Model-Informed Drug Development: Current US Regulatory Practice and Future Considerations
Corresponding Author
Yaning Wang
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Correspondence: Yaning Wang ([email protected])Search for more papers by this authorHao Zhu
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorRajanikanth Madabushi
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorQi Liu
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorShiew-Mei Huang
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorIssam Zineh
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorCorresponding Author
Yaning Wang
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Correspondence: Yaning Wang ([email protected])Search for more papers by this authorHao Zhu
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorRajanikanth Madabushi
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorQi Liu
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorShiew-Mei Huang
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorIssam Zineh
Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
Search for more papers by this authorAbstract
Model-informed drug development (MIDD) refers to the application of a wide range of quantitative models in drug development to facilitate the decision-making process. MIDD was formally recognized in Prescription Drug User Fee Act (PDUFA) VI. There have been many regulatory applications of MIDD to address a variety of drug development and regulatory questions. These applications can be broadly classified into four categories: dose optimization, supportive evidence for efficacy, clinical trial design, and informing policy. Case studies, literature papers, and published regulatory documents are reviewed in this article to highlight some common features of these applications in each category. In addition to the further development and investment in these established domains of application, new technology, and areas, such as more mechanistic models, neural network models, and real-world data/evidence, are gaining attention, and more submissions and experiences are being accumulated to expand the application of model-based analysis to a wider scope.
Conflicts of Interest
The authors declared no competing interests for this work. As an Associate Editor for Clinical Pharmacology & Therapeutics, Shiew-Mei Huang was not involved in the review or decision process for this paper.
References
- 1Zhang, L. et al. Concepts and challenges in quantitative pharmacology and model-based drug development. AAPS J. 10, 552–559 (2008).
- 2Peck, C.C. et al. Opportunities for integration of pharmacokinetics, pharmacodynamics and toxicokinetics in rational drug development. Clin. Pharmacol. Ther. 51, 465–473 (1992).
- 3Sheiner, L.B. Learning and confirming in clinical drug development. Clin. Pharmacol. Ther. 61, 275–291 (1997).
- 4Lalonde, R.L. et al. Model-based drug development. Clin. Pharmacol. Ther. 82, 21–32 (2007).
- 5Milligan, P.A. et al. Model-based drug development: a rational approach to efficiently accelerate drug development. Clin. Pharmacol. Ther. 93, 502–514 (2013).
- 6Zineh, I. & Woodcock, J. Clinical pharmacology and the catalysis of regulatory science: opportunities for the advancement of drug development and evaluation. Clin. Pharmacol. Ther. 93, 515–525 (2013).
- 7 US Food and Drug Administration (FDA) EFPIA MID3 Workgroup. Good practices in model-informed drug discovery and development: practice, application, and documentation. CPT Pharmacometrics. Syst. Pharmacol. 5, 93–122 (2016).
- 8 US Food and Drug Administration (FDA). PDUFA reauthorization performance goals and procedures fiscal years 2018 through 2022 <https://www.fda.gov/downloads/ForIndustry/UserFees/PrescriptionDrugUserFee/UCM511438.pdf>. Accessed October 19, 2018.
- 9 US Food and Drug Administration (FDA). Policy and procedure, MAPP 4000.4 Rev 1. Good review practice: clinical pharmacology review of new molecule entity (NME) new drug applications (NDA) and original biologics license applications (BLAs) <https://www.fda.gov/downloads/AboutFDA/ReportsManualsForms/StaffPoliciesandProcedures/ucm073007.pdf>. Accessed October 19, 2018.
- 10 US Food and Drug Administration (FDA). FDA clinical pharmacology review of clevidipine butyrate <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/022156s000_ClinPharmR_P1.pdf>. Accessed September 9, 2018.
- 11 US Food and Drug Administration (FDA). Product label for paricalcitol <https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021606s016s017lbl.pdf>. Accessed October 19, 2018.
- 12 US Food and Drug Administration (FDA). FDA clinical pharmacology review of paliperidone palmitate extended-release injectable suspension <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/022264s000clinpharmr.pdf>. Accessed October 19, 2018.
- 13 US Food and Drug Administration (FDA). FDA clinical pharmacology review of mirabegron <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/202611Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 14 US Food and Drug Administration (FDA). Product development under the Animal rule: guidance for industry <https://www.fda.gov/downloads/drugs/guidances/ucm399217.pdf>. Accessed October 19, 2018.
- 15 US Food and Drug Administration (FDA). Clinical pharmacology and biopharmaceutics review for NDA 20-414 <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2003/20-414_Pyridostigmine%20Bromide_biopharmr_P1.pdf>. Accessed October 19, 2018.
- 16 US Food and Drug Administration (FDA). Clinical pharmacology and biopharmaceutics review for NDAs 020634/s061, 020635/s067, and 021721/s028 <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/020634Orig1s061,%20020635Orig1s067,%20021721Orig1s028ClinPharmR.pdf>. Accessed October 19, 2018.
- 17 US Food and Drug Administration (FDA). Clinical pharmacology and biopharmaceutics review for BLA 125349 <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/125349Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 18 US Food and Drug Administration (FDA). US Package insert for Avelox <https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/021085s060,021277s056lbl.pdf>. Accessed October 19, 2018.
- 19Ma, L. et al. Dosing regimen selection under the animal rule for pegfilgrastim to treat patients with hematopoietic syndrome of acute radiation syndrome (HS-ARS). J. Pharmacokinet. Pharmacodyn. 43, S107–S108 (2016).
- 20 US Food and Drug Administration (FDA). Clinical pharmacology and biopharmaceutics review for BLA 125509 <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/125509Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 21 US Food and Drug Administration (FDA). Clinical pharmacology and biopharmaceutics review for NDA 208627 <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/208627Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 22Ma, L. et al. Application of pharmacometrics to guide regulatory decisions for drugs to treat rare diseases <http://www.go-acop.org/assets/Legacy_ACOPs/ACOP5/Poster_Abstracts/m-074.pdf>. Accessed October 19, 2018.
- 23Hajjar, J. et al. Trends in the application of pharmacometric modeling and simulation in the development of the orphan drugs in the 21st century. Presented at the 6th American Conference on Pharmacometrics (ACoP), Arlington, VA (2015).
- 24 US Food and Drug Administration (FDA). FDA Gastrointestinal Drugs Advisory Committee briefing document <https://wayback.archive-it.org/7993/20170405221622/https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/GastrointestinalDrugsAdvisoryCommittee/UCM263457.pdf>. Accessed October 19, 2018.
- 25 US Food and Drug Administration (FDA). FDA clinical pharmacology review for pasireotide <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/200677Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 26Sinha, V. et al. Physiologically based pharmacokinetic modeling: from regulatory science to regulatory policy. Clin. Pharmacol. Ther. 95, 478–480 (2014).
- 27Rekić, D. et al. Clinical drug-drug interaction evaluations to inform drug use and enable drug access. J. Pharm. Sci. 106, 2214–2218 (2017).
- 28Li, F. et al. Pharmacometrics-based dose selection of levofloxacin as a treatment for postexposure inhalational anthrax in children. Antimicrob. Agents Chemother. 54, 375–379 (2010).
- 29 US Food and Drug Administration (FDA). FDA clinical pharmacology review of raxibacumab <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/125349Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 30 US Food and Drug Administration (FDA). Product label for levocetirizine dihydrochloride <https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/022064s036lbl.pdf>. Accessed October 19, 2018.
- 31 US Food and Drug Administration (FDA). Product label for esomeprazole sodium I.V <https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/021689s033lbl.pdf>. Accessed October 19, 2018.
- 32 US Food and Drug Administration (FDA). Product label for paliperidone extended-release tablets <https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021999s034lbl.pdf>. Accessed October 19, 2018.
- 33 US Food and Drug Administration (FDA). FDA clinical pharmacology review of canakinumab <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/125319s000_ClinPharmR.pdf>. Accessed October 19, 2018.
- 34 US Food and Drug Administration (FDA). FDA clinical pharmacology review of boceprevir <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202258Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 35 US Food and Drug Administration (FDA). FDA clinical pharmacology review of telaprevir <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/204671Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 36Ruff, C.T. et al. Evaluation of the novel factor Xa inhibitor edoxaban compared with warfarin in patients with atrial fibrillation: design and rationale for the Effective aNticoaGulation with factor xA next GEneration in Atrial Fibrillation-Thrombolysis In Myocardial Infarction study 48 (ENGAGE AF-TIMI 48). Am. Heart J. 160, 635–641 (2010).
- 37 US Food and Drug Administration (FDA). FDA clinical pharmacology review of edoxaban <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/206316Orig1Orig2s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 38 US Food and Drug Administration (FDA). FDA clinical review of edoxaban <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/206316Orig1Orig2s000MedRedt.pdf>. Accessed October 19, 2018.
- 39 US Food and Drug Administration (FDA). Product label for edoxaban <https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/206316s012lbl.pdf>. Accessed October 19, 2018.
- 40Huang, S.-M. et al. Is this the dose for you?: the role of modeling. Clin. Pharmacol. Ther. 93, 159–162 (2013).
- 41Wang, Y. et al. Impact of dose-exposure-response (D-R-E) information on regulatory approval and post authorisation commitments: FDA perspective. European Medicines Agency/European Federation of Pharmaceutical Industries and Associations workshop on the importance of dose finding and dose selection for the successful development, licensing and lifecycle management of medicinal products, December 4–5, 2014 <http://www.ema.europa.eu/docs/en_GB/document_library/Presentation/2015/01/WC500179818.pdf>. Accessed September 9, 2018.
- 42 US Food and Drug Administration (FDA). FDA clinical pharmacology review of plerixafor <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/022311s000_ClinPharmR.pdf>. Accessed October 19, 2018.
- 43 US Food and Drug Administration (FDA). Product label for plerixafor <https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/022311s018lbl.pdf>. Accessed October 19, 2018.
- 44 US Food and Drug Administration (FDA). Product label for cabazitaxel <https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/201023s021lbl.pdf>. Accessed October 19, 2018.
- 45Ascierto, P.A. et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 18, 611–622 (2017).
- 46 US Food and Drug Administration (FDA). Product label for vandetanib <https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/022405s010lbl.pdf>. Accessed October 19, 2018.
- 47Simone, P.D. et al. Everolimus with reduced tacrolimus improves renal function in de novo liver transplant recipients: a randomized controlled trial. Am. J. Transplant. 12, 3008–3020 (2012).
- 48 US Food and Drug Administration (FDA). FDA guidance for industry noninferiority clinical trials <http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM202140.pdf>. Accessed October 19, 2018.
- 49Wang, Y. et al. Justification of noninferiority margin: methodology considerations in an exposure-response analysis. Clin. Pharmacol. Ther. 97, 404–410 (2015).
- 50 US Food and Drug Administration (FDA). FDA clinical pharmacology review of canagliflozin/metformin FDC tablets <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/204353Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 51 US Food and Drug Administration (FDA). FDA clinical pharmacology review of sofosbuvir <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202258Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 52Wang, Y. et al. Leveraging prior quantitative knowledge to guide drug development decisions and regulatory science recommendations: impact of FDA pharmacometrics during 2004–2006. J. Clin. Pharmacol. 48, 146–156 (2008).
- 53 US Food and Drug Administration (FDA). Guidance for industry end-of-phase 2A meetings <https://www.fda.gov/downloads/Drugs/.../Guidances/ucm079690.pdf>. Accessed October 19, 2018.
- 54Stanski, D.R. et al. Communicating with the FDA: the “third rail” of a new model for drug development. J. Clin. Pharmacol. 48, 144–145 (2008).
- 55Woosley, R.L. They are from the government and they really are here to help you. J. Clin. Pharmacol. 48, 142–143 (2008).
- 56Hammon, J. EOP2A Guidance allows that resource constraints might block meetings. Pharmaceutical Approvals Monthly. November 1, 2009.
- 57 US Food and Drug Administration (FDA). Drug development tools: fit-for-purpose initiative <https://www.fda.gov/drugs/developmentapprovalprocess/ucm505485.htm>. Accessed October 19, 2018.
- 58 US Food and Drug Administration (FDA). Guidance for industry: E14 clinical evaluation of QT/QTc interval prolongation and proarrhythmic potential for non-antiarrhythmic drugs <https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm073153.pdf>. Accessed October 19, 2018.
- 59Garnett, C.E. et al. Concentration-QT relationships play a key role in the evaluation of proarrhythmic risk during regulatory review. J. Clin. Pharmacol. 48, 13–18 (2008).
- 60Russell, T. et al. A perspective on the use of concentration-QT modeling in drug development. J. Clin. Pharmacol. 48, 9–12 (2008).
- 61Bloomfield, D. et al. Commentary on the clinical relevance of concentration/QTc relationships for new drug candidates. J. Clin. Pharmacol. 48, 6–8 (2008).
- 62Tsong, Y. et al. Statistical issues of QT prolongation assessment based on linear concentration modeling. J. Biopharm. Stat. 18, 564–584 (2008).
- 63Wang, Y. et al. Response to letter to the editor: statistical issues of QT prolongation assessment based on linear concentration modeling by Yi Tsong et al. J. Biopharm. Stat. 20, 689–692 (2010).
- 64Tsong, Y. et al. Response to letter to the editor: statistical issues of QT prolongation assessment based on linear concentration modeling by Yi Tsong et al. J. Biopharm. Stat. 20, 693–697 (2010).
- 65Garnett, C. et al. Operational characteristics of linear concentration-QT models for assessing QTc interval in the thorough QT and phase I clinical studies. Clin. Pharmacol. Ther. 100, 170–178 (2016).
- 66Darpo, B. et al. Results from the IQ-CSRC prospective study support replacement of the thorough QT study by QT assessment in the early clinical phase. Clin. Pharmacol. Ther. 97, 326–335 (2015).
- 67 US Food and Drug Administration (FDA). E14 clinical evaluation of QT/QTc interval prolongation and proarrhythmic potential for non-antiarrhythmic drugs—questions and answers (R3) <https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM073161.pdf>. Accessed October 19, 2018.
- 68Garnett, C. et al. Scientific white paper on concentration-QTc modeling. J. Pharmacokinet. Pharmacodyn. 45, 383–397 (2018).
- 69Girgis, I.G. et al. Pharmacokinetic-pharmacodynamic assessment of topiramate dosing regimens for children with epilepsy 2 to <10 years of age. Epilepsia 51, 1954–1962 (2010).
- 70 US Food and Drug Administration (FDA). Drugs for treatment of partial onset seizures: full extrapolation of efficacy from adults to pediatric patients 4 years of age and older guidance for industry <https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM596731.pdf>. Accessed September 17, 2018.
- 71 US Federal Register <https://www.federalregister.gov/documents/2018/04/17/2018-08010/pilot-meetings-program-for-model-informed-drug-development-approaches>. Accessed October 19, 2018.
- 72 US Food and Drug Administration (FDA). FDA clinical pharmacology review for natpara <https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/125511Orig1s000ClinPharmR.pdf>. Accessed October 19, 2018.
- 73 US Food and Drug Administration (FDA). Cempra Briefing Document for solithromycin oral capsule and injection, Meeting of the Antimicrobial Drugs Advisory Committee <https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Anti-InfectiveDrugsAdvisoryCommittee/UCM528874.pdf>. Accessed October 19, 2018.
- 74Bhattaram, V.A. et al. Endpoints and analyses to discern disease-modifying drug effects in early Parkinson's disease. AAPS J. 11, 456–464 (2009).
- 75Lee, J.Y. et al. Bayesian quantitative disease-drug-trial models for Parkinson's disease to guide early drug development. AAPS J. 13, 508–518 (2011).
- 76Wang, Y. et al. Elucidation of relationship between tumor size and survival in non-small-cell lung cancer patients can aid early decision making in clinical drug development. Clin. Pharmacol. Ther. 86, 167–174 (2009).
- 77William-Faltaos, D. et al. Quantification of disease progression and dropout for Alzheimer's disease. Int. J. Clin. Pharmacol. Ther. 51, 120–131 (2013).
- 78Sun, W. et al. Development of a placebo effect model combined with a dropout model for bipolar disorder. J. Pharmacokinet. Pharmacodyn. 40, 359–368 (2013).
- 79Lambin, P. et al. Radiomics: the bridge between medical imaging and personalized medicine. Nat. Rev. Clin. Oncol. 14, 749–762 (2017).
- 80Pranav, R. et al. CheXNet: radiologist-level pneumonia detection on chest X-rays with deep learning. (2017).
- 81Sun, R. et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol. 19, 1180–1191 (2018).
- 82Sherman, R.E. et al. Real-world evidence – what is it and what can it tell us? N. Engl. J. Med. 375, 2293–2297 (2016).
- 83Sherman, R.E. et al. Accelerating development of scientific evidence for medical products within the existing US regulatory framework. Nat. Rev. Drug Discov. 16, 297–298 (2017).
- 84 US Food and Drug Administration (FDA). Use of real-world evidence to support regulatory decision-making for medical devices <https://www.fda.gov/downloads/medicaldevices/deviceregulationandguidance/guidancedocuments/ucm513027.pdf>. Accessed October 19, 2018.
- 85 US Food and Drug Administration (FDA). Approval letter <https://www.accessdata.fda.gov/cdrh_docs/pdf18/DEN180044.pdf>. Accessed October 19, 2018.