US Food and Drug Administration utilization of postmarketing requirements and postmarketing commitments, 2009–2018

Background/Aims The US Food and Drug Administration outlines clinical studies as postmarketing requirements and commitments to be fulfilled following approval of new drugs and biologics (“therapeutics”). Regulators have increasingly emphasized lifecycle evaluation of approved therapeutics, and postmarketing studies are intended to advance our understanding of therapeutic safety and efficacy. However, little is known about the indications that clinical studies outlined in postmarketing requirements and commitments investigate, including whether they are intended to generate evidence for approved or other clinical indications. Therefore, we characterized US Food and Drug Administration postmarketing requirements and commitments for new therapeutics approved from 2009 to 2018. Methods We conducted a cross-sectional study of all novel therapeutics, including small-molecule drugs and biologics, receiving original US Food and Drug Administration approval from 2009 to 2018, using approval letters accessed through the Drug@FDA database. Outcomes included the number and characteristics of US Food and Drug Administration postmarketing requirements and commitments for new therapeutics at original approval, including the types of studies outlined, the indications to be investigated, and the clinical evidence to be generated. Results From 2009 to 2018, the US Food and Drug Administration approved 343 new therapeutics with 1978 postmarketing requirements and commitments. Overall, 750 (37.9%) postmarketing requirements and commitments outlined clinical studies. For 71 of 343 (20.7%) therapeutics, no postmarketing requirements or commitments for clinical studies were outlined, while at least 1 was outlined for 272 (79.3%; median 2 (interquartile range: 1–4)). Among these 272 therapeutics, the number of postmarketing requirements and commitments for clinical studies per therapeutic did not change from 2009 (median: 2 (interquartile range: 1–4)) to 2018 (median: 2 (interquartile range: 1–3)). Among the 750 postmarketing requirements and commitments for clinical studies, 448 (59.7%) outlined new prospective cohort studies, registries, or clinical trials, while the remainder outlined retrospective studies, secondary analyses, or completion of ongoing studies. Although 455 (60.7%) clinical studies investigated only original approved therapeutic indications, 123 (16.4%) enrolled from an expansion of the approved disease population and 61 (8.1%) investigated diseases unrelated to approved indications. Conclusions The US Food and Drug Administration approves most new therapeutics with at least 1 postmarketing requirement or commitment for a clinical study, and outlines investigations of safety or efficacy for both approved and unapproved indications. The median number of 2 clinical studies outlined has remained relatively constant over the last decade. Given increasing emphasis by the US Food and Drug Administration on faster approval and lifecycle evaluation of therapeutics, these findings suggest that more postmarketing requirements and commitments may be necessary to address gaps in the clinical evidence available for therapeutics at approval.

[1]  K. Lieb,et al.  Impact of physicians’ participation in non-interventional post-marketing studies on their prescription habits: A retrospective 2-armed cohort study in Germany , 2020, PLoS medicine.

[2]  H. Krumholz,et al.  Assessment of Clinical Trials Supporting US Food and Drug Administration Approval of Novel Therapeutic Agents, 1995-2017 , 2020, JAMA network open.

[3]  M. Herder Pharmaceutical Drugs of Uncertain Value, Lifecycle Regulation at the US Food and Drug Administration, and Institutional Incumbency , 2019, The Milbank quarterly.

[4]  J. Wallach,et al.  Postmarketing commitments for novel drugs and biologics approved by the US Food and Drug Administration: a cross-sectional analysis , 2019, BMC medicine.

[5]  R. Pazdur,et al.  Analysis of early mortality in randomized clinical trials evaluating anti-PD-1/PD-L1 antibodies: A systematic analysis by the United States Food and Drug Administration (FDA). , 2019, Journal of Clinical Oncology.

[6]  J. Wallach,et al.  Analysis of Postapproval Clinical Trials of Therapeutics Approved by the US Food and Drug Administration Without Clinical Postmarketing Requirements or Commitments , 2019, JAMA network open.

[7]  J. Mogil,et al.  Assessment of Pregabalin Postapproval Trials and the Suggestion of Efficacy for New Indications: A Systematic Review , 2019, JAMA internal medicine.

[8]  Lisa M. Schwartz,et al.  Timeliness of Postmarket Studies for New Pharmaceuticals Approved Between 2009 and 2012: a Cross-Sectional Analysis , 2018, Journal of General Internal Medicine.

[9]  G. Baron,et al.  Factors Associated With Postmarketing Research for Approved Indications for Novel Medicines Approved by Both the FDA and EMA Between 2005 and 2010: A Multivariable Analysis , 2018, Clinical pharmacology and therapeutics.

[10]  J. Wallach,et al.  Postmarketing commitments for novel drugs and biologics approved by the US Food and Drug Administration: a cross-sectional analysis , 2018, bioRxiv.

[11]  J. Wallach,et al.  Clinical Trial Evidence Supporting FDA Approval of Drugs Granted Breakthrough Therapy Designation , 2018, JAMA.

[12]  J. Wallach,et al.  The US Food and Drug Administration’s expedited approval programs: Evidentiary standards, regulatory trade-offs, and potential improvements , 2018, Clinical trials.

[13]  J. Wallach,et al.  The US Food and Drug Administration’s expedited approval programs: Addressing premarket flexibility with enhanced postmarket evidence generation , 2018, Clinical trials.

[14]  Lisa M. Schwartz,et al.  Postmarket studies required by the US Food and Drug Administration for new drugs and biologics approved between 2009 and 2012: cross sectional analysis , 2018, British Medical Journal.

[15]  G. Baron,et al.  Post‐marketing research and its outcome for novel anticancer agents approved by both the FDA and EMA between 2005 and 2010: A cross‐sectional study , 2018, International journal of cancer.

[16]  G. Baron,et al.  Postmarketing studies for novel drugs approved by both the FDA and EMA between 2005 and 2010: a cross-sectional study , 2017, BMJ Open.

[17]  A. Jena,et al.  The Trade-off Between Speed and Safety in Drug Approvals. , 2017, JAMA oncology.

[18]  Lisa M. Schwartz,et al.  The Fate of FDA Postapproval Studies. , 2017, The New England journal of medicine.

[19]  R. Ware,et al.  Sickle cell disease , 2017, The Lancet.

[20]  J. Ioannidis,et al.  Timing and Characteristics of Cumulative Evidence Available on Novel Therapeutic Agents Receiving Food and Drug Administration Accelerated Approval , 2017, The Milbank quarterly.

[21]  H. Krumholz,et al.  Postapproval studies of drugs initially approved by the FDA on the basis of limited evidence: systematic review , 2017, British Medical Journal.

[22]  A. Breckenridge,et al.  FDA Facilitated Regulatory Pathways: Visualizing Their Characteristics, Development, and Authorization Timelines , 2017, Front. Pharmacol..

[23]  Jonathan J. Darrow,et al.  Trends in utilization of FDA expedited drug development and approval programs, 1987-2014: cohort study , 2015, BMJ : British Medical Journal.

[24]  A. Kesselheim,et al.  FDA Policy and Cardiovascular Medicine. , 2015, Circulation.

[25]  C. Furberg,et al.  Electronic Health Data for Postmarket Surveillance: A Vision Not Realized , 2015, Drug Safety.

[26]  Harlan M. Krumholz,et al.  Clinical trial evidence supporting FDA approval of novel therapeutic agents, 2005-2012. , 2014, JAMA.

[27]  G. Alexander,et al.  The Food and Drug Administration Amendments Act and postmarketing commitments. , 2013, JAMA.

[28]  Christopher Moses,et al.  Pharmacovigilance: an active surveillance system to proactively identify risks for adverse events. , 2013, Population health management.

[29]  J. DiMasi Innovating by developing new uses of already-approved drugs: trends in the marketing approval of supplemental indications. , 2013, Clinical therapeutics.

[30]  B. Psaty,et al.  A lifecycle approach to the evaluation of FDA approval methods and regulatory actions: opportunities provided by a new IOM report. , 2012, JAMA.

[31]  G. D. Dal Pan Monitoring the Safety of Medicines Used Off‐Label , 2012, Clinical pharmacology and therapeutics.

[32]  Thomas P. Gross,et al.  The US Food and Drug Administration's Sentinel Initiative: Expanding the horizons of medical product safety , 2012, Pharmacoepidemiology and drug safety.

[33]  Asher Mullard,et al.  2010 FDA drug approvals , 2011, Nature Reviews Drug Discovery.

[34]  Bethan Hughes,et al.  2009 FDA drug approvals , 2010, Nature Reviews Drug Discovery.

[35]  Bethan Hughes,et al.  2008 FDA drug approvals , 2009, Nature Reviews Drug Discovery.

[36]  I. Cockburn,et al.  The Impact of Incremental Innovation in Biopharmaceuticals , 2006, PharmacoEconomics.

[37]  P. Lane Sickle cell disease. , 1996, Pediatric clinics of North America.

[38]  Graham R Serjeant,et al.  Sickle-cell disease , 1984, The Lancet.

[39]  Guidance for Industry Providing Clinical Evidence of Effectiveness for Human Drug and Biological Products , 1998 .