fbpx
Wikipedia

Phases of clinical research

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.[1] For drug development, the clinical phases start with testing for drug safety in a few human subjects, then expand to many study participants (potentially tens of thousands) to determine if the treatment is effective.[1] Clinical research is conducted on drug candidates, vaccine candidates, new medical devices, and new diagnostic assays.

National Cancer Institute video on clinical trial phases

Description edit

Clinical trials testing potential medical products are commonly classified into four phases. The drug development process will normally proceed through all four phases over many years.[1] When expressed specifically, a clinical trial phase is capitalized both in name and Roman numeral, such as "Phase I" clinical trial.[1]

If the drug successfully passes through Phases I, II, and III, it will usually be approved by the national regulatory authority for use in the general population.[1] Phase IV trials are 'post-marketing' or 'surveillance' studies conducted to monitor safety over several years.[1]

Summary of clinical trial phases
Phase Primary goal Dose Patient monitor Typical number of participants Success rate[2] Notes
Preclinical Testing of drug in non-human subjects to gather efficacy, toxicity and pharmacokinetic information Unrestricted Scientific researcher No human subjects, in vitro and in vivo only Includes testing in model organisms. Human immortalized cell lines and other human tissues may also be used.
Phase 0 Pharmacokinetics; particularly oral bioavailability and half-life of the drug Small, subtherapeutic Clinical researcher 10 people Often skipped for Phase I.
Phase I Dose-ranging on healthy volunteers for safety Often subtherapeutic, but with ascending doses Clinical researcher 20–100 normal healthy volunteers (or cancer patients for cancer drugs) Approx. 52% Determines whether drug is safe to check for efficacy.
Phase II Testing of drug on participants to assess efficacy and side effects Therapeutic dose Clinical researcher 100–300 participants with a specific disease Approx. 28.9% Determines whether drug can have any efficacy; at this point, the drug is not presumed to have any therapeutic effect
Phase III Testing of drug on participants to assess efficacy, effectiveness and safety Therapeutic dose Clinical researcher and personal physician 300–3,000 people with a specific disease 57.8% Determines a drug's therapeutic effect; at this point, the drug is presumed to have some effect
Phase IV Post marketing surveillance in public Therapeutic dose Personal physician Anyone seeking treatment from a physician N/A Monitor long-term effects

Preclinical studies edit

Before clinical trials are undertaken for a candidate drug, vaccine, medical device, or diagnostic assay, the product candidate is tested extensively in preclinical studies.[1] Such studies involve in vitro (test tube or cell culture) and in vivo (animal model) experiments using wide-ranging doses of the study agent to obtain preliminary efficacy, toxicity and pharmacokinetic information. Such tests assist the developer to decide whether a drug candidate has scientific merit for further development as an investigational new drug.[1]

Phase 0 edit

Phase 0 is a recent designation for optional exploratory trials conducted in accordance with the United States Food and Drug Administration's (FDA) 2006 Guidance on Exploratory Investigational New Drug (IND) Studies.[3] Phase 0 trials are also known as human microdosing studies and are designed to speed up the development of promising drugs or imaging agents by establishing very early on whether the drug or agent behaves in human subjects as was expected from preclinical studies. Distinctive features of Phase 0 trials include the administration of single subtherapeutic doses of the study drug to a small number of subjects (10 to 15) to gather preliminary data on the agent's pharmacokinetics (what the body does to the drugs).[4]

A Phase 0 study gives no data on safety or efficacy, being by definition a dose too low to cause any therapeutic effect. Drug development companies carry out Phase 0 studies to rank drug candidates to decide which has the best pharmacokinetic parameters in humans to take forward into further development. They enable go/no-go decisions to be based on relevant human models instead of relying on sometimes inconsistent animal data.[5]

Phase I edit

Phase I trials were formerly referred to as "first-in-man studies" but the field generally moved to the gender-neutral language phrase "first-in-humans" in the 1990s;[6] these trials are the first stage of testing in human subjects.[7] They are designed to test the safety, side effects, best dose, and formulation method for the drug.[8] Phase I trials are not randomized, and thus are vulnerable to selection bias.[9]

Normally, a small group of 20–100 healthy volunteers will be recruited.[10][7] These trials are often conducted in a clinical trial clinic, where the subject can be observed by full-time staff. These clinical trial clinics are often run by contract research organization (CROs) who conduct these studies on behalf of pharmaceutical companies or other research investigators.[citation needed]

The subject who receives the drug is usually observed until several half-lives of the drug have passed. This phase is designed to assess the safety (pharmacovigilance), tolerability, pharmacokinetics, and pharmacodynamics of a drug. Phase I trials normally include dose-ranging, also called dose escalation studies, so that the best and safest dose can be found and to discover the point at which a compound is too poisonous to administer.[11] The tested range of doses will usually be a fraction[quantify] of the dose that caused harm in animal testing.

Phase I trials most often include healthy volunteers. However, there are some circumstances when clinical patients are used, such as patients who have terminal cancer or HIV and the treatment is likely to make healthy individuals ill. These studies are usually conducted in tightly controlled clinics called Central Pharmacological Units, where participants receive 24-hour medical attention and oversight. In addition to the previously mentioned unhealthy individuals, "patients who have typically already tried and failed to improve on the existing standard therapies"[12] may also participate in Phase I trials. Volunteers are paid a variable inconvenience fee for their time spent in the volunteer center.

Before beginning a Phase I trial, the sponsor must submit an Investigational New Drug application to the FDA detailing the preliminary data on the drug gathered from cellular models and animal studies.[citation needed]

Phase I trials can be further divided:

Phase Ia edit

Single ascending dose (Phase Ia): In single ascending dose studies, small groups of subjects are given a single dose of the drug while they are observed and tested for a period of time to confirm safety.[7][13] Typically, a small number of participants, usually three, are entered sequentially at a particular dose.[12] If they do not exhibit any adverse side effects, and the pharmacokinetic data are roughly in line with predicted safe values, the dose is escalated, and a new group of subjects is then given a higher dose.[citation needed]

If unacceptable toxicity is observed in any of the three participants, an additional number of participants, usually three, are treated at the same dose.[12] This is continued until pre-calculated pharmacokinetic safety levels are reached, or intolerable side effects start showing up (at which point the drug is said to have reached the maximum tolerated dose (MTD)). If an additional unacceptable toxicity is observed, then the dose escalation is terminated and that dose, or perhaps the previous dose, is declared to be the maximally tolerated dose. This particular design assumes that the maximally tolerated dose occurs when approximately one-third of the participants experience unacceptable toxicity. Variations of this design exist, but most are similar.[12]

Phase Ib edit

Multiple ascending dose (Phase Ib): Multiple ascending dose studies investigate the pharmacokinetics and pharmacodynamics of multiple doses of the drug, looking at safety and tolerability. In these studies, a group of patients receives multiple low doses of the drug, while samples (of blood, and other fluids) are collected at various time points and analyzed to acquire information on how the drug is processed within the body. The dose is subsequently escalated for further groups, up to a predetermined level.[7][13]

Food effect edit

A short trial designed to investigate any differences in absorption of the drug by the body, caused by eating before the drug is given. These studies are usually run as a crossover study, with volunteers being given two identical doses of the drug while fasted, and after being fed.

Phase II edit

Once a dose or range of doses is determined, the next goal is to evaluate whether the drug has any biological activity or effect.[12] Phase II trials are performed on larger groups (50–300 individuals) and are designed to assess how well the drug works, as well as to continue Phase I safety assessments in a larger group of volunteers and patients. Genetic testing is common, particularly when there is evidence of variation in metabolic rate.[12] When the development process for a new drug fails, this usually occurs during Phase II trials when the drug is discovered not to work as planned, or to have toxic effects.[citation needed]

Phase II studies are sometimes divided into Phase IIa and Phase IIb. There is no formal definition for these two sub-categories, but generally:

  • Phase IIa studies are usually pilot studies designed to find an optimal dose and assess safety ('dose finding' studies).[14]
  • Phase IIb studies determine how well the drug works in subjects at a given dose to assess efficacy ('proof of concept' studies).[14]

Trial design edit

Some Phase II trials are designed as case series, demonstrating a drug's safety and activity in a selected group of participants. Other Phase II trials are designed as randomized controlled trials, where some patients receive the drug/device and others receive placebo/standard treatment. Randomized Phase II trials have far fewer patients than randomized Phase III trials.[citation needed]

Example: cancer design edit

In the first stage, the investigator attempts to rule out drugs that have no or little biologic activity. For example, the researcher may specify that a drug must have some minimal level of activity, say, in 20% of participants. If the estimated activity level is less than 20%, the researcher chooses not to consider this drug further, at least not at that maximally tolerated dose. If the estimated activity level exceeds 20%, the researcher will add more participants to get a better estimate of the response rate. A typical study for ruling out a 20% or lower response rate enters 14 participants. If no response is observed in the first 14 participants, the drug is considered not likely to have a 20% or higher activity level. The number of additional participants added depends on the degree of precision desired, but ranges from 10 to 20. Thus, a typical cancer phase II study might include fewer than 30 people to estimate the response rate.[12]

Efficacy vs effectiveness edit

When a study assesses efficacy, it is looking at whether the drug given in the specific manner described in the study is able to influence an outcome of interest (e.g. tumor size) in the chosen population (e.g. cancer patients with no other ongoing diseases). When a study is assessing effectiveness, it is determining whether a treatment will influence the disease. In an effectiveness study, it is essential that participants are treated as they would be when the treatment is prescribed in actual practice. That would mean that there should be no aspects of the study designed to increase compliance above those that would occur in routine clinical practice. The outcomes in effectiveness studies are also more generally applicable than in most efficacy studies (for example does the patient feel better, come to the hospital less or live longer in effectiveness studies as opposed to better test scores or lower cell counts in efficacy studies). There is usually less rigid control of the type of participant to be included in effectiveness studies than in efficacy studies, as the researchers are interested in whether the drug will have a broad effect in the population of patients with the disease.[citation needed]

Success rate edit

Phase II clinical programs historically have experienced the lowest success rate of the four development phases. In 2010, the percentage of Phase II trials that proceeded to Phase III was 18%,[15] and only 31% of developmental candidates advanced from Phase II to Phase III in a study of trials over 2006–2015.[16]

Phase III edit

This phase is designed to assess the effectiveness of the new intervention and, thereby, its value in clinical practice.[12] Phase III studies are randomized controlled multicenter trials on large patient groups (300–3,000 or more depending upon the disease/medical condition studied) and are aimed at being the definitive assessment of how effective the drug is, in comparison with current 'gold standard' treatment. Because of their size and comparatively long duration, Phase III trials are the most expensive, time-consuming and difficult trials to design and run, especially in therapies for chronic medical conditions. Phase III trials of chronic conditions or diseases often have a short follow-up period for evaluation, relative to the period of time the intervention might be used in practice.[12] This is sometimes called the "pre-marketing phase" because it actually measures consumer response to the drug.[citation needed]

It is common practice that certain Phase III trials will continue while the regulatory submission is pending at the appropriate regulatory agency. This allows patients to continue to receive possibly lifesaving drugs until the drug can be obtained by purchase. Other reasons for performing trials at this stage include attempts by the sponsor at "label expansion" (to show the drug works for additional types of patients/diseases beyond the original use for which the drug was approved for marketing), to obtain additional safety data, or to support marketing claims for the drug. Studies in this phase are by some companies categorized as "Phase IIIB studies."[17]

While not required in all cases, it is typically expected that there be at least two successful Phase III trials, demonstrating a drug's safety and efficacy, to obtain approval from the appropriate regulatory agencies such as FDA (US), or the EMA (European Union).

Once a drug has proved satisfactory after Phase III trials, the trial results are usually combined into a large document containing a comprehensive description of the methods and results of human and animal studies, manufacturing procedures, formulation details, and shelf life. This collection of information makes up the "regulatory submission" that is provided for review to the appropriate regulatory authorities[18] in different countries. They will review the submission, and if it is acceptable, give the sponsor approval to market the drug.

Most drugs undergoing Phase III clinical trials can be marketed under FDA norms with proper recommendations and guidelines through a New Drug Application (NDA) containing all manufacturing, preclinical, and clinical data. In case of any adverse effects being reported anywhere, the drugs need to be recalled immediately from the market. While most pharmaceutical companies refrain from this practice, it is not abnormal to see many drugs undergoing Phase III clinical trials in the market.[19]

Adaptive design edit

The design of individual trials may be altered during a trial – usually during Phase II or III – to accommodate interim results for the benefit of the treatment, adjust statistical analysis, or to reach early termination of an unsuccessful design, a process called an "adaptive design".[20][21][22] Examples are the 2020 World Health Organization Solidarity trial, European Discovery trial, and UK RECOVERY Trial of hospitalized people with severe COVID-19 infection, each of which applies adaptive designs to rapidly alter trial parameters as results from the experimental therapeutic strategies emerge.[23][24][25]

Adaptive designs within ongoing Phase II–III clinical trials on candidate therapeutics may shorten trial durations and use fewer subjects, possibly expediting decisions for early termination or success, and coordinating design changes for a specific trial across its international locations.[22]

Success rate edit

For vaccines, the probability of success ranges from 7% for non-industry-sponsored candidates to 40% for industry-sponsored candidates.[26]

A 2019 review of average success rates of clinical trials at different phases and diseases over the years 2005–15 found a success range of 5–14%.[27] Separated by diseases studied, cancer drug trials were on average only 3% successful, whereas ophthalmology drugs and vaccines for infectious diseases were 33% successful.[27] Trials using disease biomarkers, especially in cancer studies, were more successful than those not using biomarkers.[27]

A 2010 review found about 50% of drug candidates either fail during the Phase III trial or are rejected by the national regulatory agency.[28]

Cost of trials by phases edit

In the early 21st century, a typical Phase I trial conducted at a single clinic in the United States ranged from $1.4 million for pain or anesthesia studies to $6.6 million for immunomodulation studies.[29] Main expense drivers were operating and clinical monitoring costs of the Phase I site.[29]

The amount of money spent on Phase II or III trials depends on numerous factors, with therapeutic area being studied and types of clinical procedures as key drivers.[29] Phase II studies may cost as low as $7 million for cardiovascular projects, and as much as $20 million for hematology trials.[29]

Phase III trials for dermatology may cost as low as $11 million, whereas a pain or anesthesia Phase III trial may cost as much as $53 million.[29] An analysis of Phase III pivotal trials leading to 59 drug approvals by the US Food and Drug Administration over 2015–16 showed that the median cost was $19 million, but some trials involving thousands of subjects may cost 100 times more.[30]

Across all trial phases, the main expenses for clinical trials were administrative staff (about 20% of the total), clinical procedures (about 19%), and clinical monitoring of the subjects (about 11%).[29]

Phase IV edit

A Phase IV trial is also known as a postmarketing surveillance trial or drug monitoring trial to assure long-term safety and effectiveness of the drug, vaccine, device or diagnostic test.[1] Phase IV trials involve the safety surveillance (pharmacovigilance) and ongoing technical support of a drug after it receives regulatory approval to be sold.[7] Phase IV studies may be required by regulatory authorities or may be undertaken by the sponsoring company for competitive (finding a new market for the drug) or other reasons (for example, the drug may not have been tested for interactions with other drugs, or on certain population groups such as pregnant women, who are unlikely to subject themselves to trials).[10][7] The safety surveillance is designed to detect any rare or long-term adverse effects over a much larger patient population and longer time period than was possible during the Phase I-III clinical trials.[7] Harmful effects discovered by Phase IV trials may result in a drug being withdrawn from the market or restricted to certain uses; examples include cerivastatin (brand names Baycol and Lipobay), troglitazone (Rezulin) and rofecoxib (Vioxx).[citation needed]

Overall cost edit

The entire process of developing a drug from preclinical research to marketing can take approximately 12 to 18 years and often costs well over $1 billion.[31][32]

References edit

  1. ^ a b c d e f g h i "The drug development process". US Food and Drug Administration. 4 January 2018. Retrieved 17 August 2020.
  2. ^ "New Clinical Development Success Rates 2011–2020 Report". BIO, Informa Pharma Intelligence, and QLS Advisors. Feb 2021.
  3. ^ "Exploratory IND Studies, Guidance for Industry, Investigators, and Reviewers" (PDF). Food and Drug Administration. January 2006. Retrieved 2010-06-15.
  4. ^ The Lancet (July 2009). "Phase 0 trials: a platform for drug development?". Lancet. 374 (9685): 176. doi:10.1016/S0140-6736(09)61309-X. PMID 19616703. S2CID 30939770.
  5. ^ Burt, Tal; Young, Graeme; Lee, Wooin; Kusuhara, Hiroyuki; Langer, Oliver; Rowland, Malcolm; Sugiyama, Yuichi (2020). "Phase 0/microdosing approaches: time for mainstream application in drug development?". Nature Reviews Drug Discovery. 19 (11): 801–818. doi:10.1038/s41573-020-0080-x. ISSN 1474-1784. PMID 32901140.
  6. ^ Fisher JA (March 2015). "Feeding and Bleeding: The Institutional Banalization of Risk to Healthy Volunteers in Phase I Pharmaceutical Clinical Trials". Science, Technology, & Human Values. 40 (2): 199–226. doi:10.1177/0162243914554838. PMC 4405793. PMID 25914430.
  7. ^ a b c d e f g "Types and phases of clinical trials". American Cancer Society. 18 August 2020. Retrieved 15 September 2020.
  8. ^ "NCI Dictionary". National Cancer Institute. 2011-02-02.
  9. ^ Van den Eynde BJ, van Baren N, Baurain JF (2020). "Is There a Clinical Future for IDO1 Inhibitors After the Failure of Epacadostat in Melanoma?". Annual Review of Cancer Biology. 4: 241–256. doi:10.1146/annurev-cancerbio-030419-033635.
  10. ^ a b "Step 3. Clinical research". US Food and Drug Administration. 14 October 2016. Retrieved 1 February 2017.
  11. ^ Shamoo AE (2008). "The myth of equipoise in phase 1 clinical trials". Medscape Journal of Medicine. 10 (11): 254. PMC 2605120. PMID 19099004.(registration required)
  12. ^ a b c d e f g h i DeMets D, Friedman L, Furberg C (2010). Fundamentals of Clinical Trials (4th ed.). Springer. ISBN 978-1-4419-1585-6.
  13. ^ a b Norfleet E, Gad SC (2009). "Phase I Clinical Trials". In Gad SC (ed.). Clinical Trials Handbook. John Wiley & Sons. p. 247. ISBN 978-0-470-46635-3.
  14. ^ a b "Phase 1, 2, 3, 4 trials". Revive, Global Antiobiotic Research and Development Partnership. 2023. Retrieved 24 October 2023.
  15. ^ "New drugs failing Phase II and III clinical trials". MedCity News. 2011-06-02.
  16. ^ "Clinical Development Success Rates 2006–2015" (PDF). bio.org. Retrieved 2018-02-11.
  17. ^ "Guidance for Institutional Review Boards and Clinical Investigators". Food and Drug Administration. 1999-03-16. Retrieved 2007-03-27.
  18. ^ The regulatory authority in the US is the Food and Drug Administration; in Canada, Health Canada; in the European Union, the European Medicines Agency; and in Japan, the Ministry of Health, Labour and Welfare
  19. ^ Arcangelo VP, Peterson AM (2005). Pharmacotherapeutics for Advanced Practice: A Practical Approach. Lippincott Williams & Wilkins. ISBN 978-0-7817-5784-3.
  20. ^ 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.
  21. ^ "Adaptive Designs for Clinical Trials of Drugs and Biologics: Guidance for Industry" (PDF). U.S. Food and Drug Administration. 1 November 2019. Retrieved 3 April 2020.
  22. ^ a b Pallmann P, Bedding AW, Choodari-Oskooei B, Dimairo M, Flight L, Hampson LV, et al. (February 2018). "Adaptive designs in clinical trials: why use them, and how to run and report them". BMC Medicine. 16 (1): 29. doi:10.1186/s12916-018-1017-7. PMC 5830330. PMID 29490655.
  23. ^ Kotok A (19 March 2020). "WHO beginning Covid-19 therapy trial". Technology News: Science and Enterprise. Retrieved 7 April 2020.
  24. ^ "Launch of a European clinical trial against COVID-19". INSERM. 22 March 2020. Retrieved 5 April 2020. The great strength of this trial is its "adaptive" nature. This means that ineffective experimental treatments can very quickly be dropped and replaced by other molecules that emerge from research efforts. We will therefore be able to make changes in real time, in line with the most recent scientific data, in order to find the best treatment for our patients
  25. ^ "RECOVERY Trial". Retrieved 17 June 2020.
  26. ^ Lo A, Siah K, Wong C (14 May 2020). "Estimating probabilities of success of vaccine and other anti-infective therapeutic development programs". Harvard Data Science Review. MIT Press (Special Issue 1 – COVID-19). doi:10.1162/99608f92.e0c150e8. Retrieved 11 August 2020. we can see that the overall probability of success (PoS) for industry-sponsored vaccine development programs is 39.6%... In contrast, non-industry-sponsored vaccine development programs have an overall PoS of only 6.8%
  27. ^ a b c Wong, Chi Heem; Siah, Kien Wei; Lo, Andrew W (2018-01-31). "Estimation of clinical trial success rates and related parameters". Biostatistics. 20 (2): 273–286. doi:10.1093/biostatistics/kxx069. ISSN 1465-4644. PMC 6409418. PMID 29394327. S2CID 3277297.
  28. ^ Arrowsmith J (February 2011). "Trial watch: phase III and submission failures: 2007–2010". Nature Reviews. Drug Discovery. 10 (2): 87. doi:10.1038/nrd3375. PMID 21283095. S2CID 39480483.
  29. ^ a b c d e f 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.
  30. ^ 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.
  31. ^ Holland J (2013). "Fixing a broken drug development process". Journal of Commercial Biotechnology. 19. doi:10.5912/jcb588.
  32. ^ Adams CP, Brantner VV (2006). "Estimating the cost of new drug development: is it really 802 million dollars?". Health Affairs. 25 (2): 420–8. doi:10.1377/hlthaff.25.2.420. PMID 16522582.

phases, clinical, research, broader, coverage, this, topic, clinical, trial, phases, clinical, research, stages, which, scientists, conduct, experiments, with, health, intervention, obtain, sufficient, evidence, process, considered, effective, medical, treatme. For broader coverage of this topic see Clinical trial 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 1 For drug development the clinical phases start with testing for drug safety in a few human subjects then expand to many study participants potentially tens of thousands to determine if the treatment is effective 1 Clinical research is conducted on drug candidates vaccine candidates new medical devices and new diagnostic assays source source source source source source source source National Cancer Institute video on clinical trial phases Contents 1 Description 2 Preclinical studies 3 Phase 0 4 Phase I 4 1 Phase Ia 4 2 Phase Ib 4 3 Food effect 5 Phase II 5 1 Trial design 5 1 1 Example cancer design 5 1 2 Efficacy vs effectiveness 5 2 Success rate 6 Phase III 6 1 Adaptive design 6 2 Success rate 7 Cost of trials by phases 8 Phase IV 9 Overall cost 10 ReferencesDescription editClinical trials testing potential medical products are commonly classified into four phases The drug development process will normally proceed through all four phases over many years 1 When expressed specifically a clinical trial phase is capitalized both in name and Roman numeral such as Phase I clinical trial 1 If the drug successfully passes through Phases I II and III it will usually be approved by the national regulatory authority for use in the general population 1 Phase IV trials are post marketing or surveillance studies conducted to monitor safety over several years 1 Summary of clinical trial phases Phase Primary goal Dose Patient monitor Typical number of participants Success rate 2 NotesPreclinical wbr Testing of drug in non human subjects to gather efficacy toxicity and pharmacokinetic information Unrestricted Scientific researcher No human subjects in vitro and in vivo only Includes testing in model organisms Human immortalized cell lines and other human tissues may also be used Phase 0 wbr Pharmacokinetics particularly oral bioavailability and half life of the drug Small subtherapeutic Clinical researcher 10 people Often skipped for Phase I Phase I wbr Dose ranging on healthy volunteers for safety Often subtherapeutic but with ascending doses Clinical researcher 20 100 normal healthy volunteers or cancer patients for cancer drugs Approx 52 Determines whether drug is safe to check for efficacy Phase II wbr Testing of drug on participants to assess efficacy and side effects Therapeutic dose Clinical researcher 100 300 participants with a specific disease Approx 28 9 Determines whether drug can have any efficacy at this point the drug is not presumed to have any therapeutic effectPhase III wbr Testing of drug on participants to assess efficacy effectiveness and safety Therapeutic dose Clinical researcher and personal physician 300 3 000 people with a specific disease 57 8 Determines a drug s therapeutic effect at this point the drug is presumed to have some effectPhase IV wbr Post marketing surveillance in public Therapeutic dose Personal physician Anyone seeking treatment from a physician N A Monitor long term effectsPreclinical studies editMain article Preclinical development Before clinical trials are undertaken for a candidate drug vaccine medical device or diagnostic assay the product candidate is tested extensively in preclinical studies 1 Such studies involve in vitro test tube or cell culture and in vivo animal model experiments using wide ranging doses of the study agent to obtain preliminary efficacy toxicity and pharmacokinetic information Such tests assist the developer to decide whether a drug candidate has scientific merit for further development as an investigational new drug 1 Phase 0 editPhase 0 is a recent designation for optional exploratory trials conducted in accordance with the United States Food and Drug Administration s FDA 2006 Guidance on Exploratory Investigational New Drug IND Studies 3 Phase 0 trials are also known as human microdosing studies and are designed to speed up the development of promising drugs or imaging agents by establishing very early on whether the drug or agent behaves in human subjects as was expected from preclinical studies Distinctive features of Phase 0 trials include the administration of single subtherapeutic doses of the study drug to a small number of subjects 10 to 15 to gather preliminary data on the agent s pharmacokinetics what the body does to the drugs 4 A Phase 0 study gives no data on safety or efficacy being by definition a dose too low to cause any therapeutic effect Drug development companies carry out Phase 0 studies to rank drug candidates to decide which has the best pharmacokinetic parameters in humans to take forward into further development They enable go no go decisions to be based on relevant human models instead of relying on sometimes inconsistent animal data 5 Phase I editPhase I trials were formerly referred to as first in man studies but the field generally moved to the gender neutral language phrase first in humans in the 1990s 6 these trials are the first stage of testing in human subjects 7 They are designed to test the safety side effects best dose and formulation method for the drug 8 Phase I trials are not randomized and thus are vulnerable to selection bias 9 Normally a small group of 20 100 healthy volunteers will be recruited 10 7 These trials are often conducted in a clinical trial clinic where the subject can be observed by full time staff These clinical trial clinics are often run by contract research organization CROs who conduct these studies on behalf of pharmaceutical companies or other research investigators citation needed The subject who receives the drug is usually observed until several half lives of the drug have passed This phase is designed to assess the safety pharmacovigilance tolerability pharmacokinetics and pharmacodynamics of a drug Phase I trials normally include dose ranging also called dose escalation studies so that the best and safest dose can be found and to discover the point at which a compound is too poisonous to administer 11 The tested range of doses will usually be a fraction quantify of the dose that caused harm in animal testing Phase I trials most often include healthy volunteers However there are some circumstances when clinical patients are used such as patients who have terminal cancer or HIV and the treatment is likely to make healthy individuals ill These studies are usually conducted in tightly controlled clinics called Central Pharmacological Units where participants receive 24 hour medical attention and oversight In addition to the previously mentioned unhealthy individuals patients who have typically already tried and failed to improve on the existing standard therapies 12 may also participate in Phase I trials Volunteers are paid a variable inconvenience fee for their time spent in the volunteer center Before beginning a Phase I trial the sponsor must submit an Investigational New Drug application to the FDA detailing the preliminary data on the drug gathered from cellular models and animal studies citation needed Phase I trials can be further divided Phase Ia edit Single ascending dose Phase Ia In single ascending dose studies small groups of subjects are given a single dose of the drug while they are observed and tested for a period of time to confirm safety 7 13 Typically a small number of participants usually three are entered sequentially at a particular dose 12 If they do not exhibit any adverse side effects and the pharmacokinetic data are roughly in line with predicted safe values the dose is escalated and a new group of subjects is then given a higher dose citation needed If unacceptable toxicity is observed in any of the three participants an additional number of participants usually three are treated at the same dose 12 This is continued until pre calculated pharmacokinetic safety levels are reached or intolerable side effects start showing up at which point the drug is said to have reached the maximum tolerated dose MTD If an additional unacceptable toxicity is observed then the dose escalation is terminated and that dose or perhaps the previous dose is declared to be the maximally tolerated dose This particular design assumes that the maximally tolerated dose occurs when approximately one third of the participants experience unacceptable toxicity Variations of this design exist but most are similar 12 Phase Ib edit Multiple ascending dose Phase Ib Multiple ascending dose studies investigate the pharmacokinetics and pharmacodynamics of multiple doses of the drug looking at safety and tolerability In these studies a group of patients receives multiple low doses of the drug while samples of blood and other fluids are collected at various time points and analyzed to acquire information on how the drug is processed within the body The dose is subsequently escalated for further groups up to a predetermined level 7 13 Food effect edit A short trial designed to investigate any differences in absorption of the drug by the body caused by eating before the drug is given These studies are usually run as a crossover study with volunteers being given two identical doses of the drug while fasted and after being fed Phase II editOnce a dose or range of doses is determined the next goal is to evaluate whether the drug has any biological activity or effect 12 Phase II trials are performed on larger groups 50 300 individuals and are designed to assess how well the drug works as well as to continue Phase I safety assessments in a larger group of volunteers and patients Genetic testing is common particularly when there is evidence of variation in metabolic rate 12 When the development process for a new drug fails this usually occurs during Phase II trials when the drug is discovered not to work as planned or to have toxic effects citation needed Phase II studies are sometimes divided into Phase IIa and Phase IIb There is no formal definition for these two sub categories but generally Phase IIa studies are usually pilot studies designed to find an optimal dose and assess safety dose finding studies 14 Phase IIb studies determine how well the drug works in subjects at a given dose to assess efficacy proof of concept studies 14 Trial design edit Some Phase II trials are designed as case series demonstrating a drug s safety and activity in a selected group of participants Other Phase II trials are designed as randomized controlled trials where some patients receive the drug device and others receive placebo standard treatment Randomized Phase II trials have far fewer patients than randomized Phase III trials citation needed Example cancer design edit In the first stage the investigator attempts to rule out drugs that have no or little biologic activity For example the researcher may specify that a drug must have some minimal level of activity say in 20 of participants If the estimated activity level is less than 20 the researcher chooses not to consider this drug further at least not at that maximally tolerated dose If the estimated activity level exceeds 20 the researcher will add more participants to get a better estimate of the response rate A typical study for ruling out a 20 or lower response rate enters 14 participants If no response is observed in the first 14 participants the drug is considered not likely to have a 20 or higher activity level The number of additional participants added depends on the degree of precision desired but ranges from 10 to 20 Thus a typical cancer phase II study might include fewer than 30 people to estimate the response rate 12 Efficacy vs effectiveness edit When a study assesses efficacy it is looking at whether the drug given in the specific manner described in the study is able to influence an outcome of interest e g tumor size in the chosen population e g cancer patients with no other ongoing diseases When a study is assessing effectiveness it is determining whether a treatment will influence the disease In an effectiveness study it is essential that participants are treated as they would be when the treatment is prescribed in actual practice That would mean that there should be no aspects of the study designed to increase compliance above those that would occur in routine clinical practice The outcomes in effectiveness studies are also more generally applicable than in most efficacy studies for example does the patient feel better come to the hospital less or live longer in effectiveness studies as opposed to better test scores or lower cell counts in efficacy studies There is usually less rigid control of the type of participant to be included in effectiveness studies than in efficacy studies as the researchers are interested in whether the drug will have a broad effect in the population of patients with the disease citation needed Success rate edit Phase II clinical programs historically have experienced the lowest success rate of the four development phases In 2010 the percentage of Phase II trials that proceeded to Phase III was 18 15 and only 31 of developmental candidates advanced from Phase II to Phase III in a study of trials over 2006 2015 16 Phase III editThis phase is designed to assess the effectiveness of the new intervention and thereby its value in clinical practice 12 Phase III studies are randomized controlled multicenter trials on large patient groups 300 3 000 or more depending upon the disease medical condition studied and are aimed at being the definitive assessment of how effective the drug is in comparison with current gold standard treatment Because of their size and comparatively long duration Phase III trials are the most expensive time consuming and difficult trials to design and run especially in therapies for chronic medical conditions Phase III trials of chronic conditions or diseases often have a short follow up period for evaluation relative to the period of time the intervention might be used in practice 12 This is sometimes called the pre marketing phase because it actually measures consumer response to the drug citation needed It is common practice that certain Phase III trials will continue while the regulatory submission is pending at the appropriate regulatory agency This allows patients to continue to receive possibly lifesaving drugs until the drug can be obtained by purchase Other reasons for performing trials at this stage include attempts by the sponsor at label expansion to show the drug works for additional types of patients diseases beyond the original use for which the drug was approved for marketing to obtain additional safety data or to support marketing claims for the drug Studies in this phase are by some companies categorized as Phase IIIB studies 17 While not required in all cases it is typically expected that there be at least two successful Phase III trials demonstrating a drug s safety and efficacy to obtain approval from the appropriate regulatory agencies such as FDA US or the EMA European Union Once a drug has proved satisfactory after Phase III trials the trial results are usually combined into a large document containing a comprehensive description of the methods and results of human and animal studies manufacturing procedures formulation details and shelf life This collection of information makes up the regulatory submission that is provided for review to the appropriate regulatory authorities 18 in different countries They will review the submission and if it is acceptable give the sponsor approval to market the drug Most drugs undergoing Phase III clinical trials can be marketed under FDA norms with proper recommendations and guidelines through a New Drug Application NDA containing all manufacturing preclinical and clinical data In case of any adverse effects being reported anywhere the drugs need to be recalled immediately from the market While most pharmaceutical companies refrain from this practice it is not abnormal to see many drugs undergoing Phase III clinical trials in the market 19 Adaptive design edit The design of individual trials may be altered during a trial usually during Phase II or III to accommodate interim results for the benefit of the treatment adjust statistical analysis or to reach early termination of an unsuccessful design a process called an adaptive design 20 21 22 Examples are the 2020 World Health Organization Solidarity trial European Discovery trial and UK RECOVERY Trial of hospitalized people with severe COVID 19 infection each of which applies adaptive designs to rapidly alter trial parameters as results from the experimental therapeutic strategies emerge 23 24 25 Adaptive designs within ongoing Phase II III clinical trials on candidate therapeutics may shorten trial durations and use fewer subjects possibly expediting decisions for early termination or success and coordinating design changes for a specific trial across its international locations 22 Success rate edit For vaccines the probability of success ranges from 7 for non industry sponsored candidates to 40 for industry sponsored candidates 26 A 2019 review of average success rates of clinical trials at different phases and diseases over the years 2005 15 found a success range of 5 14 27 Separated by diseases studied cancer drug trials were on average only 3 successful whereas ophthalmology drugs and vaccines for infectious diseases were 33 successful 27 Trials using disease biomarkers especially in cancer studies were more successful than those not using biomarkers 27 A 2010 review found about 50 of drug candidates either fail during the Phase III trial or are rejected by the national regulatory agency 28 Cost of trials by phases editSee also Cost of drug development In the early 21st century a typical Phase I trial conducted at a single clinic in the United States ranged from 1 4 million for pain or anesthesia studies to 6 6 million for immunomodulation studies 29 Main expense drivers were operating and clinical monitoring costs of the Phase I site 29 The amount of money spent on Phase II or III trials depends on numerous factors with therapeutic area being studied and types of clinical procedures as key drivers 29 Phase II studies may cost as low as 7 million for cardiovascular projects and as much as 20 million for hematology trials 29 Phase III trials for dermatology may cost as low as 11 million whereas a pain or anesthesia Phase III trial may cost as much as 53 million 29 An analysis of Phase III pivotal trials leading to 59 drug approvals by the US Food and Drug Administration over 2015 16 showed that the median cost was 19 million but some trials involving thousands of subjects may cost 100 times more 30 Across all trial phases the main expenses for clinical trials were administrative staff about 20 of the total clinical procedures about 19 and clinical monitoring of the subjects about 11 29 Phase IV editA Phase IV trial is also known as a postmarketing surveillance trial or drug monitoring trial to assure long term safety and effectiveness of the drug vaccine device or diagnostic test 1 Phase IV trials involve the safety surveillance pharmacovigilance and ongoing technical support of a drug after it receives regulatory approval to be sold 7 Phase IV studies may be required by regulatory authorities or may be undertaken by the sponsoring company for competitive finding a new market for the drug or other reasons for example the drug may not have been tested for interactions with other drugs or on certain population groups such as pregnant women who are unlikely to subject themselves to trials 10 7 The safety surveillance is designed to detect any rare or long term adverse effects over a much larger patient population and longer time period than was possible during the Phase I III clinical trials 7 Harmful effects discovered by Phase IV trials may result in a drug being withdrawn from the market or restricted to certain uses examples include cerivastatin brand names Baycol and Lipobay troglitazone Rezulin and rofecoxib Vioxx citation needed Overall cost editThe entire process of developing a drug from preclinical research to marketing can take approximately 12 to 18 years and often costs well over 1 billion 31 32 References edit a b c d e f g h i The drug development process US Food and Drug Administration 4 January 2018 Retrieved 17 August 2020 New Clinical Development Success Rates 2011 2020 Report BIO Informa Pharma Intelligence and QLS Advisors Feb 2021 Exploratory IND Studies Guidance for Industry Investigators and Reviewers PDF Food and Drug Administration January 2006 Retrieved 2010 06 15 The Lancet July 2009 Phase 0 trials a platform for drug development Lancet 374 9685 176 doi 10 1016 S0140 6736 09 61309 X PMID 19616703 S2CID 30939770 Burt Tal Young Graeme Lee Wooin Kusuhara Hiroyuki Langer Oliver Rowland Malcolm Sugiyama Yuichi 2020 Phase 0 microdosing approaches time for mainstream application in drug development Nature Reviews Drug Discovery 19 11 801 818 doi 10 1038 s41573 020 0080 x ISSN 1474 1784 PMID 32901140 Fisher JA March 2015 Feeding and Bleeding The Institutional Banalization of Risk to Healthy Volunteers in Phase I Pharmaceutical Clinical Trials Science Technology amp Human Values 40 2 199 226 doi 10 1177 0162243914554838 PMC 4405793 PMID 25914430 a b c d e f g Types and phases of clinical trials American Cancer Society 18 August 2020 Retrieved 15 September 2020 NCI Dictionary National Cancer Institute 2011 02 02 Van den Eynde BJ van Baren N Baurain JF 2020 Is There a Clinical Future for IDO1 Inhibitors After the Failure of Epacadostat in Melanoma Annual Review of Cancer Biology 4 241 256 doi 10 1146 annurev cancerbio 030419 033635 a b Step 3 Clinical research US Food and Drug Administration 14 October 2016 Retrieved 1 February 2017 Shamoo AE 2008 The myth of equipoise in phase 1 clinical trials Medscape Journal of Medicine 10 11 254 PMC 2605120 PMID 19099004 registration required a b c d e f g h i DeMets D Friedman L Furberg C 2010 Fundamentals of Clinical Trials 4th ed Springer ISBN 978 1 4419 1585 6 a b Norfleet E Gad SC 2009 Phase I Clinical Trials In Gad SC ed Clinical Trials Handbook John Wiley amp Sons p 247 ISBN 978 0 470 46635 3 a b Phase 1 2 3 4 trials Revive Global Antiobiotic Research and Development Partnership 2023 Retrieved 24 October 2023 New drugs failing Phase II and III clinical trials MedCity News 2011 06 02 Clinical Development Success Rates 2006 2015 PDF bio org Retrieved 2018 02 11 Guidance for Institutional Review Boards and Clinical Investigators Food and Drug Administration 1999 03 16 Retrieved 2007 03 27 The regulatory authority in the US is the Food and Drug Administration in Canada Health Canada in the European Union the European Medicines Agency and in Japan the Ministry of Health Labour and Welfare Arcangelo VP Peterson AM 2005 Pharmacotherapeutics for Advanced Practice A Practical Approach Lippincott Williams amp Wilkins ISBN 978 0 7817 5784 3 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 Adaptive Designs for Clinical Trials of Drugs and Biologics Guidance for Industry PDF U S Food and Drug Administration 1 November 2019 Retrieved 3 April 2020 a b Pallmann P Bedding AW Choodari Oskooei B Dimairo M Flight L Hampson LV et al February 2018 Adaptive designs in clinical trials why use them and how to run and report them BMC Medicine 16 1 29 doi 10 1186 s12916 018 1017 7 PMC 5830330 PMID 29490655 Kotok A 19 March 2020 WHO beginning Covid 19 therapy trial Technology News Science and Enterprise Retrieved 7 April 2020 Launch of a European clinical trial against COVID 19 INSERM 22 March 2020 Retrieved 5 April 2020 The great strength of this trial is its adaptive nature This means that ineffective experimental treatments can very quickly be dropped and replaced by other molecules that emerge from research efforts We will therefore be able to make changes in real time in line with the most recent scientific data in order to find the best treatment for our patients RECOVERY Trial Retrieved 17 June 2020 Lo A Siah K Wong C 14 May 2020 Estimating probabilities of success of vaccine and other anti infective therapeutic development programs Harvard Data Science Review MIT Press Special Issue 1 COVID 19 doi 10 1162 99608f92 e0c150e8 Retrieved 11 August 2020 we can see that the overall probability of success PoS for industry sponsored vaccine development programs is 39 6 In contrast non industry sponsored vaccine development programs have an overall PoS of only 6 8 a b c Wong Chi Heem Siah Kien Wei Lo Andrew W 2018 01 31 Estimation of clinical trial success rates and related parameters Biostatistics 20 2 273 286 doi 10 1093 biostatistics kxx069 ISSN 1465 4644 PMC 6409418 PMID 29394327 S2CID 3277297 Arrowsmith J February 2011 Trial watch phase III and submission failures 2007 2010 Nature Reviews Drug Discovery 10 2 87 doi 10 1038 nrd3375 PMID 21283095 S2CID 39480483 a b c d e f 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 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 Holland J 2013 Fixing a broken drug development process Journal of Commercial Biotechnology 19 doi 10 5912 jcb588 Adams CP Brantner VV 2006 Estimating the cost of new drug development is it really 802 million dollars Health Affairs 25 2 420 8 doi 10 1377 hlthaff 25 2 420 PMID 16522582 Retrieved from https en wikipedia org w index php title Phases of clinical research amp oldid 1186971386 Phase I, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.