Pan-cancer population pharmacokinetics and exposure-safety and -efficacy analyses of atezolizumab in patients with high tumor mutational burden

Colby S Shemesh, Phyllis Chan, Fatema A Legrand, David S Shames, Meghna Das Thakur, Jane Shi, Lorna Bailey, Shweta Vadhavkar, Xian He, Wei Zhang, René Bruno, Colby S Shemesh, Phyllis Chan, Fatema A Legrand, David S Shames, Meghna Das Thakur, Jane Shi, Lorna Bailey, Shweta Vadhavkar, Xian He, Wei Zhang, René Bruno

Abstract

We retrospectively investigated the pharmacokinetics and exposure-efficacy/safety relationships of single-agent atezolizumab based on tissue tumor mutational burden (tTMB) status (high vs low [≥16 vs <16 mutations/megabase]) in a pan-tumor population from seven clinical trials. Data sources included the OAK, POPLAR, BIRCH, FIR, IMvigor210, IMvigor211, and PCD4989g studies; 986 of 2894 treated patients (34%) had TMB data. Exposure metrics were obtained using a prior two-compartment intravenous-infusion population-pharmacokinetics model, merged with prognostic, biomarker, efficacy, and safety variables. Baseline demographic/clinical characteristics and prognostic factors were well balanced between patients with high (n = 175) and low (n = 811) tTMB. Exposure was similar in the high- and low-tTMB subgroups, with no difference seen in the evaluable vs total treated populations. The objective response rate (ORR) was 29.7% vs 13.4%, complete response rate was 6.9% vs 3.2%, and median duration of response (95% CI) was 29.0 (18.6-NE) months vs 15.9 (12.5-20.5) months for patients with high-tTMB vs low-tTMB tumors, respectively. A flat exposure-efficacy relationship was seen for ORR in patients with high-tTMB based on the cycle 1 minimum atezolizumab concentration and area under the serum concentration time curve (AUC). A nonsignificant exposure-safety profile was seen for grade 3/4 adverse events and adverse events of special interest based on the AUC of atezolizumab in the high-tTMB population. tTMB is an additional predictive biological factor affecting response to atezolizumab, and quantitative investigations of atezolizumab exposure and relationships of exposure with safety and efficacy support the use of a 1200-mg, every 3-week regimen in a tumor-agnostic high-tTMB population.

Trial registration: ClinicalTrials.gov NCT02008227 NCT01903993 NCT02031458 NCT01846416 NCT02302807 NCT02108652 NCT01375842.

Keywords: atezolizumab; biomarkers; clinical pharmacology; mutation; pharmacokinetics; tumor.

Conflict of interest statement

All authors disclose medical writing support funded by F. Hoffmann‐La Roche Ltd. All authors are employees of Genentech, Inc (part of the Roche Group), F. Hoffmann‐La Roche Ltd., or Roche Products Ltd. and are stockholders of F. Hoffmann‐La Roche Ltd.

© 2020 The Authors. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd, British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.

Figures

FIGURE 1
FIGURE 1
Flowchart of the analysis populations. AESI, adverse event of special interest; ER, exposure‐response; mUC, metastatic urothelial carcinoma; NSCLC, non‐small cell lung cancer; popPK, population pharmacokinetics; SLD, sum of longest diameters; TMB, tumor mutational burden
FIGURE 2
FIGURE 2
Atezolizumab exposure distribution by tTMB status. Post hoc analysis of exposures across 880 patients treated with atezolizumab 1200 mg are shown, including 709 patients with tTMB

FIGURE 3

Proportion of tTMB‐high patients who…

FIGURE 3

Proportion of tTMB‐high patients who were responders (CR + PR) to atezolizumab by…

FIGURE 3
Proportion of tTMB‐high patients who were responders (CR + PR) to atezolizumab by (A) cycle 1 AUC and (B) cycle 1 Cmin and the proportion of tTMB‐high patients with grade 3/4 AEs by AUC (C) and (D) any‐grade AESI by AUC. AUC and Cmin values for each response event (yes, 1.00; no, 0) are represented by open grey circles. Solid black circles with standard error bars: proportion of response from binned observations by quartiles of the log‐transformed exposure (y value); median exposure value within the bin (x value). Black line: model‐fitted curve of the probability of response across atezolizumab exposure. Dashed lines: binning boundaries. Shaded area: 95% confidence band for the logistic regression curve. Observed data points are based on 171 and 167 patients for efficacy and safety, respectively. AESI, adverse event of special interest; AUC, area under the curve; Cmin, minimum concentration; CR, complete response; PR, partial response; tTMB, tissue tumor mutational burden

FIGURE 4

Change in SLD from baseline…

FIGURE 4

Change in SLD from baseline by TMB status and atezolizumab exposure. (A) Tumor…

FIGURE 4
Change in SLD from baseline by TMB status and atezolizumab exposure. (A) Tumor size change across 713 tTMB efficacy‐evaluable patients with available tumor scan data (gray). Mean changes in tumor size from baseline are represented by the solid black line for the tTMB‐high subgroup (≥16 mut/Mb; n = 145) and by the dotted black line for the tTMB‐low subgroup ( = 568). (B) tTMB efficacy and cycle 1 atezolizumab Cmin PK‐evaluable patients (n = 703). Mean tumor size changes from baseline as a function of cycle 1 atezolizumab Cmin exposure quartile are shown in red (n = 176), yellow (n = 176), blue (n = 176), and green (n = 175). Cmin, minimum concentration; PK, pharmacokinetics; SLD, sum of longest diameter; tTMB, tissue tumor mutational burden
FIGURE 3
FIGURE 3
Proportion of tTMB‐high patients who were responders (CR + PR) to atezolizumab by (A) cycle 1 AUC and (B) cycle 1 Cmin and the proportion of tTMB‐high patients with grade 3/4 AEs by AUC (C) and (D) any‐grade AESI by AUC. AUC and Cmin values for each response event (yes, 1.00; no, 0) are represented by open grey circles. Solid black circles with standard error bars: proportion of response from binned observations by quartiles of the log‐transformed exposure (y value); median exposure value within the bin (x value). Black line: model‐fitted curve of the probability of response across atezolizumab exposure. Dashed lines: binning boundaries. Shaded area: 95% confidence band for the logistic regression curve. Observed data points are based on 171 and 167 patients for efficacy and safety, respectively. AESI, adverse event of special interest; AUC, area under the curve; Cmin, minimum concentration; CR, complete response; PR, partial response; tTMB, tissue tumor mutational burden
FIGURE 4
FIGURE 4
Change in SLD from baseline by TMB status and atezolizumab exposure. (A) Tumor size change across 713 tTMB efficacy‐evaluable patients with available tumor scan data (gray). Mean changes in tumor size from baseline are represented by the solid black line for the tTMB‐high subgroup (≥16 mut/Mb; n = 145) and by the dotted black line for the tTMB‐low subgroup ( = 568). (B) tTMB efficacy and cycle 1 atezolizumab Cmin PK‐evaluable patients (n = 703). Mean tumor size changes from baseline as a function of cycle 1 atezolizumab Cmin exposure quartile are shown in red (n = 176), yellow (n = 176), blue (n = 176), and green (n = 175). Cmin, minimum concentration; PK, pharmacokinetics; SLD, sum of longest diameter; tTMB, tissue tumor mutational burden

References

    1. McCune JS. Rapid advances in immunotherapy to treat cancer. Clin Pharmacol Ther. 2018;103(4):540‐544.
    1. Kruger S, Ilmer M, Kobold S, et al. Advances in cancer immunotherapy 2019 ‐ latest trends. J Exp Clin Cancer Res. 2019;38(1):268.
    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(12):854‐855.
    1. Ingles Garces AH, Au L, Mason R, Thomas J, Larkin J. Building on the anti‐PD1/PD‐L1 backbone: combination immunotherapy for cancer. Expert Opin Investig Drugs. 2019;28(8):695‐708.
    1. Shindo Y, Hazama S, Tsunedomi R, Suzuki N, Nagano H. Novel biomarkers for personalized cancer immunotherapy. Cancers (Basel). 2019;11(9):1223.
    1. Zhang M, Yang J, Hua W, Li Z, Xu Z, Qian Q. Monitoring checkpoint inhibitors: predictive biomarkers in immunotherapy. Front Med. 2019;13(1):32‐44.
    1. Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA Approval summary: pembrolizumab for the treatment of microsatellite instability‐high solid tumors. Clin Cancer Res. 2019;25(13):3753‐3758.
    1. KEYTRUDA (pembrolizumab) [package insert]. Whitehouse Station, NJ: Merck Sharp & Dohme Corp; 2020.
    1. Alexandrov LB, Nik‐Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415‐421.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69‐74.
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD‐1 inhibition. N Engl J Med. 2017;377(25):2500‐2501.
    1. Marabelle A, Fakih M, Lopez J, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open‐label, phase 2 KEYNOTE‐158 study. Lancet Oncol. 2020;21(10):1353‐1365.
    1. Chan TA, Yarchoan M, Jaffee E, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30(1):44‐56.
    1. Goodman AM, Kato S, Bazhenova L, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16(11):2598‐2608.
    1. Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51(2):202‐206.
    1. Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti‐PD‐L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563‐567.
    1. Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as first‐line treatment in cisplatin‐ineligible patients with locally advanced and metastatic urothelial carcinoma: a single‐arm, multicentre, phase 2 trial. Lancet. 2017;389(10064):67‐76.
    1. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first‐line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378(24):2288‐2301.
    1. Schmid P, Adams S, Rugo HS, et al. Atezolizumab and nab‐paclitaxel in advanced triple‐negative breast cancer. N Engl J Med. 2018;379(22):2108‐2121.
    1. Horn L, Mansfield AS, Szczesna A, et al. First‐line atezolizumab plus chemotherapy in extensive‐stage small‐cell lung cancer. N Engl J Med. 2018;379(23):2220‐2229.
    1. TECENTRIQ (atezolizumab) [package insert]. South San Francisco, CA: Genentech, Inc; 2019.
    1. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non‐small‐cell lung cancer (OAK): a phase 3, open‐label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255‐265.
    1. Powles T, Duran I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum‐treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open‐label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748‐757.
    1. Legrand FA, Gandara DR, Mariathasan S, et al. Association of high tissue TMB and atezolizumab efficacy across multiple tumor types. J Clin Oncol. 2018;36(15_suppl):12000.
    1. Sheng J, Srivastava S, Sanghavi K, et al. Clinical pharmacology considerations for the development of immune checkpoint inhibitors. J Clin Pharmacol. 2017;57(suppl 10):S26‐S42.
    1. Centanni M, Moes D, Troconiz IF, Ciccolini J, van Hasselt JGC. Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors. Clin Pharmacokinet. 2019;58(7):835‐857.
    1. Johnson DB, Frampton GM, Rioth MJ, et al. Targeted next generation sequencing identifies markers of response to PD‐1 blockade. Cancer Immunol Res. 2016;4(11):959‐967.
    1. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34.
    1. Lieber DS, Kennedy MR, Johnson DB, et al. Validation and clinical feasibility of a comprehensive genomic profiling assay to identify likely immunotherapy responders through tumor mutational burden (TMB). Can Res. 2017;77:2987.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228‐247.
    1. Gandara DR, Paul SM, Kowanetz M, et al. Blood‐based tumor mutational burden as a predictor of clinical benefit in non‐small‐cell lung cancer patients treated with atezolizumab. Nat Med. 2018;24(9):1441‐1448.
    1. Stroh M, Winter H, Marchand M, et al. Clinical pharmacokinetics and pharmacodynamics of atezolizumab in metastatic urothelial carcinoma. Clin Pharmacol Ther. 2017;102(2):305‐312.
    1. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction‐corrected visual predictive checks for diagnosing nonlinear mixed‐effects models. AAPS J. 2011;13(2):143‐151.
    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(10):678‐686.
    1. Harding SD, Sharman JL, Faccenda E, et al. The IUPHARM/BPS Guide to PHARMACOLOGY in 2019: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucleic Acid Res. 2018;46:D1091‐D1106. 10.1093/nar/gkx1121
    1. Alexander SPH, Kelly E, Mathie A, et al. CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2019/20: Introduction and Other Protein Targets. Br J Pharmacol, Suppl. 2019;1:S1‐S20. 10.1111/bph.14747
    1. Nguyen TH, Mouksassi MS, Holford N, et al. Model evaluation of continuous data pharmacometric models: metrics and graphics. CPT Pharmacometrics Syst Pharmacol. 2017;6(2):87‐109.
    1. Deng R, Bumbaca D, Pastuskovas CV, et al. Preclinical pharmacokinetics, pharmacodynamics, tissue distribution, and tumor penetration of anti‐PD‐L1 monoclonal antibody, an immune checkpoint inhibitor. MAbs. 2016;8(3):593‐603.
    1. Wu Y, Xu J, Du C, et al. The predictive value of tumor mutation burden on efficacy of immune checkpoint inhibitors in cancers: a systematic review and meta‐analysis. Front Oncol. 2019;9:1161.
    1. Baretti M, Azad NS. Precision cancer trials with immunomodulatory agents: personalizing histology agnostic approaches. Cancer J. 2019;25(4):287‐295.
    1. Addeo A, Banna GL, Weiss GJ. Tumor mutation burden—from hopes to doubts. JAMA Oncol. 2019;5(7):934‐935.
    1. Stroh M, Carlile DJ, Li CC, et al. Challenges and opportunities for quantitative clinical pharmacology in cancer immunotherapy: something old, something new, something borrowed, and something blue. CPT Pharmacometrics Syst Pharmacol. 2015;4(9):495‐497.
    1. Rodallec A, Fanciullino R, Benzekry S, Ciccolini J; Group EP . Is there any room for pharmacometrics with immuno‐oncology drugs? Input from the EORTC‐PAMM course on preclinical and early‐phase clinical pharmacology. Anticancer Res. 2019;39(7):3419‐3422.
    1. Zhu J, Zhang T, Li J, et al. Association between tumor mutation burden (TMB) and outcomes of cancer patients treated with PD‐1/PD‐L1 inhibitions: a meta‐analysis. Front Pharmacol. 2019;10:673.
    1. Lee M, Samstein RM, Valero C, Chan TA, Morris LGT. Tumor mutational burden as a predictive biomarker for checkpoint inhibitor immunotherapy. Hum Vaccin Immunother. 2019;16:1‐4.
    1. Willis C, Fiander M, Tran D, et al. Tumor mutational burden in lung cancer: a systematic literature review. Oncotarget. 2019;10(61):6604‐6622.
    1. Abolhassani AR, Schuler G, Kirchberger MC, Heinzerling L. C‐reactive protein as an early marker of immune‐related adverse events. J Cancer Res Clin Oncol. 2019;145(10):2625‐2631.
    1. Marrone KA, Ying W, Naidoo J. Immune‐related adverse events from immune checkpoint inhibitors. Clin Pharmacol Ther. 2016;100(3):242‐251.
    1. Morrissey KM, Marchand M, Patel H, et al. Alternative dosing regimens for atezolizumab: an example of model‐informed drug development in the postmarketing setting. Cancer Chemother Pharmacol. 2019;84(6):1257‐1267.
    1. Bomze D, Hasan Ali O, Bate A, Flatz L. Association between immune‐related adverse events during anti‐PD‐1 therapy and tumor mutational burden. JAMA Oncol. 2019;5(11):1633‐1635.
    1. Osipov A, Lim SJ, Popovic A, et al. Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA‐4, and combination: a meta‐regression analysis. Clin Cancer Res. 2020;26(18):4842‐4851.
    1. Castle JC, Kreiter S, Diekmann J, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72(5):1081‐1091.
    1. Allgauer M, Budczies J, Christopoulos P, et al. Implementing tumor mutational burden (TMB) analysis in routine diagnostics—a primer for molecular pathologists and clinicians. Transl Lung Cancer Res. 2018;7(6):703‐715.
    1. Fancello L, Gandini S, Pelicci PG, Mazzarella L. Tumor mutational burden quantification from targeted gene panels: major advancements and challenges. J Immunother Cancer. 2019;7(1):183.
    1. Schnidrig D, Turajlic S, Litchfield K. Tumour mutational burden: primary versus metastatic tissue creates systematic bias. Immuno‐Oncol Technol. 2019;4:8‐14.
    1. Stenzinger A, Allen JD, Maas J, et al. Tumor mutational burden standardization initiatives: recommendations for consistent tumor mutational burden assessment in clinical samples to guide immunotherapy treatment decisions. Genes Chromosomes Cancer. 2019;58(8):578‐588.
    1. Merino DM, McShane LM, Fabrizio D, et al. Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project. J Immunother Cancer. 2020;8(1):e000147.
    1. Steuer CE, Ramalingam SS. Tumor mutation burden: leading immunotherapy to the era of precision medicine? J Clin Oncol. 2018;36(7):631‐632.

Source: PubMed

3
Sottoscrivi