitted to the
Director, Office of Generic Drugs in
CDER. Relevant followup information to
a bioavailability/bioequivalence safety
report must be submitted as soon as the
information is available and must be
identified as such, i.e., ‘‘Followup
bioavailability/bioequivalence safety
report.’’ Upon request from FDA, the
person conducting the study, including
any contract research organization, must
submit to FDA any additional data or
information that the agency deems
necessary, as soon as possible, but in no
case later than 15 days after receiving
the request.
(Comment 37) Some comments
requested clarification about the
requirement to submit expedited safety
reports for qualifying SADRs that arise
in human bioavailability and
bioequivalence studies that do not
require an IND. The comments
requested that the agency clarify
whether this includes studies conducted
outside of the United States and how
these reports should be submitted in the
absence of an IND.
(Response) Under § 320.31(d)(3),
sponsors of human bioequivalence or
bioavailability studies that are exempt
from the IND requirements under part
312, but are conducted in the United
States, must report any serious adverse
events from the study to FDA (to the
Office of Generic Drugs in CDER) and to
all participating investigators. These
requirements do not apply to human
bioavailability and bioequivalence
studies that are exempt from the IND
requirements under part 312 and are
conducted outside of the United States.
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
However, as part of the information
required to establish that the proposed
drug product can be expected to have
the same therapeutic effect as the
reference listed product, adverse event
reports that occurred in foreign clinical
studies must be included in the ANDA
submission (see § 314.94(a)(7)).
P. Reports to Investigators and IRBs
In proposed § 312.32(c)(1)(i) and
(c)(1)(ii), FDA proposed to require that
sponsors notify FDA and all
participating investigators in a written
IND safety report of any serious and
unexpected SADR or information
sufficient to consider product
administration changes. Although both
of these requirements have been revised
(see response to Comments 15 to 17 and
23 to 29 of this document), the
requirement that FDA and all
participating investigators receive IND
safety reports for potential serious risks
that emerge during the conduct of a
clinical investigation has not changed in
this final rule (see final § 312.32(c)(1)).
In addition, under current § 312.66,
the investigator must, among other
things, assure that he or she will
promptly report to the IRB all changes
in the research activity and all
unanticipated problems involving risk
to human subjects or others, and that he
or she will not make any changes in the
research without IRB approval, except
where necessary to eliminate apparent
immediate hazards to human subjects.
The agency did not propose any changes
to this requirement.
(Comment 38) Some comments
pointed out that the proposed rule did
not change the frequency or format for
providing clinical investigators with
information on serious, unexpected
adverse events associated with the use
of a drug. One comment agreed that it
is imperative that investigators
responsible for the conduct of studies be
informed by the sponsor of findings that
could adversely affect the safety of
study participants. However, the
comment noted that this process can be
confusing and overwhelming,
particularly for investigators of IND
studies conducted outside the United
States. Several comments proposed
alternative reporting approaches that
would provide investigators with
reports that are more useful and
efficient and less confusing. One
comment recommended that the
requirements for notifying all
participating investigators be changed to
allow a periodic summary and analysis
of qualifying SADRs rather than
individual reports that are difficult to
track, aggregate, analyze, and interpret
at the investigational site. Several
PO 00000
Frm 00022
Fmt 4700
Sfmt 4700
comments encouraged FDA to further
harmonize with CIOMS VI and the EU
Clinical Trial Directive approach for
investigator notification because: (1)
Periodic (quarterly) aggregate line
listings of suspected unexpected serious
adverse reactions (SUSARs)
accompanied by a summary of the
evolving safety profile would provide
useful information to investigators and
IRBs, especially for Phase l–3 studies;
(2) presenting all serious, unexpected,
associated events in line listings
regardless of medication administered
(e.g., active drug, comparator, or
placebo) would maintain the blind to
the investigator; and (3) significant
safety issues would be communicated as
soon as possible to the investigators.
These comments stated that
investigators would recognize that these
expedited communications represent
significant safety information that is to
be immediately reviewed and provided
to their IRBs. The comments noted that
expedited reporting to FDA and
processes for updating the investigator
brochure would remain unchanged.
In addition, one comment requested
that FDA not require investigator
notification letters for investigations of
marketed products, even if conducted
under an IND, unless the investigation
is for a patient population or indication
that is different from that approved. The
comment stated that any significant new
safety information will be evaluated by
the sponsors as part of their signal
detection process and, if necessary, will
be incorporated in the product label.
The comment recommended that FDA
allow periodic line-listings to be sent to
investigators in lieu of individual
reports.
(Response) The agency is aware that
for large, multi-center trials,
investigators have expressed concern
about receiving large numbers of
individual adverse event reports that
may not be useful. The agency believes
that these final requirements will
significantly diminish the numbers of
individual reports that, in isolation, do
not provide useful information to the
investigator. For example, the
requirement under § 312.32(c)(1)(i),
described in the response to Comment
18 of the document, makes it clear that
specific events (such as known
consequences of the underlying disease
or condition under investigation or
other events that commonly occur in the
study population independent of drug
therapy) are to be reported to FDA and
all participating investigators only if
there is evidence, based on an aggregate
analysis, to suggest a causal relationship
between the drug product and the
adverse event. The rule also makes it
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
clear that study endpoints would
ordinarily not be reported as serious,
unexpected suspected adverse reactions
(response to Comment 19 of this
document). These clarifications are
expected to reduce the number of
reports that do not contribute in a
meaningful way to the developing
profile of the drug.
FDA does not agree with the comment
that suggested that investigators not be
notified of serious, unexpected
suspected adverse reactions from
investigations of marketed products
unless the investigation is for a patient
population or indication different from
that approved. Regardless of the patient
population or indication, information
about a serious, unexpected suspected
adverse reaction may influence the
investigator’s management of a clinical
trial participant and, is therefore,
critical information for the investigator
to receive.
(Comment 39) Some comments stated
that although the IRB’s charge is to have
written procedures for reporting ‘‘any
unanticipated problems involving risks
to human subjects or others,’’ the
proposed rule is silent about sending
any information to IRBs. These
comments recommended that the
agency provide guidance to sponsors,
manufacturers, investigators, and IRBs
that clearly delineates the
responsibilities of reporting SADRs to
the IRB. One comment requested that
FDA require that the IRB receive from
the sponsor the same expedited reports
that the sponsor sends to FDA and all
participating investigators (under
proposed § 312.32(c)(1)). Other
comments pointed out that IRBs are
currently overwhelmed with IND safety
reports and recommended that sponsors
provide IRBs with routine timely
aggregated reports of listings of adverse
events instead of individual reports.
Another comment suggested that
investigators be permitted to provide
these line-listings to their IRBs in lieu of
individual reports. One comment urged
FDA to adopt the CIOMS VI
recommendations for IRB notification.
(Response) The agency concurs with
the overall sentiments expressed by the
comments and has provided
recommendations for reporting adverse
event information to IRBs in our
‘‘Guidance for Clinical Investigators,
Sponsors, and IRBs: Adverse Event
Reporting—Improving Human Subject
Protection.’’ We also expect that the
more useful individual reports
submitted by sponsors to FDA and
investigators will translate into more
useful information being provided by
investigators to their IRBs. In addition,
the agency may consider revisions to
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
investigator reporting requirements to
IRBs in a separate rulemaking initiative.
Q. Miscellaneous Comments
FDA stated in the preamble to the
proposed rule that the term ‘‘sponsors’’
would be used to describe persons
subject to the premarketing safety
reporting regulations (68 FR 12406 at
12412).
(Comment 40) Two comments asked
FDA to clarify how the safety reporting
requirements apply to investigatorinitiated studies, since such studies are
not mentioned in the agency’s definition
of ‘‘sponsors.’’
(Response) The agency considers
investigator-initiated studies to be
synonymous with studies conducted by
a sponsor-investigator. A sponsorinvestigator, as defined in § 312.3, is ‘‘an
individual who both initiates and
conducts an investigation, and under
whose immediate direction the
investigational drug is administered or
dispensed. The term does not include
any person other than an individual.
The requirements applicable to a
sponsor-investigator under this part
[312] include both those applicable to
an investigator and a sponsor.’’
Therefore, the safety reporting
requirements under § 312.32 would
apply to an investigator-initiated study.
(Comment 41) One comment
suggested that FDA request that the
National Institutes of Health (NIH) and
other Federal agencies that have agreed
to the Federal Policy for the Protection
of Human Subjects (Common Rule) also
adopt the proposed regulations. The
comment stated that all participants in
the research enterprise must be fully
committed to the protection of research
participants, and fostering better and
more complete safety reporting will
support that commitment.
(Response) This final rule would
apply to FDA-regulated research
conducted by NIH and other Federal
agencies. The agency agrees that
improved safety reporting should
enhance the protection of human
subjects participating in clinical trials.
(Comment 42) FDA proposed that the
final rule would become effective 180
days after its date of publication in the
Federal Register, except for any final
rule regarding the proposal to require
that postmarketing SADRs in the
individual case safety reports be coded
using the Medical Dictionary for
Regulatory Activities (MedDRA), which
would become effective 1 year after its
date of publication in the Federal
Register.
Many comments expressed concern
that the proposed timeline for
implementing the new requirements is
PO 00000
Frm 00023
Fmt 4700
Sfmt 4700
59955
too aggressive, given its impact on
systems and processes (e.g., to develop,
test, and validate a new system). Some
comments did not believe 180 days was
sufficient implementation time unless
the final rule was significantly
modified. One comment requested that
FDA allow for a transition period for
ongoing clinical trials if FDA continues
with its interpretation of ‘‘related,’’ as
used in the proposed SADR definition.
One comment agreed with the adoption
of MedDRA for premarketing safety
reporting for clinical trials, but did not
believe that the 1-year proposed
timeline was realistic. Comments
requested other implementation
schedules, ranging from 12 to 18 months
for all the requirements.
(Response) The agency does not agree
that an effective date of 180 days after
the date of publication in the Federal
Register is too aggressive. The agency
believes that the revisions to the
requirements in this final rule will
streamline adverse event reporting and
are crucial to ensuring that timely and
accurate safety information about
clinical trials is received, analyzed, and
disseminated. Therefore, as proposed,
the agency has retained the effective
date for the final rule to be 180 days
after the date of publication in the
Federal Register. The concerns raised
by the comments about the agency’s
interpretation of ‘‘related’’ are no longer
an issue because the agency did not
adopt the SADR definition. In addition,
the agency did not propose, and is not
requiring in this final rule, the use of
MedDRA for IND safety reporting.
R. Initial Analysis of Impacts and
Paperwork Burden Estimates
For the initial analysis of impacts,
FDA estimated the costs of adding the
new premarketing safety reporting
requirements (68 FR 12406 at 12456 and
12457, table 14) (see section VI of this
document for discussion). For the initial
paperwork burden estimates, FDA
estimated the total annual reporting
burden associated with the
premarketing safety reporting
requirements, accounting for not only
the additional burdens associated with
the proposed new requirements, but
also for burdens already approved by
the Office of Management and Budget
(OMB) for requirements under thencurrent §§ 312.32 and 312.64 (68 FR
12406 at 12470, table 21) (see section
VII of this document for further
discussion).
For narrative reports based on
information sufficient to consider a
change in product administration
(discussed in section III.G of this
document), for the initial analysis of
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59956
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
impacts, FDA estimated that sponsors
would spend an additional 4 hours per
report for up to 600 IND safety reports.
For the paperwork burden, however, for
the same 600 IND safety reports, FDA
estimated that sponsors would spend a
total of 8 hours per report. The 4-hour
per report estimate in the initial analysis
of impacts accounted only for the
incremental burden of the proposed
reports from in vitro studies,
epidemiological studies, and clinical
studies and did not account for required
reports of ‘‘any finding from tests in
laboratory animals that suggests a
significant risk in human subjects’’
under then-current § 312.32(c)(1)(i)(B).
However, the 8-hour per report
paperwork burden estimate accounted
not only for the burden of complying
with the new proposed requirements,
but also the then-current requirement to
submit reports from animal tests.
(Comment 43) Comments from
industry stated that FDA
underestimated the number of IND
safety reports and that the proposed
SADR definition could increase the
volume of IND safety reports from 2-fold
to 10-fold. Furthermore, comments
claimed that any additional reports
would be uninformative. An increase in
the number of uninformative safety
reports would create an additional
burden on investigators and IRBs
without a corresponding benefit.
Comments noted that FDA’s analysis
failed to account for the potential
impact of these additional reports on
IRBs and investigators. Moreover, in
some cases, additional uninformative
reports could force sponsors to
unnecessarily break the blind of a
clinical trial, potentially reducing the
power of double-blind clinical trials to
detect safety issues and imposing
additional burdens to industry.
(Response) As discussed in response
to Comment 1 of this document, the
agency has decided not to adopt the
proposed SADR definition, and instead
adopted definitions for the terms
‘‘adverse event’’ and ‘‘suspected adverse
reaction.’’ In addition, FDA clarified the
circumstances under which IND safety
reports need to be submitted. With these
changes, we expect fewer reports.
Therefore, the comments stating that
FDA underestimated the number of IND
safety reports have been addressed.
(Comment 44) Some industry
comments stated that FDA
underestimated the number of hours
required to prepare a narrative report
based on information sufficient to
consider changes in product
administration or risk profile. These
comments stated that preparing a
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
narrative report requires more than 8
hours.
(Response) Although comments stated
that preparing a narrative report
requires more than 8 hours, none of
these comments provided estimates for
a specific number of hours. Without
other information, we are unable to
respond directly to these comments.
Nevertheless, we recognize that there
may be some situations and types of
findings that would require sponsors to
spend more time preparing a narrative
report. Therefore, to capture the
uncertainty of this estimate, FDA has
decided to use a range of hours (from 4
to 12 hours) to estimate the incremental
burden of this requirement instead of
the 4-hour estimate used in our initial
analysis of impacts (section VI of this
document) or the total 8-hour estimate
used in the initial paperwork burden
analysis (section VII of this document).
IV. Legal Authority
The premarket approval provisions of
the act authorize FDA to require that
drug labeling provide the practitioner
with adequate information to permit
safe and effective use of the drug
product. Section 505 of the Federal
Food, Drug, and Cosmetic Act (the act)
(21 U.S.C. 355) requires us to weigh
evidence of effectiveness and safety to
determine whether the evidence
supports drug approval, whether data
are adequate to permit a clinical
investigation to proceed under the IND
regulations, and/or whether a product is
appropriately labeled. Section
351(a)(2)(C)(i)(1)) of the Public Health
Service Act (the PHS Act) (42 U.S.C.
262(a)(2)(C)(i)(I)) authorizes the agency
to approve a biologics license
application (BLA) only if the applicant
demonstrates that the product is safe,
pure, and potent. Section 351(a)(2)(A) of
the PHS Act authorizes the agency to
establish, by regulation, requirements
for the approval, suspension, and
revocation of biologics licenses. Section
701(a) of the act (21 U.S.C. 371(a))
authorizes FDA to issue regulations for
the efficient enforcement of the act.
These statutory provisions authorize us
to issue regulations requiring sponsors
to submit safety information to the
agency to support an IND, NDA, ANDA,
or BLA.
V. Environmental Impact
The agency has determined under 21
CFR 25.30(h) that this action is of a type
that does not individually or
cumulatively have a significant effect on
the human environment. Therefore,
neither an environmental assessment
nor an environmental impact statement
is required.
PO 00000
Frm 00024
Fmt 4700
Sfmt 4700
VI. Analysis of Impacts
FDA has examined the impacts of the
final rule under Executive Order 12866
and the Regulatory Flexibility Act (5
U.S.C. 601–612), and the Unfunded
Mandates Reform Act of 1995 (Public
Law 104–4). Executive Order 12866
directs agencies to assess all costs and
benefits of available regulatory
alternatives and, when regulation is
necessary, to select regulatory
approaches that maximize net benefits
(including potential economic,
environmental, public health and safety,
and other advantages; distributive
impacts; and equity). The agency
believes that this final rule is not a
significant regulatory action under the
Executive order.
The Regulatory Flexibility Act
requires agencies to analyze regulatory
options that would minimize any
significant impact of a rule on small
entities. Because the new reporting
requirements are likely to impose a
minimal burden on small entities (less
than 0.2 percent of the average value of
shipments of entities with less than 10
employees), the agency believes that the
final rule will probably not have a
significant economic impact on a
substantial number of small entities.
Section 202(a) of the Unfunded
Mandates Reform Act of 1995 requires
that agencies prepare a written
statement, which includes an
assessment of anticipated costs and
benefits, before proposing ‘‘any rule that
includes any Federal mandate that may
result in the expenditure by State, local,
and tribal governments, in the aggregate,
or by the private sector, of $100,000,000
or more (adjusted annually for inflation)
in any one year.’’ The current threshold
after adjustment for inflation is $135
million, using the most current (2009)
Implicit Price Deflator for the Gross
Domestic Product. FDA does not expect
this final rule to result in any 1-year
expenditure that would meet or exceed
this amount.
In accordance with Executive Order
12866, FDA has previously analyzed the
potential economic effects of the
proposed rule. Although FDA
determined that the proposed rule was
an economically significant rule as
described in the Executive order, the
final rule covers a smaller subset of the
proposed regulatory actions and is only
related to premarket safety reporting
and safety reporting for certain
bioavailability and bioequivalence
studies. Consequently, the annual
estimated costs of this final rule are
projected to equal less than $0.7
million. We are unable to quantify the
benefits of the final rule, but expect that
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
the potential benefits of harmonized and
improved safety reporting will justify
the minimal costs of this rule.
A. Need for the Regulation
Ambiguous regulatory requirements
may cause sponsors to unnecessarily
submit certain IND safety reports to
FDA and investigators. As described in
section I of this document, lack of
clarity about definitions and regulatory
reporting requirements may create
uncertainty about when to submit an
IND safety report and may lead to overor underreporting to FDA and
investigators. Uncertainty about safety
reporting requirements can result in
reports being submitted for adverse
events when there is little evidence of
a causal relationship between the drug
and the adverse event. Such reports can
produce so-called ‘‘noise’’ in the system
and hinder the development of the
premarket safety profile of an
investigational drug. Conversely,
exempting certain bioavailability and
bioequivalence studies from safety
reporting requirements may lead to
underreporting of some serious adverse
events.
The rule will finalize definitions and
IND safety reporting standards that are
as consistent as possible with ICH
documents, require expedited reporting
of study findings suggesting a
significant risk to humans, and establish
reporting requirements for certain
bioavailability and bioequivalence
studies. Moreover, the final rule clarifies
when certain safety information, such as
study endpoints, should be reported,
potentially reducing the number of
uninformative reports sent to FDA,
participating investigators, and IRBs.
B. Costs of the Regulation (to Prepare
and Submit Safety Reports)
erowe on DSK5CLS3C1PROD with RULES
1. Number of Reports
For the initial analysis of impacts, we
estimated that sponsors would submit
up to 200 reports per year to comply
with the new requirement for safety
reporting of bioavailability and
bioequivalence studies under proposed
§ 320.31(d). No comments were received
on this estimate. Consequently, in the
final analysis of impacts, we retain our
original estimate of 200 reports per year.
In the initial analysis of impacts, we
estimated that sponsors would submit
up to 600 written IND safety reports
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
annually based on information
sufficient to consider a change in
product administration (proposed
§ 312.32(c)(1)(ii))3. Consistent with ICH
recommendations for IND safety
reporting, the proposed rule would have
clarified that sponsors should submit
written IND safety reports when they
receive information suggesting
significant human risk sufficient to
consider changes in the conduct of a
clinical trial or product administration.
Information suggesting a significant
human risk could come from animal
studies, in vitro studies,
epidemiological studies, or clinical
studies. We received no comments on
this estimate.
In contrast to the ICH
recommendation that sponsors rapidly
report an increase in the rate of
occurrence of an expected, serious
SADR, the preamble of the proposed
rule noted that sponsors should submit
this type of information in IND annual
reports under § 312.33(b)(1) (68 FR at
12406 at 12425). Because no changes to
the IND annual reports were proposed,
FDA did not estimate the incremental
impact of these reports. For the final
rule, however, increases in the
occurrence rates of serious suspected
adverse reactions over that listed in the
protocol or investigator brochure must
be reported as expedited IND safety
reports. We have insufficient
information to determine the potential
impact of reporting increases in
occurrence rates of serious suspected
adverse reactions over that listed in the
protocol or investigator brochure as
expedited reports as opposed to
including this information in annual
reports. As part of good clinical
practice, sponsors routinely review and
analyze the incidence rates of serious
and nonserious adverse events of their
investigational drugs. Therefore, we
expect that the incremental burden of
this requirement will be minimal and
3 The proposed premarketing reporting
requirement revised the existing requirements and
expanded the types of findings that sponsors should
report as expedited narrative IND safety reports. As
discussed in sections III.R and VII of this document,
the estimated average incremental burden of the
regulatory action in the initial analysis of impacts
(i.e., 4 hours) accounted for then-current
compliance (i.e., reports based on findings from
animal tests) under then-current
§ 312.32(c)(1)(i)(B)).
PO 00000
Frm 00025
Fmt 4700
Sfmt 4700
59957
estimate that sponsors will submit up to
10 additional reports per year.
Furthermore, the final rule clarifies
the definition of a suspected adverse
reaction for reporting purposes
(§ 312.32(a)) and adds a requirement
that sponsors only submit reports of
study endpoints in unusual
circumstances not described in the
protocol (§ 312.32(c)(5)). We anticipate
that by clarifying what is a suspected
adverse reaction for reporting purposes
and the circumstances under which
study endpoints should be submitted as
expedited reports, the number of
uninformative expedited reports will be
reduced, thus reducing the burden on
sponsors, investigators, IRBs, and FDA.
However, we have no information to
estimate the magnitude of this reduced
burden.
Last, the final rule clarifies safety
reporting requirements for investigators
to report to sponsors (§ 312.64(b)).
Instead of requiring that investigators
promptly report any adverse event
reasonably caused or probably caused
by the drug, the final rule requires that
investigators immediately report any
serious adverse event to the sponsor and
include an assessment of whether there
is a reasonable possibility that the drug
caused the event. Because it is common
practice for sponsors to outline similar
reporting responsibilities in their
clinical trial protocols, we assume that
this final requirement will impose no
additional burden.
2. Costs to Prepare and Submit Safety
Reports
As shown in table 3 of this document,
we estimate that it takes an average of
14 hours to prepare a safety report for
a bioavailability and bioequivalence
study. Based on 2007 hourly median
wages for the pharmaceutical
manufacturing industry, each of these
reports will cost sponsors about $950.
As discussed in Comment 44 of this
document, the additional time needed
to prepare a report of findings
suggesting a significant risk in humans
may vary. We estimate that sponsors
could spend from 4 to 12 hours
additional time to prepare a narrative
IND safety report. The average
incremental cost of a narrative IND
safety report ranges from $250 to $750
(table 3 of this document).
E:\FR\FM\29SER1.SGM
29SER1
59958
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
TABLE 3.—ESTIMATED INCREMENTAL BURDEN AND UNIT COSTS FOR IND SAFETY REPORTS
Burden (hours) and Type of Expertise Required
Type of Report
Epidemiology
and Clinical
Medicine2
Clerical1
Bioavailability and Bioequivalence Safety Reports
Regulatory
Affairs3
Total Burden
(hours)
Total Cost ($)4
2
1
11
14
950
estimate5
1
1
2
4
250
IND Safety Reports—upper estimate5
3
3
6
12
750
IND Safety Reports—lower
Numbers are rounded.
Source: U.S. Department of Labor, Bureau of Labor Statistics, May 2007 (Ref. 4).
1 Based on median hourly wages for Office and Administrative Support Occupations (43–0000) and 40 percent benefits ($24.43 = $17.44 x
1.4).
2 Based on median hourly wages for Medical and Health Services Managers (11–9111) and 40 percent benefits ($75.03 = $53.59 x 1.4).
2 Based on median hourly wages for Medical and Health Services Managers (11–9111) and 40 percent benefits ($75.03 = $53.59 x 1.4).
3 Based on median hourly wages for Management Occupations (11–0000) and 40 percent benefits ($74.96 = $53.54 x 1.4).
4 Unit costs are rounded.
5 Includes reports based on findings suggesting a significant risk in humans from epidemiological studies, pooled analysis of multiple studies,
other clinical studies, or in vitro testing. Reports from animal testing are not included (see footnote 3 of this document).
Table 4 of this document summarizes
the estimated total costs of the final
rule. Annually, sponsors will submit up
to 200 safety reports for bioavailability
and bioequivalence studies and up to
610 IND safety reports. We estimate that
the total costs of the final rule will equal
less than $0.7 million annually.
TABLE 4.—ESTIMATED TOTAL COSTS OF THE FINAL RULE
Type of Report
Unit Costs ($)
Bioavailability and Bioequivalence Safety Reports1
Annual No. of Reports
Total Annual Costs ($)
950
190,000
250 to 750
IND Safety Reports2
200
610
150,000 to 460,000
Total Costs
340,000 to 650,000
Numbers are rounded; total costs are rounded to the nearest ten thousand dollar increment.
1 We received no comments that provided sufficient information to revise our initial estimate. Because these events occur sporadically and the
number of reports will vary from year to year, these numbers represent reasonable estimates of the annual average number of reports.
2 The annual number of IND safety reports includes the proposed 600 reports of information suggesting a significant human risk (from epidemiological studies, pooled analysis of multiple studies, other clinical studies, or in vitro testing, but not from animal testing (see footnote 3 of this
document)) and an additional 10 reports of increases in the occurrence rates of serious suspected adverse reactions over that listed in the protocol or investigator brochure.
erowe on DSK5CLS3C1PROD with RULES
C. Benefits of the Regulation
Benefits for the initial analysis of
impacts were based on potential
improvements in public health from
better postmarket safety reporting and
surveillance. The definitions and other
requirements of the final rule provide a
standardized framework against which
adverse events and adverse reactions
can be evaluated, reducing ambiguity
and uncertainty about when and how to
submit IND safety reports.
The final rule adds a requirement to
submit safety reports for certain
bioavailability and bioequivalence
studies that have been exempt from
safety reporting. These studies have
been exempted from safety reporting
requirements because serious adverse
events in these types of studies are rare.
As described elsewhere in this
document, most serious adverse events
would be listed in the labeling of the
reference listed drug and thus would
not meet the threshold for expedited
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
IND safety reporting. However,
reporting such unusual events would
alert FDA to serious adverse events
occurring in these trials. For this reason,
it is prudent that FDA review such
safety information. However, we lack
sufficient information to estimate the
magnitude of these potential benefits.
The revised IND safety reporting
requirements will clarify when a
sponsor should send a narrative IND
safety report to FDA and participating
investigators. Regardless of who
conducts a study or whether a study is
conducted under an IND, any finding
that suggests a significant risk to
humans must be reported as an
expedited report. A risk is considered
significant if it will ordinarily result in
a safety-related change in the protocol,
informed consent, investigator brochure,
or conduct of the clinical investigation.
Findings of a significant risk to humans
can come from many sources, including
epidemiological studies, pooled analysis
PO 00000
Frm 00026
Fmt 4700
Sfmt 4700
of multiple studies, clinical studies,
animal testing, or in vitro testing.
Expedited reports of important safety
information will enable FDA to more
quickly review and monitor the safety
profile of investigational drugs.
However, because we lack estimates of
the impact of expedited reporting on
drug safety, we are not able to estimate
the potential benefits of this reporting
requirement.
The final rule includes a new
requirement to report clinically
important increases in the occurrence
rates of serious suspected adverse
reactions over that listed in the protocol
or investigator brochure as expedited
IND safety reports. Because these
reports are usually based on incidence
rates from clinical trials (i.e., known
exposure rates), such reports can alert
FDA to previously undetected human
safety risks. Although these reports can
occur sporadically, such reports can
provide important information that
E:\FR\FM\29SER1.SGM
29SER1
59959
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
could affect drug safety profiles.
However, we lack sufficient information
to estimate the magnitude of these
potential benefits.
Uncertainty about reporting
requirements can lead sponsors to
overreport or underreport safety events.
Overreporting can introduce so-called
‘‘noise’’ that can delay the detection of
possible safety problems.
Underreporting potential safety
problems can also delay identification
of an important new risk. We expect
that the final rule will remove some of
the uncertainty that may lead sponsors
to over- and underreport adverse events.
In addition, we expect that FDA will
receive expedited reports of safety
information that suggest a significant
risk in humans. Such reports can
promote timely review of important
drug safety information. Although we
are unable to make a quantitative
estimate of the benefits of the final rule,
we believe that the potential benefits
realized through more informative,
accurate, and timely safety reports will
justify the minimal costs of the final
rule.
D. Final Regulatory Flexibility Analysis
This final rule will harmonize certain
FDA safety reporting requirements with
international initiatives and improve the
quality of safety reporting for IND
products and certain marketed products.
According to the Table of Small
Business Size Standards, the U.S. Small
Business Administration (SBA)
considers pharmaceutical preparation
manufacturing entities (NAICS 325412)
with 750 or fewer employees and
biological product manufacturing
entities (NAICS 325414) with 500 or
fewer employees to be small. Statistics
on the classification of firms by
employment size from the U.S. Bureau
of the Census show that in 2005, at least
85 percent of pharmaceutical
manufacturing and biological product
manufacturing entities had fewer than
500 employees and would have been
considered small by SBA.
Entities have sufficient expertise to
comply with the new safety reporting
requirements. As shown in table 5 of
this document, the unit costs of a safety
report total less than 0.2 percent of the
average value of shipments for the
smallest entities. As further explained
previously, the agency does not believe
that this final rule will have a
significant economic impact on a
substantial number of small entities, but
the impact is uncertain. Although some
final requirements extend to
investigators, we anticipate no
additional burden on investigators who
would meet the SBA definition of small
entity.
TABLE 5.—UNIT COSTS OF SAFETY REPORTS AS A PERCENTAGE OF THE AVERAGE VALUE OF SHIPMENTS FOR VERY
SMALL ESTABLISHMENTS
Pharmaceutical Preparation Manufacturing (NAICS 325412)1
No. of employees
<5
Total value of shipments ($1,000)
Biological Product Manufacturing
(NAICS 325414)2
<10
<5
<10
187,933
561,636
32,011
115,307
228
339
67
109
824,268
1,656,743
477,776
1,057,862
0.0% to 0.1%
0.0% to 0.0%
0.1% to 0.2%
0.0% to 0.1%
0.1%
0.1%
0.2%
0.1%
No. of establishments
Average value of shipments ($)
Unit costs of an IND safety report as a percentage of the average value of shipments3
Unit costs of a bioavailability or bioequivalence report as a
percentage of the average value of shipments4
Numbers are rounded.
1 Source: U.S. Department of Commerce, Bureau of the Census, 2002 (Ref. 5).
2 Source: U.S. Department of Commerce, Bureau of the Census, 2002 (Ref. 6).
3 Based on a unit cost ranging from $250 to $750.
4 Based on a unit cost = $950.
erowe on DSK5CLS3C1PROD with RULES
VII. Paperwork Reduction Act of 1995
This final rule contains information
collection requirements that are subject
to review by OMB under the Paperwork
Reduction Act of 1995 (44 U.S.C. 3501–
3520) (the PRA). The title, description,
and respondent description of the
information collection provisions are
shown in the following paragraphs with
an estimate of the annual reporting
burden. Our estimate includes the time
for reviewing instructions, searching
existing data sources, gathering and
maintaining the data needed, and
completing and reviewing each
collection of information, not accounted
for under then-current § 312.32 or
§ 312.64, already approved by OMB
(OMB control number 0910–0014).
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
Title: Investigational New Drug Safety
Reporting Requirements for Human
Drug and Biological Products and Safety
Reporting Requirements for
Bioavailability and Bioequivalence
Studies in Humans
Description: The final rule clarifies
the agency’s expectations for timely
review, evaluation, and submission of
relevant and useful safety information
and implements internationally
harmonized definitions and reporting
standards for IND safety reports. The
final rule also subjects bioavailability
and bioequivalence studies to safety
reporting requirements. The final rule is
intended to improve the utility of IND
safety reports, expedite FDA’s review of
critical safety information, better protect
human subjects enrolled in clinical
PO 00000
Frm 00027
Fmt 4700
Sfmt 4700
trials, and harmonize safety reporting
requirements internationally.
The Final Rule and Estimates of
Reporting Burden
The rule finalizes revisions to the IND
safety reporting requirements found in
part 312 and the safety reporting
requirements for bioavailability and
bioequivalence studies found in part
320. For the initial PRA analysis for the
proposed rule, FDA estimated for the
annual reporting burdens for collections
of information for the entire proposal
(i.e., pre- and postmarketing safety
reporting requirements). For this PRA
analysis, FDA has estimated only for the
annual reporting burdens for collections
of information included in this final
rule (i.e., requirements found in
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59960
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
§§ 312.32, 312.64, and 320.31). In
addition, in the initial PRA analysis for
the proposed rule, FDA estimated for
the total reporting burden associated
with the proposed reporting
requirements in §§ 312.32, 312.64, and
320.31 (as opposed to only the increased
burdens associated with the proposed
rule). Because OMB has approved
paperwork burdens for many of the
reporting requirements found in
§§ 312.32 and 312.64, for purposes of
this final rule and this PRA analysis,
FDA is providing estimates for only the
additional burdens not already
approved by OMB for §§ 312.32, 312.64,
and 320.31 (OMB control number 0910–
0014). The following provisions of the
final rule contain collections of
information and the following burden
estimates are based on those discussed
in the Analysis of Impacts (section VI.B
of this document).
Section 312.32(c)(1)(i) specifies the
requirements for reporting to FDA in an
IND safety report potential serious risks
from clinical trials within 15 calendar
days for reports of serious and
unexpected suspected adverse reactions
and provides examples of what
evidence supports a suggestion that
there is a causal relationship between
the drug and the adverse event. For
purposes of this final rule, there is no
new information collection because the
reporting burden is unchanged from
former § 312.32 and the information
collection is already approved by OMB
(OMB control number 0910–0014).
Section 312.32(c)(1)(ii) requires
reporting to FDA in an IND safety report
potential serious risks from clinical
trials within 15 calendar days for
findings from epidemiological studies,
pooled analyses of multiple studies, or
other clinical studies that suggest a
significant risk in humans exposed to
the drug. This reporting requirement
was not included in former § 312.32.
Section 312.32(c)(1)(iii) specifies the
requirements for reporting to FDA in an
IND safety report potential serious risks
from clinical trials within 15 calendar
days for findings from animal or in vitro
testing that suggest a significant risk to
humans. While reports from in vitro
testing that suggest a significant risk to
humans were not required to be
reported under former § 312.32, reports
from any finding from tests in laboratory
animals were required to be reported
(former § 312.32(c)(1)(i)(B)). For
purposes of this final rule, for the
provisions that are unchanged from
former § 312.32, the information
collection is already approved by OMB
(OMB control number 0910–0014). For
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
the additional reporting requirements
(i.e., the proposed narrative reports
excluding animal testing) in the initial
PRA analysis, FDA estimated that
sponsors would spend a total of 8 hours
per report to prepare and submit these
narrative reports. In response to
comments, FDA has revised the estimate
from an incremental 4 hours to a range
from 4 hours to 12 hours per report.
Given this range, the upper estimate of
additional paperwork burden associated
with this requirement for each applicant
could be an additional 12 hours to
prepare each narrative report. Therefore,
for an additional 600 reports, FDA
estimates the total annual reporting
burden of this final rule could be as
high as 7,200 hours.
Section 312.32(c)(1)(iv) requires
reporting to FDA in an IND safety report
within 15 calendar days any clinically
important increase in the rate of
occurrence of serious suspected adverse
reactions over that listed in the protocol
or investigator brochure
(§ 312.32(c)(1)(iv)). These reports were
not required to be submitted within 15
days under former § 312.32. FDA
estimates that the minimal incremental
burden for this requirement to be
approximately 10 reports per year.
Using the same upper estimate for the
burden as discussed previously (i.e., 12
hours to prepare each report), FDA
estimates the additional burden
associated with this requirement could
be as high as 120 hours. We request
industry to comment on whether the
requirement will impose an increased
burden and if so, provide an estimate of
the reporting burden.
Section 312.32(c)(2) requires reporting
within 7 days any unexpected fatal or
life-threatening suspected adverse
reaction. For purposes of this final rule,
there is no new information collection
because the reporting burden is
unchanged from former § 312.32 and the
information collection is already
approved by OMB (OMB control
number 0910–0014).
Section 312.32(c)(4) requires a
sponsor of a clinical study of a drug
marketed or approved in the United
States that is conducted under an IND
to submit safety reports for suspected
adverse reactions that are observed in
the clinical study. For purposes of this
final rule, there is no new information
collection because the reporting burden
is unchanged from former § 312.32 and
the information collection is already
approved by OMB (OMB control
number 0910–0014).
Section 312.32(c)(5) clarifies the
circumstances under which study
PO 00000
Frm 00028
Fmt 4700
Sfmt 4700
endpoints should be submitted to FDA.
FDA believes that these clarifications to
former § 312.32 are likely to result in a
reduction in the number of expedited
reports that currently are accounted for
by OMB. However, FDA has insufficient
information to provide an estimate and
was unable to ascertain from industry
an estimate for such a reduction.
Therefore, FDA requests that industry
comment on the impact of this
provision on reporting burdens. Any
reduction in reports will be reflected the
next time the information collection for
§ 312.32 (OMB control number 0910–
0014) is extended.
Section 312.32(d)(1)-(3) requires
followup reporting requirements. For
purposes of this final rule, there is no
new information collection because the
reporting burden is unchanged from
former § 312.32 and the information
collection is already approved by OMB
(OMB control number 0910–0014).
Section 312.64(b) requires
investigators to report immediately to
the sponsor any serious adverse event
and include an assessment of whether
there is a reasonable possibility that the
drug caused the event. FDA revised
former § 312.64(b) for clarity and to
reflect current practices for investigator
reporting to sponsors. For purposes of
this final rule, there is no new
information collection because we
believe that the reporting burden is
unchanged from former § 312.64 and the
information collection is already
approved by OMB (OMB control
number 0910–0014).
Finally, § 320.31(d)(3) subjects
bioavailability and bioequivalence
studies to safety reporting requirements.
This reporting requirement was not
included in former § 320.31. Therefore,
all of these reports would be new. For
purposes of the initial PRA analysis and
this PRA analysis, FDA estimated up to
200 new safety reports required under
§ 320.31(d) from bioavailability and
bioequivalence studies. For these 200
reports, FDA estimates that it could take
applicants an additional 14 hours to
prepare and submit each report. The
burden for bioavailability and
bioequivalence safety reporting
requirements would total 2,800 hours
per year as a result of this final rule.
Description of Respondents: Business
or other for-profit organizations.
Table 6 of this document presents the
estimated annualized reporting burden
of the final rule, providing estimates for
those safety reports not already
approved under OMB control number
0910–0014.
E:\FR\FM\29SER1.SGM
29SER1
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
59961
TABLE 6.—ESTIMATED ANNUAL REPORTING BURDEN OF THE FINAL RULE1
21 CFR Section
No. of
Respondents
No. of Responses
per Respondent
Total Annual
Responses
Hours per
Response
Total Hours
320.31(d) Bioavailability and
Bioequivalence Safety Reports
10
20
200
14
2,800
312.32(c)(1)(ii) and (c)(1)(iii)
IND Safety Reports2
100
6
600
12
7,200
10
1
10
12
120
312.32(c)(1)(iv) IND Safety Reports3
1 There
are no capital costs or operating and maintenance costs associated with this collection. The estimates are for the additional burdens
beyond those already approved for then-current §§ 312.32 and 312.64.
2 Includes reports based on findings suggesting a significant risk in humans from epidemiological studies, pooled analysis of multiple studies,
other clinical studies, or in vitro testing. Reports from animal testing are not included (see footnote 3 of this document).
3 Includes reports of clinically important increases in the rate of occurrence of serious suspected adverse reactions over that listed in the protocol or investigator brochure.
The information collection provisions
of this final rule have been submitted to
OMB for review. Prior to the effective
date of this final rule, FDA will publish
a notice in the Federal Register
announcing OMB’s decision to approve,
modify, or disapprove the information
collection provisions in this final rule.
An agency may not conduct or sponsor,
and a person is not required to respond
to, a collection of information unless it
displays a currently valid OMB control
number.
erowe on DSK5CLS3C1PROD with RULES
VIII. Executive Order 13132:
Federalism
FDA has analyzed this final rule in
accordance with the principles set forth
in Executive Order 13132. FDA has
determined that the rule does not
contain policies that have substantial
direct effects on the States, on the
relationship between the National
Government and the States, or on the
distribution of power and
responsibilities among the various
levels of government. Accordingly, the
agency has concluded that the final rule
does not contain policies that have
federalism implications as defined in
the Executive order and, consequently,
a federalism summary impact statement
is not required.
IX. References
The following references have been
placed on display in the Division of
Dockets Management (HFA–305), Food
and Drug Administration, 5630 Fishers
Lane, rm. 1061, Rockville, MD 20852,
and may be seen by interested persons
between 9 a.m. and 4 p.m., Monday
through Friday. (FDA has verified the
Web site addresses, but FDA is not
responsible for any subsequent changes
to the Web sites after this document
publishes in the Federal Register.)
1. Enterprise Directorate-General, European
Commission, ‘‘Detailed Guidance on the
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
Collection, Verification and Presentation of
Adverse Reaction Reports Arising From
Clinical Trials on Medicinal Products for
Human Use,’’ revision 2, Brussels, ENTR/CT
3, April 2006 (https://ec.europa.eu/enterprise/
index_en.htm).
2. Council for International Organizations
of Medical Sciences, ‘‘Guidelines for
Preparing Core Clinical-Safety Information
on Drugs. Second Edition, Including New
Proposals for Investigator’s Brochures,’’
Report of CIOMS Working Groups III and V,
Geneva, 1999.
3. Council for International Organizations
of Medical Sciences, ‘‘Management of Safety
Information From Clinical Trials,’’ Report of
CIOMS Working Group VI, Geneva, 2005.
4. U.S. Department of Labor, Bureau of
Labor Statistics, National Industry-Specific
Occupational Employment and Wage
Estimates. NAICS 325400 - Pharmaceutical
and Medicine Manufacturing, May 2007,
extracted September 3, 2008, https://
www.bls.gov/oes/current/naics4_325400.htm.
5. U.S. Department of Commerce, Bureau
of the Census, Economic Census,
Manufacturing Industry Series,
Pharmaceutical Preparation Manufacturing,
Table 4, EC02–311–325412 (RV), 2002.
6. U.S. Department of Commerce, Bureau
of the Census, Economic Census,
Manufacturing Industry Series, Biological
Product Manufacturing, Table 4, EC02–311–
325414 (RV), 2002.
List of Subjects
21 CFR Part 312
Drugs, Exports, Imports,
Investigations, Labeling, Medical
research, Reporting and recordkeeping
requirements, Safety.
21 CFR Part 320
Drugs, Reporting and recordkeeping
requirements.
■ Therefore, under the Federal Food,
Drug, and Cosmetic Act, the Public
Health Service Act, and under authority
delegated to the Commissioner of Food
and Drugs, 21 CFR parts 312 and 320 are
amended as follows:
PO 00000
Frm 00029
Fmt 4700
Sfmt 4700
PART 312— INVESTIGATIONAL NEW
DRUG APPLICATION
1. The authority citation for 21 CFR
part 312 continues to read as follows:
■
Authority: 21 U.S.C. 321, 331, 351, 352,
353, 355, 360bbb, 371; 42 U.S.C. 262.
2. Section 312.32 is revised to read as
follows:
■
§ 312.32
IND safety reporting.
(a) Definitions. The following
definitions of terms apply to this
section:
Adverse event means any untoward
medical occurrence associated with the
use of a drug in humans, whether or not
considered drug related.
Life-threatening adverse event or lifethreatening suspected adverse reaction.
An adverse event or suspected adverse
reaction is considered ‘‘life-threatening’’
if, in the view of either the investigator
or sponsor, its occurrence places the
patient or subject at immediate risk of
death. It does not include an adverse
event or suspected adverse reaction that,
had it occurred in a more severe form,
might have caused death.
Serious adverse event or serious
suspected adverse reaction. An adverse
event or suspected adverse reaction is
considered ‘‘serious’’ if, in the view of
either the investigator or sponsor, it
results in any of the following
outcomes: Death, a life-threatening
adverse event, inpatient hospitalization
or prolongation of existing
hospitalization, a persistent or
significant incapacity or substantial
disruption of the ability to conduct
normal life functions, or a congenital
anomaly/birth defect. Important medical
events that may not result in death, be
life-threatening, or require
hospitalization may be considered
serious when, based upon appropriate
medical judgment, they may jeopardize
the patient or subject and may require
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
59962
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
medical or surgical intervention to
prevent one of the outcomes listed in
this definition. Examples of such
medical events include allergic
bronchospasm requiring intensive
treatment in an emergency room or at
home, blood dyscrasias or convulsions
that do not result in inpatient
hospitalization, or the development of
drug dependency or drug abuse.
Suspected adverse reaction means
any adverse event for which there is a
reasonable possibility that the drug
caused the adverse event. For the
purposes of IND safety reporting,
‘‘reasonable possibility’’ means there is
evidence to suggest a causal relationship
between the drug and the adverse event.
Suspected adverse reaction implies a
lesser degree of certainty about causality
than adverse reaction, which means any
adverse event caused by a drug.
Unexpected adverse event or
unexpected suspected adverse reaction.
An adverse event or suspected adverse
reaction is considered ‘‘unexpected’’ if it
is not listed in the investigator brochure
or is not listed at the specificity or
severity that has been observed; or, if an
investigator brochure is not required or
available, is not consistent with the risk
information described in the general
investigational plan or elsewhere in the
current application, as amended. For
example, under this definition, hepatic
necrosis would be unexpected (by virtue
of greater severity) if the investigator
brochure referred only to elevated
hepatic enzymes or hepatitis. Similarly,
cerebral thromboembolism and cerebral
vasculitis would be unexpected (by
virtue of greater specificity) if the
investigator brochure listed only
cerebral vascular accidents.
‘‘Unexpected,’’ as used in this definition,
also refers to adverse events or
suspected adverse reactions that are
mentioned in the investigator brochure
as occurring with a class of drugs or as
anticipated from the pharmacological
properties of the drug, but are not
specifically mentioned as occurring
with the particular drug under
investigation.
(b) Review of safety information. The
sponsor must promptly review all
information relevant to the safety of the
drug obtained or otherwise received by
the sponsor from foreign or domestic
sources, including information derived
from any clinical or epidemiological
investigations, animal or in vitro
studies, reports in the scientific
literature, and unpublished scientific
papers, as well as reports from foreign
regulatory authorities and reports of
foreign commercial marketing
experience for drugs that are not
marketed in the United States.
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
(c)(1) IND safety reports. The sponsor
must notify FDA and all participating
investigators (i.e., all investigators to
whom the sponsor is providing drug
under its INDs or under any
investigator’s IND) in an IND safety
report of potential serious risks, from
clinical trials or any other source, as
soon as possible, but in no case later
than 15 calendar days after the sponsor
determines that the information
qualifies for reporting under paragraph
(c)(1)(i), (c)(1)(ii), (c)(1)(iii), or (c)(1)(iv)
of this section. In each IND safety
report, the sponsor must identify all IND
safety reports previously submitted to
FDA concerning a similar suspected
adverse reaction, and must analyze the
significance of the suspected adverse
reaction in light of previous, similar
reports or any other relevant
information.
(i) Serious and unexpected suspected
adverse reaction. The sponsor must
report any suspected adverse reaction
that is both serious and unexpected. The
sponsor must report an adverse event as
a suspected adverse reaction only if
there is evidence to suggest a causal
relationship between the drug and the
adverse event, such as:
(A) A single occurrence of an event
that is uncommon and known to be
strongly associated with drug exposure
(e.g., angioedema, hepatic injury,
Stevens-Johnson Syndrome);
(B) One or more occurrences of an
event that is not commonly associated
with drug exposure, but is otherwise
uncommon in the population exposed
to the drug (e.g., tendon rupture);
(C) An aggregate analysis of specific
events observed in a clinical trial (such
as known consequences of the
underlying disease or condition under
investigation or other events that
commonly occur in the study
population independent of drug
therapy) that indicates those events
occur more frequently in the drug
treatment group than in a concurrent or
historical control group.
(ii) Findings from other studies. The
sponsor must report any findings from
epidemiological studies, pooled analysis
of multiple studies, or clinical studies
(other than those reported under
paragraph (c)(1)(i) of this section),
whether or not conducted under an IND,
and whether or not conducted by the
sponsor, that suggest a significant risk in
humans exposed to the drug. Ordinarily,
such a finding would result in a safetyrelated change in the protocol, informed
consent, investigator brochure
(excluding routine updates of these
documents), or other aspects of the
overall conduct of the clinical
investigation.
PO 00000
Frm 00030
Fmt 4700
Sfmt 4700
(iii) Findings from animal or in vitro
testing. The sponsor must report any
findings from animal or in vitro testing,
whether or not conducted by the
sponsor, that suggest a significant risk in
humans exposed to the drug, such as
reports of mutagenicity, teratogenicity,
or carcinogenicity, or reports of
significant organ toxicity at or near the
expected human exposure. Ordinarily,
any such findings would result in a
safety-related change in the protocol,
informed consent, investigator brochure
(excluding routine updates of these
documents), or other aspects of the
overall conduct of the clinical
investigation.
(iv) Increased rate of occurrence of
serious suspected adverse reactions.
The sponsor must report any clinically
important increase in the rate of a
serious suspected adverse reaction over
that listed in the protocol or investigator
brochure.
(v) Submission of IND safety reports.
The sponsor must submit each IND
safety report in a narrative format or on
FDA Form 3500A or in an electronic
format that FDA can process, review,
and archive. FDA will periodically issue
guidance on how to provide the
electronic submission (e.g., method of
transmission, media, file formats,
preparation and organization of files).
The sponsor may submit foreign
suspected adverse reactions on a
Council for International Organizations
of Medical Sciences (CIOMS) I Form
instead of a FDA Form 3500A. Reports
of overall findings or pooled analyses
from published and unpublished in
vitro, animal, epidemiological, or
clinical studies must be submitted in a
narrative format. Each notification to
FDA must bear prominent identification
of its contents, i.e., ‘‘IND Safety Report,’’
and must be transmitted to the review
division in the Center for Drug
Evaluation and Research or in the
Center for Biologics Evaluation and
Research that has responsibility for
review of the IND. Upon request from
FDA, the sponsor must submit to FDA
any additional data or information that
the agency deems necessary, as soon as
possible, but in no case later than 15
calendar days after receiving the
request.
(2) Unexpected fatal or lifethreatening suspected adverse reaction
reports. The sponsor must also notify
FDA of any unexpected fatal or lifethreatening suspected adverse reaction
as soon as possible but in no case later
than 7 calendar days after the sponsor’s
initial receipt of the information.
(3) Reporting format or frequency.
FDA may require a sponsor to submit
IND safety reports in a format or at a
E:\FR\FM\29SER1.SGM
29SER1
erowe on DSK5CLS3C1PROD with RULES
Federal Register / Vol. 75, No. 188 / Wednesday, September 29, 2010 / Rules and Regulations
frequency different than that required
under this paragraph. The sponsor may
also propose and adopt a different
reporting format or frequency if the
change is agreed to in advance by the
director of the FDA review division that
has responsibility for review of the IND.
(4) Investigations of marketed drugs.
A sponsor of a clinical study of a drug
marketed or approved in the United
States that is conducted under an IND
is required to submit IND safety reports
for suspected adverse reactions that are
observed in the clinical study, at
domestic or foreign study sites. The
sponsor must also submit safety
information from the clinical study as
prescribed by the postmarketing safety
reporting requirements (e.g., §§ 310.305,
314.80, and 600.80 of this chapter).
(5) Reporting study endpoints. Study
endpoints (e.g., mortality or major
morbidity) must be reported to FDA by
the sponsor as described in the protocol
and ordinarily would not be reported
under paragraph (c) of this section.
However, if a serious and unexpected
adverse event occurs for which there is
evidence suggesting a causal
relationship between the drug and the
event (e.g., death from anaphylaxis), the
event must be reported under
§ 312.32(c)(1)(i) as a serious and
unexpected suspected adverse reaction
even if it is a component of the study
endpoint (e.g., all-cause mortality).
(d) Followup. (1) The sponsor must
promptly investigate all safety
information it receives.
(2) Relevant followup information to
an IND safety report must be submitted
as soon as the information is available
and must be identified as such, i.e.,
‘‘Followup IND Safety Report.’’
(3) If the results of a sponsor’s
investigation show that an adverse event
not initially determined to be reportable
under paragraph (c) of this section is so
reportable, the sponsor must report such
suspected adverse reaction in an IND
safety report as soon as possible, but in
no case later than 15 calendar days after
the determination is made.
(e) Disclaimer. A safety report or other
information submitted by a sponsor
under this part (and any release by FDA
of that report or information) does not
necessarily reflect a conclusion by the
sponsor or FDA that the report or
information constitutes an admission
that the drug caused or contributed to
an adverse event. A sponsor need not
admit, and may deny, that the report or
information submitted by the sponsor
constitutes an admission that the drug
caused or contributed to an adverse
event.
■ 3. Section 312.64 is amended by
revising paragraph (b) to read as follows:
VerDate Mar<15>2010
14:30 Sep 28, 2010
Jkt 220001
§ 312.64
Investigator reports.
*
*
*
*
*
(b) Safety reports. An investigator
must immediately report to the sponsor
any serious adverse event, whether or
not considered drug related, including
those listed in the protocol or
investigator brochure and must include
an assessment of whether there is a
reasonable possibility that the drug
caused the event. Study endpoints that
are serious adverse events (e.g., allcause mortality) must be reported in
accordance with the protocol unless
there is evidence suggesting a causal
relationship between the drug and the
event (e.g., death from anaphylaxis). In
that case, the investigator must
immediately report the event to the
sponsor. The investigator must record
nonserious adverse events and report
them to the sponsor according to the
timetable for reporting specified in the
protocol.
*
*
*
*
*
PART 320—BIOAVAILABILITY AND
BIOEQUIVALENCE REQUIREMENTS
59963
study, including any contract research
organization, must also notify FDA of
any fatal or life-threatening adverse
event from the study as soon as possible
but in no case later than 7 calendar days
after becoming aware of its occurrence.
Each notification under this paragraph
must be submitted to the Director,
Office of Generic Drugs in the Center for
Drug Evaluation and Research at FDA.
Relevant followup information to a
bioavailability/bioequivalence safety
report must be submitted as soon as the
information is available and must be
identified as such, i.e., ‘‘Followup
bioavailability/bioequivalence safety
report.’’ Upon request from FDA, the
person conducting the study, including
any contract research organization, must
submit to FDA any additional data or
information that the agency deems
necessary, as soon as possible, but in no
case later than 15 calendar days after
receiving the request.
Dated: September 23, 2010.
Leslie Kux,
Acting Assistant Commissioner for Policy.
[FR Doc. 2010–24296 Filed 9–28–10; 8:45 am]
4. The authority citation for 21 CFR
part 320 continues to read as follows:
■
Authority: 21 U.S.C. 321, 351, 352, 355,
371.
5. Section 320.31 is amended in
paragraphs (d)(1) and (d)(2) by removing
the word ‘‘shall’’ and by adding in its
place the word ‘‘must,’’ and by removing
‘‘and’’ at the end of paragraph (d)(1) and
replacing ‘‘this chapter.’’ at the end of
paragraph (d)(2) with ‘‘this chapter;
and’’, and by adding paragraph (d)(3) to
read as follows:
■
§ 320.31 Applicability of requirements
regarding an ‘‘Investigational New Drug
Application.’’
*
*
*
*
*
(d) * * *
(3) The person conducting the study,
including any contract research
organization, must notify FDA and all
participating investigators of any serious
adverse event, as defined in § 312.32(a),
observed during the conduct of the
study as soon as possible but in no case
later than 15 calendar days after
becoming aware of its occurrence. Each
report must be submitted on FDA Form
3500A or in an electronic format that
FDA can process, review, and archive.
FDA will periodically issue guidance on
how to provide the electronic
submission (e.g., method of
transmission, media, file formats,
preparation and organization of files).
Each report must bear prominent
identification of its contents, i.e.,
‘‘bioavailability/bioequivalence safety
report.’’ The person conducting the
PO 00000
Frm 00031
Fmt 4700
Sfmt 4700
BILLING CODE 4160–01–S
DEPARTMENT OF HOMELAND
SECURITY
Coast Guard
33 CFR Part 165
[Docket No. USCG–2010–0620]
RIN 1625–AA00
Safety Zone: Monte Foundation
Firework Display, Monterey, CA
Coast Guard, DHS.
Temporary final rule.
AGENCY:
ACTION:
The Coast Guard is
establishing a temporary safety zone in
the navigable waters of Monterey Bay
off the fishing pier of Seacliff State
Beach, Santa Cruz, CA in support of the
Monte Foundation Firework Display.
This safety zone is established to ensure
the safety of participants and spectators
from the dangers associated with the
pyrotechnics. Unauthorized persons and
vessels are prohibited from entering
into, transiting through, or remaining in
the safety zone without permission from
the Captain of the Port or her designated
representative.
DATES: This rule is effective from 7 a.m.
through 9:30 p.m. on October 8, 2010.
ADDRESSES: Documents indicated in this
preamble as being available in the
docket are part of docket USCG–2010–
0620 and are available online by going
SUMMARY:
E:\FR\FM\29SER1.SGM
29SER1
Agencies
[Federal Register Volume 75, Number 188 (Wednesday, September 29, 2010)]
[Rules and Regulations]
[Pages 59935-59963]
From the Federal Register Online via the Government Printing Office [www.gpo.gov]
[FR Doc No: 2010-24296]
=======================================================================
-----------------------------------------------------------------------
DEPARTMENT OF HEALTH AND HUMAN SERVICES
Food and Drug Administration
21 CFR Parts 312 and 320
[Docket No. FDA-2000-N-0108] (formerly Docket No. 00N-1484)
RIN 0910-AG13
Investigational New Drug Safety Reporting Requirements for Human
Drug and Biological Products and Safety Reporting Requirements for
Bioavailability and Bioequivalence Studies in Humans
AGENCY: Food and Drug Administration, HHS.
ACTION: Final rule.
-----------------------------------------------------------------------
SUMMARY: The Food and Drug Administration (FDA) is amending its
regulations governing safety reporting requirements for human drug and
biological products subject to an investigational new drug application
(IND). The final rule codifies the agency's expectations for timely
review, evaluation, and submission of relevant and useful safety
information and implements internationally harmonized definitions and
reporting standards. The revisions will improve the utility of IND
safety reports, reduce the number of reports that do not contribute in
a meaningful way to the developing safety profile of the drug, expedite
FDA's review of critical safety information, better protect human
subjects enrolled in clinical trials, subject bioavailability and
bioequivalence studies to safety reporting requirements, promote a
consistent approach to safety reporting internationally, and enable the
agency to better protect and promote public health.
DATES: This rule is effective March 28, 2011.
FOR FURTHER INFORMATION CONTACT:
For information on IND safety reporting for human drug products:
Janet Norden, Center for Drug Evaluation and Research, Food and Drug
Administration, 10903 New Hampshire Ave., Bldg. 51, rm. 6324, Silver
Spring, MD 20993-0002, 301-796-2500.
For information on IND safety reporting for human biological
products: Laura Rich, Center for Biologics Evaluation and Research,
Food and Drug Administration,1401 Rockville Pike, suite 200N,
Rockville, MD 20852-1448, 301-827-6210.
SUPPLEMENTARY INFORMATION:
Table of Contents
I. Background
A. Rationale for Rulemaking
B. The Proposed Rule
II. Overview of the Final Rule
A. Definitions
B. Review of Safety Information
C. Reporting Requirements
III. Comments on the Proposed Rule
A. Definitions--Proposed Sec. 312.32(a)
B. Review of Safety Information--Proposed Sec. 312.32(b)
C. IND Safety Reports (Requirement for Minimum Data Set)--Proposed
Sec. 312.32(c)
D. Serious and Unexpected SADR--Proposed Sec. 312.32(c)(1)(i)
E. Alternative Reporting Arrangements
F. Unblinding
G. Information Sufficient to Consider Product Administration
Changes--Proposed Sec. 312.32(c)(1)(ii)
H. Submission of Written Reports--Proposed Sec. 312.32(c)(1)(iii)
I. Telephone and Facsimile Transmission Safety Reports--Proposed
Sec. 312.32(c)(2)
J. Investigations of Marketed Drugs--Proposed Sec. 312.32(c)(4)
K. Followup--Proposed Sec. 312.32(d)
L. Disclaimer--Proposed Sec. 312.32(e)
M. Annual Reports
N. Investigator Reports--Proposed Sec. 312.64(b)
O. Bioavailability and Bioequivalence Requirements--Proposed Sec.
320.31(d)
P. Reports to Investigators and IRBs
Q. Miscellaneous Comments
R. Initial Analysis of Impacts and Paperwork Burden Estimates
IV. Legal Authority
V. Environmental Impact
VI. Analysis of Impacts
A. Need for the Regulation
B. Costs of the Regulation (to Prepare and Submit Safety Reports)
C. Benefits of the Regulation
D. Final Regulatory Flexibility Analysis
VII. Paperwork Reduction Act of 1995
VIII. Executive Order 13132: Federalism
IX. References
I. Background
In the Federal Register of March 14, 2003 (68 FR 12406), FDA issued
a proposed rule to revise its regulations governing pre- and
postmarketing safety
[[Page 59936]]
reporting for human drug and biological products\1\, which appear in
parts 310, 312, 314, 320, 600, 601, and 606 (21 CFR parts 310, 312,
314, 320, 600, 601, and 606). The proposed revisions represented a
major effort to clarify and integrate several safety reporting rules
and guidance documents that had been issued by international
organizations and by FDA dating back to the 1990s. The background for
and description of these regulations and guidance documents are
described in the preamble of the proposed rule (68 FR 12406 at 12407 to
12410, Figure 1). The proposal called for the submission of comments by
July 14, 2003. At the request of industry, and to provide all
interested persons additional time to comment, the comment period was
extended until October 14, 2003 (68 FR 36527, June 18, 2003).
---------------------------------------------------------------------------
\1\ For the purposes of this document, unless otherwise
specified, all references to ``drugs'' or ``drug products'' include
human drug products and biological products that are also drugs.
---------------------------------------------------------------------------
FDA received numerous comments in response to the proposed rule,
many of which stated that the proposal would not meet its stated goals
and requested that the agency reevaluate specific aspects of the
proposal. FDA agreed with some of these comments and has reevaluated
and revised aspects of the proposal. To make the rulemaking process
more manageable, FDA has decided to issue revisions to the premarketing
and postmarketing safety reporting regulations in two separate
rulemakings. By separating these rules, the agency has been able to
reevaluate and refine each requirement in the premarketing and
postmarketing settings to better ensure that the rules will achieve
their goals.
This rule finalizes revisions to the IND safety reporting
regulations found in part 312 and the safety reporting requirements for
bioavailability and bioequivalence studies found in part 320. The
agency is working on revisions to the postmarketing safety reporting
regulations found in parts 310, 314, 600, 601, and 606 separately, and
will address these sections in a future rule. Therefore, revisions to
and comments about postmarketing safety reporting requirements found in
parts 310, 314, 600, 601, and 606 are not addressed in this rulemaking.
This document discusses information relevant to and comments about the
proposed revisions found in parts 312 and 320.
A. Rationale for Rulemaking
In the proposed rule (68 FR 12406 at 12412 to 12415), FDA described
its goals for the proposed rulemaking. Many of the stated goals were
primarily applicable to postmarketing safety reporting, but revising
and clarifying the IND safety reporting requirements was also a
critical component of FDA's stated efforts to: (1) Improve the overall
quality of safety reporting, thereby strengthening the agency's ability
to review critical safety information, (2) monitor the safety of human
drug and biological products, and (3) harmonize safety reporting
internationally. Each of these is discussed in turn in this document.
First, the revisions to the IND safety reporting requirements will
improve the overall quality of safety reporting and the agency's
ability to review critical safety information by ensuring that the
information that FDA receives in an IND safety report is relevant and
useful. Under former regulations, there may have been over-reporting of
serious adverse events for which there was little reason to believe
that the drug had caused the event, complicating or delaying FDA's
ability to detect a safety signal. In this final rule, FDA clarifies
definitions, provides examples of the types of evidence that suggest a
causal relationship for purposes of reporting a suspected adverse
reaction to the IND and participating investigators, and revises the
requirements for expedited reporting of serious and unexpected
suspected adverse reactions to the IND. The final rule also allows
sponsors to arrange alternative formats and/or frequencies for
reporting and provides that study endpoints must not be submitted as
IND safety reports except in unusual cases. These revisions not only
have an impact on which reports are sent to FDA and participating
investigators, but also affect the reports that are sent by
investigators to Institutional Review Boards (IRBs). These revisions
and clarifications will minimize reports that do not contribute to
FDA's understanding of the developing safety profile of the drug and
decrease the number of uninterpretable reports (so-called ``noise'') in
the system. In addition, the revisions and clarifications will help to
make clear under what circumstances the study blind should be broken
and when unblinding is unnecessary. Ultimately, these revisions and
clarifications should contribute toward more useful adverse reaction
information and more effective monitoring of clinical trials.
Second, by requiring expedited reports of certain safety
information that was not reported expeditiously under former IND safety
reporting requirements or bioavailability or bioequivalence
requirements, the final rule will help FDA monitor the safety of human
drug and biological products and better protect human subjects enrolled
in clinical trials. Under the final rule, FDA will receive expedited
reports of:
Findings from clinical studies, epidemiological studies or
pooled analyses of multiple studies that suggest a significant risk in
humans exposed to the drug,
Serious suspected adverse reactions that occur at an
increased rate than listed in the protocol or investigator brochure,
and
Serious adverse events from bioavailability and
bioequivalence studies.
By receiving these reports expeditiously, FDA will be better able
to monitor and evaluate the drug's safety.
Finally, FDA had proposed certain revisions to its IND safety
reporting requirements to harmonize the regulations with
recommendations by the International Conference on Harmonisation of
Technical Requirements for Registration of Pharmaceuticals for Human
Use (ICH) and by the World Health Organization's Council for
International Organizations of Medical Sciences (CIOMS), and which have
been adopted by the European Union (EU) (Ref. 1). In the preamble to
the proposed rule (68 FR 12406 at 12415, table 4), FDA detailed the
specific proposed revisions to the definitions and reporting standards
based on international recommendations in the ICH guidance ``E2A
Clinical Safety Data Management: Definitions and Standards for
Expedited Reporting'' (60 FR 11284, March 1, 1995) (ICH E2A guidance).
FDA received numerous comments, described in more detail in section III
of this document, stating that certain of FDA's proposed revisions were
inconsistent with how the provisions are interpreted and implemented in
other member ICH nations. After reviewing the comments and after
discussions with our ICH partners, FDA has revised the definitions and
reporting standards to be as consistent as possible with international
definitions and standards, recognizing that there may be
inconsistencies within ICH documents and among the other member ICH
nations' interpretations of these definitions and standards.
B. The Proposed Rule
The following describes the proposed revisions to the requirements
in parts 312 and 320. FDA proposed the following revisions to Sec.
312.32 on IND safety reports:
Replace the defined phrase ``associated with the use of
the drug''
[[Page 59937]]
with the term ``suspected adverse drug reaction (SADR),''
Require submission of expedited reports of ``information
sufficient to consider product administration changes,''
Make it clear that safety reports of overall findings or
data in the aggregate must be submitted in a narrative format,
Permit the determination that an SADR is life-threatening
to be based on the opinion of either the investigator or sponsor (as
opposed to only the investigator),
Require that the sponsor notify FDA and all participating
investigators of each SADR that is both serious and unexpected, based
on the opinion of either the investigator or sponsor (as opposed to
only the sponsor),
Require a ````minimum data set'' for each report of an
SADR submitted to FDA, and
Clarify the sources of information that sponsors must
review for safety surveillance and reporting purposes.
FDA proposed the following revision to Sec. 312.64(b):
Make it clear that the investigator must report to the
sponsor any serious SADR immediately and any other SADR promptly,
unless otherwise specified in the protocol or investigator's brochure.
FDA proposed the following revision to Sec. 320.31(d):
Make bioavailability and bioequivalence studies subject to
IND safety reporting requirements.
II. Overview of the Final Rule
This final rule amends parts 312 and 320 of FDA regulations by
revising the requirements for IND safety reporting and for
bioavailability and bioequivalence studies. This final rule reflects
revisions the agency made in response to comments on the March 2003
proposal (addressed in detail in section III of this document) and
other revisions, including editorial changes to clarify provisions and
support the agency's plain language initiative (addressed in this
section).
A. Definitions
The definitions section for the IND safety reporting regulations
(Sec. 312.32(a)) now includes the following five terms:
Adverse event,
Life-threatening adverse event or life-threatening
suspected adverse reaction,
Serious adverse event or serious suspected adverse
reaction,
Suspected adverse reaction, and
Unexpected adverse event or unexpected suspected adverse
reaction.
FDA has revised and clarified terms and definitions that were in
the proposed rule. First, as discussed in detail in section III of this
document, the two terms ``adverse event'' and ``suspected adverse
reaction'' replace the proposed definition of ``suspected adverse drug
reaction (SADR).'' The definitions ``adverse event'' and ``suspected
adverse reaction'' also replace the phrase ``associated with the use of
the drug'' defined in former Sec. 312.32(a). The definitions of the
terms ``adverse event'' and ``suspected adverse reaction'' make clear a
distinction in the degree of evidence of a causal relationship between
the drug and the adverse event within these terms.
Second, the final rule requires that the determination for
reporting purposes about whether an adverse event or suspected adverse
reaction is ``life-threatening'' or ``serious'' be based on the opinion
of either the investigator or sponsor. FDA had proposed this revision
for the definition of ``life-threatening SADRs,'' and the agency
decided that the determination about whether an adverse event or
suspected adverse reaction is ``serious'' is comparable to the
determination of whether it is life-threatening. Therefore, FDA revised
the definition ``serious adverse event or serious suspected adverse
reaction'' to specify that the determination of seriousness be based on
the opinion of either the investigator or sponsor. In addition, FDA
eliminated the definition of ``disability'' as a separate term and
includes the meaning of the term in the definition of ``serious adverse
event or serious suspected adverse reaction.''
Third, the final rule makes clear what adverse events or suspected
adverse reactions are considered unexpected. The proposed definition of
``unexpected SADR'' included the following sentence from the then-
current definition for ``unexpected adverse drug experience'' (with
minor clarification): ```Unexpected' as used in this definition, refers
to an SADR that has not been previously observed (e.g., in the
investigator brochure); it does not refer to an SADR that might be
anticipated from the pharmacological properties of the drug product.''
To this clarification, FDA proposed to add the following new sentence:
``SADRs that are mentioned in the investigator's brochure as occurring
with a class of drugs but not specifically mentioned as occurring with
the particular drug are considered unexpected.'' In this final rule,
FDA combined these proposed sentences to read as follows:
```Unexpected,' as used in this definition, also refers to adverse
events or suspected adverse reactions that are mentioned in the
investigator brochure as occurring with a class of drugs or as
anticipated from the pharmacological properties of the drug, but are
not specifically mentioned as occurring with the particular drug under
investigation.'' This revision makes clear that adverse events that
have not been previously observed with the drug under investigation,
but are predicted to occur based on the class of the drug or
pharmacological properties of the drug are considered ``unexpected''
for reporting purposes.
B. Review of Safety Information
The final rule clarifies what safety information must be reviewed
under Sec. 312.32(b). The proposal would have required sponsors to
review ``reports from foreign regulatory authorities that have not been
previously reported to FDA by the sponsor.'' FDA has deleted the phrase
``that have not been previously reported to FDA by the sponsor,''
because it confuses the review with the reporting requirements. FDA
expects sponsors to review all information, but to avoid duplicate
reporting to the agency. In addition, the final rule clarifies the
agency's expectations for analysis of previous, similar reports (Sec.
312.32(c)(1)).
C. Reporting Requirements
In Sec. 312.32(c), the final rule clarifies how and when to submit
IND safety reports to FDA and participating investigators, including
the requirement in Sec. 312.32(c)(1)(v) that certain reports be
submitted in a narrative format (proposed Sec. 312.32(c)(1)(iii)). It
provides examples of the kinds of evidence that suggest a causal
relationship between the drug and the adverse event when determining
whether a serious and unexpected adverse event qualifies for expedited
reporting (Sec. 312.32(c)(1)(i)). The final rule also requires that
sponsors submit expedited reports of findings from clinical studies,
epidemiological studies, or pooled analyses of multiple studies that
suggest a significant risk in humans (Sec. 312.32(c)(1)(ii)); findings
from animal or in vitro testing that suggests a significant risk in
humans (Sec. 312.32(c)(1)(iii)); and reports of an increased rate of
occurrence of serious suspected adverse reactions over that listed in
the protocol or investigator brochure (Sec. 312.32(c)(1))(iv)). The
final rule also provides for alternative reporting arrangements (Sec.
312.32(c)(3)) and provides that study endpoints not be reported except
in unusual cases (Sec. 312.32(c)(5)).
Furthermore, FDA has made it clear in Sec. 312.32(c)(1)(v) that
the period of time for submitting additional data requested by the
agency is 15 calendar
[[Page 59938]]
days (i.e., the same period of time that is allowed for submitting
followup information under Sec. 312.32(d)(3)). In addition, the agency
revised several provisions to allow for electronic submission of
reports. First, in Sec. 312.32(c)(1)(v) ``Submission of IND safety
reports,'' FDA renamed and revised proposed Sec. 312.32(c)(1)(iii)
``Submission of written reports.'' Second, FDA revised proposed Sec.
312.32(c)(2) ``Telephone and facsimile transmission safety reports'' to
eliminate the specificity that unexpected fatal or life-threatening
reports be submitted only by telephone or facsimile transmission so
that other means of rapid communication (e.g., e-mail) may be accepted
in the future. FDA also renamed the provision to ``Unexpected fatal or
life-threatening suspected adverse reaction reports.'' Last, in Sec.
320.31(d)(3), FDA revised the proposed requirement for submission of
IND safety reports and unexpected fatal or life-threatening reports
from bioavailability and bioequivalence studies to mirror these
revisions.
The final rule allows for alternative reporting arrangements, as
provided in former Sec. 312.32(c)(3). However, the agency revised the
statement, ``FDA may request a sponsor to submit IND safety reports in
a format or at a frequency different than that required under this
paragraph'' by replacing the word ``request'' with ``require'' to
reflect the existing process. In addition, the final rule clarifies the
reporting requirements for clinical investigations of drug products
that are marketed in the United States (Sec. 312.32(c)(4)).
The final rule makes minor editorial changes to Sec. 312.32(d)(2)
to clarify the followup reporting requirements. In addition, the agency
eliminated the redundant submission requirements for information
amendments and annual reports under Sec. 312.32(d)(4) because they are
already contained in Sec. Sec. 312.31 and 312.33.
The final rule clarifies the requirements for investigators to
submit reports of serious adverse events to the sponsor and clarifies
the requirement for reporting study endpoints that are serious adverse
events (Sec. 312.64(b)).
Finally, the final rule requires that applicants submit to FDA
reports of serious adverse events from bioavailability and
bioequivalence studies. Proposed Sec. 320.31(d) would have required
that these studies be subject to the proposed IND safety reporting
requirements, thereby requiring all reports under proposed Sec. 312.32
(e.g., reports of serious and unexpected SADRs, reports of information
sufficient to consider product administration changes). FDA has
tailored the rule to require only those reports that FDA believes would
be most informative (i.e., reports of all serious adverse events). FDA
also revised this provision to make it consistent with the final
revisions for submission of IND safety reports and reports of any fatal
or life-threatening adverse event. The final rule requires that reports
must be submitted to the Office of Generic Drugs.
Table 1 of this document identifies the changes from the proposed
rule in the IND safety reporting requirements that the agency made in
this final rule.
Table 1--Changes Made by the Final Rule From the Proposed Rule
----------------------------------------------------------------------------------------------------------------
Description of Change See comment or section of this document
21 CFR Section in Final Rule (identified in parentheses) for more detailed information regarding
the change.
----------------------------------------------------------------------------------------------------------------
312.32(a) Adverse event Added definition for ``adverse event'' (1)
----------------------------------------------------------------------------------------------------------------
312.32(a) Life-threatening adverse event Made minor editorial revisions for clarity, including
or life-threatening suspected adverse language changes to accommodate deletion of ``SADR'' definition and
reaction use of alternative terminology (2)
----------------------------------------------------------------------------------------------------------------
312.32(a) Serious adverse event or Changed language to accommodate deletion of ``SADR''
serious suspected adverse reaction definition and use of alternative terminology (6)
Incorporated the definition from former Sec. 312.32(a) of
``disability'' within the definition of ``serious'' (III.A.2)
Revised so that the seriousness determination is based on the
opinion of either the sponsor or investigator (6)
----------------------------------------------------------------------------------------------------------------
312.32(a) Suspected adverse reaction Replaced the term ``SADR'' with the term ``suspected adverse
reaction,'' clarifying the meaning of ``reasonable possibility''
within the definition (1)
----------------------------------------------------------------------------------------------------------------
312.32(a) Unexpected adverse event or Revised to make clear that ``unexpected'' adverse events or
unexpected suspected adverse reaction suspected adverse reactions include those that may be anticipated
from the pharmacological properties of the drug, or that occur with
members of the drug class, but that have not previously been observed
with the drug under investigation (8)
----------------------------------------------------------------------------------------------------------------
312.32(b) Review of safety information Made minor editorial changes for clarity and deleted the
phrase ``that have not been previously reported to FDA by the
sponsor'' (II)
----------------------------------------------------------------------------------------------------------------
312.32(c)(1) IND safety reports Withdrew the proposed requirement for each report of an SADR
to contain a minimum data set and to maintain records of efforts to
obtain a minimum data set (5, 13, and 14)
----------------------------------------------------------------------------------------------------------------
312.32(c)(1)(i) Serious and unexpected Clarified agency's expectation for analysis of previous,
suspected adverse reactions similar reports or any other relevant information (16)
Withdrew the requirement that the causality assessment be
based on the opinion of the investigator or the sponsor (15)
Provided examples of the types of evidence that suggest a
causal relationship between the drug and the adverse event (18 to 21)
----------------------------------------------------------------------------------------------------------------
312.32(c)(1)(ii) Findings from other Revised proposed reports of ``Information sufficient to
studies consider product administration changes'' to clarify agency
expectations of reports from clinical studies, epidemiological
studies or pooled analyses of multiple studies that suggest a
significant risk in humans (23 to 25)
----------------------------------------------------------------------------------------------------------------
[[Page 59939]]
312.32(c)(1)(iii) Findings from animal Revised proposed reports of ``Information sufficient to
or in vitro testing consider product administration changes'' to clarify agency
expectations of reports from animal or in vitro testing that suggests
a significant risk in humans (26 to 29)
----------------------------------------------------------------------------------------------------------------
312.32(c)(1)(iv) Increased rate of Added the requirement for reports of any clinically important
occurrence of serious suspected adverse increase in the rate of a serious suspected adverse reaction over
reactions that listed in the protocol or investigator brochure (32)
----------------------------------------------------------------------------------------------------------------
312.32(c)(1)(v) Submission of IND safety Revised to allow for electronic submission of IND safety
reports reports and clarified time period for reporting additional data or
information requested by FDA (II)
----------------------------------------------------------------------------------------------------------------
312.32(c)(2) Unexpected fatal or life- Revised to eliminate the specificity that unexpected fatal or
threatening suspected adverse reaction life-threatening suspected adverse reaction reports be submitted only
reports by telephone or facsimile transmission and renamed the requirement
(II)
----------------------------------------------------------------------------------------------------------------
312.32(c)(3) Reporting format or Replaced ``request'' with ``require'' (20)
frequency
----------------------------------------------------------------------------------------------------------------
312.32(c)(4) Investigations of marketed Clarified requirements for investigations of marketed drugs
drugs (31)
----------------------------------------------------------------------------------------------------------------
312.32(c)(5) Reporting study endpoints Added requirement that study endpoints (e.g., mortality or
major morbidity) must be reported according to the protocol instead
of as IND safety reports except when there is evidence suggesting a
causal relationship between the drug and the event (19 and 21)
----------------------------------------------------------------------------------------------------------------
312.32(d) Followup Deleted provision that required safety information to be
submitted in an information amendment or annual report and made minor
editorial changes for clarity (III.K)
----------------------------------------------------------------------------------------------------------------
312.64(b) Investigator reports Clarified requirements for investigator reports (35 and 36)
----------------------------------------------------------------------------------------------------------------
320.31(d) Applicability of requirements Revised to require that persons conducting bioavailability
regarding an ``Investigational New Drug and bioequivalence studies report all serious adverse events (II)
Application'' Revised to make consistent with requirements for submission
of IND safety reports and reports of any fatal or life-threatening
adverse event (II)
----------------------------------------------------------------------------------------------------------------
III. Comments on the Proposed Rule
The agency received 110 comments in the docket for the March 14,
2003, proposed rule on premarket and postmarket safety reporting
revisions. Comments were received from prescription and nonprescription
drug manufacturers and related companies; trade organizations
representing drug manufacturers and other interested parties; blood
banks and transfusion facilities; international organizations and non-
U.S. agencies; professional associations and organizations;
consultants; contract research organizations; academic institutions;
health care and consumer advocacy organizations, individual physicians,
pharmacists, and consumers; and others.
To make it easier to identify comments and our responses, the word
``Comment,'' in parentheses, appears before the comment's description,
and the word ``Response,'' in parentheses, appears before our response.
We have numbered each comment to help distinguish between different
comments. Similar comments are grouped together under the same number.
The number assigned to each comment is purely for organizational
purposes and does not signify the comment's value or importance or the
order in which it was received. Comments addressing the proposed
requirements for IND safety reporting and bioavailability and
bioequivalence studies and the agency's responses follow:
A. Definitions--Proposed Sec. 312.32(a)
1. Suspected Adverse Drug Reaction (SADR)
FDA proposed to add the term ``suspected adverse drug reaction
(SADR)'' and define the term as follows: ``A noxious and unintended
response to any dose of a drug product for which there is a reasonable
possibility that the product caused the response. In this definition,
the phrase `a reasonable possibility' means that the relationship
cannot be ruled out.''
(Comment 1) Nearly all of the comments overwhelmingly opposed the
agency adopting the proposed definition of SADR and strongly encouraged
the agency to abandon the proposed definition for many reasons,
including the following:
Many comments did not agree that ``reasonable
possibility'' should be defined as ``the relationship cannot be ruled
out.'' Most comments stated that this interpretation makes the
definition overly broad and will lead to reporting almost every
serious, unexpected adverse event because no event could ever be
completely ruled out.
Many comments stated that although the proposed definition
was similar to the definition contained in the ICH E2A guidance, the
agency's interpretation was inconsistent with the guidance. The ICH E2A
guidance makes clear that a causality assessment is required for
clinical investigations and that a ``reasonable causal relationship''
is meant to convey in general that there are facts (evidence) or
arguments to suggest a causal relationship. The comments expressed
concern that the agency's interpretation of ``reasonable possibility''
would lead to inconsistencies in globally conducted studies and
reports.
Many comments asserted that the significantly increased
numbers of expedited reports that could result from the proposed
definition might dilute real safety signals, making them harder to
detect. The lengthy in depth investigations needed to rule out the
increased number of false positive associations would take away
resources from other safety surveillance efforts and potentially lead
to a delay in identification of real signals.
[[Page 59940]]
Several comments expressed concern that the proposed
definition would have a negative impact on the conduct of clinical
trials. In addition to sharply increasing the number of reports of
cases from clinical trials that would need to be sent to FDA in an
expedited manner, sponsors and investigators would have to break the
blind for nearly all subjects with serious, unexpected SADRs because
the relationship between drug and the event could not definitively be
ruled out. Increased unblinding would compromise the integrity of well-
regulated clinical investigations, lead to fewer patients completing a
trial, necessitate larger patient enrollment, and lengthen the timeline
for new product development, possibly leading to higher costs for
marketed drugs. One comment expressed concern that, to minimize
unblinding, studies would be designed to exclude patients with serious
medical conditions who are likely to experience serious adverse events
during the study period, thereby limiting the applicability of study
results.
Many comments also stated that the proposed definition would result
in significant increases in meaningless individual expedited reports
being sent to already overburdened IRBs and investigators. The comments
pointed out that an unintended effect of the increase in volume of
reports may be to reduce an investigator's and IRB's vigilance in
detecting adverse events.
Several comments expressed concern that the proposed
definition would dilute the utility of drug product labeling because
many more events would be regarded as ``drug related'' even though the
likelihood of a true causal relationship is minimal.
Several comments stated that the ``S'' abbreviation for
``suspected'' in SADR could be confused with the ``S'' abbreviation for
``serious'' in SAE (serious adverse event).
The majority of the comments recommended that reporting adverse
events from clinical trials should be based on a scientific or medical
judgment that there is a possible causal relationship between the drug
and the event, rather than simply being unable to unequivocally exclude
a drug's role. The comments suggested several alternatives to the
agency's proposed definition, including the following:
Several comments recommended that the definition of an
adverse reaction encompass all of the concepts presented within the ICH
E2A guidance, which are supported by CIOMS and presented in the
European Union Clinical Trial Directive. Comments recommended that the
definition of reasonable possibility be technically consistent with the
ICH E2A guidance definition and clearly delineate the concept of
``reasonable causal relationship'' as conveying in general that there
are facts (evidence) or arguments to suggest a causal relationship.
Some comments supported retaining FDA's former definition
of ``associated with the use of the drug'' as ``there is a reasonable
possibility that the experience may have been caused by the drug.''
Three comments supported adopting the proposed definition because
they considered it an inclusive, conservative approach to adverse event
reporting.
(Response) Based on the comments, and on review of definitions and
terminology used in the ICH E2A guidance and in former Sec. 312.32,
the agency has decided not to adopt the proposed definition for
``suspected adverse drug reaction (SADR).'' The agency agrees with the
comments stating that there should be a causality assessment applied
and that the threshold for reporting should be that there is a
``reasonable possibility'' that the drug caused the adverse event. The
agency also believes that it is important to use definitions that are
clear and consistent, and in harmony with those used internationally.
The agency believes that the comments raised legitimate concerns
that the proposed definition was too broad and could have a negative
impact on clinical trials, IRBs, investigators, signal detection, and
drug labeling. Instead of adopting the proposed definition, the agency
has adopted the terms for ``adverse event'' and ``suspected adverse
reaction'' in the definition section of this final rule, which
addresses these concerns. The definitions of these terms should
contribute to harmonization of safety reporting to regulatory
authorities worldwide because they are consistent with the concepts and
definitions adopted by the ICH E2A guidance and CIOMS. The terms are
defined as follows:
``Adverse event'' means any untoward medical occurrence
associated with the use of a drug in humans, whether or not considered
drug related. (For the purposes of this definition, ``untoward'' means
unfavorable, negative, or harmful).
``Suspected adverse reaction'' means any adverse event for which
there is a reasonable possibility that the drug caused the adverse
event. For the purposes of IND safety reporting, ``reasonable
possibility'' means there is evidence to suggest a causal relationship
between the drug and the adverse event. Suspected adverse reaction
implies a lesser degree of certainty about causality than adverse
reaction, which means any adverse event caused by a drug.
These definitions reflect the varying degrees of certainty that are
part of a causality assessment. For example:
An adverse event (also referred to as an ``adverse
experience'') is any event observed or reported that is associated with
the use of the drug, without regard to causality.
A suspected adverse reaction is a subset of all adverse
events in which there is a reasonable possibility that the drug caused
the event.
An adverse reaction, described within the definition, is a
subset of all suspected adverse reactions for which there is reason to
conclude that the drug caused the event.
With this change from the proposed definition, the basis that the
agency has established for assessing the degree of certainty about
causality between a drug and an adverse event for the purposes of
expedited IND safety reporting has not changed from former Sec.
312.32(c). The sponsor must continue to evaluate the evidence and use
its judgment to determine whether an adverse event meets the definition
of suspected adverse reaction and qualifies for expedited reporting
under Sec. 312.32(c). The agency has also clarified the requirements
for reporting a serious and unexpected suspected adverse reaction under
Sec. 312.32(c)(1)(i) to assist sponsors with making this determination
(see Comment 18 of this document).
Finally, the agency has concluded that abbreviations are
potentially confusing (e.g., the ``S'' abbreviation for ``suspected''
in SADR could be mistaken for an abbreviation of the term ``serious'').
Although the agency has retained the term ``suspected'' in ``suspected
adverse reaction,'' our preferred approach is to avoid use of any
abbreviation (e.g., ``SAR'' for ``suspected adverse reaction''). The
agency believes that sponsors are familiar with the term ``suspected''
and its use by the European Commission and CIOMS (e.g., the acronym
``SUSAR'' means ``suspected, unexpected, serious adverse reaction'' in
guidance documents and working group reports (for example, see Ref.
1)).
Because the agency is not adopting the proposed definition of
``suspected adverse drug reaction (SADR),'' other proposed definitions
(e.g., ``serious SADR,'' ``life-threatening SADR'') and requirements
that used this terminology have been revised in this final rule to use
the terms ``adverse event'' or ``suspected adverse reaction'' as
appropriate.
[[Page 59941]]
2. Disability
The proposed rule included a definition of the term ``disability''
to mean a substantial disruption of a person's ability to conduct
normal life functions. Because the term ``disability'' appeared only
within the definition of ``serious SADR'' in the proposed rule, the
agency eliminated the definition of ``disability'' as a separate term
in this final rule. Instead, the agency revised the definition of
``serious adverse event or serious suspected adverse reaction'' in this
final rule to incorporate the definition of ``disability'' by replacing
the phrase ``a persistent or significant disability/incapacity'' with
``a persistent or significant incapacity or substantial disruption of
the ability to conduct normal life functions.'' Thus, in the final
rule, the term disability is replaced by the proposed definition in the
one place where it appeared, and the definition itself has been
deleted.
3. Life-Threatening Suspected Adverse Drug Reaction (SADR)
FDA proposed the term ``life-threatening suspected adverse drug
reaction (SADR)'' to mean any SADR that, in the view of the
investigator or sponsor, places the patient or subject at immediate
risk of death from the SADR as it occurred. It does not include an SADR
that, had it occurred in a more severe form, might have caused death.
(Comment 2) Several comments agreed with FDA's proposal to add the
term ``or sponsor'' to the definition of life-threatening SADR. SADRs
would be reported as life-threatening if either the investigator or
sponsor considered them to be life-threatening. However, several
comments expressed concern with FDA's proposal. The comments stated
that a trained investigator is most qualified to make the sometimes
subjective assessment of whether an event is life-threatening and that
this determination often is best made by the health-care professional
or the reporter who is in direct contact with the patient. These
comments also stated that sponsors may exercise medical and scientific
judgment in deciding whether expedited reporting is appropriate. One
comment stated that allowing a sponsor to determine severity would
change the nature of the assessment and result in increased reporting
of events assessed by those with often incomplete information. One
comment pointed out that FDA's rationale for expanding the role of the
sponsor is not supported by the quote from the ICH E2A guidance in the
preamble to the proposed rule (68 FR 12406 at 12419) because the ICH
E2A guidance quote refers to causality assessment, not assessment of
seriousness.
(Response) The agency agrees with the comments that support
expanding this definition to include reporting of an adverse event as
life-threatening if either the investigator or the sponsor considers it
to be life-threatening. The agency believes that, in some cases, the
sponsor may not agree with the investigator's assessment that an
adverse event does not qualify as life-threatening. In such cases,
because these events are critically important for the identification of
significant safety problems, the agency believes that broadening the
definition to allow sponsors to also make this assessment is prudent
and appropriate. While the agency agrees with the comment that pointed
out that the preamble to the proposed rule misinterpreted the quote
from the ICH E2A guidance, we nonetheless believe that the revision to
the definition is consistent with the overall intent of the ICH E2A
guidance.
(Comment 3) Several comments disagreed with the agency's position
articulated in the preamble to the proposed rule that reasons for any
differences of opinion between the investigator and sponsor regarding a
determination that an SADR is life-threatening would be included in the
IND safety report (68 FR 12406 at 12419). The comments argued that this
adds no value and is not appropriate or necessary in all cases. In
addition, comments stated that obtaining the investigator's view when
he or she deems the event non-life-threatening would be difficult.
(Response) The agency agrees that reasons for differences of
opinion between the sponsor and investigator are not always important
and, therefore, not necessary to include in the IND safety report in
all cases. Therefore, in this final rule, the agency does not require
including the reasons for differences of opinion in the IND safety
report. However, it is important that any adverse event or suspected
adverse reaction considered life-threatening by either the sponsor or
the investigator be reported as such.
(Comment 4) Some comments suggested that FDA clarify the definition
of life-threatening to take into account the role of other study staff
making safety observations. The comments suggested that the definition
be clarified to state that investigators or sponsors must evaluate
information communicated to them or recorded by their qualified staff
or agents and transmit reportable information to the sponsor or FDA.
One comment recommended that the definition be modified to include
contractors as well as sponsors.
(Response) The agency does not agree that the recommended revisions
to the definition are necessary because taking the observations of
staff into account is inherent in the obligations of the investigator.
Any qualified study staff could make pertinent safety observations, and
it is the investigator's responsibility in supervising the conduct of
the clinical investigation (see Sec. Sec. 312.53 and 312.60) to report
adverse experiences to the sponsor in accordance with Sec. 312.64.
Further information on the supervisory responsibilities of
investigators can be obtained in the agency's guidance for industry
entitled ``Investigator Responsibilities: Protecting the Rights,
Safety, and Welfare of Study Subjects'' (74 FR 55052, October 26,
2009).\2\ The agency does not believe that it is necessary to change
the definition to include contractors because, under Sec. 312.52, a
contract research organization that assumes any obligation of a sponsor
must comply with the applicable regulation.
---------------------------------------------------------------------------
\2\ Draft and final guidances for the Center for Drug Evaluation
and Research (CDER)-related information are posted on the Internet
at https://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm. The Center for Biologics Evaluation and
Research (CBER)-related information is posted at https://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htm (21 U.S.C. 371(h), 21 CFR 10.115).
---------------------------------------------------------------------------
4. Minimum Data Set
Under Sec. 312.32(a), FDA proposed the term ``minimum data set''
to mean that ``the report includes an identifiable patient, an
identifiable reporter, a suspect drug product, and an SADR.''
(Comment 5) Two comments requested further clarification regarding
the meaning of ``identifiable'' with respect to the kind and amount of
information needed to meet the criteria for an ``identifiable patient''
and ``identifiable reporter.'' One comment questioned whether patient
characteristics, such as age or gender, would be adequate, or if the
ability to contact the patient is necessary.
(Response) As discussed in comments 13 and 14 of this document,
because the four elements of the minimum data set are generally readily
available in the clinical trial setting, the agency has determined that
the definition and the requirement are unnecessary and has decided not
to require a minimum data set for IND safety reports as proposed in
Sec. 312.32(c). Because the agency is not adopting this definition in
the IND safety reporting requirements, the comments requesting
clarification about
[[Page 59942]]
the elements of the definition are no longer relevant.
5. Serious SADR
FDA proposed to define ``serious SADR'' in the same way as the
then-current definition of ``serious adverse drug experience'' under
Sec. 312.32(a) as follows: ``Serious SADR means any SADR that results
in any of the following outcomes: Death, a life-threatening SADR,
inpatient hospitalization or prolongation of existing hospitalization,
a persistent or significant disability/incapacity, or a congenital
anomaly/birth defect. Important medical events that may not result in
death, be life-threatening, or require hospitalization may be
considered a serious SADR when, based upon appropriate medical
judgment, they may jeopardize the patient or subject and may require
medical or surgical intervention to prevent one of the outcomes listed
in this definition. Examples of such medical events include allergic
bronchospasm requiring intensive treatment in an emergency room or at
home, blood dyscrasias or convulsions that do not result in
hospitalization, or the development of drug dependency or drug abuse.''
(Comment 6) One comment suggested that the definition of ``serious
SADR'' be revised to expressly allow the sponsor to determine if an
adverse event is serious, in the absence of a reporter's assessment of
seriousness.
(Response) For reasons similar to those stated in Comment 2 of this
document (definition of life-threatening), the agency agrees that the
definition of ``serious adverse event or serious suspected adverse
reaction'' should be revised to allow the determination that an adverse
event or suspected adverse reaction is ``serious'' if either the
investigator or sponsor considers it serious. Therefore, the agency has
revised this definition to add the phrase ``in the view of either the
investigator or sponsor.''
6. Unexpected SADR
FDA proposed that the definition of ``unexpected SADR'' be the same
as the then-current definition for ``unexpected adverse drug
experience'' under Sec. 312.32(a), except that the following sentence
was added to make clear which SADRs are considered unexpected: ``SADRs
that are mentioned in the investigator's brochure as occurring with a
class of drugs but not specifically mentioned as occurring with the
particular drug are considered unexpected.''
(Comment 7) One comment stated that in the proposed definition, the
``severity'' standard is vague, leaving the determination of
``expectedness'' to the investigator's judgment.
(Response) Unless a sponsor-investigator is responsible for the
clinical trial, the sponsor, rather than the investigator, generally
determines if a suspected adverse reaction is unexpected for reporting
purposes. However, the agency acknowledges that judgment is needed to
decide if the severity of a suspected adverse reaction is greater than
described in the investigator brochure. The definition of ``unexpected
adverse event or unexpected suspected adverse reaction'' in the final
rule includes an example of a suspected adverse reaction that would be
considered unexpected by virtue of its greater severity than other
suspected adverse reactions mentioned in the investigator brochure
(i.e., hepatic necrosis would be considered unexpected where the
investigator brochure includes elevated hepatic enzymes or hepatitis).
(Comment 8) Another comment recommended that FDA provide guidance
on what should be considered ``expected'' for regulatory reporting
purposes, in particular, what safety information to include in the
investigator brochure and what subset of such information would be
considered ``expected'' (i.e., only those for which a causal
relationship is suspected, reasonably established, or inferred based on
evidence). Some comments stated that if the basis for evaluating
expectedness is that an event is listed in the investigator's brochure,
sponsors may add long lists of adverse events, thereby delaying
important safety reports from being submitted to FDA. One comment
recommended that FDA require that, until the applicable reference
safety information document is officially updated (e.g., reprinted and
distributed) to include a new serious, suspected adverse reaction
(thereby making it expected), all subsequent reports of similar serious
adverse drug reactions be submitted expeditiously as an IND safety
report. Another comment suggested adopting use of the Developmental
Core Safety Information (DCSI) document, proposed by a CIOMS Working
Group, as the reference for ``expectedness'' instead of the
investigator brochure because the DCSI document contains only those
adverse events that, after careful analysis are believed by the company
to be likely related to the drug (Refs. 2 and 3).
(Response) The purpose of the investigator brochure is to provide
the investigator with information (clinical and nonclinical) about the
investigational drug that is relevant to study of the drug in human
subjects. The investigator brochure should include the information that
is important for the investigator, who is administering the drug to
human subjects, to know and understand. The investigator brochure is
required to include information about the drug substance and
formulation, pharmacological and toxicological effects of the drug in
animals (and in humans, if known), pharmacokinetics and biological
disposition of the drug in animals (and in humans, if known),
information relating to safety and effectiveness in humans obtained
from prior clinical studies, and information about possible risks and
side effects to be anticipated on the basis of prior experience with
the drug under investigation or with related drugs, and precautions or
special monitoring to be done as part of the investigational use of the
drug (see Sec. 312.23(a)(5)).
In general, the investigator brochure lists those adverse events
that have been observed with the investigational drug and for which a
causal relationship with the drug is suspected or confirmed. It is not
appropriate for sponsors to add long lists of adverse events that are
unlikely to have been caused by the drug to the investigator brochure
because such lists could dilute the importance of clinically meaningful
risk information and as a result, may put subjects at risk. The sponsor
needs to exercise judgment when deciding if the threshold has been
reached for adding a newly observed adverse event to the investigator
brochure. This decision usually depends on the strength of the evidence
from individual or multiple cases and previous knowledge about the drug
or drug class. In some cases, the threshold for including an adverse
event may be lower if it could result in a significant adverse outcome
for trial participants.
The investigator brochure describes adverse events that may be
predicted to occur based on the pharmacological properties of the drug.
For reporting purposes, if an adverse event occurs that has not
previously been observed with the drug under investigation, the event
is considered ``unexpected.'' To make clear that such predicted adverse
events are considered ``unexpected,'' the final rule revises the
proposed definition of ``unexpected'' to state explicitly that the term
also refers to adverse events or suspected adverse reactions that are
mentioned in the investigator brochure as occurring with a class of
drugs or as anticipated from the pharmacological properties of the
drug, but are not specifically mentioned as occurring with the
particular drug under investigation.
[[Page 59943]]
The agency expects the sponsor to update the investigator brochure
on an ongoing basis with new important safety information. However, the
agency agrees with the comment that, until the investigator brochure
and other applicable reference safety information are updated to
include a new serious, suspected adverse reaction, subsequent reports
of similar serious, suspected adverse reactions must be submitted
expeditiously in IND safety reports.
Finally, sponsors submit and the agency accepts a variety of
formats for the investigator brochure. For this reason, we are not
formally adopting use of the DCSI document in this final rule. However,
we agree that a sponsor could incorporate a document such as the DCSI
into the investigator brochure for use as the reference for
``expectedness'' for reporting purposes if the DCSI contains the
required safety information about the investigational drug.
B. Review of Safety Information--Proposed Sec. 312.32(b)
IND safety reporting regulations in former Sec. 312.32(b) required
that sponsors promptly review all information relevant to the safety of
the drug obtained or otherwise received by the sponsor from any source,
foreign or domestic. Examples of potential sources of information in
the former regulation included information derived from any clinical or
epidemiological investigations, animal investigations, commercial
marketing experience, reports in the scientific literature, as well as
unpublished scientific papers, and reports from foreign regulatory
authorities that had not been previously reported to FDA by the
sponsor. Proposed Sec. 312.32(b) would have amended this requirement
to include in vitro studies as another example of a potential source of
information and to clarify that ``reports from commercial marketing
experience'' is intended to apply only to reports from foreign
commercial marketing experience for drugs that are not marketed in the
United States. As proposed, reports from IND studies of drugs that are
marketed in the United States would be required to be reported as
described under Sec. 312.32(c)(4), if applicable.
(Comment 9) One comment stated that reportable information can come
from a wider variety of media or sources than those listed in the
proposed rule. The comment maintained that investigators or sponsors
participating in public or private meetings or conferences can learn of
reportable events from colleagues or other professionals. The comment
recommended that the list of potential sources of reportable
information include such alternative sources.
(Response) The sponsor is required to ``promptly review all
information relevant to the safety of the drug obtained or otherwise
received by the sponsor from foreign or domestic sources, including
information derived from any clinical or epidemiological
investigations, animal or in vitro studies * * *'' (emphasis added).
The sources listed in the requirement are not all inclusive, but
represent examples of the variety of sources that may yield safety
information. Therefore, the agency agrees that reportable information
can come from sources other than those listed in Sec. 312.32(b) and
that one such source could be from public or private meetings. However,
the agency does not believe that it is necessary to amend the
requirement to provide additional examples.
(Comment 10) One comment agreed with the clarification that
reporting from commercial marketing experience applies only to foreign
commercial marketing experience for drugs that are not marketed in the
United States. The comment requested that FDA further make it clear
that expedited reporting under Sec. 312.32 is not required for reports
from foreign commercial marketing experience for a different
formulation of the same active moiety as a drug product that is
lawfully marketed in the United States and that those reports should be
submitted to the most appropriate new drug application (NDA) for the
active moiety.
(Response) As described further in Comment 31 of this document, IND
safety reports are required under Sec. 312.32(c)(4) for suspected
adverse reactions observed in clinical studies that are being conducted
under an IND for a drug marketed or approved in the United States. In
general, an expedited report from domestic or foreign commercial
marketing experience for a drug lawfully marketed in the United States
would not be submitted to the IND, but instead, must be submitted in
accordance with the relevant postmarketing reporting requirements
(e.g., Sec. Sec. 310.305, 314.80, and 600.80). Similarly, a report of
a suspected adverse reaction from foreign marketing experience for a
different formulation of the drug product (same active moiety) that is
lawfully marketed in the United States must be submitted in accordance
with the relevant postmarketing reporting requirements.
(Comment 11) One comment agreed with the proposal to add in vitro
studies to the list of information that should be reviewed by the
sponsor in its ongoing assessment of the safety of an investigational
drug. Some comments stated that it would be helpful if FDA could
provide examples, in addition to carcinogenicity, mutagenicity and
teratogenicity, of when safety data from in vitro studies would yield
relevant, important information that should be reviewed for IND
reporting purposes.
(Response) Data from in vitro microsusceptibility, drug
interaction, or genotoxicity studies are examples of other data from in
vitro studies that may yield important safety information.
(Comment 12) One comment expressed concern that once a sponsor
provides FDA with the animal and in vitro studies, emails, and reports
from foreign regulatory authorities and any other information it
reviewed in determining whether to report safety information, FDA may
have to make the information publicly available under the Freedom of
Information Act (FOIA). The comment stated that, before implementing
the requirement, FDA should explain why these additional data are
needed and how they will be handled for FOIA purposes. The comment
requested t