Citation: Sharon Lu. Clinical pharmacology to support monoclonal antibody drug development[J]. AIMS Medical Science, 2022, 9(2): 322-341. doi: 10.3934/medsci.2022014
Quantitative systems pharmacology
Biologics license application
Pharmacokinetics
Pharmacodynamics
Drug-drug interaction
First in human
No observed adverse effect level
Minimum anticipated biological effect level
Human equivalent dose
Dose severely toxic to 10% rodent
Maximum tolerated dose
Pharmacology activity
Receptor occupancy
Highest non-severely toxic dose
Target mediated drug disposition
Model-informed drug discovery and development
Integrated model-informed drug discovery and development
Thorough QT/QTc study
[1] | Mullard A (2021) FDA approves 100th monoclonal antibody product. Nat Rev Drug Discov 20: 491-495. https://doi.org/10.1038/d41573-021-00079-7 |
[2] | Frei JC, Lai JR (2016) Protein and antibody engineering by phage display. Methods Enzymol 580: 45-87. https://doi.org/10.1016/bs.mie.2016.05.005 |
[3] | Zhao J, Nussinov R, Wu WJ, et al. (2018) In silico methods in antibody design. Antibodies (Basel) 7: 22. https://doi.org/10.3390/antib7030022 |
[4] | Booth BJ, Ramakrishnan B, Narayan K, et al. (2018) Extending human IgG half-life using structure-guided design. MAbs 10: 1098-1110. https://doi.org/10.1080/19420862.2018.1490119 |
[5] | Zou P, Yu Y, Zheng N, et al. (2012) Applications of human pharmacokinetic prediction in first-in-human dose estimation. AAPS J 14: 262-281. https://doi.org/10.1208/s12248-012-9332-y |
[6] | Tam K (2013) Estimating the “first in human” dose—a revisit with particular emphasis on oncology drugs. ADMET DMPK 1: 63-75. https://doi.org/10.5599/admet.1.4.10 |
[7] | U.S. Food & Drug Administration, Guidance for industry: Estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers, 2005. Available from: https://www.fda.gov/media/72309/download |
[8] | European Medicines Agency, Guideline on strategies to identify and mitigate risks for first-in-human clinical trials with investigational medicinal products, 2007. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-strategies-identify-mitigate-risks-first-human-clinical-trials-investigational-medicinal_en.pdf |
[9] | Senderowicz AM (2010) Information needed to conduct first-in-human oncology trials in the United States: a view from a former FDA medical reviewer. Clin Cancer Res 16: 1719-1725. https://doi.org/10.1158/1078-0432.CCR-09-2766 |
[10] | Olson H, Betton G, Robinson D, et al. (2000) Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol 32: 56-67. https://doi.org/10.1006/rtph.2000.1399 |
[11] | Tibbitts J, Cavagnaro JA, Haller CA, et al. (2010) Practical approaches to dose selection for first-in-human clinical trials with novel biopharmaceuticals. Regul Toxicol Pharmacol 58: 243-251. https://doi.org/10.1016/j.yrtph.2010.06.007 |
[12] | Saber H, Gudi R, Manning M, et al. (2016) An FDA oncology analysis of immune activating products and first-in-human dose selection. Regul Toxicol Pharmacol 81: 448-456. https://doi.org/10.1016/j.yrtph.2016.10.002 |
[13] | Mittapalli RK, Liao K, Williams J, et al. (2021) Optimal dose selection in oncology biologics development: Need for a paradigm shift. J Clin Oncol 39: 3093. https://doi.org/10.1200/JCO.2021.39.15_suppl.3093 |
[14] | Paccaly AJ, Migden MR, Papadopoulos KP, et al. (2021) Fixed dose of cemiplimab in patients with advanced malignancies based on population pharmacokinetic analysis. Adv Ther 38: 2365-2378. https://doi.org/10.1007/s12325-021-01638-5 |
[15] | Brahmer JR, Drake CG, Wollner I, et al. (2010) Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 28: 3167-3175. https://doi.org/10.1200/JCO.2009.26.7609 |
[16] | Patnaik A, Kang SP, Rasco D, et al. (2015) Phase I study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res 21: 4286-4293. https://doi.org/10.1200/JCO.2009.26.7609 |
[17] | Pink Sheet, US FDA's “Project Optimus” will encourage move away from conventional dose-finding for modern cancer therapies by Rawson K, 2021. Available from: https://pink.pharmaintelligence.informa.com/PS144387/US-FDAs-Project-Optimus-Will-Encourage-Move-Away-From-Conventional-Dose-Finding-For-Modern-Cancer-Therapies |
[18] | U.S. Food & Drug Administration, Guidance for Industry: Immunogenicity testing of therapeutic protein products—developing and validating assays for anti-drug antibody detection, 2019. Available from: https://www.fda.gov/media/119788/download |
[19] | U.S. Food & Drug Administration, Guidance for Industry: Immunogenicity assessment for therapeutic protein products, 2014. Available from: https://www.fda.gov/media/85017/download |
[20] | European Medicines Agency, Guideline on Immunogenicity assessment of therapeutic proteins, 2017. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-immunogenicity-assessment-therapeutic-proteins-revision-1_en.pdf |
[21] | Lu S, Ronald RB, Clancy A, et al. (2021) An integrated analysis of dostarlimab immunogenicity. AAPS J 23: 96. https://doi.org/10.1208/s12248-021-00624-7 |
[22] | Bettsa A, Keunecke A, Steegb TJ, et al. (2018) Linear pharmacokinetic parameters for monoclonal antibodies are similar within a species and across different pharmacological targets: A comparison between human, cynomolgus monkey and hFcRn Tg32 transgenic mouse using a population-modeling approach. MAbs 10: 751-764. https://doi.org/10.1080/19420862.2018.1462429 |
[23] | Bai S, Jorga K, Xin Y, et al. (2012) A guide to rational dosing of monoclonal antibodies. Clin Pharmacokinet 51: 119-135. https://doi.org/10.2165/11596370-000000000-00000 |
[24] | Hendrikx J, Haanen J, Voest EE, et al. (2017) Fixed dosing of monoclonal antibodies in oncology. Oncologist 22: 1212-1221. https://doi.org/10.1634/theoncologist.2017-0167 |
[25] | Ryman JT, Meibohm B (2017) Pharmacokinetics of monoclonal antibodies. CPT Pharmacometrics Syst Pharmacol 6: 576-588. https://doi.org/10.1002/psp4.12224 |
[26] | Section 12.3 Pharmacokinetics of prescribing information: KEYTRUDA (pembrolizumab) injection. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125514s110lbl.pdf |
[27] | Section 12.3 Pharmacokinetics of prescribing information: OPDIVO (nivolumab) injection. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125554s112lbl.pdf |
[28] | Section 12.3 Pharmacokinetics of prescribing information: LIBTAYO (cemiplimab-rwlc) injection. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761097s007lbl.pdf |
[29] | Melhem M, Hanze E, Lu S, et al. (2022) Population pharmacokinetics and exposure-response of anti-programmed cell death protein-1 monoclonal antibody dostarlimab in advanced solid tumors. Br J Clin Pharmacol . https://doi.org/10.1111/bcp.15339 |
[30] | Section 12.3 Pharmacokinetics of prescribing information: TECENTRIQ (atezolizumab) injection. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/761034s043lbl.pdf |
[31] | Section 12.3 Pharmacokinetics of prescribing information: BAVENCIO (avelumab) injection. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761049s005lbl.pdf |
[32] | Section 12.3 Pharmacokinetics of prescribing information: IMFINZI (durvalumab) injection. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761069s028lbl.pdf |
[33] | Sheng J, Srivastava S, Sanghavi K, et al. (2017) Clinical pharmacology considerations for the development of immune checkpoint inhibitors. J Clin Pharmacol 57: S26-S42. https://doi.org/10.1002/jcph.990 |
[34] | Liu L (2018) Pharmacokinetics of monoclonal antibodies and Fc-fusion proteins. Protein Cell 9: 15-32. https://doi.org/10.1007/s13238-017-0408-4 |
[35] | Melhem M, Hanze E, Lu S, et al. (2021) Population pharmacokinetics and exposure-response analysis of the anti-programmed cell death-1 receptor monoclonal antibody dostarlimab in recurrent/advanced solid tumors with mismatch repair-deficiency. ACoP12 . |
[36] | Liu C, Yu J, Li H, et al. (2017) Association of time-varying clearance of nivolumab with disease dynamics and its implications on exposure response analysis. Clin Pharmacol Ther 101: 657-666. https://doi.org/10.1002/cpt.656 |
[37] | Turner DC, Kondic AG, Anderson KM, et al. (2018) Pembrolizumab exposure-response assessments challenged by association of cancer cachexia and catabolic clearance. Clin Cancer Res 24: 5841-5849. https://doi.org/10.1158/1078-0432.CCR-18-0415 |
[38] | U.S. Food & Drug Administration, Drug-drug interaction assessment for therapeutic proteins—Guidance for industry, 2020. Available from: https://www.fda.gov/media/140909/download |
[39] | Lee J, Zhang L, Men AY, et al. (2010) CYP-mediated therapeutic protein-drug interactions: clinical findings, proposed mechanisms and regulatory implications. Clin Pharmacokinet 49: 295-310. https://doi.org/10.2165/11319980-000000000-00000 |
[40] | Coutant DE, Hall SD (2018) Disease-drug interactions in inflammatory states via effects on CYP-mediated drug clearance. J Clin Pharmacol 58: 849-863. https://doi.org/10.1002/jcph.1093 |
[41] | Abuqayyas L, Balthasar JP (2012) Pharmacokinetic mAb-mAb interaction: Anti-VEGF mAb decreases the distribution of anti-CEA mAb into colorectal tumor xenografts. AAPS J 14: 445-455. https://doi.org/10.1208/s12248-012-9357-2 |
[42] | Pastuskovas CV, Mundo EE, Williams SP, et al. (2012) Effects of anti-VEGF on pharmacokinetics, biodistribution, and tumor penetration of trastuzumab in a preclinical breast cancer model. Mol Cancer Ther 11: 752-762. https://doi.org/10.1158/1535-7163.MCT-11-0742-T |
[43] | Kiessling P, Lledo-Garcia R, Watanabe S, et al. (2017) The FcRn inhibitor rozanolixizumab reduces human serum IgG concentration: A randomized phase 1 study. Sci Transl Med 9: 1208. https://doi.org/10.1126/scitranslmed.aan1208 |
[44] | Paglialunga S, Offman E, Ichhpurani N, et al. (2017) Update and trends on pharmacokinetic studies in patients with impaired renal function: practical insight into application of the FDA and EMA guidelines. Expert Rev Clin Pharmacol 10: 273-283. https://doi.org/10.1080/17512433.2017.1274651 |
[45] | U.S. Food & Drug Administration, Guidance for Industry: Pharmacokinetics in patients with impaired renal function—study design, data analysis, and impact on dosing, 2020. Available from: https://www.fda.gov/media/78573/download |
[46] | European Medicines Agency, Guideline on the clinical investigation of the pharmacokinetics of therapeutic proteins, 2007. Available from: https://www.ema.europa.eu/en/clinical-investigation-pharmacokinetics-therapeutic-proteins |
[47] | U.S. Food & Drug Administration, Guidance for Industry: Pharmacokinetics in patients with impaired hepatic function: Study design, data analysis, and impact on dosing and labeling. Available from: https://www.fda.gov/media/71311/download |
[48] | Ryman JT, Meibohm B (2017) Pharmacokinetics of monoclonal antibodies. CPT Pharmacometrics Syst Pharmacol 6: 576-588. https://doi.org/10.1002/psp4.12224 |
[49] | Sun Q, Seo S, Zvada S, et al. (2020) Does hepatic impairment affect the exposure of monoclonal antibodies?. Clin Pharmacol Ther 107: 1256-1262. https://doi.org/10.1002/cpt.1765 |
[50] | Yao LP (2018) Extrapolation in pediatric drug development: An evolving science.FDA Biopharm Workshop. |
[51] | Karesh A FDA Pediatric drug development: Regulatory expectations (BASIC). Available from: https://www.fda.gov/files/drugs/published/Pediatric-Drug-Development--Regulatory-Expectations---BASIC.pdf |
[52] | U.S. Food & Drug Administration, Pediatric study plans: Content of and process for submitting initial pediatric study plans and amended initial pediatric study plans—Guidance for industry, 2020. Available from: https://www.fda.gov/media/86340/download |
[53] | U.S. Food & Drug Administration, FDARA Implementation guidance for pediatric studies of molecularly targeted oncology drugs: Amendments to Sec. 505B of the FD &C Act—Guidance for Industry, 2021. Available from: https://www.fda.gov/media/133440/download |
[54] | Ollivier C, Mulugeta YL, Ruggieri L, et al. (2019) Paediatric extrapolation: A necessary paradigm shift. Br J Clin Pharmacol 85: 675-679. https://doi.org/10.1111/bcp.13809 |
[55] | Sun H, Temeck JW, Chambers W, et al. (2017) Extrapolation of efficacy in pediatric drug development and evidence-based medicine: Progress and lessons learned. Ther Innov Regul Sci 2017: 1-7. |
[56] | International Council on Harmonisation of TechnicalICH Harmonised Guideling: Addendum to ICH E11: Clinical investigation of medicinal prodicinal products in the pediatric population population E11 (R1) (2017). Available from: https://www.cfdi.org.cn/resource/attachment/ueFile/2018/01/TW9uIEphbiAwOCAwOTo1OToxOSAyMDE4MjQ3MDQ=.pdf |
[57] | European Medicines Agency, Reflection paper on the use of extrapolation in the development of medicines for paediatrics, 2018. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/adopted-reflection-paper-use-extrapolation-development-medicines-paediatrics-revision-1_en.pdf |
[58] | Robbie GJ, Zhao L, Mondick J, et al. (2012) Population pharmacokinetics of palivizumab, a humanized anti-respiratory syncytial virus monoclonal antibody, in adults and children. Antimicrob Agents Chemother 56: 4927-4936. https://doi.org/10.1128/AAC.06446-11 |
[59] | Hardiansyah D, Ng CM (2018) Effects of the FcRn developmental pharmacology on the pharmacokinetics of therapeutic monoclonal IgG antibody in pediatric subjects using minimal physiologically-based pharmacokinetic modelling. MAbs 10: 1144-1156. https://doi.org/10.1080/19420862.2018.1494479 |
[60] | Pan X, Stader F, Abduljalil K, et al. (2020) Development and application of a physiologically-based pharmacokinetic model to predict the pharmacokinetics of therapeutic proteins from full-term neonates to adolescents. AAPS J 22: 76. https://doi.org/10.1208/s12248-020-00460-1 |
[61] | U.S. Food & Drug Administration, FDA draft guidance for industry: Bioavailability and bioequivalence studies submitted in NDAs or INDs—General Considerations, 2014. Available from: https://www.fda.gov/media/88254/download |
[62] | U.S. Food & Drug Administration, Guidance for industry: Statistics approaches to establishing bioequivalence, 2001. Available from: https://www.fda.gov/media/70958/download |
[63] | Li ZY, Easton R (2018) Practical considerations in clinical strategy to support the development of injectable drug-device combination products for biologics. MAbs 10: 18-33. https://doi.org/10.1080/19420862.2017.1392424 |
[64] | European Medicines Agency, Guideline on comparability of biotechnology-derived medicinal products after a change in the manufacturing process: Non-clinical and clinical issues, 2007. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-comparability-biotechnology-derived-medicinal-products-after-change-manufacturing-process_en.pdf |
[65] | U.S. Food & Drug Administration, Human factors studies and related clinical study considerations in combination product design and development—Draft guidance for industry and FDA staff, 2016. Available from: https://www.fda.gov/media/96018/download |
[66] | Pecoraro V, De Santis E, Melegari A, et al. (2017) The impact of immunogenicity of TNFα inhibitors in autoimmune inflammatory disease. A systematic review and meta-analysis. Autoimmun Rev 16: 564-575. https://doi.org/10.1016/j.autrev.2017.04.002 |
[67] | Bahrami AA, Payandeh Z, Khalili S, et al. (2019) Immunoinformatics: in silico approaches and computational design of a multi-epitope, immunogenic protein. Int Rev Immunol 38: 307-322. https://doi.org/10.1080/08830185.2019.1657426 |
[68] | Seyhan AA (2019) Lost in translation: the valley of death across preclinical and clinical divide-identification of problems and overcoming obstacles. Transl Med Commun 4: 18. https://doi.org/10.1186/s41231-019-0050-7 |
[69] | Fleming N (2018) How artificial intelligence is changing drug discovery. Nature 557: S55-S57. https://doi.org/10.1038/d41586-018-05267-x |
[70] | Marshall S, Madabushi R, Manolis E, et al. (2019) Model-informed drug discovery and development: Current industry good practice and regulatory expectations and future perspectives. CPT Pharmacometrics Syst Pharmacol 8: 87-96. https://doi.org/10.1002/psp4.12372 |
[71] | Chelliah V, Lazarou G, Bhatnagar S, et al. (2021) Quantitative systems pharmacology approaches for immuno-oncology: Adding virtual patients to the development paradigm. Clin Pharmacol Ther 109: 605-618. https://doi.org/10.1002/cpt.1987 |
[72] | Ma H, Wang H, Sové RJ, et al. (2020) A quantitative systems pharmacology model of T cell engager applied to solid tumor. AAPS J 22: 85. https://doi.org/10.1208/s12248-020-00450-3 |
[73] | Wang H, Ma H, Sové RJ, et al. (2021) Quantitative systems pharmacology model predictions for efficacy of atezolizumab and nab-paclitaxel in triple-negative breast cancer. J Immunother Cancer 9: e002100. https://doi.org/10.1136/jitc-2020-002100 |
[74] | Gong C, Milberg O, Wang B, et al. (2017) A computational multiscale agent-based model for simulating spatio-temporal tumour immune response to PD1 and PDL1 inhibition. J R Soc Interface 14: 20170320. https://doi.org/10.1098/rsif.2017.0320 |
[75] | Lou H, Hageman MJ (2021) Machine learning attempts for predicting human subcutaneous bioavailability of monoclonal antibodies. Pharm Res 38: 451-460. https://doi.org/10.1007/s11095-021-03022-y |
[76] | Terranova N, Venkatakrishnan K, Benincosa LJ (2021) Application of machine learning in translational medicine: Current status and future opportunities. AAPS J 23: 74. https://doi.org/10.1208/s12248-021-00593-x |
[77] | Mawdsley D, Bennetts M, Dias S, et al. (2016) Model-based network meta-analysis: A framework for evidence synthesis of clinical trial data. CPT Pharmacometrics Syst Pharmacol 5: 393-401. https://doi.org/10.1002/psp4.12091 |
[78] | Mandema JM, Cox E, Alderman J (2005) Therapeutic benefit of eletriptan compared to sumatriptan for the acute relief of migraine pain—results of a model-based meta-analysis that accounts for encapsulation. Cephalalgia 25: 715-725. https://doi.org/10.1111/j.1468-2982.2004.00939.x |
[79] | Zineh I (2019) Quantitative systems pharmacology: A regulatory perspective on translation. CPT Pharmacometrics Syst Pharmacol 8: 336-339. https://doi.org/10.1002/psp4.12403 |
[80] | Liu XY, Liu C, Huang RH, et al. (2021) Long short-term memory recurrent neural network for pharmacokinetic-pharmacodynamic modeling. Int J Clin Pharmacol Ther 59: 138-146. https://doi.org/10.5414/CP203800 |
[81] | Hart T, Xie L (2016) Providing data science support for systems pharmacology and its implications to drug discovery. Expert Opin Drug Discov 11: 241-256. https://doi.org/10.1517/17460441.2016.1135126 |