[Published by the Office for Protection from Research Risks (now the Office of Human Research Protections), 1993]


 

Institutional Review Board Guidebook

* CHAPTER V *
BIOMEDICAL AND BEHAVIORAL RESEARCH:
AN OVERVIEW


 

A. Introduction

 

Biomedical Research

Behavioral Research                                           

Fieldwork                               

Social Policy Experimentation

 

B. Drug Trials

Introduction

Definitions

Overview

IRB Considerations

Points to Consider

Applicable Laws and Regulations

 

C. Vaccine Trials

Introduction

Definitions

IRB Considerations

Points to Consider

Applicable Laws and Regulations

 

D. Medical Devices

Introduction

Definitions

Overview

IRB Considerations

Points to Consider

Applicable Law and Regulations

 

E. Use of Radioactive Materials and X-Rays

Introduction

Definitions

Overview

IRB Considerations

Points to Consider

Applicable Laws and Regulations

 

F. AIDS/HIV-Related Research

Introduction

IRB Considerations

Clinical Trials of HIV-Related Therapies

Vaccines             

Informing Subjects of Their HIV Serostatus

Behavioral Research

Vulnerability of Subjects

Availability of Drugs and Other Therapeutic Agents

Points to Consider

 

G. Transplants

Introduction

Overview

IRB Considerations

Points to Consider

Applicable Laws and Regulations

 

H. Human Genetic Research

Introduction

Definitions

IRB Considerations

Pedigree Studies

Identifying and Deciphering Genes

Research On Genetic Testing

Gene Therapy Research

Points to Consider

Applicable Laws and Regulations

 

I. Alcohol and Drug Research

Introduction

Definitions

IRB Considerations

Risk/Benefit.

Incentives for Participation

Informed Consent

Subject Selection       

Privacy and Confidentiality

IRB Membership

Points to Consider

Applicable Laws and Regulations

 

Suggestions for Further Reading

 


 

A. INTRODUCTION

Most of the research reviewed by IRBs falls within the broad categories of biomedical or behavioral research. IRBs should be sensitive to specific aspects of biomedical and behavioral research in their review of protocols.

Biomedical research includes both studies designed primarily to increase the scientific base of information about normal or abnormal physiology and development and studies primarily intended to evaluate the safety, effectiveness or usefulness of a medical product, procedure, or intervention. The terms "behavioral research" or "the behavioral sciences" may be used to refer either to studies of the behavior of individuals, or to studies of the behavior of aggregates such as groups, organizations, or societies. The broad objective of the behavioral and social sciences is similar to that of the biomedical sciences: to establish a body of demonstrable, replicable facts and theory that contributes to knowledge and to the amelioration of human problems.

It is neither possible nor necessary to draw a clean line between biomedical and behavioral research. Some biomedical research pertains to behavior (e.g., in psychiatry, neurology, or epidemiology), and many of the methods used in behavioral research, such as observation and the questioning of subjects, are also used in biomedical research. Research may be designed to evaluate the behavioral changes that result from a biomedical intervention (e.g., lessening of depression after taking a particular medication or changes in psychiatric disorders following hemodialysis) or to examine physiological responses to behavioral interventions (e.g., lowering of blood pressure through biofeedback or weight loss through hypnosis). Some studies involve functions that are not easily defined as either behavioral or physiological (e.g., sleep, exercise, or diet). Thus, although it is sometimes useful to refer to biomedical or behavioral and social research as if they involve distinct activities, there is considerable overlap among the three areas. (While the use of such terms as "behavioral and social research" may imply that social research is distinct from behavioral research, this distinction generally has little utility for the work of IRBs and is not applied here.) The questions that are of concern to IRBs stem not from the label attached to the research but from the nature of the interventions and the characteristics of the subjects in any given study. It is for this reason that institutions and federal agencies are concerned that IRB members be knowledgeable about the various types of research reviewed by that IRB.

Return to Top of Page

BIOMEDICAL RESEARCH

Biomedical research employs many methods and research designs. Studies designed to evaluate the safety, effectiveness, or usefulness of an intervention include research on therapies (e.g., drugs, diet, exercise, surgical interventions, or medical devices), diagnostic procedures (e.g., CAT scans or prenatal diagnosis through amniocentesis, chorionic villi testing, and fetoscopy), and preventive measures (e.g., vaccines, diet, or fluoridated toothpaste). Research on normal human functioning and development can include studies of the human body while exercising, fasting, feeding, sleeping, or learning, or responding to such things as stress or sensory stimulation. Some studies compare the functioning of a particular physiological system at different stages of development (e.g., infancy, childhood, adolescence, adulthood, or old age). Others are directed at defining normal childhood development so that deviations from normal can be identified. Sometimes research, particularly records research, is used to develop and refine hypotheses. Research on specific disease processes is often needed before improved methods of prevention, diagnoses, and treatment can be developed (e.g., research on the biochemical changes associated with AIDS or schizophrenia, or the neurological changes associated with senile dementia of the Alzheimer type). Research on the human genome and genetic markers is expected to create new avenues for understanding disease processes and their eventual control.

Subjects of some biomedical studies engage in ordinary tasks (e.g., exercise, learn a series of words, or respond to various sensory stimuli) while measurements of physiological and bodily functions are made. Although many procedures used in biomedical research are similar to those used in routine physical examinations, at times more invasive procedures (e.g., "spinal taps," skin or muscle biopsies, or X-rays used in conjunction with contrast dyes) must be used if a desired measurement is to be made. Although research designed to generate information about normal physiology or a disease process is not concerned with evaluating a medical intervention, it may still require the use of invasive procedures. When the research deals with subjects whose condition is not normal, the research can have either therapeutic or nontherapeutic purposes.

Other biomedical studies do not involve human subjects or are exempt from the human subjects regulations, and, therefore, do not require IRB review. This category includes research with animals and research on preexisting samples of materials (tissue, blood, or urine) collected for other purposes, where the information is recorded by the investigator in such a manner that subjects cannot be identified, directly or through identifiers linked to the subjects [Federal Policy §___.101(b)(4)]. It also includes research based on records, when the data are recorded in such a manner that the individuals to whom the records pertain cannot be identified, either directly or through identifiers linked to them [Federal Policy §___.101(b)(4)]. [See Guidebook Chapter 4, "Considerations of Research Design."]

Some biomedical studies, particularly those conducted to evaluate new therapies or treatments, use such rigorous experimental methods as random assignment to treatment and control groups. Other studies, such as those directed at establishing the normal range of some element in the blood, may involve no experimental intervention and no assignment of subjects to groups. [See Guidebook Chapter 4, "Considerations of Research Design."]

The fact that much biomedical research is conducted for the purpose of evaluating new therapies or treatments leads to two problems for IRBs. The first is to some degree a problem of IRB jurisdiction; the second is a problem of risk/benefit assessment. The distinction between research and treatment can become blurred in patient care settings, as well as in some educational and training settings. This distinction raises questions of IRB jurisdiction over the research: Is the proposed activity one that requires IRB review (pursuant either to federal regulations or institutional policy)? A discussion of this issue appears in the Guidebook in Chapter 1, Section A, "Jurisdiction of the Institutional Review Board."

The second distinction between research and therapies that may pose a problem for IRBs concerns risk/benefit assessments in research on therapies. Often, the risks of a study may seem justified by a therapy provided as part of the study. IRBs should determine, however, whether the anticipated therapeutic benefits would be available to persons who are not participating in a study that presents additional risks. As is discussed in the Guidebook Section on risk/benefit analysis [Chapter 3, Section A], such benefits should not be used to justify risks presented by the research.

The IRB's general responsibilities in reviewing biomedical research are discussed in other chapters of the Guidebook. [See Chapter 3, "Basic IRB Review," and Chapter 4, "Considerations of Research Design."] Special concerns arising in the conduct of certain types of biomedical research are discussed in the following Sections of this chapter on "Drug Trials," "Vaccine Trials," "Medical Devices," "Use of Radioactive Materials and X-Rays," "HIV-Related Research," "Transplants," "Human Genetic Research," and "Alcohol and Drug Research." The additional IRB responsibilities that arise when the subjects of biomedical research are other than healthy, normal adults are set forth in Chapter 6, "Special Classes of Subjects."

Return to Top of Page

 

 

BEHAVIORAL RESEARCH

The scope and diversity of research areas in the behavioral and social sciences is quite broad. Some research is readily applicable to human affairs; other studies may broaden understanding without any apparent or immediate application. Some research is designed to test hypotheses derived from theory; other research is primarily descriptive. Still other research may be directed at evaluating an intervention or social program.

Theories and methods vary both across and within disciplines; the same problems may be approached by researchers trained in different disciplines. For example, some research psychologists work in laboratories studying the neurology, anatomy, and physiology that underlies perception, learning, instinctual behavior, and emotional responses. Other psychologists may perform survey research, observational studies, or small group experiments that differ little from work done by some sociologists. Within anthropology, physical anthropology overlaps with paleontology, anatomy, and genetics, while the social or cultural anthropologist studies the organization, institutions, and belief and value systems of societies or groups of people.

Behavioral research involving human subjects generates data by means of questionnaires, observation, studies of existing records, and experimental designs involving exposure to some type of stimulus or intervention. Many variations of these four basic methods are used. Questions may be asked in person, over the telephone, or by means of a questionnaire. Observation may or may not be covert, and the observer may or may not be a participant in the activity being studied. Records studied in research may be public (e.g., vital statistics, motor vehicle registrations, or court records) or non-public and sensitive (e.g., medical or educational records in which the subjects are identified). Experimental studies may be conducted in public places, in private settings (e.g., a clinic or therapist's office), or in laboratories. Interventions in such studies range from the innocuous, such as varying the package design of commercial products, to the potentially significant, such as varying behavior modification techniques in studying the treatment of alcoholism. Not all behavioral research involves human subjects. Studies of human migration are often undertaken using anonymous U.S. Census data, and much research in behavioral psychology is done with animals. In addition, many categories of behavioral research that do involve human subjects are exempt from the federal regulations for protection of human subjects. [See Federal Policy §___.101.] This exemption does not imply that investigators have no ethical responsibilities to subjects in such research; it means only that IRB review and approval of the research is not required by federal regulations.

Most behavioral research involves no physical intervention and no physical risk. However, some studies do present a risk of social harm (e.g., harm to a subject's reputation, which is sometimes a danger if confidentiality is not maintained) or psychological harm, which may occur if the research involves deception or provides subjects with unwelcome and disturbing information about themselves. When deception is involved, the IRB needs to be satisfied that the deception is necessary and that, when appropriate, the subjects will be debriefed. (Debriefing may be inappropriate, for example, when the debriefing itself may present an unreasonable risk of harm without a counterveiling benefit.) The IRB should also make sure that the proposed subject population is suitable. [See Guidebook Chapter 3, Section A, "Risk/Benefit Analysis."]

Some studies involve the possibility of a moral wrong, which is what some commentators have labeled the ethical problems posed by deception of subjects or invasions of their privacy. Although some psychologists have overemphasized the value and necessity of using deception, deception or incomplete disclosure may be the only scientifically valid approach for certain research. An example of such research would be a study designed to determine the effect of group pressure (i.e., responses of others) on a subject's estimate of the length of a series of lines. In some groups, pseudo-subjects would be told in advance to give incorrect answers to questions about the length of the lines to determine the effect of such misinformation on the real subjects' responses. Obviously, if the subjects were told all about the research design and its purpose in advance, it would not be possible to do the research. IRBs need to determine whether any deception or invasion of privacy involved in a research protocol is justified.

Some social and behavioral researchers are concerned that IRB judgments at times seem to be influenced more by the subject matter of the study than by concerns about informed consent or risks to subjects. Researchers cite examples of studies that involve minimal risk and pose no consent questions, but that encounter difficult with some IRBs, particularly IRBs in medical settings. Some researchers believe that IRBs are more likely to object to research on the behavior or values of the powerful (e.g., physicians, professors, or managers) than to research using similar methods but on subjects of lower status (e.g., patients, students, or workers). Other researchers believe that IRBs sometimes perceive research on controversial topics, such as deviant sexual behavior or fraud in science, as presenting ethical problems because of the nature of the activity being studied, rather than because of research methods, risks, or the rights of subjects. Still others complain of a less specific prejudice against social and behavioral research on the grounds that it is "soft" or concerned with trivial questions.

Some behavioral research involves human subjects in studies of heredity and human behavior, genetics, race and IQ, psychobiology, or sociobiology. Vigorous ethical debates about these studies arise out of the fear that scientific data may be used to justify social stratification and prejudice, or that certain groups will appear to be genetically inferior. The possible use — or misuse — of research findings, however, should not be a matter for IRB review, despite the importance of this question.

The incidence of such problems may well have decreased because the regulations exempt much social research and provide additional flexibility regarding informed consent. IRBs should resist placing restrictions on research because of its subject matter; IRBs should instead be concerned about research methods and the rights and welfare of research subjects. IRBs must differentiate disapproving a research proposal because of qualms about the subject being explored or its possible findings, such as genetic differences in intelligence, from disapproving research involving the performance of illegal or unethical acts. The former raises serious issues of academic freedom; the latter is quite different and appropriate. Whatever the propriety of institutional administrators prohibiting research to protect the institutions from being associated with controversial or sensitive subjects, it is generally agreed that this is not an appropriate concern for an IRB, whose function is to protect human subjects.

Return to Top of Page

FIELDWORK

Fieldwork, or ethnographic research, involves observation of and interaction with the persons or group being studied in the group's own environment, often for long periods of time. Since fieldwork is a research process that gains shape and substance as the study progresses, it is difficult, if not impossible, to specify detailed contents and objectives in a protocol.

After gaining access to the fieldwork setting, the ongoing demands of scientifically and morally sound research involve gaining the approval and trust of the persons being studied. These processes, as well as the research itself, involve complex, continuing interactions between researcher and hosts that cannot be reduced to an informed consent form. Thus, while the idea of consent is not inapplicable in fieldwork, IRBs and researchers need to adapt prevailing notions of acceptable protocols and consent procedures to the realities of fieldwork. IRBs should keep in mind the possibility of granting a waiver of informed consent.

Return to Top of Page

SOCIAL POLICY EXPERIMENTATION

Social policy experimentation involves interventions in social or economic systems for use in planning public policy. Such experimentation often involves studying the costs and benefits of alternative ways of providing health, educational, or welfare services at national, state, or local levels. Some of this research may be exempt from IRB review under §___.101(b)(5) of the Federal Policy. That section exempts research and demonstration projects that are conducted by or subject to the approval of department or agency heads, and that are designed to study, evaluate, or otherwise examine: (1) public benefit or service programs; (2) procedures for obtaining benefits or services under those programs; (3) possible changes in or alternatives to those programs or procedures; or (4) possible changes in methods or levels of payment for benefits or services under those programs.

Return to Top of Page

B. DRUG TRIALS

INTRODUCTION

Drug trials provide the transition from promising basic or laboratory research to helpful therapeutic or diagnostic procedures for patients. New drugs that offer the hope of some beneficial response in afflicted patients are first tested in animal models. But animal trials do not necessarily demonstrate what the physiological, pharmacological, or toxicological effects of a new drug will be in human beings. Only by careful testing in human subjects can the safety and effectiveness of a new drug be evaluated. The Food and Drug Administration (FDA) is responsible for monitoring the testing of new drugs in humans, for determining whether a new drug can be marketed, and for observing drugs after marketing to be sure that they are safe, effective, and properly labeled [21 CFR 312 and 21 CFR 314].

See also Guidebook Chapter 4, Section H, "Clinical Trials," and Section J, "Assignment of Subjects to Experimental and Control Groups."

DEFINITIONS

Clinical Trial: A controlled study involving human subjects, designed to evaluate prospectively the safety and effectiveness of new drugs or devices or of behavioral interventions.

Drug: Any chemical compound that may be used on or administered to humans as an aid in the diagnosis, treatment, cure, mitigation, or prevention of disease or other abnormal conditions.

Investigational New Drug or Device: A drug or device permitted by FDA to be tested in humans, but not yet determined to be safe and effective for a particular use in the general population, and not yet licensed for marketing.

Investigator: In clinical trials, an individual who actually conducts an investigation [21 CFR 312.3]. Any interventions (e.g., drugs) involved in the study are administered to subjects under the immediate direction of the investigator. (See also: Principal Investigator.)

Phase 1, 2, 3, 4 Drug Trials: Different stages of testing drugs in human, from first application in humans (Phase 1) through limited and broad clinical tests (Phase 3), to postmarketing studies (Phase 4).

Phase 1 Drug Trial: Phase 1 trials include the initial introduction of an investigational new drug into humans. These studies are typically conducted with healthy volunteers; sometimes, where the drug is intended for use in patients with a particular disease, however, such patients may participate as subjects. Phase 1 trials are designed to determine the metabolic and pharmacological actions of the drug in humans, the side effects associated with increasing doses (to establish a safe dose range), and, if possible, to gain early evidence of effectiveness; they are typically closely monitored. The ultimate goal of Phase 1 trials is to obtain sufficient information about the drug's pharmacokinetics and pharmacological effects to permit the design of well-controlled, sufficiently valid Phase 2 studies. Other examples of Phase 1 studies include studies of drug metabolism, structure-activity relationships, and mechanisms of actions in humans, as well as studies in which investigational drugs are used as research tools to explore biological phenomena or disease processes. The total number of subjects involved in Phase 1 investigations is generally in the range of 20-80.

Phase 2 Drug Trial: Phase 2 trials include controlled clinical studies conducted to evaluate the drug's effectiveness for a particular indication in patients with the disease or condition under study, and to determine the common short-term side effects and risks associated with the drug. These studies are typically well-controlled, closely monitored, and conducted with a relatively small number of patients, usually involving no more than several hundred subjects.

Phase 3 Drug Trial: Phase 3 trials involve the administration of a new drug to a larger number of patients in different clinical settings to determine its safety, effectiveness, and appropriate dosage. They are performed after preliminary evidence of effectiveness has been obtained, and are intended to gather necessary additional information about effectiveness and safety for evaluating the overall benefit-risk relationship of the drug, and to provide an adequate basis for physician labeling. In Phase 3 studies, the drug is used the way it would be administered when marketed. When these studies are completed and the sponsor believes that the drug is safe and effective under specific conditions, the sponsor applies to FDA for approval to market the drug. Phase 3 trials usually involve several hundred to several thousand patient-subjects.

Phase 4 Drug Trial: Concurrent with marketing approval, FDA may seek agreement from the sponsor to conduct certain postmarketing (Phase 4) studies to delineate additional information about the drug's risks, benefits, and optimal use. These studies could include, but would not be limited to, studying different doses or schedules of administration than were used in Phase 2 studies, use of the drug in other patient populations or other stages of the disease, or use of the drug over a longer period of time [21 CFR §312.85].

Principal Investigator: The scientist or scholar with primary responsibility for the design and conduct of a research project. (See also: Investigator.)

Sponsor: A person or entity that initiates a clinical investigation of a drug — usually the drug manufacturer or research institution that developed the drug. The sponsor does not actually conduct the investigation but rather distributes the new drug to investigators and physicians for clinical trials. The drug is administered to subjects under the immediate direction of an investigator who is not also a sponsor. A clinical investigator may, however, serve as a sponsor-investigator. The sponsor assumes responsibility for investigating the new drug, including responsibility for compliance with applicable laws and regulations. The sponsor, for example, is responsible for obtaining FDA approval to conduct a trial and for reporting the results of the trial to the FDA.

Sponsor-Investigator: An individual who both initiates and actually conducts, alone or with others, a clinical investigation. Corporations, agencies or other institutions do not qualify as sponsor-investigators.

OVERVIEW

Once a chemical (drug) is identified as having a potential effect on a disease state, it is subjected to testing in animals. Initial animal tests are designed to see whether the chemical has any desired drug effects, what dosage levels are poisonous, what the safe dosage range might be in humans, and whether there is a reason to test the chemical in humans. Additional animal tests may be required as human tests progress. If initial animal tests indicate that the drug can be safely tested in humans and that the chemical may be therapeutically useful, the drug sponsor will submit an Investigational New Drug Application (IND) to the FDA. In the IND, the sponsor must describe the complete composition of the drug, its source, and how it is made. In addition, the sponsor must submit the results of all animal studies that support the drug's potential usefulness in humans and that define its toxicity in animals. The data should indicate that no human subject will be exposed to an unreasonable risk. The IND must also include a protocol describing the plan for testing in humans. To permit the FDA to review the materials and make sure subjects will not be exposed to unreasonable risks, the sponsor may not begin clinical tests for 30 days after submitting the IND. At the end of that period, the sponsor may begin the proposed clinical trial unless the FDA has asked for a delay because of a potential safety problem involving use of the drug.

Clinical trials are conducted by clinical investigators (usually physicians) who have entered into an agreement with a sponsor to conduct the study. All physicians administering an investigational drug agree to conditions regarding the conduct of the study outlined by FDA regulations. Clinical investigators agree to these conditions by signing an FDA form that certifies that the investigator has obtained IRB review and approval prior to conducting the study.

Investigational new drugs may be available outside of a clinical trial, through a treatment protocol, to patients with life-threatening or other serious diseases for which no satisfactory alternative drug or other therapy exists. Established by the FDA in 1987, the Treatment Investigational New Drug exemption (Treatment IND) is a treatment protocol that is added to an existing IND. The Treatment IND allows physicians to treat qualifying patients according to the protocol. Treatment INDs are discussed in greater detail in Guidebook Chapter 2, Section B, "Food and Drug Administration Regulations and Policies."

For further information concerning human subjects research to which FDA regulations apply, contact:

Mr. Richard M. Klein
Office of Health Affairs (HFY-20)
Food and Drug Administration
5600 Fishers Lane
Rockville, MD 20857
Tel: (301) 443-1382

IRB CONSIDERATIONS

In reviewing proposed drug research, IRBs must first consider whether the protocol is scientifically sound. Since this decision is not the IRB's primary concern, however, an IRB may rely on the FDA, institutions, scientific review committees, funding agencies (e.g., NIH), or others for this determination. [See the Introduction to Guidebook Chapter 4, "Considerations of Research Design" for a discussion of this question.] Evaluating the risks and benefits of drug trials requires IRBs to consider many aspects of the study design, paying special attention to the study population, the trial phase, and mechanisms for data analysis and surveillance. Risk/benefit analysis and review of the procedure for obtaining informed consent must be performed in all IRB reviews. [See Guidebook Chapter 3, Section A, "Risk/Benefit Analysis," and Chapter 3, Section B, "Informed Consent."] In addition, subjects participating in studies involving investigational drugs must be told that the FDA may have access to their medical records as they pertain to the study.

The obligation of IRBs and investigators to assure that subjects understand the purposes, methods, and possible hazards of the research is more difficult to fulfill when prospective subjects are seriously ill and in need of therapy. The consent process may require additional efforts and attention for research involving particularly vulnerable subjects such as the seriously ill. [See Chapter 6, Section G, "Terminally Ill Patients."]

Phase 1 trials are historically safest because they usually involve administering a single dose to healthy volunteers. However, Phase 1 trials may pose the highest level of unknown risk because they involve the drug's first administration to humans. (With highly toxic drugs such as cancer chemotherapies, Phase 1 trials are usually conducted with cancer patients as subjects.) Insofar as possible, risks should be identified from previous laboratory experiments and animal trials. The FDA, which reviews Phase 1 trials submitted in the initial IND application, may have valuable information and recommendations on particular protocols.

Subjects in Phase 2 trials are usually patients with the condition that the new drug is intended to detect or treat. IRBs should recognize that although Phase 2 testing is preceded by earlier clinical trials, the physiological responses of healthy volunteers to a therapeutic drug may not be reliable indicators of how safe the drug is for persons who are ill, taking other medication, or have immunodeficiencies. Since the primary purpose of a Phase 2 trial is to test the drug's effectiveness in achieving its purpose, the responses of subjects receiving the drug are usually compared with those of subjects who are not receiving the drug (control subjects). Whether control subjects receive some existing therapy or a placebo is a research design issue with serious ethical implications. Where an alternate safe and effective drug is available for a serious condition being studied, it should generally be given to the control subjects; however, existing therapies may be inadequate because they are of limited effectiveness against the disease, they have relatively high levels of toxicity, or because they are inconvenient to administer. When determining the acceptability of a proposed research design, IRBs must examine the risks and effectiveness of existing therapies, as well as the risks associated with providing no therapy (or a placebo). [See Chapter 4, "Considerations of Research Design."]

While most drug trials involve agents that the FDA has not yet approved for marketing, some drugs may be the subject of further testing concurrent with or following FDA approval. Post-marketing investigations, also called Phase 4 trials, are conducted to develop further information about the article's safety or effectiveness. Such studies might, for example, seek to establish the safety or effectiveness of using the drug for a new indication, with a new dosage level or a new route of administration [21 CFR §312.85].

Phase 4 studies should be distinguished from use of a marketed product by a physician for an indication not in the approved labeling as part of the "practice of medicine." Investigational use of a marketed product differs from such uses by physicians in that the principal intent of the investigational use of a test article is to develop information about its safety or efficacy; the submission of an IND or IDE may therefore be required. The criteria for submission of an IND or IDE for investigational use of a marketed product is described in the FDA's IRB Information Sheet entitled, "Investigational Use of Marketed Products," (1989, pp. 70-71).

Throughout drug trials, the distinction between therapy and research must be maintained. A physician who participates in research by administering a new drug to consenting patients must ensure that the patients understand and remember that the drug is experimental, and that its benefits for the condition under study are unproven. Furthermore, whereas the principal investigator's primary allegiance is to the protocol, the physician's allegiance is to the patient. Where an individual is both an investigator and the subject's treating physician, these two allegiances may conflict. The subject must recognize that the person with whom he or she is dealing may have such conflicting interests. The IRB should be aware of the need to inform the patient of the potential conflict.

If the trial is to collect accurate and timely data concerning the drug's safety and effectiveness, procedures for identifying positive and negative responses to the drug should be in place, and all participating physicians should be well integrated into a reporting system. The principal investigator is responsible for keeping all subjects informed of material changes in the design and conduct of the research, and must communicate new information that might affect their willingness to continue as subjects [Federal Policy §___.116]. The IRB may assist the investigator in deciding when information from accumulating data should be disclosed to participating or prospective subjects. The disclosure of information gained during the conduct of the trial is especially important with patients entering a study when it is nearing completion.

As part of their determination of the appropriate methods for conducting continuing reviews of ongoing studies, IRBs should be aware of the arrangements made for monitoring the study results. In FDA-regulated clinical investigations, arrangements for data monitoring are the sponsor's responsibility. The sponsor may designate an independent person or group (often called a data and safety monitoring board) to assume this responsibility. An IRB may function in such a capacity; however, most IRBs do not have the necessary expertise. Independent monitoring is most appropriate when the study is double-masked (i.e., neither the subjects nor the investigators know which drug a subject is receiving) or if the trial is multicentered. Ongoing monitoring of drug trials includes review of data on therapeutic effects, side effects and the effects of any changes in the study design. [See also Guidebook Chapter 3, Section E, "Monitoring and Observation."] Sponsors must notify the FDA and all participating investigators of any adverse experiences associated with the use of an investigational new drug that is both serious and unexpected [21 CFR 312.32].

Occasionally, hazards are discovered after a trial is concluded. If the drug has since been marketed, the FDA and the drug manufacturer are usually responsible for notifying users and physicians.

POINTS TO CONSIDER

1. Is the proposed research scientifically sound?

2. Has sufficient information been obtained from the literature, experimental and animal studies, and the FDA to define, as far as possible, the potential risks of and the precise need for studies involving human subjects?

3. Does the principal investigator have the appropriate qualifications, experience, and facilities to ensure that all aspects of the trial and follow-up will be conducted rigorously and with due regard for the safety and well-being of the subjects?

4. Have appropriate measures been adopted to ensure that subjects understand the objectives and consequences, particularly the risks, of their participation?

5. Are sufficient safeguards provided to ensure the confidentiality of data generated during research?

6. Are adequate procedures provided for the ongoing surveillance of the drug's effectiveness and safety, and for notifying subjects and physicians of significant risks?

7. Has appropriate FDA review and clearance been obtained?

APPLICABLE LAWS AND REGULATIONS

Federal Policy for the protection of human subjects

21 CFR 50 [FDA: Informed consent]
21 CFR 56 [FDA: IRB review and approval]
21 CFR 312 [FDA: Investigational new drugs]
21 CFR 52 [FDA: Sponsor and monitor (proposed)]
21 CFR 54 [FDA: Clinical investigators (proposed)]

Return to Top of Page

 

 

 

C. VACCINE TRIALS

INTRODUCTION

Vaccines are used to prevent infectious diseases. Successful vaccine trials have resulted in the development of safe and effective vaccines for polio, measles, rubella, hepatitis B, pneumococcal pneumonia, and other serious diseases. Currently, vaccines are being evaluated to prevent infectious diseases such as AIDS (or transmission of HIV), malaria, tuberculosis, trachoma, cytomegalovirus, herpes simplex, and influenza. Vaccines must undergo clinical testing prior to approval and licensure by the FDA. The regulations governing the conduct of clinical trials on investigational vaccines are the same as those governing the conduct of investigational new drug research [see Guidebook Chapter 5, Section B, "Drug Trials"]; however, the risks and benefits associated with vaccine trials may differ from those of drug trials.

A vaccine is a biologic; its use in trials involving human subjects is similar to the use of any drug. Vaccines do, however, differ from therapeutic drugs in two important ways. As used here, they are not designed to diagnose or cure disease in afflicted individuals; their purpose is to prevent a particular disease in healthy human beings. Vaccines are also used to protect people with a high statistical risk for contracting a particular disease or for suffering especially serious consequences from a disease. Vaccines trigger the body's normal immune response, producing antibodies that protect against future infection. Some vaccines (e.g., those containing active microorganisms or live-attenuated vaccines) have a small but real disease-producing capacity. Thus, one rare risk of a new vaccine is the possibility of infecting a healthy subject with the very disease researchers are seeking to prevent. More often, however, subjects involved in vaccine trials temporarily suffer from some of the symptoms and effects of the disease (e.g., polio, German measles) as they acquire immunity.

DEFINITIONS

  • Biologic: Any therapeutic serum, toxin, anti-toxin, or analogous microbial product applicable to the prevention, treatment, or cure of diseases or injuries.
  • Purity: The relative absence of extraneous matter in a vaccine that may or may not be harmful to the recipient or deleterious to the product.
  • Sterility: The absence of viable contaminating microorganisms; aseptic state.
  • Vaccine: A biologic product generally made from an infectious agent or its components — a virus, bacterium or other microorganism — that is killed (inactive) or live-attenuated (active, although weakened). Vaccines may also be biochemically synthesized or made through recombinant DNA techniques.

IRB CONSIDERATIONS

The development of vaccines is of considerable benefit to society, especially in the case of devastating or highly infectious diseases. The direct benefit to the individual subject receiving a new vaccine is the possibility of immunity (i.e., protection against future disease). The benefits of such immunity will vary depending on: (1) the severity of the disease to be avoided; (2) the likelihood that the subject will be exposed to the infectious disease; and (3) in the case of certain diseases, the likelihood that the subject would suffer adverse consequences should he or she contract the disease. Some populations will be at greater risk of contracting an infectious disease than others, either because they are more likely to be exposed to the disease or because they have an increased susceptibility to it. Among those who contract an infectious disease, there may be some sub-groups that are particularly vulnerable to adverse consequences (e.g., children, persons of advanced age, or persons suffering from other illnesses).

For most diseases, participation in vaccine trials carries the generally small risk of contracting the disease. [In some vaccine trials (e.g., HIV) there is no such risk. In the case of HIV vaccine research, the lack of risk is due to the manner in which the vaccine is derived.] The risks of participating in a vaccine trial also include adverse effects unrelated to the disease in question (e.g., slight fever, headache, muscle soreness, or muscle aches). Such side effects are usually short-lived, tolerable, and not life-threatening. Again, the degree of risk associated with participating in a vaccine trial varies depending on the subjects' vulnerability to the adverse side effects of the vaccine. Some subjects may have an allergic or anaphylactic (i.e., a decrease rather than an increase in immunity) reaction to the vaccine. Anaphylactic reactions to vaccines cause the recipient to be hypersusceptible to the disease. Such reactions are generally unpredictable, and may be serious or potentially life-threatening.

The IRB should be aware of other risks associated with vaccine trials, including the possibility that vaccines produced synthetically or using recombinant DNA techniques may present risks as yet unknown, that groups often most likely to benefit from receiving a vaccine are often the most vulnerable to coercion (e.g., institutionalized persons or children), and that subjects in control groups may erroneously assume that they have been immunized.

When determining whether the risks are reasonable in relation to the benefits, IRBs should consider the severity of the disease, the risk of contracting the disease, and any special vulnerability of the subject population to the potential adverse effects of the vaccine. The most difficult cases are those in which the subjects most likely to benefit from participating in the vaccine trial are also the subjects at the greatest risk of suffering from the vaccine's potential adverse effects.

Some of the risks inherent in vaccine trials can be minimized. Before a vaccine is approved for testing with human subjects, IRBs should receive satisfactory evidence that animal trials and laboratory tests have, to the extent possible, demonstrated its safety. Since the sponsor must submit such information to the FDA as part of its investigational new drug application (IND), IRBs can readily obtain evidence of safety as well.

Mechanisms for protecting human subjects from some risks can be built into the vaccine study design. For example, with careful screening, investigators can avoid enrolling persons who may be susceptible to certain adverse reactions. Furthermore, trials can be designed to involve subjects who are most likely to be exposed to the infectious agent and who stand to benefit most from the protection afforded by the vaccine. Selecting subjects in this way avoids exposing those who may not be in need of its protective benefits to the risks of the vaccine. In many situations, however, Phase 1 trials should be designed to evaluate low risk subjects. For example, an effective hepatitis B vaccine already exists. It would therefore be appropriate to determine that an investigational vaccine for hepatitis B is immunogenic in humans prior to use in high risk subjects.

Vaccine trials require careful monitoring of human subjects for both immune status and adverse reactions. The monitoring reflects the dual goals of any trial to determine both the effectiveness and the safety of the investigational substance or device. Although subjects in vaccine trials should be advised beforehand of known or anticipated side effects, rare or unknown reactions may occur. FDA regulations require that subjects be provided with written instructions about whom to contact in the event of serious adverse reactions or research-related injury.

IRBs should also be aware that large-scale field trials of a vaccine may involve many thousands of subjects, making monitoring difficult. The IRB should make sure that the sponsor has made provisions for monitoring the progress of the research, the immune status of participants, and side effects reported. Maintaining careful records is important both for monitoring the safety and effectiveness of the vaccine and for locating subjects for follow-up. If a vaccine either does not immunize the subject or does so for too limited a time, subjects may erroneously assume they are protected and fail to seek necessary medical attention. In addition, members of a control group may (incorrectly) assume they are immune from the disease because they believe they have received an effective vaccine (which they have not). IRBs sometimes require that control group subjects be given the first opportunity to receive the vaccine once its safety and effectiveness have been established. If such arrangements are not part of the research design, at the end of the trial control subjects should be informed of both their status vis a vis the vaccine, and the outcome of the trial: e.g., that the vaccine was shown to be safe and effective, but that they either did not receive the vaccine or did not receive an effective dose of the vaccine.

For a discussion of ethical issues related to the clinical testing of AIDS vaccines, see Guidebook Chapter 5, Section F, "AIDS/HIV-Related Research."

POINTS TO CONSIDER

1. Has appropriate FDA clearance and an approved IND been obtained?

2. Is there evidence that the vaccine has been adequately tested in animal trials and in the laboratory?

3. Where appropriate, are subjects clearly told in the consent process that they might receive a placebo or ineffective dose of the vaccine, and thus may not be protected against the disease?

4. Does the protocol provide adequate plans to monitor all subjects for immune status and adverse reactions, respond to problems, and disseminate results?

5. Will subjects be informed about what to do and whom to contact in case of a serious adverse reaction or research-related injury?

APPLICABLE LAWS AND REGULATIONS

Federal Policy for the protection of human subjects

21 CFR 50 [FDA: Informed consent]
21 CFR 56 [FDA: IRB review and approval]
21 CFR 312 [FDA: Investigational new drug research]
21 CFR 600-800 [FDA: Standards for biological products]
21 CFR 630 [FDA: Standards for viral vaccines]

Return to Top of Page

 

 

D. MEDICAL DEVICES

INTRODUCTION

Comprehensive federal regulations governing investigations involving medical devices are comparatively new. In addition to their other duties, IRBs reviewing certain device investigations must also determine whether a device study presents a significant or nonsignificant risk to the human subjects participating in the study. When making determinations of significant versus nonsignificant risk, IRBs must consider not only the risks associated with use of the device itself, but also the risks associated with the investigational device study as a whole.

DEFINITIONS

510(k) Device: A medical device that is considered substantially equivalent to a device that was or is being legally marketed. A sponsor planning to market such a device must submit notification to the FDA 90 days in advance of placing the device on the market. If the FDA concurs with the sponsor, the device may then be marketed. 510(k) is the section of the Food, Drug and Cosmetic Act that describes premarket notification; hence the designation "510(k) device."

General Controls: Certain FDA statutory provisions designed to control the safety of /marketed drugs and devices. The general controls include provisions on adulteration, misbranding, banned devices, good manufacturing practices, notification and record keeping, and other sections of the Medical Device Amendments to the Food, Drug and Cosmetic Act [21 U.S. Code §360(c) (Food, Drug and Cosmetic Act §513)].

Investigational Device Exemptions (IDE): Exemptions from certain regulations found in the Medical Device Amendments that allow shipment of unapproved devices for use in clinical investigations.

Medical Device: A diagnostic or therapeutic article that does not achieve any of its principal intended purposes through chemical action within or on the body. Such devices include diagnostic test kits, crutches, electrodes, pacemakers, arterial grafts, intraocular lenses, and orthopedic pins or other orthopedic equipment.

Nonsignificant Risk Device: An investigational medical device that does not present significant risk to the patient. (See also: Significant Risk Device.)

Postamendments Devices: Medical devices marketed after enactment of the 1976 Medical Device Amendments.

Preamendments Devices: Medical devices marketed before the enactment of the 1976 Medical Device Amendments.

Predicate Devices: Currently legally marketed devices to which new devices may be found substantially equivalent under the 510(k) process.

Premarket Approval: Process of scientific and regulatory review by the FDA to ensure the safety and effectiveness of Class III devices.

Significant Risk Device: An investigational medical device that presents a potential for serious risk to the health, safety, or welfare of the subject. Such a device is:

• intended for use as an implant and presents a potential for serious risk to the health, safety, or welfare of the subject; or
• purported or represented to be of use in supporting or sustaining human life and presents a potential for serious risk to the health, safety, or welfare of the subject; or
• intended for a use that is of substantial importance in diagnosing, curing, mitigating, or treating disease, or otherwise preventing impairment of human health, and presents a potential for serious risk to the health, safety, or welfare of the subject; or
• otherwise presents a potential for serious risk to the health, safety, or welfare of a subject.

OVERVIEW

The 1976 Medical Device Amendments (the Amendments) to the Federal Food, Drug and Cosmetic Act (the Act) were passed to give the FDA additional authority to assure safety and effectiveness in devices intended for human use. New medical devices must be cleared by the FDA prior to being placed on the market. As part of the clearance process, all medical devices are classified into one of three categories by the FDA based on the extent of control necessary to ensure the safety and effectiveness of each device [21 U.S. Code §360(c) (Food, Drug and Cosmetic Act §513)].

Medical devices are classified as Class I, Class II, or Class III devices depending on several criteria. Devices are classified as Class I medical devices if their safety and effectiveness can be assured by the general controls of the Amendments. The general controls include the provisions of the Act pertaining to adulteration, misbranding, banned devices, notification, repair, replacement or refund, records and reports, and restricted devices. In addition, general controls require device manufacturers or other designated persons, unless specifically exempted, to register their establishment, list their device, submit a premarket notification application, and be in compliance with the good manufacturing practices (GMPs). If a device cannot be classified as a Class I device because the general controls are insufficient to provide reasonable assurance of the safety and effectiveness of the device, the device may qualify for Class II classification. A Class II device must comply with general controls, and, in addition, the sponsor must provide sufficient information about the device to establish special controls that are sufficient to provide such assurance. Examples of special controls include the promulgation of performance standards, postmarket surveillance, the establishment of patient registries, and the development and dissemination of guidelines.

Devices are classified as Class III devices when: (1) their safety and effectiveness cannot be reasonably assured through either general or special controls; and (2) they are life-sustaining, life-supporting, implanted in the body, or of substantial importance in preventing impairment to health.

A new device that a manufacturer claims is substantially equivalent to a currently legally marketed device may be marketed after the FDA is notified of the intent to market, and the agency concurs with the manufacturer's claim of equivalence to other marketed devices. If the FDA determines that the new device is not substantially equivalent to a predicate device, the new device is automatically placed in Class III, and the manufacturer must obtain premarket approval from the FDA. Alternatively, the sponsor (or others) may petition the FDA to reclassify the device into Class I or II.

Investigational devices are medical devices that are the object of clinical research to determine their safety or effectiveness. Clinical investigations are necessary to support a request for premarket approval. Studies involving human subjects that are undertaken to develop safety and effectiveness data for medical devices must be conducted according to the requirements of the Investigational Device Exemption regulations [21 CFR 812] or Investigational Exemptions for Intraocular Lenses [21 CFR 813]. An approved IDE exempts a device from certain sections of the Act (e.g., misbranding under §502; registration, listing, and premarket notification under §510; special controls under §513; premarket approval under §515; banned devices under §516; records and reports under §519; restricted devices under §520(e); good manufacturing practices under §520(f); and color additive requirements under §706).

The IDE regulation describes two types of device investigations: significant risk device studies and nonsignificant risk device studies. Clinical trials involving significant risk devices require both FDA and IRB approval; sponsors must meet the full IDE requirements, including obtaining an FDA-approved IDE. Approval of studies involving nonsignificant risk devices require only IRB approval; no IDE is required to be formally submitted to the FDA. However, the sponsor must comply with the abbreviated regulatory requirements for such devices [21 CFR 812.2(b)]. The FDA may overturn IRB determinations that a device presents no significant risk.

IRB CONSIDERATIONS

In reviewing studies involving medical devices, IRBs should recognize that they must make two determinations: (1) whether a device study presents significant or nonsignificant risk; and (2) whether the study should be approved. These questions should be considered separately because the issues involved in making these decisions are quite different. Determining whether a device study poses a significant risk is based solely on considerations of risk to subjects, while IRB approval of the study is based on many factors. The discussion in this Section first considers IRB determinations of significant risk.

The FDA reviews and approves IDEs for significant risk device studies; it exercises less regulatory control over nonsignificant risk device studies. The initial responsibility for making the nonsignificant risk assessment for studies lies with the sponsor. If the sponsor believes that a particular device study presents a nonsignificant risk, the sponsor should provide the IRB with the study proposal, an explanation of why the device study presents a nonsignificant risk, and any other supporting information, such as reports of prior investigations. The sponsor should also tell the IRB whether the FDA or any other IRB has made a risk assessment and what the results of those assessments were. The IRB reviews the information, and may or may not agree with the sponsor's determination. If the IRB finds that the device study presents a nonsignificant risk, the investigation may begin without submission of an IDE application to the FDA. If the IRB disagrees with the sponsor's determination that a device study presents nonsignificant risk to human subjects, the sponsor must so notify the FDA, whether or not the sponsor ultimately conducts the study at that institution.

If the study comes to the attention of the FDA, the agency's Office of Device Evaluation may reach a different conclusion on the risk presented by a device study than that reached by the IRB. If the FDA overrules an IRB's decision that a device study presents nonsignificant risk, the sponsor must then submit an IDE application to the FDA. The IRB must then review the investigation as a significant risk device study, and the investigator will be subject to more stringent recordkeeping and reporting requirements.

In determining whether a device study presents a significant or a nonsignificant risk, both the risks of the device and the risks associated with the procedure for using the device (e.g., surgery for installing an implant) must be considered. The comparison of risks is the basis for the other decision the IRB must make: whether to approve the research.

The clinical investigator should provide the IRB with adequate information about a device's regulatory status and the results of any risk assessment the FDA may have made. The IRB may also ask the sponsor whether other IRBs have reviewed the study and what determinations were made. IRBs may also request the sponsor or clinical investigator to provide documentation of appropriate FDA clearances, and may consult the FDA for its opinion on risk.

In the past, clinical investigations of intraocular lenses (IOLs) differed from other medical device studies in that there were few restrictions on the total number of subjects in an IOL investigation. Unlimited "adjunct" studies were phased out when enough approved IOLs became commercially available. IOL studies are now limited in enrollment size, as are other medical device studies.

Clinical investigations involving IOLs that commenced before July 27, 1981, are exempt from investigational device requirements [21 CFR 812], since they are subject to specific regulations on intraocular lenses [21 CFR 813], which specify procedures for IRB review and informed consent.

The IRB's second responsibility is to decide whether to approve the proposed research. In general, full IRB review is required for both significant and nonsignificant risk studies. However, some studies involving nonsignificant risk devices may also be considered minimal risk studies, and thus may be reviewed through the expedited review procedure established by the IRB.

IRBs need to keep in mind the difference between the risk/benefit evaluation made in the context of approving the research and the IRB's assessment of whether use of the device poses significant or nonsignificant risk. The latter decision categorizes the degree of risk of harm based upon the seriousness of the harm that may result from the use of the device; the former is a balancing of those risks (plus the risks of the research process) against the potential benefits to be gained from conducting the research.

The criteria for deciding whether a medical device study should be approved are the same as those used to evaluate research involving any FDA-regulated product. The IRB should determine that risks to subjects are minimized and are reasonable in relation to anticipated benefits and knowledge to be gained, that subject selection is equitable, informed consent procedures and documentation are adequate, and that provisions for monitoring the study and protecting subjects' privacy and confidentiality of data are acceptable. As in other clinical investigations, an IRB's decision to approve the research must take into account the risks and benefits of the investigational device as compared with the other available therapies. However, the IRB should not simply consider the increase in risk over standard treatment, but rather the risk of the procedure as a whole.

For further information and guidance on studies involving medical devices, contact:

Dr. Michael J. Blackwell
Chief, IDE Section (HFZ-403)
Office of Device Evaluation
Center for Devices and Radiological Health
Food and Drug Administration
1390 Piccard Drive
Rockville, MD 20850
Tel: (301) 427-1190

Mr. Richard M. Klein
Health Assessment Policy Staff
Office of Health Affairs (HFY-20)
Food and Drug Administration
Room 11-44, 5600 Fishers Lane
Rockville, MD 20857
Tel: (301) 443-1382

POINTS TO CONSIDER

1. What risks are presented by the device? Are they significant or nonsignificant?

2. Have other IRBs reviewed and made decisions regarding this device? (Such information should be available from the sponsor or clinical investigator.)

3. What is the status of the device with the FDA? Has the device been approved for marketing? Is the device approved for other indications? Is it now being studied for a different indication? Is an IDE needed for this device? If so, has it been approved?

APPLICABLE LAW AND REGULATIONS

Federal Policy for the protection of human subjects

The Food, Drug and Cosmetic Act, as amended [codified at U.S. Code, Title 21]

The Medical Device Amendments of 1976 [P.L. 94-295, 90 Stat. 539 (May 28, 1976)]

The Safe Medical Devices Act of 1990 [P.L. 101-629]

21 CFR 50 [FDA: Informed consent]
21 CFR 56 [FDA: IRB review and approval]
21 CFR 812 [FDA: Investigational device exemptions]
21 CFR 813 [FDA: Investigational exemptions for intraocular lenses]

Return to Top of Page

 

E. USE OF RADIOACTIVE MATERIALS AND X-RAYS

INTRODUCTION

Radiopharmaceuticals and X-rays are widely used in medicine today for both diagnostic and therapeutic purposes. Certain aspects of human physiology can only be studied through exposure to radiation, or can be studied more safely by radiation than by alternative methods.

The types of radiation used most frequently in medical investigations and treatments are X-rays, gamma rays, and beta radiation. In addition to passing X-rays through the body to produce an image, some procedures use contrast agents to outline or define the shape of internal structures, or to image metabolic processes. Nuclear medicine uses procedures in which radioactive materials (i.e., radiopharmaceuticals) are injected, ingested, or inhaled into the body. Most medical institutions have a radiation safety committee responsible for evaluating the risks of medical projects involving radiation and limiting the radiation exposure of employees and patients. Nevertheless, IRBs should have an understanding of radiation and its biological effects so they can evaluate the relative risks and benefits of research proposals utilizing radioactive materials or X-rays.

DEFINITIONS

Radioactive Drug: Any substance defined as a drug in §201(b)(1) of the Federal Food, Drug and Cosmetic Act that exhibits spontaneous disintegration of unstable nuclei with the emission of nuclear particles or photons [21 CFR 310.3(n)]. Included are any nonradioactive reagent kit or nuclide generator that is intended to be used in the preparation of a radioactive drug and "radioactive biological products," as defined in 21 CFR 600.3(ee). Drugs such as carbon-containing compounds or potassium-containing salts containing trace quantities of naturally occurring radionuclides are not considered radioactive drugs.

Radioactive Drug Research Committee (RDRC): An FDA-approved institutional committee responsible for the use of radioactive drugs in human subjects for certain research purposes [21 CFR 361.1]. Research involving human subjects that proposes to use radioactive drugs must be approved by the RDRC and must meet various FDA requirements, including limitations on the pharmacological dose and the radiation dose. The research must be basic research, not intended for diagnosis or treatment of a disease. Furthermore, the exposure to radiation must be justified by the quality of the study and the importance of the information it seeks to obtain. The committee is also responsible for continuing review of the drug use to ensure that the research continues to comply with FDA requirements, including reporting obligations. The committee must include experts in nuclear medicine as well as other medical and scientific members.

Radiopaque Contrast Agents: Materials that stop or attenuate radiation that is passed through the body, creating an outline on film of the organ(s) being examined. Contrast agents, sometimes called "dyes," do not contain radioisotopes. When such agents are used, exposure to radiation results only from the X-ray equipment used in the examination. The chemical structure of radiopaque contrast agents can produce a variety of adverse reactions, some of which may be severe — and possibly life-threatening — in certain individuals.

Radiopharmaceuticals: Radioactive drugs that are labeled or tagged with a radioisotope. These materials are largely physiological or subpharmacological in action, and, in many cases, function much like materials found in the body. The principal risk associated with these materials is the consequent radiation exposure to the body or to specific organ systems when they are introduced into the body.

REM: Acronym for Roentgen Equivalent in Man; the unit of measurement for a dose of an ionizing radiation that produces the same biological effect as a unit of absorbed dose (1 rad) of ordinary X-rays. One millirem is equal to 1/1000 of a rem.

OVERVIEW

The quantity of natural background radiation to which we are exposed varies considerably (e.g., radiation exposures are much lower at sea level than they are at higher altitudes). The average annual natural background radiation from all sources in the United States is approximately 100 to 125 millirems (mrem) per year, while some individual exposures may be more than 400 mrem per year. Diagnostic medical procedures are the most likely source of additional radiation exposure. Estimates suggest that medical procedures increase the total exposure by 50 to 70 mrem per person per year.

Experts disagree, however, over the fundamental concepts that affect how radiation risks from medical procedures and other sources are estimated. The disagreements include debate about the existence of a theoretical threshold level below which no harmful effects occur. The National Council for Radiation Protection and Measurement (NCRPM) takes the position that there is no absolutely safe radiation dose. Generally, only approximations of risk from exposure are available; they are based on extrapolations from known exposures to high levels of radiation. The NCRPM has recommended dose standards; the Nuclear Regulatory Commission (NRC) has established occupational dose limits. The occupational dose limits vary according to the part of the body exposed to radiation.

The NRC is responsible for those radioactive materials considered to be "source material," "byproduct material," or "special nuclear material" [10 CFR Parts 30, 40, and 70]. The NRC directly regulates these materials in 21 states; the other 29 states, known as "Agreement States," have entered into an agreement with the NRC to regulate uses within their states of byproduct material, source material, or special nuclear material involving less than certain quantities. Agreement States may have unique policies or standards concerning the use of radioactive materials in research that could, in some cases, be more restrictive than those of the NRC. Naturally-occurring or accelerator-produced radioactive materials (NARM), such as Thallium-201, are not covered by the Atomic Energy Act; therefore they are not regulated by the NRC. Those radioactive materials (NARM) may be dealt with under specific state regulations (in both Agreement States as well as non-Agreement States) governing the use of radioactive materials.

The FDA requires investigators to submit an Investigational New Drug Application (IND) for radioactive drugs, kits, or generators that are to be used for investigational diagnostic or therapeutic purposes (including testing to establish their safety and effectiveness). An exception is made for radioactive drugs to be used in certain research designed to study the metabolism of the drug or to gather information about human physiology, pathophysiology, or biochemistry, but not intended for immediate therapeutic, diagnostic, or similar purposes [21 CFR 361.1]. If the radiation dose will not exceed the limits set forth in these regulations, the study design meets other research criteria, and the protocol is approved by a Radioactive Drug Research Committee (RDRC), the investigator does not need to submit an IND. Current radiation limits for the use of such drugs in research (including radiation doses from X-ray procedures that would not have occurred but for the study) are as follows [21 CFR 361.1]:

• For an adult research subject, radiation to the whole body, active blood-forming organs, the lens of the eye, or the gonads may not exceed a single dose of 3 rems or an annual cumulative dose of 5 rems.
• The amount of radiation to other organs may not exceed a single dose of 5 rems or an annual cumulative dose of 15 rems.
• Permissible doses for children (persons under age 18) are 10 percent of those for adults. The FDA must approve studies involving children before the study begins.

[See also 21 CFR 312.2(b), providing certain exemptions from IND application requirements.]

In addition to the RDRC, most medical institutions also have an Institutional Radiation Safety Committee, which assesses the risks that may be associated with exposure to radiation, both for research subjects and employees. In some states or institutions, review by the Radiation Safety Committee is mandated by law or policy; in others, the committee's review is offered as an opinion to the IRB to help it assess the risks and benefits of a given study involving radiation exposure.

IRB CONSIDERATIONS

An IRB should distinguish between radiation exposure resulting from routine medical management of a patient and radiation exposure that is part of research, including a clinical investigation. Although the occupational dose limits may not necessarily be appropriate when applied in a research setting, they do provide some guidance when exposure to radiation for research purposes is contemplated.

The likelihood of adverse effects associated with radiation exposure is generally considered to be low, but adverse effects can be serious when they do occur. Some effects rarely present themselves until many years after the subject has been exposed to radiation. The two adverse effects most commonly associated with radiation exposure are certain types of cancer and genetic damage.

The increased risk of genetic damage is of particular concern because exposure to radiation may involve substantial risk to the subject's unborn offspring. When the proposed research poses risk of genetic damage, an IRB should pay particular attention to the subject selection criteria. The human embryo is known to be particularly susceptible to damage from exposure to radiation; research involving pregnant or possibly pregnant women has therefore been of particular concern. Pregnancy tests could be required where doubt exists as to the presence of pregnancy, or the subject might be asked to use an effective contraceptive method during the course of the research. [See Guidebook Chapter 3, Section C, "Selection of Subjects," and Chapter 6, Section B, "Women."] Recent studies have suggested that male sperm cells are also adversely affected by radiation. Thus, no radiation dose should be considered risk-free if it is directed toward, or absorbed by, the reproductive organs.

Research involving radiation may also pose risks to lab personnel, nursing staff, and family members. This increased risk usually results from exposure to nuclear sources of radiation used in a medical device or nuclear medicine or radiotherapy. For example, when nuclear-powered artificial heart implants were under consideration, a federal panel expressed concern over the possible exposure and resultant risk to the patient's spouse.

Additional risk may be associated with the intravascular administration of contrast agents used in X-ray procedures (e.g., intravenous pylograms (IVP), venograms, and cardiac catheterizations). The risks vary depending on the dose of the contrast agents, the chemical nature of the contrast agent used, and the age and disease state of the subject. Conditions such as advanced age, renal disease, diabetes, cardiac, or cerebrovascular disease, asthma, or chronic obstructive pulmonary disease may greatly increase the risk associated with the proposed study. Unsuspected anaphylactic reactions may also, although rarely, occur.

Radiopharmaceuticals present relatively low risks of adverse reactions unrelated to their radioactivity. The principal risks associated with radiopharmaceuticals are posed by the radioisotope's energy, its half-life, the radiosensitivity of the organ system being studied, and the radiation dose to the target organ, adjacent organs, and the whole body. Other factors are, however, also relevant. For example, the dose of a labeled brain receptor agent or the status of a subject's brain receptors must be considered.

In addition to determining the level of risk associated with exposure to radiation, IRBs must be concerned with informed consent. Specifically, IRBs must determine what subjects should be told: how properly to communicate the uncertainty about the risk of harm posed by exposure to the level of radiation involved in the study. Since subjects must be given sufficient information on which to decide whether to participate, consent should be based on information that the subjects may reasonably be expected to want to know. The question for the IRB is how much risk must there be before a "reasonable volunteer" would want to know about it. Given the sensitivity of our society to the uncertainty surrounding the risks associated with radiation exposure, IRBs should require that subjects be told that participation in the research involves exposure to radiation.

Several ways of explaining the risks associated with exposure to radioactive materials to potential subjects have been suggested, but none are totally satisfactory. One method used is comparing the risk of death from radiation exposure to that of more familiar activities such as air travel or cigarette smoking. A second method compares the incidence of death per year from radiation exposure with the mortality rates of various occupations. Comparisons may also be made between the proposed research exposure and the dose received from cosmic and background radiation to which a subject is naturally exposed. The proposed research exposure may also be compared with the annual maximum permissible exposures suggested by the NCRPM for occupational workers. Finally, the research exposure can be compared with exposures from more familiar medical procedures, such as chest X-rays.

The major problem with expressing risks in comparative terms is that the actual risk from low levels of exposure is not known. This uncertainty should be communicated to research subjects. Even in cases where the risks from exposure are considered to be minimal and not reasonably foreseeable, the IRB may determine that the information concerning exposure and its possible effects is something that research subjects might reasonably want to know.

The IRB should ensure that the risks of radiation exposure are minimized. In an attempt to minimize radiation exposure, experts have developed a principle known as ALARA: As Low As Reasonably Achievable. IRBs should ensure that the ALARA principle is observed. [See also 21 CFR 361.1(b)(3) (limit on radiation dose).]

 

 

POINTS TO CONSIDER

1. Can the information to be gained from the research project be gathered using methods that do not expose subjects to more radiation than that to which they would naturally be exposed?

2. Could the research be performed on patients undergoing the procedures for diagnostic or therapeutic purposes?

3. Will the smallest exposure (dose) possible be used in the study?

4. Have investigators taken steps to avoid re-exposure? Are procedures in place to ensure that investigators will use a minimum number of re-exposures in the event that the study needs to be repeated?

5. Are adequate radiation safety measures being taken to protect research subjects and others who may be exposed to radiation?

6. Have the investigators taken adequate precautions to screen subjects and exclude those not essential to the research project and those at increased risk from exposure to radiation or contrast agents?

7. Will both men and women be informed of the risks to future offspring due to possible genetic damage?

8. Will women of childbearing potential be adequately informed of the risks to an embryo associated with radiation exposure in early pregnancy, and of the importance of disclosing a possible pregnancy to the investigator? Does the protocol make adequate provisions for detecting pregnancies?

APPLICABLE LAWS AND REGULATIONS

Federal Policy for the protection of human subjects

10 CFR 19 [NRC: Notices, instructions, and reports to workers; inspections]
10 CFR 20 [NRC: Standards for protection against radiation]
10 CFR 35 [NRC: Medical use of byproduct material]
21 CFR 50 [FDA: Informed consent]
21 CFR 56 [FDA: IRB review and approval]
21 CFR 361.1 [FDA: Radioactive drugs for certain research uses]
21 CFR 312 [FDA: Investigational new drug application]

State laws regarding radioactive materials licensure

Return to Top of Page

 

 

 

F. AIDS/HIV-RELATED RESEARCH

INTRODUCTION

The human immunodeficiency virus (HIV) is a pathogenic retrovirus that causes acquired immunodeficiency syndrome (AIDS) and its related diseases in humans. Because of its high rate of mortality, AIDS has become the center of worldwide attention; research into the development of safe and effective therapies, as well as methods of prevention of this fatal disease, is currently a national public health priority.

HIV-related research centers on both biomedical and behavioral questions. Biomedical research has been characterized as falling into five major scientific categories: "(1) the study of the distribution of HIV infection and AIDS in the population (epidemiology) and the pattern of disease progression (natural history); (2) the identification and characterization of the virus that causes AIDS (etiologic agent); (3) delineation of the mechanisms by which the virus destroys the immune system and produces disease (pathogenesis); (4) the development and testing of potential therapies for HIV infection and its complications; and (5) the development and evaluation of potential AIDS vaccines" [Hamburg and Fauci (1989), p. 22].

Behavioral research on HIV focuses on: (1) identifying the social, psychological, and behavioral conditions of disease transmission and prevention; (2) the effects of psychological state on immunosuppression; and (3) the role of psychology in alleviating the distress experienced by persons affected by HIV infection (including families, friends, and persons at risk).

IRB CONSIDERATIONS

Research designed to answer the many biomedical and behavioral questions presented by HIV poses numerous ethical concerns. Primary among them are considerations of privacy, confidentiality, and justice (fairness in the distribution of the benefits and risks of research). The subjects involved in HIV-related research, HIV-infected individuals, and persons at risk of HIV infection, are particularly vulnerable, both because of their disease status, and because the disease disproportionately affects certain populations: male homosexuals and bisexuals, intravenous drug users, minorities, and, increasingly, women and children. [See Guidebook Chapter 6, "Special Classes of Subjects."]

An overriding concern in HIV research is confidentiality. Subjects included in HIV-related studies are understandably concerned about the confidentiality of the data, since breaches in confidentiality could have severe adverse consequences such as loss of employment or insurance coverage, or criminal charges. OPRR guidance on HIV studies states that:

where identifiers are not required by the design of the study, they are not to be recorded. If identifiers are recorded, they should be separated, if possible, from data and stored securely, with linkage restored only when necessary to conduct the research. No lists should be retained identifying those who elected not to participate. Participants must be given a fair, clear explanation of how information about them will be handled.

As a general principle, information is not to be disclosed without the subject's consent. The protocol must clearly state who is entitled to see records with identifiers, both within and outside the project. This statement must take account of the possibility of review of records by the funding agency.... [OPRR Reports, Dear Colleague Letter (December 26, 1984), p.3.]

IRBs should also consider whether and how information from HIV-related studies will be recorded in subjects' medical records, and may decide to impose limits on the recording of such data. Before agreeing to participate in an HIV study, subjects should be informed of exactly what information will be recorded, and whether any state laws require the reporting of HIV infection or other disclosures of information. The research protocol should also deal with the possibility of attempts under compulsory legal process to force disclosure of records, how such attempts will be responded to, and whether individuals will be notified of such attempts. [See also the Guidebook Chapter 3, Section D, "Privacy and Confidentiality," which deals with certificates of confidentiality and subpoenas.] The protocol should specifically set forth how to respond to requests by third parties who have authorizations for disclosure of information signed by subjects. An extensive set of guidelines for confidentiality in research on HIV has been developed by a group of prominent scholars, practitioners, and community members, and may be helpful to IRBs considering HIV-related protocols. [See Bayer, Levine, and Murray (1984).]

The PHS has an established policy on the issuance of certificates of confidentiality to projects that are subject to the reporting of communicable diseases to state and local health departments. The policy applies to projects that intend routinely to determine whether its subjects have communicable diseases, and that are required to report them under state law. Certificates will be issued: (1) where the referring treating physicians assure the project that they have complied with reporting requirements; (2) the investigator has reached an agreement with the health department about how he or she will cooperate with the department to help serve the purposes of the reporting requirements (unless the investigator can show why such cooperation is precluded); and (3) only where disclosures of identifiable information about subjects comply with regulations on subject protection, and are explained clearly to subjects prior to their participation [Mason (August 9, 1991)]. [See also Guidebook Chapter 3, Section D, "Privacy and Confidentiality."]

The giving of voluntary consent, axiomatic to all research involving human subjects, applies equally in HIV-related research. Complicating the consent issue, however, is that HIV-related illness, particularly in its later stages, can cause dementia, thus affecting the ability of subjects to give consent or continue to consent to ongoing research. Research protocols should deal with this possibility; IRBs should ensure that subjects in this particularly vulnerable condition are adequately protected. [See also Guidebook Chapter 6, Section D, "Cognitively Impaired."]

Research on vaccines and treatments poses some of the most difficult questions, including the level of acceptable risk to subjects when the disease is fatal and no effective therapy is available; whether HIV-infected patients can be used as a placebo group that is not given experimental treatments; how subjects should be selected to receive experimental therapies; whether and under what circumstances healthy and at-risk but not-yet-HIV-infected persons can ethically be asked to participate in vaccine trials.

Clinical Trials of HIV-Related Therapies. Randomized clinical trials (RCTs) and the ethical problems surrounding their use is discussed in Guidebook Chapter 4, Section H and related Guidebook Sections. This Section will focus on questions of particular concern for research involving HIV-infected individuals.

Randomized, controlled clinical trials are considered the research design most likely to yield valid scientific results for the evaluation of the safety and effectiveness of experimental therapies. Ethical use of RCTs depends on the existence of both the ability to state a null hypothesis (also called "theoretical equipoise") and that there be no other therapy known to be more effective than the one being studied in the RCT. A report produced by a working group on clinical HIV research convened by the American Foundation for AIDS Research argues, however, that when no known effective alternative therapy exists, as is presently the case with HIV, it may be justified to consider the use of other forms of controls such as historical controls (that is, to compare the effects of the therapy in the trial population with the treatment experiences of patients with the same disease before use of the experimental therapy) [Levine, Dubler, and Levine (1991), pp. 3, 6]. The justification for this position is that the conditions of "clinical equipoise" (a situation in which there is a "current or likely dispute among expert members of the clinical community as to which of two or more therapies is superior in all relevant respects," and which is also necessary for an RCT to be ethical) are not satisfied [id.]. The working group issued a document that included 57 recommendations on the conduct of clinical research on HIV, which IRBs may wish to consult [id.].

The use of placebo controls is particularly problematic. As a general matter, where the disease is lethal or seriously debilitating, as in the case of HIV, the use of placebo controls in place of an active control is difficult to justify ethically, despite the possibility that the experimental therapy is harmful (e.g., toxic) rather than therapeutic. In the language of the Belmont Report, the question of the use of control groups in this situation is one of beneficence: Are potential benefits maximized in all arms of the trial? The fatal nature of the disease leaves patients in a desperate position in which many seek any promising treatment. It has been suggested that the question may be resolved in favor of placebo controls only under two conditions: (1) when there is either no known effective therapy that can be used as an active control, or subjects are persons who cannot tolerate a known effective therapy; and (2) the trial therapy is "so scarce that only a limited number of patients can receive it" [Levine, Dubler, and Levine (1991), p. 8]. A fair way to then assign subjects to the active and control arm(s) is through a lottery [id.] [See also Macklin and Friedland (1986), pp. 277-79, and Guidebook Chapter 4, Section H, "Clinical Trials," and related Guidebook Sections.]

Once there is sufficient evidence of either a beneficial therapeutic effect, unacceptable side effects, or indication that there is a very low probability of establishing statistically significant research results, the trial should be stopped or the protocol should be modified [Macklin and Friedland (1986), pp. 177-78]. Where an experimental therapy is shown to have a beneficial therapeutic effect, the control group should be offered access to the experimental therapy. Prospective subjects should be informed of the probability of being assigned to the control group, the risks associated with being assigned to either the treatment or control group, the criteria that will be used for determining a beneficial effect sufficient to discontinue the control arm of the trial, and the consequences of discontinuing the control arm (e.g., will control subjects be added to the experimental group, will they be given the experimental therapy on a treatment basis, will they be offered the experimental therapy only if they pay for its cost, or will they be dropped from the study without access to the experimental therapy). It should be made clear to prospective subjects that the likelihood of the experimental therapy having harmful effects may well be as great as the likelihood of its having beneficial effects.

The selection and recruitment of subjects is also of concern. Subjects for clinical trials are often recruited on the recommendation of treating physicians. Unable or unwilling to obtain medical care, many individuals have been excluded from participation in trials. Others, not aware of the existence of trials, are also left out. Care should be taken to ensure the appropriate inclusion of women, children and adolescents, and minority groups in HIV-related clinical trials. Note also that IRBs must follow the additional protections provided in the DHHS regulations wherever applicable. [See Subpart B (fetuses, pregnant women, and human in vitro fertilization), Subpart C (prisoners), and Subpart D (children).]

When reviewing protocols involving HIV-infected or at-risk individuals or persons, IRBs should consider including (as consultants, if they are not already members) persons knowledgeable about and experienced in working with such subjects [Federal Policy §___.107]. Some investigatory groups have used "community advisory committees" as a means both of better understanding the concerns of the subject population and of educating the HIV-infected community about clinical research.

Vaccines. The testing of AIDS/HIV vaccines in human subjects raises substantial ethical issues. First and foremost is the question of risks and benefits. Limited availability of animal data means that many of the risks that might be associated with an AIDS/HIV vaccine (e.g., vaccine-induced immunotoxicity) are unknown. Nonetheless, the importance of developing an AIDS/HIV vaccine is felt to outweigh these uncertainties. From the standpoint of protecting the welfare of human subjects, however, the lack of knowledge about risk and the potential for the existence of serious risk must be clearly communicated and consented to by prospective subjects.

While all viral vaccines pose risks, HIV vaccines may, in addition, increase the risk of acquiring the disease when subsequently exposed to HIV. Also, because of potential immune tolerance, subjects may not be able to be vaccinated with a different AIDS/HIV vaccine if the experimental one proves ineffective. Persons with whom the subject is in close contact may also be at risk of transmission of recombinant viruses (through the injection site). IRBs should consider the degree to which investigators have minimized these risks, and ensure that subjects are adequately informed of and consent to these and other potential physical risks.

Another issue about which subjects must be informed is the effect of participation in the trial on their HIV serostatus and the potential social ramifications of changes in HIV serostatus. Just as persons infected with HIV through more usual means of transmission (e.g., sexual activity, the use of intravenous drugs, or blood transfusions) will test positive on antibody screening tests, so too will persons immunized with experimental AIDS/HIV vaccines. There may be limited access to diagnostic methods for distinguishing between persons who are HIV-infected and persons who have received HIV vaccinations. One way to help alleviate this problem is for trial sponsors to follow the lead of the National Institute of Allergy and Infectious Diseases (NIAID), and provide subjects with documentation certifying participation in the vaccine trial. Nonetheless, participation in AIDS/HIV vaccine trials in itself may carry a social stigma.

Informing Subjects of Their HIV Serostatus. Some research protocols involve screening blood samples for HIV seroprevalence or other procedures through which subjects' HIV serostatus will be discovered. In addition to ensuring that the confidentiality of this information and all research data is scrupulously provided for, and that subjects will be informed that they will be tested and of the risks and benefits involved, IRBs will need to consider the circumstances under which subjects should or must be told of their HIV serostatus. PHS policy requires that where HIV testing is conducted or supported by the PHS, individuals whose test results are associated with personal identifiers must be informed of their own test results and provided the opportunity to receive appropriate counseling unless the situation calls for an exception under the special circumstances set forth in the policy. Under the PHS policy, individuals may not be given the option "not to know" their test results, either at the time of consenting to be tested or thereafter. The acceptable "special circumstances" include such compelling and immediate reasons as an indication that a given individual would attempt suicide if informed that he or she was HIV seropositive; that extremely valuable knowledge might be gained from research involving subjects who would be expected to refuse to learn their HIV antibody results; or research activities conducted at foreign sites where cultural norms, the health resource capabilities, and official health policies of the host country preclude informing subjects of their HIV serostatus. Subjects should also be informed early in the consent process of any plans to notify subjects' sexual or needle-sharing partners. [See OPRR Reports ("Dear Colleague" letters dated December 26, 1984 and June 10, 1988).] Several commentators have taken issue with the position that subjects should be told of their serostatus regardless of their wishes. [See, e.g., Novick (1986) and Dubler (1986); compare Landesman (1986).] While this issue may be controversial, opportunities for early intervention weigh in favor of policies that require informing subjects of their HIV serostatus.

Counseling. Whenever subjects will be informed of their HIV serostatus, appropriate pretest and post test counseling must be provided. Counselors should be qualified to provide HIV test counseling and partner notification services. IRBs should ensure that such provisions are made. [See OPRR Reports ("Dear Colleague" letters dated December 26, 1984 and June 10, 1988)]

See also Guidebook Chapter 2, Section B, "Food and Drug Administration Regulations and Policies" (discussing expanded availability of investigational agents), and Chapter 4, "Considerations of Research Design."

Behavioral Research. Research on behavioral questions related to HIV often centers on what behavioral factors contribute to disease transmission and dissemination, as well as other psychosocial factors related to HIV (e.g., the relationship of stress to immunosuppression). The American Psychological Association has expressed concerns for subjects' privacy, protections against the intrusive nature of behavioral research (because research on risk factors and modes of disease transmission often probes intimate details of subjects' lives such as sexual practices and past history of illicit drug use), confidentiality, and the need to carefully debrief subjects.

Vulnerability of Subjects. In addition to the ethical issues raised by the conduct of HIV-related research itself, the involvement of HIV-infected subjects presents special concerns to which IRBs should be sensitive. As noted above, homosexual and bisexual men, intravenous drug users, minorities, and, increasingly, women and children constitute the bulk of the HIV-infected population. Their vulnerability as subjects arises primarily because their HIV status presents special concerns of confidentiality and privacy. Knowledge of a person's HIV status can lead to discriminatory practices on the part of employers, landlords, insurance companies, and others. That HIV disproportionately affects certain populations heightens the threat of inappropriate disclosure of HIV-related data. In addition, characteristics of the progression of AIDS, which can include both physical incapacity and loss of mental capacity, can impinge on subjects' ability to exercise their right to autonomy in the course of the research. IRBs can ensure that AIDS patients and other HIV-infected subjects are adequately protected by viewing each subject first and foremost as an individual. Researchers working with HIV-infected persons must be capable of dealing with social, emotional, and psychological, as well as physical factors. Taking such a multifaceted approach to working with this subject population is a means of incorporating the various necessary cultural and filial influences into the research relationship. Researchers should seek the advice and consultation of experts in these and other relevant fields as necessary.

Another factor that heightens the vulnerability of HIV-infected individuals is the lack of available treatment alternatives. At present, HIV infection is believed uniformly to progress to AIDS; no available treatment cures AIDS, although some therapies postpone the onset and severity of opportunistic infection. Prospective subjects in HIV-related studies may, therefore, agree to participate in research out of a hope for a cure, which may or may not be realistic. But while IRBs should protect subjects against exposure to excessive risk, they must also guard against paternalism. Despite the fatal nature of the disease, there may be risks to which individuals should not be asked to subject themselves; despite their vulnerability, however, prospective subjects should be given the opportunity to participate and obtain whatever benefits may be available. IRBs should consider protocols and make their evaluation of the requisite factors (i.e., the level of risk involved, a positive risk/benefit ratio, equitable selection of subjects, informed consent, and protection of privacy and confidentiality) with this concern in mind. The additional protection that IRBs can provide is to ensure that the protocol, its goals, and the research benefits and risks are clearly and simply delineated and communicated to the subject. It is important that participation in the research not engender either false hopes or a sense of hopelessness. Furthermore, IRBs should try to ensure that access to health care does not serve as a lure for participation.

IRBs need to review participant eligibility requirements closely and extensively monitor the data collection and analysis process. The consent process should also be carefully considered, with special attention to provisions for determining mental capacity to consent and alternative means for obtaining consent, where necessary. [See Guidebook Chapter 6, Section D, "Cognitively Impaired."] The duration of any health care to be rendered through participation, including counseling, should be thoroughly reviewed with subjects. As noted above, subjects must be clearly and explicitly informed of any applicable law or policy that requires either partner notification or notification to health authorities of subjects' HIV serostatus or disease status.

Finally, many HIV-infected persons are economically and/or educationally disadvantaged, and may need adjunct services or other help to be able to participate in research. To ensure that all affected groups have an adequate opportunity to participate, IRBs should give some thought to how investigators might meet these needs, thereby encouraging a broader distribution of the risks and benefits of HIV-related research.

Availability of Drugs and Other Therapeutic Agents for AIDS and HIV-Related Conditions. The availability of experimental drugs and other therapeutic agents for the treatment of AIDS and other HIV-related conditions has been highly controversial. Two mechanisms, Treatment INDs, and a subset of Treatment INDs, Parallel Track programs, have been developed by the FDA to meet this concern. They are discussed in the Guidebook in Chapter 2, Section B, "Food and Drug Administration Regulations and Policies."

POINTS TO CONSIDER

1. Pre-screening clinical study participants for HIV antibody status: See the list of questions provided in OPRR Reports, "Points to Consider for Institutional Review Boards (IRBs) Regarding the Screening of Volunteers for HIV Antibody Status," (circa August, 1989).

2. Is the composition of the IRB membership appropriate for an adequate review of the protocol? Should the IRB seek consultation with laypersons, persons with AIDS or who are HIV-infected, or members of the HIV-affected community?

3. Are subjects' privacy and confidentiality adequately protected? Are certificates of confidentiality appropriate?

4. Does the consent process provide adequately for the special needs of subjects participating in HIV-related research, including subjects with impaired mental capacities and the difficulties of communicating the risks presented by drug and vaccine trials?

5. Will the informed consent process clearly inform the subject of all pertinent information (e.g., the circumstances under which the investigator may terminate the subject's participation without the subject's consent; the circumstances under which the subject may withdraw from participation and the costs associated with withdrawal; the financial costs of participation; how medical care will be handled in the event of injury or onset of opportunistic illness; whether partner notification and/or disease status reporting to health authorities will occur)?

6. Is there a mechanism for dealing with changes in mental capacity and continuing consent? Who will give consent in the event of diminished mental capacity or lack of majority (in the case of children)? Is it necessary to obtain subjects' assent?

7. Are protections against coercion in place?

8. If the protocol involves a clinical trial, have appropriate FDA clearances and an approved IND been obtained?

9. Does the protocol provide for adequate monitoring of all subjects for adverse reactions? Are provisions made for early termination?

10. Will subjects be informed about what to do and whom to contact in case of a serious adverse reaction or research-related injury?

11. Will subjects involved in behavioral research be adequately debriefed? Are intrusions into subjects' privacy minimized?

APPLICABLE LAWS AND REGULATIONS

Federal Policy for the protection of human subjects

21 CFR 50 [FDA: Informed consent]
21 CFR 56 [FDA: IRB review and approval]
21 CFR 312 [FDA: New drugs for investigational use]
45 CFR 46, Subparts B-C-D [DHHS: Protection of human subjects]

Federal Register 57 (April 15, 1992): 13250-13259 [FDA: Parallel track policy]

State and local laws concerning the reporting of HIV-related information

Public Health Service policies related to AIDS research:

U.S. Public Health Service. National Institutes of Health. "Guidance for Institutional Review Boards for AIDS Studies" [Dear Colleague Letter]. OPRR Reports (December 26, 1984).

U.S. Public Health Service. National Institutes of Health. "Policy on Informing Those Tested About HIV Serostatus" [Dear Colleague Letter]. OPRR Reports (June 10, 1988).

U.S. Public Health Service. National Institutes of Health. "Points to Consider for Institutional Review Boards (IRBs) Regarding the Screening of Volunteers for HIV Antibody Status" [Dear Colleague Letter] OPRR Reports [circa August, 1989].

James O. Mason [Assistant Secretary for Health]. "Certificates of Confidentiality — Disease Reporting" [Memorandum]. (August 9, 1991.)

Return to Top of Page

G. TRANSPLANTS

INTRODUCTION

Numerous ethical issues confront IRBs considering research that involves the transplantation of organs or tissues into human subjects. Transplanted organs may be either natural or artificial; natural organs or tissue may be of either human or animal origin. Ethical issues include the physical and psychological risks to the donor and recipient, informed consent, coercion, and the selection of recipient-subjects (i.e., the distribution of organs or tissue to needy recipients).

The ethical considerations surrounding the transplantation of organs concern two basic problems: the scientific basis of the procedure (i.e., risk to the recipient-subject) and the procurement of organs for transplantation. The first problem raises issues with which IRBs are familiar: determining whether the proposed research poses an acceptable risk; balancing that risk with the potential benefits; ensuring that the patient-subject and the donor give their informed consent; and ensuring that the decision to participate is free from coercion and undue influence. The second problem has several facets, including the appropriate selection of recipient-subjects and the obtaining of organs. Equitable subject selection for research on transplantation raises unique questions because of the involvement of the donor in the process and because of the scarcity of appropriate materials (e.g., organs, tissue, or bone marrow) for the transplant procedure. The use of fetal tissue in transplantation is dealt with in Guidebook Chapter 6, Section A, "Fetuses and Human In Vitro Fertilization."

OVERVIEW

Experimental transplants are performed using a number of techniques: An organ or tissue can be obtained from a living relative, a living nonrelative, or a deceased person (usually a nonrelative). Transplants can also be performed using organs or tissue from animals (called xenografts); portions of organs have also been transplanted from living relatives into patient-subjects. The use of artificial implants is another method of replacing diseased organs that has been pursued.

The transplant procedure requires the matching of various factors between donor and recipient (e.g., blood and tissue types). To increase the likelihood of a match (i.e., to decrease the likelihood that the organ or tissue will be rejected by the recipient's system), living relatives are a preferred source of organs or tissue. For some organs, such as a heart, such an arrangement is obviously impossible. Furthermore, the subject may not have a living relative who provides an appropriate match or who is willing to donate the organ or tissue.

IRB CONSIDERATIONS

Candidates for experimental transplant procedures are usually under threat of imminent death; experimental transplant procedures are a last hope for survival. The highly vulnerable status of potential subjects makes stringent review of proposed transplant research essential. Transplant investigations involving children as subjects are governed by Subpart D of the DHHS regulations [45 CFR 46.401-409]. [See Guidebook Chapter 6, Section C, "Children and Minors."]

The first issue with which IRBs must concern themselves is whether the risk of the transplant procedure is outweighed by the potential benefits of the research. [See Guidebook Chapter 3, Section A, "Risk/Benefit Analysis."] The benefits take two forms: intended therapeutic benefit for the individual subject and the benefit to society from the knowledge gained from the research. An important factor when considering the benefit to individual subjects is the availability and quality of therapeutic alternatives for potential subjects. The subjects' prospects for survival and quality of life, with or without the transplant, will be particularly relevant to the IRB's decision.

Transplants involving living donors present a second level of risk that must be evaluated: the risk of obtaining the organ from the donor. That risk entails the risk of the removal procedure itself, plus the long-term risks of living without the donated organ or tissue. When balancing those risks against the potential benefits, one can see that the relationship of the donor to the recipient may be relevant. The donor will not therapeutically benefit from the donation; quite the contrary. The benefit comes, rather from the direct good the donor gives the recipient. In this regard, the living related donor will benefit more directly than will the living nonrelated donor: He or she is increasing the likelihood that the relative (about whom he or she presumably cares more than would a nonrelated donor) will live longer.

As with any research involving human subjects, IRBs need to ensure that subjects give informed consent that is free from coercion or undue influence. Potential subjects for studies involving experimental transplants must be clearly informed of the highly experimental nature of the procedure, including the state of knowledge about the prospects for long-term viability of the organ or tissue.

Complicating the question of consent when the research involves transplants is the involvement of a donor. Where the donor is living, his or her consent must be obtained; the regulations concerning research subjects apply fully to the donor as well as to the recipient. Where the donor is deceased, his or her next of kin must be consulted: State and federal Required Request Laws mandate that the treating physician ask if the family wishes to donate organs from the patient upon his or her death; the deceased may also have indicated a desire to donate his or her organs in the event of death by, for instance, signing an organ donation card.

Technological innovations that allow for the preservation of cadavers and organs has led to concerns about treating brain dead persons as research objects. Some question exists whether deceased donors come within the jurisdiction of IRBs because the federal regulations define subjects as "living individual[s]" [Federal Policy §___.102(f)]. Nevertheless, the President's Commission [(1983), p. 41] suggested that IRBs consider requiring review of research on brain dead persons "to determine whether...it is consistent with 'commonly held convictions about respect for the dead.'" [See also Levine (1986), p. 78.] Considerable controversy surrounds the use of anencephalic infants as a source of organs for donation, with most commentators arguing against their use.

The involvement of living related donors also raises concerns of coercion and undue influence. The pressure on relatives to donate needed organs or tissues is unquestionably great; IRBs must carefully scrutinize the proposed consent process. Some investigators have provided for both medical and psychiatric evaluations and counseling as part of the donor consent process, as well as a waiting period (if feasible) before the transplantation, during which the donor may withdraw consent. Some investigators have also provided for a consent advocate for the donor who is not directly involved in the donor's operation. [See, e.g., Singer, et. al. (1989).]

A further complication to the consent process for organ donors is the minor who is a potential donor for a relative — a sibling, for instance. Where the donor is a minor, the regulations concerning children and minors as research subjects apply [45 CFR 46.401-46.409]. Organ donations from minors raise concerns about the ability of the minor to comprehend the risks of donation, as well as the possibility of coercion or undue influence. [See Guidebook Chapter 6, Section C, "Children and Minors."] IRBs may want to consider requesting the guidance of a court of law before allowing a given donation to be made.

Experimental xenografts have been particularly controversial. The celebrated Baby Fae case, in which an infant received the transplanted heart of a baboon, raised serious questions about IRB review of research involving human subjects. Any research involving transplants should be carefully reviewed by an IRB regardless of the source of funding. The extremely risky nature of the procedure and the special vulnerability of the subjects demand that their welfare be scrupulously protected. Subjects must be clearly informed of the state of knowledge about the long-term viability of the transplant, of alternatives to the procedure, and of all possible physical and psychological effects that may result from the transplant and any other procedures that will be undertaken as a part of the transplant. Consent to the transplant must be carefully documented. [See Caplan (1985), p. 3343].

POINTS TO CONSIDER

1. Does the consent process adequately protect both the donor and the recipient? Is sufficient information provided regarding the risks of all procedures involved? Is adequate provision made for incompetent subjects by providing for trustworthy proxy decision makers?

2. Have both donors and recipients been adequately protected against coercion and undue influence?

3. Are special regulatory provisions applicable, e.g., Subpart D governing children as subjects?

APPLICABLE LAWS AND REGULATIONS

Federal Policy §___.111(a)(3) [Criteria for IRB approval of research: equitable selection of subjects]

Omnibus Budget Reconciliation Act of 1986 (Pub. L. 99-509) enacted sec. 1138, Social Security Act (Required Request Law)

The Uniform Anatomical Gift Act

The Uniform Definition of Death Act

State and local laws pertaining to organ donation

Return to Top of Page  

H. HUMAN GENETIC RESEARCH

INTRODUCTION

Human genetic research involves the study of inherited human traits. Much of this research is aimed at identifying DNA mutations that can help cause specific health problems, developing methods of identifying those mutations in patients, and improving the interventions available to help patients address those problems. The identification of genetic mutations enables clinicians to predict the likelihood that persons will develop a given health problem in the future or pass on a health risk to their children. For many disorders, however, there will be a considerable time lag between the ability to determine the likelihood of disease and the ability to treat the disease.

Efforts to isolate DNA mutations involved in disease in order to understand the origins of the pathophysiological process are becoming increasingly common across the broad sweep of biomedical research, from cardiology to oncology to psychiatry. IRBs should expect to see more of these kinds of studies in the future. The U.S. Human Genome Project (part of the worldwide research effort known as the Human Genome Initiative) is one of the genetic tool making efforts that is facilitating this growth, through the production of better genetic maps and sequencing technology.

The ethical issues raised by this scientific trend primarily concern the management of psychosocially potent personal genetic information. For researchers and IRBs, the major challenge in addressing these issues is the fact that genetic studies typically involve families; the research subjects involved in genetic studies are usually related to each other. As a result, research findings about individual subjects can have direct implications for other subjects, information flow between subjects is increased, and participation decisions are not made entirely independently. A second set of ethical issues emerge in cases in which the results of these studies are used to develop therapeutic interventions at the genetic level. Such concerns involve the special safety precautions and subject selection considerations required for gene therapy research.

Some of the areas described in this Section present issues for which no clear guidance can be given at this point, either because not enough is known about the risks presented by the research, or because no consensus on the appropriate resolution of the problem yet exists. IRBs need to become familiar with the issues and be prepared to address them in the context in which they arise in their particular research setting. Because of the uncertainties involved in genetic research, IRBs may not, for some time, be able to set clear standards for investigators. What IRBs can do, however, is ensure that investigators have thought through the factors that may affect the rights and welfare of human subjects (e.g., risks to privacy, psychological risks, employment and insurance risks). IRBs should require investigators to explain their thoughts on these problems, how they plan to handle them, and how they plan to communicate them to subjects.

IRBs would do well to work together with investigators, so that investigators see the IRB, as they should, as a partner in developing research protocols that adequately protect the participants. For example, IRBs may want to sponsor workshops within their institutions to help inform investigators of what the IRB will be looking for, or invite investigators to consult with the IRB prior to developing genetic research protocols.

Voluntary organizations involved in supporting research on various genetic disorders (e.g., genetic disease support groups and voluntary health associations) can also be useful sources of information for IRBs. Through consultation with voluntary organizations, IRBs can obtain useful information on the human subjects concerns of prospective research participants. These groups can also help act as intermediaries for subject recruitment, which may be particularly helpful for family studies, and can help provide counseling and support services. Not only can they help IRBs (e.g., in subject recruitment), but both IRBs and voluntary organizations benefit when their constituent publics are well-informed about what IRBs do (and do not do).

The issues raised in this Section of the Guidebook are addressed with particular reference to genetic research. General discussion of these issues (e.g., risk/benefit analysis, informed consent, privacy and confidentiality, and vulnerable populations) will also be useful to IRBs, and are found in other chapters of the Guidebook (primarily Chapter 3, "Basic IRB Review," Chapter 4, "Considerations of Research Design," and Chapter 6, "Special Classes of Subjects").

DEFINITIONS

Lod Score: An expression of the probability that a gene and a marker are linked.

Genotype: The genetic constitution of an individual.

Phenotype: The physical manifestation of a gene function.

Proband: The person whose case serves as the stimulus for the study of other members of the family to identify the possible genetic factors involved in a given disease, condition, or characteristic.

IRB CONSIDERATIONS

It may be useful to think of genetic research as being carried out on a continuum comprising four stages: (1) pedigree studies (to discover the pattern of inheritance of a disease and to catalog the range of symptoms involved); (2) positional cloning studies (to localize and identify specific genes); (3) DNA diagnostic studies (to develop techniques for determining the presence of specific DNA mutations); and (4) gene therapy research (to develop treatments for genetic disease at the DNA level).

Unlike the risks presented by many biomedical research protocols considered by IRBs, the primary risks involved in the first three types of genetic research are risks of social and psychological harm, rather than risks of physical injury. Genetic studies that generate information about subjects' personal health risks can provoke anxiety and confusion, damage familial relationships, and compromise the subjects' insurability and employment opportunities. For many genetic research protocols, these psychosocial risks can be significant enough to warrant careful IRB review and discussion. The fact that genetic studies are often limited to the collection of family history information and blood drawing should not, therefore, automatically classify them as "minimal risk" studies qualifying for expedited IRB review.

Pedigree Studies

When investigators attempt to document and study the natural history of an inherited disease, condition, or characteristic, they do so by identifying individual members of families presenting the disease, condition, or characteristic and obtaining information about them and the other members of their family. The result is a pedigree analysis, which, in addition to tracing the natural history of a disease and documenting the range of symptoms involved, may also reveal information about family members that individual members may not have known about previously (e.g., the existence of previously unknown relatives or the presence of stigmatizing diseases, such as mental illness). It may also reveal information about the likelihood that individual members of the family either are carriers of genetic defects or will be affected by the disease.

Subject Recruitment and Retention. The familial nature of the research cohorts involved in pedigree studies can pose challenges for ensuring that recruitment procedures are free of elements that unduly influence decisions to participate. The very nature of the research exerts pressure on family members to take part, because the more complete the pedigree, the more reliable the resulting information will be. For example, revealing who else in the family has agreed to participate may act as an undue influence on an individual's decision, as may recruiting individuals in the presence of other family members. (Both would also constitute a breach of confidentiality. The problem of confidentiality will be dealt with later in this Section.)

Recruitment plans, some of which are described here, can attempt to address these problems; each approach has its own strengths and weaknesses. One strategy is to use the proband as the point of contact for recruiting. This approach insulates families from pressure by the investigator, but presents the risk that the proband may be personally interested in the research findings and exert undue pressure on relatives to enroll in the study. Furthermore, the proband may not want to act as a recruiter for fear that other family members will then know that he or she is affected by the disease. Another approach is direct recruitment by the investigator through letters or telephone calls to individuals whose identity is supplied by the proband. Direct recruitment by the investigator may, however, be seen as an invasion of privacy by family members. (Similar issues arise in epidemiologic research. See Guidebook Chapter 4, Section E.) A third approach is to recruit participants through support groups or lay organizations. Adopting this strategy requires investigator and IRB confidence that these organizations will be as scrupulous in their own efforts to protect subjects as the investigator would be. A fourth possibility is to contact individuals through their personal physicians. Prospective subjects contacted by their physician may, however, feel that their health care will be compromised if they do not agree to participate. In the end, the IRB must ensure that the recruitment plan minimizes the possibility of coercion or undue influence [Federal Policy §___.116].

In contrast to inappropriate pressure placed on prospective participants to join the study is the possibility that a subject may agree to participate out of a misguided effort to obtain therapy. The purposes of the research and how subjects will or will not benefit by participation must be clearly explained. (See discussion below on informed consent).

Investigators and IRBs need to consider each of these concerns in arriving at a recruitment strategy that protects these various interests.

Defining Risks and Benefits. Potential risks and benefits should be discussed thoroughly with prospective subjects. In genetic research, the primary risks, outside of gene therapy, are psychological and social (referred to generally as "psychosocial") rather than physical. IRBs should review genetic research with such risks in mind.

Psychological risk includes the risk of harm from learning genetic information about oneself (e.g., that one is affected by a genetic disorder that has not yet manifested itself). Complicating the communication of genetic information is that often the information is limited to probabilities. Furthermore, the development of genetic data carries with it a margin of error; some information communicated to subjects will, in the end, prove to be wrong. In either event, participants are subjected to the stress of receiving such information. For example, researchers involved in developing presymptomatic tests for Huntington Disease (HD) have been concerned that the emotional impact of learning the results may lead some subjects to attempt suicide. They have therefore asked whether prospective participants should be screened for emotional stability prior to acceptance into a research protocol.

Note that these same disclosures of information can also be beneficial. One of the primary benefits of participation in genetic research is that the receipt of genetic information, however imperfect, can reduce uncertainty about whether participants will likely develop a disease that runs in their family (and possibly whether they have passed the gene along to their children). Where subjects learn that they will likely develop or pass along the disease, they might better plan for the future.

To minimize the psychological harms presented by pedigree research, IRBs should make sure that investigators will provide for adequate counseling to subjects on the meaning of the genetic information they receive. Genetic counseling is not a simple matter and must be done by persons qualified and experienced in communicating the meaning of genetic information to persons participating in genetic research or persons who seek genetic testing.

Social risks include stigmatization, discrimination, labelling, and potential loss of or difficulty in obtaining employment or insurance. Changes in familial relationships are also social ramifications of genetic research. For example, an employer who knew that an employee had an 80 percent chance of developing HD in her 40s might deny her promotion opportunities on the calculation that their investment in training would be better spent on someone without this known likelihood. Of course, the company may be acting irrationally (the other candidate might be hit by a car the next day, or have some totally unknown predisposition to debilitating disease), but the risk for our subject of developing HD is real, nonetheless. One problem with allowing third-parties access to genetic information is the likelihood that information, poorly understood, will be misused. Likewise, an insurer with access to genetic information may be likely to deny coverage to applicants when risk of disease is in an unfavorable balance. Insuring against uncertain risks is what insurance companies do; when the likelihood of disease becomes more certain, they may refuse to accept the applicant's "bet."

Debate about the social policy implications of genetic information is vitally important and is occurring on a national and international level, but is not literally a concern for IRBs. The IRB's concern is, first, to ensure that these risks will be disclosed to subjects, and, second, to protect subjects against unwarranted disclosures of information.

See also Guidebook Chapter 3, Section A, "Risk/Benefit Analysis," and Chapter 3, Section B, "Informed Consent," for further discussion of these issues.

Privacy and Confidentiality Protections. Special privacy and confidentiality concerns arise in genetic family studies because of the special relationship between the participants. IRBs should keep in mind that within families, each person is an individual who deserves to have information about him- or herself kept confidential. Family members are not entitled to each other's diagnoses. Before revealing medical or personal information about individuals to other family members, investigators must obtain the consent of the individual.

Another problem that arises in genetic family studies that is also common in other areas of research involving interviews with subjects is the provision by a subject of information about another person. In pedigree studies, for example, the proband or other family member is usually asked to provide information about other members of the family. The ethical question presented by this practice is whether that information can become part of the study without the consent of the person about whom the data pertains. While no consensus on this issue has yet been reached, IRBs may consider collection of data in this manner acceptable, depending on the nature of the risks and sensitivities involved. It may be helpful, for example, to draw a distinction between information about others provided by a subject that is also available to the investigator through public sources (e.g., family names and addresses) and other personal information that is not available through public sources (e.g., information about medical conditions or adoptions).

IRBs should require investigators to establish ahead of time what information will be revealed to whom and under what circumstances, and to communicate these conditions to subjects in clear language. For example, if the pedigree is revealed to the study participants, family members will learn not only about themselves but about each other. The possibility that family members who did not participate might also learn of the pedigree data should not be overlooked. Subjects should know and agree ahead of time to what they might learn (and what they will not learn), both about themselves and others, and what others might learn about them. One approach would be never to reveal the pedigree to participating subjects. Many investigators record their pedigrees using code numbers rather than names. IRBs should note, however, that when a study involves a rare disease or a "known" family, the substitution of numbers for names does not eliminate the problem.

Even where the protocol calls for providing certain information to subjects, participants in genetic studies should be given the option of not receiving genetic information about themselves or others that they do not wish to receive. In genetic research, the potential for psychosocial harm accruing to persons who express a desire not to receive information gained through the study and the uncertainties surrounding the disease-predictive value of the early phases of contemporary genetic research is felt to outweigh benefits of required disclosure. (A possible exception involving circumstances where early treatment of genetically-linked disease improves prognoses is discussed in the section on identifying and deciphering genes, below.)

Data must be stored in such a manner that does not directly identify individuals. In general, except where directly authorized by individual subjects, data may not be released to anyone other than the subject. An exception to requiring explicit authorization for the release of data may be secondary research use of the data, where the data are not especially sensitive and where confidentiality can be assured. IRBs should exercise their discretion in reviewing protocols that call for the secondary use of genetic data. Furthermore, when reviewing a consent documents, IRBs should note agreements made by investigators not to release information without the express consent of subjects. Subsequent requests for access to the data are subject to agreements made in the consent process. For studies involving socially sensitive traits or conditions, investigators might also consider requesting a certificate of confidentiality. [See Guidebook Chapter 3, Section D, "Privacy and Confidentiality."]

Informed Consent. The information presented to subjects in the informed consent process should be as specific as possible. Subjects should be told both the known risks, as well as the uncertainty surrounding the risks of participation. Among the uncertainties is the likelihood that useful information will result from the study (it may not). Prospective participants often come into genetic studies with unrealistic expectations of how they will benefit from the study, and without an appreciation of low-probability risks that are not well-understood by anyone. To the extent possible, unrealistic expectations should be dispelled in the informed consent process.

The provision of relevant information should take place as a thoughtful discussion with prospective subjects. Through this process, subjects should be informed:

• about the kind of information they will be provided (e.g., that they will receive only information the investigator feels is significant and reliable, or that no genetic information will be provided) and at what point in the study they will receive that information;
• that they may find out things about themselves or their family that they did not really want to know, or that they may be uncomfortable knowing;
• that information about themselves may be learned by others in their family;
• whether information they learn or information generated about them during the study may compromise their insurability;
• that actions they may take as a result of their participation may expose them to risks (e.g., submitting insurance claim forms for reimbursement for costs of genetic counseling or procedures whose costs are not covered by the protocol);
• about what assurances can be given to protect confidentiality and what lack of assurance can be given;
• about the rights they retain and the rights they must give up regarding control over what can be done with tissue they donate (e.g., blood samples);
• what the consequences of withdrawal from the study will be; and
• of any costs associated with participation (including, for example, the cost of genetic and/or psychological counseling, if those costs will not be covered by the investigator or the institution).

Information should be given to subjects in clear language, suitable to their age, cultural background, and physical and mental capabilities. Accommodations should be made for persons with learning disabilities (as distinguished from persons who suffer diminished mental capacity). The consent process should take place in the subject's native language, through an interpreter, if necessary; consent documents should be translated into the subject's native language. The IRB should satisfy itself that great care will be taken by the investigator to ensure that prospective subjects fully understand the risks and benefits involved in participation.

Disposition of DNA Samples. When tissue samples are to be collected for later DNA analysis, numerous issues must be addressed by investigators and IRBs. Primary among them are through what mechanism samples should be collected, who can have access to the samples and for what purposes, who owns the DNA, and how incorrect genetic information (due, for example, to faulty laboratory analysis) can be corrected. The American Society of Human Genetics' Ad Hoc Committee on DNA Technology has published a set of Points to Consider on DNA banking and DNA analysis (1987), with which IRBs may wish to acquaint themselves. While not all of the Society's recommendations may be directly applicable to the IRB's concerns, it is worth noting the importance the Society places on appropriate counseling and limited access to familial genotypes.

The genetic information (and tissue samples, where applicable) collected under a research protocol are of continuing importance to the families involved in the research. An important question for IRBs to consider is what will happen to the data (and samples) when funding for the research ends. Particular attention should be paid to protecting the confidentiality of the data and obtaining consent from the participants for any use of the data (and samples) that is not strictly within the original uses to which the participants agreed.

Withdrawal from Participation. Attention should be paid to subjects' rights when they decide to withdraw from participation in the study. The federal regulations clearly require that subjects be free to withdraw from participation without penalty or loss of benefits to which they are otherwise entitled [Federal Policy §___.116(a)(8)]. What the regulations do not address, however, is how to treat data or tissue samples obtained from subjects who subsequently withdraw from the study. A similar question was addressed by the California Supreme Court in the Moore case [John Moore v. The Regents of the University of California (1990)]. While Moore constitutes binding legal authority only in California and has not, as of this writing, been adopted in other jurisdictions, the court's findings may be helpful for exploring possible approaches to handling the treatment of data and tissue samples when a subject withdraws from a genetic study.

In Moore, the California Supreme Court held that cell lines established from a donated sample are not the property of the person who donated the sample. Extrapolating to the broader context of genetic research generally, the underlying principle would be that withdrawal releases the subject from providing further information or tissue samples, and perhaps requires the removal of the subject's identity from all research records, but does not require the investigator to eliminate the resulting data from the study or to destroy the cell line.

In pedigree studies, for example, investigators may respond to a request to withdraw by removing all information about that person and his or her spouse and children from the pedigree, but it is not clear that removal of the information is required by the human subjects regulations or any other legal principle.

Secondary Use of Tissue Samples. Where a new study proposes to use samples collected for a previously conducted study, IRBs should consider whether the consent given for the earlier study also applies to the new study. Where the purposes of the new study diverge significantly from the purposes of the original protocol, and where the new study depends on the familial identifiability of the samples, new consent should be obtained.

Vulnerable Populations. IRBs should ensure that the investigator conduct the research with sensitivity to the specific mental and physical manifestations of the particular disorders being investigated. Depending on the disease, and, therefore, the likely presenting population, investigators should be prepared to communicate effectively and with sensitivity with persons who have physical limitations (e.g., deafness or blindness), learning disabilities, cognitive impairments, or any other life circumstance that may affect their participation (e.g., severe pain).

The nature of genetic research raises some special concerns when the research will involve children, physically or cognitively impaired persons, older persons, or any subject population likely to have special needs. Not only must the IRB ensure that their participation is fully voluntary and informed, IRBs must also be sure to evaluate the risks and benefits of the research as they apply to these special populations. The risk of participation for an adult differs from that of children; persons who suffer from diminished mental capacities may be subject to different risks than persons who do not. If children will be involved in the research, IRBs should seriously consider consulting with experts in child development and others knowledgeable about risks to children and families. Similarly, if physically or cognitively impaired persons will be involved in the research, IRBs should consider consulting with experts who can advise them on the special concerns their participation raises. Where applicable, 45 CFR 46 Subparts B, C, and D (pertaining to women, fetuses, prisoners, and children) must be followed. [See also Guidebook Chapter 6, Section C, "Children and Minors," Section D, "Cognitively Impaired Persons," Section G, "Terminally Ill Patients," Section H, "Elderly/Aged Persons," and Section I, "Minorities."]The involvement of children in genetic research raises many concerns, including pressure brought by family members on the child to participate and the potential for harm that may result from disclosure of genetic or incidental information. Even seemingly harmless research may actually present serious risks of harm to children. For example, interviewing children for genetic research on psychological disorders, such as schizophrenia or depression, or on alcoholism may inadvertently convey information about family members (the child may well wonder why he or she is being asked about alcoholism in the family) or cause self-doubt or stigmatization on the part of the child. Furthermore, disclosures of data to third-parties may result in children being labelled or stigmatized as, for example, potential alcohol abusers. IRBs must look carefully at both the questions that will be asked of children and the information that will be directly conveyed to them to determine whether the research involves more than minimal risk. The advisability of including children in studies of untreatable, fatal disorders such as HD has been strongly questioned [MacKay (1984), p. 3].

IRBs should also consider the mental capacities of participants in genetic research. In some diseases, such as Alzheimer Disease, patients will suffer loss of mental capacity over a period of time. In addition, it is possible that a family member might be comatose or legally incompetent for reasons unrelated to the disease under study. Special attention should be paid to methods of ensuring voluntary consent by the subject or the subject's legally authorized representative [Federal Policy §§___.102(c), ___.116]. Under the regulations, a "legally authorized representative" is defined as "an individual or judicial or other body authorized under applicable law to consent on behalf of a prospective subject to the subject's participation in the procedure(s) involved in the research" [Federal Policy §§___.102(c)] IRBs should pay particular attention to state and local laws relating to persons authorized to give permission for participation in research on behalf of prospective subjects, noting that such "proxy" consent to participation in research that does not involve a direct medical benefit may differ from consent to receive medical treatment. Where possible, the subject's assent should be sought; his or her dissent should be honored. [See also Guidebook Chapter 6, Section D, "Cognitively Impaired."]

In appropriate circumstances the IRB might consider granting waivers of consent or alteration of the consent process. [See MacKay (1984), pp. 3-4, and Levine (1986).] The federal regulations allow for waivers or alterations in the consent process where the IRB finds that: (1) the research involves no more than minimal risk; (2) the waiver or alteration will not adversely affect the rights and welfare of the subject; (3) the research could not practicably be carried out without the waiver or alteration; and (4) whenever appropriate, the subjects will be provided with additional pertinent information after participation [Federal Policy §___.116(d)]. Again, IRBs should carefully consider whether the research qualifies as "minimal risk."

Publication Practices. One final issue involving consent is the publication of research data. The publication of pedigrees can easily result in the identification of study participants. Where a risk of identification exists, participants must consent, in writing, to the release of personal information. Various authors have noted the problem of obtaining consent for the publication of identifying data, and have recommended that consent to the publication be obtained immediately prior to the publication, rather than as part of the consent to treatment or participation in research. [See, e.g., Rost and Cohen (1976) and Murray and Pagon (1984).] It is worth noting, however, that to address this concern, IRBs must also resolve the following questions: Who determines the risk of identification, and on what grounds? Who are defined as participants (is it everyone listed in the pedigree, some of whom have been contacted by investigators, some of whom have had information about them provided by a family member)?

While IRBs must be careful to avoid inappropriate restrictions on investigators' research publications, some evaluation of publication plans is important as part of the IRB's overall interest in preserving the confidentiality of research subjects. One approach for investigators to use in evaluating their publication plans might be to work in a step-wise fashion: First, is publication of the pedigree essential? If publication of the pedigree or other identifying data (e.g., case histories, photographs, or radiographs) is essential, can some identifying data be omitted without changing the scientific message? (The practice of altering data — such as changing the birth order and gender — is controversial, and no clear professional consensus yet exists as to whether this is an appropriate practice.) Finally, if the pedigree must be published, and if identifying data cannot be omitted in an appropriate manner without changing the scientific message, subjects must give their permission for publication of data that may reveal their identity.

Another concern about publication is the potential for publicity of the research results, and the exposure of participants to such publicity. Consent by individuals to such publicity does not resolve the question. Because genetic research involves families, the agreement of one subject to participate in releases of information to the media (including interviews and the like) has significant implications for other members of the family, particularly where the research is of a sensitive nature. IRBs should ensure that the investigator has addressed this possibility.

Expedited Review and Exemption from Review. The expedited review process is available for minimal risk research where the research activity is limited to one of a specified category (as published in the Federal Register), including the provision of blood samples [Federal Policy §___.110; Federal Register 46 (January 26, 1981): 8392]. In genetic studies that involve a blood draw, the additional psychosocial risks are likely to raise the risk beyond the "minimal risk" level allowable for expedited review. When an expedited review is requested, IRBs should review the question of minimal risk carefully.

With respect to exemption from review, the development of a pedigree through interviews with family members is likely to create identifying information, even where individuals will not be identified. Such research would not, therefore, qualify for exemption from review under the federal regulations [Federal Policy §___.101(b)(2)].

Identifying and Deciphering Genes

Research focusing on identifying the specific genetic component of a particular disease, condition, or characteristic relies upon DNA analysis of tissue samples taken from the members of families in which the condition appears. Many issues raised by pedigree analysis are relevant to this stage of research as well: pressure or coercion in recruiting subjects; informing prospective subjects of the possible harms; minimizing psychological harm through counseling and education; protection of confidentiality (which is particularly problematic when family members constitute the subject population); control over the use of DNA tissue samples; and protecting particularly vulnerable persons, all of which were discussed in the previous section. Additional issues include: determining when the data constitute "information;" additional risks presented by this stage of research (e.g., the possibility of incidental findings); and possible conflicts between subjects' rights and investigators' duties with respect to revealing the results of the study to subjects [i.e., telling subjects whether they (or their relatives) carry the defect, and the meaning of their status with respect to the likelihood of suffering from the disease or passing it along to their children].

Access to Data: Interim Findings. An issue that must be resolved prior to beginning any genetic study is who will have access to the data and the stage in the research at which they will have access. The issue of information transfer is vitally important in all genetic research, but particularly in the first three stages of investigation. A crucial question investigators and IRBs must address is whether (and which) interim findings will be communicated to subjects.

Experts disagree about whether interim or inconclusive findings should be communicated to subjects, although most agree that they should not (that only confirmed, reliable findings constitute "information"). Persons who oppose revealing interim findings argue that the harms that could result from revealing preliminary data whose interpretation changes when more precise or reliable data become available are serious, including anxiety or irrational — and possibly harmful — medical interventions. They argue that such harms are avoidable by controlling the flow of information to subjects and limiting communications to those that constitute reliable information. MacKay (1984), writing about the development of genetic tests, argues against revealing interim findings, contending that preliminary results do not yet constitute "information" since "until an initial finding is confirmed, there is no reliable information" to communicate to subjects, and that "even...confirmed findings may have some unforseen limitations" [p. 3]. He argues that subjects should not be given information about their individual test results until the findings have been confirmed through the "development of a reliable, accurate, safe and valid presymptomatic test" [pp. 2-3; see also Fost and Farrell (1990)]. Others have argued that all interim results should be shared with subjects, based on the principle of autonomy — that subjects have a right to know what has been learned about them.

These arguments are equally relevant at any of the first three stages of genetic research. IRBs should consider these arguments, weighing the possible harms and benefits. Investigators should determine, prior to initiation of the study, the point at which the data will be considered solid enough to be constitute information that should be provided to subjects. Investigators should further consider coding the data and separating the research records from individuals' medical records, so that neither the investigators nor the subjects may gain access to them [MacKay (1984), p. 3].

Reilly (1980) suggests that IRBs develop general policies governing the disclosure of information to subjects, to help make these determinations. He suggests that at least the following three factors be considered: "(1) the magnitude of the threat posed to the subject, (2) the accuracy with which the data predict that the threat will be realized, and (3) the possibility that action can be taken to avoid or ameliorate the potential injury" [p. 5]. IRBs should ask investigators to define three categories of disclosure: (1) "findings that are of such potential importance to the subject that they must be disclosed immediately;" (2) "data that are of importance to subjects..., but about which [the investigator] should exercise judgment about the decision to disclose....[i]n effect, these are data that trigger a duty to consider the question of disclosure;" and (3) "data that do not require special disclosure" [pp. 5, 12].

IRBs should consider whether the investigator's approach appropriately balances the risks and benefits involved in providing access to the data. Subjects should be told, as part of the consent process, whether, when, and what information they will receive. Any disclosures of genetic information should be accompanied by appropriate counseling by trained genetic counselors. However the IRB resolves this question, investigators should explain to prospective subjects the basis according to which they will decide which data will be disclosed to whom, and when those disclosures will be made.

Access to Data: The Subjects' "Right Not to Know." Subjects generally retain the right not to receive information about the results of a study that reveals their genetic status. A possible exception involves circumstances where early treatment of genetically-linked disease could improve the subject's prognosis. In such circumstances, investigators may have a duty to inform the subject about the existence of the genetic defect and to advise him or her to seek medical advice. [See, e.g., Andrews (1991).] (As of this writing, a legal duty of investigators to inform subjects about the existence of genetic defects has not been firmly established.)

Furthermore, the existence of a genetic defect that is linked to disease may have important implications for family members; can or should the confidentiality of subjects' data be compromised to allow other family members to be warned? The President's Commission (1983), addressed this question with respect to information generated from genetic screening. The Commission's discussion may also be relevant to information obtained as the result of genetic research, at stages that precede genetic screening. The Commission concluded that:

[the] ethical duty of [providing confidentiality] can be overridden only if several conditions are satisfied: (1) reasonable efforts to elicit voluntary consent to disclosure have failed; (2) there is a high probability both that harm will occur if the information is withheld and that the disclosed information will actually be used to avert harm; (3) the harm that identifiable individuals would suffer would be serious; and (4) appropriate precautions are taken to ensure that only the genetic information needed for diagnosis and/or treatment of the disease in question is disclosed [p. 44].

The Commission further advised that, to the extent possible, persons undergoing genetic screening should be asked to consent in advance to the disclosure of genetic information to relatives in the event that such useful information is discovered [pp. 43-44]. Whether a legal duty exists to warn relatives of possible genetic defects has not yet been established. [See Robertson (1992), pp. 92-94.]

Access to Data: Incidental Findings. IRBs should also ensure that investigators adequately deal with how they will handle incidental findings; that is, what will be done with genetic information that is learned during the course of the study that does not directly relate to the research. For example, in intergenerational pedigree analyses, questions of paternity or parentage can come up. DNA analysis will reveal information indicating that an individual's biological parents are not who he or she thought they were; blood typing may reveal similar information. DNA analysis may also reveal information about diseases or conditions other than the disease or condition under study. Prospective subjects should be informed during the consent process that the discovery of such information is possible. Appropriate counseling should be provided to educate subjects about the meaning of the genetic information they have received, and to assist them in coping with any psychosocial effects of participation.

Access to Data: Secondary Use. Investigators should also address secondary use of research data (e.g., by other investigators, or by themselves for different research purposes). Where secondary uses can be foreseen, consent to the use should be sought. Express consent to access to data for secondary uses should be obtained for sensitive data and for circumstances under which confidentiality cannot be assured.

Research on Genetic Testing

Testing individuals to determine the presence of genetic defects falls into four basic categories:

  • Testing newborns to detect serious genetic diseases. The screening of newborns is considered to be of value to the extent that infants can benefit from early intervention. In the case of phenylketonuria (PKU), for example, a genetic disease for which a test is available, a special diet can prevent most of the serious effects of the disease (which include brain damage).

·         Testing for carrier status to identify individuals whose genetic makeup includes a gene or a chromosome abnormality that might have serious health implications for their children. Carrier testing is usually requested by adults who have some indication that they may be carriers of a genetically-linked disorder (e.g., because they are members of an ethnic group known to have a high incidence of the disorder, because a relative has a genetic disease, or because a spouse knows that he or she is a carrier). Testing will provide such persons information about the risks of being a carrier and of passing on either the disease or abnormal genes to their children. For recessive diseases, for example, a carrier will pass on the disease to their children only if the other biological parent is also a carrier of the same defective gene.

·         Prenatal testing is aimed at detecting the presence of genetic or chromosomal abnormalities in fetuses. Examination of the genetic makeup of the fetus is done through amniocentesis, chorionic villi sampling, blood sampling from the umbilical cord and blood samples from the mother.

·         Risk assessment testing (sometimes referred to as "presymptomatic testing") determines the probability that a person will develop a genetically-linked disease at some point in the future. The degree of certainty with which risk assessment tests can predict the likelihood of disease differs depending on the disease. For some diseases the actual gene has been located, making tests more accurate than for diseases for which only a marker has been found. Further, some markers are more closely linked to the gene than are others, thereby having a more predictive quality than others.

Protocols involving genetic testing raise somewhat different issues, depending on whether the tests are under development or are already established as reliable. IRBs are concerned with research aimed at developing genetic tests.

The ethical issues raised by the various kinds of genetic testing largely concern the concept of autonomy or self-determination. Before consenting to undergo genetic tests, whether new tests that are being developed, or already-established genetic tests, subjects should fully understand what it is they are going to learn about themselves, what they are not going to learn about themselves, and how reliable the information will be. Subjects must have information on which to base their decisions whether or not to go ahead with the testing. When the research involves the development of a genetic test, however, the uncertainties involved make the consent process problematic: How does one adequately alert subjects to the psychosocial risks of testing when the point of the study is to try to help define those risks? Research on pre-test education in effect experiments with the informed consent process. Can subjects consent to research knowing that one of the risks is that they may not be adequately informed about what they are agreeing to? The federal regulations allow IRBs to approve consent procedures that do not include or that alter some or all of the elements of informed consent; one of the requirements is that the research must involve no more than minimal risk [Federal Policy §___.116(d)]. Research that involves deliberate withholding of information or deception is reviewed pursuant to those provisions. Even where it is permitted, purposeful nondisclosure of pertinent information is allowed only to the extent necessary to conduct the study (e.g., when disclosure of the information would affect the outcome of the study). Furthermore, subjects must consent to the nondisclosure; that is, they must be told that there is some relevant information about the study that they will not be told prior to consenting to participate. [See Levine (1986, p. 117) and discussion in Guidebook Chapter 3, Section B, "Informed Consent."] In genetic testing research, however, the nondisclosure is not purposeful; rather, the nature and extent of the psychosocial risks involved is simply unknown. IRBs must look carefully at such studies to ensure that subjects are adequately protected. Investigators should provide the IRB their assessment of unknown risks. Subjects should be informed, in clear, understandable language, of the possibility of undisclosed risks, including any information the investigator has about their possible nature and extent.

Research results should be communicated to the subject by someone who possesses the appropriate medical and counseling expertise with which to explain the meaning of the test results. That person should ensure that the subject comprehends the information that has been provided to him or her, regardless of the time that may be involved. Furthermore, it may be appropriate to provide counseling not just for the subjects themselves, but also for their families. Consent to involve family members, should the need arise, should be sought as part of the consent to be tested.

Smurl and Weaver (1987) have developed a set of proposed ethical guidelines for the clinical testing of risk assessment tests for HD. IRBs reviewing investigations of risk assessment genetic tests should find their recommendations helpful. Many of their recommendations follow the arguments set forth in the discussions in the Guidebook on pedigree analysis and identifying and deciphering genes.

The misuse of genetic information due to misunderstanding its meaning is a risk faced by all participants in genetic research. Investigators can minimize this risk by working to educate not only subjects, but also the medical profession and the public about genetic testing. The term "diagnostic" is often used, but the term does not really apply. Genetic tests identify risks rather than "diagnose" the presence of disease. Discrimination in employment or in obtaining insurance are two areas that are of major concern, particularly where the genetic trait is one that is thought to indicate a predisposition to diseases or conditions caused by exposure to environmental agents. Significant damage has been done by, for example, misperceptions about what it means to be a carrier of sickle cell trait. Persons who carry the sickle cell trait have been denied jobs or have been otherwise discriminated against. Education, together with protecting subjects against disclosure of genetic information, can help minimize the risk of discrimination.

Gene Therapy Research

Gene therapy attempts to treat genetic disease by altering an individual's cells. Gene therapy can involve treatment of either somatic (nonreproductive) cells or germline (reproductive) cells. Genetic changes made to somatic cells affect only the individual who has received treatment; genetic changes made to germline cells may be passed on to the patient's descendants. A distinction must be made between gene therapy designed to treat or eliminate disease or serious medical, psychological, or behavioral conditions (e.g., cystic fibrosis), and the "improvement" of human characteristics (e.g., height).

Gene therapy techniques involving somatic cells are aimed at curing genetic disease in individuals by inserting properly functioning genes into the individual's somatic cells [Walters (1989), pp. 220-221]. The approach for making genetic changes to germ line cells is to add new DNA to early embryos in an attempt to change the genes not only in the individual, but also the genes passed on to his or her progeny. Walters (1989) has described the process as follows:

In studies involving mice, for example, genes have been added to one-cell mouse embryos after the sperm had penetrated the egg but before the genetic material from the sperm and egg are joined within the same nucleus. If the experiment is successful, these added genes are then adopted by the embryo. As the embryo grows and the number of embryonic cells increases, the added genes become part of every new embryonic cell. Later, when the sperm or egg cells of the mouse develop, the added genes are included in approximately half of these reproductive cells. Thus, when the mouse reproduces, some of its progeny receive the added genes, and so on through the generations [p. 221].

After being reviewed and approved by the IRB and the local institutional biosafety committee, gene therapy protocols for research conducted at or sponsored by an institution that receives any support for recombinant DNA research from NIH must be reviewed by the Recombinant DNA Advisory Committee (RAC) at NIH. At present, the RAC will consider human somatic cell gene therapy protocols, but not germline cell gene therapy protocols. The process of review is as follows: The Human Gene Therapy Subcommittee conducts a public review of the protocol, then submits its recommendation to the RAC; if the RAC approves the protocol, it is forwarded to the director of NIH for final approval.

The RAC, through a Points to Consider Subcommittee, has established "Points to Consider in the Design and Submission of Protocols for the Transfer of Recombinant DNA into Human Subjects." Among the ethical concerns that investigators must address are subject selection, informed consent, and privacy and confidentiality. Investigators must also justify the use of recombinant DNA techniques against alternative methodologies and delineate the risks and benefits of the research. A summary of the Points to Consider follows; IRBs would be well-served to follow a similar line of inquiry when reviewing protocols that involve the transfer of recombinant DNA into human subjects.

The RAC Points to Consider. The following points should be addressed by all protocols involving somatic cell gene therapy:

A. The proposed research should be fully described.

1. Where recombinant DNA will be used for therapeutic purposes:

a. Why is the disease a good candidate for gene therapy?

b. After reviewing the natural history and range of expression of the disease (including the available objective and/or quantifiable measures of disease activity), are the usual effects of the disease predictable enough to allow for meaningful assessment of gene therapy?

c. What is the therapeutic goal of the research: to prevent all manifestations of the disease, to halt the progression of the disease after symptoms have begun to appear, or to reverse manifestations of the disease in seriously ill persons?

d. What alternative therapies exist? In what groups of patients are these therapies effective? What are their relative advantages and disadvantages as compared with the proposed gene therapy?

2. When recombinant DNA will be transferred for nontherapeutic purposes:

a. Into what cells will the recombinant DNA be transferred? Why is the transfer of recombinant DNA necessary for the proposed research? What questions can be answered by using recombinant DNA?

b. What alternative methodologies exist? What are their relative advantages and disadvantages as compared to the use of recombinant DNA?

B. The research design and anticipated risks and benefits should be described.

1. A full description of the methods and reagents to be employed for gene delivery and the rationale for their use should be provided to the IRB (a list of specific points to be addressed is provided in the Points to Consider).

2. Previous pre-clinical studies, including risk-assessment studies, that support the investigator claims about safety and effectiveness should be described. The investigator should explain why the model chosen is the most appropriate (a list of specific issues to be addressed is provided in the Points to Consider).

3. The treatment to be administered to patients and the diagnostic methods that will be used to monitor the success or failure of the treatment should be described, including any relevant previous clinical studies using similar methods that have been performed (specific issues to be addressed are provided in the Points to Consider).

4. Any potential benefits and hazards to persons other than the patients should be described.

5. The qualifications of the investigator(s) and the adequacy of the clinical facilities should be given.

C. Methods for patient selection should be described, including the numbers of patients, the recruitment procedures that will be used, the eligibility criteria that will be used (both exclusion and inclusion criteria), and how the investigator will select among eligible patients if it is not possible to include all who desire to participate.

D. Methods for obtaining informed consent should be described. Where the study involves pediatric or mentally handicapped patients, investigators should describe the procedures for seeking the permission of parents or guardians, and, where applicable, the assent of each patient (in keeping with the requirements of 45 CFR 46). In particular, investigators should address:

1. How the major points covered in A-C will be disclosed to potential participants in understandable language;

2. How the innovative character and the possible (including theoretically possible) adverse effects of the experiments will be discussed with patients; how the potential adverse effects will be compared with the consequences of the disease; and what will be said to convey that some of these adverse effects, if they occur, could be irreversible;

3. How the financial costs to the subject of the experiment and any available alternatives will be explained to the patients;

4. How patients will be informed that they may be subjected to media attention as a result of participating; and

5. How patients will be informed about the irreversible consequences of some of the procedures; about any adverse medical consequences that might occur if they withdraw from the study once it has begun; and about any preconditions to participation, such as their willingness to cooperate in long-term follow-up and autopsy in the event of a patient's death following gene transfer.

E. Measures that will be taken to protect the privacy of patients and their families and for maintaining the confidentiality of research data should be described.

IRB Considerations. The potential risks associated with gene therapy may weigh against the involvement of human subjects in gene therapy trials. As Walters (1989) has described it:

There are clearly some risks and some unknowns associated with even this simplest type of gene therapy [somatic cell gene therapy]. For example, it is not presently possible to control where the retroviral vectors will "touch down" when they reach the nuclei of the patient's...cells. In other words, currently-available vectors are unguided missiles. There is some concern among researchers that the vectors may disrupt properly-functioning genes and therefore kill some cells or, more seriously, that the vectors may activate some previously dormant cancer-causing genes. It is also possible that the domesticated retroviral vectors will recombine with other DNA or other viruses and so recapture their native capacity to produce more retroviruses and to infect large number of cells [p. 222].

IRBs need to consider the risks and benefits of gene therapy carefully, and, if a protocol is approved, ensure that subjects will be thoroughly informed of the risks and benefits involved in the procedure. It must be made clear to subjects that the investigation has both a therapeutic intent and the goal of acquiring scientific knowledge. Investigators should be careful not to raise unrealistically the hopes of the subjects and their families [Fletcher (1985) p. 298].

Protocols involving children are subject to the special provisions of 45 CFR 46 Subpart D. IRBs should pay particular attention to protecting children, including obtaining assent from child-participants, wherever possible. IRBs may want to consider using a consultant, an IRB member, or a "group consent auditor" to oversee the consent process, ensuring that the child's best interests have been carefully considered [Fletcher (1985), pp. 298-99]. Fletcher argues that IRBs should refrain from allowing the hopeless situation of subjects to overshadow the consideration of acceptable risk. He concludes that "even if the degree of risk does not approach the level of 'dangerous'...[d]esperation about [a] child's condition is not a sound premise for experimental gene therapy. Children in imminent danger of death should not be selected as subjects for the first trials" [p. 297]. [See also Guidebook Chapter 6, Section C, "Children and Minors."]

Special attention should also be paid to the possibility of mental impairment. [See Guidebook Chapter 6, Section D, "Cognitively Impaired."]

POINTS TO CONSIDER

1. Does the proposed study population comprise family members? Has the appropriateness of various strategies for recruiting subjects (e.g., recruiting by the proband or other family members, by the investigator, through support groups, or through prospective subjects' personal physicians) been considered? Does the proposed strategy for recruiting subjects sufficiently protect prospective subjects from the possibility of coercion or undue influence?

2. Does the investigator plan to use the proband or the proband's clinical medical records as a source of research data about other persons (e.g., other family members)? If so, must their consent be obtained before their data can be included, or is the permission of the person providing the information sufficient?

3. Has the investigator established clear guidelines for disclosure of information, including interim or inconclusive research results, to the subjects? Will subjects be informed, in clear language, about what information they are entitled to receive at what point in the research? Will subjects receive an explanation of the meaning of the information they receive?

4. Will family members be protected against disclosures of medical or other personal information about themselves to other family members? Will they be given the option not to receive information about themselves? Will limits on such protections be clearly communicated to subjects, including obtaining advance consent to such disclosures (e.g., when family members will be warned about health risks)?

5. Will the possible psychological and social risks of genetic research be adequately considered in the consent process? Will appropriate counseling be provided, both as part of the consent process and when communicating test or other research results to subjects?

6. Will subjects be informed about the possibility that incidental findings may be made (e.g., paternity, diseases, or conditions other than the one(s) under study)?

7. Will the data be protected from disclosure to third parties, such as employers and insurance companies? Will the data be stored in a secure manner? Will the data be coded so as to protect the identity of subjects? Is a request for a certificate of confidentiality appropriate?

8. Does the investigator plan to disclose research findings to subjects' physicians for clinical use? Are such plans appropriate? Will the possibility of such disclosures be discussed with and consented to by prospective subjects?

9. Will vulnerable populations (e.g., children, persons with impaired mental capacities) be adequately protected? Under what circumstances can a research subject serve to grant permission to involve a minor child or an incapacitated adult in a study?

10. Have adequate provisions been made for protecting against misuse of tissue samples (e.g., confidentiality, obtaining consent for any use not within the original purpose for which the samples were collected)? What agreements with subjects are necessary to use stored materials for new studies or for clinical diagnoses?

11. Have adequate provisions been made for the treatment of data and tissue samples in the event of subject withdrawal from the study?

12. Do the investigator's publication plans threaten the privacy or confidentiality of subjects? Has adequate consideration been given to ways in which subjects' privacy and confidentiality can be protected (e.g., providing for consent to publication of identifying information)?

13. Have the RAC's Points to Consider for gene therapy protocols been considered?

Some of the questions arising from the conduct of large pedigree studies were addressed at an NIH workshop jointly sponsored by the National Center for Human Genome Research (NCHGR) and the Office for Protection from Research Risks (OPRR) in October of 1992. For further information on the availability of the papers presented at the workshop (listed in "Suggestions for Further Reading," below), contact:

Dr Eric T. Juengst
National Center for Human Genome Research
National Institutes of Health
Building 38A, Room 617
Bethesda, MD 20892
Tel: (301) 402-0911

Dr. Joan P. Porter
Office for Protection from Research Risks
National Institutes of Health
Building 31, Room 5B63
Bethesda, MD 20892
Tel: (301) 496-7005

APPLICABLE LAWS AND REGULATIONS

Federal Policy for the protection of human subjects

Federal Policy §___.116 [Informed consent]

45 CFR 46 Subpart D [DHHS: Additional protections for children involved as subjects in research]

Federal, state, and local laws or regulations governing confidentiality of information

Federal, state, and local laws or regulations pertaining to insurance

There are currently no laws or regulations specifically governing the involvement of human subjects in genetic research, but the following guidelines may be useful:

U.S. Department of Health and Human Services. Public Health Service. National Institutes of Health. "Federal Guidelines for Recombinant DNA Molecule Research." 51 Federal Register (May 7, 1986): 16958.

U.S. Department of Health and Human Services. Public Health Service. National Institutes of Health. "Recombinant DNA Molecule Research, Proposed Actions under Guidelines; Notice." 50 Federal Register (August 19, 1985): 33462-33467.

U.S. Department of Health and Human Services. Public Health Service. National Institutes of Health. Recombinant DNA Advisory Committee. Points to Consider Subcommittee. "Points to Consider in the Design and Submission of Protocols for the Transfer of Recombinant DNA into Human Subjects." Recombinant DNA Technical Bulletin 12 (No. 3, September 1989): 151-170.

Return to Top of Page

I. ALCOHOL AND DRUG RESEARCH

INTRODUCTION

Alcohol and drug research focuses on use, abuse, and dependence on abuse-liable substances, and may or may not involve the administration of an abusable substance. It may seek to investigate physiological responses to alcohol or drugs, or may be aimed at the treatment of alcohol or drug abuse. Treatment protocols may be behavioral or biomedical, including the administration of medications.

DEFINITIONS

Abuse-liable: Pharmacological substances that have the potential for creating abusive dependency. Abuse-liable substances can include both illicit drugs (e.g., heroin) and licit drugs (e.g., methamphetamines).

IRB CONSIDERATIONS

Research on alcohol or drug use raises special concerns for IRBs because the research often involves the administration of abuse-liable substances. Further, subjects' capacity to provide informed consent is often compromised. Institutionalization may also have an impact on the prospective subject's ability to choose freely to participate in research.

Other issues IRBs need to consider are the selection of subjects and confidentiality. With respect to confidentiality, federal, state, and local laws regarding criminal behavior must be considered, because legal requirements may impinge on the ability of the researcher to guarantee confidentiality. Finally, researchers may face ethical problems with the study design, such as the morality of giving alcohol to alcoholics, or the problems associated with studies that include placebo arms.

The National Advisory Council on Alcohol Abuse and Alcoholism (the Council) has issued guidelines entitled Recommended Council Guidelines on Ethyl Alcohol Administration in Human Experimentation (1989). Many of the recommendations apply equally well to studies involving abuse-liable drugs. The Council's recommendations contain a series of questions and answers about research involving alcohol administration to human subjects, and should be consulted by IRBs reviewing protocols involving alcohol- or drug-related research.

Risk/Benefit. A number of issues raised by alcohol and drug research focus on considerations of risk and benefit. Where alcohol or drugs will be administered to subjects, IRBs should consider, for example, whether the subjects are drug or alcohol abusers, and whether participation of the proposed populations is likely to expose the subjects to harm [see the Recommended Council Guidelines (1989), pp. 5-6]. Investigators must adequately assess the potential for toxicity, and make provisions for clinical care of subjects where it will likely be needed. Further, the need for access to medical backup services (i.e., the presence of a nurse or physician during conduct of the research, or the availability of a physician "on call") should be considered.

Other risks presented by some drug or alcohol research are those inherent in self-administration of abuse-liable substances. IRBs should consider such risks, including the possibility that subjects may self-administer an increasing amount of the drug to levels higher than those to which they are accustomed, and the possible harms that might result.

Where the study has a placebo arm, investigators need to consider the various effects the use of placebos might have and provide mechanisms for dealing with them so that subjects are adequately protected. For example, in a study in which subjects are told that the investigational agent blocks the effect of an abuse-liable drug, a subject, believing herself not to be in the placebo arm, might self-administer sufficiently large doses of the drug to fatally overdose. Investigators should be prepared to address this issue (e.g., through informed consent, monitoring, use of an in-patient subject population, or other means).

Adequate provisions must be made to eliminate the risk of drug or alcohol impairment before the subject leaves the research site.

The Council Guidelines describe investigators' obligations to facilitate the entry of alcoholics who are current, active drinkers into treatment programs [p. 6]. The Guidelines go on to point out that where potential subjects "have completed the initial phase of treatment and progressed into rehabilitation or recovery," their involvement in research in which alcohol will be administered requires "extremely strong scientific justification and risk/benefit assessment" [p. 6]. Further, Council policy holds that "it is considered inappropriate to administer alcohol to any recovering alcoholic who is abstinent and living a sober life in the community" [p. 6].

In addition to the risk/benefit questions discussed here, the Council Guidelines also consider such issues as the age of subjects, the involvement of alcohol-naïve subjects, deception or incomplete disclosure, medical and psychological evaluation of subjects prior to participation, and follow-up of subjects.

Incentives for Participation. IRBs should consider whether any remuneration offered to subjects for their participation is appropriate. Any remuneration (e.g., money, food, lodging, or medical care) should be commensurate with the burden of participation, and should not be such that it constitutes an undue inducement or is coercive.

The possible involvement of drug or alcohol abusers in drug and alcohol research has led some investigators not to offer any remuneration to their subjects for fear of unfairly inducing their participation. Many potential participants are unemployed or otherwise economically disadvantaged, so that concern over this issue is appropriate. Nonetheless, to assume that any remuneration given to alcohol and drug abusing constitutes an undue influence is also unfair. IRBs should consider this issue carefully.

Informed Consent. In drug and alcohol research, concerns about the consent process focus on determining the competence of subjects to consent to the research and effectively communicating the information necessary to obtain informed consent. With respect to competence, IRBs should ensure that competence is assessed rather than assumed. Because there are no generally accepted standards for determining competence to consent to research, the IRB plays an important role in assessing the researcher's proposed procedures. IRBs should take an active part in helping researchers formulate appropriate criteria and procedures, taking into consideration the degree of risk presented. The same or similar considerations as those discussed Guidebook Chapter 6, Section D, "Cognitively Impaired," would apply, noting, however, that the capacity to consent to research may fluctuate, depending on the subject's state of inebriation.

The Council recommendations on competency to consent state that:

due consideration should be given to the cognitive, physiologic and motivational states of the individuals in terms of their ability to fully understand the context of the informed consent. Individuals who are severely intoxicated or in a confusional withdrawal state are unable to give true informed consent. Alternatively, a blood alcohol concentration (BAC) of zero for the potential subject may not be a required prerequisite, depending upon cognitive capabilities of the individual at that time. If there is a question of a potential subject's ability to give meaningful informed consent, an independent clinician, ethical consultant, or uninvolved third party with appropriate qualifications may be asked to evaluate this ability [p. 4].

In drug research, the lack of physical measures for levels of drugs means that investigators must rely (as must alcohol researchers, in many instances) on clinical judgments based on other indications of mental competence (e.g., through evaluating the prospective subject's ability to converse or observing his or her motor skills).

The consent document must use language that is understandable to the subject population, including ethnic sensitivities. Further, the Council states its belief that "it [is] appropriate that every informed consent form should indicate that the drug, alcohol, is a toxin and a reinforcing agent which may cause changes in behavior, including repetitive or excessive consumption. Such a statement would appropriately acknowledge that alcohol is not an innocuous substance, and that everyone who drinks alcohol is at some risk" [p.4].

Investigators should be aware of federal, state, and local laws regarding criminal behavior, and any possible reporting requirements, whether they relate to criminal activity or other issues, such as HIV serostatus (see Guidebook Chapter 5, Section F, "AIDS/HIV-Related Research" ). They should give the IRB evidence that they have considered these requirements and provide a means of dealing with them. The IRB should seek legal advice if necessary. The consent document should explain any limits on the investigator's ability to provide confidentiality of the data, including any required reporting to law enforcement or health authorities. [See also discussion of certificates of confidentiality below and in Guidebook Chapter 3, Section D, "Privacy and Confidentiality."]

Subject Selection. The question of subject selection also requires IRB attention. Drug- or alcohol-dependent individuals should not be taken advantage of. As the Council states, researchers must "avoid using subjects merely because of their easy availability, low social or economic status, or limited capacity to understand the nature of the research." Furthermore, IRBs should ensure that the proposed subject population is appropriate in terms of "age, sex, familial or genetic background, prior alcohol use, other drug use, and general medical and psychological condition, including, if appropriate, alcoholism recovery status." IRBs should consult the Council's recommendations, which describe these issues in greater detail.

IRBs must take into consideration the fact that the subject population of alcohol abusers and users of illicit drugs might include a significant number of adolescents. The protocol must address this issue. If subjects who are not adults participate, the additional protections for of 45 CFR 46 Subpart D apply. [See Guidebook Chapter 6, Section C, "Children and Minors."]

Privacy and Confidentiality. Records indicating alcohol abuse or illicit drug use are of an obviously sensitive nature, and must be provided appropriate confidentiality. Methods for assuring adequate protection of the privacy of subjects and the confidentiality of the information gathered about them (including the fact of participation in a drug or alcohol treatment program) should also be described by the investigator. Individually identifiable information that is "sensitive" should be safeguarded; requests for the release of such information to others, for research or auditing, should be allowed only when continued confidentiality is guaranteed.

To protect data against compelled disclosure, investigators may request a certificate of confidentiality from an appropriate federal official [42 CFR 2 and 2A]. For example, the directors of the National Institute of Mental Health, the National Institute on Alcohol Abuse and Alcoholism and the National Institute on Drug Abuse are authorized to grant such protection for research on mental disorders or the use and effects of alcohol and other psychoactive drugs.

IRBs and investigators should note, however, that certificates of confidentiality protect research data from compelled disclosure; they do not cover voluntary disclosures (e.g., reporting of communicable diseases or suspected child abuse). The consent document should not, therefore, promise that "no information will ever be released," but should clearly spell out what can and cannot be released.

For information on certificates of confidentiality, contact:

National Institute on Alcohol Abuse and Alcoholism

Dr. Fulton Caldwell
National Institute on Alcohol Abuse and Alcoholism
16C-05 Parklawn Building
5600 Fishers Lane
Rockville, MD 20850
Tel: (301) 443-0796

National Institute on Drug Abuse

Ms. Jacqueline R. Porter
National Institute on Drug Abuse
10-42 Parklawn Building
5600 Fishers Lane
Rockville, MD 20850
Tel: (301) 443-2755

Cmdr. Lura S. Oravec
National Institute on Drug Abuse
5600 Fishers Lane, Room 10A-55
Rockville, MD 20850
Tel: (301) 443-6071

National Institute of Mental Health

Dr. Anthony Pollitt
National Institute of Mental Health
9-97 Parklawn Building
5600 Fishers Lane
Rockville, MD 20850
Tel: (301) 443-4673

Other health research

Mr. John P. Fanning
Office of Health Planning and Evaluation
Public Health Service
740G Humphrey Building
U.S. Department of Health and Human Services
Washington, DC 20201
Tel: (202) 690-7911
Fax: (202) 690-6603
BITNET: JFANNING%PHSHHH7@NIHCU
INTERNET: JFANNING%PHSHHH7@CU.NIH.GOV

For further discussion of certificates of confidentiality, see Guidebook Chapter 3, Section D, "Privacy and Confidentiality."

IRB Membership. IRBs that regularly review research involving vulnerable subjects are required by DHHS and FDA regulations to consider including among their members one or more individuals who are knowledgeable about and experienced in working with those subjects [45 CFR 46.107; 21 CFR 56.107]. In addition, the IRB must be sure that additional safeguards are in place to protect the rights and welfare of these subjects [45 CFR 46.111(b);  21 CFR 56.111(b)].

For Further Information. Investigators and IRB members wishing to discuss drug research involving human subjects should contact:

Cmdr. Lura S. Oravec
National Institute on Drug Abuse
5600 Fishers Lane, Room 10A-55
Rockville, MD 20850
Tel: (301) 443-6071

 

POINTS TO CONSIDER

1. Does the IRB's membership include sufficient expertise for reviewing the protocol?

2. Will the subject's drug or alcohol dependency create a deficient mental status which will preclude the subject's continuing ability to consent to participation in research? Will prospective subjects be in either a state of intoxication or withdrawal when approached to consent to participation? What mechanisms are proposed for evaluating subjects' competence to consent? Are they adequate?

3. Have additional safeguards been implemented to minimize risks (e.g., pregnancy tests or procedures for ensuring that subjects cannot leave the research site while intoxicated)?

4. Are there federal, state, or local laws regarding criminal behavior or reporting requirements that must be considered? How will they be dealt with? Will prospective subjects be informed of any reporting requirements?

5. Have the investigators provided for maintaining subjects' privacy and confidentiality? Should the investigators obtain a certificate of confidentiality to protect against compelled disclosure of their data?

6. Are adolescents potential participants? Have the additional requirements of 45 CFR 46 Subpart D been met? [See Guidebook Chapter 6, Section C, "Children and Minors."]

APPLICABLE LAWS AND REGULATIONS

Federal Policy for the protection of human subjects

Federal Policy §___.116 [Informed consent]

21 CFR 50.20 and 50.25 [FDA: Informed consent]

45 CFR 46 Subpart D [DHHS: Additional protections for children involved as subjects in research]

Federal, state, and local laws governing disclosure or reporting of criminal behavior (e.g., use of illicit drugs)

Federal, state, and local laws governing confidentiality of information

Return to Top of Page

SUGGESTIONS FOR FURTHER READING

A. Introduction

  • Brett, A., and Grodin, M. "Ethical Aspects of Human Experimentation in Health Services Research." Journal of the American Medical Association 265 (No. 14, April 10, 1991): 1854-1857.
  • Daedalus: Proceedings of the American Academy of Arts and Sciences 107 (No. 2, Spring 1978). "Limits of Scientific Inquiry." Special issue devoted to this theme.
  • Drew, Clifford J., and Hardman, Michael L. "Ethical Issues in Conducting Research." In Designing and Conducting Behavioral Research, edited by Clifford J. Drew and Michael L. Hardman, pp. 29-48. New York: Pergamon Press, 1985.
  • Fredrickson, Donald S. "Biomedical Research in the 1980s." New England Journal of Medicine 304 (No. 9, February 26, 1981): 509-517.
  • Imber, Stanley D. et al. "Ethical Issues in Psychotherapy Research: Problems in a Collaborative Clinical Trials Study." American Psychologist 41 (No. 2, February 1986): 137-146.
  • Kelman, Herbert C. "Research, Behavioral." In Encyclopedia of Bioethics (Vol. 4), edited by Warren T. Reich, pp. 1470-1481. New York: The Free Press, 1978.
  • Kravitz, Richard. "Serving Several Masters: Conflicting Responsibilities in Health Services Research." Journal of General Internal Medicine 5 (No. 2, March/April 1990): 170-174.
  • Lind, Stuart E. "The Institutional Review Board: An Evolving Ethics Committee." The Journal of Clinical Ethics 3 (No. 4, Winter 1992): 278-282. See also commentary in the same issue by Troyen A. Brennan ("Researcher as Witness," dealing with health services research, pp. 308-309).
  • Macklin, Ruth. "The Paradoxical Case of Payment as Benefit to Subjects." IRB 11 (No. 6, November/December 1989): 1-3.
  • Mahler, D. Mark. "When to Obtain Informed Consent in Behavioral Research: A Study of Mother-Infant Bonding." IRB 8 (No. 3, May/June 1986): 7-11.
  • Marshall, Patricia A. "Research Ethics in Applied Anthropology." IRB 14 (No. 6, November/December 1992): 1-5.
  • Robertson, John A. "The Scientist's Right to Research: A Constitutional Analysis." Southern California Law Review 51 (1978): 1203-1279.
  • Sieber, Joan E. "On Studying the Powerful (or Fearing to Do So): A Vital Role for IRBs." IRB 11 (No. 5, September/October 1989): 1-6.
  • Sieber, Joan E., and Stanley, Barbara. "Ethical and Professional Dimensions of Socially Sensitive Research." American Psychologist 43 (No. 1, January 1988): 49-55.
  • Sieber, Joan E., ed. The Ethics of Social Research: Surveys and Experiments. New York: Springer-Verlag, 1982.
  • Sieber, Joan E., ed. The Ethics of Social Research: Fieldwork, Regulation, and Publication. New York: Springer-Verlag, 1982.
  • Stanley, Barbara. "Ethical Considerations in Biological Research on Suicide." In Psychobiology of Suicidal Behavior, edited by J. Mann and M. Stanley, pp. 42-46. New York: New York Academy of Sciences, 1986.
  • U.S. Congress. Office of Technology Assessment. Assessing the Efficacy and Safety of Medical Technologies. Washington, D.C.: U.S. Government Printing Office, 1978.
  • U.S. National Academy of Sciences. Behavioral and Social Science Research: A National Resource. Washington: National Academy Press, 1982.
  • VandenBos, Gary R., Guest Editor. "Psychotherapy Research." American Psychologist 41 (No. 2, February 1986). Entire volume is devoted to this topic.
  • Webb, Eugene J.; Campbell, Donald T.; Schwartz, Richard D.; and Sechrest, Lee. Unobtrusive Measures: Nonreactive Research in the Social Sciences. Chicago: Rand McNally, 1966.

B. Drug Trials

  • Cowan, Dale H. "Scientific Design, Ethics and Monitoring: IRB Review of Randomized Clinical Trials." IRB 2 (No. 9, November 1980): 1-4.
  • Curran, William J. Rights and Responsibilities in Drug Research. Washington: Medicine in the Public Interest, Inc., 1977.
  • Friedman, Lawrence, and DeMets, David. "The Data Monitoring Committee: How It Operates and Why," IRB 2 (No. 4, April 1981): 6-8.
  • Friedman, L.F.; Furberg, C.; and DeMets, D. Fundamentals of Clinical Trials. Boston: Wright-PSG, 1981.
  • Nightingale, Stuart. "Drug Regulation and Policy Formulation." Milbank Memorial Fund Quarterly 59 (No. 3, Summer 1981): 412-444.
  • Ruse, Michael, and De Vries, Andre. "At What Level of Statistical Uncertainty Ought a Random Clinical Trial to be Interrupted?" In The Use of Human Beings in Research, edited by Stuart F. Spicker, Ilai Alan, Andre de Vries, and H. Tristam Engelhardt, Jr., pp. 189-225. Boston: Kluwer Academic, 1988.
  • Sheps, Mindel C. "Problems in Clinical Evaluation of Drug Therapy." Perspectives in Biology and Medicine 5 (No. 3, Spring 1962): 308-323.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. "Guideline for the Monitoring of Clinical Investigations," January 1988.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. General Consideration for the Clinical Evaluation of Drugs. Washington, D.C.: U.S. Government Printing Office, 1977. HEW Publication No. (FDA) 77-3040. (Under revision.)

C. Vaccine Trials

  • Bjune, Gunnar, and Arnesen, Øyvind. "Problems Related to Informed Consent from Young Teenagers Participating in Efficacy Testing of a New Vaccine." IRB 14 (No. 5, September/October 1992): 6-9.
  • Bjune, Gunnar, and Gedde-Dahl, Truls W. "Some Problems Related to Risk-Benefit Assessments in Clinical Testing of New Vaccines." IRB 15 (No. 1, January/February 1993): 1-5.
  • Porter, Joan P.; Glass, Marta J.; and Koff, Wayne C. "Ethical Considerations in AIDS Vaccine Testing." IRB 11 (No. 3, May/June 1989): 1-4.
  • Sawyer, Leigh A.; Katzenstein, David A.; and Quinnan, Gerald V. "Regulatory Concerns Regarding AIDS Vaccine Development." AIDS and Public Policy Journal 3 (No. 3, Summer 1988): 36-45.
  • U.S. Congress. Office of Technology Assessment. A Review of Selected Federal Vaccine and Immunization Policies. Washington, D.C.: U.S. Government Printing Office, 1979.
  • U.S. Congress. Office of Technology Assessment. Compensation for Vaccine-Related Injuries. Washington, D.C.: U.S. Government Printing Office, 1980.
  • U.S. Department of Health, Education and Welfare. Public Health Service. National Center for Health Services Research. Office of the Assistant Secretary for Health. Estimated Economic Costs of Selected Medical Events Known or Suspected to be Related to the Administration of Common Vaccines: Final Report. Washington, D.C.: U.S. Government Printing Office, 1979.
  • U.S. Department of Health, Education and Welfare. National Immunization Work Groups. Reports and Recommendations. McLean, Va.: J.R.B. Associates, 1977.
  • U.S. Department of Health, Education and Welfare. Centers for Disease Control. Immunization Practices Advisory Committee (ACIP). "Recommendations on Poliomyelitis Prevention." Morbidity and Mortality Weekly Reports, November 22, 1979, p. 510.
  • Voller, A., and Friedman, H. New Trends and Developments in Vaccines. Baltimore, MD: University Park Press, 1978.
  • Weibel, R.E. et al. "Studies in Human Subjects of Polyvalent Pneumococcal Vaccines." Proceedings of the Society for Experimental Biology and Medicine. 156 (No. 1, October 1977): 144-150.

D. Medical Devices

  • Annas, George J. "Made in the U.S.A.: Legal and Ethical Issues in Artificial Heart Experimentation." Law, Medicine and Health Care 14 (Nos. 3-4, September 1986): 164-171.
  • Heath, Erica. "Information Sought about 'Significant Risk' Decisions." IRB 3 (No. 2, February 1981): 11.
  • Holder, Angela. "The FDA's Final Regulations: IRBs and Medical Devices." IRB 2 (No. 6, June/July 1980): 1-4.
  • Kennedy, Robert S. "Clinical Investigations With Medical Devices." Journal of the American Medical Association 245 (No. 20, May 22/29, 1980): 2052-2055.
  • Kyper, C. "The 510(k) Route to the Marketplace." MD&MI (January 1982).
  • Ryan, Mary Kay. "Research Involving Medical Devices." In Human Subjects Research: A Handbook for Institutional Review Boards, edited by Robert A. Greenwald, Mary Kay Ryan, and James E. Mulvihill, pp. 107-123. New York: Plenum Press, 1982.
  • Swazey, Judith P.; Watkins, Judith C.; and Fox, Renee. "Assessing the Artificial Heart: The Clinical Moratorium Revisited." International Journal of Technology Assessment in Health Care 2 (No. 3, July 1986): 387-410.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. Bureau of Medical Devices. Investigational Device Exemptions Manual. Washington, D.C.: U.S. Government Printing Office, June 1992. DHHS Publication No. (FDA) 92-4159.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. Center for Devices and Radiological Health. "Highlights of the Safe Medical Devices Act of 1990." August 1991 (mimeo).
  • U.S. Department of Health, Education and Welfare. Food and Drug Administration. Bureau of Medical Devices. All You Ever Wanted to Know About Medical Devices But Were Afraid to Ask, 3d ed. Washington, D.C.: U.S. Government Printing Office, August 1990. DHHS Publication No. (FDA) 90-4173.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. "Guidance to Significant and Nonsignificant Risk Device Studies." FDA IRB Information Sheets, February 1989.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. "IRBs and Medical Devices." FDA IRB Information Sheets, February 1989.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. Center for Devices and Radiological Health. An Introduction to Medical Device Regulations. Washington, D.C.: U.S. Government Printing Office, 1987. DHHS Publication No. (FDA) 87-4222.
  • U.S. Department of Health and Human Services. Public Health Service. Food and Drug Administration. Center for Devices and Radiological Health. Office of Device Evaluation. Guidance Document on Significant and Nonsignificant Risk Device Studies. Washington, D.C.: U.S. Government Printing Office, 1986.

E. Use of Radioactive Materials and X-Rays

  • Alazracki, Naomi. "Evaluating Risk from Radiation for Research Subjects." IRB 4 (No. 1, January 1982): 1-3.
  • Bolsen, Barbara. "CT Scanning of the Brain: A Revolution in Only Eight Years." Journal of the American Medical Association 246 (No. 23, December 11, 1981): 2667-2669, 2675.
  • Crawford-Brown, D.J. "Truth and Meaning in the Determination of Radiogenic Risk." IRB 5 (No. 5, September/October 1983): 1-5, 12.
  • Huda, Walter, and Scrimger, John W. "Irradiation of Volunteers in Nuclear Medicine." The Journal of Nuclear Medicine 30 (No. 2, February 1989): 260-264. Comment in Journal of Nuclear Medicine 30 (No. 12, December 1989): 2062-2063.
  • Lyman, James D. Nuclear Terms: A Glossary, 2d ed. Oak Ridge, Tenn.: U.S. Atomic Energy Commission, Office of Information Services, 1974.
  • Masse, Francis X., and Miller, Tracy. "Exposure to Radiation and Informed Consent." IRB 7 (No. 4, July/August 1985): 1-4. Includes description of the biological effects of ionizing radiation and examples of radiation risk statements appropriate for inclusion in the risk section of informed consent statements.
  • U.S. Congress. House. Committee on Energy and Commerce. Subcommittee on Energy Conservation and Power. American Nuclear Guinea Pigs: Three Decades of Radiation Experiments on U.S. Citizens. [Report, Committee Print.] Washington: U.S. Government Printing Office, November 1986.
  • U.S. National Research Council. Committee on the Biological Effects of Ionizing Radiations (BEIR III). "The Effects on Populations of Exposure to Low Levels of Ionizing Radiation," Washington, D.C.: National Academy Press, 1980.
  • Veatch, Robert M. "The Ethics of Research Involving Radiation." IRB 4 (No. 1, January 1982): 3-5. Reprinted in The Patient as Partner: A Theory of Human-Experimentation Ethics, Chapter 18. Bloomington and Indianapolis: Indiana University Press, 1987.

F. AIDS/HIV-Related Research

  • Ackerman, Terrence F. "Protectionism and the New Research Imperative in Pediatric AIDS." IRB 12 (No. 5, September/October 1990): 1-5.
  • American Psychological Association. Committee for the Protection of Human Participants in Research. "Ethical Issues in Psychological Research on AIDS." IRB 8 (No. 4, July/August 1986): 8-10.
  • Arvino, Andrew, and Lo, Bernard. "To Tell or Not to Tell: The Ethical Dilemmas of HIV Test Notification in Epidemiologic Research." American Journal of Public Health 79 (No. 11, 1989): 1544-1548.
  • Barry, Michele. "Ethical Dilemmas with Economic Studies in Less-Developed Countries: AIDS Research Trials." IRB 13 (No. 4, July/August 1991): 8-9.
  • Bayer, Ronald. "The Ethics of Research on HIV/AIDS in Community-Based Settings." AIDS 4 (No. 12, December 1990): 1287-1288.
  • Bayer, Ronald; Levine, Carol; and Murray, Thomas H. "Guidelines for Confidentiality in Research on AIDS." IRB 6 (No. 6, November/December 1984): 1-9.
  • Byer, David P. et al. "Design Considerations for AIDS Trials." New England Journal of Medicine 323 (No. 19, November 8, 1990): 1343-1348.
  • Carter, Michele; McCarthy, Charles R.; and Wichman, Alison. "Regulating AIDS Research on Infants and Children: The Moral Task of Institutional Review Boards." Bridges 2 (No. 1/2, Spring/Summer 1990): 63-73.
  • Christakis, Nicholas A.; Lynn, Lorna A.; and Castelo, Aduato. "Case Study — Clinical AIDS Research that Evaluates Cost Effectiveness in the Developing World." IRB 13 (No. 4, July/August 1991): 6-7.
  • Christakis, Nicholas A. "The Ethical Design of an AIDS Vaccine Trial in Africa." Hastings Center Report 18 (No. 3, June/July 1988): 31-37.
  • Corey, Lawrence, and Fleming, Thomas R. "Treatment of HIV Infection — Progress in Perspective." New England Journal of Medicine 326 (No. 7, February 13, 1992): 484-486.
  • Culliton, B.J. "Human Experimentation: AIDS Trials Questioned." Nature 350 (No. 6316, March 28, 1991): 263.
  • Curran, William J. "AIDS Research and 'The Window of Opportunity.'" New England Journal of Medicine 312 (No. 14, April 4, 1985): 903-904.
  • DeJarlais, Don C., and Friedman, Samuel R. "AIDS Prevention Among IV Drug Users: Potential Conflicts Between Research Design and Ethics." IRB 9 (No.1, January/February 1987): 6-8.
  • Dubler, Nancy Neveloff, and Sidel, Victor W. "On Research on HIV Infection in Correctional Institutions." Milbank Quarterly 67 (No. 2, 1989): 171-207.
  • Dubler, Nancy Neveloff. "Treating Research Subjects Fairly." IRB 8 (No. 5, September/October 1986): 7-9.
  • Freedman, Benjamin. "Placebo-Controlled Trials and the Logic of Clinical Purpose." IRB 12 (No. 6, November/December 1990):1-6.
  • Freedman, Benjamin. "Equipoise and the Ethics of Clinical Research." New England Journal of Medicine 317 (July 16, 1987): 141-145.
  • Gray, Joni N., and Melton, Gary B. "The Law and Ethics of Psychosocial Research on AIDS." Nebraska Law Review 64 (No. 4, 1985): 637-688.
  • Hamburg, Margaret A., and Fauci, Anthony S. "AIDS: The Challenge to Biomedical Research." Daedalus 18 (Spring 1989): 19-39.
  • Jones, James H. "The Tuskegee Legacy: AIDS and the Black Community." The Hastings Center Report 22 (No. 6, November/December 1992): 38-40.
  • Jonsen, A.R. "The Ethics of Using Human Volunteers for High-Risk Research." Journal of Infectious Diseases 160 (No. 2, August 1989): 205-208. Comment by Tacket and Edelman, "Ethical Issues Involving Volunteers in AIDS Vaccine Trials," Journal of Infectious Diseases 161 (No. 2, February 1990): 356.
  • Landesman, Sheldon M. "Against the Option Not to be Informed of HIV Status." IRB 8 (No. 5, September/October 1986): 9.
  • Leikin, Sanford L. "Immunodeficiency Virus Infection, Adolescents, and the Institutional Review Board." Journal of Adolescent Health Care 10 (November 1989): 500-505.
  • Levine, Carol. "Women and HIV/AIDS Research: The Barriers to Equity." IRB 13 (No. 1-2, January-April 1991): 1-17.
  • Levine, Carol. "Children in HIV/AIDS Clinical Trials: Still Vulnerable after All These Years." Law, Medicine and Health Care 19 (No. 3-4, Fall/Winter 1991): 231-37.
  • Levine, Carol. "Has AIDS Changed the Ethics of Human Subjects Research?" Law, Medicine and Health Care 16 (No. 3-4, Fall/Winter 1988): 167-173.
  • Levine, Carol; Dubler, Nancy Neveloff; and Levine, Robert J. "Building a New Consensus: Ethical Principles and Policies for Clinical Research on HIV/AIDS." IRB 13 (No. 1-2, January/February 1991):1-17.
  • Levine, Robert J. "The Use of Placebos in Randomized Clinical Trials." IRB 7 (No. 2, March/April 1985): 1-4. Comment by Arthur Schafer, p. 4.
  • Macklin, Ruth, and Friedland, Gerald. "AIDS Research: The Ethics of Clinical Trials." Law, Medicine and Health Care 14 (No. 5-6, December 1986): 273-280.
  • Mariner, Wendy K. "New FDA Drug Approval Policies and HIV Vaccine Development." American Journal of Public Health 80 (No. 3, March 1990): 336-341.
  • Martin, Judith M., and Sacks, Henry S. "Do HIV-Infected Children in Foster Care Have Access to Clinical Trials of New Treatments?" AIDS and Public Policy Journal 5 (No. 1, Winter 1990): 3-8.
  • Melton, Gary B. et al. "Community Consultation in Socially Sensitive Research: Lessons from Clinical Trials of Treatments for AIDS." American Psychologist 43 (No. 7, July 1988): 573-581.
  • Merigan, Thomas C. "You Can Teach an Old Dog New Tricks: How AIDS Trials are Pioneering New Strategies." New England Journal of Medicine 323 (No. 19, November 8, 1990): 1341-1343.
  • Meyers, Karin, and Dunton, Alan W. "Case Study: Applying 'An Ethical Framework' to a Proposed HIV Antibody Screening Program." IRB 10 (No. 1, January/February 1988): 6-8.
  • Nolan, Kathleen. "AIDS and Pediatric Research." Evaluation Review 14 (No. 5, October 1990): 464-481.
  • Novick, Alvin. "Noncompliance in Clinical Trials: I. Subjects." AIDS and Public Policy Journal 5 (No. 2, Spring 1990): 94-96.
  • Novick, Alvin. "Clinical Trials with Vulnerable or Disrespected Subjects." AIDS and Public Policy Journal 4 (No. 2, 1989): 125-130.
  • Novick, Alvin. "Some Ethical Issues Associated with HIV Vaccine Trials." AIDS and Public Policy Journal 3 (No. 3, Summer 1988): 46-48.
  • Novick, Alvin. "Why the Burdensome Knowledge Need Not Be Imposed." IRB 8 (No. 5, September/October 1986): 6-7.
  • Novick, Alvin. "At Risk for AIDS: Confidentiality in Research and Surveillance." IRB 6 (No. 6, November/December 1984): 10-11.
  • Perry, Samuel W. "Pharmacological and Psychological Research on AIDS: Some Ethical Considerations." IRB 9 (No. 5, September/October, 1987): 8-10.
  • Porter, Joan P.; Glass, Marta J.; and Koff, Wayne C. "Ethical Considerations in AIDS Vaccine Testing." IRB 11 (No. 3, May/June 1989): 1-4.
  • Richman, Douglas D. "Public Access to Experimental Drug Therapy: AIDs Raises Yet Another Conflict between Freedom of the Individual and Welfare of the Individual and Public." Journal of Infectious Diseases 159 (No. 3, March 1989): 412-415.
  • Sawyer, Leigh A.; Katzenstein, David A.; and Quinnan, Gerald V. "Regulatory Concerns Regarding AIDS Vaccine Development." AIDS and Public Policy Journal 3 (No. 3, Summer 1988): 36-45.
  • Sieber, Joan E., and Sorensen, James L. "Conducting Social and Behavioral AIDS Research in Drug Treatment Clinics." IRB 14 (No. 5, September/October 1992): 1-5.
  • Spiers, Herbert R. "Community Consultation and AIDS Clinical Trials, Part I." IRB 13 (No. 3, May/June 1991): 7-10.
  • Spiers, Herbert R. "Community Consultation and AIDS Clinical Trials, Part II" IRB 13 (No. 4, July/August 1991): 1-6.
  • Spiers, Herbert R. "Community Consultation and AIDS Clinical Trials, Part III." IRB 13 (No. 5, September/October 1991): 3-7.
  • Thomas, S.B., and Quinn, S.C. "The Tuskegee Syphilis Study, 1932-1972: Implications for HIV Education and AIDS Risk Education Programs in the Black Community." American Journal of Public Health 81 (No. 11, November 1991): 1498-1505.
  • U.S. Department of Health and Human Services. Public Health Service. "U.S. Public Health Service Consultation on International Collaborative Human Immunodeficiency Virus (HIV)." Reprinted in Law, Medicine and Health Care 19 (No. 3-4, Fall/Winter 1991): 259-263.
  • Valdiserri, Ronald O.; Tama, Geraldine Maiatico; and Ho, Monto. "The Role of Community Advisory Committees in Clinical Trials of Anti-HIV Agents." IRB 10 (No. 4, July/August 1988): 5-7.
  • Veatch, Robert M. "Drug Research in Humans: The Ethics of Nonrandomized Access." Clinical Pharmacy 8 (No. 5, May 1989): 366-370.

G. Transplants

  • American Medical Association. Council on Scientific Affairs. "Xenografts: Review of the Literature and Current Status." Journal of the American Medical Association 254 (No. 23, December 20, 1985): 3353-3357.
  • Arras, John D., and Shinnar, Shlomo. "Anencephalic Newborns as Organ Donors: A Critique." Journal of the American Medical Association 259 (No. 15, April 15, 1988): 2284-2285.
  • Bailey, Leonard L. "Organ Transplantation: A Paradigm of Medical Progress." The Hastings Center Report 20 (No. 1, January/February 1990): 24-28.
  • Beauchamp, Tom L., and Childress, James F. Principles of Biomedical Ethics, 3d ed. New York: Oxford University Press, 1989.
  • Caplan, Arthur L. "Is Xenografting Morally Wrong?" Transplantation Proceedings 24 (No. 2, April 1992): 722-727.
  • Caplan, Arthur L. "Ethical Issues Raised by Research Involving Xenografts." Journal of the American Medical Association 254 (No. 23, December 20, 1985): 3339-3343.
  • Caplan, Arthur L. "Ethical and Policy Issues in the Procurement of Cadaver Organs for Transplantation." New England Journal of Medicine 311 (No. 15, October 11, 1984): 981-983.
  • Carney, Bridget. "Bone Marrow Transplantation: Nurses' and Physicians' Perceptions of Informed Consent." Cancer Nursing 10 (No. 5, October 1987): 252-259.
  • Cowan, Dale; Kantorowitz, Jo Ann; Moskowitz, Jay; and Rheinstein, Peter H., eds. Human Organ Transplantation: Societal, Medical-Legal, Regulatory, and Reimbursement Issues. Ann Arbor, MI: Health Administration Press, 1987.
  • Diethelm, Arnold G. "Ethical Decisions in the History of Organ Transplantation." Annals of Surgery 211 (No. 5, May 1990): 505-520.
  • Fost, Norman. "Organs from Anencephalic Infants: An Idea Whose Time Has Not Yet Come." The Hastings Center Report 18 (No. 5, October/November 1988): 5-10.
  • Gil, Gideon. "The Artificial Heart Juggernaut." The Hastings Center Report 19 (No. 2, March/April 1989): 24-31.
  • Holder, Angela Roddey. "The Minor as Research Subject or Transplant Donor." In Legal Issues in Pediatrics and Adolescent Medicine, 2d ed., by Angela Roddey Holder, pp. 146-178. New Haven, CT: Yale University Press, 1985.
  • Levine, Robert J. Ethics and Regulation of Clinical Research. 2d ed. Baltimore: Urban and Schwarzenberg, 1986.
  • Lind, Stuart E. "The Institutional Review Board: An Evolving Ethics Committee." The Journal of Clinical Ethics 3 (No. 4, Winter 1992): 278-282. See also commentary in the same issue by John B. Dossetor ("An Ethics Issue for Cadaver Renal Transplantation," p. 309-311).
  • McCarthy, Charles R. "The Role of the Institutional Review Board in Research Relating to Human Organ Transplantation." In Human Organ Transplantation: Societal, Medical-Legal, Regulatory, and Reimbursement Issues, edited by Dale H. Cowan et al., pp. 90-95. Ann Arbor, MI: 1987.
  • Moore, Francis D. "The Desperate Case: CARE (Costs, Applicability, Research, Ethics)." Journal of the American Medical Association 261 (No. 10, March 10, 1989): 1483-1484.
  • Moore, Francis D. "Three Ethical Revolutions: Ancient Assumptions Remodeled Under Pressure of Transplantation." Transplantation Proceedings 20 (No. 1, Supp. 1, February 1988): 1061-1067.
  • Reemtsma, K. "Ethical Aspects of Xenotransplantation." Transplantation Proceedings 22 (No. 3, June 1990): 1042-1043.
  • Robertson, John A. "Patient Selection for Organ Transplantation: Age, Incarceration, Family Support, and Other Factors." Transplantation Proceedings 21 (No. 3, June 1984): 3397-3402.
  • Shewmon, D. Alan et al. "The Use of Anencephalic Infants as Organ Sources: A Critique." Journal of the American Medical Association 261 (No. 12, March 24/31, 1989): 1773-1781.
  • Shewmon, D. Alan. "Anencephaly: Selected Medical Aspects." The Hastings Center Report 18 (No. 5, October/November 1988): 11-19.
  • Singer, Peter A. et al. "Ethics of Liver Transplantation with Living Donors." New England Journal of Medicine 321 (No. 9, August 31, 1989): 620-622.
  • Starzl, Thomas E. "Ethical Problems in Organ Transplantation: A Clinician's Point of View." Annals of Internal Medicine 67 (No. 3, September 1967): 32-36.
  • Transplant Policy Center. Ethics and Social Impact Committee. "Anencephalic Infants as Sources of Transplantable Organs." The Hastings Center Report 18 (No. 5, October/November 1988): 28-30.
  • U.S. President's Commission for the Study of Ethical Problems in Medicine and Biomedical and Behavioral Research. Implementing Human Subjects Regulations. Washington, D.C.: U.S. Government Printing Office, 1981.
  • Walters, James W., and Ashwal, Stephen. "Organ Prolongation in Anencephalic Infants: Ethical and Medical Issues." The Hastings Center Report 18 (No. 5, October/November 1988): 19-27.

H. Human Genetic Research

  • American Association for the Advancement of Science-American Bar Association National Conference of Lawyers and Scientists and the American Association for the Advancement of Science Committee on Scientific Freedom and Responsibility. The Genome, Ethics and the Law: Issues in Genetic Testing. Washington, D.C.: American Association for the Advancement of Science, 1992.
  • American Society of Human Genetics. Ad Hoc Committee on DNA Technology. "DNA Banking and DNA Analysis: Points to Consider." American Journal of Human Genetics 42 (No. 5, May 1988): 781-783. Adopted October 9, 1987.
  • Anderson, W.F. "Human Gene Therapy: Scientific and Ethical Considerations." Journal of Medicine and Philosophy 10 (No. 3, August 1985): 275-277.
  • Anderson, W.F., and Fletcher, John C. "Gene Therapy in Human Beings: When is it Ethical to Begin?" New England Journal of Medicine 303 (1980): 1293-1296.
  • Andrews, Lori B. "Legal Aspects of Genetic Information." The Yale Journal of Biology and Medicine 64 (No. 1, January/February 1991): 29-40.
  • Appelbaum, Paul S., and Rosenbaum, Alan. "Tarasoff and the Researcher: Does the Duty to Protect Apply in the Research Setting?" American Psychologist 44 (No. 6, June 1989): 885-894.
  • Australia. National Medical Council. Report on Ethics in Epidemiological Research. Canberra: Australian Government Publishing Service, 1985.
  • Baum, Rudy M. "Genetic Screening: Medical Promise Amid Legal and Ethical Questions." Chemical and Engineering News 67 (August 7, 1989): 10-16.
  • Beauchamp, Tom L., and Childress, James F. Principles of Biomedical Ethics, 3d ed. New York: Oxford University Press, 1989. See especially pp. 333-342.
  • Billings, Paul R. "Brewing Genes and Behavior: The Potential Consequences of Genetic Screening for Alcoholism." In Banbury Report 33: Genetics and Biology of Alcoholism, pp. 333-350. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press, 1990.
  • Bishop, Jerry E., and Waldholz, Michael. "The Search for the Perfect Child." The Wall Street Journal, 19 March 1986, p. 30.
  • Bloch, M., and Hayden, M. R. "Opinion: Predictive Testing for Huntington Disease in Childhood: Challenges and Implications." American Journal of Human Genetics 46 (1990): 1-4.
  • Brody, Howard. "Ethics, Technology, and the Human Genome Project." The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 278-281.
  • Busch, David B. et al. "Recontacting Subjects in Mutagen Exposure Monitoring Studies." IRB 8 (No. 6, November/December 1986): 1-4.
  • Capron, Alexander M. "Which Ills to Bear?: Reevaluating the 'Threat' of Modern Genetics." Emory Law Journal 39 (Summer 1990): 665-696. See Sections III-IV.
  • Collins, Francis S. "Medical and Ethical Consequences of the Human Genome Project." The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 260-267.
  • DeGrazia, David. "The Ethical Justification for Minimal Paternalism in the Use of the Predictive Test for Huntington's Disease." The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 219-228.
  • Dorozynski, Alexander. "Privacy Rules Blindside French Glaucoma Effort." Science 250 (April 1991): 369-370.
  • Emory Law Journal 39 (No. 3, Summer 1990). The entire issue is devoted to issues in genetics and the law.
  • European Parliament. "Resolution on the Ethical and Legal Problems of Genetic Engineering." Resolution of March 16, 1989. Official Journal of the European Communities 17.4.89, pp. C 96/165-170. Reprinted (slightly abridged) in Bulletin of Medical Ethics 57 (April 1990): 8-10.
  • Fletcher, John C. "Ethical Issues In and Beyond Prospective Clinical Trials of Human Gene Therapy." Journal of Medicine and Philosophy 10 (No. 3, August 1985): 293-309.
  • Fletcher, John C.; Berg, Kare; and Tranoy, Knut Erik. "Ethical Aspects of Medical Genetics: A Proposal for Guidelines in Genetic Counseling, Prenatal Diagnosis and Screening." Clinical Genetics 27 (No. 2, February 1985): 199-205.
  • Fost, Norman, and Farrell, Philip M. "A Prospective Randomized Trial of Early Diagnosis and Treatment of Cystic Fibrosis: A Unique Ethical Dilemma." Clinical Research 37 (No. 3, September 1989): 495-500.
  • Fost, N. and Cohen, S. "Ethical Issues Regarding Case Reports: To Publish or Perish the Thought." Clinical Research 24 (1976): 269-273.
  • Friedmann, Theodore. "The Evolving Concept of Gene Therapy." Gene Therapy 1 (1990): 175-181.
  • Glover, Jonathan. "Questions About Some Uses of Genetic Engineering." In Contemporary Issues in Bioethics, edited by Tom Beauchamp and LeRoy Walters, pp. 525-535. Belmont, CA: Wadsworth Publishing Co., 1978.
  • Goldstein, David J., ed. Genetics for the Medically Oriented: Medical Genetics Core Book. Indianapolis: Indiana University School of Medicine, Medical Genetics, 1983.
  • Gordis, Enoch; Babakoff, Boris; Goldman, David; and Berg, Kate. "Finding the Gene(s) for Alcoholism." Journal of the American Medical Association 263 (No. 15, April 18, 1990): 2094-2095.
  • Hayden, Michael R. "Predictive Testing for Huntington Disease: Are We Ready for Widespread Community Implementation?" American Journal of Medical Genetics 40 (1991): 515-517.
  • Hayes, Catherine V. "Genetic Testing for Huntington's Disease — A Family Issue." New England Journal of Medicine 327 (No. 20, November 12, 1992): 1449-1451.
  • Holtzman, Neil A. et al. "The Effect of Education on Physicians' Knowledge of a Laboratory Test: The Case of Maternal Serum Alpha-Fetoprotein Screening." The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 243-247.
  • Holtzman, Neil A. Proceed with Caution: Predicting Genetic Risks in the Recombinant DNA Era. Baltimore: Johns Hopkins University Press, 1989.
  • Howe, Edmund G., III; Kark, John A.; and Wright, Daniel G. "Studying Sickle Cell Trait in Healthy Army Recruits: Should the Research Be Done?" Clinical Research 31 (No. 2, April 1983): 119-125.
  • Huggins, M. et al. "Ethical and Legal Dilemmas Arising During Predictive Testing for Adult-Onset Disease: The Experience of Huntington Disease." American Journal of Human Genetics 47 (1990): 4-12.
  • International Committee of Medical Journal Editors. "Statements from the International Committee of Medical Journal Editors." Journal of the American Medical Association 265 (No. 20, May 22/29, 1991):2697-2698. Requiring the informed consent of patients when a risk of identification in published descriptions of research exists.
  • Karson, Evelyn and Anderson, W. French. "Prospects for Gene Therapy." In The Unborn Patient: Prenatal Diagnosis and Treatment, 2d ed., edited by Michael R. Harrison, Mitchell S. Golbus and Roy A. Filly, pp. 481-494. Philadelphia: Saunders, 1991.
  • Kolata, Gina. "Genetic Screening Raises Questions for Employers and Insurers." Science 232 (April 16, 1986): 317-319.
  • Lappe, Marc; Gustafson, James M.; and Roblin, Richard. "Ethical and Social Issues in Screening for Genetic Disease." New England Journal of Medicine 286 (No. 21, May 25, 1972): 1129-1132.
  • Levine, Robert J. Ethics and Regulation of Clinical Research. 2d ed. Baltimore: Urban and Schwarzenberg, 1986.
  • Lewis, Ricki. "Genetic-Marker Testing: Are We Ready for It?" Issues in Science and Technology 4 (No. 1, Fall 1987): 76-82.
  • MacKay, Charles R. "The Physician as Fortune Teller: A Commentary on 'The Ethical Justification for Minimal Paternalism.'" The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 228-238.
  • MacKay, Charles R. "Ethical Issues in Research Design and Conduct: Developing a Test to Detect Carriers of Huntington's Disease." IRB 6 (No. 4, July/August 1984): 1-5.
  • Meissen, Gregory H. et al. "Understanding the Decision To Take the Predictive Test for Huntington Disease." American Journal of Medical Genetics 39 (No. 4, June 15, 1991): 404-410.
  • Milewski, Elizabeth A., comp. "Discussions on Human Gene Therapy." Recombinant DNA Technical Bulletin 9 (No. 2, June 1986): 88-130. Compilation of excerpts from meetings of the National Institutes of Health's Recombinant DNA Advisory Committee and its working groups on human gene therapy and viruses.
  • Milewski, Elizabeth A. "Development of A Points to Consider Document for Human Somatic Cell Gene Therapy." Recombinant DNA Technical Bulletin 8 (No. 4, December 1985): 176-180.
  • Miller, Bruce. "Autonomy and Proxy Consent." In Alzheimer's Dementia: Dilemmas in Clinical Research, edited by Vijaya L. Melnick and Nancy N. Dubler, pp. 239-263. Clifton, NJ: Humana Press, 1985.
  • Milunsky, Aubrey, and Annas, George J., eds. Genetics and the Law, III: Proceedings of the Third National Symposium on Genetics and the Law, Boston, 1984. New York: Plenum Press, 1985.
  • Murray, Jeffrey C., and Pagon, Roberta A. "Informed Consent for Research Publication of Patient-Related Data." Clinical Research 32 (No. 4, October 1984): 404-408.
  • Murray, Thomas H. "Warning: Screening Workers for Genetic Risk." Hastings Center Report 13 (No. 1, February 1983): 5-8.
  • Nolan, Kathleen, and Sevenson, Sara. "New Tools, New Dilemmas: Genetics Frontiers." Hastings Center Report (October/November, 1988): 40-46.
  • Quaid, Kimberly A. "Predictive Testing for HD: Maximizing Patient Autonomy." The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 238-240.
  • Reilly, Philip. "When Should an Investigator Share Raw Data with the Subjects?" IRB 2 (No. 9, November 1980): 4-5, 12.
  • Reilly, Philip. Genetics, Law and Social Policy. Cambridge, Mass.: Harvard University Press, 1977.
  • Riis, Povl and Nylenna, Magne. "Patients Have a Right to Privacy and Anonymity in Medical Publication." Journal of the American Medical Association 265 (No. 10, May 22/29 1991): 2720.
  • Robertson, John A. "Legal Issues in Genetic Testing." In The Genome, Ethics and the Law: Issues in Genetic Testing, published by the American Association for the Advancement of Science-American Bar Association National Conference of Lawyers and Scientists and the American Association for the Advancement of Science Committee on Scientific Freedom and Responsibility, pp. 79-110. Washington, D.C.: American Association for the Advancement of Science, 1992.
  • Robertson, John A. "Genetic Alteration of Embryos: The Ethical Issues." In Genetics and the Law III, edited by Aubrey Milunsky and George J. Annas, pp. 115-127. New York: Plenum Press, 1985.
  • Schmeck, Harold M., Jr. "Scientists Now Doubt They Found Faulty Gene Linked to Mental Illness." The New York Times, 7 November 1989, p. 18.
  • Shaw, Margery W. "Testing for the Huntington Gene: A Right to Know, A Right Not to Know, or a Duty to Know." American Journal of Medical Genetics 26 (1987): 243-246.
  • Smurl, James F., and Weaver, David D. "Presymptomatic Testing for Huntington Chorea: Guidelines for Moral and Social Accountability." American Journal of Medical Genetics 26 (1987): 247-257.
  • Stich, Stephen P. "Lessons to be Learned from the Recombinant DNA Controversy." In Research Ethics, edited by Kare Berg and Knut Erik Tranoy, pp. 75-86. New York: Alan R. Liss, 1983.
  • Tibbles, Lance. "Derived Consent, Proxy Consent: Legal Issues." In Alzheimer's Dementia: Dilemmas in Clinical Research, edited by Vijaya L. Melnick and Nancy N. Dubler, pp. 265-294. Clifton, NJ: Humana Press, 1985.
  • U. S. Congress. Office of Technology Assessment. "Human Gene Therapy: Background Paper." Washington, D.C.: U.S. Government Publications Office, 1984. OTA-BP-BA-32.
  • U. S. Department of Health and Human Services. Public Health Service. National Institutes of Health; and U. S. Department of Energy. Office of Energy Research. Office of Health and Environmental Research. Understanding Our Genetic Inheritance, The U.S. Human Genome Project: The First Five Years FY 1991-1995. Springfield, Va.: NTIS, April 1990.
  • U.S. Department of Health and Human Services. Public Health Service. National Institutes of Health. "Recombinant DNA Molecule Research, Proposed Actions under Guidelines; Notice." Federal Register 50 (August 19, 1985): 33462-33467.
  • U. S. Department of Health and Human Services. Public Health Service. National Institutes of Health. Recombinant DNA Advisory Committee. Points to Consider Subcommittee. "Points to Consider in the Design and Submission of Protocols for the Transfer of Recombinant DNA into Human Subjects." Recombinant DNA Technical Bulletin 12 (No. 3, September 1989): 151-170.
  • U.S. Department of Health and Human Services. Public Health Service. National Institutes of Health. Workshop on Human Subjects in Genetic Research Involving Families. (October 5-6, 1992.) (Unpublished manuscripts.)

Botkin, Jeffrey. "Disclosure of Interim Results to Research Subjects."
Kass, Nancy. "Participation in Pedigree Studies and Risk of Impeded Access to Health Insurance."
Langfelder, Elinor J. "The Search for a Disease Gene: An Overview of the Methodology."
Parker, Lisa S., and Lidz, Charles W. "Family Law, Family Dynamics and the Possibility of Coercion in Family Studies."
Powers, Madison. "Publication-Related Risks to Privacy: The Ethical Implications of Pedigree Studies."
Quaid, Kimberly A. "Withdrawal from Genetic Research: Practical Consequences of Subject Control Over Specimens."

  • U. S. President's Commission for the Study of Ethical Problems in Medicine and Biomedical and Behavioral Research. Screening and Counseling for Genetic Conditions. Washington, D.C.: U.S. Government Printing Office, 1983.
  • U. S. President's Commission for the Study of Ethical Problems in Medicine and Biomedical and Behavioral Research. Splicing Life: A Report on the Social and Ethical Issues of Genetic Engineering with Human Beings. Washington, D.C.: U.S. Government Printing Office, 1982.
  • Walters, LeRoy. "Ethical Issues in Human Gene Therapy." The Journal of Clinical Ethics 2 (No. 4, Winter 1991): 267-274.
  • Walters, LeRoy. "Genetics and Reproductive Technologies." In Medical Ethics, edited by Robert M. Veatch, pp. 201-228. Boston: Jones and Bartlett, 1989.
  • Walters, LeRoy. "The Ethics of Human Genome Therapy." Nature 320 (March 20, 1986): 225-227.
  • Weiss, Rick. "Predisposition and Prejudice." Science News 135 (January 21, 1989): 40-42.
  • Wertz, Dorothy C., and Fletcher, John C. "Fatal Knowledge? Prenatal Diagnosis and Sex Selection." Hastings Center Report 19 (No. 3, May/June 1989): 21-27.
  • WGBH Educational Foundation. "Decoding the Book of Life." Nova. Program Number 1615, October 31, 1989.
  • Wiggins, Sandi et al. "The Psychological Consequences of Predictive Testing for Huntington's Disease." New England Journal of Medicine 327 (No. 20, November 12, 1992): 1402-1405.
  • World Federation of Neurology. "Ethical Issues Policy Statement on Huntington's Disease Molecular Genetics Predictive Test." Journal of Medical Genetics 27 (1990): 34-38; also, Journal of the Neurological Sciences 94 (1989): 327-332.

I. Alcohol and Drug Research

  • Appelbaum, Paul S., and Rosenbaum, Alan. "Tarasoff and the Researcher: Does the Duty to Protect Apply in the Research Setting?" American Psychologist 44 (No. 6, June 1989): 885-894.
  • Mendelson, J.H. "Protection of Participants and Experimental Design in Clinical Abuse Liability Testing." British Journal of Addiction 86 (No. 12, December 1991): 1543-1548.
  • Nahas, G.G. "The Experimental Use of Cocaine in Human Subjects." Bulletin on Narcotics 42 (No. 1, 1990): 57-62.
  • Sieber, Joan E., and Sorensen, James L. "Conducting Social and Behavioral AIDS Research in Drug Treatment Clinics." IRB 14 (No. 5, September/October 1992): 1-5.
  • U.S. Department of Health and Human Services. Public Health Service. National Institute on Alcohol Abuse and Alcoholism. National Advisory Council on Alcohol Abuse and Alcoholism. Recommended Council Guidelines on Ethyl Alcohol Administration in Human Experimentation. Revised June 1989.

Return to Top of Page

 

________________________
Chapter V: Research: An Overview