Population pharmacokinetics, exposure-safety, and immunogenicity of atezolizumab in pediatric and young adult patients with cancer

Colby S Shemesh, Pascal Chanu, Kris Jamsen, Russ Wada, Gianluca Rossato, Francis Donaldson, Amit Garg, Helen Winter, Jane Ruppel, Xin Wang, Rene Bruno, Jin Jin, Sandhya Girish, Colby S Shemesh, Pascal Chanu, Kris Jamsen, Russ Wada, Gianluca Rossato, Francis Donaldson, Amit Garg, Helen Winter, Jane Ruppel, Xin Wang, Rene Bruno, Jin Jin, Sandhya Girish

Abstract

Background: The iMATRIX-atezolizumab study was a phase I/II, multicenter, open-label study designed to assess the safety and pharmacokinetics of atezolizumab in pediatric and young adult patients. We describe the pharmacokinetics (PK), exposure-safety, and immunogenicity of atezolizumab in pediatric and young adults with metastatic solid tumors or hematologic malignancies enrolled in this study.

Methods: Patients aged < 18 years (n = 69) received a weight-adjusted dose of atezolizumab (15 mg/kg every 3 weeks [q3w]; maximum 1200 mg); those aged ≥ 18 years (n = 18) received a flat dose (1200 mg q3w). A prior two-compartment intravenous infusion input adult population-PK (popPK) model of atezolizumab was used as a basis to model pediatric data.

Results: A total of 431 atezolizumab serum concentrations from 87 relapse-refractory pediatric and young adult patients enrolled in the iMATRIX-atezolizumab study were used for the popPK analysis. The dataset comprised predominantly patients aged < 18 years, including two infants aged < 2 years, with a wide body weight and age range. The clearance and volume of distribution estimates of atezolizumab were 0.217 L/day and 3.01 L, respectively. Atezolizumab geometric mean trough exposures were ~ 20% lower in pediatric patients versus young adults; this was not clinically meaningful as both groups achieved the target concentration (6 μg/mL). Safety was similar between pediatric and young adult patients with no exposure-safety relationship observed. Limited responses (4/87) precluded an exposure-response assessment on outcomes. A comparable rate (13% vs 11%) of atezolizumab anti-drug antibodies was seen in pediatric and young adult patients.

Conclusions: These findings demonstrate a similar exposure-safety profile of atezolizumab in pediatric and young adult patients, supportive of weight-based dosing in pediatric patients.

Trial registration: NCT02541604.

Keywords: Atezolizumab; Cancer immunotherapy; Clinical pharmacology; Exposure-safety; Immune checkpoint inhibitor; Pediatric oncology; Population pharmacokinetics.

Conflict of interest statement

CSS, PC, GR, FD, JR, XW, RB, JJ, and SG are employees and stockholders of Genentech, Inc. and F. Hoffmann-La Roche Ltd. AG and HW were employed by Genentech at the time of these analyses. KJ and RW are employees of Certara and have provided consulting services for this study.

Figures

Fig. 1
Fig. 1
(a) Prediction-corrected visual predictive check, (b) goodness of fit diagnostic plots, (c) Eta distributions, and (d) random effect correlations to covariates. Prediction-corrected visual predictive check (a): the gray solid and dashed lines represent the observed median and the 10th and 90th percentiles, respectively, while the two shades of blue represent overlap between the empirical 95% prediction intervals. Goodness of fit diagnostic plots (b): the gray solid line indicates fitted values from a nonparametric smoother. Dashed lines indicate the line of unity (top plots), or zero lines and boundary lines for conditional weighted residuals (bottom). Eta distributions (c): the blue solid line represents a density curve. Random effect correlations to covariates (d): for continuous covariates, the blue solid line represents fitted values from a nonparametric smoother. The dashed line indicates the zero line, the box-plot indicates the median and interquartile range (25th to 75th percentile), the whiskers indicate 1.5 times the interquartile range. Abbreviations: ADA anti-drug antibody, CL clearance, V1 volume of the central compartment, V2 volume of the peripheral compartment
Fig. 2
Fig. 2
Cycle 1 and steady-state (cycle 10) exposure metrics by age group: (a) Cmax, (b) Cmin, and (c) AUC. Expected interquartile range (IQR) from simulated distributions (n = 1000) based on reported geometric means and %CVs. The box-plots indicate the median and IQR (25th to 75th percentile). The whiskers indicate 1.5 times the IQR. Abbreviations: AUC area under the curve, Cmin minimum concentration, Cmax maximum concentration
Fig. 3
Fig. 3
Post-hoc exposures at cycle 1 (a) and steady-state (cycle 10) (b). Exposures across 69 patients aged < 18 years (including two infants < 2 years, 29 children 2 to < 12 years, and 38 adolescents 12 to < 18 years) and 18 young adults aged 18 to < 29 years. The dotted line indicates the therapeutic target exposure of 6 μg/mL. The height of the bar represents the number of patients within that concentration range. A cumulative distribution trend (red line) is superimposed over the frequency distribution histogram. Abbreviation: Cmin minimum concentration
Fig. 4
Fig. 4
Incidence of grade ≥ 3 AEs (a) and any-grade AESI (b). AEs and AESI are displayed by open blue circles. Solid black circles with standard error bars (y-value: binned probability of having an event from observations; x-value: median exposure value within the bin). Red line: mean model fitted curve (obtained from averaging the fitted curve for each exposure record in the data set). Dashed green lines: binning boundaries. Exposure levels are binned based on the quantiles of the log transformed exposure variable levels. Blue shaded area: based on 100 bootstrap replicates, depicting the 90% confidence band for the mean model fitted curve. Plot is based on 69 patients. Abbreviations: AE adverse event, AESI adverse event of special interest, AUC area under the curve

References

    1. Wedekind MF, Denton NL, Chen CY, Cripe TP. Pediatric cancer immunotherapy: opportunities and challenges. Paediatr Drugs. 2018;20:395–408. doi: 10.1007/s40272-018-0297-x.
    1. Tang J, Yu JX, Hubbard-Lucey VM, Neftelinov ST, Hodge JP, Lin Y. Trial watch: the clinical trial landscape for PD1/PDL1 immune checkpoint inhibitors. Nat Rev Drug Discov. 2018;17:854–855. doi: 10.1038/nrd.2018.210.
    1. Kabir TF, Chauhan A, Anthony L, Hildebrandt GC. Immune checkpoint inhibitors in pediatric solid tumors: status in 2018. Ochsner J. 2018;18:370–376. doi: 10.31486/toj.18.0055.
    1. Shebley M, Menon RM, Gibbs JP, Dave N, Kim SY, Marroum PJ. Accelerating drug development in pediatric oncology with the clinical pharmacology storehouse. J Clin Pharmacol. 2019;59:625–637. doi: 10.1002/jcph.1359.
    1. Yanni S. Disposition and interaction of biotherapeutics in pediatric populations. Curr Drug Metab. 2012;13:882–900. doi: 10.2174/138920012802138624.
    1. Van den Berg H, Tak N. Licensing and labelling of drugs in a paediatric oncology ward. Br J Clin Pharmacol. 2011;72:474–481. doi: 10.1111/j.1365-2125.2011.03977.x.
    1. Mehrotra N, Bhattaram A, Earp JC, Florian J, Krudys K, Lee JE, et al. Role of quantitative clinical pharmacology in pediatric approval and labeling. Drug Metab Dispos. 2016;44:924–933. doi: 10.1124/dmd.116.069559.
    1. Zhang Y, Wei X, Bajaj G, Barrett JS, Meibohm B, Joshi A, et al. Challenges and considerations for development of therapeutic proteins in pediatric patients. J Clin Pharmacol. 2015;55:S103–S115. doi: 10.1002/jcph.382.
    1. United States Food and Drug Administration. General clinical pharmacology considerations for pediatric studies for drugs and biological products. Guidance for industry. December 2014. . Accessed 30 July 2019.
    1. Commission of the European Communities Regulation (EC) No 1901/2006 on medicinal products for paediatric use and amending Regulation (EC) No 1768/92, Directive 2001/20/EC, Directive 2001/83/EC and Regulation (EC) No 726/2004. Off J Eur Communities L378 27/12 20061–19.
    1. Van Hasselt JG, van Ejkelenburg NK, Beijnen JH, Schellens JH, Huitema AD. Optimizing drug development of anti-cancer drugs in children using modelling and simulation. Br J Clin Pharmacol. 2013;76:30–47. doi: 10.1111/bcp.12062.
    1. Korth-Bradley JM. The path to perfect pediatric posology – drug development in pediatrics. J Clin Pharmacol. 2018;58:S48–S57. doi: 10.1002/jcph.1081.
    1. Liu L. Pharmacokinetics of monoclonal antibodies and fc-fusion proteins. Protein Cell. 2018;9:15–32. doi: 10.1007/s13238-017-0408-4.
    1. Davda JP, Dodds MG, Gibbs MA, Wisdom W, Gibbs J. A model-based meta-analysis of monoclonal antibody pharmacokinetics to guide optimal first-in-human study design. MAbs. 2014;6:1094–1102. doi: 10.4161/mabs.29095.
    1. Pasqualini C, Rialland F, Valteau-Couanet D, Michon J, Minard-Colin V. New perspectives in immunotherapy for pediatric cancers. Cancer Bull. 2018;105:S68–S79. doi: 10.1016/S0007-4551(18)30392-8.
    1. McCune JS. Rapid advances in immunotherapy to treat cancer. Clin Pharmacol Ther. 2018;103:540–544. doi: 10.1002/cpt.985.
    1. Hamilou Z, Lavaud P, Loriot Y. Atezolizumab in urothelial bladder carcinoma. Future Oncol. 2018;14:331–341. doi: 10.2217/fon-2017-0433.
    1. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–2301. doi: 10.1056/NEJMoa1716948.
    1. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108–2121. doi: 10.1056/NEJMoa1809615.
    1. Horn L, Mansfield AS, Szczęsna A, Havel L, Krzakowski M, Hochmair MJ, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 2018;379:2220–2229. doi: 10.1056/NEJMoa1809064.
    1. Pinto Navin, Park Julie R., Murphy Erin, Yearley Jennifer, McClanahan Terri, Annamalai Lakshmanan, Hawkins Douglas S., Rudzinski Erin R. Patterns of PD-1, PD-L1, and PD-L2 expression in pediatric solid tumors. Pediatric Blood & Cancer. 2017;64(11):e26613. doi: 10.1002/pbc.26613.
    1. Anderson John. Unleashing the immune response against childhood solid cancers. Pediatric Blood & Cancer. 2017;64(10):e26548. doi: 10.1002/pbc.26548.
    1. Majzner RG, Simon JS, Grosso JF, Martinez D, Pawel BR, Santi M, et al. Assessment of programmed death-ligand 1 expression and tumor-associated immune cells in pediatric cancer tissues. Cancer. 2017;123:3807–3815. doi: 10.1002/cncr.30724.
    1. Edlund H, Melin J, Parra-Guillen ZP, Kloft C. Pharmacokinetics and pharmacokinetic-pharmacodynamic relationships of monoclonal antibodies in children. Clin Pharmacokinet. 2015;54:35–80. doi: 10.1007/s40262-014-0208-4.
    1. Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. Developmental pharmacology—drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349:1157–1167. doi: 10.1056/NEJMra035092.
    1. Edginton AN, Schmitt W, Willmann S. Development and evaluation of a generic physiologically based pharmacokinetic model for children. Clin Pharmacokinet. 2006;45:1013–1034. doi: 10.2165/00003088-200645100-00005.
    1. Stroh M, Winter H, Marchand M, Claret L, Eppler S, Ruppel J, et al. Clinical pharmacokinetics and pharmacodynamics of atezolizumab in metastatic urothelial carcinoma. Clin Pharmacol Ther. 2017;102:305–312. doi: 10.1002/cpt.587.
    1. Morrissey Kari M., Marchand Mathilde, Patel Hina, Zhang Rong, Wu Benjamin, Phyllis Chan H., Mecke Almut, Girish Sandhya, Jin Jin Y., Winter Helen R., Bruno René. Alternative dosing regimens for atezolizumab: an example of model-informed drug development in the postmarketing setting. Cancer Chemotherapy and Pharmacology. 2019;84(6):1257–1267. doi: 10.1007/s00280-019-03954-8.
    1. Deng R, Bumbaca D, Pastuskovas CV, Boswell CA, West D, Cowan KJ, et al. Preclinical pharmacokinetics, pharmacodynamics, tissue distribution, and tumor penetration of anti-PD-L1 monoclonal antibody, an immune checkpoint inhibitor. MAbs. 2016;8:593–603. doi: 10.1080/19420862.2015.1136043.
    1. Geoerger B, Zwaan CM, Marshall LV, Michon J, Bordeaut F, Casanova M, et al. Atezolizumab for children and young adults with previously treated solid tumours, non-Hodgkin lymphoma, and Hodgkin lymphoma (iMATRIX): a multicentre phase 1–2 study. Lancet Oncol. 2019. Manuscript in press.
    1. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction-corrected visual predictive checks for diagnosing nonlinear mixed-effects models. AAPS J. 2011;13:143–151. doi: 10.1208/s12248-011-9255-z.
    1. Jamsen KM, Patel K, Nieforth K, Kirkpatrick CMJ. A regression approach to visual predictive checks for population pharmacometric models. CPT Pharmacometrics Syst Pharmacol. 2018;7:678–686. doi: 10.1002/psp4.12319.
    1. Centanni M, Moes DJAR, Troconiz IF, Ciccolini J, van Hasselt JGC. Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors. Clin Pharmacokinet. 2019;58:835–857. doi: 10.1007/s40262-019-00748-2.
    1. Xu Z, Davis HM, Zhou H. Rational development and utilization of antibody-based therapeutic proteins in pediatrics. Pharmacol Ther. 2013;137:225–247. doi: 10.1016/j.pharmthera.2012.10.005.
    1. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011.
    1. Malik P, Edginton A. Pediatric physiology in relation to the pharmacokinetics of monoclonal antibodies. Expert Opin Drug Metab Toxicol. 2018;14:585–599. doi: 10.1080/17425255.2018.1482278.
    1. Glassman PM, Balthasar JP. Physiologically-based pharmacokinetic modeling to predict the clinical pharmacokinetics of monoclonal antibodies. J Pharmacokinet Pharmacodyn. 2016;43:427–446. doi: 10.1007/s10928-016-9482-0.
    1. Malik PRV, Hamadeh A, Phipps C, Edginton AN. Population PBPK modelling of trastuzumab: a framework for quantifying and predicting inter-individual variability. J Pharmacokinet Pharmacodyn. 2017;44:277–290. doi: 10.1007/s10928-017-9515-3.
    1. Sheng J, Srivastava S, Sanghavi K, Lu Z, Schmidt BJ, Bello A, et al. Clinical pharmacology considerations for the development of immune checkpoint inhibitors. J Clin Pharmacol. 2017;57:S26–S42. doi: 10.1002/jcph.990.
    1. Lucchesi M, Sardi I, Puppo G, Chella A, Favre C. The dawn of “immune-revolution” in children: early experiences with checkpoint inhibitors in childhood malignancies. Cancer Chemother Pharmacol. 2017;80:1047–1053. doi: 10.1007/s00280-017-3450-2.
    1. Marrone KA, Ying W, Naidoo J. Immune-related adverse events from immune checkpoint inhibitors. Clin Pharmacol Ther. 2016;100:242–251. doi: 10.1002/cpt.394.
    1. Tecentriq (atezolizumab) package insert. South San Francisco: Genentech, Inc.; 2019.
    1. Ceppi F, Beck-Popovic M, Bourquin JP, Renella R. Opportunities and challenges in the immunological therapy of pediatric malignancy: a concise snapshot. Eur J Pediatr. 2017;176:1163–1172. doi: 10.1007/s00431-017-2982-0.
    1. Foran AE, Nadel HR, Lee AF, Savage KJ, Deyell RJ. Nivolumab in the treatment of refractory pediatric Hodgkin lymphoma. J Pediatr Hematol Oncol. 2017;39:e263–e266. doi: 10.1097/MPH.0000000000000703.
    1. Opdivo (nivolumab) package insert. Princeton, NJ: Bristol-Myers Squibb; 2018.
    1. Bavencio (avelumab) package insert. NY, NY: EMD Serono, Inc. and Pfizer Inc.; 2019.
    1. Keytruda (pembrolizumab) package insert. Whitehouse Station, NJ: Merck & Co. Inc.; 2019.
    1. Park JA, Cheung NV. Limitations and opportunities for immune checkpoint inhibitors in pediatric malignancies. Cancer Treat Rev. 2017;58:22–33. doi: 10.1016/j.ctrv.2017.05.006.

Source: PubMed

3
订阅