Model-based meta-analysis of non-small cell lung cancer with standard of care PD-1 inhibitors and chemotherapy for early development decision making

David C Turner, Russ Wada, Helen Zhou, Xiaowei Wang, Rik de Greef, Chandni Valiathan, Lindsey Zhang, Nancy Zhang, Mita Kuchimanchi, Tai-Tsang Chen, Marc Ballas, Sandra A G Visser, David C Turner, Russ Wada, Helen Zhou, Xiaowei Wang, Rik de Greef, Chandni Valiathan, Lindsey Zhang, Nancy Zhang, Mita Kuchimanchi, Tai-Tsang Chen, Marc Ballas, Sandra A G Visser

Abstract

Single-arm cohorts/trials are often used in early phase oncology programs to support preliminary clinical activity assessments for investigational products, administered alone or in combination with standard of care (SOC) agents. Benchmarking clinical activity of those combinations against other treatments, including SOC, requires indirect comparisons against external trials, which presents challenges including cross-study differences in trial populations/other factors. To facilitate such nonrandomized comparisons, we developed a comprehensive model-based meta-analysis (MBMA) framework to quantitatively adjust for factors related to efficacy in metastatic non-small cell lung cancer (mNSCLC). Data were derived from 15 published studies assessing key programmed cell death protein-1 (PD-1) inhibitors pembrolizumab (n = 8) and nivolumab (n = 7), representing current SOC in mNSCLC. In the first stage, a mixed-effects logistic regression model for overall response rate (ORR) was developed accounting for effects of various population covariates on ORR. The ORR model results indicated an odds ratio (OR) of 1.02 for squamous versus non-squamous histology and OR of 1.20 for PD-ligand 1 tumor proportion score (TPS) per every 10% increase of TPS level. Next, a nonparametric mixed-effects model for overall survival (OS) was developed with ORR/other clinical covariates as input. Subsequently, MBMA simulations of relevant hypothetical scenarios involving single-arm trial design predicted OS hazard ratios as a function of ORR with matched patient characteristics. Findings from this MBMA and derived parameter estimates can be generally applied by the reader as a framework for interpreting efficacy data from early phase trials to support ORR-based go/no-go decisions and futility rules, illustrated through examples in this report.

Conflict of interest statement

D.C.T., H.Z., X.W., C.V., M.K., T.T.C., M.B., and S.A.G.V. were employees of and stockholders in GSK at the time this work was completed. D.C.T. is a former employee of GSK and currently employed by Genentech. R.W. was an employee in Certara (Certara was a consultant to GSK) at the time this work was completed, and is currently employed at QuanTx Consulting. X.W. is a former employee of GSK and currently employed by Moderna. C.V. is a former employee of GSK and currently employed by J&J. M.B. is a former employee of GSK and currently employed by Novocure. R.G., L.Z., and N.Z. are employees of Certara, and consultants for GSK.

© 2023 GSK, GlaxoSmithKline. CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

FIGURE 1
FIGURE 1
The NSCLC MBMA framework defines ORR and ORR‐to‐OS relationships across lines of therapy and treatment types for generating synthetic control arms and projecting OS. Chemo, chemotherapy; MBMA, model‐based meta‐analysis; nivo, nivolumab; NME, new molecular entity; NSCLC, non‐small cell lung cancer; ORR, overall response rate; OS, overall survival; Ph, phase; pembro, pembrolizumab; SOC, standard of care.
FIGURE 2
FIGURE 2
Illustration of conditional probability method for modeling aggregate survival curves. Pj, conditional probability; Sj, survival up to the beginning of the interval j.
FIGURE 3
FIGURE 3
Forest plot of the impact of ORR model covariates for PD‐1 inhibitors and relevant chemotherapy comparators from selected pembrolizumab and nivolumab clinical trials (n = 15). The center line represents the modeled ORR value for a patient with 50% squamous histology and 50% PD‐L1 expression receiving 2L PD‐1 inhibitor with chemotherapy. Each point illustrates the modeled ORR rate for the univariate change described, with the indicated 90% CI. “PD‐L1 superhigh” is defined as greater than or equal to 90% PD‐L1 tumor expression and corresponds to imputed continuous PD‐L1 expression value of 95%. “PD‐L1 high” is defined as greater than or equal to 50% PD‐L1 tumor expression and corresponds to imputed continuous PD‐L1 expression value of 70%. “PD‐L1 weak” is defined as 1%–49% PD‐L1 tumor expression and corresponds to imputed continuous PD‐L1 expression value of 25%. “PD‐L1 negative” is defined as less than 1% PD‐L1 tumor expression and corresponds to imputed continuous PD‐L1 expression value of 0%. 1L, first line; ≥2L, second line or greater; CI, confidence interval; combo, combination therapy; ORR, overall response rate; PD‐1, programmed cell death protein‐1; PD‐L1, programmed death‐ligand 1.
FIGURE 4
FIGURE 4
Exploratory ORR‐to‐OS correlation of SOC PD‐1 inhibitor and chemotherapy alone for (a) median OS and (b) OS at 1‐year. Any IO, any immuno‐oncology therapy containing arm; chemo alone, chemotherapy alone; ORR, overall response rate; OS, overall survival; PD‐1, programmed cell death protein‐1; SOC, standard of care.
FIGURE 5
FIGURE 5
Forest plot of the impact of covariates on OS. The center line represents the modeled OS value for a patient with 50% squamous histology and 50% PD‐L1 tumor expression receiving 2L PD‐1 inhibitor in combination with chemotherapy. Each point illustrates the modeled OS rate for the univariate change described with whiskers representing a 90% confidence interval. “PD‐L1 superhigh” is defined as 95% PD‐L1 tumor expression. “PD‐L1 high” is defined as 70% PD‐L1 tumor expression. “PD‐L1 weak” is defined as 25% PD‐L1 tumor expression. “PD‐L1 negative” is defined as 0% PD‐L1 tumor expression. 1L, first line; ≥2L, second line or greater; IO, immuno‐oncology; OS, overall survival; PD‐L1, programmed death‐ligand 1.
FIGURE 6
FIGURE 6
ORR difference achieved for hypothetical phase I/II cohorts of IO and chemotherapy in (a) “base case” (b) “low responder” (c), and “high responder” scenarios. A “base case” scenario for patients with 20% squamous histology, 30% continuous PD‐L1 TPS. A “low responder” scenario for patients with 10% squamous histology, 15% continuous PD‐L1 TPS. A “high responder” scenario for patients with 30% squamous histology, 50% continuous PD‐L1 TPS. HR, hazard ratio; IO, immuno‐oncology; ORR, overall response rate; PD‐L1, programmed death‐ligand 1; sq, squamous histology, TPS, tumor proportion score.

References

    1. Xia L, Liu Y, Wang Y. PD‐1/PD‐L1 blockade therapy in advanced non‐small‐cell lung cancer: current status and future directions. Oncologist. 2019;24:S31‐s41.
    1. Food and Drug Administration . FDA approves cemiplimab‐rwlc for non‐small cell lung cancer with high PD‐L1 expression. Accessed January 18, 2023.
    1. Snyders K, Cho D, Hong JH, et al. Benchmarking single‐arm studies against historical controls from non‐small cell lung cancer trials ‐ an empirical analysis of bias. Acta Oncol. 2020;59:90‐95.
    1. Thorlund K, Dron L, Park JJH, Mills EJ. Synthetic and external controls in clinical trials ‐ a primer for researchers. Clin Epidemiol. 2020;12:457‐467.
    1. Food and Drug Administration . Submitting Documents Using Real‐World Data and Real‐World Evidence to FDA for Drugs and Biologics Guidance for Industry. Accessed January 18, 2023.
    1. EMA . Committee for Medicinal Products for human use (CHMP): guideline on clinical trials in small populations. Accessed January 18, 2023.
    1. Mandema JW, Gibbs M, Boyd RA, Wada DR, Pfister M. Model‐based meta‐analysis for comparative efficacy and safety: application in drug development and beyond. Clin Pharmacol Ther. 2011;90:766‐769.
    1. Shameer K, Zhang Y, Jackson D, et al. Correlation between early endpoints and overall survival in non‐small‐cell lung cancer: a trial‐level meta‐analysis. Front Oncol. 2021;11:672916.
    1. Nie RC, Chen FP, Yuan SQ, et al. Evaluation of objective response, disease control and progression‐free survival as surrogate end‐points for overall survival in anti‐programmed death‐1 and anti‐programmed death ligand 1 trials. Eur J Cancer. 2019;106:1‐11.
    1. Mushti SL, Mulkey F, Sridhara R. Evaluation of overall response rate and progression‐free survival as potential surrogate endpoints for overall survival in immunotherapy trials. Clin Cancer Res. 2018;24:2268‐2275.
    1. Kaufman HL, Schwartz LH, William WN Jr, et al. Evaluation of classical clinical endpoints as surrogates for overall survival in patients treated with immune checkpoint blockers: a systematic review and meta‐analysis. J Cancer Res Clin Oncol. 2018;144:2245‐2261.
    1. Goring S, Varol N, Waser N, et al. Correlations between objective response rate and survival‐based endpoints in first‐line advanced non‐small cell lung cancer: a systematic review and meta‐analysis. Lung Cancer. 2022;170:122‐132.
    1. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non‐small‐cell lung cancer. N Engl J Med. 2015;372:2018‐2028.
    1. Hui R, Garon EB, Goldman JW, et al. Pembrolizumab as first‐line therapy for patients with PD‐L1‐positive advanced non‐small cell lung cancer: a phase 1 trial. Ann Oncol. 2017;28:874‐881.
    1. Chatterjee M, Turner DC, Felip E, et al. Systematic evaluation of pembrolizumab dosing in patients with advanced non‐small‐cell lung cancer. Ann Oncol. 2016;27:1291‐1298.
    1. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD‐L1‐positive, advanced non‐small‐cell lung cancer (KEYNOTE‐010): a randomised controlled trial. Lancet. 2016;387:1540‐1550.
    1. Langer CJ, Gadgeel SM, Borghaei H, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non‐squamous non‐small‐cell lung cancer: a randomised, phase 2 cohort of the open‐label KEYNOTE‐021 study. Lancet Oncol. 2016;17:1497‐1508.
    1. Gadgeel SM, Stevenson JP, Langer CJ, et al. Pembrolizumab and platinum‐based chemotherapy as first‐line therapy for advanced non‐small‐cell lung cancer: phase 1 cohorts from the KEYNOTE‐021 study. Lung Cancer. 2018;125:273‐281.
    1. Reck M, Rodríguez‐Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD‐L1‐positive non‐small‐cell lung cancer. N Engl J Med. 2016;375:1823‐1833.
    1. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD‐L1‐expressing, locally advanced or metastatic non‐small‐cell lung cancer (KEYNOTE‐042): a randomised, open‐label, controlled, phase 3 trial. Lancet. 2019;393:1819‐1830.
    1. Gandhi L, Rodríguez‐Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non‐small‐cell lung cancer. N Engl J Med. 2018;378:2078‐2092.
    1. Paz‐Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non‐small‐cell lung cancer. N Engl J Med. 2018;379:2040‐2051.
    1. Gettinger S, Rizvi NA, Chow LQ, et al. Nivolumab monotherapy for first‐line treatment of advanced non‐small‐cell lung cancer. J Clin Oncol. 2016;34:2980‐2987.
    1. Gettinger SN, Horn L, Gandhi L, et al. Overall survival and long‐term safety of nivolumab (anti‐programmed death 1 antibody, BMS‐936558, ONO‐4538) in patients with previously treated advanced non‐small‐cell lung cancer. J Clin Oncol. 2015;33:2004‐2012.
    1. Rizvi NA, Hellmann MD, Brahmer JR, et al. Nivolumab in combination with platinum‐based doublet chemotherapy for first‐line treatment of advanced non‐small‐cell lung cancer. J Clin Oncol. 2016;34:2969‐2979.
    1. Hellmann MD, Rizvi NA, Goldman JW, et al. Nivolumab plus ipilimumab as first‐line treatment for advanced non‐small‐cell lung cancer (CheckMate 012): results of an open‐label, phase 1, multicohort study. Lancet Oncol. 2017;18:31‐41.
    1. Hellmann MD, Paz‐Ares L, Bernabe Caro R, et al. Nivolumab plus ipilimumab in advanced non‐small‐cell lung cancer. N Engl J Med. 2019;381:2020‐2031.
    1. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous‐cell non‐small‐cell lung cancer. N Engl J Med. 2015;373:123‐135.
    1. Carbone DP, Reck M, Paz‐Ares L, et al. First‐line nivolumab in stage IV or recurrent non‐small‐cell lung cancer. N Engl J Med. 2017;376:2415‐2426.
    1. Borghaei H, Paz‐Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non‐small‐cell lung cancer. N Engl J Med. 2015;373:1627‐1639.
    1. Rizvi NA, Mazières J, Planchard D, et al. Activity and safety of nivolumab, an anti‐PD‐1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non‐small‐cell lung cancer (CheckMate 063): a phase 2, single‐arm trial. Lancet Oncol. 2015;16:257‐265.
    1. Aguilar EJ, Ricciuti B, Gainor JF, et al. Outcomes to first‐line pembrolizumab in patients with non‐small‐cell lung cancer and very high PD‐L1 expression. Ann Oncol. 2019;30:1653‐1659.
    1. Abdel‐Rahman O. Correlation between PD‐L1 expression and outcome of NSCLC patients treated with anti‐PD‐1/PD‐L1 agents: a meta‐analysis. Crit Rev Oncol Hematol. 2016;101:75‐85.
    1. Jansen JP. Network meta‐analysis of survival data with fractional polynomials. BMC Med Res Methodol. 2011;11:61.
    1. Guyot P, Ades AE, Ouwens MJNM, Welton NJ. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan‐Meier survival curves. BMC Med Res Methodol. 2012;12:9.
    1. Borcoman E, Kanjanapan Y, Champiat S, et al. Novel patterns of response under immunotherapy. Ann Oncol. 2019;30:385‐396.
    1. Huo G, Liu W, Chen P. Inhibitors of PD‐1 in non‐small cell lung cancer: a meta‐analysis of clinical and molecular features. Front Immunol. 2022;13:875093.

Source: PubMed

3
Abonner