Skip to main content

Advertisement

Log in

A Framework for Safety Evaluation Throughout the Product Development Life-Cycle

  • Review Article
  • Published:
Therapeutic Innovation & Regulatory Science Aims and scope Submit manuscript

Abstract

Evaluation of the safety profile of medicines is moving from a more reactive approach, where safety experts and statisticians have been primarily focusing on the review of clinical trial data and spontaneous reports, to a more proactive endeavor with cross-functional teams strategically evolving their understanding of the safety profile. They do this by anticipating the ultimate benefit–risk profile and its related risk management implications from the start of development. The proposed approach is based on assessments of integrated program-level safety data. These data stem from multiple sources such as preclinical information; clinical and spontaneous adverse event reports; epidemiological, real-world, and registry data; as well as, potentially, data from social media. Blended qualitative and quantitative evaluations allow integration of data from diverse sources. Adding to this, a collaborative multidisciplinary view, which is focused on continuous learning and decision-making via diverse safety management teams, ensures that companies look at their growing safety database and associated risk management implications from every relevant perspective. This multifaceted and iterative approach starts early in the development of a new medicine, continues into the post-marketing setting, and wanes as the product matures and the safety profile becomes more well understood. Not only does this satisfy regulatory requirements but, crucially, it provides the healthcare system and treated patients with a better understanding of the drug’s safety profile.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Yoshioka A. Use of randomization in the Medical Research Council’s clinical trial of streptomycin in pulmonary tuberculosis in the 1940’s. BMJ. 1998;317:1220–3.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Calverley PMA, Anderson JA, Celli B, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775–89.

    CAS  PubMed  Google Scholar 

  3. Ziagen 300 mg film-coated tablets. Summary of product characteristics. https://www.ema.europa.eu/en/documents/product-information/ziagen-epar-product-information_en.pdf. Accessed 27 Aug 2019

  4. Novartis. 2018 Annual review: go big on data and digital. https://www.annualreview.novartis.com/at-a-glance/what-we-do/strategic-priorities/go-big-on-data-and-digital.html. Accessed 27 Aug 2019

  5. Chainey J. CDISC US interchange 2018: CDISC standards in the real world. https://www.cdisc.org/system/files/all/event/restricted/2018_US/1_Keynote_Chainey.pdf. Accessed 27 Aug 2019

  6. Weisberg HI. Willful ignorance: the mismeasure of uncertainty. Hoboken: Wiley; 2014.

    Google Scholar 

  7. Tukey JW. Exploratory data analysis. Boston: Addison Wesley; 1977.

    Google Scholar 

  8. Hill B. The environment and disease: association or causation? Proc R Soc Med. 1965;58:295–300.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Council for International Organizations of Medical Sciences. Guidelines for preparing core clinical-safety information on drugs (second edition): report of CIOMS working groups III and V. 1999.

  10. European Commission. Communication from the commission—detailed guidance on the collection, verification and presentation of adverse event/reaction reports arising from clinical trials on medicinal products for human use (‘CT-3’). Off J Eur Union. 2011;172:01–2011.

    Google Scholar 

  11. US Food and Drug Administration. Final rule: investigational new drug safety reporting requirements for human drug and biological products and safety reporting requirements for bioavailability and bioequivalence studies in humans. September 2010.

  12. US Food and Drug Administration. Safety reporting requirements for INDs and BA/BE studies. December 2012.

  13. European Medicines Agency. ICH guideline E2F on development safety update report. September 2011.

  14. Clinical Trial Facilitation Group CTFG. Q&A document—reference safety information. November 2017.

  15. US Food and Drug Administration. Guidance for industry: safety assessment for IND safety reporting (draft). December 2015.

  16. Rosen CJ. The rosiglitazone story—lessons from an FDA advisory committee meeting. N Engl J Med. 2007;357:844–6.

    CAS  PubMed  Google Scholar 

  17. US Food and Drug Administration. Drug safety communication: FDA requires removal of some prescribing and dispensing restrictions for rosiglitazone-containing diabetes medicines. https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-fda-requires-removal-some-prescribing-and-dispensing-restrictions. Accessed 29 July 2019

  18. Ball G, Kurek R, Hendrickson BA, et al. Global regulatory landscape for aggregate safety assessments: recent developments and future directions. Ther Innov Regul Sci.

  19. Ball G, Piller L, Silverman M. Continuous safety monitoring for randomized controlled clinical trials with blinded treatment information. Contemp Clin Trials. 2011;32:2–10.

    Google Scholar 

  20. Schnell P, Ball G. A Bayesian exposure-time method for clinical trial safety monitoring with blinded data. Ther Innov Regul Sci. 2016;50(6):833–45.

    PubMed  Google Scholar 

  21. Gould AL, Wang WB. Monitoring potential adverse event rate differences using data from blinded trials: the canary in the coal mine. Stat Med. 2017;36(1):92–104.

    PubMed  Google Scholar 

  22. Mukhopadhyay S, Waterhouse B, Hartford A. Bayesian detection of potential risk using inference on blinded safety data. Pharm Stat. 2018;17(6):823–34.

    PubMed  Google Scholar 

  23. Ball G, Lievano F. The importance of cross-disciplinary scientific engagement in the development of quantitative procedures for aggregate safety assessments. Pharm Stat. (online).

  24. Council for International Organizations of Medical Sciences. Management of safety information from clinical trials: report of CIOMS working group VI. 2005.

  25. Comfort S, Dorrell D, Meiris S, Fine J. Modified NARanjo causality scale for ICSRs (MONARCSi): a decision support tool for safety scientists. Drug Saf. 2018;41(11):1073–85.

    PubMed  PubMed Central  Google Scholar 

  26. Chuang-Stein C, Beltangady M. Reporting cumulative proportion of subjects with an adverse event based on data from multiple studies. Pharm Stat. 2011;10(1):3–7.

    PubMed  Google Scholar 

  27. Crowe B, Chuang-Stein C, Lettis S, Brueckner A. Reporting adverse drug reactions in product labels. Ther Innov Regul Sci. 2016;50(4):455–63.

    PubMed  Google Scholar 

  28. Wittes J, Crowe B, Chuang-Stein C, et al. The FDA’s final rule on expedited safety reporting: statistical considerations. Stat Biopharm Res. 2015;7(3):174–90.

    PubMed  PubMed Central  Google Scholar 

  29. Duke SP, Kleoudis C, Polinkovsky M, et al. Quantitative methods for safety monitoring of rare serious adverse events. Pharm Med. 2017;31(2):113–8.

    Google Scholar 

  30. Tukey JW. The future of data analysis. Ann Math Stat. 1962;33(1):13.

    Google Scholar 

  31. Crowe BJ, Xia HA, Berlin JA, et al. Recommendations for safety planning, data collection, evaluation and reporting during drug, biologic and vaccine development: a report of the safety planning, evaluation, and reporting team. Clin Trials. 2009;6(5):430–40.

    PubMed  Google Scholar 

  32. Ibrahim H, Saad A, Abdo A, Eldin AS. Mining association patterns of drug-interactions using post marketing FDA’s spontaneous reporting data. J Biomed Inf. 2016;60:294–308.

    Google Scholar 

  33. International Conference on Harmonization. E1: the extent of population exposure to assess clinical safety for drugs intended for long-term treatment of non-life threatening conditions. October 1994.

  34. US Food and Drug Administration. Premarketing risk assessment. March 2005.

  35. Yang F, Wittes J, Pitt B. Beware of on-treatment safety analyses. Clin Trials. 2019;16(1):63–70.

    PubMed  Google Scholar 

  36. Unkel S, Amiri M, Benda N, et al. On estimands and the analysis of adverse events in the presence of varying follow-up times within the benefit assessment of therapies. Pharm Stats. 2019;18:166–83.

    Google Scholar 

  37. Rockhold FW, Lindblad A, Siegel JP, Molenberghs G. University of Pennsylvania 11th annual conference on statistical issues in clinical trials: estimands, missing data and sensitivity analysis (morning panel session). Clin Trials. 2019;16:350–62.

    PubMed  Google Scholar 

  38. International Conference on Harmonization. Final concept paper E19: optimization of safety data collection. July 2017.

  39. US Food and Drug Administration. Determining the extent of safety data collection needed in late-stage premarket and postapproval clinical investigations. February 2016.

  40. European Medicines Agency. Guideline on good pharmacovigilance practices (GVP) Module V: risk management systems. March 2017.

  41. US Food and Drug Administration. Framework for FDA’s real-world evidence program. December 2018.

  42. Haque A, Daniel S, Maxwell T, Boerstoel M. Postmarketing surveillance studies—an industry perspective on changing global requirements and implications. Clin Ther. 2017;39(4):675–85.

    PubMed  Google Scholar 

  43. US Food and Drug Administration. Best practices for conducting and reporting pharmacoepidemiologic safety studies using electronic healthcare data. May 2013.

  44. Ball R, Robb M, Anderson SA, Dal Pan G. The FDA’s sentinel initiative: a comprehensive approach to medical product surveillance. Clin Pharmacol Ther. 2016;99(3):265–8.

    CAS  PubMed  Google Scholar 

  45. Franklin JM, Eddings W, Austin PC, et al. Comparing the performance of propensity score methods in healthcare database studies with rare outcomes. Stats Med. 2017;36(12):1946–63.

    Google Scholar 

  46. Benedetto U, Head SJ, Angelini GD, Blackstone EH. Statistical primer: propensity score matching and its alternatives. Eur J Cardio-Thoracic Surg. 2018;53(6):1112–7.

    Google Scholar 

  47. Gliklich RE, Dreyer NA, Leavy MB, editors. Registries for evaluating patient outcomes: a user’s guide. 3rd ed. Rockville: Agency for Healthcare Research and Quality; 2014.

    Google Scholar 

  48. Jonker CJ, van den Berg HM, Kwa MSG, Hoes AW, Mol PGM. Registries supporting new drug applications. Pharmacoepidemiol Drug Saf. 2017;26:1451–7.

    PubMed  PubMed Central  Google Scholar 

  49. Corrigan-Curay J, Sacks L, Woodcock J. Real-world evidence and real-world data for evaluating drug safety and effectiveness. JAMA. 2018;320(9):867–8.

    PubMed  Google Scholar 

  50. Tremmel L, Hendrickson B, Buchanan J, et al. Joint statistical meetings (JSM) 2018: drug safety monitoring in a complex world-wide regulatory environment—strategy meets methodologies. https://ww2.amstat.org/meetings/jsm/2018/onlineprogram/ActivityDetails.cfm?SessionID=215471. Accessed 27 Aug 2019

Download references

Acknowledgements

We would like to acknowledge Jacqueline Corrigan-Curay, Bruce Binkowitz, Jay Herson, and Janet Wittes for helping us to refine our framework.

Disclaimer

This article reflects the views of the individual authors and should not be construed to represent the views or policies of their companies.

Funding

No funding sources.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Greg Ball.

Ethics declarations

Conflict of interest

Dr. Schnell reports personal fees from Merck & Co, Inc, outside the submitted work. Dr. Rockhold reports grants from AstraZeneca, grants and personal fees from Janssen, personal fees from Merck Research Labs, personal fees from Merck Healthcare KGaA, personal fees from Novo Nordisk, personal fees from Rhythm, personal fees from KLSMC, personal fees from Aldeyra, personal fees from Complexa, personal fees from Sarepta, grants and personal fees from Eidos, grants from American Regent, grants from Reneuron, personal fees from Phathom, other from Athira, other from Spencer Healthcare, other from Datavant, grants from BMS, outside the submitted work.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ball, G., Reblin, T., Buchanan, J. et al. A Framework for Safety Evaluation Throughout the Product Development Life-Cycle. Ther Innov Regul Sci 54, 821–830 (2020). https://doi.org/10.1007/s43441-019-00021-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s43441-019-00021-5

Keywords

Navigation