Skip to main content
Log in

Innovations in Clinical Development in Rare Diseases of Children and Adults: Small Populations and/or Small Patients

  • Review Article
  • Published:
Pediatric Drugs Aims and scope Submit manuscript

Abstract

Many of the afflictions of children are rare diseases. This creates numerous drug development challenges related to small populations, including limited information about the disease state, enrollment challenges, and diminished incentives for pediatric development of novel therapies by pharmaceutical and biotechnology sponsors. We review selected innovations in clinical development that may partially mitigate some of these difficulties, starting with the concept of development efficiency for individual clinical trials, clinical programs (involving multiple trials for a single drug), and clinical portfolios of multiple drugs, and decision analysis as a tool to optimize efficiency. Development efficiency is defined as the ability to reach equally rigorous or more rigorous conclusions in less time, with fewer trial participants, or with fewer resources. We go on to discuss efficient methods for matching targeted therapies to biomarker-defined subgroups, methods for eliminating or reducing the need for natural history data to guide rare disease development, the use of basket trials to enhance efficiency by grouping multiple similar disease applications in a single clinical trial, and the use of alternative data sources including historical controls to augment or replace concurrent controls in clinical studies. Greater understanding and broader application of these methods could lead to improved therapies and/or more widespread and rapid access to novel therapies for rare diseases in both children and adults.

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Orphan Drug Act of 1983, Public Law 97-414, Stat. 2049 (1983) amended by public law 98-551 (1984) to add a numeric prevalence threshold.

  2. Utilizing innovative statistical methods and trial designs in rare diseases. In: health policy. https://healthpolicy.duke.edu/sites/default/files/2020-03/backgrounder_10_11_16.pdf. Accessed 23 Dec 2021.

  3. Information packet, rare diseases day 2022. In: Rare disease day. https://download2.rarediseaseday.org/2022/campaign_materials/infopack_2022.pdf Accessed 23 Dec 2021.

  4. Beckman RA, Clark JC, Chen C. Integrating predictive biomarkers and classifiers into oncology drug development programs. Nat Rev Drug Discovery. 2011;10:735–49.

    Article  CAS  PubMed  Google Scholar 

  5. Schwartz, J. Research in rare disease: the nature and extent of evidence needed for decision. In: 51st annual meeting of the Drug information Association, session #318 (track 17), 2015, Washington, DC.

  6. Ondra T, Jobjörnsson S, Beckman RA, Burman CF, König F, Stallard N, et al. Optimizing trial designs for targeted therapies. PLoS ONE. 2016;11:0163726.

    Article  Google Scholar 

  7. United States Food and Drug Administration, Rare diseases: common issues in drug development, guidance for industry (draft) (2015). https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM458485.pdf. Accessed 23 Dec 2021.

  8. Ghadessi M, Tang R, Zhou J, Liu R, Wang C, Toyoizumi K, et al. A roadmap to using historical controls in clinical trials—by Drug Information Association Adaptive Design Scientific Working Group (DIA-ADSWG). Orphanet J Rare Dis. 2020;15:69–87.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Beckman RA, Chen C, Posch M, Zohar S. Trial designs for rare diseases and small sample sizes in oncology. In: Halabi S, Michiels S, editors. Textbook of clinical trials in oncology: a statistical perspective. Boca Rotan: Chapman & Hall/CRC Press, Taylor and Francis Group; 2019. p. 297–316.

    Chapter  Google Scholar 

  10. Gamalo M, Bucci-Rechtweg C, Nelson RM, Vanh L, Porcella A, Thackray H, et al. Extrapolation as a default strategy in pediatric drug development. Ther Innov Regul Sci. 2022. https://doi.org/10.1007/s43441-021-00367-9.

    Article  PubMed  Google Scholar 

  11. Patel N, Ankoleka S. Dynamically optimizing budget allocation for phase 3 drug development portfolios incorporating uncertainty in the pipeline. In: Antonijevic Z, editor. Optimization of Pharmaceutical R&D Programs and Portfolios; design and investment strategy. Cham: Springer; 2004. p. 181–200.

    Google Scholar 

  12. Patel N, Ankolekar S, Antonijevic Z, Rajicic N. A mathematical model for maximizing the value of pharmaceutical portfolios incorporating budget constraints and risk. Stat Med. 2013;32:1763–77.

    Article  PubMed  Google Scholar 

  13. Ondra T, Jobjörnsson S, Beckman RA, Burman CF, König F, Stallard N, et al. Optimized adaptive enrichment designs. Stat Methods Med Res. 2019;28:2096–111.

    Article  PubMed  Google Scholar 

  14. Antonijevic Z, Wang Z. Optimal approach for addressing multiple stakeholders’ requirements in drug development. In: Antonijevic Z, Beckman RA, editors. Platform trials in drug development: umbrella trials and basket trials. Boca Raton: Chapman & Hall/CRC Press, Taylor and Francis Group; 2018. p. 153–65.

    Chapter  Google Scholar 

  15. Chen C, Beckman RA. Optimal cost-effective designs of phase II proof of concept trials and associated Go-No Go decisions. J Biopharm Stat. 2009;19:424–36.

    Article  PubMed  Google Scholar 

  16. Chen C, Beckman RA. Optimal cost-effective Go-No Go decisions in late-stage oncology drug development. Stat Biopharm Res\. 2009;1:159–69.

    Article  Google Scholar 

  17. Chen C, Beckman RA. Maximizing return on socioeconomic investment in Phase II Proof-of-Concept trials. Clin Cancer Res. 2014;20:1730–4.

    Article  PubMed  Google Scholar 

  18. Antonijevic Z. Impact of adaptive design on pharmaceutical portfolio optimization. Therap Innov Regulat Sci. 2016;50:615–9.

    Article  Google Scholar 

  19. He L, Du L, Antonijevic Z, Posch M, Korostyshevskiy V, Beckman RA. Efficient two-stage sequential arrays of proof of concept studies for pharmaceutical portfolios. Stat Methods Med Res. 2020;30:396–410.

    Article  PubMed  Google Scholar 

  20. Beckman RA, Burman CF, Chen C, Jobjörnsson S, König F, Stallard N, et al. Decision analysis from the perspective of single and multiple stakeholders. In: Antonijevic Z, Beckman RA, editors., et al., Platform trials in drug development: umbrella trials and basket trials. Boca Raton: Chapman & Hall/CRC Press, Taylor and Francis Group; 2018. p. 141–52.

    Chapter  Google Scholar 

  21. Antonijevic Z, Mills E, Häggström J, Thorlund K. Impact of platform trials on pharmaceutical frameworks. In: Antonijevic Z, Beckman RA, editors. Platform trials in drug development: umbrella trials and basket trials. Boca Raton: Chapman & Hall/CRC Press, Taylor and Francis Group; 2018. p. 73–83.

    Chapter  Google Scholar 

  22. Burman CF, Wiklund SJ. Modelling and simulation in the pharmaceutical industry—some reflections. Pharm Stat. 2011;10:508–16.

    Article  PubMed  Google Scholar 

  23. Hee SW, Hamborg T, Day S, Madan J, Miller F, Posch M, et al. Decision-theoretic designs for small trials and pilot studies: a review. Stat Methods Med Res. 2016;25:1022–38.

    Article  PubMed  Google Scholar 

  24. Stallard N, Miller F, Day S, Hee SW, Madan J, Zohar S, et al. Determination of the optimal sample size for a clinical trial accounting for the population size. Biom J. 2017;59(4):609–25.

    Article  PubMed  Google Scholar 

  25. Miller F, Zohar S, Stallard N, Madan J, Posch M, Hee SW, et al. Approaches to sample size calculation for clinical trials in rare diseases. Pharm Stat. 2018;17:214–30.

    Article  PubMed  Google Scholar 

  26. Pearce M, Hee SW, Madan J, Posch M, Day S, Miller F, et al. Value of information methods to design a clinical trial in a small population to optimise a health economic utility function. BMC Med Res Methodol. 2018;18:20.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Chen C, Deng Q, He L, Mehrotra DV, Rubin EH, Beckman RA. How many tumor indications should be initially studied in clinical development of next generation immunotherapies? Contemp Clin Trials. 2017;59:113–7.

    Article  PubMed  Google Scholar 

  28. Chaturvedi PR, Antonijevic Z, Mehta C. Practical considerations for a two-stage confirmatory adaptive clinical trial design and its implementation: ADVENT trial. In: He W, Pinheiro J, Kuznetsova OM, editors. Practical considerations for adaptive trial design and implementation. New York: Springer; 2014. p. 383–411.

    Chapter  Google Scholar 

  29. Chen C, Anderson K, Mehrotra DV, Rubin EH, Tse A. A 2-in-1 adaptive phase 2/3 design for expedited oncology drug development. Contemp Clin Trials. 2018;64:238–42.

    Article  PubMed  Google Scholar 

  30. Beckman RA, Chen C. Efficient, Adaptive Clinical Validation of Predictive Biomarkers in Cancer Therapeutic Development. In: Scatena R, editor. Advances in Cancer Biomarkers (Advances in Experimental Medicine and Biology Series, #867). Heidelberg, Germany; Springer Netherlands; 2015. pp. 81-90.

  31. Chen C, Beckman RA. Hypothesis testing in a confirmatory phase III trial with a possible subset effect. Stat Biopharm Res. 2009;1:431–40.

    Article  Google Scholar 

  32. Chen C, Li X, Li W, Beckman RA. Adaptive expansions of biomarker populations in phase 3 clinical trials. Contemp Clin Trials. 2018;71:181–5.

    Article  PubMed  Google Scholar 

  33. Ballarini NM, Burnett T, Jaki T, Jennison C, Koenig F, Posch M. Optimizing subgroup selection in two-stage adaptive enrichment and umbrella designs. Stat Med. 2021;40:2939–56.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Chen C, Li N, Shentu Y, Pang L, Beckman RA. Adaptive informational design of confirmatory phase III trials with an uncertain biomarker effect to improve the probability of success. Stat Biopharm Res. 2016;8:238–47.

    Article  Google Scholar 

  35. Woodcock J, LaVange L. Master protocols to study multiple therapies, multiple diseases, or both. N Engl J Med. 2017;377:62–70.

    Article  CAS  PubMed  Google Scholar 

  36. Barker AD, Sigman CC, Kelloff GJ, Hylton NM, Berry DA, Esserman LJ. I-Spy 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin Pharmacol Ther. 2009;86:97–100.

    Article  CAS  PubMed  Google Scholar 

  37. Chen C, Beckman RA. Informational design of confirmatory Phase III trials. Biopharm Rep. 2016;23:1–16.

    Google Scholar 

  38. Van der Ploeg AT, Clemens PR, Corzo D, Escolar DM, Florence J, Groeneveld GJ, et al. A randomized study of alglucosidase alfa in late-onset Pompe’s disease. N Engl J Med. 2010;362(15):1396–406.

    Article  PubMed  Google Scholar 

  39. Antonijevic Z, Beckman RA, editors. Platform trials in drug development: umbrella trials and basket trials. Boca Raton: Chapman & Hall; 2018.

    Google Scholar 

  40. Parsons DW, Janeway KA, Patton D, Coffey B, Williams PM, Hamilton SR, et al. Identification of targeted molecular alterations in the NCI-COG Pediatric MATCH trial. J Clin Onc. 2019;37(15_suppl):10011.

    Article  Google Scholar 

  41. Collignon O, Gartner C, Haidich AB, Hemmings RJ, Hofner B, Pétavy F, et al. Current statistical considerations and regulatory perspectives on the planning of confirmatory basket, umbrella, and platform trials. Clin Pharmacol Ther. 2020;107:1059–67.

    Article  PubMed  Google Scholar 

  42. He L, Ren Y, Chen H, Guinn D, Parashar D, Chen C, et al. Efficiency of a randomized confirmatory basket trial design constrained to control the family wise error rate by indication. Stat Methods Med Res. 2022;13:1207–23.

    Article  Google Scholar 

  43. Chen C, Zhou H, Li W, Beckman RA. How many cohorts should be considered in an exploratory master protocol? Stat Biopharm Res. 2020;13:280–5.

    Article  Google Scholar 

  44. Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, et al. Unresponsiveness of colon cancer to BRAF (V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100–4.

    Article  CAS  PubMed  Google Scholar 

  45. Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbovic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19:58–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Berry SM, Broglio KR, Groshen S, Berry DA. Bayesian hierarchical modeling of patient subpopulations: efficient designs of phase II oncology clinical trials. Clin Trials. 2013;10:720–34.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Simon RM, Geyer S, Subramanian J, Roychowdhury S. The Bayesian basket design for genomic variant-driven Phase II trials. Sem Onc. 2016;43:13–8.

    Article  Google Scholar 

  48. Beckman RA, Antonijevic Z, Kalamegham R, Chen C. Adaptive design for a confirmatory basket trial in multiple tumor types based on a putative predictive biomarker. Clin Pharmacol Ther. 2016;100:617–25.

    Article  CAS  PubMed  Google Scholar 

  49. Chen C, Li N, Yuan S, Antonijevic Z, Kalamegham R, Beckman RA. Statistical design and considerations of a Phase 3 basket trial for simultaneous investigation of multiple tumor types in one study. Stat Biopharm Res. 2016;8:248–57.

    Article  Google Scholar 

  50. Cunanan KM, Iasonos A, Shen R, Begg CB, Gönen M. An efficient basket trial design. Stat Med. 2017;36:1568–79.

    PubMed  PubMed Central  Google Scholar 

  51. Chu Y, Yuan Y. A Bayesian basket trial design using a calibrated Bayesian hierarchical model. Clin Trials. 2018;15:149–58.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Lengliné E, Peron J, Vanier A, Gueyffier F, Kouzan S, Dufour P, et al. Basket clinical trial design for targeted therapies for cancer: a French National Authority for Health Statement for health technology assessment. Lancet Oncol. 2021;22:e430–4.

    Article  PubMed  Google Scholar 

  53. European Medicines Agency (EMA), ICH topic E10 Choice of Control Group in Clinical Trials http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500002925.pdf Accessed 9 Apr 2018.

  54. Center for Drug Evaluation and Research for Biologics Evaluation and Research, Guidance for industry E10: Choice of control group and related issues. In: Clinical trials guidance for industry. 2001; http://www.fda.gov/cder/guidance/index.htm. Accessed 9 Apr 2018.

  55. Jahanshahi M, Gregg K, Davis G, Ndu A, Miller V, Vockley J, et al. The use of external controls in FDA regulatory decision making. Ther Innov Regul Sci. 2021;55:1019–35.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Pocock SJ. The combination of randomized and historical controls in clinical trials. J Chronic Dis. 1976;29:175–88.

    Article  CAS  PubMed  Google Scholar 

  57. Friede T, Röver C, Wandel S, Neuenschwander B. Meta-analysis of few small studies in orphan diseases. Res Synth Methods. 2017;8(1):79–91.

    Article  PubMed  Google Scholar 

  58. Li YR, Li J, Zhao SD, Bradfield JP, Mentch FD, Maggadottir SM, et al. Meta-analysis of shared genetic architecture across ten pediatric autoimmune diseases. Nat Med. 2015;21(9):1018–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

No external funding was used in the production of this manuscript. This work is the product of a volunteer organization, the Drug Information Association Innovative Design Scientific Working Group (DIA-IDSWG). We thank Dr Martin Posch for insightful discussions in the area of decision analysis.

Funding

No funding was used in this work.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert A. Beckman.

Ethics declarations

Conflict of interest

RAB is a consultant for AstraZeneca Pharmaceuticals, and the Chief Scientific Officer of Onco-Mind, LLC, which owns issued and pending patents on personalized strategic cancer treatment and dynamic precision medicine of cancer. Other authors are employees of for-profit firms as listed in their affiliations.

Ethics approval

Not applicable.

Consent to participate

Not applicable.

Consent for publication

Not applicable

Availability of data and materials

Not applicable.

Code availability

Not applicable.

Author contributions

RAB developed the overall manuscript concept and contributed to all sections. All other authors contributed to specific sections. All authors reviewed and approved the manuscript.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Beckman, R.A., Antonijevic, Z., Ghadessi, M. et al. Innovations in Clinical Development in Rare Diseases of Children and Adults: Small Populations and/or Small Patients. Pediatr Drugs 24, 657–669 (2022). https://doi.org/10.1007/s40272-022-00538-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40272-022-00538-7

Navigation