UNIT VI



History of regulatory affairs

 Regulatory affairs (RA), also called government affairs, is a profession within regulated industries, such as pharmaceuticals, medical devices, energy, and banking. Regulatory affairs also has a very specific meaning within the healthcare industries (pharmaceuticals, medical devices, biologics and functional foods).

Regulatory affairs professionals (aka regulatory professionals) usually have responsibility for the following general areas:

Ensuring that their companies comply with all of the regulations and laws pertaining to their business.

Working with federal, state, and local regulatory agencies and personnel on specific issues affecting their business. i.e. working with such agencies as the Food and Drug Administration or European Medicines Agency (pharmaceuticals and medical devices); The Department of Energy; or the Securities and Exchange Commission (banking).

Advising their companies on the regulatory aspects and climate that would affect proposed activities. i.e. describing the "regulatory climate" around issues such as the promotion of prescription drugs


The regulatory function in healthcare industries is vital in making safety and effective healthcare products available worldwide. Individuals who ensure regulatory compliance and prepare submissions, as well as those whose main job function is clinical affairs or quality assurance are all considered regulatory professionals.
Regulatory professionals are employed in industry, government and academia and are involved with a wide range of products, including:

pharmaceuticals
medical devices
in vitro diagnostics
biologics and biotechnology
nutritional products
cosmetics
veterinary products

The regulatory professional's roles and responsibilities often begin in the research and development phases, moving into clinical trials and extending through premarket approvals, manufacturing, labeling and advertising and postmarket surveillance.

The healthcare industries were the first to be significantly regulated in the modern era. Much of this regulation has stemmed from avoiding the repetition of disasters, and has tended to be led by the USA due to size of the market and its technological lead:
Diphtheria Epidemic led to 1902 Biologics Control Act
Publication of The Jungle by Upton Sinclair led to 1906 Pure Food and Drugs Act
Elixir of Sulfanilamide led to the 1938 Food Drug and Cosmetic Act
Thalidomide led to the 1962 Kefauver-Harris Amendments
Dalkon Shield led to the 1976 Medical Device Amendments
Bjork-Shiley Heart Valves led to the 1990 Safe Medical Devices Act
In the USA, this regulation is largely written directly into law and codified in Title 21 of the Code of Federal Regulations.


Main concepts QSE

The ICH topics are divided into four categories and ICH topic codes are assigned according to these categories.

1.       Quality Guidelines
Harmonisation achievements in the Quality area include pivotal milestones such as the conduct of stability studies, defining relevant thresholds for impurities testing and a more flexible approach to pharmaceutical quality based on Good Manufacturing Practice (GMP) risk management.
Stability Q1A - Q1F
Analytical Validation Q2
Impurities Q3A - Q3D
Pharmacopoeias Q4 - Q4B
Quality of Biotechnological Products Q5A - Q5E
Specifications Q6A- Q6B
Good Manufacturing Practice Q7
Pharmaceutical Development Q8
Quality Risk Management Q9
Pharmaceutical Quality System Q10
Development and Manufacture of Drug Substances Q11
Cross-cutting Topics

2.       Safety Guidelines

ICH has produced a comprehensive set of safety Guidelines to uncover potential risks like . testing strategy for assessing the QT interval prolongation liability: the single most important cause of drug withdrawals in recent years.
Zip file with all ICH Safety Guidelines in Word format
Carcinogenicity Studies S1A - S1C
Genotoxicity Studies S2
Toxicokinetics and Pharmacokinetics S3A - S3B
Toxicity Testing S4
Reproductive Toxicology S5
Biotechnological Products S6
Pharmacology Studies S7A - S7B
Immunotoxicology Studies S8
Nonclinical Evaluation for Anticancer Pharmaceuticals S9
Photosafety Evaluation S10
Cross-cutting Topics

3.      Efficacy Guidelines
The work carried out by ICH under the Efficacy heading is concerned with the design, conduct, safety and reporting of clinical trials.  It also covers novel types of medicines derived from biotechnological processes and the use of pharmacogenetics/ pharmacogenomics techniques to produce better targeted medicines.
Zip file with all Efficacy Guidelines in Word format
Clinical Safety E1 - E2F
Clinical Study Reports E3
Dose-Response Studies E4
Ethnic Factors E5
Good Clinical Practice E6
Clinical Trials E7 - E11
Clinical Evaluation by Therapeutic Category E12
Clinical Evaluation E14
Pharmacogenomics E15 - E16
Cross-cutting Topics
(Ref:   http://www.ich.org/products)


Sources of information Regulatory affairs for studies in human subjects

The Office of Research Compliance and Regulatory Affairs (“ORCRA”) provides
administrative support to the Institutional Review Boards and the other research review
committees and compliance functions. With respect to human subjects’ research, these
committees include the Radiation Safety Committee, which reviews and provides
approval for all human research protocols involving the use of ionizing radiation and the
Institutional Biosafety Committee, which reviews and approves all human research
protocols involving the use of infectious agents or recombinant DNA. The University’s
Compliance Hotline program for anonymous reporting of allegations of non compliance
in any aspect of research or physician billing is administered by ORCRA. The Quality
Assurance/Quality Improvement program is also a service of ORCRA. It provides postapproval monitoring of human subjects’ research projects. The office is directed by the  Research Compliance Officer, who reports to the Vice President for Research.

The Institutional Review Board

The University of Cincinnati has established three Institutional Review Board (“IRB”)
panels to review all proposed research involving human subjects to ensure that the rights
and welfare of participants in research are adequately protected. The University of
Cincinnati IRB serves as the IRB of record for the University of Cincinnati, University
Hospital, Inc., The Shriners’ Institute for Burned Children, The Cincinnati Veterans
Affairs Medical Center, and The Drake Center. The IRBs are composed primarily of
faculty members from disciplines in which research involving human subjects is integral
to that discipline's work, as well as several members from the community whose primary
interest is in non-scientific areas. The Board(s) membership, policies, and procedures are
governed by an Assurance agreement with the Federal government.

Human subjects’ research means any activity intended to obtain and record information
from or about individuals for research purposes. Any undertaking in which students,
faculty, or staff investigate and/or collect data on human subjects or use existing data or
specimens collected from living human subjects for research purposes, requires review by
the Institutional Review Board prior to initiation of the project. This includes both funded
and non-funded research, including dissertations, masters’ theses, pilot studies, class
projects, and non-funded, faculty-directed research if the following conditions are met:

• the research is sponsored by the University, or
• the research is conducted by or under the direction of any University employee or
agent of this institution in connection with his/her institutional responsibilities, or
• the research is conducted by or under the direction of any University employee or
agent of this institution using any University property or facility, or
• the research involves the use of the University's non-public information to
identify or contact human research subjects or prospective subjects

Studies involving drugs, devices, or other invasive interventions will be reviewed by the
Medical IRBs. Those studies involving surveys, interviews and observation techniques
will be reviewed by the Behavioral and Social Sciences Board. The human subjects
review process is administered through the Institutional Review Board Office for the
University

The Office of Research Compliance Training is responsible for developing and delivering
workshops and training classes, as well as designing computer-based training courses on
research regulatory compliance issues. In addition to delivering training that meets the
needs of the research investigators, courses are developed in response to changing
regulatory and accreditation requirements, e.g., HIPAA. Efforts are coordinated with the
Regulatory Compliance Officer, the multiple Institutional Review Boards and IRB
support staff, the Institutional Animal Care and Use Committee and IACUC support staff,
the Institutional Biosafety Committee and IBC support staff, and others, as needed. Many
programs are eligible for continuing education credits.



The following documents must be on file at HJF before funding is released for any research involving human subjects:

Initial local Institutional Review Board (IRB) approval letter with signature(s) (electronically signed is acceptable).

Second-level/Third-level approval letter with signature(s) (electronically signed is acceptable) [A second-level review may be required depending on the funding source and where Principal Investigator (PI) /Associate Investigator (AI) is located/billeted. If second-level/third-level approval is required, all documentation must be submitted through the Office of Regulatory Affairs and Research Compliance, who in turn will forward to the appropriate agency/department for review/and approval.

Scientific committee review approval letter with signature(s) (electronically signed is acceptable)
Protocol application form and final approved version of protocol with signature(s) where indicated (electronically signed is acceptable).

Copy of approved stamped informed consent form(s) and HIPAA authorization form (as applicable). If the IRB has granted a waiver of informed consent, provide approval letter with signatures (electronically signed is acceptable).
All protocol appendices: Questionnaires, instruments, surveys and data collection forms (as applicable), etc.

For protocols and associated documents that are conducted internationally and are not in the English language, the following additional documentation is required:
A translation letter certifying that the foreign version of the protocol and informed consent is an accurate and true translation of the English version. The letter should include the signature, name, address, phone number, fax number, and email of the translator, and the date translated.

Copies of all documents in the foreign language

Current copies of HIPAA compliant CVs for PI, AIs and all personnel listed on the protocol. All personal identifiers should be deleted from the CVs. The PHS 398/2590 Biographical Sketch format can be used as a reference.
Human subjects protection training certificates for all personnel listed on a protocol and for all other personnel involved in the research. Course must have been completed within the past three years. Human subjects training can be completed online athttp://www.citiprogram.org. For HJF employees/HJF contractors: Select HJF as your affiliation.
Copies of advertisements for recruiting human subjects and associated IRB approvals for such advertisements.

Current assurances from rDNA, radiation, biosafety committees, environmental safety committee, and/or relevant impact statements (Directorate of Information Management, laboratory and personnel), if required.
Copies of current state licenses and Basic Life Support (BLS) certification for physicians, nurses, nurse practitioners, physician's assistants and any other licensed health care provider(s) listed on the protocol.

For studies that involve an Investigation New Drug (IND) or Investigational Device Exemption (IDE), the following additional documents are required:
a copy of the signed form FDA-1572
a copy of the investigator’s brochure (for drug studies)/product brochure (for device studies)
Good Clinical Practice (GCP) training certification for personnel listed on protocol and the FDA 1572.
For studies with material transfers (biological materials, such as select agents, cell lines, plasmids, and vectors, chemical compounds, data, and even some types of software and machinery), a copy of the Material Transfer Agreement (MTA).
Copy of any Data Use Agreements (DUAs) between collaborating parties.
Documents Required Until Study Closure

Continuing review/annual progress reporting:
Copies of continuing review/annual progress report (APR) with signature(s)(electronically signed is acceptable) submitted to the IRB.
IRB approval letter for continuing review
Stamped informed consent form(s) and HIPAA form from continuing review (if applicable).
Amendment documents:

Copies of protocol amendment request(s) with signature(s) (electronically signed is acceptable) with all supporting documents submitted to the IRB.
IRB approval letter and any approved forms for amended protocols (informed consent forms, HIPAA, etc.).

Copies of advertisement(s) (not previously submitted) for recruiting human subjects and IRB approvals with signature(s) (electronically signed is acceptable) for such advertisements.
Copies of all adverse event reports and protocol deviations submitted to the IRB(s) and corresponding approval/acknowledgement by the IRB(s). All adverse events must be reported as indicated in the protocol and according to the institutions’ policies where the research is conducted, Department of Defense requirements, and federal regulations.

Copies of approved presentations and publications resulting from the study. This includes all submissions, whether or not they are accepted for publication. Approval from the relevant IRB(s) and other departments as required by the institution where study is conducted. Approval must be received prior to submission of data for publication or presentation.Copy of updated Investigator Brochure /Product brochure (as required for studies conducted under an IND or IDE application).

Copies of any audit reports received from the IRBs, clinical investigations departments, Food and Drug Administration or other regulatory agencies. If you are notified that you will be audited, please contact HJF Regulatory Affairs as soon as possible, so that we are aware of the audit and can help you prepare.

Final report submitted to IRB and IRB approval letter of study closure with signature(s) (electronically signed is acceptable) and second and third level approvals as required.


  
What data is needed

Current Federal law requires that a drug be the subject of an approved marketing application before it is transported or distributed across state lines. Because a sponsor will probably want to ship the investigational drug to clinical investigators in many states, it must seek an exemption from that legal requirement. The IND is the means through which the sponsor technically obtains this exemption from the FDA.
During a new drug's early preclinical development, the sponsor's primary goal is to determine if the product is reasonably safe for initial use in humans, and if the compound exhibits pharmacological activity that justifies commercial development. When a product is identified as a viable candidate for further development, the sponsor then focuses on collecting the data and information necessary to establish that the product will not expose humans to unreasonable risks when used in limited, early-stage clinical studies.
FDA's role in the development of a new drug begins when the drug's sponsor (usually the manufacturer or potential marketer) having screened the new molecule for pharmacological activity and acute toxicity potential in animals, wants to test its diagnostic or therapeutic potential in humans.  At that point, the molecule changes in legal status under the Federal Food, Drug, and Cosmetic Act and becomes a new drug subject to specific requirements of the drug regulatory system.
There are three IND types:
An Investigator IND is submitted by a physician who both initiates and conducts an investigation, and under whose immediate direction the investigational drug is administered or dispensed.  A physician might submit a research IND to propose studying an unapproved drug, or an approved product for a new indication or in a new patient population.
Emergency Use IND  allows the FDA to authorize use of an experimental drug in an emergency situation that does not allow time for submission of an IND in accordance with  21CFR , Sec. 312.23 or Sec. 312.34.  It is also used for patients who do not meet the criteria of an existing study protocol, or if an approved study protocol does not exist.
Treatment IND is submitted for experimental drugs showing promise in clinical testing for serious or immediately life-threatening conditions while the final clinical work is conducted and the FDA review takes place.
There are two IND categories:
Commercial
Research (non-commercial)

The IND application must contain information in three broad areas:
Animal Pharmacology and Toxicology Studies - Preclinical data to permit an assessment as to whether the product is reasonably safe for initial testing in humans.  Also included are any previous experience with the drug in humans (often foreign use).
Manufacturing Information -  Information pertaining to the composition, manufacturer, stability, and controls used for manufacturing the drug substance and the drug product.  This information is assessed to ensure that the company can adequately produce and supply consistent batches of the drug.
Clinical Protocols and Investigator Information - Detailed protocols for proposed clinical studies to assess whether the initial-phase trials will expose subjects to unnecessary risks.  Also, information on the qualifications of clinical investigators--professionals (generally physicians) who oversee the administration of the experimental compound--to assess whether they are qualified to fulfill their clinical trial duties.  Finally, commitments to obtain informed consent from the research subjects, to obtain review of the study by an institutional review board (IRB), and to adhere to the investigational new drug regulations.

Once the IND is submitted, the sponsor must wait 30 calendar days before initiating any clinical trials.  During this time, FDA  has an opportunity to review the IND for safety to assure that research subjects will not be subjected to unreasonable risk.
 (ref: http://www.fda.gov/drugs/)

Current and future European requirements and procedures

On 17 July, the European Commission released the long-awaited proposal for a Regulation on
clinical trials on medicinal products for human use (the Proposal). The future regulation will
replace the Clinical Trials Directive 2001/20/EC, the revision of which has been advocated by
the pharmaceutical industry, academia, and patients for years. Directive 2001/20/EC aims at
harmonising the requirements throughout the EU but has not prevented divergent national
requirements that, especially for multinational clinical trials, substantially hinder a swift handling
of clinical trial applications and generate a very burdensome and costly administration. The
Proposal primarily seeks to cut the red tape and to facilitate the conduct of multinational trials
and restore competitiveness in clinical research in Europe.
The future Regulation will be more complete and detailed than Directive 2001/20/EC (more than
90 articles and five annexes) and will revise current rules, in particular as regards the
authorisation procedures, and introduce new principles, such as co-sponsoring. Overall, the new
regime should reduce administrative costs for industry and better reflect the variety of clinical
trials. The Commission chose a Regulation as legislative instrument to ensure that identical rules
will apply throughout European Union. This will increase harmonisation of the rules, but certain
issues, such as ethics and sponsor liability, will remain governed by national law.
The submission of the Proposal to the European Parliament and Council is the first step of the
legislative process. The Proposal will probably be heavily debated and substantially amended, in
particular by the Council as the Member States are keen on preserving their powers over clinical
trials conducted in their territories. It is difficult to determine how long the legislative process will last. The Commission expects the new rules to come into effect in 2016 after a transitional
period of two years.

Scope
The future regulation has the same scope as Directive 2001/20/EC, i.e., interventional clinical
trials and investigational medicinal products (IMPs). The Proposal, however, amends the existing
definitions (clinical trial, non-interventional clinical trial) and introduces new definitions, such as
“clinical study”, “low-intervention clinical trial”, or “auxiliary medicinal product” (to replace the
concept of “non-investigational medicinal product”).
The concept of “normal clinical practice” at the Member State level is key for distinguishing
between a clinical study and a clinical trial and between a standard clinical trial and a lowintervention clinical trial. Although legally defined, the term refers to the clinical practice as it exists in each Member State and could lead to national divergences on the qualification of a
study.

Risk Categorization

The proposed new rules show a risk-based approach to clinical trials and distinguish between
low-intervention clinical trials and other clinical trials. A “low-intervention clinical trial” is a clinical  trial where the IMPs are authorised and used in-label (or their use is a standard treatment in any  f the Member States concerned) and any additional diagnostic or monitoring procedures pose  minimal additional risk or burden for the subjects compared to normal clinical practice in “any” Member State concerned. The reference to ”any of the Member States concerned” suggests that  a trial which qualifies as a low-interventional clinical trial in one Member State should also qualify as such in other Member States, but that does not seem to be the intention (as illustrated by the German version of the Proposal).  Because of their very low risk profile, low-intervention clinical trials benefit from less burdensome  requirements (safety reporting, labelling, and insurance) and shorter authorisation timelines.

Streamlined Authorisation Procedures

Currently, a clinical trial is approved by a competent national authority and an ethics committee
in each Member State where the sponsor wants to conduct the trial, and the authorisation
procedure is conducted in parallel in all the Member States. These parallel national procedures
are replaced by a form of decentralised procedure , which also applies to purely national trials.

Authorisation

The key features of the new authorisation procedure for clinical trials are the following:
 Submission of a single and harmonised application dossier through an EU portal linked to an
EU database. The content and format of the dossier are defined in an annex to the Proposal
(which can be amended by delegated acts).
 Choice of a Reporting Member State (RMS) by the sponsor. The RMS is the interface
between the sponsor and the other Member States for the assessment of Part I aspects of
the trial.
 Validation of the application by the RMS.
 Two-part assessment of the application:
o Each part covers specific aspects that are exhaustively listed in the Proposal.
o Part I covers general aspects, such as the anticipated therapeutic and public health
benefits, the risks and inconveniences for the trial subjects, or the investigator brochure.
It is to be assessed by the RMS in coordination with the other involved Member States.
o Part II covers ethical, local, and national aspects, such as the informed consent or the
arrangements for compensating investigators or recruiting trial subjects. It is to be
assessed separately by each Member State.
o Parallel assessment of Part I and Part II.
 Single decision by each Member State, which covers Part I and Part II and must be notified to
the sponsor within 10 days of the assessment.
 Tacit approval system based on the positive assessment of Part I by the RMS:
 The RMS assessment of Part I is binding on the other Member States unless they refuse the
assessment (“opt-out”). The refusal can only be based on two, mainly ethical, grounds.
 In the absence of a notification of the decision within the deadline, the conclusion of the RMS
assessment of Part I qualifies as the authorisation decision of the Member State on the
application (also for Part II).
 The impact of a negative RMS assessment of Part I is unclear.
 Timelines are set out for each phase of the procedure. They vary depending on the nature of
the clinical trial (shorter for low-intervention trials) or the medicinal product (longer for
advanced therapy medicinal products). Except for low-intervention clinical trials, they are
longer than under Directive 2001/20, but the tacit approval system provides for better
compliance.
 The Commission manages the EU portal, and supports the cooperation of the Member States
with the assistance of a Clinical Trials Coordination and Advisory Group (group of the national
contact points). There is no role for the EMA.


Addition of a new Member States

The procedure for extending the trial to a new Member State is similar to the authorisation
procedure, but with shorter timelines. The RMS is the same as for the authorisation procedure,
but its role is limited as there is no additional assessment of Part I aspects.

Substantial Amendments

After having been authorised, a clinical trial may be modified. The modification is subject to
authorisation where it has a substantial impact on the safety or rights of the subjects or on the
reliability and robustness of the data generated. Non-substantial amendments should simply be
entered into the EU database. The Proposal does not define or further describe what constitutes
a substantial amendment, and the current Commission guideline may remain applicable. The
deadlines for authorisation are shorter than for the authorisation procedure. The RMS is the
same as for the main authorisation procedure.

Streamlined Safety Reporting

The rules on safety reporting are streamlined and simplified. So, reporting of certain adverse
events by the investigator to the sponsor can be excluded in the protocol, and suspected
unexpected serious adverse reactions (SUSARs) can be reported by the sponsor directly to
EudraVigilance (EU pharmacovigilance database). More detailed rules on safety reporting, which
partly codify the current Commission guidance, are contained in an annex to the Proposal.

Conduct of Clinical Trials Outside of the EU

An increasing number of clinical trials are conducted outside the EU and the results are also used
to support marketing authorisation applications in the EU. Building on an EMA reflection paper,
the Proposal implicitly requires that clinical trials conducted outside the EU comply with
regulatory requirements at least equivalent to those in the EU. This obligation however is already
included in Annex I to Directive 2001/83/EC. Also, it is unclear how the equivalence between
foreign and EU clinical trial regimes will be assessed and, more importantly, how third countries
will be convinced to adopt rules that are similar to the EU rules.

Indemnity and Insurance

Directive 2001/20 introduced an obligation for the sponsors to compensate trial subjects for
damages resulting from clinical trials (in accordance with national law) by way of an indemnity or
insurance. Under the Proposal, low-intervention clinical trials are exempted from the mandatory
indemnity or insurance requirement. For the other (interventional) clinical trials, the sponsor
must ensure compensation through an insurance or an indemnification mechanism, and the
Proposal requires the Member States to set up a national indemnification mechanism that works
on a not-for-profit basis (and may require payment of a fee by commercial sponsors).

Transparency

Under Directive 2001/20, clinical trials are registered in a central database (EudraCT). That
database is not publicly available except for paediatric trials, but protocol-related information and
trial results are indirectly made public through another database (EudraPharm). The Proposal
makes EudraCT fully accessible to the public, subject to personal data, commercially confidential
information, and the need for effective supervision of the conduct of the clinical trial by a
Member State. This increased clinical trial transparency is in line with the general European
trend in the pharmaceutical sector, but adequate attention is needed to protect business
secrets.

(Ref: http://www.cov.com/files/Publication/b7133e14-05a4-47c4-9229  39b21218825/Presentation/PublicationAttachment/5f11bb6d-be8f-47c2-8818-28c45a6bd135/New_Rules_for_Clinical_Trials_Proposed_in_Europe.pdf)


US perspective

Since at least the mid-1990s, college and university students, faculty, and staff who conduct oral history interviews have increasingly found their interviewing protocols subject to review by their local Institutional Review Board (commonly referred to as an IRB), a body charged by the federal government with protecting the rights, interests, and dignity of human research subjects – or, as some prefer, research participants. The review has generally not been a constructive process for oral historians, indeed, in many cases has been quite contentious as principles and practices developed within biomedical and behavioral frameworks have been applied to a more humanistic form of inquiry. The following sections will:

outline the regulatory framework and historical context for IRB review of research involving what are termed “living individuals;”
explain why IRBs claim authority to review oral history and suggest why they are increasingly choosing to do so; identify primary difficulties oral historians face when submitting interview protocols for IRB review; explain efforts to address these problems made by professional bodies including the Oral History Association; and suggest what to do when facing IRB review of oral history interviewing projects.

Framework and Context: Authority to review research involving living individuals resides with the federal government, as authorized by Title II of the National Research Act of 1974 (P.L. 93-348) and codified in Title 45 (Public Welfare), Part 46 (Protection of Human Subjects) of the Code of Federal Regulations, commonly referred to as 45 CFR 46 or the Common Rule. The Office of Human Research Protections (OHRP) within the U.S. Department of Health and Human Services (HHS) oversees implementation of 45 CFR 46; however, actual responsibility falls to local, typically campus-based IRBs. IRBs are empowered by 45 CFR 46 to approve or disapprove research protocols – or more typically, request their revision – based on their conformity to the principles and practices delineated by the regulations, as well as subsequent guidance issued by OHRP.

The result of lengthy and at times contentious deliberations within government and professional circles, 45 CFR 46 was first codified in 1981 and, in its current form, in 1991. However, its origins date to the 1960s and 1970s, with policies established first by the U.S. Public Health Service and then by the entire U.S. Department of Health, Education, and Welfare (HEW, predecessor to HHS) requiring independent review of research involving human subjects funded by these agencies. More broadly, concern for the welfare of human subjects of research was provoked, on the one hand, by the explosion of government funded medical research in the years after World War II; and on the other, by outrage at the violation of human rights demonstrated by certain medical experiments, including Nazi doctors’ experiments on Holocaust victims and the Tuskegee Syphilis Study.

Three ethical principles underlie 45 CFR 46, each codified in procedures to ensure adherence to the principle. These were first articulated in The Belmont Report, issued in 1979 by the federally mandated National Commission for the Protection of Human Subjects of Biomedical and Behavioral Research, which was charged with identifying the ethical principles underlying the conduct of research involving living human beings. These principles are: a respect for persons as autonomous individuals capable of making decisions about their own behavior, or, in the case of individuals with “diminished autonomy,” as needing specific protections, codified in 45 CFR 46 as procedures for securing informed consent from research subjects; beneficence, or the obligation to minimize harm and maximize benefits to research subjects, codified as a directive to assess and balance risks and benefits of research; and justice, or the equitable selection of research subjects, codified as terms for selection of subjects.

Oral History: The Common Rule defines “research” as “a systematic investigation, including research development, testing and evaluation, designed to develop or contribute to generalizable knowledge;” and “human subject” as “a living individual about whom an investigator (whether professional or student) conducting research obtains (1) data through intervention or interaction with the individual, or (2) identifiable private information” (46.102[d] and [f]). Most IRBs – and oral historians – assume that oral history interviewing is “research” (even though it doesn’t exactly fit the above definition) and because it involves “interaction,” further defined as “communication or interpersonal contact between investigator and subject (46.102 [f]),” that it is subject to IRB review.

However, 45 CFR 46 also exempts most oral history from regulatory oversight. The regulations state that “research involving . . . interview procedures . . . [is exempt from this policy] unless (i) information obtained is recorded in such a manner that human subjects can be identified directly or through identifiers linked to the subjects and [this “and” is all important] (ii) disclosure of the human subjects’ responses outside the research could reasonably place the subjects at risk of criminal or civil liability, or be damaging to the subjects’ financial standing, employability, or reputation” ( 46.101 [b] [2]). In other words, while most oral history narrators can be identified if by no other means than their recorded voice, most do not reveal information that could “reasonably place [them] at risk.” However, following advice issued by OHRP in 1995, the IRB – not the researcher – determines exemption, so we find ourselves in the position of submitting a description of our oral history research protocols to an IRB in order to apply for an exemption from applying to the IRB.

Oral history’s relationship to IRB review is further complicated by the fact that in 1998 ORHP, in an effort to ease the regulatory burden increasingly imposed upon oral history – and in a perverse example of the cure being worse than the disease – issued guidance that included oral history in a list of research modes that could enjoy “expedited review,” which means that the full IRB does not need to review a research proposal, that review can be delegated to one or more members of the Board. “Exempt” or “expedited”? – a contradiction that OHRP has never clarified.

Federal and institutional policies also support IRB review of oral history. The Common Rule itself applies only to research funded by the seventeen federal agencies that subscribe to its terms – and notably, the National Endowment for the Humanities, which is the federal agency most likely to support research in history and other humanities fields, is not among them. In fact, however, most universities and research centers that receive funding from any of these seventeen agencies – and very few do not – apply the terms of 45 CFR 46 to all research conducted within or sponsored by the institution, whether or not the research receives funding from one of these agencies, or is funded at all. For it is in an institution’s self-interest to apply the same oversight to all research conducted within the institution. The regulations themselves also require assurance that ethical safeguards are in place for all human subjects research conducted within an institution for that institution to be eligible for funds from any of the seventeen agencies subscribing to the Common Rule. Most institutions simply choose to extend the terms of the Common Rule – and hence IRB review – to all such research.

Yet another reason for the extension of IRB review to oral history interviewing is what some have termed “mission creep.” Within the last several years, egregious violations of requirements for human subjects review in biomedical research have led to the suspension of all human subjects research at several major institutions. Simultaneously, colleges and universities have become increasingly fearful of litigation or bad press over dangerous or controversial research. As a result, IRBs, already risk-averse bodies, have tended to become hyper-vigilant, extending review to forms of minimal risk research in the social sciences and humanities that previously had escaped their purview. Arguably, oral history’s increasing visibility and acceptance within the academy during this same period have also served to bring it to the attention of local IRBs.

Difficulties: Not surprisingly given the abuses that formed the context within which 45 CFR 46 was developed, the regulations assume a scientific, largely biomedical, model of research. While the social sciences have always been included within the regulatory embrace, their inclusion has been disputed by critics among both policy makers and scholars since the 1960s. The lack of fit between the epistemologies of various social science disciplines and the terms of regulation has never been given due consideration. Psychology is the discipline that has most concerned regulators and with which they are most familiar; history – and the humanities in general – have been a “foreign language.”2 As a result, IRB review of oral history is an awkward and at times contentious affair.

The problems oral historians confront cluster in two areas: 1.) the potential an interview might have for inflicting psychological harm on the narrator within the interview; and 2.) maintaining narrator privacy in any subsequent use of the interview. Concern about psychological harm derives rather awkwardly from that section of the Common Rule that defines minimal risk as the “harm or discomfort . . . encountered . . . during the performance of routine physical or psychological examinations or tests” (46.102 [i]). The Guidebook issued by OHRP explicates the notion of psychological harm, stating that “stress and feelings of guilt or embarrassment may arise simply from thinking or talking about one’s own behavior or attitudes on sensitive topics.” In fact, the assumption that harm can result from talking about sensitive topics is contradicted by both anecdotal and more scientific evidence, which suggests that talking about even the most difficult of subjects is generally not perceived as harmful but indeed as having a salutary effect.3 And insofar as oral history narrators freely agree to be interviewed about past experiences (one area in which oral historians and IRBs do agree is the need for informed consent!), it seems patronizing to assume they need to be protected from talking about those experiences, that they cannot decide which experiences, including traumatic experiences, they wish to talk about and which they do not.

Still, concerns about psychological harm have led some IRBs to require oral historians to submit lists of interview questions for review, advise against or prevent them from asking questions about potentially sensitive subjects, and require interviewers who do ask potentially sensitive questions to make referrals to counseling services available to narrators at the conclusion of the interview. These proscriptions make little sense to oral historians: our inquiries are open-ended dialogues that cannot be confined to a prescripted set of questions and we do not always know in advance if an inquiry will enter into a sensitive area – or what might even be a sensitive area for any given narrator. Our practice often requires a series of ethically sensitive decisions within the context of a specific interview relationship.4 The Oral History Association’s Evaluation Guidelines, which codifies the fundamental principles of oral history, provides more appropriate guidance, stating that interviewers “should encourage interviewees . . . to address issues that reflect their concerns” and “must respect the rights of interviewees to refuse to discuss certain subjects,” and “clearly explain [this option] to all interviewees.”

The second area of concern, privacy, also arises from language in the Common Rule, which does not exempt – and hence raises concern about – interviews for which “disclosure of the human subjects’ responses outside the research could reasonably place the subjects at risk of criminal or civil liability, or be damaging to the subjects’ financial standing, employability, or reputation” ( 46.101 [b] ). In an effort to protect narrators from such potential risk or damage, IRBs frequently require that they remain anonymous; and that researchers either retain completed interviews in their possession or, preferably, destroy them after the research is completed. For those who come to oral history via anthropology or sociology, maintaining confidentiality of sources is quite compatible with normal disciplinary practice. Yet requiring anonymity violates a fundamental principle of oral history. For historians, anonymous sources lack credibility – knowing the identity of a narrator allows the historian to gauge that person’s relationship to the topic at hand and hence assess the perspective from which he speaks. While OHA’s Evaluation Guidelines do allow interviewees to choose anonymity “under extreme circumstances,” when failure to do so could have adverse consequences, the operating assumption is for narrators to be identified and most, in fact, choose to be. Typically, narrators are proud of having contributed their story to the permanent record and wish to be associated with it.

Furthermore, historians neither keep to themselves nor destroy evidence but rather are enjoined by their own disciplinary ethics to provide open access to sources, so that others can evaluate and build upon their scholarship. Oral history is fundamentally an archival practice, defined by the assumption that interviews are conducted for the permanent record and are to be made publicly available. Again to cite the Evaluation Guidelines: “With the permission of interviewees, interviewers should arrange to deposit their interviews in an archival repository that is capable of both preserving the interviews and eventually making them available for general use.”

For all matters of privacy, “with the permission of interviewees” is the operative phrase within oral history, not the terms of 45 CFR 46: Because oral history interviews are a copyrightable document, owned by the narrator, he or she must sign over – to either an individual researcher or a public archive – rights to the interview via a legal release form. Without this, no one, including the interviewer, can legally use the interview. This release allows the narrator to define the terms with which the interview can be used, including if he/she wishes to remain anonymous or restrict access to the interview for a period of years. The Evaluation Guidelines also recognize the potential for exploitation in oral history, if not “risk of criminal or civil liability,” or damage to a narrator’s “financial standing, employability, or reputation.” They state that “interviewers should guard against possible exploitation of interviewees and be sensitive to the ways in which their interviews might be used;” and “should be sensitive to the communities from which they have collected oral histories, taking care not to reinforce thoughtless stereotypes or to bring undue notoriety to them.” In fact, these principles are more expansive – and more appropriate to oral history – than the terms of 45 CFR 46.

Still, it must also be said that for historians, a deep disjunction exists between the Common Rule’s concern for privacy and the canons of historical inquiry. At times information in an interview, if made public, can indeed place a person at risk of criminal or civil liability, or be damaging to his financial standing, employability, or reputation. Yet historians’ deepest responsibility is to follow the evidence where it leads, to discern and make sense of the past in all its complexity; not necessarily to protect individuals from their past actions. The American Historical Association’s (AHA) “Statement on Standards of Professional Conduct” puts it this way: “Professional integrity in the practice of history requires awareness of one’s own biases and a readiness to follow sound method and analysis wherever they may lead” (bold face in the original). And the OHA Guidelines suggest a similarly critical approach, calling on both interviewees and interviewers to “mutually strive to record candid information of lasting value” and enjoining interviewers to “strive to prompt informative dialogue through challenging and perceptive inquiry.” In this we are akin to journalists and unlike medical professionals, who are indeed enjoined to do no harm.

Historians are not alone in their concern that 45 CFR 46 can be used to constrain critical inquiry. Legal scholar Philip Hamburger has argued that insofar as it requires researchers to get permission to conduct research, 45 CFR 46 violates First Amendment rights of freedom of speech and the press.5 In a 2006 report, “Research on Human Subjects: Academic Freedom and the Institutional Review Board,” the American Association of University Professors recommends that “research on autonomous adults whose methodology consists entirely in collecting data by surveys, conducting interviews, or observing behavior in public places, be exempt from the requirement of IRB review—straightforwardly exempt, with no provisos, and no requirement of IRB approval of the exemption.”6 The Illinois White Paper, “Improving the System for Protecting Human Subjects: Counteracting IRB ‘Mission Creep’“, produced by the Center for Advanced Study at the University of Illinois, concludes that “most journalism and oral history cannot be appropriately reviewed under the Common Rule.”

Efforts to Address the Problems: Responding to increasing concerns about the lack of congruence between the terms of Common Rule and the practice of oral history, in 2003 the OHA and AHA, after a series of discussions, secured the Office of Human Research Protections’ concurrence with a policy statement that excluded most oral history interviewing from IRB review on the grounds that it does not conform to the regulatory definition of research as seeking “generalizable knowledge,” that is to say historians “do not reach for generalizable principles of historical or social development; nor do they seek underlying principles or laws of nature that have predictive value and can be applied to other circumstances for the purpose of controlling outcomes.”

Some IRBs, however, questioned this policy and subsequent commentary from OHRP, developed in response to inquiries from the University of California Los Angeles IRB, refined its concurrence with a policy of exclusion by attempting to distinguish between interviews that are not intended “to draw conclusions, inform policy, or generalize findings” and hence are not subject to IRB review, and those that are so intended and hence are subject to review, including “creat[ion of an] archives for the purpose of providing a resource for others to do research.” To complicate matters even more, in response to further questions raised by OHA and AHA, OHRP also affirmed its original policy statement.7 If this sounds contradictory, it is: Oral history’s status vis-à-vis 45 CFR 46 and hence its relationship to IRB review remains unresolved at the federal level.8 Meanwhile, a few IRBs have attempted to address apparently contradictory advice from OHRP and adopted reasonable policies regarding IRB review of oral history.9 Others exclude interviews conducted for classroom assignments, if they are not made public beyond the classroom, on the grounds that they are pedagogy, not research. Most IRBs, however, which have considerable decision-making autonomy, continue to require review of oral history without parsing OHRP’s statements and without considered attention either to ways oral history differs from biomedical or social science research or to historians’ concerns. The AHA continues to monitor the situation and responds on behalf of all historians to requests by OHRP for public comment on various proposed policy changes.

What To Do: Some oral historians, it must be said, are supportive – or at least tolerant – of IRB review and willingly submit interview protocols. Others, with less vigilant IRBs, simply fly below the regulatory radar. And some researchers have insisted that the regulations do not, or should not, apply to them and have refused compliance with apparently no adverse consequence.11 Still, while a tenured professor might ignore or refuse compliance with impunity, this may not be an acceptable course of action for many and is especially ill advised for graduate students, whose dissertations can – indeed have – been held up at the administrative level for failure to comply with IRB review.

So, for a college or university affiliated student, scholar, or staff member confronting IRB review for a forthcoming oral history project, the following might be a reasonable course of action:

Inform yourself of the federal regulations governing research on human subjects, the context within which they were developed, and recent critiques of human subjects review in the humanities and social sciences.
Seek allies within your department or relevant administrative unit as you develop an approach to the IRB.

Take a proactive approach with the IRB, informing it of the principles and practices governing history in general and oral history in particular and insisting that it conform to the federal requirement that an IRB include or consult with individuals who can knowledgeably review any proposed research, in this case an individual with adequate knowledge of oral history.12  Oral historians might also consider cooperating with the IRB to develop a review policy appropriate to oral history, one that might include, as some have suggested, departmental review of oral history research projects with some minimal reporting to the IRB .
  
(ref: http://www.oralhistory.org/about/do-oral-history/oral-history-and-irb-review/)
  

Recognizing why clinical research has to meet the needs of regulatory affairs

The regulatory landscape has continued to evolve in response to product safety, regulatory
compliance, new technologies, improved understanding of disease states, stakeholder and
customer needs and global imperatives, with new and increasing regulations, regulatory guidance and oversight. The global regulatory affairs group and the regulatory professionals in the
biopharmaceutical industry occupy a central and pivotal role to all the functional groups. The
regulatory team is charged with a strong leadership role that ensures compliance with regulations and enables understanding and interpretation of the dynamic regulatory landscape,
while creating opportunities in the highly-regulated and complex environment.
The challenging landscape is largely a result of the success of the biopharmaceutical industry
in delivering medical therapies for many disease states, safety catastrophes from use of products
post-approval; and product quality compliance issues. These have sharpened regulatory authorities’ focus on product benefit/risk profiles and related stakeholders’ views on cost effectiveness
and patient access. The regulatory professional has to be equipped and poised to effectively guide
the organization to success with a credible voice, informed strategic guidance and objective evaluation.
The demand for globally acceptable products heightens the imperative for harmonization of
regulatory requirements to lend efficiency and cost effectiveness to the process of product development, manufacturing and expediency to global access. In addition, biopharmaceutical companies have continued to expand their frontiers to attain a global reach, with presence in many  regions and countries, and therefore exposed to myriad, and sometimes diverse, regulatory requirements and operating standards. The challenges of globalization in a heterogeneous world
with an evolving regulatory landscape and expectations of multiple stakeholders have increased
the complexity, unpredictability and intensity of the biopharmaceutical product development
and registration process. These challenges likewise reinforce the crucial role of the regulatory
team and underscore the need for enhanced global regulatory function with regulatory professionals who are strong leaders, business partners and strategic contributors.
The scope of the regulatory affairs group function spans the entire spectrum of product development, manufacturing, registration, post-marketing activities and lifecycle optimization. This span of involvement and responsibility is sometimes referred to as bench to bedside and beyond,  from cradle to grave, from inception through lifecycle optimization, from laboratory to launch, etc. The regulatory team and professional hold a unique position of importance with impressive diversity in function and significant breadth and depth of responsibility.
This chapter focuses on the role of the global regulatory affairs team in the biopharmaceutical industry and draws upon its crucial and unique position to highlight the strong leadership function, critical business partner, strategic contributor and honest broker for some of industry’s most important decisions.

Global regulatory affairs organization

Functional units

Global regulatory affairs groups in industry provide worldwide, strategic leadership in and
comprehensive direction of the government regulatory requirements for product introduction
and commercialization, utilizing regulatory knowledge to ensure compliance and regulatory
intelligence and create opportunities in a highly-regulated environment.
The global regulatory affairs group has strategic and operational functions, both requiring
precision and flawless execution, as a company’s success relies on, in part, the guidance and rigorous assessment provided by the regulatory affairs team. The key product development, manufacturing and registration milestones (such as Investigational New Drug filing, end of phase II, phase III, New Drug Application submissions, regulatory agency review timetable and product approvals), which are the corporate valuation and decision points, are based on critical regulatory interface goalposts and milestones. This underscores the importance of strategic regulatory contribution to the industry. In order to be valuable contributors and critical business partners, the global regulatory affairs organization must develop mechanisms and establish systems to ensure it has the right people and talent, proficient processes, prolific productivity, phenomenal performance and precision in planning.

What comprises global regulatory affairs

The global regulatory affairs groups within different pharmaceutical companies have different organizational structures and functional reporting. However, most are comprised of fundamental units that house the various specialists or experts within the regulatory function.
The global regulatory affairs group has traditionally resided within the research and development group of biopharmaceutical companies, but that, too, is evolving. Many companies
have recognized the central role of the group to all technical functions—development, manufacturing and marketing—and therefore are aligning the group’s reporting to a central and
neutral executive. In some companies, the global regulatory group reports to the Chief Execu tive Officer—a statement of the importance of this function. The typical functional units are
represented and may vary from company to company; therefore, this chapter will address the
typical functional units of importance.
The key functions of the global regulatory affairs group include:
● Leading and providing strategic regulatory guidance and delivering the global regulatory strategy for product development, manufacturing and registration
● Building and maintaining a credible relationship with regulatory authorities with effective written and verbal communication
● Establishing an efficient repository and archive for correspondence compliant with
regulatory standards for audits
● Providing high-quality, complete user/reviewer-friendly documents electronically
transmissible and reproducible
● Developing and maintaining product information and label
● Providing regulatory intelligence, from paying attention to regulatory environment and
changing landscape, to participating in external industry partnerships and developing
policy with regulatory agencies
● Ensuring published information and promotional/advertising material meet regulatory
requirements
● Ensuring functional units comply with regulatory requirements and good regulatory
practices.

Role of global regulatory affairs

The enhanced role of the regulatory affairs professional
The past 10 years have brought both an exciting evolution and challenging times for the biopharmaceutical industry with an increasingly complex and demanding regulatory environment, creating opportunities for both the regulatory profession and the pharmaceutical industry. This evolution in the regulatory function has been driven largely by the expanding scope
and global reach of the industry, global regulatory environment and intelligence, scientific
breakthrough and innovation, cutting-edge technologies including e-submissions, complexity of disease area targets for development and multiple stakeholder demand for rigorous and
competitive product differentiation. These have heightened the need for highly-skilled regulatory professionals with area specialization and broad leadership capability.
In this changing and enhanced regulatory role, the regulatory team is expected to provide
regulatory leadership and excellence in a changing environment. It requires the regulatory team
to have at the core of its focus the end deliverable goal, which is expeditious patient access to highquality, well-differentiated products.
This end goal drives the need for the regulatory team to:
● Deliver innovative, breakthrough regulatory strategies for product development and
registration.
● Become more anticipatory of the company success imperatives.
● Be proactive and forward thinking, provide timely, comprehensive and robust global
regulatory guidance.
● Understand the biopharmaceutical environment and regulatory actions on precedents
and utilize such regulatory intelligence.
● Forge new standards to deliver more predictable outcomes.
● Increase focus on building and strengthening relationship with regulatory authorities to
provide timely expert input into product development, manufacturing and registration.
(ref: http://www.centerwatch.com/pdfs/s11701_ch2_sample.pdf)