Are tumor size changes predictive of survival for checkpoint blockade based immunotherapy in metastatic melanoma?

Meihua Wang, Cong Chen, Thomas Jemielita, James Anderson, Xiaoyun Nicole Li, Chen Hu, S Peter Kang, Nageatte Ibrahim, Scot Ebbinghaus, Meihua Wang, Cong Chen, Thomas Jemielita, James Anderson, Xiaoyun Nicole Li, Chen Hu, S Peter Kang, Nageatte Ibrahim, Scot Ebbinghaus

Abstract

Background: In oncology clinical development, objective response rate, disease control rate and early tumor size changes are commonly used as efficacy metrics for early decision-making. However, for immunotherapy trials, it is unclear whether these early efficacy metrics are still predictive of long-term clinical benefit such as overall survival. The goal of this paper is to identify appropriate early efficacy metrics predictive of overall survival for immunotherapy trials.

Methods: Based on several checkpoint blockade based immunotherapy studies in metastatic melanoma, we evaluated the predictive value of early tumor size changes and RECIST-based efficacy metrics at various time points on overall survival. The cut-off values for tumor size changes to predict survival were explored via tree based recursive partitioning and validated by external data. Sensitivity analyses were performed for the cut-offs.

Results: The continuous tumor size change metric and RECIST-based trichotomized response metric at different landmark time points were found to be statistically significantly associated with overall survival. The predictive values were higher at Week 12 and 18 than those at Week 24. The percentage of tumor size changes appeared to have comparable or lower predictive values than the RECIST-based trichotomized metric, and a cut-off of approximately 10% tumor reduction appeared to be reasonable for predicting survival.

Conclusions: An approximate 10% tumor reduction may be a reasonable cut-off for early decision-making while the RECIST-based efficacy metric remains the primary tool. Early landmark analysis is especially useful for decision making when accrual is fast. Composite response rate (utilizing different weights for PR/CR and SD) may be worth further investigation.

Trial registration: Clinical trials gov, NCT01295827 , Registered February 15, 2011; NCT01704287 , Registered October 11, 2012; NCT01866319 , Registered May 31, 2013.

Keywords: Cut-off values; Early efficacy metrics; Early tumor size changes; Immunotherapy trials; RECIST.

Conflict of interest statement

Ethics approval and consent to participate

All three study protocols and amendments were approved by appropriate institutional review boards and ethics committees at each institution. The studies were done in accordance with the protocols, Good Clinical Practice guidelines, and the declaration of Helsinki. All patients provided written informed consent. The studies were registered on ClinicalTrials.gov.

Consent for publication

Not applicable.

Competing interests

MW, CC, TJ, JA, NL, PK, NI and SE are employed by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, and hold stock in the company. CH serves as a research consultant for Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
c-index (95%CI) and AIC from Survival Models with OS Based on KEYNOTE-002 and -006. Lines correspond to c-index with 95% CI (leftside of Y axis); Triangles correspond to AIC (rightside of Y axis)
Fig. 2
Fig. 2
Overall Survival Curves Based on Validation Datasets (KEYNOTE-001 Melanoma Patients). Left panel shows the survival curves with 10% cut-off and the right panel shows survival curves with two cutoffs of 10 and 30% tumor reduction

References

    1. Eisenhauer E, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R., et al.. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1). Eur J Cancer, 2009;45(2):228–247.
    1. Karrison TG, Maitland ML, Stadler WM, Ratain MJ. Design of Phase II Cancer trials using a continuous endpoint of change in tumor size: application to a study of Sorafenib and Erlotinib in non–small-cell NSCLC Cancer. J Natl Cancer Inst. 2007;99(19):1455–1461. doi: 10.1093/jnci/djm158.
    1. Jaki T, Andre V, Su TL, Whitehead J. Designing exploratory cancer trials using change in tumour size as primary endpoint. Stat Med. 2013;32(15):2544–2554. doi: 10.1002/sim.5716.
    1. Suzuki C, Blomqvist L, Sundin A, Jacobsson H, Byström P, Berglund Å, et al. The initial change in tumor size predicts response and survival in patients with metastatic colorectal cancer treated with combination chemotherapy. Ann Oncol. 2012;23(4):948–954. doi: 10.1093/annonc/mdr350.
    1. Piessevaux H, Buyse M, Schlichting M, Van Cutsem E, Bokemeyer C, Heeger S, et al. Use of early tumor shrinkage to predict long-term outcome in metastatic colorectal cancer treated with cetuximab. J Clin Oncol. 2013;31(30):3764–3775. doi: 10.1200/JCO.2012.42.8532.
    1. An MW, Dong X, Meyers J, Han Y, Grothey A, Bogaerts J, et al. Evaluating Continuous Tumor Measurement-Based Metrics as Phase II Endpoints for Predicting Overall Survival. J Natl Cancer Inst. 2015;107(11):djv239. doi: 10.1093/jnci/djv239.
    1. An MW, Han Y, Meyers JP, Bogaerts J, Sargent DJ, Mandrekar SJ. Clinical utility of metrics based on tumor measurements in phase II trials to predict overall survival outcomes in phase III trials by using resampling methods. J Clin Oncol. 2015;33(34):4048–4057. doi: 10.1200/JCO.2015.60.8778.
    1. An MW, Mandrekar SJ, Branda ME, Hillman SL, Adjei AA, Pitot HC, et al. Comparison of continuous vs categorical tumor measurement-based metrics to predict overall survival in cancer treatment trials. Clin Cancer Res. 2011;17(20):6592–6599. doi: 10.1158/1078-0432.CCR-11-0822.
    1. Claret L, Gupta M, Han K, Joshi A, Sarapa N, He J, et al. Evaluation of tumour-size response metrics to predict overall survival in Western and Chinese patients with first-line metastatic colorectal Cancer. J Clin Oncol. 2013;31(17):2110–2114. doi: 10.1200/JCO.2012.45.0973.
    1. Taieb J, Rivera F, Siena S, Karthaus M, Valladares-Ayerbes M, Gallego J, et al. Exploratory analyses assessing the impact of early tumour shrinkage and depth of response on survival outcomes in patients with RAS wild-type metastatic colorectal cancer receiving treatment in three randomised panitumumab trials. J Cancer Res Clin Oncol. 2018;144(2):321–335. doi: 10.1007/s00432-017-2534-z.
    1. Heinemann V, Stintzing S, Modest DP, Giessen-Jung C, Michl M, Mansmann UR. Early tumour shrinkage (ETS) and depth of response (DpR) in the treatment of patients with metastatic colorectal cancer (mCRC) Eur J Cancer. 2015;51(14):1927–1936. doi: 10.1016/j.ejca.2015.06.116.
    1. Lee CK, Kim SS, Park S, Kim C, Heo SJ, Lim JS, et al. Depth of response is a significant predictor for long-term outcome in advanced gastric cancer patients treated with trastuzumab. Oncotarget. 2017;8(19):31169–31179.
    1. Kaufman H, Schwartz LH, William WN, Sznol M, Aguila M, Whittington C, et al. Evaluation of clinical endpoints as surrogates for overall survival in patients treated with immunotherapies. J Clin Oncol. 35(15_suppl - published online before print). 10.1200/JCO.2017.35.15_suppl.e14557.
    1. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbé C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15(23):7412–7420. doi: 10.1158/1078-0432.CCR-09-1624.
    1. Hodi FS, Hwu WJ, Kefford R, Weber JS, Daud A, Hamid O, et al. Evaluation of immune-related response criteria and RECIST v1.1 in patients with advanced melanoma treated with pembrolizumab. J Clin Oncol. 2016;34(13):1510–1517. doi: 10.1200/JCO.2015.64.0391.
    1. Blumenthal GM, Zhang L, Zhang H, Kazandjian D, Khozin S, Tang S, et al. Milestone Analyses of Immune Checkpoint Inhibitors, Targeted Therapy, and Conventional Therapy in Metastatic Non-Small Cell Lung Cancer Trials: A Meta-analysis. JAMA Oncol. 2017;3(8):e171029. doi: 10.1001/jamaoncol.2017.1029.
    1. Mushti S, 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;1902:2017. doi: 10.1158/1078-0432.CCR-17-1902.
    1. Krajewski KM, Franchetti Y, Nishino M, Fay AP, Ramaiya N, Van den Abbeele AD, et al. 10% tumor diameter shrinkage on the first follow-up computed tomography predicts clinical outcome in patients with advanced renal cell carcinoma treated with angiogenesis inhibitors: a follow-up validation study. Oncologist. 2014;19:507–514. doi: 10.1634/theoncologist.2013-0391.
    1. Krajewski KM, Guo M, Van den Abbeele AD, Yap J, Ramaiya N, Jagannathan J, et al. Comparison of four early post therapy imaging changes (EPTIC; RECIST 1.0, tumor shrinkage, computed tomography tumor density, Choi criteria) in assessing outcome to vascular endothelial growth factor-targeted therapy in patients with advanced renal cell carcinoma. Eur Urol. 2011;59:856–862. doi: 10.1016/j.eururo.2011.01.038.
    1. Sakamaki K, Kito Y, Yamazaki K, Izawa N, Tsuda T, Morita S, et al. Exploration of time points and cut-off values for early tumour shrinkage to predict survival outcomes of patients with metastatic colorectal cancer treated with first-line chemotherapy using a biexponential model for change in tumour size. ESMO Open. 2017;2(5):e000275. doi: 10.1136/esmoopen-2017-000275.
    1. Luo Y, Chen J. Optimisation of the Size Variation Threshold for CT Evaluation of Response in Advanced Gastroenteropancreatic Nauroendocrine Tumors Treated with Octreotide LAR. ENETS Annual Coference 2017; abstract K11.
    1. Lamarca A, Barriuso J, Kulke M, Borbath I, Lenz HJ, Raoul JL, et al. Determination of an optimal response cut-off able to predict progression-free survival in patients with well-differentiated advanced pancreatic neuroendocrine tumours treated with sunitinib: an alternative to the current RECIST-defined response. Br J Cancer. 2018;118(2):181–188. doi: 10.1038/bjc.2017.402.
    1. Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–918. doi: 10.1016/S1470-2045(15)00083-2.
    1. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–2532. doi: 10.1056/NEJMoa1503093.
    1. Schachter J, Ribas A, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006) Lancet. 2017;390(10105):1853–1862. doi: 10.1016/S0140-6736(17)31601-X.
    1. Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA. 2016;315(15):1600–1609. doi: 10.1001/jama.2016.4059.
    1. Joseph R, Elassaiss-Schaap J, Kefford R, Hwu W, Wolchok J, Joshua A, et al. Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with Pembrolizumab. Clin Cancer Res. 2018;24(20):4960–4967. doi: 10.1158/1078-0432.CCR-17-2386.
    1. Anderson J, Cain K, Gelber R. Analysis of survival by tumor response. J Clin Oncol. 1983;1(11):710–719. doi: 10.1200/JCO.1983.1.11.710.
    1. Breiman L, Friedman J, Olshen R, Stone C. Classification and regression trees. Belmont, Ca: Wadsworth; 1984.
    1. Breiman L. Bagging predictors. Mach Learn. 1996;24(2):123–140.
    1. Blanche P, Dartigues J, Jacqmin-Gadda H. Estimating and comparing time dependent areas under receiver operating characteristic curves for censored event times with competing risks. Stat in med. 2013;32(30):5381–5397. doi: 10.1002/sim.5958.
    1. Li L, Greene T, Hu B. A simple method to estimate the time-dependent receiver operating characteristic curve and the area under the curve with right censored data. Stat methods Med Res. 2016;27(8):2264-2278.
    1. Claeskens G, Hjort N. Model Selection and Model Averaging. Cambridge; Cambridge University Press; 2008.
    1. Harrell FE, Califf RM, Pryor DB, Lee KL, Rosati RA. Evaluating the yield of medical tests. JAMA. 1982;247:2543–2546. doi: 10.1001/jama.1982.03320430047030.
    1. Harrell FE, Lee KL, Califf RM, Pryor DB, Lee KL, Rosati RA. Regression modeling strategies for improved prognostic prediction. Stat in Med. 1984;3:143–152. doi: 10.1002/sim.4780030207.
    1. Harrell FE, Lee KL, Mark DB. Tutorial in biostatistics: multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat in Med. 1996;15:361–387. doi: 10.1002/(SICI)1097-0258(19960229)15:4<361::AID-SIM168>;2-4.
    1. Hastie T, Tibshirani R, Friedman JH. The elements of statistical learning: data mining, inference, and prediction. 2. New York: Springer; 2009.

Source: PubMed

3
订阅