CCRP virtual course Flashcards

1
Q

Assuring the safety, effectiveness, quality and security of human drugs, vaccines, biological products and medical devices fall under the direction of: LIST ALL LEVELS

A

United States Department of Health and Human Services
-FDA
–Center for Drug Evaluation and Research (CDER)
–Center for Biologics Evaluation and Research (CBER)
–Center for Devices and Radiological Health (CDRH)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Who develops SOPs? Who approves SOPs? What is the goal? Who is binded by the SOP

A

Developed by the organization
2. Sponsor, Investigational Site, Contracting Research Organization (CRO)
3. Compliance of regulations
4. Binding only on the organization.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

CFR Title 21 Part 11?

A

Electronic Records; Electronic Signatures

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

CFR Title 21 Part 50?

A

Informed consent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

CFR Title 21 Part 54

A

Financial Disclosure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

CFR Title 21 Part 56

A

Institutional Review Boards

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

CFR Title 21 Part 312

A

Investigational New Drug Application

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

CFR Title 21 Part 314

A

New drug appication

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

CFR Title 21 Part 812

A

Investigational Device Exemption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

CFR Title 21 Part 814

A

Premarket Approval of Medical Devices

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

CFR Title 45 Part 46

A

Federal research

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

45 CFR 46 part A is?

A

Common Rule

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is the largest grant making agency in the US

A

HHS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

The Common rule provides what

A

set of protections for research subjects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

The HHS regulations regarding human subject protection at
45 CFR 46 differ in limited but significant ways from the
FDA regulations regarding human subject protection at 21
CFR 50 and 56.
 When a research activity is governed by both sets of
regulations, then there are certain regulatory provisions that
are allowable under 45 CFR 46 that are not allowable under
21 CFR 50 and 56, and thus cannot be applied to the
research.

What are those provisions

A
  1. The application of the exempt research categories
  2. provision of waiver of consent
  3. Waiver of documentation of consent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is ICH

A

International conference of harmonizaiton

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What does ICH do

A

brings together the regulatory authorities and
pharmaceutical industry to discuss scientific and
technical aspects of pharmaceuticals and develop ICH
guidelines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Mission of ICH

A

achieve greater harmonisation
worldwide to ensure that safe, effective and high quality
medicines are developed, and registered and maintained
in the most resource efficient manner whilst meeting high
standards.
2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are the following ICH E series
E2A
E3
E6
E7
E8
E9
E11
Q
S
M

A

 E2A – Clinical Safety Data Management
 E3 – Clinical Study Reporting
 E6 – Good Clinical Practice
 E7 – Geriatric Populations
 E8 – General Considerations for Clinical Trials
 E9 – Statistical Principles
 E11 – Pediatric Populations
Q -Quality Guidelines
S- Safety
M- Multidisciplinary

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

 International Conference on Harmonization (ICH)
Good Clinical Practice (GCP) E6(R2) is

A

international ethical and scientific quality
standard for designing, conducting, recording
and reporting trials that involve the
participation of human subjects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

ICH GCP is a unified standard for who, and to do what?

What other jurisdictions did it cover

A

European Union, Japan and US.

facilitate the mutual
acceptance of clinical data by the regulatory
authorities in these jurisdictions

well as those of Australia, Canada,
the Nordic countries, and the World Health
Organization.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the focus of the ICH E6(R2) addendum to ICH E6 (R1)

Who does it affect the most?

A

focus of the revisions is on increasing
human subject protections and data integrity mainly
through better study design and conduct.

Therefore,
most of the changes affect the sponsor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is at the core of Nuremberg Code?

A

Informed consent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are the 10 research principles for the research subject from the nuremberg code?

A
  1. The voluntary consent of the human subject is
    absolutely essential.
  2. The experiment should yield fruitful results for the
    good of society, unprocurable by other methods or
    means of study, and not random and unnecessary in
    nature.
  3. The experiment should be designed and based on the results of animal experimentation and a knowledge
    of the natural history of the disease or other
    problem under study, that the anticipated results
    will justify the performance of the experiment.
  4. The experiment should be so conducted as to avoid all unnecessary physical and mental suffering and
    injury.
  5. No experiment should be conducted, where there is
    reason to believe that death or disabling injury will
    occur.
  6. The degree of risk to be taken should never exceed
    that determined by the humanitarian importance of
    the problem to be solved by the experiment.
  7. Proper preparations should be made and adequate
    facilities provided to protect the experimental subject
    against even remote possibilities of injury, disability, or
    death.
  8. The experiment should be conducted only by
    scientifically qualified persons with the highest
    degree of skill and care.
  9. During the course of the experiment, the human
    subject should be at liberty to bring the experiment
    to an end, if he/she has reached the physical or mental
    state, where continuation of the experiment seemed to
    him to be impossible.
  10. During the course of the experiment, the scientist in
    charge must be prepared to terminate the experiment at any stage, if he has probable cause to believe, in the
    exercise of the good faith, superior skill and careful
    judgement required of him, that a continuation of the
    experiment is likely to result in injury, disability, or death to the experimental subject.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the Declaration of Helsinki

A

 A statement of ethical principles for medical research
involving human subjects, including research on
identifiable human material and data.
 The Declaration is addressed primarily to physicians.
The WMA encourages others who are involved in
medical research involving human subjects to adopt
these principles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are ethical foundations of the Declaration of Helsinki?

A

 It is the duty of the physician to promote and safeguard
the health, well-being and rights of patients, including
those who are involved in medical research.
 Medical progress is based on research that ultimately
must include studies involving human subjects.
 The primary purpose of medical research involving
human subjects is to understand the causes,
development and effects of diseases and improve
preventive, diagnostic and therapeutic interventions
(methods, procedures and treatments).
 Even the best proven interventions must be evaluated
continually through research for their safety,
effectiveness, efficiency, accessibility and quality.
 Medical research is subject to ethical standards that
promote and ensure respect for all human subjects and
protect their health and rights.
 While the primary purpose of medical research is to
generate new knowledge, this goal can never take
precedence over the rights and interests of individual
research subjects.
 The responsibility for the protection of research
subjects must always rest with the physician or other
health care professionals and never with the
research subjects, even though they have given
consent.
 Research on patients or healthy volunteers requires the
supervision of a competent and appropriately qualified
physician or other health care professional.
 Groups that are underrepresented should be provided
appropriate access to participation in research. All
vulnerable groups and individuals should receive
specifically considered protection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

How does the Belmont report give boundaries to practice?

A

Practice refers to interventions that are designed solely
to enhance the well-being of an individual patient or
client that has a reasonable expectation of success. The
purpose of medical or behavioral practice is to provide
diagnosis, preventive treatment or therapy to particular
individuals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

How does the Belmont Report give boundaries to research?

A

Research designates an activity designed to test an
hypothesis, permit conclusions to be drawn, and
develop or contribute to generalizable knowledge.
Research is usually described in a formal protocol that sets
forth an objective and a set of procedures designed to reach
that objective.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are the three basic principles for the belmont report?

A

Respect of persons
beneficence
justice

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the specifics for respect of persons in Belmont report?

A
  1. Individuals should be treated as autonomous agents
    capable of deliberation about personal goals. To respect
    autonomy is to give weight to autonomous persons’ opinions
    and choices while refraining from obstructing their actions
    unless they are clearly detrimental to others.
     Persons with diminished autonomy are entitled to extensive
    protection. The extent of protection afforded should depend
    upon the risk of harm and the likelihood of benefit.
     Respect for persons demands that subjects enter into
    research voluntarily and with adequate information

Specifically in Informed Consent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are the specifics of Beneficence in Belmont report

A

 Persons are treated in an ethical manner not only by
respecting their decisions and protecting them from
harm, but also by making efforts to secure their wellbeing.
 Beneficence is an obligation:
 Do not harm
 Maximize possible benefits and minimize possible
harms

Assess risks and Benefits

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Specifics of Justice in Belmont report

A

 Fairness in distribution of burdens and benefits:
 To each person an equal share
 To each person according to individual need
 To each person according to individual effort
 To each person according to societal contribution
 To each person according to merit

Selection of Subjects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What are the sponsor responsibilities for study

A

 Select qualified investigators and monitors
 Provide information needed to conduct the
investigation properly
 Ensure Proper Monitoring, IRB Review and
Approval
 Ensure investigation conducted in
accordance with the general investigational
plan and protocols contained in the IND/IDE;
maintain effective IND/IDE
 Ensure FDA and all participating investigators are
promptly informed on significant new adverse
effects or risks with respect to the investigational
product
 Obtain agreement from the investigator/
institution for adherence to the protocol, to obtain
IRB approval, and GCP compliance
 Notify all involved parties, if warranted, of new
safety information adversely affecting subject
safety or IRB favorable opinion
 Secure Compliance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

When the sponsor obtains a signed 1572 (pharmaceutical
investigation) or Investigator Agreement (device
investigation) what does it include:

A
  1. CV
  2. Statement of Investigators relevant experience
  3. Explanation of termination circumstances
  4. Statement of investigator commitment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

If the sponsor wants to transfer any or all of their responsibilities to a contract research organization what do they have to do

A

 The transferred responsibilities must be described in
writing
 Any responsibility that is not described is assumed to
remain with the Sponsor
 The CRO assuming the responsibilities of a sponsor
must comply with the applicable regulations and is
subject to the same regulatory consequences as the
Sponsor for noncompliance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are monitoring plans that the sponsor creates?

A

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.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is a risk-based approach in monitoring?

A

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. is dynamic,
more readily facilitating continual improvement
in trial conduct and oversight.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the differences between on-site monitoring, remote monitoring and centralized monitoring

A

 On-site Monitoring: periodic site visits by a
clinical research associate conducted in-person
 Remote Monitoring: replace on-site with remote
activity
 Centralized Monitoring: electronic data capture
involving analytical evaluation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are the roles of a medical monitor?

A

provide medical expertise and
oversite to ensure clinical integrity and safety
accountability, and respond to subject safety and
trial management (e.g., inclusion and exclusion
criteria, unblinding procedure)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are data safety monitoring boards

A

independent
group who conducts periodic review and
evaluates study data for patient safety, study
conduct and progress – will have initiated “event
triggers” prior to study start

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the sponsor responsibilities around investigational product?

A

 Manufacturing, packaging, labeling, and
coding of the investigational product
 Providing the investigational product only to
investigators participating in an investigation
 Maintain drug and device accountability
records from manufacturing through use,
return and destruction

 Submit Safety Reports (Expedited
Reporting), if appropriate
 Provide pre-clinical and clinical study
reports in information amendments to
the IND/IDE
 Submit an annual report to the IND/IDE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What does the sponsor have to do if they determine the investigational product presents an unreasonable and significant risk to subjects?

A

 Discontinue all studies that present the risk
 Notify FDA, all investigators involved in the study(ies)
and their IRBs
 Assure return and accounting for all investigational
product, its return to the Sponsor (or on-site
destruction)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

A sponsor who discovers that an investigator is not
complying with the signed agreement, the investigational
plan, the IND/IDE requirements, any other applicable FDA
regulations, or any conditions of approval imposed by the
reviewing IRB or FDA must

A

 Promptly either secure compliance, or discontinue
shipments of the investigational product to the
investigator and terminate the investigator’s participation
in the investigation.
 A sponsor must also require that the investigator dispose
of or return the investigational product, unless this action
would jeopardize the rights, safety, or welfare of a
subject.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Under 21 CFR 54 requires financial disclosures; why does the sponsor require applicants to send these in?

A

Requires any clinical investigator conducting clinical studies covered by the regulation to certify the absence of certain financial interests and arrangements of clinical investigators that could affect the reliability of data submitted to FDA, or to disclose those financial interests and
arrangements to the agency and identify steps taken to
minimize the potential for bias (21 CFR § 54.4(a)).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Which financial disclosure form do clinical investigators have to submit if they do not have disclosable financial interests?

A

FDA 3454

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Which financial disclosure form do clinical investigators need to file if the individual has participated in financial arrangements of holds financial interests?

A

FDA 3455

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Who is responsible for registering trials and submitting results?

A

The responsible party for an applicable clinical trial (ACT)
must register the trial and submit results information. The
responsible party is defined as:
 The sponsor of the clinical trial, as defined in 21 CFR 50.3; or
 The principal investigator (PI) of such clinical trial if so
designated by a sponsor, grantee, contractor, or awardee, so
long as the PI is responsible for conducting the trial, has
access to and control over the data from the clinical trial, has
the right to publish the results of the trial, and has the ability to
meet all of the requirements for the submission of clinical trial
information.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

ACTs generally include interventional studies (with one or
more arms) of FDA-regulated drug, biological, or device
products that meet one of the following conditions:

A

 The trial has one or more sites in the United States
 The trial is conducted under an FDA investigational new drug application or investigational device exemption
 The trial involves a drug, biological, or device product
that is manufactured in the United States or its territories
and is exported for research

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

The overall purpose of monitoring by CRA is to verify what?

A

 The rights and well-being of human subjects are
protected
 Reported trial data are accurate, complete, and
verifiable from source documents
 The study is conducted in compliance with the
study protocol, the GCP guidelines, and
applicable regulations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What should monitoring visits reports include?

A

 the investigator’s name and site location
 date of the visit
 monitor’s name
 site personnel contacted
 summary of what information was reviewed
 significant findings and corrective actions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are other monitor responsibilities for visits?

A

 Review and follow-up by the sponsor on corrective
actions should be documented
 Identify missing data, inconsistent data, data outliers,
unexpected lack of variability and protocol deviations
 Examine data trends such as the range, consistency,
and variability of data within and across sites
 Evaluate for systematic or significant errors in data
collection and reporting at a site or across sites; or
potential data manipulation or data integrity problems
 Analyze site characteristics and performance metrics
 Select sites and/or processes for targeted on-site
monitoring.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What FDA form is for the statement of investigator or investigator agreement?

A

FDA 1572

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is required on the 1572

A

The name and address of the investigational
site(s)
 The name and address of the responsible IRB
 The name and address of clinical laboratories, if
any
 For devices, the corresponding requirements are
contained in an investigator’s agreement (no form
comparable to Form FDA 1572)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

After the investigator signs the 1572 what CRF TITLE WOULD THEY HAVE TO ENSURE RELATING TO OBTAINING INFORMED CONSENT AND WHICH ONE FOR IRB review and approval?

A

21 CRF 50

21 CFR 56

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

After the investigator signs the 1572 what CFR would they have to follow for reporting adverse events during the study

A

21 CFR 312.64

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

After the investigator signs the 1572 what CFR would they have to follow for maintaining adequate records and make study records available for inspection

A

21 CFR 312.62
21 CFR 312.68

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

How should study records be kept/ Which acronym helps remmeber?

A

Attributable, legible, contemporaneous original, accurate and complete (AKCoA-C)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are GCP for investigators to verify via accountability records

A

 Doses specified in the protocol were provided to
subjects.
 Drug/device was provided only to study subjects.
 Use by study subjects reconciles the amounts received
and returned to the sponsor.
 Product was stored as specified by the sponsor and in
compliance with applicable regulations.
 Device was used for its intended purpose.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are financial disclosures by clinical investigators for 21 CFR 54

A

 Any compensation made to the investigator by any sponsor
of the covered clinical study in which the value of
compensation could be affected by study outcome.

 A proprietary interest in the tested product including, but not limited to, a patent, trademark, copyright or licensing agreement.

 Any equity interest in any sponsor of the covered clinical study, i.e., any ownership interest, stock options, or other financial interest whose value cannot be readily determined through reference to public prices. The requirement applies to interests held during the time the clinical investigator is carrying out the study and for one year following completion of the study.

 Any equity interest in any sponsor of the covered study if the sponsor is a publicly held company and the
interest exceeds $50,000 in value. The requirement
applies to interests held during the time the clinical
investigator is carrying out the study and for one year
following completion of the study.
 Significant payments of other sorts are payments that
have a cumulative monetary value of $25,000 or more
and are made by any sponsor of a covered study to the
investigator or the investigator’s institution during the time the clinical investigator is carrying out the study and for one year following completion of the study.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

what is involved in drug discovery steps for drug development in non-clinical/pre-clinical activities:

A

 Identify potential new compounds
 Test in appropriate laboratory and animal
models to assess potential activity in humans
 Screening may involve thousands of
molecules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What steps are included during the develop the dosage form for drug development?

A

 Manufacture pure, stable drug substance
(active ingredient)
 Test various formulations to optimize the drug
product (active ingredient and excipients)
 Stability testing in packaging planned to be
used for clinical trials and the marketed
product
 Begin defining product labeling expectations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is the goal of safety pharmacology studies?

A

 Studies that investigate the potential
undesirable pharmacodynamic effects of a
substance on physiological functions in
relation to exposure in the therapeutic range
and above.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What are the goals of animal studies?

A

 Establish general safety in multiple species
 Summarize the toxicities observed
 Determine the safety margins between the
planned human dose range and toxic effects
in animals
 Optimize the dose range, formulation, and
frequency of administration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What are results used for in single-dose toxicity studies in multiple animal species?

A

results used to select doses for
repeated dose studies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What results are used for Repeated dose studies to assess toxicity after
multiple administrations

A

results used to
determine doses used for chronic
administration, e.g. – carcinogenicity studies in
animals

66
Q

There are minimum requirements before clinical trials may begin.

What are included in applications to initiate clinical trials IND?

A

 Summaries of all acute toxicity studies
 28-day studies in one rodent and one non-rodent
species, usually rats and dogs
 Regulatory authorities can require more information
 Non-clinical studies have to support the
conclusion that the drug can be administered
safely to human subjects

67
Q

What CFR correlates to general principles of IND submmission?

A

21 CFR 312.22

68
Q

What are general principles of IND submissions (21 CFR 312.22) for Phases 2 and 3, FDA:

A

 helps assure the quality of the scientific evaluation
is adequate to permit evaluation of the drug’s
safety and efficacy
 determines if studies in Phases 2 and 3 are likely
to yield data capable of meeting regulatory
standards for marketing approval

69
Q

During drug development IND general Considerations (21 CFR 312.22) the amount of information needed depends on:

A

 Novelty of the drug, e.g. – a new mechanism of
action
 Extent the drug has been studied previously
 Known or suspected risks
 Phase of development

70
Q

What does the Initial IND focus on?

A

 The general investigational plan
 Summary of preclinical studies
 Protocols for specific human studies

71
Q

 Reasons for IND amendments include:

A

 New protocols or amendments to studies in
progress
 Submission of reports for completed preclinical
and clinical studies, as well as relevant preclinical
and clinical information in the public domain
 Expedited reporting of serious adverse events
 Changes in ingredients or manufacturing process
for the active ingredient or final drug product
 Submission of the annual report

72
Q

What CFR is associated with IND format and Content

A

(21 CFR 312.23)

73
Q

A sponsor initiated IND must contain:

A

 Cover sheet (Form FDA 1571)
 Table of contents
 Introductory statement and general investigational
plan
 Investigator’s brochure
 Protocol(s) for clinical trials
 Chemistry and manufacturing information
 Pharmacology and toxicology information
 Previous human experience with
investigational drug
 Additional information, e.g. – drug
dependence and abuse potential, exposure
to radiation, plans for pediatric studies

74
Q

What CFR is associated with administrative actions

A

(21 CFR 312.40)

75
Q

Under administrative action 21 CFR 312.40 an IND can go into effect when…

A

30 days after the FDA acknowledges receipt
unless FDA notifies sponsor of clinical hold
 Upon earlier notification clinical investigations
may begin

76
Q

What CFR is associated with clinical holds and requests for modifications

A

(21 CFR 312.42)

77
Q

what is a clinical hold, what is put on, and who issues it

A

is an order issued by the FDA to the
Sponsor to delay a proposed clinical investigation or
to suspend an ongoing investigation

78
Q

If a clinical hold happens to a proposed study, what happens
/

A

subjects may not be given the
investigational drug

79
Q

If a clinical hold happens to an ongoing study, what happens?

A

no new subjects may be given the
investigational drug and subjects already taking
the drug should be discontinued unless
continuation is specifically permitted by FDA

80
Q

What are grounds for clinical holds on Phase 1

A

 Human subjects are or would be exposed to an
unreasonable and significant risk of illness or
injury
 The clinical investigators named in the IND are not
qualified by reason of their scientific training and
experience to conduct the investigation described
in the IND;
 Investigator’s brochure is misleading, inaccurate,
or materially incomplete
 The IND does not contain sufficient information
required under 312.23 to assess risk to subjects of
the proposed studies

81
Q

What are additional grounds for clinical hold phases 2 and 3

A

The protocol is clearly deficient in design to meet
its stated objectives

82
Q

what CFR is associated with withdrawal of an IND

A

(21 CFR 312.38)

83
Q

A sponsor may withdraw an IND at any time
without prejudice by:

A

 Notifying the FDA
 Stopping all studies and notifying the
investigators
 All drug is returned to the sponsor or destroyed
as directed by the sponsor
 If withdrawn for safety reasons, the sponsor must
notify the investigators and the IRBs of those
reasons

84
Q

What are protocol amendments allowed under an IND

A

 New protocols
 When a sponsor wants to conduct a study not
already covered by the IND
 Changes in current protocols
 Any change in Phase I that significantly affects
safety of subjects
 Any change in Phase II or III that affects safety of
subjects, scope of investigation, or scientific quality
of study
 New investigator added to a current study

85
Q

What CFR is associated with information amendments with IND amendments and what are they?

A

312.31

Information Amendments (312.31):
 Chemistry/Microbiology
 Pharmacology/Toxicology
 Clinical

86
Q

What are other amendments

A

 IND Safety Reports
 Response to FDA Request for Information
 Response to Clinical Hold
 General Correspondence
 Annual Report
 Request for Re-instatement of IND

87
Q

A new Drug Application for a drug or biologic is filed when the sponsor considers that there is sufficient info to meet the regulatory requirements for approval

A

 Chemistry, manufacturing and controls (CMC), full
reports appended
 Summaries of all non-clinical studies, full reports
appended
 Summaries of all clinical studies , full reports
appended
 Integrated summary of safety (ISS)
 Integrated summary of efficacy (ISE)
 Risk evaluation and mitigation strategy (REMS)

88
Q

When is a decision on the NDA supposed to be made?

A

Within 12 months after submission

89
Q

What is expanded access

A

is a potential pathway for a patient with an immediately
life-threatening condition or serious disease or
condition to gain access to an investigational medical
product (drug, biologic, or medical device) for treatment
outside of clinical trials when no comparable or satisfactory
alternative therapy options are available

90
Q

What are the different types of expanded access

A

Emergency use
Compassionate USe
Treatment Use
Continued Access

91
Q

When are expanded releases appropriate?

A

 Patient has a serious disease or condition, or whose life is
immediately threatened by their disease or condition.
 There is no comparable or satisfactory alternative therapy
to diagnose, monitor, or treat the disease or condition.
 Patient enrollment in a clinical trial is not possible.
 Potential patient benefit justifies the potential risks of
treatment.
 Providing the investigational medical product will not
interfere with investigational trials that could support a
medical product’s development or marketing approval for
the treatment indication.

92
Q

What are characteristics of phase 1 studies

A

Normal volunteers or subjects with condition
under study
 Usually conducted at Phase 1 units, tightly
controlled, in-patient setting
 Usually single-center studies
 Small numbers of subjects/study and overall:
“…generally in the range of 20 to 80.”

93
Q

What are characteristics of phase 2 clinical trials

A

 Initial demonstration of efficacy in subjects
with the condition under investigation
 Obtain short-term safety data
 Multicenter, well-controlled studies
 Relatively small number of subjects per
study
 Combined population: “…usually involving
no more than several hundred subjects.”

94
Q

What are characteristics of phase 3 clinical trials

A

 Confirmation of short-term efficacy and
safety
 Establish long-term efficacy and safety,
as appropriate
 Assess overall therapeutic value
 Expanded controlled and uncontrolled
studies
 Additional evidence of efficacy and safety
 Establish overall benefit-risk relationship
 Supports the final labeling content
 Combined population from several hundred
to several thousand subjects.”

95
Q

What are characteristics of phase 4 clinical trials

A

(Post-marketing):
 Address FDA requirements for additional
information not in NDA
 Delineate additional information about
the drug’s risk, benefits and optimal use;
continue assessing overall therapeutic
value
 Surveillance for less common adverse
events (spontaneous reports, registries)
 Design and number of subjects
depends on study objective
 May be similar to Phase 2 or Phase 3
studies in design

96
Q

What are the regulatory submission paths for pharmaceutical companies

A

Preclinical > IND> PHase I, II, III > NDA

97
Q

wHAT IS THE REGULATORY SUBMISSION PATH FOR MEDICAL DEVICE PLAC

A

If needed, Pre-IDE > If clinical trials needed: IDE via IRB approval or FDA approval > Pilot, Pivotal > LEtter to file, 510(k), OMA

98
Q

What is the development timeline for drugs/biologics

A

Has to have Phase I, II, III and has thousands of subjects over 10-12 years long

99
Q

What is the development timeline for devices

A

Pilot study, Pivotal Stdy, hundreds of subjects 1+ years

100
Q

What is the definition of a device

A

An instrument, apparatus, implement, machine, contrivance,
implant, in vitro reagent, or other similar article, including any
component, part, or accessory which is:
 Recognized in the official National Formulary, or the United States
pharmacopeia, or any supplement to them
 Intended for use in the diagnosis of disease or conditions, or in
the cure, mitigation, treatment, or prevention of disease in man
or other animals
 Intended to affect t

101
Q

What is the definition of intended use

A

The general purpose of the device or
its function. The intended use of a device
encompasses the indications for use.

102
Q

What is the definition of indications for use

A

The disease or condition the
device will diagnose, treat, prevent, cure or mitigate,
including a description of the patient population for
which the device is intended.

103
Q

What established FDA’s jurisdiction over costmetics and medical devies

A

Federal Food, Drug and Cosmetic ACt 1938

104
Q

What amendment was the first major amendment specifically for devices and established three regulatory classes for medical devices based on degree of control necessary to assure that the various types of devices are safe and effective?

A

Medical Device Amendments 1976

105
Q

What is the CFR for Medical Device Reporting

A

21 CFR 803

106
Q

What is the CFR for INvestigational Device Exemption (counterpart to IND requirements 21 CFR 312)

A

21 CFR 812

107
Q

Premarket Approval of Medical
Devices (counterpart to NDA requirements 21 CFR 314)

A

21 CFR 814

108
Q

Medical Device Classification PRocedures

A

21 CFR 860

109
Q

During device development there are distinguished classes according to risk factors, what are those classes and describe them

A

 Class I: Lowest risk, clinical trials generally not
required
 Class II: Moderate risk, usually requires 510(k),
Might require PMA
Generally requires some clinical data
 Class III: Highest risk; PMA usually required,
clinical trials absolutely necessary

110
Q

What general controls are regulatory class I device developments required to follow

A

 Prohibition against adulterated or misbranded devices
 Pre-market notification 510(k) requirements
 GMPs
 Registration of manufacturing facilities
 Listing of device types
 Generally do not require clinical trials

Examples include: elastic bandages, examination gloves, hand-held surgical instruments

111
Q

For Class I regulatory for device developments describe the general controls

A

General controls alone are sufficient to
provide reasonable assurance of the safety
and effectiveness of the device

112
Q

Regulatory Class II for device development

A

Are devices for which general controls alone are
insufficient to assure safety and effectiveness,
and existing methods are available to provide
such assurances.

113
Q

In addition to complying with general controls, Class II devices are also subject to special controls, what are they?

A

 Special controls may include:
 Special labeling requirements
 Mandatory performance standards
 Post-market surveillance
Examples: powered wheelchairs, infusion pumps,
and surgical drapes

114
Q

Regulatory class II device deleopment

A

devices are those for which insufficient information exists to assure safety and effectiveness solely through general or special controls

examples include: replacement heart valves, silicone gel-filled breast implants,
implanted cerebella stimulators, and implantable pacemaker pulse
generators

115
Q

What is an IDE and what does it do

A

investigational device exemption

is the mechanism the sponsor uses to submit documents to the FDA; protocols reports correspondence. IT applies to all clinical investigations of devices to determine safety and effectiveness

An approved IDE permits a device to be shipped lawfully for the purpose of conducting investigations of the device

116
Q

Significant Risk (SR) and Non Significant Risk (NSR)
determination is based on risk of the device to the subject ______________________________

A

as it will be used in the study

117
Q
  • A SR device study is defined as a study of a
    device that presents a potential for serious risk to
    the health, safety, or welfare of a subject:
A
  • is an implant,
  • is used in supporting or sustaining human life,
  • is of substantial importance in diagnosing, curing,
    mitigating or treating disease, or otherwise prevents
    impairment of human health, or
  • otherwise presents a potential for serious risk to the
    health, safety, or welfare of a subject.
  • Misdiagnosis and/or error in treatment caused by
    inaccurate test results would be considered a significant
    risk.
118
Q

A NSR device study is defined as a study _____________

A

that
does not meet SR criteria

119
Q

The sponsor makes an itial determination for SR or NSR studies… what are the next steps for NSR

A

 If NSR, the sponsor submits device description, rationale for determining NSR status, and protocol to the investigator who provides to the IRB
 If IRB concurs, the sponsor sends device description,
rationale for determining NSR status, IRB concurrence and protocol to the FDA

120
Q

The sponsor makes an intial determination for SR or NSR studies… what are the next steps for

A

 If either the sponsor or IRB consider the device to be SR,
the sponsor consults CDRH about submitting an IDE
 FDA can override the determination by either sponsor or
IRB; FDA determination of SR or NSR is final

121
Q
  • NSR Abbreviated Requirements (Sponsors):
A
  • Label device
  • Maintain records
  • Ensure investigators maintain records and make
    reports
  • Obtain IRB approval; SR vs. NSR designation
  • Ensures informed consent is obtained (IRB can waive
    if minimal risk)
  • Submit reports
  • Monitor the study
  • Refrain from promotion and other practices
122
Q

Yes. Under § 812.5 an investigational device or its
immediate package must bear a label with the following
information:

A

 the name and place of business of the manufacturer,
packer, or distributor;
 the quantity of contents, if appropriate; and
 the statement, “CAUTION Investigational device.
Limited by Federal (or United States) law to
investigational use.”
 The label must also describe all relevant contraindications,
hazards, adverse effects, interfering substances or devices,
warnings, and precautions.

123
Q

 Sponsor must report the results of an evaluation of
an unanticipated adverse device effect to FDA
and all reviewing IRBs and investigators within _____
working days after the Sponsor first received
notice of the adverse effect.

A

10

124
Q

Sponsor must report the following when?

Withdrawal of IRB approval

A

5WD

125
Q

Sponsor must report the following when?
Withdrawal of FDA Approval

A

5WD

126
Q

Sponsor must report the following when?
Current List of Investigators

A

(every 6 months)

127
Q

Sponsor must report the following when?
Progress Reports

A

at least yearly to IRB
at least yearly to FDA for SR

128
Q

The legally marketed device(s) to which equivalence
is drawn is known as the

A

predicate device

129
Q

________________________ is the FDA process of
scientific and regulatory review to evaluate the
safety and effectiveness of Class III medical
devices.

A
  • Premarket approval (PMA)
130
Q

Medical device manufacturers as well as other firms
involved in the distribution of devices must follow certain
requirements and regulations once devices are on the
market to include:

A

 Implementing tracking systems
 Reporting device malfunctions
 Reporting serious injuries or deaths
 Registering establishments where devices are produced
or distributed

131
Q

What are the required elements of informed consent

A

The study involves research
 Explain purposes of the research
 Expected duration of the subject’s participation
 Description of procedures to be followed
 Identification of experimental procedures
2. Description of any reasonably foreseeable risks or
discomforts to the subject
3. Description of any benefits to the subject or to others
which may reasonably be expected from the research
Disclosure of appropriate alternative
procedures or courses of treatment, if any,
that might be advantageous to the subject
5. Statement describing extent, if any, to
which confidentiality of records identifying
the subject will be maintained:
 FDA regulations require notice that the Food and Drug
Administration may inspect the records
 ICH extends this notice to “regulatory authority(ies)”, the
Sponsor and/or its representatives, and IRB
representatives
Explanation as to whether any compensation
and whether medical treatments are
available if injury occurs
7. Explanation of who to contact for pertinent
questions regarding:
 The research project
 Research subjects’ rights
 Research-related injury
8. Participation is voluntary
 Refusal will involve no penalty or loss of benefits to
which the subject is otherwise entitled
 The subject may discontinue participation at any time
without penalty or loss of benefits to which the subject
is otherwise entitled

132
Q

What is 21 CFR 50.27

A

Short form consent

133
Q

Who signs what when using a short form consent

A

 Oral consent followed by subject and
witness signing and dating short form
 Person obtaining consent and witness sign
written summary
 Copy of written summary and short form
given to subject or representative

134
Q

What is 21 CFR 50.23

A

Informed Consent Exception from General
Requirements

135
Q

 If, in the investigator’s opinion, immediate use of the test
article is required to save the subject’s life and time is
not sufficient to obtain independent confirmation before
using the test article:

A

 The investigator makes the determination
 Within 5 working days after use, a physician not
otherwise involved in the clinical trial will review the
use and make a written evaluation.
 In both scenarios, the required documentation must be
submitted to the IRB within 5 working days after the use
of the test article.

136
Q

What is 21 CFR 50.24

A

Informed Consent - Exception from
Informed Consent Requirements for
Emergency Research

137
Q

 21 CFR 50 Subpart D -
 45 CFR 46 Subpart B -
 45 CFR 46 Subpart D -

A

Children
Pregnant minors, Neonates
Children

138
Q

The IRB can waive assent requirements if:

A

 clinical trial involves no more than minimal risk
 waiver will not adversely affect the rights and welfare
of the subjects
 clinical trial could not practically be carried out without
the waiver
 When appropriate, pertinent information is given to
the subjects after participation

139
Q

Rules of characteristics of those on an IRB

A

-minimum 5 members
- varied backgrounds
-diversity and expertise
-no gender discrimination
-at least one non scientific member
-at least one not affiliated with institution
 No IRB member may participate in initial or
continuing review in any project that might
involve conflict of interest
 Experts may assist the board, but may not vote

140
Q

What are the vulnerable populations

A

 prisoners,
 pregnant women, children,
 handicapped,
 mentally disabled,
 economically or educationally disadvantaged

141
Q

The following should be submitted to the
IRB for review:

A

 Protocol and Amendments
 Written informed consent
 Subject recruitment procedures
(advertisements)
 Investigators brochure
 Available safety data
Information about payments and
compensation to subjects
 Current curriculum vitae and/or other
documentation of qualifications for
investigator and sub investigators (critical
staff)
 Additional material as individual IRBs may
find necessary

142
Q

What are the key topics for IRB continuing review?

A

1) Risk Assessment;
2) Adequacy of Informed Consent;
3)Local Issues, and
4) Trial Progress

143
Q

Progress report should include:

A

 Risk/benefit assessment (e.g. Data Monitoring Cttee Report)
 The number of subjects entered (locally and study wide)
 Summary of:
 amendments since initial or last continuing review
 experiences ( including benefits and adverse events)
 unanticipated problems
 complaints
 subject withdrawals and reason for withdrawals
 Research results thus far

144
Q

what is 21 CFR 56.110

A

Expedited Review

145
Q

What are the four types of control groups

A

 Placebo concurrent control
 Active-treatment concurrent control
 Dose-comparison concurrent control
 No-treatment concurrent control

146
Q

What is the trial design for placebo concurrent control

A

 Placebo is expected to provide no or only minimal
efficacy.
 This type of study is intended to show the investigational
product is better than a presumed inactive treatment
regarding efficacy.
 Placebo also provides a valuable reference for
determining which safety endpoints (AEs, abnormal
laboratory, vital signs, ECGs, physical examinations,
etc.) might be associated with the investigational product.
 Generally used when the treatment period is short, and
lack of treatment of the medical condition is not expected
to have clinically relevant impact on the subject’s overall
health (example: hypertension treated for 6-8 weeks)

147
Q

Trial design for active-treatment concurrent control

A

 Active controls generally are used when it is
unethical to use placebo, e.g. – anti-infective
drugs, chemotherapy, other conditions where
non-treatment could result in significant
health consequences, even in the short term
 Generally are intended to demonstrate
equivalence (neither better nor worse than
the comparator) or non-inferiority (at least as
good as the comparator)

148
Q

Trial Design:
Dose-comparison Concurrent Control

A

 Multiple dose levels of the investigational
product might be evaluated in the same
study, with prospective randomization to
treatment groups
 These studies might be conducted using the
dose-escalation design
 Primary objectives are to compare the
efficacy and safety at multiple doses of the
investigational product

149
Q

Trial Design:
No-treatment Concurrent Control

A

 Subjects are randomly assigned to test
treatment or no (i.e., absence of) study
treatment.
 The principle difference between this design and
a placebo-controlled trial is that subjects and
investigators are not blind to treatment
assignment.
 Usually used only when it is difficult or
impossible to double-blind (e.g., treatments with
easily recognized toxicity).

150
Q

______________________________is an adverse reaction that is
both serious and unexpected

A

suspected and unexpected adverse reaction

151
Q

 IND Safety Reports
 Sponsor must notify the FDA and all participating
investigators of any adverse events______ that are both __________
 This should be done as soon as possible but no
more than ______ after the sponsor’s
initial receipt of information

A

associated
with the use of a drug

both serious and
unexpected

15 calendar days

152
Q
  • An unexpected fatal or life threatening
    experience associated with the use of the drug
    must be reported to FDA in writing no later than
    ________ after the receipt of information
  • Followed by a written report no more than ________ after the initial receipt of
    information
A

7 calendar days

15 calendar days

153
Q

what is FDA 21 CFR 312.62

A

Investigator recordkeeping and record retention

154
Q

 The IRB should retain all relevant records
for a period of ______years after completion of
the trial and make them available upon
request from the regulatory authorities

A

3

155
Q

What are the types of FDA investigator inspections

A

PDUFA-related inspections
Directed inspections

156
Q

When would PDFUA-related inspections occur

A

 “Routine” inspections
 Triggered by submission of an NDA (NME, pivotal
studies not conducted in US, only foreign data)

157
Q

When would directed inspections occur

A

 “For cause”
 Triggered by allegations that raise concerns regarding
data integrity or the rights, welfare and safety of study
subjects have been compromised

158
Q

what are some triggers for directed inspections

A

 Participation in a pivotal study
 High enrollment
 Irregularities in drug management
 High percentage of subjects excluded from the perprotocol population

159
Q

If an IRB’s contact or chair person information changes, the IRB must
revise its registration information by submitting any changes in that information within ______ days of the change.

A

90

160
Q

An IRB’s decision to
review new types of FDA-regulated products (such as a decision to review studies pertaining to food additives whereas the IRB
previously reviewed studies pertaining to drug products), or to discontinue reviewing clinical investigations regulated by FDA is a
change that must be reported within ________ days of the change.

A

30

161
Q

An IRB’s decision to disband is a change that must be reported within
_____ days of permanent cessation of the IRB’s review of research.

A

30