Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-28T00:55:16.064Z Has data issue: false hasContentIssue false

Nanomedicine First-in-Human Research: Challenges for Informed Consent

Published online by Cambridge University Press:  01 January 2021

Extract

First-in-human (FIH) research has several characteristics that require special attention with respect to ethics and human subjects protections. At least some nanomedical technologies may also have characteristics that merit special attention in clinical research, as other papers in this symposium show. This paper considers how to address these characteristics in the consent form and process for FIH nanomedicine research, focusing principally on experimental nanotherapeutic interventions but also considering nanodiagnostic interventions.

It is essential, as a starting point, to recognize that the consent form and process are by no means the primary protectors of human subjects (although they are sometimes so regarded). Instead, consideration of the form and content of informed consent becomes relevant only after a clinical trial has been reviewed and deemed scientifically and ethically acceptable.

Two convergent types of challenges to informed consent are posed by nanomedicine FIH research. First, some issues appear generally applicable to FIH research, but have specific nanomedicine implications.

Type
Symposium
Copyright
Copyright © American Society of Law, Medicine and Ethics 2012

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

Emanuel, E. Wendler, D., and Grady, C., “What Makes Clinical Research Ethical?” JAMA 283, no. 20 (2000): 27012711.CrossRefGoogle Scholar
U.S. Department of Health and Human Services, “Common Rule, 45 CFR 46.101–124,” Federal Register 56, no. 117 (June 18, 1991): 2801228022.Google Scholar
King, N. M. P. Henderson, G. E. Churchill, L. R. Davis, A. M. Hull, S. C., and Nelson, D. K. et al., “Consent Forms and the Therapeutic Misconception: The Example of Gene Transfer Research,” IRB: Ethics & Human Subjects Research 27, no. 1 (2005): 18.CrossRefGoogle Scholar
Fatehi, L. and Wolf, S. M. et al., “Recommendations for Nanomedicine Human Subjects Research Oversight: An Evolutionary Approach for an Emerging Field,” Journal of Law, Medicine & Ethics 40, no. 4 (2012): 716750; Resnik, D. B. and Tinkle, S. S., “Ethical Issues in Clinical Trials Involving Nanomedicine,” Contemporary Clinical Trials 28, no. 4 (2007): 433–441.CrossRefGoogle Scholar
Maynard, A. D. Warheit, D. B., and Philbert, M. A., “The New Toxicology of Sophisticated Materials: Nanotoxicology and Beyond,” Toxicological Sciences 120, no. S1 (2011): S109S129.CrossRefGoogle Scholar
See Fatehi, et al., supra note 4.Google Scholar
Kimmelman, J. and London, A. J., “Predicting Harms and Benefits in Translational Trials: Ethics, Evidence, and Uncertainty,” PLoS Medicine 8, no. 3 (2011): E1001010 (5 pages), available at <http://dx.doi.org/10.1371%2Fjournal.pmed.1001010> (last visited November 12, 2012); Kimmelman, J., “Recent Developments in Gene Transfer: Risk and Ethics,” BMJ 330, no. 7482 (2005): 79–82.CrossRefGoogle Scholar
McGinn, R., “Ethical Responsibilities of Nanotechnology Researchers: A Short Guide,” Nanoethics 4, no. 1 (2010): 112; Wolf, S. M. Ramachandran, G. Kuzma, J., and Paradise, J., eds., “Symposium: Developing Oversight Approaches to Nanobiotechnology: The Lessons of History,” Journal of Law, Medicine & Ethics 37, no. 4 (2009): 543–789.CrossRefGoogle Scholar
Kimmelman, J., “Missing the Forest: Further Thoughts on the Ethics of Bystander Risk in Medical Research,” Cambridge Quarterly of Healthcare Ethics 16, no. 4 (2007b): 483–90; Kimmelman, J., “Predicting Ethical and Safety Risks to Bystanders from Nanomedicine Research,” Journal of Law, Medicine & Ethics 40, no. 4 (2012): 841–847; Marchant, G., Applying Prudent Precaution to Nanomedicine Clinical Trials, this symposium; Ramachandran, G. Howard, J. Maynard, A., and Philbert, M., “Handling Worker and Third-Party Exposures in Nanomedicine Research,” Journal of Law, Medicine & Ethics 40, no. 4 (2012): 831–840.CrossRefGoogle Scholar
McAulife, M. E. and Perry, M. J., “Are Nanoparticles Potential Male Reproductive Toxicants? A Literature Review,” Nanotoxicology 1, no. 3 (2007): 204–10; Wiwanitkit, V. Sereemaspun, A., and Rojanathanes, R., “Effect of Gold Nanoparticles on Spermatozoa: The First World Report,” Fertility and Sterility 91, no. 1 (2009): E7-e8, available at <http://www.sciencedirect.com/science/article/B6T6K-4RB5BNR-2/2/33a12feb54d3316276cbabaa2d9492b0> (last visited November 12, 2012).CrossRefGoogle Scholar
Wolf, S. M. Paradise, J. Nelson, C. A. Kahn, J. P. Lawrenz, F., “Symposium: Incidental Findings in Human Subjects Research: From Imaging to Genomics,” Journal of Law, Medicine & Ethics 36, no. 2 (2008): 216383.CrossRefGoogle Scholar
Jotterand, F. and Alexander, A. A., “Managing the ‘Known Unknowns’: Theranostic Cancer Nanomedicine and Informed Consent,” in Biomedical Nanotechnology: Methods and Protocols (New York: Springer-Verlag, 2011): At Chapter 26.Google Scholar
King, N. M. P., “Defining and Describing Benefit Appropriately in Clinical Trials,” Journal of Law, Medicine & Ethics 28, no. 4 (2000): 332343.CrossRefGoogle Scholar
Dresser, R., “First-in-Human Trial Participants: Not a Vulnerable Population, but Vulnerable Nonetheless,” Journal of Law, Medicine & Ethics 37, no. 1 (2009): 3850; King, N. M. P. and Cohen-Haguenauer, O., “En Route to Ethical Recommendations for Gene Transfer Clinical Trials,” Molecular Therapy 16, no. 3 (2008): 432–438.CrossRefGoogle Scholar
Kass, N. Taylor, H. Fogarty, L. Sugarman, J. Goodman, S. N. Goodwin-Landher, A. Carducci, M., and Hurwitz, H., “Purpose and Benefits of Early Phase Cancer Trials: What Do Oncologists Say? What Do Patients Hear?” Journal of Empirical Research on Human Research Ethics 3, no. 3 (2008): 5768; see King, et al., supra note 3.CrossRefGoogle Scholar
See King, , supra note 13.Google Scholar
See King, et al., supra note 3.Google Scholar
Appelbaum, P. S. Roth, L. H., and Lidz, C., “The Therapeutic Misconception: Informed Consent in Psychiatric Research,” International Journal of Law and Psychiatry 5, no. 3-4 (1982): 319–29.CrossRefGoogle Scholar
Churchill, L. R. Nelson, D. K. Henderson, G. E. King, N. M. P. Davis, A. M. Leahey, E., and Wilfond, B. S., “Assessing Benefits in Clinical Research: Why Diversity in Benefit Assessment Can Be Risky,” IRB: Ethics & Human Research 25, no. 3 (2003): 18; Dresser, R., “The Ubiquity and Utility of the Therapeutic Misconception,” Social Philosophy & Policy 19, no. 2 (2002): 271–294; Henderson, G. E. Easter, M. M., and Zimmer, C. et al., “Therapeutic Misconception in Early Phase Gene Transfer Trials,” Social Science and Medicine 62, no. 1 (2006): 239–253; Kimmelman, J., “The Therapeutic Misconception at 25: Treatment, Research, and Confusion,” Hastings Center Report 37, no. 6 (2007): 36–42.CrossRefGoogle Scholar
See Dresser, , supra note 14; King, and Cohen-Haguenauer, , supra note 14; King, et al., supra note 3; Fatehi, et al., supra note 4.Google Scholar
Henderson, G. E. Churchill, L. R. Davis, A. M. Grady, C. Joffe, S., and Kass, N. et al., “Clinical Trials and Medical Care: Defining the Therapeutic Misconception,” PLoS Medicine 4, no. 11 (2007): 17351738.CrossRefGoogle Scholar
See Henderson, et al., supra note 19; King, et al., supra note 3.Google Scholar
Churchill, L. R. Collins, M. L. King, N. M. P. Pemberton, S., and Wailoo, K., “Genetic Research as Therapy: Implications of ‘Gene Therapy’ for Informed Consent,” Journal of Law Medicine & Ethics 26, no. 1 (1998): 3847.CrossRefGoogle Scholar
See Kass, et al., supra note 15; King, et al., supra note 3.Google Scholar
Horng, S. and Grady, C., “Misunderstanding in Clinical Research: Distinguishing Therapeutic Misconception, Therapeutic Misestimation, and Therapeutic Optimism,” IRB: Ethics & Human Research 25, no. 1 (2003): 1116; Sulmasy, D. P. Astrow, A. B. He, M. K. Sells, D. M. Meropol, N. J. Micco, E., and Weinfurt, K. P., “The Culture of Faith and Hope: Patients' Justifications for Their High Estimates of Expected Therapeutic Benefit When Enrolling in Early Phase Oncology Trials,” Cancer 116, no. 15 (2010): 3702–3711.CrossRefGoogle Scholar
See Common Rule, supra note 2.Google Scholar
National Institutes of Health, “NIH Guidance on Informed Consent for Gene Transfer Research,” available at <http://www4.od.nih.gov/oba/RAC/ic> (last visited November 12, 2012).+(last+visited+November+12,+2012).>Google Scholar
See Kimmelman, , supra note 7; King, and Cohen-Haguenauer, , supra note 14.Google Scholar
See McGinn, , supra note 8.Google Scholar
Schenk-Braat, E. A. van Mierlo, M. M. Wagemaker, G. Bangma, C. H., and Kaptein, L. C., “An Inventory of Shedding Data from Clinical Gene Therapy Trials,” Journal of Gene Medicine 9, no. 10 (2007): 910921; King, and Cohen-Haguenauer, , supra note 14.CrossRefGoogle Scholar
See sources cited in supra note 10.Google Scholar
Hacein-Bey-Abina, S. Von Kalle, C. Schmidt, M. McCormack, M. P. Wulffraat, N., and Leboulch, P. et al., “LMO2-Associated Clonal T Cell Proliferation in Two Patients after Gene Therapy for SCID-X1,” Science 302, no. 5645 (2003): 415419.CrossRefGoogle Scholar
Capron, A., “Informed Consent in Catastrophic Disease Research and Treatment,” University of Pennsylvania Law Review 123, no. 2 (1974): 340438.CrossRefGoogle Scholar