Getting the Dose Right in Drug Development for Rare Diseases: Barriers and Enablers.

Item request has been placed! ×
Item request cannot be made. ×
loading   Processing Request
  • Additional Information
    • Source:
      Publisher: Wiley Country of Publication: United States NLM ID: 0372741 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1532-6535 (Electronic) Linking ISSN: 00099236 NLM ISO Abbreviation: Clin Pharmacol Ther Subsets: MEDLINE
    • Publication Information:
      Publication: 2015- : Hoboken, NJ : Wiley
      Original Publication: St. Louis : C.V. Mosby
    • Subject Terms:
    • Abstract:
      In the relentless pursuit of optimizing drug development, the intricate process of determining the ideal dosage unfolds. This involves "dose-finding" studies, crucial for providing insights into subsequent registration trials. However, the challenges intensify when tackling rare diseases. The complexity arises from poorly understood pathophysiologies, scarcity of appropriate animal models, and limited natural history understanding. The inherent heterogeneity, coupled with challenges in defining clinical end points, poses substantial challenges, hindering the utility of available data. The small affected population, low disease awareness, and restricted healthcare access compound the difficulty in conducting dose-finding studies. This white paper delves into critical dose selection aspects, focusing on key therapeutic areas, such as oncology, neurology, hepatology, metabolic rare diseases. It also explores dose selection challenges posed by pediatric rare diseases as well as novel modalities, including enzyme replacement therapies, cell and gene therapies, and oligonucleotides. Several examples emphasize the pivotal role of clinical pharmacology in navigating the complexities associated with these diseases and emerging treatment modalities.
      (© 2024 The Author(s). Clinical Pharmacology & Therapeutics © 2024 American Society for Clinical Pharmacology and Therapeutics.)
    • References:
      Abuasal, B. et al. Clinical pharmacology in drug development for rare diseases in neurology: contributions and opportunities. Clin. Pharmacol. Ther. 111, 786–798 (2022).
      Haendel, M. et al. How many rare diseases are there? Nat. Rev. Drug Discov. 19, 77–78 (2020).
      Tambuyzer, E. et al. Therapies for rare diseases: therapeutic modalities, progress and challenges ahead. Nat. Rev. Drug Discov. 19, 93–111 (2020).
      Wang, L. et al. Dose‐finding studies in drug development for rare genetic diseases. Orphanet J. Rare Dis. 17, 156 (2022).
      Han, Q. et al. Research advances in treatment methods and drug development for rare diseases. Front. Pharmacol. 13, 971541 (2022).
      Rothrock, N.E., Kaiser, K.A. & Cella, D. Developing a valid patient‐reported outcome measure. Clin. Pharmacol. Ther. 90, 737–742 (2011).
      Ghaffar Nia, N., Kaplanoglu, E. & Nasab, A. Evaluation of artificial intelligence techniques in disease diagnosis and prediction. Discov. Artif. Intell. 3, 5 (2023).
      Bhathena, A. & Spear, B.B. Pharmacogenetics: improving drug and dose selection. Curr. Opin. Pharmacol. 8, 639–646 (2008).
      Oates, J.T. & Lopez, D. Pharmacogenetics: an important part of drug development with a focus on its application. Int J Biomed Investig 1, 1–16 (2018).
      US Food and Drug Administration. Rare Diseases: Considerations for the Development of Drugs and Biological Products (2023) Accessed 06/19/2024.
      Bouwman, L., Sepodes, B., Leufkens, H. & Torre, C. Trends in orphan medicinal products approvals in the European Union between 2010–2022. Orphanet J. Rare Dis. 19, 91 (2024).
      Madabushi, R., Benjamin, J., Zhu, H. & Zineh, I. The US Food and Drug Administration's model‐informed drug development meeting program: from pilot to pathway. Clinical Pharmacology & Therapeutics 116, 278–281 (2024).
      Li, R.J. et al. Model‐informed approach supporting drug development and regulatory evaluation for rare diseases. J. Clin. Pharmacol. 62(Suppl 2), S27–s37 (2022).
      DeSantis, C.E., Kramer, J.L. & Jemal, A. The burden of rare cancers in the United States. CA Cancer J. Clin. 67, 261–272 (2017).
      Murphy, R., Halford, S. & Symeonides, S.N. Project Optimus, an FDA initiative: considerations for cancer drug development internationally, from an academic perspective. Front. Oncol. 13, 1144056 (2023).
      US Food and Drug Administration. Optimizing the dosage of human prescription drugs and biological products for the treatment of oncologic diseases. Guidance for Industry (2023). Accessed 01/31/2024.
      Harmonisation, T.I.C.f. Guideline for industry: dose‐response information to support drug registration (1994).
      Mitra, A., Lee, J.B., Steinbach, D., Hazra, A. & Krishna, R. Rare oncology therapeutics: review of clinical pharmacology package of drug approvals (2019‐2023) by US FDA, best practices and recommendations. J. Pharmacokinet. Pharmacodyn. 50, 475–493 (2023).
      US Food and Drug Administration. NDA 215358 Multi‐Disciplinary Review and Evaluation (2021). Accessed 01/31/2024.
      Schoepfer, J. et al. Discovery of Asciminib (ABL001), an allosteric inhibitor of the tyrosine kinase activity of BCR‐ABL1. J. Med. Chem. 61, 8120–8135 (2018).
      Xia, H. et al. Use of patient‐reported outcomes (PRO) data to complement exposure‐response analysis in early clinical cancer drug development. J Patient Rep Outcomes 7, 116 (2023).
      Zhu, R. et al. Exposure‐response‐based product profile‐driven clinical utility index for Ipatasertib dose selection in prostate cancer. CPT Pharmacometrics Syst. Pharmacol. 8, 240–248 (2019).
      Archie, S.R., Al Shoyaib, A. & Cucullo, L. Blood‐brain barrier dysfunction in CNS disorders and putative therapeutic targets: an overview. Pharmaceutics 13, 1779 (2021).
      Miller, T. et al. Phase 1‐2 trial of antisense oligonucleotide Tofersen for SOD1 ALS. N. Engl. J. Med. 383, 109–119 (2020).
      Monine, M., Norris, D., Wang, Y. & Nestorov, I. A physiologically‐based pharmacokinetic model to describe antisense oligonucleotide distribution after intrathecal administration. J. Pharmacokinet. Pharmacodyn. 48, 639–654 (2021).
      Kriegermeier, A. & Green, R. Pediatric cholestatic liver disease: review of bile acid metabolism and discussion of current and emerging therapies. Front. Med. 7, 1–15 (2020).
      Poupon, R.E., Poupon, R., Balkau, B. & the UDCA‐PBC Study Group. Ursodiol for the long‐term treatment of primary biliary cirrhosis. N. Engl. J. Med. 330, 1342–1347 (1994).
      Administration, U.F.a.D. Obeticholic Acid Clinical Pharmacology and Biopharmaceutics Review (2016).
      Center for Drug Evaluation and Research. Application Number: 207999orig1s000. Summary Review Accessed 01/21/2024.
      Ravenstijn, P., Chetty, M., Manchandani, P., Elmeliegy, M., Qosa, H. & Younis, I. Design and conduct considerations for studies in patients with hepatic impairment. Clin. Transl. Sci. 16, 50–61 (2023).
      Edwards, J.E. et al. Modeling and experimental studies of Obeticholic acid exposure and the impact of cirrhosis stage. Clin. Transl. Sci. 9, 328–336 (2016).
      FDA. Drug Safety Communication (2021). Accessed 1/21/2024.
      Eaton, J.E., Vuppalanchi, R., Reddy, R., Sathapathy, S., Ali, B. & Kamath, P.S. Liver injury in patients with cholestatic liver disease treated with Obeticholic acid. Hepatology 71, 1511–1514 (2020).
      Jeyaraj, R., Maher, E.R. & Kelly, D. Paediatric research sets new standards for therapy in paediatric and adult cholestasis. Lancet Child Adolesc Health 8, 75–84 (2024).
      Baumann, U. et al. Effects of odevixibat on pruritus and bile acids in children with cholestatic liver disease: phase 2 study. Clin. Res. Hepatol. Gastroenterol. 45, 101751 (2021).
      Mayo, M.J. et al. A randomized, controlled, phase 2 study of Maralixibat in the treatment of itching associated with primary biliary cholangitis. Hepatol Commun 3, 365–381 (2019).
      Gonzales, E. et al. Efficacy and safety of maralixibat treatment in patients with Alagille syndrome and cholestatic pruritus (ICONIC): a randomised phase 2 study. Lancet 398, 1581–1592 (2021).
      Gambello, M.J. & Li, H. Current strategies for the treatment of inborn errors of metabolism. J. Genet. Genomics 45, 61–70 (2018).
      Yue, W.W., Mackinnon, S. & Bezerra, G.A. Substrate reduction therapy for inborn errors of metabolism. Emerging topics in life sciences 3, 63–73 (2019).
      Ginocchio, V.M. & Brunetti‐Pierri, N. Progress toward improved therapies for inborn errors of metabolism. Hum. Mol. Genet. 25, R27–R35 (2016).
      Hon, Y.Y. et al. Dose selection for biological enzyme replacement therapy indicated for inborn errors of metabolism. Clin. Transl. Sci. 16, 2438–2457 (2023).
      US Food and Drug Administration. BLA 125513 clinical pharmacology and biopharmaceutics review (2015). Accessed 01/21/2024.
      Kulkarni, J.A. et al. The current landscape of nucleic acid therapeutics. Nat. Nanotechnol. 16, 630–643 (2021).
      US Food and Drug Administration. Clinical Pharmacology Considerations for the Development of Oligonucleotide Therapeutics Draft Guidance (2022). Accessed 01/21/2024.
      Yin, W. & Rogge, M. Targeting RNA: a transformative therapeutic strategy. Clin. Transl. Sci. 12, 98–112 (2019).
      Jing, X., Arya, V., Reynolds, K.S. & Rogers, H. Clinical pharmacology of RNA interference‐based therapeutics: a summary based on Food and Drug Administration‐approved small interfering RNAs. Drug Metab. Dispos. 51, 193–198 (2023).
      US Food and Drug Administration. NDA 211172 Clinical Pharmacology and Biopharmaceutics Review (2017). Accessed 01/21/2024.
      US Food and Drug Administration. NDA 214012 Clinical Pharmacology and Biopharmaceutics Review (2020). Accessed 01/21/2024.
      US Food and Drug Administration. NDA 209531 Clinical Pharmacology and Biopharmaceutics Review (2016). Accessed 01/21/2024.
      US Food and Drug Administration. NDA 212194 Clinical Pharmacology and Biopharmaceutics Review (2019). Accessed 01/21/2024.
      US Food and Drug Administration. NDA 214012 Clinical Pharmacology and Biopharmaceutics Review (2021). Accessed 01/21/2024.
      Kim, J. et al. Patient‐customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 381, 1644–1652 (2019).
      Synofzik, M. et al. Preparing n‐of‐1 antisense oligonucleotide treatments for rare neurological diseases in Europe: genetic, regulatory, and ethical perspectives. Nucleic Acid Ther 32, 83–94 (2022).
      US Food and Drug Administration. IND Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life‐Threatening Diseases: Clinical Recommendations (2021). Accessed 01/31/2024.
      US Food and Drug Administration. Nonclinical Testing of Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life‐Threatening Diseases Guidance for Sponsor‐Investigators (2021). Accessed 01/31/2024.
      Chen, N., Sun, K., Chemuturi, N.V., Cho, H. & Xia, C.Q. The perspective of DMPK on recombinant adeno‐associated virus‐based gene therapy: past learning, current support, and future contribution. AAPS J. 24, 31 (2022).
      Sun, K. & Liao, M.Z. Clinical pharmacology considerations on recombinant adeno‐associated virus‐based gene therapy. J. Clin. Pharmacol. 62(Suppl 2), S79–s94 (2022).
      Zou, P. First‐in‐patient dose prediction for adeno‐associated virus‐mediated hemophilia gene therapy using allometric scaling. Mol. Pharm. 20, 758–766 (2023).
      US Food and Drug Administration. Considerations for the Design of Early‐Phase Clinical Trials of Cellular and Gene Therapy Products (2015). Accessed 02/09/2024.
      US Food and Drug Administration. Cellular & Gene Therapy Guidances Accessed 02/09/2024.
      European Medicines Agency. Multidisciplinary gene therapy guidelines Accessed 02/09/2024.
      Kast, J. et al. Recent advances and clinical pharmacology aspects of chimeric antigen receptor (CAR) T‐cellular therapy development. Clin. Transl. Sci. 15, 2057–2074 (2022).
      Mueller, K.T. et al. Clinical pharmacology of Tisagenlecleucel in B‐cell acute lymphoblastic leukemia. Clin. Cancer Res. 24, 6175–6184 (2018).
      Raje, N. et al. Anti‐BCMA CAR T‐cell therapy bb2121 in relapsed or refractory multiple myeloma. N. Engl. J. Med. 380, 1726–1737 (2019).
      Munshi, N.C. et al. Idecabtagene Vicleucel in relapsed and refractory multiple myeloma. N. Engl. J. Med. 384, 705–716 (2021).
      Singh, A.P. et al. Development of a quantitative relationship between CAR‐affinity, antigen abundance, tumor cell depletion and CAR‐T cell expansion using a multiscale systems PK‐PD model. MAbs 12, 1688616 (2020).
      Batshaw, M.L., Groft, S.C. & Krischer, J.P. Research into rare diseases of childhood. JAMA 311, 1729–1730 (2014).
      US Food and Drug Administration. Draft Guidance for Industry: Ethical Considerations for Clinical Investigations of Medical Products Involving Children (2022). Accessed 01/21/2024.
      ICH guideline. E11A on pediatric extrapolation (2022). Accessed 01/21/2024.
      Ahmed, M.A., Burnham, J., Dwivedi, G. & AbuAsal, B. Achieving big with small: quantitative clinical pharmacology tools for drug development in pediatric rare diseases. J. Pharmacokinet. Pharmacodyn. 50, 429–444 (2023).
      Durairaj, C. & Bhattacharya, I. Challenges, approaches and enablers: effectively triangulating towards dose selection in pediatric rare diseases. J. Pharmacokinet. Pharmacodyn. 50, 445–459 (2023).
      US Food and Drug Administration. BLA 761261 Integrated Review (2021). Accessed 01/21/2024.
      Bhattacharya, I., Manukyan, Z., Chan, P., Heatherington, A. & Harnisch, L. Application of quantitative pharmacology approaches in bridging pharmacokinetics and pharmacodynamics of Domagrozumab from adult healthy subjects to pediatric patients with Duchenne muscular disease. J. Clin. Pharmacol. 58, 314–326 (2018).
      US Food and Drug Admnistration. NDA 205494 Review (2015). Accessed 01/21/2024.
      US Food and Drug Administration. NDA 205494 Division Director Summary Review (2014). Accessed 01/21/2024.
      Lukina, E. et al. A phase 2 study of eliglustat tartrate (Genz‐112638), an oral substrate reduction therapy for Gaucher disease type 1. Blood 116, 893–899 (2010).
      Peterschmitt, M.J. et al. Safety, tolerability, and pharmacokinetics of eliglustat tartrate (Genz‐112638) after single doses, multiple doses, and food in healthy volunteers. J. Clin. Pharmacol. 51, 695–705 (2011).
      Table of Pharmacogenomic Biomarkers in Drug Labeling. Accessed 01/21/2024.
      Rare Disease Cures Accelerator. Accessed 01/21/2024.
      Clinical Pharmacogenomics. Premarket Evaluation in Early‐Phase Clinical Studies and Recommendations for Labeling (2013). Accessed 01/21/2024.
      Babrak, L.M. et al. Traditional and digital biomarkers: two worlds apart? Digit Biomark 3, 92–102 (2019).
      Stephenson, D. et al. Precompetitive consensus building to facilitate the use of digital health technologies to support Parkinson disease drug development through regulatory science. Digit Biomark 4, 28–49 (2020).
      EMA. Qualification Opinion for Stride velocity 95th centile as primary endpoint in studies in ambulatory Duchenne Muscular Dystrophy studies. EMADOC‐1700519818‐112713 (2023). Accessed 01/21/2024.
      Servais, L. et al. First regulatory qualification of a digital primary endpoint to measure treatment efficacy in DMD. Nat. Med. 29, 2391–2392 (2023).
    • Publication Date:
      Date Created: 20240816 Date Completed: 20241115 Latest Revision: 20241118
    • Publication Date:
      20241119
    • Accession Number:
      10.1002/cpt.3407
    • Accession Number:
      39148459