Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-15T11:27:58.853Z Has data issue: false hasContentIssue false

First-in-Human Trial Participants: Not a Vulnerable Population, but Vulnerable Nonetheless

Published online by Cambridge University Press:  01 January 2021

Extract

Translational science is a 21st century mission. Government officials and industry leaders are making huge investments in an attempt to transform more basic science discoveries into therapeutic applications. Scientists and policymakers express great excitement about the medical advances that could come with the current bench-to-bedside campaign.

A key step in translational science is the move from animal and other preclinical studies to initial human testing. Researchers ability to predict human effects is limited, and first-in-human (FIH) tests present significant uncertainty. Participants in this form of research face risks and can experience serious, even lethal, harm. Well-known incidents involving Jolee Mohr and Jesse Gelsinger, as well as subjects in the 2006 study of the investigational agent TGN1412, show the dangers that can arise in early human research.

The bench-to-bedside campaign will need many volunteers to participate in early human testing. Investigators and regulators must not allow the policy enthusiasm for translational science to overshadow the commitment to protect human subjects. Participants in exploratory research are diverse and as a group lack characteristics usually associated with vulnerable populations, such as impaired decisional ability or economic or educational disadvantage.

Type
Symposium
Copyright
Copyright © American Society of Law, Medicine and Ethics 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

See, e.g., Westfall, J. M., “Practice-Based Research — ‘Blue Highways’ on the NIH Roadmap,” JAMA 297, no. 4 (2007): 403406; Zerhouni, E. A., “Translational and Clinical Science — Time for a New Vision,” New England Journal of Medicine 353, no. 15 (2005): 1621–1623.CrossRefGoogle Scholar
Gilbert, S., “Trials and Tribulations,” Hastings Center Report 38, no. 2 (2008): 1418.CrossRefGoogle Scholar
Steinbrook, R., “The Gelsinger Case,” in Emanuel, E., eds., The Oxford Textbook of Clinical Research Ethics (New York: Oxford University Press, 2008): At 110–120.Google Scholar
Suntharalingam, G., “Cytokine Storm in a Phase I Trial of the Anti-CD28 Monoclonal Antibody TGN1412,” New England Journal of Medicine 355, no. 10 (2006): 10181028.CrossRefGoogle Scholar
Seidenfeld, J. Horstmann, E. Emanuel, E. J. Grady, C., “Participants in Phase I Oncology Research Trials: Are They Vulnerable?” Archives of Internal Medicine 168, no. 1 (2008): 1620.CrossRefGoogle Scholar
O'Donnell, D. O., “First Time in Man Studies: A Contract Research Organization Perspective,” Journal of Clinical Pharmacology 30, no. 3 (1990): 217–11, at 217.Google Scholar
This article considers the ethics of FIH trials in adult subjects. For discussion of the special issues involved in pediatric FIH trials, see Kimmelman, J., “Staunch Protections: The Ethics of Haemophilia Gene Transfer Research,” Haemophilia 14, no. 1 (2008): 514. For a discussion of issues raised by FIH trials in pregnant women and fetuses, see Recombinant DNA Advisory Committee, Prenatal Gene Transfer: Scientific, Medical, and Ethical Issues, 2000, available at <http://www4.od.nih.gov/oba/rac/gtpcreport.pdf> (last visited November 20, 2008). For an analysis of issues raised by FIH trials in the developing world, see London, A. J. Kimmelman, J., “Justice in Translation: From Bench to Bedside in the Developing World,” The Lancet 372, no. 9632 (2008): 82–85; Kimmelman, J., “Clinical Trials and SCID Row: The Ethics of Phase I Trials in Developing World,” Developing World Bioethics 7, no. 3 (2007): 128–135.Google Scholar
21 Code of Federal Regulations § 312.21. See also A.Nada, Somberg, J., “First-in-Man (FIM) Clinical Trials Post-TeGenero: A Review of the Impact of the TeGenero Trial on the Design, Conduct, and Ethics of FIM Trials,” American Journal of Therapeutics 14, no. 6 (2007): 594604, at 595–596 (first-in-human trials “aim to assess the safety and tolerability of new drug compounds by identifying their side effects, including their intensity, duration, and reversibility as a function of dose and corresponding dose/plasma concentration relationships after administration of initially single, then multiple, doses of a new drug compound without compromising participant safety”).CrossRefGoogle Scholar
Bergstrom, R. F. Lemberger, L., “First in Man Studies: An Industrial Perspective,” Journal of Clinical Pharmacology 30, no. 3 (1990): 212217.Google Scholar
Wood, A. J. Darbyshire, J., “Injury to Research Volunteers — The Clinical-Research Nightmare,” New England Journal of Medicine 354, no. 18 (2006): 18701871.CrossRefGoogle Scholar
Horstmann, E., “Risks and Benefits of Phase I Oncology Trials, 1991 through 2002,” New England Journal of Medicine 352, no. 9 (2005): 895904.CrossRefGoogle Scholar
See Bergstrom, Lemberger, , supra note 9.Google Scholar
Aguilar, L. K. Aguilar-Cordova, E., “Evolution of a Gene Therapy Trial,” Journal of Neuro-Oncology 65, no. 3 (2003): 307315; Spink, J. Geddes, D., “Gene Therapy Progress and Prospects: Bringing Gene Therapy into Medical Practice: The Evolution of International Ethics and the Regulatory Environment,” Gene Therapy 11, no. 22 (2004): 1611–1616.CrossRefGoogle Scholar
Food and Drug Administration, Guidance for Industry, Investigators, and Reviewers: Exploratory IND Studies, January 2006, available at <www.fda.gov/cder/guidance/7086fnl.pdf> (last visited November 20, 2008).+(last+visited+November+20,+2008).>Google Scholar
European Medicines Agency, Note for Guidance on Non-Clinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals, July 2008, available at <http://www.emea.europa.eu/pdfs/human/ich/028695en.pdf> (last visited November 20, 2008).+(last+visited+November+20,+2008).>Google Scholar
European Medicines Agency, Draft Guideline on Requirements for First-in-Man Clinical Trials for Potential High-Risk Medicinal Products, March 2007, available at <www.emea.europa.eu/pdfs/human/swp/2836707en.pdf> (last visited November 20, 2008).+(last+visited+November+20,+2008).>Google Scholar
See Nada, Somberg, , supra note 8.Google Scholar
See, e.g., National Commission for the Protection of Human Subjects of Biomedical and Behavioral Research, The Belmont Report: Ethical Principles and Guidelines for the Protection of Human Subjects of Research, National Institutes of Health, Bethesda, Maryland, 1979, available at <http://www.hhs.gov/ohrp/humansubjects/guidance/belmont.htm> (last visited November 20, 2008); World Medical Association, “Declaration of Helsinki: Ethical Principles for Medical Research Involving Human Subjects,” JAMA 284, no. 23 (2000): 30433045; “The Nuremberg Code,” New England Journal of Medicine 337, no. 20 (1997): 1436. See also Joffe, S. Miller, F. G., “Bench to Bedside: Mapping the Moral Terrain of Clinical Research,” Hastings Center Report 38, no. 2 (2008): 30–42.Google Scholar
The Code recognizes a possible exception when the researcher acts as the experimental subject. See Id., (“Nuremberg Code”) at 1436. In the past, this practice was not unusual among researchers, and it reportedly persists in the modern era. See Altman, L. K., Who Goes First? The Story of Self-Experimentation in Medicine (Berkeley: University of California Press, 1998).CrossRefGoogle Scholar
See “Nuremberg Code,” supra note 18, at 1436.Google Scholar
Joint ABPI/BIA Report, “Early Stage Clinical Trial Task Force,” in Department of Health, Expert Group on Phase I Clinical Trials, Final Report, Appendix D, at 18–19, December 2006, available at <www.dh.gov.uk> (last visited November 20, 2008).+(last+visited+November+20,+2008).>Google Scholar
See Nada, Somberg, , supra note 8.Google Scholar
Dowsing, T. Kendall, M. J., “The Northwick Park Tragedy — Protecting Healthy Volunteers in Future First-in-Man Trials,” Journal of Clinical Pharmacy and Therapeutics 32, no. 3 (2007): 203207.CrossRefGoogle Scholar
See Nada, Somberg, , supra note 8.Google Scholar
Stroke Therapy Academic Industry Roundtable, “Recommendations for Standards Regarding Preclinical Neuroprotective and Restorative Drug Development,” Stroke 30, no. 12 (1999): 27522758, at 2752.CrossRefGoogle Scholar
See Weihrauch, M. R. Diehl, V., “Artificial Sweeteners — Do They Bear a Carcinogenic Risk?” Annals of Oncology 15, no. 10 (2004): 14601465.CrossRefGoogle Scholar
Marincola, F. M., “Translational Medicine: A Two-Way Road,” Journal of Translational Medicine 1, no. 1 (2003): 12, available at <http://www.translational-medicine.com/content/1/1/1> (last visited November 20, 2008). In an effort to improve animal models in cancer research, investigators study companion animals with naturally occurring disease. The hope is that these animals are a closer model for human cancer patients than are laboratory animals whose disease was purposely induced. See Porrello, A. Cardelli, P. Spugnini, E. P., “Oncology of Companion Animals as a Model for Humans: An Overview of Tumor Histotypes,” Journal of Experimental and Clinical Cancer Research 25, no. 1 (2006): 97–105; Porrello, A. Cardelli, P. Spugnini, E. P., “Pet Models in Cancer Research: General Principles,” Journal of Experimental and Clinical Cancer Research 23, no. 2 (2004): 181–183.CrossRefGoogle Scholar
See Aguilar and Aguilar-Cordova, supra note 13, at 311.Google Scholar
Davenport, R. J., “On Trial,” SAGE KE, June 16, 2004, available at <http://sageke.sciencemag.org> (last visited July 1, 2008).Google Scholar
Sabroe, I., “Identifying and Hurdling Obstacles to Translational Research,” Nature Reviews Immunology 7, no. 1 (2007): 7782.CrossRefGoogle Scholar
See Aguilar and Aguilar-Cordova, supra note 13.Google Scholar
See Marincola, , supra note 28.Google Scholar
Hackam, D. G. Redelmeier, A., “Translation of Research Evidence from Animals to Humans,” JAMA 296, no. 14 (2006): 17311732, at 1732.CrossRefGoogle Scholar
Bebarta, V. Luyten, D. Heard, K., “Emergency Medicine Animal Research: Does Use of Randomization and Blinding Affect the Results?” Academic Emergency Medicine 10, no. 6 (2003): 684687. The authors note that even though laboratory animal research typically involves less subject heterogeneity and opportunity for biased assessment than human research does, there are still variations among animals and observer biases that could affect animal study results.CrossRefGoogle Scholar
Hampshire, V. DeRenzo, E., “Moving Research from the Cage to the Bedside: The Need for IACUC/IRB Cooperation,” Lab Animal 31, no. 4 (2002): 2731, at 30.Google Scholar
Hampshire and DeRenzo describe how additional animal studies could have produced better information on potential human risks of the gene transfer agent in the trial involving Jesse Gelsinger. See id., at 27. They also point out that negative findings from animal studies often remain unpublished, which can deprive investigators planning human studies of potentially important information. Id., at 29.Google Scholar
Kimmelman, J., Lost in Translation: Gene Transfer and the Ethics of First in Human Studies (Cambridge University Press, forthcoming). Kimmelman suggests that the preliminary work could include minimal risk “proof of principle” human studies to test physiological parameters or validate hypotheses related to the investigational intervention.Google Scholar
Kimmelman, J., “Ethics at Phase O: Clarifying the Issues,” Journal of Law, Medicine & Ethics 35, no. 4 (2007): 727733. As Kimmelman observes, it remains to be seen whether exploratory INDs and other translational innovations will help sponsors engage in more efficient product development.CrossRefGoogle Scholar
See Wood, Darbyshire, , supra note 10, at 1870.Google Scholar
See Nada, Somberg, , supra note 8, at 596.Google Scholar
This produces what is known as the “Minimal Anticipated Biological Effect Level.” See European Medicines Agency, supra note 16, at 7.Google Scholar
See Food and Drug Administration, supra note 14, at 9.Google Scholar
See Nada, Somberg, , supra note 8, at 597.Google Scholar
See European Medicines Agency, supra note 16, at 9.Google Scholar
See Nada, Somberg, , supra note 8, at 598.Google Scholar
See Bergstrom, Lemberger, , supra note 9.Google Scholar
See European Medicines Agency, supra note 16, at 9.Google Scholar
Shamoo, A. E. Resnik, D. B., “Strategies to Minimize Risks and Exploitation in Phase I Trials on Healthy Subjects,” American Journal of Bioethics 6, no. 3 (2006): W1W13, at W5.CrossRefGoogle Scholar
Id., at W6.Google Scholar
See Nada, Somberg, , supra note 8, at 599–60.Google Scholar
See Wood, Darbyshire, , supra note 10.Google Scholar
See Department of Health, Expert Group on Phase I Clinical Trials, Final Report, December 2006, available at <www.dh.gov.uk> (last visited July 2, 2008); Nada, Somberg, , supra note 8; Levine, C. Sugarman, J., “After the TGN1412 Tragedy: Addressing the Right Questions at the Right Time for Early-Phase Testing,” Hastings Center Bioethics Forum, April 17, 2006, available at <http://www.bioethicsforum.org/20060417clevinejsugarman.asp> (last visited November 20, 2008).Google Scholar
See Nada, Somberg, , supra note 8, at 599.Google Scholar
Dawson, L., “Safety Issues in Cell-Based Intervention Trials,” Fertility and Sterility 80, no. 5 (2003): 10771085; see European Medicines Agency, supra note 16, at 8.CrossRefGoogle Scholar
Id., at 1083.Google Scholar
See Nada, Somberg, , supra note 8, at 599.Google Scholar
See Joint ABPI/BIA Report, supra note 21, at 2324.Google Scholar
Steinbrook, R., “Protecting Research Subjects – The Crisis at Johns Hopkins,” New England Journal of Medicine 346, no. 9 (2002): 716720.CrossRefGoogle Scholar
Kimmelman, J., “Stable Ethics: Enrolling Non-Treatment-Refractory Volunteers in Novel Gene Transfer Trials,” Molecular Therapy 15, no. 11 (2007): 19041906.CrossRefGoogle Scholar
Sugarman, J., “Ethical Considerations in Leaping from Bench to Bedside,” Science 285, no. 5436 (1999): 20712072.CrossRefGoogle Scholar
Emanuel, E. J. Miller, F. G., “Money and Distorted Judgments about Research: Ethical Assessment of the TeGenero TGN1412 Trial,” American Journal of Bioethics 7, no. 2 (2007): 7681.CrossRefGoogle Scholar
See Kimmelman, , “Staunch Protections,” supra note 7.Google Scholar
See Kimmelman, , supra note 38.Google Scholar
See Sugarman, , supra note 65, at 2071.Google Scholar
See Steinbrook, , supra note 3, at 113. Some commentators also criticized the choice of healthy volunteers for the TGN1412 trial. In that case, the test agent was being developed as a potential therapy for leukemia and autoimmune disease. Some said that it would have been better to recruit patients with the relevant diseases in whom approved therapies had failed. See Emanuel, Miller, , supra note 66, at 77.Google Scholar
See Committee on Ethical Considerations for Revisions to DHHS Regulations for Protection of Prisoners Involved in Research, Ethical Considerations for Research Involving Prisoners (Washington, D.C.: National Academies Press, 2006).Google Scholar
Elliott, C., “Guinea-Pigging,” New Yorker, January 7, 2008, at 36–41, at 36.Google Scholar
For examples of such deception, see McHugh, J., “Drug Test Cowboys: The Secret World of Pharmaceutical Trial Subjects,” Wired Magazine, April 24, 2007, available at <http://www.wired.com/wired/archive/15.05/feat_drugtest.html> (last visited November 20, 2008); Cohen, L., “Stuck for Money: To Screen New Drugs for Safety, Lilly Pays Homeless Alcoholics,” Wall Street Journal, November 14, 1996, at A1.Google Scholar
See Joint ABPI/BIA Report, supra note 21, at 2425.Google Scholar
Tishler, C. L. Bartholomae, S., “Repeat Participation among Normal Healthy Research Volunteers: Professional Guinea Pigs in Clinical Trials?” Perspectives in Biology and Medicine 46, no. 4 (2003): 508520, at 512.CrossRefGoogle Scholar
Elliott, C. Abadie, R., “Exploiting a Research Underclass in Phase I Clinical Trials,” New England Journal of Medicine 358, no. 22 (2008): 23162317.CrossRefGoogle Scholar
See Tishler, Bartholomae, , supra note 75.Google Scholar
See Shamoo, Resnik, , supra note 50, at W9.Google Scholar
Id., at W10; see Emanuel, Miller, , supra note 66.Google Scholar
See Elliott, , supra note 72, at 37.Google Scholar
Id., at 40.Google Scholar
Nuffield Council on Bioethics Comment, in Department of Health, Expert Group on Phase I Clinical Trials, Final Report, Appendix D, at 61, December 2006, available at <www.dh.gov.uk> (last visited November 20, 2008). See also Grant, R. W. Sugarman, J., “Ethics in Human Subject Research: Do Incentives Matter?” Journal of Philosophy and Medicine 29, no. 6 (2004): 717738.Google Scholar
Dickert, N. Grady, C., “What's the Price of a Research Subject? Approaches to Payment for Research Participation,” New England Journal of Medicine 341, no. 3 (1999): 198203. The general view is that increased payment should not be offered for higher-risk studies. Nada and Somberg think that increased payment should be considered, but say that high payment could not legitimize unreasonably risky research. See Nada, Somberg, , supra note 8, at 601.CrossRefGoogle Scholar
Id. (Nada and Somberg); Joffe, Miller, , supra note 18; Shamoo, Resnik, , supra note 50.Google Scholar
In an interview, one participant said, “I've worked as an electrician and seen guys get electrocuted. Being a lab rat is the only work situation where you've got round-the-clock medical attention. It's the safest job I've been in.” See McHugh, , supra note 73, at 3.Google Scholar
See Nada, Somberg, , supra note 8, at 601.Google Scholar
See Tishler, Bartholomae, , supra note 75.Google Scholar
See Dickert, Grady, , supra note 84.Google Scholar
See Nada, Somberg, , supra note 8, at 601.Google Scholar
See Kimmelman, , supra note 64; Morreim, E. H., “High-Profile Research and the Media: The Case of the AbioCor Artificial Heart,” Hastings Center Report 34, no. 1 (2004): 1124; Albers, G. W. Zivin, J. A. Choi, D. W., “Ethical Standards in Phase I Trials of Neuroprotective Agents for Stroke,” Stroke 29, no. 8 (1998): 1493–1494.Google Scholar
See Dawson, , supra note 56, at 1082.Google Scholar
Chen, E. X. Tannock, I. F., “Risks and Benefits of Phase I Clinical Trials Evaluating New Anticancer Agents,” JAMA 292, no. 17 (2004): 21502151.CrossRefGoogle Scholar
See Horstmann, , supra note 11.Google Scholar
Id. For another evaluation of risks and benefits in phase I oncology trials, see Roberts, T. G., “Trends in the Risks and Benefits to Patients with Cancer Participating in Phase I Clinical Trials,” JAMA 292, no. 17 (2004): 21302140.CrossRefGoogle Scholar
Agrawal, M. Emanuel, E. J., “Ethics of Phase I Oncology Studies,” JAMA 290, no. 8 (2003): 10751082, at 1076.CrossRefGoogle Scholar
See Horstmann, , supra note 11.Google Scholar
Rothschild, B. King, N., “Phase I Clinical Trials in Oncology,” New England Journal of Medicine 352, no. 23 (2005): 2451.Google Scholar
Kuehn, B. M., “Industry, FDA Warm to ‘Adaptive’ Trials,” JAMA 296, no. 16 (2006): 19551957; see Chen, Tannock, , supra note 94, at 2151. It is not clear that this approach will improve the risk-benefit ratio, however. A review of oncology trials published between 2002 and 2004 found that such design strategies were not associated with increased response rates and that subjects in such trials were at greater risk of experiencing severe or life-threatening toxicity. Koyfman, S. A., “Risks and Benefits Associated with Novel Phase I Oncology Trial Designs,” Cancer 110, no. 5 (2007): 1115–1124.CrossRefGoogle Scholar
See Chen, Tannock, , supra note 94; Degos, L., “Phase I Trials in Cancer Treatment. The Maximum Tolerated Dose: A Barbarian Guideline,” Hematology Journal 1, no. 4 (2000): 219.Google Scholar
See Agrawal, Emanuel, , supra note 97, at 1077. For analysis questioning whether this kind of benefit should “count” in risk-benefit evaluations, see King, N. Churchill, L., “Assessing and Comparing Potential Benefits and Risks or Harm,” in Emanuel, E., eds., The Oxford Textbook of Clinical Research Ethics (New York: Oxford University Press, 2008): 514526.Google Scholar
See Chen, Tannock, , supra note 94, at 2150.Google Scholar
See Joffe, Miller, , supra note 18, at 37–39.Google Scholar
Horng, S., “Descriptions of Benefits and Risks in Consent Forms for Phase I Oncology Trials,” New England Journal of Medicine 347, no. 26 (2002): 21342140.CrossRefGoogle Scholar
Grady, C. Emanuel, E. Horng, S., “Consent Forms for Oncology Trials,” New England Journal of Medicine 348, no. 15 (2003): 14961497, at 1497.Google Scholar
See Horng, , supra note 105, at 2139.Google Scholar
King, N., “Consent Forms and the Therapeutic Misconception,” IRB: Ethics & Human Research 27, no. 1 (2005): 18, at 7.CrossRefGoogle Scholar
Id., at 2. Studies indicate that many participants overestimate the chance of personal benefit in trials. For review and discussion, see Flory, J. H. Wendler, D. Emanuel, E. J., “Empirical Issues in Informed Consent for Research,” in Emanuel, E., eds., The Oxford Textbook of Clinical Research Ethics (New York: Oxford University Press, 2008): 645660, at 651–52.Google Scholar
Henderson, G. E., “Uncertain Benefit: Investigators' Views and Communications in Early Phase Gene Transfer Trials,” Molecular Therapy 10, no. 2 (2004): 225231.CrossRefGoogle Scholar
Id., at 226227.Google Scholar
Ryan, D. P., “Reality Testing in Cancer Treatment: The Phase I Trial of Endostatin,” The Oncologist 4 (1999): 501508, at 505.CrossRefGoogle Scholar
Annas, G. J., “The Changing Landscape of Human Experimentation: Nuremberg, Helsinki, and Beyond,” Health Matrix 2, no. 2 (1992): 119140; Gordon, J., “when Consent to Serve as a Research Subject Is Not Informed,” Lahey Clinic Medical Ethics (Winter 2008): at 3.Google Scholar
Miller, M., “Phase I Cancer Trials: A Collusion of Misunderstanding,” Hastings Center Report 30, no. 4 (2000): 3442.CrossRefGoogle Scholar
Dresser, R., When Science Offers Salvation: Patient Advocacy and Research Ethics (New York: Oxford University Press, 2001): At 45–70, 129–50.Google Scholar
The therapeutic misconception arises when research participants confuse the objective of research, which is to generate knowledge, with the objective of therapy, which is to benefit the individual patient. In this situation, some may enroll in studies with the mistaken belief that each study component is aimed at patient care, rather than knowledge production. See Henderson, G. E., “Clinical Trials and Medical Care: Defining the Therapeutic Misconception,” PLoS Medicine 4, no. 11 (2007): e324, available at <http://medicine.plosjournals.org/perlserv/?request=get-document&doi=10.1371/journal.pmed.0040324&ct=1> (last visited November 20, 2008). In a discussion of translational science, one group advises scientists to “learn to articulate not only the promise of science, but also the difficulties that are associated with moving an idea along to the product stage, so that unrealistic expectations of perfect medicines for every disease in short time frames are not raised.” See Sabroe, , supra note 31, at 79.CrossRefGoogle Scholar
Kong, W. M., “Legitimate Requests and Indecent Proposals: Matters of Justice in the Ethical Assessment of Phase I Trials Involving Competent Patients,” Journal of Medical Ethics 31, no. 4 (2005): 205208.CrossRefGoogle Scholar
Id., at 207.Google Scholar
See Annas, , supra note 113, at 138.Google Scholar
E.g., Seidenfeld, , supra note 5; Agrawal, Emanuel, , supra note 97.Google Scholar
Id. (Agrawal and Emanuel), at 1077.Google Scholar
See King, , supra note 108.Google Scholar
See Gordon, , supra note 113.Google Scholar
See Kimmelman, , “Staunch Protections,” supra note 7, at 2.Google Scholar
See Kimmelman, , supra note 64, at 1904.Google Scholar
King, N., “The Healthy-Patient Paradox in Clinical Trials,” Atrium: The Report of the Northwestern Medical Humanities and Bioethics Program, no. 5 (2008): 911.Google Scholar
See Kimmelman, , supra note 64, at 1905.Google Scholar
Id. See also Dresser supra note 115, at 2143.Google Scholar
Id. (Kimmelman), at 1905.Google Scholar