The Food and Drug Administration (FDA) requires researchers to provide specific data about new drug candidates before the drug can to proceed to human clinical trials. The IND submission package includes preclinical data on animal pharmacology and toxicology as a proof that the drug candidate is reasonably safe for human trials; manufacturing information to ensure consistent quality of the product; and clinical protocols and investigator information. The application also includes information on clinical professionals and a commitment to obtain informed consent and a review by the institutional review board (Food and Drug Administration, 2017).
The pharmacology/toxicology package contains pharmacology studies, acute, subchronic and chronic toxicity studies, special toxicity, carcinogenicity, reproduction toxicity, mutagenicity and absorption, distribution, metabolism, and excretion (ADME) studies (Food and Drug Administration, 2017). This information generally comes from in vitro tests, computer modeling, and from experiments on laboratory animals. All experiments on animals in the U.S. have to conform to Part 58 of Title 21 of the Code of Federal Regulations Good Clinical Practice for Nonclinical Laboratory Studies (eCFR — Code of Federal Regulations, 2017).
From a business risk perspective, compliance and due diligence are the easy parts. The hard part is the predictive power of this information, whether or not the drug candidate has the potential to show safety and efficacy in clinical trials and obtain approval. There is a universal pressure to conduct these tests in the timeliest manner practicable, to pass the IND submission landmark. This step typically indicates the company's willingness to undertake a huge investment in clinical trials, without any guarantees of success. The impact of IND submission on stock price is typically neutral (Picardo, 2017).
The dropout rate is especially high for new molecular entities with the potential of becoming first-in-class. The dropout rate in phase III is especially worrying.
The low reproducibility of preclinical research is an important source of assumptions made in the early stages of preclinical research. Freedman et al. (2015) argue that 50% of all preclinical research is irreproducible. Research that produces no useful results exceeds US$28 billion in the United States only. The reasons include study design, methodology, materials, reference standards and reagents used, laboratory protocols, and analysis and reporting (Freedman, Cockburn and Simcoe, 2015).
The predictive power of data from preclinical research is apparent from the attrition rate in clinical trials. The Tufts Center for the Study of Drug Development estimates the cost of a newly approved drug to be at about US$2.6 billion. On average, about 60% drop out in Phase I of clinical research, and only 8% percent of drug candidates from the discovery phase reach the market. The attrition rate in the preclinical stage is approximately 70%. Unsurprisingly, the highest attrition rate is among first-in-class drugs (Booth, 2017).
References
eCFR — Code of Federal Regulations (2017). eCFR — Code of Federal Regulations: Title 21, Chapter I, Subchapter A, Part 58: Good Laboratory Practice for non-clinical laboratory studies. [online] Ecfr.gov. Available at: https://www.ecfr.gov/cgi-bin/text-idx?SID=57e31d5c0911f0938111f968430b4fd5&mc=true&tpl=/ecfrbrowse/Title21/21cfr58_main_02.tpl [Accessed 1 Jun. 2017].
Food and Drug Administration (2017). Investigational New Drug (IND) Application. [online] Fda.gov. Available at: https://www.fda.gov/drugs/developmentapprovalprocess/howdrugsaredevelopedandapproved/approvalapplications/investigationalnewdrugindapplication/default.htm [Accessed 2 Jun. 2017].
Food and Drug Administration (2017). Drugs: Guidance, Compliance & Regulatory Information - Guidances (Drugs) - Pharmacology / Toxicology. [online] Food and Drug Administration. Available at: https://www.fda.gov/drugs/guidancecomplianceregulatoryinformation/guidances/ucm065014.htm [Accessed 2 Jun. 2017].
Shanley, A. (2017). Preventing Phase III Failures. [online] Pharmtech.com. Available at: http://www.pharmtech.com/preventing-phase-iii-failures [Accessed 2 Jun. 2017].
World Health Organization (2017). ICTRP Search Portal. [online] Apps.who.int. Available at: http://apps.who.int/trialsearch/ [Accessed 1 Jun. 2017].
US. National Institute of Health (2017). Home - ClinicalTrials.gov. [online] Clinicaltrials.gov. Available at: https://clinicaltrials.gov/ [Accessed 1 Jun. 2017].
Sheckler, R. (2017). All organizational dysfunctions track directly to leadership failings. (Part 2). [online] Arete-Zoe. Available at: https://www.aretezoe.com/single-post/2017/02/19/All-organizational-dysfunctions-track-directly-to-leadership-failings-Part-2 [Accessed 2 Jun. 2017].
Wyss Institute (2017). Human Organs-on-Chips. [online] Wyss Institute. Available at: https://wyss.harvard.edu/technology/human-organs-on-chips/ [Accessed 2 Jun. 2017].
ter Riet, G., Korevaar, D., Leenaars, M., Sterk, P., Van Noorden, C., Bouter, L., Lutter, R., Elferink, R. and Hooft, L. (2012). Publication Bias in Laboratory Animal Research: A Survey on Magnitude, Drivers, Consequences and Potential Solutions. PLoS ONE, 7(9), p.e43404.
Matosin, N., Frank, E., Engel, M., Lum, J. and Newell, K. (2014). Negativity towards negative results: a discussion of the disconnect between scientific worth and scientific culture. Disease Models & Mechanisms, 7(2), pp.171-173.
Freedman, L., Cockburn, I. and Simcoe, T. (2015). The Economics of Reproducibility in Preclinical Research. PLOS Biology, 13(6), p.e1002165.
Booth, B. (2017). A Billion Here, A Billion There: The Cost Of Making A Drug Revisited | LifeSciVC. [online] LifeSciVC. Available at: https://lifescivc.com/2014/11/a-billion-here-a-billion-there-the-cost-of-making-a-drug-revisited/ [Accessed 1 Jun. 2017].
Manning, F. and Swartz, M. (1995). Review of the Fialuridine (FIAU) clinical trials. 1st ed. Washington, DC: National Academy Press.
Attarwala, H. (2010). TGN1412: From Discovery to Disaster. Journal of Young Pharmacists, 2(3), pp.332-336.
Kerbrat, A., Ferré, J., Fillatre, P., Ronzière, T., Vannier, S., Carsin-Nicol, B., Lavoué, S., Vérin, M., Gauvrit, J., Le Tulzo, Y. and Edan, G. (2016). Acute Neurologic Disorder from an Inhibitor of Fatty Acid Amide Hydrolase. New England Journal of Medicine, 375(18), pp.1717-1725.
European Medicines Agency (2017). European Medicines Agency - News and Events - Proposals to revise guidance on first-in-human clinical trials. [online] Ema.europa.eu. Available at: http://www.ema.europa.eu/ema/index.jsp?curl=pages/news_and_events/news/2016/07/news_detail_002572.jsp&mid=WC0b01ac058004d5c1 [Accessed 1 Jun. 2017].
Abaci, H. and Shuler, M. (2015). Human-on-a-chip design strategies and principles for physiologically based pharmacokinetics/pharmacodynamics modeling. Integr. Biol., 7(4), pp.383-391.
Oleaga, C., Bernabini, C., Smith, A., Srinivasan, B., Jackson, M., McLamb, W., Platt, V., Bridges, R., Cai, Y., Santhanam, N., Berry, B., Najjar, S., Akanda, N., Guo, X., Martin, C., Ekman, G., Esch, M., Langer, J., Ouedraogo, G., Cotovio, J., Breton, L., Shuler, M. and
Hickman, J. (2016). Multi-Organ toxicity demonstration in a functional human in vitro system composed of four organs. Scientific Reports, 6(1).