Jump to content

Drug development

From Wikipedia, the free encyclopedia
Drug discovery cycle schematic

Drug developmentis the process of bringing a newpharmaceutical drugto the market once alead compoundhas been identified through the process ofdrug discovery.It includespreclinical researchon microorganisms and animals, filing for regulatory status, such as via the United StatesFood and Drug Administrationfor aninvestigational new drugto initiateclinical trialson humans, and may include the step of obtainingregulatory approvalwith anew drug applicationto 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]

New chemical entity development

[edit]

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

Timeline showing the various drug approval tracks and research phases[5]

Pre-clinical

[edit]

New chemical entities(NCEs, also known as new molecular entities or NMEs) are compounds that emerge from the process ofdrug discovery.These have promising activity against a particular biological target that is important in disease. However, little is known about the safety,toxicity,pharmacokinetics,andmetabolismof 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 amedicinal chemistproducing milligrams, to manufacturing on the kilogram andtonscale. They further examine the product for suitability to package ascapsules,tablets,aerosol, intramuscular injectable, subcutaneous injectable, orintravenousformulations.Together, these processes are known in preclinical and clinical development aschemistry, manufacturing, and control(CMC).[citation needed]

Many aspects of drug development focus on satisfying the regulatory requirements for anew 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 usingin vitromethods (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 theFDA), as anInvestigational New Drug(IND) application. If the IND is approved, development moves to the clinical phase.

Clinical phase

[edit]

Clinical trialsinvolve four steps:[7]

  • Phase I trials, usually in healthy volunteers, determine safety and dosing.
  • Phase II trials are used to get an initial reading of efficacy and further explore safety in small numbers of patients having the disease targeted by the NCE.
  • Phase III trials are large, pivotal trials to determine safety and efficacy in sufficiently large numbers of patients with the targeted disease. If safety and efficacy are adequately proved, clinical testing may stop at this step and the NCE advances to thenew drug application(NDA) stage.
  • Phase IV trials are post-approval trials that are sometimes a condition attached by the FDA, also called post-market surveillance studies.

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 toxicside effects(50% failure of Phase IIcardiologytrials), 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

[edit]

There are a number of studies that have been conducted to determine research and development costs: notably, recent studies from DiMasi[15]and Wouters[16]suggest pre-approval capitalized cost estimates of $2.6 billion and $1.1 billion, respectively. The figures differ significantly based on methodologies, sampling and timeframe examined. Several other studies looking into specific therapeutic areas or disease types suggest as low as $291 million[17]for orphan drugs, $648 million[18]for cancer drugs or as high as $1.8 billion for cell and gene therapies[19].

The average cost (2013 dollars) ofeach stage of clinical researchwas US$25 million for a Phase I safety study, $59 million for a Phase IIrandomized controlled efficacy study,and $255 million for apivotal Phase III trialto demonstrate its equivalence or superiority to an existing approved drug,[20]possibly as high as $345 million.[21]The average cost of conducting a 2015–16 pivotal Phase III trial on an infectious disease drug candidate was $22 million.[21]

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][22][21][23]In a 2016 review of 106 drug candidates assessed through clinical trials, the totalcapital expenditurefor 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%.[20]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 forPhase IV trials for continuous safety surveillance.[24]

Alternatives to conventional drug development have the objective for universities, governments, and thepharmaceutical industryto collaborate and optimize resources.[25]An example of a collaborative drug development initiative isCOVID Moonshot,an internationalopen-scienceproject started in March 2020 with the goal of developing an un-patentedoralantiviral drugto treatSARS-CoV-2.[26][27]

Valuation

[edit]

The nature of a drug development project is characterised by highattrition 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 arerisk-adjusted net present value(rNPV),decision trees,real options,orcomparables.[citation needed]

The most important value drivers are the cost of capital or discount rate that is used,phaseattributes 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 thecash flowsestimation.[28][29]

Success rate

[edit]

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.[30]A study conducted by theTufts Center for the Study of Drug Developmentcovering the 1980s and 1990s found that only 21.5 percent of drugs that started Phase I trials were eventually approved for marketing.[31]In the time period of 2006 to 2015, the success rate was 9.6%.[32]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.[33]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.[34]

Computing initiatives

[edit]

Novel initiatives include partnering between governmental organizations and industry, such as the EuropeanInnovative Medicines Initiative.[35]The USFood and Drug Administrationcreated the"Critical Path Initiative"to enhance innovation of drug development,[36]and theBreakthrough Therapydesignation 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.[37]

In March 2020, theUnited States Department of Energy,National Science Foundation,NASA,industry, and nine universities pooled resources to access supercomputers fromIBM,combined with cloud computing resources fromHewlett Packard Enterprise,Amazon,Microsoft,andGoogle,for drug discovery.[38][39]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.[38][39][40]In May 2020, the OpenPandemics – COVID-19 partnership betweenScripps Researchand IBM'sWorld Community Gridwas 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".[41]

See also

[edit]

References

[edit]
  1. ^abStrovel 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.PMID22553881.
  2. ^abTaylor 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.ISBN978-1-78262-189-8.
  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.ISSN0362-1642.PMID27483339.Retrieved2 November2021.
  4. ^abcd"The Drug Development Process".U.S.Food and Drug Administration(FDA). 4 January 2018.Retrieved21 March2020.
  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.PMID7871409.
  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.PMC7202182.PMID31621554.
  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.PMID24796999.S2CID205100166.
  8. ^abcdStrovel 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.PMID22553881.
  9. ^"Vaccine Product Approval Process".U.S.Food and Drug Administration(FDA). 30 January 2018.Retrieved21 March2020.
  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.PMC6609997.PMID31312766.
  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.PMC6092479.PMID30112460.
  12. ^"R&D costs are on the rise".Medical Marketing and Media.38(6): 14. June 1, 2003. Archived fromthe originalon October 18, 2016.
  13. ^ab"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.S2CID17171991.
  15. ^DiMasi, Joseph A.; Grabowski, Henry G.; Hansen, Ronald W. (2016-05-01)."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.ISSN0167-6296.
  16. ^Wouters, Olivier J.; McKee, Martin; Luyten, Jeroen (2020-03-03)."Estimated Research and Development Investment Needed to Bring a New Medicine to Market, 2009-2018".JAMA.323(9): 844–853.doi:10.1001/jama.2020.1166.ISSN0098-7484.
  17. ^Jayasundara, Kavisha; Hollis, Aidan; Krahn, Murray; Mamdani, Muhammad; Hoch, Jeffrey S.; Grootendorst, Paul (2019-01-10)."Estimating the clinical cost of drug development for orphan versus non-orphan drugs".Orphanet Journal of Rare Diseases.14(1): 12.doi:10.1186/s13023-018-0990-4.ISSN1750-1172.PMC6327525.PMID30630499.{{cite journal}}:CS1 maint: unflagged free DOI (link)
  18. ^jamanetwork.comhttps://jamanetwork.com/journals/jamainternalmedicine/fullarticle/2653012.Retrieved2024-10-04.{{cite web}}:Missing or empty|title=(help)
  19. ^Sabatini, Marco T.; Chalmers, Mark (September 2023)."The Cost of Biotech Innovation: Exploring Research and Development Costs of Cell and Gene Therapies".Pharmaceutical Medicine.37(5): 365–375.doi:10.1007/s40290-023-00480-0.ISSN1179-1993.PMID37286928.
  20. ^abDiMasi 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.PMID26928437.
  21. ^abcMoore 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.PMC6248200.PMID30264133.
  22. ^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.ISBN978-1-78262-189-8.
  23. ^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.PMID26908540.S2CID24308679.
  24. ^Herper M (11 August 2013)."The cost of creating a new drug now $5 billion, pushing Big Pharma to change".Forbes.Retrieved17 July2016.
  25. ^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.PMID27558048.
  26. ^Whipple, Tom (October 23, 2021)."Moonshot is the spanner in the Covid-19 works the country needs".The Times.Retrieved5 November2021.
  27. ^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.S2CID244170138.Retrieved1 November2021.
  28. ^Boris Bogdan and Ralph Villiger, "Valuation in Life Sciences. A Practical Guide", 2008, 2nd edition, Springer Verlag.
  29. ^Nielsen, Nicolaj Hoejer "Financial valuation methods for biotechnology", 2010."Archived copy"(PDF).Archived fromthe original(PDF)on 2012-03-05.Retrieved2014-11-25.{{cite web}}:CS1 maint: archived copy as title (link)
  30. ^Stratmann HG (September 2010). "Bad Medicine: When Medical Research Goes Wrong".Analog Science Fiction and Fact.CXXX(9): 20.
  31. ^"R&D costs are on the rise".Medical Marketing and Media.38(6): 14. June 1, 2003. Archived fromthe originalon October 18, 2016.
  32. ^"Clinical Development Success Rates 2006-2015"(PDF).BIO Industry Analysis.June 2016.
  33. ^Wang Y. (2012)."Extracting Knowledge from Failed Development Programmes".Pharm Med.26(2): 91–96.doi:10.1007/BF03256897.S2CID17171991.
  34. ^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.S2CID15782597.Archived fromthe originalon 2012-06-16.Retrieved2012-06-12.
  35. ^"About the Innovative Medicines Initiative".European Innovative Medicines Initiative. 2020.Retrieved24 January2020.
  36. ^"Critical Path Initiative".US Food and Drug Administration. 23 April 2018.Retrieved24 January2020.
  37. ^"Breakthrough Therapy".US Food and Drug Administration. 4 January 2018.Retrieved24 January2020.
  38. ^abShankland S (2020-03-23)."Sixteen supercomputers tackle coronavirus cures in the US".CNET.ViacomCBS.Retrieved27 April2020.
  39. ^ab"The COVID-19 High Performance Computing Consortium".The COVID-19 High Performance Computing Consortium. 2020.Retrieved2020-04-27.
  40. ^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: Pharmacometrics & Systems Pharmacology.8(2): 87–96.doi:10.1002/psp4.12372.PMC6389350.PMID30411538.
  41. ^"OpenPandemics – COVID-19".IBM. 2020.Retrieved18 May2020.
[edit]