Development of Comprehensive Monitoring Plan

Guidance for Industry on Oversight of Clinical Investigations: A Risk-Based Approach to Monitoring

FINAL GUIDANCE 0733 1-4-2016

Development of Comprehensive Monitoring Plan

OMB: 0910-0733

Document [pdf]
Download: pdf | pdf
Guidance for Industry
Oversight of Clinical
Investigations —
A Risk-Based Approach to
Monitoring

U.S. Department of Health and Human Services
Food and Drug Administration
Center for Drug Evaluation and Research (CDER)
Center for Biologics Evaluation and Research (CBER)
Center for Devices and Radiological Health (CDRH)
Office of Good Clinical Practice (OGCP)
Office of Regulatory Affairs (ORA)
August 2013
Procedural

OMB Control No. 0910-0733
Expiration Date: 03/31/2016
See additional PRA statement in section VII of this guidance.

Guidance for Industry
Oversight of Clinical Investigations — A
Risk-Based Approach to Monitoring
Additional copies are available from:
Office of Communications
Division of Drug Information, WO51, Room 2201
Center for Drug Evaluation and Research
Food and Drug Administration
10903 New Hampshire Ave.
Silver Spring, MD 20993-0002
Phone: 301-796-3400; Fax: 301-847-8714
[email protected]
http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm
or
Office of Communication, Outreach and Development, HFM-40
Center for Biologics Evaluation and Research
Food and Drug Administration
1401 Rockville Pike, Suite 200N, Rockville, MD 20852-1448
[email protected] Phone: 800-835-4709 or 301-827-1800
http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/default.htm
or
Office of Communication and Education
Division of Small Manufacturers, International and Consumer Assistance
Center for Devices and Radiological Health
Food and Drug Administration
10903 New Hampshire Ave., Bldg. 66, rm. 4613
Silver Spring, MD 20993-0002
(Tel) 800-638-2041 or 301-796-7100; (Fax) 301-847-8149; (E-mail) [email protected]
http://www.fda.gov/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/default.htm

U.S. Department of Health and Human Services
Food and Drug Administration
August 2013
Procedural

Contains Nonbinding Recommendations
TABLE OF CONTENTS
I.

INTRODUCTION............................................................................................................. 1

II.

BACKGROUND ............................................................................................................... 2
A.

Current Monitoring Practices and FDA Guidance .................................................................... 3

B.

FDA’s Rationale for Risk-Based Monitoring .............................................................................. 5

III.

OVERVIEW OF MONITORING METHODS ............................................................. 6

A. On-Site and Centralized Monitoring ........................................................................................... 6
1. On-Site Monitoring .......................................................................................................................... 6
2. Centralized Monitoring .................................................................................................................... 7
B.
1.
2.
3.

IV.

Examples of Alternative Monitoring Techniques ....................................................................... 7
Communication with Study Site Staff ............................................................................................. 9
Review of Site’s Processes, Procedures, and Records ..................................................................... 9
Source Data Verification and Corroboration ................................................................................. 10

RISK-BASED MONITORING...................................................................................... 10

A.

Identify Critical Data and Processes to be Monitored.............................................................. 11

B.

Risk Assessment ........................................................................................................................... 12

C.

Factors to Consider when Developing a Monitoring Plan ....................................................... 13

D.
1.
2.
3.
4.
5.

Monitoring Plan ........................................................................................................................... 14
Description of Monitoring Approaches.......................................................................................... 15
Communication of Monitoring Results ......................................................................................... 15
Management of Noncompliance .................................................................................................... 16
Ensuring Quality Monitoring ......................................................................................................... 16
Monitoring Plan Amendments ....................................................................................................... 17

V.

DOCUMENTING MONITORING ACTIVITIES ...................................................... 17

VI.

ADDITIONAL STRATEGIES TO ENSURE STUDY QUALITY ............................ 17

A.

Protocol and Case Report Form Design .................................................................................... 17

B.

Clinical Investigator Training and Communication ................................................................. 18

C.

Delegation of Monitoring Responsibilities to a CRO ................................................................ 18

D.

Clinical Investigator and Site Selection and Initiation ............................................................. 19

VII.

PAPERWORK REDUCTION ACT OF 1995 .............................................................. 19

Contains Nonbinding Recommendations

Guidance for Industry1
Oversight of Clinical Investigations — A Risk-Based
Approach to Monitoring

This guidance represents the Food and Drug Administration’s (FDA’s) current thinking on this topic. It
does not create or confer any rights for or on any person and does not operate to bind FDA or the public.
You can use an alternative approach if the approach satisfies the requirements of the applicable statutes
and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for
implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate
number listed on the title page of this guidance.

I.

INTRODUCTION

This guidance assists sponsors of clinical investigations in developing risk-based monitoring
strategies and plans for investigational studies of medical products, including human drug and
biological products, medical devices, and combinations thereof. The overarching goal of this
guidance is to enhance human subject protection and the quality of clinical trial data by focusing
sponsor oversight on the most important aspects of study conduct and reporting.
This guidance makes clear that sponsors can use a variety of approaches to fulfill their
responsibilities for monitoring clinical investigator (CI) conduct and performance in
investigational new drug (IND) studies conducted under 21 CFR part 312 or investigational
device exemption (IDE) studies conducted under 21 CFR part 812. The guidance describes
strategies for monitoring activities that reflect a modern, risk-based approach that focuses on
critical study parameters and relies on a combination of monitoring activities to oversee a study
effectively. For example, the guidance specifically encourages greater use of centralized
monitoring methods where appropriate.
FDA’s guidance documents, including this guidance, do not establish legally enforceable
responsibilities. Rather, guidances describe the Agency’s current thinking on a topic and should
be viewed only as recommendations, unless specific regulatory or statutory requirements are
cited. The use of the word should in Agency guidances means that something is suggested or
recommended, but not required.

1

This guidance has been prepared by the Office of Medical Policy in the Center for Drug Evaluation and Research
(CDER) in cooperation with CDER’s Office of Scientific Investigations in the Office of Compliance, CBER’s
Office of Compliance and Biologics Quality, CDRH’s Office of Compliance, Office of the Commissioner’s Office
of Good Clinical Practice, and the Office of Regulatory Affairs (ORA).

1

Contains Nonbinding Recommendations
II.

BACKGROUND

Effective monitoring of clinical investigations by sponsors is critical to the protection of human
subjects and the conduct of high-quality studies. Sponsors of clinical investigations involving
human drugs, biological products, medical devices, and combinations thereof are required to
provide oversight to ensure adequate protection of the rights, welfare, and safety of human
subjects and the quality of the clinical trial data submitted to FDA.2 FDA’s regulations require
sponsors to monitor the conduct and progress of their clinical investigations.3,4 The regulations
are not specific about how sponsors are to conduct such monitoring and are therefore compatible
with a range of approaches to monitoring (see section III) that will vary depending on multiple
factors (see section IV.C).
During the past two decades, the number and complexity of clinical trials have grown
dramatically. These changes create new challenges to clinical trial oversight, particularly
increased variability in clinical investigator experience, site infrastructure, treatment choices, and
standards of health care,5 as well as challenges related to geographic dispersion. At the same
time, increasing use of electronic systems and records and improvements in statistical
assessments, present opportunities for alternative monitoring approaches (e.g., centralized
monitoring) that can improve the quality and efficiency of sponsor oversight of clinical
investigations. FDA encourages sponsors to develop monitoring plans that manage important
risks to human subjects and data quality and address the challenges of oversight in part by taking
advantage of the innovations in modern clinical trials. A risk-based approach to monitoring does
not suggest any less vigilance in oversight of clinical investigations. Rather, it focuses sponsor
oversight activities on preventing or mitigating important and likely risks to data quality and to
processes critical to human subject protection and trial integrity. Moreover, a risk-based
approach is dynamic, more readily facilitating continual improvement in trial conduct and
oversight. For example, monitoring findings should be evaluated to determine whether
additional actions (e.g., training of clinical investigator and site staff, clarification of protocol
requirements) are necessary to ensure human subject protection and data quality across sites.
This guidance focuses principally on monitoring, which is one aspect of the processes and
procedures needed to ensure clinical trial quality and subject safety. Monitoring is a quality
control tool for determining whether study activities are being carried out as planned, so that
deficiencies can be identified and corrected. Monitoring, or oversight, alone cannot ensure
quality. Rather, quality is an overarching objective that must be built into the clinical trial
enterprise. FDA recommends a quality risk management approach to clinical trials and is
considering the need for additional guidance describing this approach.
2

21 CFR part 312, subpart D generally (Responsibilities of Sponsors and Investigators) and 21 CFR part 812,
subpart C generally (Responsibilities of Sponsors).
3
21 CFR 312.50 requires a sponsor to, among other things, ensure “proper monitoring of the investigation(s)” and
“that the investigation(s) is conducted in accordance with the general investigational plan and protocols contained in
the IND.” 21 CFR 812.40 states that sponsors are responsible for, among other things, “ensuring proper monitoring
of the investigation, …”
4
See also 21 CFR 312.53(d), 312.56(a), 812.43(d), and 812.46.
5
Glickman et al. Ethical and Scientific Implications of the Globalization of Clinical Research. NEJM. 360: 816-823
(2009).

2

Contains Nonbinding Recommendations
We are aware that the term monitoring is used in different ways in the clinical trial context. It
can refer to the assessment of CI conduct, oversight, and reporting of findings of a clinical trial;
to the ongoing evaluation of safety data and the emerging benefit-risk profile of an
investigational product; and to the monitoring of internal sponsor and contract research
organization (CRO) processes and systems integral to proposing, designing, performing,
recording, supervising, reviewing, or reporting clinical investigations.
For purposes of this guidance, monitoring refers to the methods used by sponsors of
investigational studies, or CROs delegated responsibilities for the conduct of IND studies, to
oversee the conduct of, and reporting of data from, clinical investigations, including appropriate
CI supervision of study site staff and third party contractors. Monitoring activities include
communication with the CI and study site staff; review of the study site’s processes, procedures,
and records; and verification of the accuracy of data submitted to the sponsor.
A.

Current Monitoring Practices and FDA Guidance

A survey conducted through the Clinical Trials Transformation Initiative (CTTI)6 indicated that a
range of practices has been used to monitor the conduct of clinical trials. These practices vary in
intensity, focus, and methodology and include centralized monitoring of clinical data by
statistical and data management personnel; targeted on-site visits to higher risk CIs (e.g., where
centralized monitoring suggests problems at a site); and frequent, comprehensive on-site visits to
all CI sites by sponsor personnel or representatives (e.g., clinical monitors or clinical research
associates).7 See definitions of on-site and centralized monitoring in section III.A.
Although survey participants reported a range of monitoring methods, periodic, frequent visits to
each CI site to evaluate study conduct and review data for each enrolled subject remain the
predominant mechanism by which pharmaceutical, biotechnology, and medical device
companies monitor the progress of clinical investigations. For major efficacy trials, companies
typically conduct on-site monitoring visits at approximately 4- to 8-week intervals,8 at least
partly because of the perception that the frequent on-site monitoring visit model, with 100%
verification of all data, historically has been FDA’s preferred way for sponsors to meet their
monitoring obligations. In contrast, academic coordinating centers, cooperative groups, and
government organizations use on-site monitoring less extensively. For example, some
government agencies and oncology cooperative groups typically visit sites only once every 2 or 3
years to qualify or certify clinical study sites9 to ensure they have the resources, training, and
safeguards to conduct clinical trials. FDA also recognizes that regulators and practitioners have
relied on data from critical outcome studies (e.g., many National Institutes of Health-sponsored
trials, Medical Research Council-sponsored trials in the United Kingdom, ISIS (International
6

CTTI is a public-private partnership involving FDA, academia, industry representatives, patient and consumer
representatives, professional societies, investigator groups, and other government agencies, initiated in 2008. CTTI’s
mission is to identify practices that will increase the quality and efficiency of clinical trials.
7
Morrison et al. Monitoring the Quality of Conduct of Clinical Trials: A Survey of Current Practices. Clin Trials. 8:
342-349 (2011).
8
Usher, R. PhRMA BioResearch Monitoring Committee Perspective on Acceptable Approaches for Clinical Trial
Monitoring. Drug Inf J. 44: 477-483 (2010).
9
Id.

3

Contains Nonbinding Recommendations
Study of Infarct Survival) trials,10 and GISSI11), which had no regular on-site monitoring and
used primarily centralized and other alternative monitoring methods.12 These examples suggest
that use of alternative monitoring approaches should be considered by all sponsors, including
commercial sponsors, when developing risk-based monitoring strategies and plans.
The 1996 International Conference on Harmonisation of Technical Requirements for
Registration of Pharmaceuticals for Human Use (ICH) guidance on good clinical practice (ICH
E6) and the 2011 International Standards Organization (ISO) Clinical investigation of medical
devices for human subjects – good clinical practice (ISO 14155:2011) address monitoring. Both
ICH E6 and ISO 14155:2011 specifically provide for flexibility in how trials are monitored. ICH
E6 and ISO 14155:2011 advise sponsors to consider the objective, design, complexity, size, and
endpoints of a trial in determining the extent and nature of monitoring for a given trial.13,14 The
ISO standard further states that a sponsor’s assessment of these factors should be used to develop
a monitoring plan, a recommendation consistent with FDA’s recommendation for monitoring
plan development in this guidance. Although the ICH guidance and ISO standard specifically
provide for the possibility of reduced, or even no, on-site monitoring, they also make clear that it
would be appropriate to rely entirely on centralized monitoring only in exceptional
circumstances.
FDA has communicated the goals of, and recommendations for, risk-based monitoring to FDA
staff in review, inspection, and compliance functions. FDA’s bioresearch monitoring
compliance program guidance manuals (CPGMs) for sponsors, CROs, and monitors (CPGM
7348.810)15 and for CIs and sponsor-investigators (CPGM 7348.811)16 are compatible with the
approaches described in this guidance. For example, CPGM 7348.810 informs FDA field staff
that the regulations do not prescribe a specific monitoring technique. While CPGM 7348.810
refers to site visits and does not discuss centralized monitoring, the focus is on the review of
monitoring activities through documentation and whether these activities were carried out in
accordance with the sponsor’s (or CRO’s) monitoring procedures.
10

Califf et al. Developing Systems for Cost-Effective Auditing of Clinical Trials. Controlled Clinical Trials. 18:
651-660 (1997).
11
Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico-Italian group for the study of the
survival of myocardial infarction.
12
Temple, R. Policy Developments in Regulatory Approval. Statistics in Medicine. 21: 2939-2948 (2002).
13
Guidance for industry, E6 Good Clinical Practice: Consolidated Guidance, 1996, section 5.18.3. We update
guidances periodically. To make sure you have the most recent version of a guidance, check the guidance Web site.
CDER guidance documents can be found at
http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm.
CBER guidance documents can be found at
http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htm.
CDRH guidance documents can be found at
http://www.fda.gov/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/default.htm.
14
ISO 14155:2011, Clinical investigation of medical devices for human subjects – Good clinical practice, sections
5.7 and 6.3.
15
CPGM 7348.810: Sponsors, Contract Research Organizations and Monitors (March 11, 2011), available at:
http://www.fda.gov/ICECI/EnforcementActions/BioresearchMonitoring/ucm133777.htm.
16
CPGM 7348.811: Clinical Investigators and Sponsor-Investigators (December 8, 2008), available at:
http://www.fda.gov/ICECI/EnforcementActions/BioresearchMonitoring/ucm133562.htm.

4

Contains Nonbinding Recommendations
B.

FDA’s Rationale for Risk-Based Monitoring

FDA is issuing this guidance to provide FDA’s current recommendations regarding monitoring
practices and to encourage sponsors to consider a change in approach to monitoring. FDA
believes that risk-based monitoring could improve sponsor oversight of clinical investigations.
This guidance is therefore intended to make it clear that risk-based monitoring, including the
appropriate use of centralized monitoring (see section III.A.2 for discussion of centralized
monitoring) and reliance on technological advances (e.g., e-mail, webcasts, online training
modules), can meet statutory and regulatory requirements under appropriate circumstances.
There is a growing consensus that risk-based approaches to monitoring, focused on risks to the
most critical data elements and processes necessary to achieve study objectives, are more likely
than routine visits to all clinical sites and 100% data verification to ensure subject protection and
overall study quality.17,18,19,20 For example, incorporation of centralized monitoring practices,
where appropriate, should improve a sponsor’s ability to ensure the quality of clinical trial data.
Several publications suggest that certain data anomalies (e.g., fraud, including fabrication of
data, and other non-random data distributions) may be more readily detected by centralized
monitoring techniques than by on-site monitoring.21, 22, 23 It has been suggested that a statistical
approach to central monitoring can “help improve the effectiveness of on-site monitoring by
prioritizing site visits and by guiding site visits with central statistical data checks,” an approach
that is supported by illustrative examples using actual trial datasets.24 A recent review of on-site
monitoring findings collected during a multi-center international trial also suggests that
centralized monitoring can identify the great majority of on-site monitoring findings. The review
determined that centralized monitoring activities could have identified more than 90% of the
findings identified during on-site monitoring visits.25
FDA encourages sponsors to tailor monitoring plans to the needs of the trial (see section IV).
FDA recognizes that this guidance places greater emphasis on centralized monitoring than
appeared feasible at the time ICH E6 was finalized. However, FDA considers the approach to
monitoring described in this guidance to be consistent with ICH E6 and ISO 14155:2011. FDA
17

Usher, R. PhRMA BioResearch Monitoring Committee Perspective on Acceptable Approaches for Clinical Trial
Monitoring. Drug Inf J. 44: 477-483 (2010).
18
FDA, Concept Paper: Quality in FDA-Regulated Clinical Research; Background to HSP/BIMO Workshop 5/105/11/07, (4/26/07).
19
Brosteanu et al. Risk Analysis and Risk Adapted On-Site Monitoring in Noncommercial Clinical Trials. Clin
Trials. 6: 585-595 (2009).
20
Tantsyura et al. Risk-Based Source Data Verification Approaches: Pros and Cons. Drug Inf J. 44: 745-756 (2010).
21
Usher, R. PhRMA BioResearch Monitoring Committee Perspective on Acceptable Approaches for Clinical Trial
Monitoring. Drug Inf J. 44: 477-483 (2010).
22
Baigent et al. Ensuring Trial Validity by Data Quality Assurance and Diversification of Monitoring Methods. Clin
Trials. 5: 49-55 (2008).
23
Buyse et al. The Role of Biostatistics in the Prevention, Detection and Treatment of Fraud in Clinical Trials.
Statistics in Medicine. 18: 3435-51 (1999).
24
Venet et al. A Statistical Approach to Central Monitoring of Data Quality in Clinical Trials. Clin Trials. 0: 1-9
(2012).
25
Bakobaki et al. The Potential for Central Monitoring Techniques to Replace On-Site Monitoring: Findings from
an International Multi-Centre Clinical Trial. Clin Trials. 9: 257-264 (2012).

5

Contains Nonbinding Recommendations
believes it is reasonable to conclude that the flexibility described in ICH E6 and ISO 14155:2011
was intended to permit innovative approaches to improve the effectiveness of monitoring.
Notably, the advancement in electronic systems and increasing use of electronic records (i.e.,
electronic data capture (EDC) systems) facilitate remote access to electronic data and,
increasingly, to some source data (see section III.B.2.b for further discussion of access to
electronic source data). Additionally, statistical assessments using data submitted on paper CRFs
or via EDC may permit timely identification of clinical sites that require additional training,
monitoring, or both. We expect that the pharmaceutical and device industries will, for the
foreseeable future, continue to use some amount of on-site monitoring, but we anticipate
decreased use of on-site monitoring with evolving monitoring methods and technological
capabilities.
The following sections reflect FDA’s current thinking on monitoring and include
recommendations on how to develop and implement a study-specific monitoring plan as well as
how to document monitoring activities. FDA acknowledges that there are limited empirical data
to support the utility of the various methods employed to monitor clinical investigations (e.g.,
superiority of one method versus another), including data to support on-site monitoring.26 As a
result, the recommendations are based, in part, on FDA’s experience from the review of
protocols during the IND or IDE phase, data submitted in pre-approval applications, results of
inspections conducted to ensure human subject protection and data integrity, and information
obtained from public outreach efforts conducted under the auspices of the CTTI.
III.

OVERVIEW OF MONITORING METHODS

A.

On-Site and Centralized Monitoring

This section is intended to assist sponsors in identifying and designing monitoring practices
appropriate to a given clinical trial. It describes some of the capabilities of on-site and
centralized monitoring processes and factors to consider in determining which monitoring
practices may be appropriate for a given clinical trial. See section IV.C for a discussion of
factors to consider when determining the types, frequency, and extent of monitoring activities
and section IV.D.1 for examples of events or results that would trigger a change in planned
monitoring activities.
1.

On-Site Monitoring

On-site monitoring is an in-person evaluation carried out by sponsor personnel or representatives
at the sites at which the clinical investigation is being conducted. On-site monitoring can
identify data entry errors (e.g., discrepancies between source records and case report forms
(CRFs)) and missing data in source records or CRFs; provide assurance that study
documentation exists; assess the familiarity of the site’s study staff with the protocol and
required procedures; and assess compliance with the protocol and investigational product
26

Two studies are on-going as of June 2013 that compare the effectiveness of on-site to alternative (e.g., centralized)
monitoring methods (OPTIMON study (https://ssl2.isped.u-bordeaux2.fr/optimon/Default.aspx) and ADAMON
study (http://ctj.sagepub.com/content/6/6/585.full.pdf+html)).

6

Contains Nonbinding Recommendations
accountability. On-site monitoring can also provide a sense of the quality of the overall conduct
of the trial at a site (e.g., attention to detail, thoroughness of study documentation, appropriate
delegation of study tasks, appropriate CI supervision of site staff performing critical study
functions). On-site monitoring can therefore be particularly helpful early in a study, especially if
the protocol is complex and includes novel procedures with which CIs may be unfamiliar.
Findings at the site may lead to training efforts at both the site visited and elsewhere (see section
VI.B).
2.

Centralized Monitoring

Centralized monitoring is a remote evaluation carried out by sponsor personnel or
representatives (e.g., clinical monitors, data management personnel, or statisticians) at a location
other than the sites at which the clinical investigation is being conducted. Centralized
monitoring processes can provide many of the capabilities of on-site monitoring as well as
additional capabilities.
FDA encourages greater use of centralized monitoring practices, where appropriate, than has
been the case historically, with correspondingly less emphasis on on-site monitoring. The types
of monitoring activities and the extent to which centralized monitoring practices can be
employed depend on various factors, including the sponsor’s use of electronic systems; the
sponsor’s access to subjects’ electronic records, if applicable; the timeliness of data entry from
paper CRF, if applicable; and communication tools available to the sponsor and study site.
These may vary by study and by site. Sponsors who plan to use centralized monitoring
processes should ensure that the processes and expectations for site record keeping, data entry,
and reporting are well-defined and ensure timely access to clinical trial data and supporting
documentation.27 If sponsors intend to rely heavily on centralized monitoring practices, they
should identify, in the monitoring plan, when one or more on-site monitoring visits would be
indicated.
B.

Examples of Alternative Monitoring Techniques

As discussed in section II, monitoring activities broadly include communication with the CI and
study site staff; review of the study site’s processes, procedures, and records; and verification of
the accuracy of data submitted to the sponsor. This section highlights areas for which centralized
monitoring techniques could be considered. For certain monitoring activities, centralized
monitoring techniques can be considered in place of, or to complement, traditional monitoring
techniques. Specific techniques used should be prospectively included in the monitoring plan
and should be informed by the risk assessment (see section IV.B for discussion of risk
assessment).
Centralized monitoring techniques should be used to the extent appropriate and feasible to:

27

See guidances for industry: Part 11, Electronic Records; Electronic Signatures – Scope and Application and
Computerized Systems Used in Clinical Investigations.

7

Contains Nonbinding Recommendations


Supplement or reduce the frequency and extent of on-site monitoring with monitoring
activities that can be done as well or better remotely or with monitoring activities that can be
accomplished using centralized processes only. Examples include:
o Monitor data quality through routine review of submitted data to identify and
follow-up on missing data, inconsistent data, data outliers, and potential
protocol deviations that may be indicative of systemic or significant errors in
data collection and reporting at a site
o Conduct statistical analyses to identify data trends not easily detected by onsite monitoring, such as


Standard checks of range, consistency, and completeness of data



Checks for unusual distribution of data within and between study sites,
such as too little variance28

o Analyze site characteristics, performance metrics (e.g., high screen failure or
withdrawal rates, high frequency of eligibility violations, delays in reporting
data), and clinical data to identify trial sites with characteristics correlated
with poor performance or noncompliance
o Verify critical source data remotely as described in the monitoring plan, in
cases where such source data are accessible, or where CRF data are, according
to the protocol, source data
o Complete administrative and regulatory tasks. Such tasks include, for
example, verifying continuous institutional review board (IRB) approval by
reviewing electronic IRB correspondence, if available; performing portions of
investigational product accountability, such as comparison of randomization
and CRF data, to preliminarily assess whether the subject was administered or
dispensed the assigned product and to evaluate consistency between
investigational product receipt, use, and disposition records; and verifying
whether previously requested CRF corrections were made.
Centralized techniques, including routine review of submitted data and statistical and other
analyses, may also be used to identify significant concerns (e.g., need for clarification of a
protocol procedure, indications of data fabrication) with non-critical data that may not have
otherwise been a focus of monitoring (e.g., source document verification).


Target on-site monitoring by identifying higher risk clinical sites (e.g., sites with data
anomalies or a higher frequency of errors, protocol violations, or dropouts relative to other
sites), through the activities described above. Such findings, whether related to critical or
non-critical data, may warrant more intensive and consideration of on-site monitoring.

The following sections provide additional descriptions of alternative monitoring techniques.

28

Collins, Rory. (2010, October) Quality Design of Clinical Trials. Presentation at CTTI work stream 3 expert
meeting. Available at: https://www.ctti-clinicaltrials.org/websiteadministration/documents/COLLINS%20FDA%20trial%20quality%200811%20FINAL_no%20animation.pdf/view

8

Contains Nonbinding Recommendations
1.

Communication with Study Site Staff

Communication between the monitor and the study site staff is an essential component of
monitoring. Various modes of communication (e.g., teleconferences, videoconferencing, email)
could be considered for specific study time points (e.g., study initiation) and activities (e.g., to
discuss findings of a monitor’s eCRF review, training of new site staff).
2.

Review of Site’s Processes, Procedures, and Records

Techniques for monitoring informed consent and site records are included here as examples of
approaches to monitoring site’s processes, procedures, and records.
a. Informed Consent
Verification of subjects’ informed consent is a critical activity that should be monitored (see
section IV.A). Alternatives to the traditional approach (monitors verifying the original signature
on the consent form for each subject at the site) may be more effective in identifying
inadequacies in the consent process and may be more efficient. For example, the study site
electronically sends (e.g., fax, e-mail) the signed page(s) of consent forms to the monitor, or the
monitor performs remote comparison of dates of study procedures and documentation of
informed consent on CRFs. An internet portal that enables the site staff to upload signed consent
forms and enables access by designated monitors is a tool that can be considered. Use of
electronic informed consent may also facilitate sponsor oversight of human subject protection.
We recognize that sponsors must attend to privacy and confidentiality concerns when
considering techniques for monitoring informed consent remotely.
b. Site’s Records
A growing portion of source documents (e.g., laboratory and radiology reports, source
documents submitted by the CI for other purposes such as health records documenting serious
adverse events or adjudicated events) are electronic and may be available to the sponsor
remotely. Furthermore, consistent with ICH E6 and ISO 14155:2011, original observations can
be entered directly into the eCRF or transmitted to the eCRF from various locations, devices, or
instruments.29 We recognize that sponsors may not have remote access to electronic health
records maintained by hospitals, universities, and other institutions because of data privacy and
security concerns as well as technological challenges. Sponsors should consider risk-based
approaches to monitoring using the format of study information (i.e., electronic, paper, or
combination of electronic and paper), tools, and other resources available to them.
As discussed in this guidance, a variety of centralized monitoring techniques can be used to
replace, supplement, and target on-site monitoring activities. The majority of these techniques
29

Section 6.4.9 of ICH E6 provides that the trial design description should include “The identification of any data to
be recorded directly on the CRFs (i.e., no prior written or electronic record of data), and to be considered to be
source data.” ISO 14155:2011, section 6.8.2, provides that the clinical investigation plan “shall specify which data
can be recorded directly in the CRFs.”

9

Contains Nonbinding Recommendations
(e.g., checks for completeness of data, sites with a higher frequency of protocol violations
relative to other sites, sites with high screen failure rates) can be performed regardless of the
extent of use of electronic records in the study. For example, the majority of these techniques
can be performed using CRF data collected either using electronic data capture systems or
entered into a database from a paper CRF collected by the sponsor. A recent publication
discusses statistical techniques for identifying various types of data errors.30 We recognize that
the statistical techniques described in this guidance may not be routinely used by all sponsors
and may not be appropriate for every trial, but they are included in this guidance as examples of
monitoring techniques that may be considered by sponsors.
Additional monitoring techniques, such as routine review of data as they are submitted, are
possible for studies that use electronic CRFs. Although not a monitoring technique, another
method of ensuring data quality routinely implemented in eCRFs is the use of electronic prompts
in the eCRF to minimize errors and omissions at the time of data entry, particularly if data are
entered directly into the eCRF.
3.

Source Data Verification and Corroboration

The sponsor should consider the quantity and types of source data that need to be verified against
CRFs or corroborated against other records (e.g., review of medical record to corroborate a
subject’s response of “no hospitalizations” since the previous visit on a CRF) during the
sponsor’s identification of critical data and processes or in the risk assessment, or both. The
sponsor should include a description of the quantity and types of source records to verify or
corroborate in the monitoring plan. The sponsor should consider which source records are likely
to provide the most meaningful information about a subject’s participation and the CI’s conduct
and oversight. For example, for a particular study, there may be minimal benefit in comparing
100% of the source data for each subject to the CRFs for each study visit. Rather, it may be
sufficient to compare the most critical data points for a sample of subjects and study visits as an
indicator of data accuracy. Similarly, for a particular study, although collection of all
concomitant medications, body temperature, and body weight are required by the protocol and
are documented in the medical record and transcribed to a CRF, they may not be identified by
the sponsor as critical data, because a small error rate in those variables would not affect the
outcome of the trial. In the absence of information indicating potential concerns with the data
(e.g., sites with data anomalies, inconsistent data), source document verification or corroboration
of these non-critical data may not provide significantly useful information to the sponsor.

IV.

RISK-BASED MONITORING

No single approach to monitoring is appropriate or necessary for every clinical trial. FDA
recommends that each sponsor design a monitoring plan that is tailored to the specific human
subject protection and data integrity risks of the trial. Ordinarily, such a risk-based plan would
30

Venet et al. A Statistical Approach to Central Monitoring of Data Quality in Clinical Trials. Clin Trials. 0: 1-9
(2012).

10

Contains Nonbinding Recommendations
include a mix of centralized and on-site monitoring practices. The monitoring plan should
identify the various methods intended to be used and the rationale for their use (see section IV.D
for recommendations on the components of a monitoring plan).
Monitoring activities should focus on preventing or mitigating important and likely sources of
error in the conduct, collection, and reporting of critical data and processes necessary for human
subject protection and trial integrity. Sponsors should prospectively identify critical data and
processes, then perform a risk assessment to identify and understand the risks that could affect
the collection of critical data or the performance of critical processes, and then develop a
monitoring plan that focuses on the important and likely risks to critical data and processes.
A.

Identify Critical Data and Processes to be Monitored

Sponsors should prospectively identify critical data and processes that if inaccurate, not
performed, or performed incorrectly, would threaten the protection of human subjects or the
integrity of the study results. As examples, the following types of data and processes should
ordinarily be identified as critical:


Verification that informed consent was obtained appropriately



Adherence to protocol eligibility criteria designed to exclude individuals for whom the
investigational product may be less safe than the protocol intended and to include only
subjects from the targeted study population for whom the test article is most appropriate



Procedures for documenting appropriate accountability and administration of the
investigational product (e.g., ensuring the integrity of randomization at the site level, where
appropriate)



Conduct and documentation of procedures and assessments related to



–

study endpoints

–

protocol-required safety assessments

–

evaluating, documenting, and reporting serious adverse events and unanticipated adverse
device effects, subject deaths, and withdrawals, especially when a withdrawal may be
related to an adverse event

Conduct and documentation of procedures essential to trial integrity, such as ensuring the
study blind is maintained, both at the site level and at the sponsor level, as appropriate,
referring specified events for adjudication, and allocation concealment

11

Contains Nonbinding Recommendations
Other types of data (e.g., covariates such as concomitant treatments or demographic
characteristics, routine laboratory tests performed as part of subject monitoring that do not
address protocol specified safety or efficacy endpoints) and processes (e.g., a hospital
pharmacy’s storage of an investigational product with no specific critical handling instructions)
identified by the sponsor as non-critical often may be monitored less intensively.
There is increasing recognition that some types of errors in a clinical trial are more important
than others.31 For example, a low, but non-zero rate of errors in capturing certain baseline
characteristics of enrolled subjects (e.g., age, concomitant treatment, or concomitant illness) will
not, in general, have a significant effect on study results if the errors are distributed randomly. In
contrast, a small number of errors related to study endpoints (e.g., not following protocolspecified definitions) can profoundly affect study results, as could failure to report rare but
important adverse events. Based on FDA’s inspection and review experience, infrequent errors
in non-critical data are unlikely to alter FDA’s conclusions about whether a product is safe and
effective and whether participants’ safety was appropriately monitored.
B.

Risk Assessment

This guidance discusses the risk assessment, a component of risk management, as applied in the
context of clinical monitoring. Risk assessment generally involves identifying risks, analyzing
risks, and then determining whether risks need to be modified by implementing controls (e.g.,
processes, policies, or practices). The risk assessment recommended in this guidance to inform
development of a monitoring plan may also support efforts to manage risks across a clinical trial
(e.g., through modifying the protocol design or implementation) or development program. This
guidance does not provide comprehensive detail on how to perform a risk assessment. There are
many risk assessment methodologies and tools from a variety of industries that can be applied to
clinical trials. 32,33
Following the identification of critical data and processes (section IV.A), sponsors should
perform a risk assessment to identify and understand the nature, sources, and potential causes of
risks that could affect the collection of critical data or the performance of critical processes.
Risks to critical data and processes most merit consideration during risk assessment, to ensure
that monitoring efforts are focused on preventing or mitigating important and likely sources of
error in their conduct, collection and reporting.
Risk identification for monitoring purposes should generally consider the types of data to be
collected, the specific activities required to collect these data, and the range of potential safety
and other human subject protection concerns that are inherent to the clinical investigation (e.g.,
based on trial design or investigational product).

31

Baigent et al. Ensuring Trial Validity by Data Quality Assurance and Diversification of Monitoring Methods. Clin
Trials. 5: 49-55 (2008).
32
Guidance for industry, Q9 Quality Risk Management, June 2006.
33
ISO 31010:2009 Risk Management – Risk Assessment Techniques.

12

Contains Nonbinding Recommendations
The identified risks should be assessed and prioritized by considering the following:




the likelihood of errors occurring
the impact of such errors on human subject protection and trial integrity
the extent to which such errors would be detectable

Sponsors should use the results of the risk assessment in developing the monitoring plan (e.g.,
determining which risks may be addressed through monitoring, determining the types and
intensity of monitoring activities best suited to addressing these risks). Sponsors may also
determine that some risks are better managed through activities other than monitoring, for
example, modifying the protocol to remove the source of the risk. Sponsors should periodically
evaluate emerging risks and whether monitoring activities require modification to effectively
oversee the risks.
C.

Factors to Consider when Developing a Monitoring Plan

A monitoring plan ordinarily should focus on preventing or mitigating important and likely risks,
identified by the risk assessment, to critical data and processes.  The types (e.g., on-site,
centralized), frequency (e.g., early, for initial assessment and training versus throughout the
study), and extent (e.g., comprehensive (100% data verification) versus targeted or random
review of certain data (less than 100% data verification)) of monitoring activities will depend to
some degree on a range of factors, considered during the risk assessment, including the
following:


Complexity of the study design

More intensive monitoring (e.g., increased frequency and extent of review) may be necessary as
study design complexity increases. Examples may include studies with adaptive designs,
stratified designs, complex dose titrations, or multiple device placement studies.


Types of study endpoints

Endpoints that are more interpretative or subjective may require on-site visits to assess the
totality of subject records and to review application of protocol definitions with the CI. More
objective endpoints (e.g., death, hospitalization, or clinical laboratory values and standard
measurements) may be more suitable for remote verification. Endpoints for which inappropriate
subject withdrawal or lack of follow-up may impede study evaluation are likely to need more
intensive monitoring to identify the reason(s) subjects are withdrawing and to determine
whether follow-up can be improved.


Clinical complexity of the study population

A study that involves a population that is seriously ill or vulnerable may require more intensive
monitoring and consideration of on-site monitoring visits to be sure appropriate protection is
being provided.


Geography

Sites in geographic areas where there are differences in standards of medical practice or subject
demographics, or where there is a less established clinical trial infrastructure may require more
intensive monitoring and consideration of on-site monitoring visits.
13

Contains Nonbinding Recommendations


Relative experience of the CI and of the sponsor with the CI

CIs who lack significant experience in conducting and overseeing investigations, using a novel
or innovative medical device, or with the surgical procedure associated with medical device use
may benefit from more intensive monitoring and frequent communication to ensure CI
understanding of responsibilities. In addition, the relative experience of a sponsor with the CI
may be a factor in determining an appropriate monitoring plan.


Electronic data capture

Use of EDC systems with the capability to assess quality metrics (e.g., missing data, data error
rates, protocol violations) in real-time could help identify potentially higher risk sites for the
purpose of targeting sites in need of more intensive monitoring.


Relative safety of the investigational product

A study of a product that has significant safety concerns or for which there is no prior experience
in human clinical trials (e.g., a phase 1 pharmaceutical investigation or a device feasibility study)
may require more intensive monitoring and consideration of on-site monitoring visits to ensure
appropriate CI oversight of subject safety.


Stage of the study

A tapered approach to monitoring may be used where appropriate, with more intensive
monitoring at initiation and during early stages of a trial. For example, a tapered approach could
be used for a complex study where more intensive and on-site monitoring might be required
early, but where, once procedures are established, less intensive monitoring might suffice.
Similarly, a tapered approach could be used for relatively inexperienced CIs.


Quantity of data

Some centralized monitoring tools may be more useful as the quantity of data (e.g., size or
duration of trial, number of sites) collected increases.
D.

Monitoring Plan

For each clinical trial, the sponsor should develop a monitoring plan that describes the
monitoring methods, responsibilities, and requirements for the trial. The monitoring plan should
include a brief description of the study, its objectives, and the critical data and study procedures,
with particular attention to data and procedures that are unusual in relation to clinical routine and
require training of study site staff. The plan should also communicate the specific risks to be
addressed by monitoring and should provide those involved in monitoring with adequate
information to effectively carry out their duties. A monitoring plan may reference existing
policies and procedures (e.g., standard operating procedure describing general monitoring
processes or issue investigation and resolution). All sponsor and CRO personnel involved with
monitoring, including those who review or determine appropriate action regarding potential
issues identified through monitoring, should review the monitoring plan and associated
documents (e.g., standard operating procedures or other documents referenced in the monitoring
plan).

14

Contains Nonbinding Recommendations
Sponsors of device studies wishing to solicit feedback on their monitoring procedures prior to the
submission of the application may either submit a Pre-Submission,34 or contact CDRH’s
Division of Bioresearch Monitoring.35
Sponsors of drug studies may include specific questions about a monitoring plan in a request for
a formal meeting with FDA (e.g., end of phase 2 meeting).
The components of a monitoring plan might include the following:
1.

Description of Monitoring Approaches



A description of each monitoring method to be employed during the study and how it will be
used to address important risks and ensure the validity of critical data



Criteria for determining the timing, frequency, and extent of planned monitoring activities



Specific activities required for each monitoring method employed during the study, including
reference to required tools, logs, or templates



Definitions of events or results (e.g., findings from central monitoring activities) that would
trigger changes in planned monitoring activities for a particular CI
For example, if it is determined that a CI differs markedly from other CIs in making safetyrelated findings or other key safety metrics, in rate of enrollment, in the number of protocol
deviations, or in the rate of missing CRFs, the CI’s site should be considered for targeted onsite visits. The establishment of acceptable variation for particular critical data and processes
would facilitate identification of significant deviations.



Identification of possible deviations or failures that would be critical to study integrity and
how these are to be recorded and reported
For example, sponsors may wish to establish a specific mechanism for tracking and notifying
key study personnel of deviations related to collection or reporting of data necessary to
interpret the primary endpoint, regardless of which monitoring method identified a concern.

The study monitoring plan should also describe how various monitoring activities will be
documented, regardless of whether they are conducted on-site or centrally (see section V).
2.

Communication of Monitoring Results

 Format, content, timing, and archiving requirements for reports and other documentation of
monitoring activities (see section V)
 Process for appropriate communication
34

For more information, see FDA’s draft guidance Medical Devices: The Pre-Submission Program and Meetings
with FDA Staff. When final, this guidance will represent the FDA’s current thinking on this topic.
35
IDE regulations (21 CFR 812.25(e)) require that written monitoring procedures be submitted as part of the IDE
application.

15

Contains Nonbinding Recommendations
–

of routine monitoring results to management and other stakeholders (e.g., CRO, data
management)

–

of immediate reporting of significant monitoring issues to appropriate parties (e.g.,
sponsor management, CI and site staff, IRB, FDA), as necessary

–

from study management and other stakeholders to monitors

For example, data management personnel may provide monitors with routine reports of
outstanding CRFs or of common data queries at or across sites that may enable effective
targeting of monitoring activities.
3.

Management of Noncompliance



Processes for addressing unresolved or significant issues (e.g., significant non-compliance
with the investigational plan, suspected or confirmed data falsification) identified by
monitoring, whether at a particular site or across study sites



Processes to ensure that root cause analyses are conducted where important deviations are
discovered and that appropriate corrective and preventive actions (e.g., additional training on
a study or site level) are implemented to address issues identified by monitoring



Other quality management practices applicable to the clinical investigation (e.g., reference to
any other written documents describing appropriate actions regarding non-compliance)
4.

Ensuring Quality Monitoring



Description of any specific training required for personnel carrying out monitoring activities,
including personnel conducting internal data monitoring, statistical monitoring, or other
centralized review activities. Training should include principles of clinical investigations and
human subject protection. In addition, study-specific training should include discussion of
the trial design, protocol requirements, the study monitoring plan, applicable standard
operating procedures, appropriate monitoring techniques, and applicable electronic systems.



Planned audits of monitoring to ensure that sponsor and CRO staff conduct monitoring
activities in accordance with the monitoring plan, applicable regulations, guidance, and
sponsor policies, procedures, templates, and other study plans.  Auditing is a quality
assurance tool that can be used to evaluate the effectiveness of monitoring to ensure human
subject protection and data integrity.36



Many sponsors have successfully implemented on-site co-monitoring visits (i.e., monitoring
visits performed by both a study monitor and the monitor’s supervisor or another evaluator
designated by the sponsor or CRO) to evaluate whether monitors are effectively carrying out
visit activities, in compliance with the study monitoring plan. These visits may be conducted
either for randomly selected monitors or may be targeted to specific monitors, based upon
questions arising from review of monitoring visit documentation.

36

See ICH E6, section 5.19 and ISO 14155:2011, section 6.11 for additional information on audits.

16

Contains Nonbinding Recommendations
5.

Monitoring Plan Amendments

Sponsors should consider what events would indicate a need for review and revision of the
monitoring plan and establish processes to permit timely updates where necessary. For example,
a protocol amendment, change in the definition of significant protocol deviations, or
identification of new risks to study integrity could result in a change to the monitoring plan.
V.

DOCUMENTING MONITORING ACTIVITIES

Documentation of monitoring activities should generally include the following:


The date of the activity and the individual(s) conducting and participating in it



A summary of the data or activities reviewed



A description of any noncompliance, potential noncompliance, data irregularities, or other
deficiencies identified



A description of any actions taken, to be taken, or recommended, including the person
responsible for completing actions and the anticipated date of completion

Documentation of monitoring should include sufficient detail to allow verification that the
monitoring plan was followed.
Monitoring documentation should be provided to appropriate management in a timely manner
for review and follow-up, as indicated.
VI.

ADDITIONAL STRATEGIES TO ENSURE STUDY QUALITY

Although the focus of this guidance is on monitoring the oversight and conduct of, and reporting
of data from, clinical investigations, FDA considers monitoring to be just one component of a
multi-factor approach to ensuring the quality of clinical investigations. Many other factors
contribute to the quality of a clinical investigation. This section highlights additional areas that
complement monitoring and can affect study quality.
A fundamental component of ensuring quality monitoring is a sponsor’s compliance with
monitoring plans and any accompanying procedures.
A.

Protocol and Case Report Form Design

The most important tool for ensuring human subject protection and high-quality data is a welldesigned and articulated protocol. A poorly designed or ambiguous protocol may introduce
systemic errors that can render a clinical investigation unreliable despite rigorous monitoring.
Additionally, the complexity of the trial design and the type and amount of data collected may
influence data quality.37 The CRF, which captures the data required by the protocol, is another
37

Sponsors are encouraged to consult the appropriate review division within FDA's medical product centers with
questions about quality aspects of clinical trial design.

17

Contains Nonbinding Recommendations
critical tool for which design directly affects the quality of trial data. Care should be taken to
ensure that the CRF captures data accurately (e.g., as required by the protocol) and that the CRF
design and instructions facilitate consistent data collection across CI sites.
B.

Clinical Investigator Training and Communication

Clinical trial monitors conducting on-site visits have historically played an important role in
training the CI and site staff during a study. On-site visits also have served as a primary means
of providing feedback to CIs and study personnel on study conduct. Without meaningful
training prior to the conduct of a study and of appropriate instruction during the study (e.g., when
changes are made to the protocol), CIs and their staff may have difficulty carrying out a trial
correctly. Sponsors who plan less frequent or limited on-site monitoring should consider the
following:


Monitoring activities should include sufficient time for discussion of CI’s and site staff’s
responsibilities, feedback, and additional training, if needed, during the conduct of the study.



It may be necessary to implement alternative training (e.g., teleconferences, webcasts, online
training modules) and communication methods (see section III.B.1) for providing and
documenting ongoing, timely training and feedback, as well as to provide notification of
significant changes to study conduct or other important information.

C.

Delegation of Monitoring Responsibilities to a CRO

If a sponsor of an IND study delegates the responsibility for ensuring proper monitoring to a
CRO, FDA regulations (21 CFR 312.52) require the written transfer of any obligations from a
sponsor to a CRO and require the CRO to comply with the regulations.38 Although sponsors can
transfer responsibilities for monitoring to a CRO(s), they retain responsibility for oversight of the
work completed by the CRO(s) that assume this responsibility. Sponsors should evaluate CRO
compliance with regulatory requirements and contractual obligations in an ongoing manner. For
example, sponsor oversight of monitoring performed by a CRO may include the sponsor’s
periodic review of monitoring reports and vendor performance or quality metrics and
documented communication between the sponsor and CRO regarding monitoring progress and
findings.
Sponsors and CROs should consider additional factors when a sponsor transfers responsibilities
for monitoring to a CRO. Sponsors and CROs should prospectively establish a clear
understanding of both parties’ responsibilities and of the expectations for the conduct of the
transferred obligations. Sponsors should share information with a CRO that may inform
decisions a CRO may make regarding the monitoring practices for a trial (e.g., findings of a risk
assessment). Sponsors should prospectively evaluate monitoring procedures and monitoring
plans developed by a CRO to ensure the monitoring approach is consistent with applicable
aspects of the trial. In addition, sponsors and CROs should have processes in place for timely
38

The regulations for investigational device exemptions (21 CFR part 812) do not contain a provision for delegation
to a contract research organization.

18

Contains Nonbinding Recommendations
exchange of relevant information (e.g., significant monitoring findings, significant changes in
risk for a trial).
D.

Clinical Investigator and Site Selection and Initiation

In addition to regulatory requirements for CI selection, sponsors should consider factors such as
sponsor’s previous experience with the CI or site, workload of the CI and study staff, and
resource availability at the study site during CI and site selection.
Site initiation is a critical study activity that often involves sponsor personnel from a range of
disciplines, including monitors. Key components of site initiation include ensuring the CIs and
site staff understand their responsibilities, including applicable regulatory requirements as well
as study processes and procedures, including the sponsor’s processes for monitoring the
investigation. Communication and documentation tools for monitoring discussed in this
guidance can also be used for site selection and initiation activities.
VII.

PAPERWORK REDUCTION ACT OF 1995

This guidance contains information collection provisions that are subject to review by the Office of
Management and Budget (OMB) under the Paperwork Reduction Act of 1995 (44 U.S.C. 35013520).
The time required to complete this information collection is estimated to average 4 hours per
response, including the time to review instructions, search existing data resources, gather the data
needed, and complete and review the information collection. Send comments regarding this burden
estimate or suggestions for reducing this burden to:
Food and Drug Administration
Center for Drug Evaluation and Research
Office of Medical Policy
10903 New Hampshire Avenue, Bldg. 51, rm. 6352
Silver Spring, MD 20993-0002
This guidance also refers to previously approved collections of information found in FDA
regulations. The collections of information in part 312, including certain provisions under
subpart D, and part 812 have been approved under OMB Control Numbers 0910-0014, and
0910-0078. 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. The OMB
control number for this information collection is 0910-0733 (expires 03/31/2016).

19


File Typeapplication/pdf
File TitleGuidance for Industry Oversight of Clinical Investigations — A Risk-Based Approach to Monitoring
SubjectGuidance for Industry Oversight of Clinical Investigations — A Risk-Based Approach to Monitoring
AuthorFDA/CDER
File Modified2016-01-04
File Created2013-08-02

© 2024 OMB.report | Privacy Policy