Drug development

Last updated

Drug development is the process of bringing a new pharmaceutical drug to the market once a lead compound has been identified through the process of drug discovery. It includes preclinical research on microorganisms and animals, filing for regulatory status, such as via the United States Food and Drug Administration for an investigational new drug to initiate clinical trials on humans, and may include the step of obtaining regulatory approval with a new drug application to market the drug. [1] [2] The entire process—from concept through preclinical testing in the laboratory to clinical trial development, including Phase I–III trials—to approved vaccine or drug typically takes more than a decade. [3] [1] [2] [4]

Contents

New chemical entity development

Broadly, the process of drug development can be divided into preclinical and clinical work.

Timeline showing the various drug approval tracks and research phases Drug Evaluation Process.jpg
Timeline showing the various drug approval tracks and research phases

Pre-clinical

New chemical entities (NCEs, also known as new molecular entities or NMEs) are compounds that emerge from the process of drug discovery. These have promising activity against a particular biological target that is important in disease. However, little is known about the safety, toxicity, pharmacokinetics, and metabolism of this NCE in humans. It is the function of drug development to assess all of these parameters prior to human clinical trials. A further major objective of drug development is to recommend the dose and schedule for the first use in a human clinical trial ("first-in-human" [FIH] or First Human Dose [FHD], previously also known as "first-in-man" [FIM]).[ citation needed ]

In addition, drug development must establish the physicochemical properties of the NCE: its chemical makeup, stability, and solubility. Manufacturers must optimize the process they use to make the chemical so they can scale up from a medicinal chemist producing milligrams, to manufacturing on the kilogram and ton scale. They further examine the product for suitability to package as capsules, tablets, aerosol, intramuscular injectable, subcutaneous injectable, or intravenous formulations. Together, these processes are known in preclinical and clinical development as chemistry, manufacturing, and control (CMC).[ citation needed ]

Many aspects of drug development focus on satisfying the regulatory requirements for a new drug application. These generally constitute a number of tests designed to determine the major toxicities of a novel compound prior to first use in humans. It is a legal requirement that an assessment of major organ toxicity be performed (effects on the heart and lungs, brain, kidney, liver and digestive system), as well as effects on other parts of the body that might be affected by the drug (e.g., the skin if the new drug is to be delivered on or through the skin). Such preliminary tests are made using in vitro methods (e.g., with isolated cells), but many tests can only use experimental animals to demonstrate the complex interplay of metabolism and drug exposure on toxicity. [6]

The information is gathered from this preclinical testing, as well as information on CMC, and submitted to regulatory authorities (in the US, to the FDA), as an Investigational New Drug (IND) application. If the IND is approved, development moves to the clinical phase.

Clinical phase

Clinical trials involve four steps: [7]

The process of defining characteristics of the drug does not stop once an NCE is advanced into human clinical trials. In addition to the tests required to move a novel vaccine or antiviral drug into the clinic for the first time, manufacturers must ensure that any long-term or chronic toxicities are well-defined, including effects on systems not previously monitored (fertility, reproduction, immune system, among others). [8] [9]

If a vaccine candidate or antiviral compound emerges from these tests with an acceptable toxicity and safety profile, and the manufacturer can further show it has the desired effect in clinical trials, then the NCE portfolio of evidence can be submitted for marketing approval in the various countries where the manufacturer plans to sell it. [4] In the United States, this process is called a "new drug application" or NDA. [4] [8]

Most novel drug candidates (NCEs) fail during drug development, either because they have unacceptable toxicity or because they simply do not prove efficacy on the targeted disease, as shown in Phase II–III clinical trials. [4] [8] Critical reviews of drug development programs indicate that Phase II–III clinical trials fail due mainly to unknown toxic side effects (50% failure of Phase II cardiology trials), and because of inadequate financing, trial design weaknesses, or poor trial execution. [10] [11]

A study covering clinical research in the 1980–1990s found that only 21.5% of drug candidates that started Phase I trials were eventually approved for marketing. [12] During 2006–2015, the success rate of obtaining approval from Phase I to successful Phase III trials was under 10% on average, and 16% specifically for vaccines. [13] The high failure rates associated with pharmaceutical development are referred to as an "attrition rate", requiring decisions during the early stages of drug development to "kill" projects early to avoid costly failures. [13] [14]

Cost

One 2010 study assessed both capitalized and out-of-pocket costs for bringing a single new drug to market at about US$1.8 billion and $870 million, respectively. [15] A median cost estimate of 2015–16 trials for development of 10 anti-cancer drugs was $648 million. [16] In 2017, the median cost of a pivotal trial across all clinical indications was $19 million. [17]

The average cost (2013 dollars) of each stage of clinical research was US$25 million for a Phase I safety study, $59 million for a Phase II randomized controlled efficacy study, and $255 million for a pivotal Phase III trial to demonstrate its equivalence or superiority to an existing approved drug, [18] possibly as high as $345 million. [17] The average cost of conducting a 2015–16 pivotal Phase III trial on an infectious disease drug candidate was $22 million. [17]

The full cost of bringing a new drug (i.e., new chemical entity) to market—from discovery through clinical trials to approval—is complex and controversial. [8] [19] [17] [20] In a 2016 review of 106 drug candidates assessed through clinical trials, the total capital expenditure for a manufacturer having a drug approved through successful Phase III trials was $2.6 billion (in 2013 dollars), an amount increasing at an annual rate of 8.5%. [18] Over 2003–2013 for companies that approved 8–13 drugs, the cost per drug could rise to as high as $5.5 billion, due mainly to international geographic expansion for marketing and ongoing costs for Phase IV trials for continuous safety surveillance. [21]

Alternatives to conventional drug development have the objective for universities, governments, and the pharmaceutical industry to collaborate and optimize resources. [22] An example of a collaborative drug development initiative is COVID Moonshot, an international open-science project started in March 2020 with the goal of developing an un-patented oral antiviral drug to treat SARS-CoV-2. [23] [24]

Valuation

The nature of a drug development project is characterised by high attrition rates, large capital expenditures, and long timelines. This makes the valuation of such projects and companies a challenging task. Not all valuation methods can cope with these particularities. The most commonly used valuation methods are risk-adjusted net present value (rNPV), decision trees, real options, or comparables.[ citation needed ]

The most important value drivers are the cost of capital or discount rate that is used, phase attributes such as duration, success rates, and costs, and the forecasted sales, including cost of goods and marketing and sales expenses. Less objective aspects like quality of the management or novelty of the technology should be reflected in the cash flows estimation. [25] [26]

Success rate

Candidates for a new drug to treat a disease might, theoretically, include from 5,000 to 10,000 chemical compounds. On average about 250 of these show sufficient promise for further evaluation using laboratory tests, mice and other test animals. Typically, about ten of these qualify for tests on humans. [27] A study conducted by the Tufts Center for the Study of Drug Development covering the 1980s and 1990s found that only 21.5 percent of drugs that started Phase I trials were eventually approved for marketing. [28] In the time period of 2006 to 2015, the success rate was 9.6%. [29] The high failure rates associated with pharmaceutical development are referred to as the "attrition rate" problem. Careful decision making during drug development is essential to avoid costly failures. [30] In many cases, intelligent programme and clinical trial design can prevent false negative results. Well-designed, dose-finding studies and comparisons against both a placebo and a gold-standard treatment arm play a major role in achieving reliable data. [31]

Computing initiatives

Novel initiatives include partnering between governmental organizations and industry, such as the European Innovative Medicines Initiative . [32] The US Food and Drug Administration created the "Critical Path Initiative" to enhance innovation of drug development, [33] and the Breakthrough Therapy designation to expedite development and regulatory review of candidate drugs for which preliminary clinical evidence shows the drug candidate may substantially improve therapy for a serious disorder. [34]

In March 2020, the United States Department of Energy, National Science Foundation, NASA, industry, and nine universities pooled resources to access supercomputers from IBM, combined with cloud computing resources from Hewlett Packard Enterprise, Amazon, Microsoft, and Google, for drug discovery. [35] [36] The COVID-19 High Performance Computing Consortium also aims to forecast disease spread, model possible vaccines, and screen thousands of chemical compounds to design a COVID-19 vaccine or therapy. [35] [36] [37] In May 2020, the OpenPandemics – COVID-19 partnership between Scripps Research and IBM's World Community Grid was launched. The partnership is a distributed computing project that "will automatically run a simulated experiment in the background [of connected home PCs] which will help predict the effectiveness of a particular chemical compound as a possible treatment for COVID-19". [38]

See also

Related Research Articles

An orphan drug is a pharmaceutical agent that is developed to treat certain rare medical conditions. An orphan drug would not be profitable to produce without government assistance, due to the small population of patients affected by the conditions. The conditions that orphan drugs are used to treat are referred to as orphan diseases. The assignment of orphan status to a disease and to drugs developed to treat it is a matter of public policy that depends on the legislation of the country.

<span class="mw-page-title-main">Pharmaceutical industry</span> Industry involved with discovery, development, production and marketing of drugs

The pharmaceutical industry is an industry in medicine that discovers, develops, produces, and markets pharmaceutical drugs for use as medications to be administered to patients, with the aim to cure and prevent diseases, or alleviate symptoms. Pharmaceutical companies may deal in generic or brand medications and medical devices. They are subject to a variety of laws and regulations that govern the patenting, testing, safety, efficacy using drug testing and marketing of drugs. The global pharmaceuticals market produced treatments worth $1,228.45 billion in 2020 and showed a compound annual growth rate (CAGR) of 1.8%.

<span class="mw-page-title-main">Vaccine trial</span> Clinical trial

A vaccine trial is a clinical trial that aims at establishing the safety and efficacy of a vaccine prior to it being licensed.

<span class="mw-page-title-main">Preclinical development</span> Stage of drug development

In drug development, preclinical development is a stage of research that begins before clinical trials and during which important feasibility, iterative testing and drug safety data are collected, typically in laboratory animals.

<span class="mw-page-title-main">Investigational New Drug</span> USFDA permission to test

The United States Food and Drug Administration's Investigational New Drug (IND) program is the means by which a pharmaceutical company obtains permission to start human clinical trials and to ship an experimental drug across state lines before a marketing application for the drug has been approved. Regulations are primarily at 21 CFR 312. Similar procedures are followed in the European Union, Japan, and Canada.

An approved drug is a medicinal preparation that has been validated for a therapeutic use by a ruling authority of a government. This process is usually specific by country, unless specified otherwise.

Safety pharmacology is a branch of pharmacology specialising in detecting and investigating potential undesirable pharmacodynamic effects of new chemical entities (NCEs) on physiological functions in relation to exposure in the therapeutic range and above.

A drug pipeline is the set of drug candidates that an individual pharmaceutical company or the entire pharmaceutical industry collectively has under discovery or development at any given point in time. The drug pipeline is also sometimes restricted to a particular drug class or extended to mean the process of discovering drugs. The R&D pipeline involves various phases that can broadly be grouped in 4 stages: discovery, pre-clinical, clinical trials and marketing. Pharmaceutical companies usually have a number of compounds in their pipelines at any given time.

<span class="mw-page-title-main">Tasimelteon</span> Wakefulness medication

Tasimelteon, sold under the brand name Hetlioz, is a medication approved by the U.S. Food and Drug Administration (FDA) in January 2014, for the treatment of non-24-hour sleep–wake disorder. In June 2014, the European Medicines Agency (EMA) accepted an EU filing application for tasimelteon and in July 2015, the drug was approved in the European Union for the treatment of non-24-hour sleep-wake rhythm disorder in totally blind adults, but not in the case of non-24 in sighted people.

FV-100, also known as Cf1743, is an orally available nucleoside analogue drug with antiviral activity. It may be effective against shingles.

<span class="mw-page-title-main">Phases of clinical research</span> Clinical trial stages using human subjects

The phases of clinical research are the stages in which scientists conduct experiments with a health intervention to obtain sufficient evidence for a process considered effective as a medical treatment. For drug development, the clinical phases start with testing for drug safety in a few human subjects, then expand to many study participants to determine if the treatment is effective. Clinical research is conducted on drug candidates, vaccine candidates, new medical devices, and new diagnostic assays.

The cost of drug development is the full cost of bringing a new drug to market from drug discovery through clinical trials to approval. Typically, companies spend tens to hundreds of millions of U.S. dollars on drug development. One element of the complexity is that the much-publicized final numbers often not only include the out-of-pocket expenses for conducting a series of Phase I-III clinical trials, but also the capital costs of the long period during which the company must cover out-of-pocket costs for preclinical drug discovery. Additionally, companies often do not report whether a given figure includes the capitalized cost or comprises only out-of-pocket expenses, or both.

<span class="mw-page-title-main">COVID-19 drug repurposing research</span> Drug repurposing research related to COVID-19

Drug repositioning is the repurposing of an approved drug for the treatment of a different disease or medical condition than that for which it was originally developed. This is one line of scientific research which is being pursued to develop safe and effective COVID-19 treatments. Other research directions include the development of a COVID-19 vaccine and convalescent plasma transfusion.

<span class="mw-page-title-main">COVID-19 drug development</span> Preventative and therapeutic medications for COVID-19 infection

COVID-19 drug development is the research process to develop preventative therapeutic prescription drugs that would alleviate the severity of coronavirus disease 2019 (COVID-19). From early 2020 through 2021, several hundred drug companies, biotechnology firms, university research groups, and health organizations were developing therapeutic candidates for COVID-19 disease in various stages of preclinical or clinical research, with 419 potential COVID-19 drugs in clinical trials, as of April 2021.

<span class="mw-page-title-main">Solidarity trial</span> Accelerated multinational clinical trial program to identify therapies against COVID-19

The Solidarity trial for treatments is a multinational Phase III-IV clinical trial organized by the World Health Organization (WHO) and partners to compare four untested treatments for hospitalized people with severe COVID-19 illness. The trial was announced 18 March 2020, and as of 6 August 2021, 12,000 patients in 30 countries had been recruited to participate in the trial.

<span class="mw-page-title-main">Molnupiravir</span> Antiviral medication

Molnupiravir, sold under the brand name Lagevrio, is an antiviral medication that inhibits the replication of certain RNA viruses. It is used to treat COVID‑19 in those infected by SARS-CoV-2. It is taken by mouth.

A human challenge study, also called a challenge trial or controlled human infection model (CHIM), is a type of clinical trial for a vaccine or other pharmaceutical involving the intentional exposure of the test subject to the condition tested. Human challenge studies may be ethically controversial because they involve exposing test subjects to dangers beyond those posed by potential side effects of the substance being tested.

<span class="mw-page-title-main">Jason McLellan</span> American structural biologist

Jason S. McLellan is a structural biologist, professor in the Department of Molecular Biosciences and Robert A. Welch Chair in Chemistry at The University of Texas at Austin who specializes in understanding the structure and function of viral proteins, including those of coronaviruses. His research focuses on applying structural information to the rational design of vaccines and other therapies for viruses, including SARS-CoV-2, the novel coronavirus that causes COVID-19, and respiratory syncytial virus (RSV). McLellan and his team collaborated with researchers at the National Institute of Allergy and Infectious Diseases’ Vaccine Research Center to design a stabilized version of the SARS-CoV-2 spike protein, which biotechnology company Moderna used as the basis for the vaccine mRNA-1273, the first COVID-19 vaccine candidate to enter phase I clinical trials in the U.S. At least three other vaccines use this modified spike protein: those from Pfizer and BioNTech; Johnson & Johnson and Janssen Pharmaceuticals; and Novavax.

<span class="mw-page-title-main">Adaptive design (medicine)</span> Concept in medicine referring to design of clinical trials

In an adaptive design of a clinical trial, the parameters and conduct of the trial for a candidate drug or vaccine may be changed based on an interim analysis. Adaptive design typically involves advanced statistics to interpret a clinical trial endpoint. This is in contrast to traditional single-arm clinical trials or randomized clinical trials (RCTs) that are static in their protocol and do not modify any parameters until the trial is completed. The adaptation process takes place at certain points in the trial, prescribed in the trial protocol. Importantly, this trial protocol is set before the trial begins with the adaptation schedule and processes specified. Adaptions may include modifications to: dosage, sample size, drug undergoing trial, patient selection criteria and/or "cocktail" mix. The PANDA provides not only a summary of different adaptive designs, but also comprehensive information on adaptive design planning, conduct, analysis and reporting.

<span class="mw-page-title-main">History of COVID-19 vaccine development</span> Scientific work to develop a vaccine for COVID-19

SARS-CoV-2, the virus that causes COVID-19, was isolated in late 2019. Its genetic sequence was published on 11 January 2020, triggering an urgent international response to prepare for an outbreak and hasten the development of a preventive COVID-19 vaccine. Since 2020, vaccine development has been expedited via unprecedented collaboration in the multinational pharmaceutical industry and between governments. By June 2020, tens of billions of dollars were invested by corporations, governments, international health organizations, and university research groups to develop dozens of vaccine candidates and prepare for global vaccination programs to immunize against COVID‑19 infection. According to the Coalition for Epidemic Preparedness Innovations (CEPI), the geographic distribution of COVID‑19 vaccine development shows North American entities to have about 40% of the activity, compared to 30% in Asia and Australia, 26% in Europe, and a few projects in South America and Africa.

References

  1. 1 2 Strovel J, Sittampalam S, Coussens NP, Hughes M, Inglese J, Kurtz A, et al. (July 1, 2016). "Early Drug Discovery and Development Guidelines: For Academic Researchers, Collaborators, and Start-up Companies". Assay Guidance Manual. Eli Lilly & Company and the National Center for Advancing Translational Sciences. PMID   22553881.
  2. 1 2 Taylor D (2015). "The Pharmaceutical Industry and the Future of Drug Development". Issues in Environmental Science and Technology. Royal Society of Chemistry: 1–33. doi:10.1039/9781782622345-00001. ISBN   978-1-78262-189-8.
  3. Everts, Maaike; Cihlar, Tomas; Bostwick, J. Robert; Whitley, Richard J. (6 January 2017). "Accelerating Drug Development: Antiviral Therapies for Emerging Viruses as a Model". Annual Review of Pharmacology and Toxicology. 57 (1): 155–169. doi:10.1146/annurev-pharmtox-010716-104533. ISSN   0362-1642. PMID   27483339 . Retrieved 2 November 2021.
  4. 1 2 3 4 "The Drug Development Process". U.S. Food and Drug Administration (FDA). 4 January 2018. Retrieved 21 March 2020.
  5. Kessler DA, Feiden KL (March 1995). "Faster evaluation of vital drugs". Scientific American. 272 (3): 48–54. Bibcode:1995SciAm.272c..48K. doi:10.1038/scientificamerican0395-48. PMID   7871409.
  6. Madorran E, Stožer A, Bevc S, Maver U (2020). "In vitro toxicity model: Upgrades to bridge the gap between preclinical and clinical research". Bosnian Journal of Basic Medical Sciences. 20 (2): 157–68. doi:10.17305/bjbms.2019.4378. PMC   7202182 . PMID   31621554.
  7. Ciociola AA, Cohen LB, Kulkarni P (May 2014). "How drugs are developed and approved by the FDA: current process and future directions". The American Journal of Gastroenterology. 109 (5): 620–3. doi:10.1038/ajg.2013.407. PMID   24796999. S2CID   205100166.
  8. 1 2 3 4 Strovel J, Sittampalam S, Coussens NP, Hughes M, Inglese J, Kurtz A, et al. (July 1, 2016). "Early Drug Discovery and Development Guidelines: For Academic Researchers, Collaborators, and Start-up Companies". Assay Guidance Manual. Eli Lilly & Company and the National Center for Advancing Translational Sciences. PMID   22553881.
  9. "Vaccine Product Approval Process". U.S. Food and Drug Administration (FDA). 30 January 2018. Retrieved 21 March 2020.
  10. Van Norman GA (June 2019). "Phase II Trials in Drug Development and Adaptive Trial Design". JACC. Basic to Translational Science. 4 (3): 428–437. doi:10.1016/j.jacbts.2019.02.005. PMC   6609997 . PMID   31312766.
  11. Fogel DB (September 2018). "Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: A review". Contemporary Clinical Trials Communications. 11: 156–164. doi:10.1016/j.conctc.2018.08.001. PMC   6092479 . PMID   30112460.
  12. "R&D costs are on the rise". Medical Marketing and Media. 38 (6): 14. June 1, 2003. Archived from the original on October 18, 2016.
  13. 1 2 "Clinical development success rates: 2006–2015" (PDF). BIO Industry Analysis. June 2016.
  14. Wang Y (2012). "Extracting knowledge from failed development programmes". Pharmaceutical Medicine. 26 (2): 91–96. doi:10.1007/BF03256897. S2CID   17171991.
  15. Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH, Lindborg SR, Schacht AL (March 2010). "How to improve R&D productivity: the pharmaceutical industry's grand challenge". Nature Reviews. Drug Discovery. 9 (3): 203–14. doi: 10.1038/nrd3078 . PMID   20168317. S2CID   1299234.
  16. Prasad V, Mailankody S (November 2017). "Research and Development Spending to Bring a Single Cancer Drug to Market and Revenues After Approval". JAMA Internal Medicine. 177 (11): 1569–1575. doi:10.1001/jamainternmed.2017.3601. PMC   5710275 . PMID   28892524.
  17. 1 2 3 4 Moore TJ, Zhang H, Anderson G, Alexander GC (November 2018). "Estimated Costs of Pivotal Trials for Novel Therapeutic Agents Approved by the US Food and Drug Administration, 2015-2016". JAMA Internal Medicine. 178 (11): 1451–1457. doi:10.1001/jamainternmed.2018.3931. PMC   6248200 . PMID   30264133.
  18. 1 2 DiMasi JA, Grabowski HG, Hansen RW (May 2016). "Innovation in the pharmaceutical industry: New estimates of R&D costs". Journal of Health Economics. 47: 20–33. doi:10.1016/j.jhealeco.2016.01.012. hdl: 10161/12742 . PMID   26928437.
  19. Taylor D (2015). "The Pharmaceutical Industry and the Future of Drug Development". Issues in Environmental Science and Technology. Royal Society of Chemistry: 1–33. doi:10.1039/9781782622345-00001. ISBN   978-1-78262-189-8.
  20. Sertkaya A, Wong HH, Jessup A, Beleche T (April 2016). "Key cost drivers of pharmaceutical clinical trials in the United States". Clinical Trials. 13 (2): 117–26. doi: 10.1177/1740774515625964 . PMID   26908540. S2CID   24308679.
  21. Herper M (11 August 2013). "The cost of creating a new drug now $5 billion, pushing Big Pharma to change". Forbes . Retrieved 17 July 2016.
  22. Maxmen A (August 2016). "Busting the billion-dollar myth: how to slash the cost of drug development". Nature. 536 (7617): 388–90. Bibcode:2016Natur.536..388M. doi: 10.1038/536388a . PMID   27558048.
  23. Whipple, Tom (October 23, 2021). "Moonshot is the spanner in the Covid-19 works the country needs". The Times. Retrieved 5 November 2021.
  24. Lee, Alpha; Chodera, John; von Delft, Frank (27 September 2021). "Why we are developing a patent-free Covid antiviral therapy". Knowable Magazine. doi: 10.1146/knowable-092721-1 . S2CID   244170138 . Retrieved 1 November 2021.
  25. Boris Bogdan and Ralph Villiger, "Valuation in Life Sciences. A Practical Guide", 2008, 2nd edition, Springer Verlag.
  26. Nielsen, Nicolaj Hoejer "Financial valuation methods for biotechnology", 2010. "Archived copy" (PDF). Archived from the original (PDF) on 2012-03-05. Retrieved 2014-11-25.{{cite web}}: CS1 maint: archived copy as title (link)
  27. Stratmann HG (September 2010). "Bad Medicine: When Medical Research Goes Wrong". Analog Science Fiction and Fact. CXXX (9): 20.
  28. "R&D costs are on the rise". Medical Marketing and Media. 38 (6): 14. June 1, 2003. Archived from the original on October 18, 2016.
  29. "Clinical Development Success Rates 2006-2015" (PDF). BIO Industry Analysis. June 2016.
  30. Wang Y. (2012). "Extracting Knowledge from Failed Development Programmes". Pharm Med. 26 (2): 91–96. doi:10.1007/BF03256897. S2CID   17171991.
  31. Herschel, M. (2012). "Portfolio Decisions in Early Development: Don't Throw Out the Baby with the Bathwater". Pharm Med. 26 (2): 77–84. doi:10.1007/BF03256895. S2CID   15782597. Archived from the original on 2012-06-16. Retrieved 2012-06-12.
  32. "About the Innovative Medicines Initiative". European Innovative Medicines Initiative. 2020. Retrieved 24 January 2020.
  33. "Critical Path Initiative". US Food and Drug Administration. 23 April 2018. Retrieved 24 January 2020.
  34. "Breakthrough Therapy". US Food and Drug Administration. 4 January 2018. Retrieved 24 January 2020.
  35. 1 2 Shankland S (2020-03-23). "Sixteen supercomputers tackle coronavirus cures in the US". CNET. ViacomCBS. Retrieved 27 April 2020.
  36. 1 2 "The COVID-19 High Performance Computing Consortium". The COVID-19 High Performance Computing Consortium. 2020. Retrieved 2020-04-27.
  37. Marshall S, Madabushi R, Manolis E, Krudys K, Staab A, Dykstra K, Visser SA (February 2019). "Model-Informed Drug Discovery and Development: Current Industry Good Practice and Regulatory Expectations and Future Perspectives". CPT. 8 (2): 87–96. doi: 10.1002/psp4.12372 . PMC   6389350 . PMID   30411538.
  38. "OpenPandemics – COVID-19". IBM. 2020. Retrieved 18 May 2020.