Sensitivity of treatment-free survival to subgroup analyses in patients with advanced melanoma treated with immune checkpoint inhibitors

Charlene M Mantia, Lillian Werner, Brian Stwalley, Corey Ritchings, Ahmad A Tarhini, Michael B Atkins, David F McDermott, Meredith M Regan, Charlene M Mantia, Lillian Werner, Brian Stwalley, Corey Ritchings, Ahmad A Tarhini, Michael B Atkins, David F McDermott, Meredith M Regan

Abstract

Patients with advanced melanoma treated with immune checkpoint inhibitors can experience ongoing disease control after treatment discontinuation without subsequent systemic anticancer therapy. We previously defined a novel outcome, treatment-free survival (TFS), as the time between protocol therapy cessation and subsequent therapy initiation/death. We assessed the effect of established prognostic variables [lactate dehydrogenase (LDH), programmed death ligand 1 status, BRAF mutation status, performance status, and sex] on TFS in different treatment scenarios: treatment until toxicity/progression with frequent early cessation (nivolumab plus ipilimumab), treatment until toxicity/progression with a well-tolerated regimen (nivolumab), and treatment for a short fixed duration (ipilimumab). Data were pooled from 1077 patients with advanced melanoma treated in the CheckMate 069 and 067 trials. TFS was defined as the area between the Kaplan-Meier curves for time to therapy cessation and time to subsequent therapy initiation/death. TFS was estimated by restricted mean (r-mean) survival time at 36 months since randomization. Clinically meaningful TFS (r-mean TFS 3.7-12.7 months) was observed across all patient subgroups. TFS was longest in patients treated with nivolumab plus ipilimumab. The largest differences in r-mean TFS were observed with LDH in the nivolumab plus ipilimumab and ipilimumab treatment groups (TFS difference 4.7 and 4.9 months, respectively). In the nivolumab group, there was little difference in TFS across subgroups (r-mean TFS 3.7-5.5 months). TFS was sensitive to prognostic subgroup differences; however, duration of treatment affected the sensitivity of TFS. These results provide further support for TFS as a clinical outcome measure.

Trial registration: ClinicalTrials.gov NCT01927419 NCT01844505.

Conflict of interest statement

L.W. has worked as a consultant to Bristol Myers Squibb. B.S. is an employee of and stockholder in Bristol Myers Squibb. C.R. is an employee of Bristol Myers Squibb. A.A.T. has received institutional research grants from Bristol Myers Squibb, Genentech/Roche, Merck, and OncoSec; and served as a consultant/advisor for Array Biopharma, BioNTech, Bristol Myers Squibb, EMD Serono, Genentech/Roche, Immunocore, Merck, NewLink Genetics, Novartis, Partner Therapeutics, Pfizer, and Sanofi-Genzyme/Regeneron. M.B.A. has served as an advisor for Arrowhead, Aveo, Bristol Myers Squibb, Eisai, Elpis, Genetech/Roche, Leads, Merck, Novartis, Pfizer, Pneuma, Pyxis Oncology, Werewolf; and as a consultant to Adagene, Agenus, Apexigen, Exelixis, Idera, ImmunoCore, Iovance, and Neoleuken. D.F.M. has served as a consultant to Alkermes, Bristol Myers Squibb, Eisai, Eli Lilly and Company, EMD Serono, Iovance, Merck, and Pfizer; received research support from Alkermes, Bristol Myers Squibb, Exelixis, Genentech, Merck, Pfizer, and X4 Pharma. M.M.R. has served as a consultant to Ipsen/Debiopharm; received research support from Bayer and Bristol Myers Squibb; received institutional research support from Bristol Myers Squibb, Ipsen, Ferring, Merck, Novartis, Pfizer, Pierre Fabre, Roche, and TerSera; received personal fees from Bristol Myers Squibb and Tolmar Pharmaceuticals; and received nonfinancial support from Bristol Myers Squibb. For the remaining author, there is no conflicts of interest.

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

Fig. 1
Fig. 1
Schematic illustration of the end points that partition the area under the OS curve into TFS and other resulting health states. ICI, immune checkpoint inhibitor; OS, overall survival; TFS, treatment-free survival. Adapted from J Clin Oncol [7] under Creative Commons License 4.0 [CC BY 4.0].
Fig. 2
Fig. 2
Estimates of TFS and other health states over the 36-month period in patients treated with nivolumab plus ipilimumab according to patient subgroups based on (a) LDH status, (b) PD-L1 status and (c) BRAF mutation status; r-mean times (months) are annotated on the health state areas. ICI, immune checkpoint inhibitor; LDH, lactate dehydrogenase; PD-L1, programmed death 1; TFS, treatment-free survival.
Fig. 3
Fig. 3
Estimates of TFS and other health states over the 36-month period in patients treated with nivolumab according to patient subgroups based on (a) LDH status, (b) PD-L1 status and (c) BRAF mutation status; r-mean times (months) are annotated on the health state areas. ICI, immune checkpoint inhibitor; LDH, lactate dehydrogenase; PD-L1, programmed death 1; TFS, treatment-free survival.
Fig. 4
Fig. 4
Estimates of TFS and other health states over the 36-month period in patients treated with ipilimumab according to patient subgroups based on (a) LDH status, (b) PD-L1 status and (c) BRAF mutation status; r-mean times (months) are annotated on the health state areas; r-mean times (months) are annotated on the health state areas. ICI, immune checkpoint inhibitor; LDH, lactate dehydrogenase; PD-L1, programmed death 1; TFS, treatment-free survival.

References

    1. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, et al. . Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017; 377:1345–1356.
    1. Hodi FS, Chesney J, Pavlick AC, Robert C, Grossmann KF, McDermott DF, et al. . Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol 2016; 17:1558–1568.
    1. Robert C, Ribas A, Hamid O, Daud A, Wolchok JD, Joshua AM, et al. . Durable complete response after discontinuation of pembrolizumab in patients with metastatic melanoma. J Clin Oncol 2018; 36:1668–1674.
    1. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. . Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol 2019; 30:582–588.
    1. Robert C, Ribas A, Schachter J, Arance A, Grob JJ, Mortier L, et al. . Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol 2019; 20:1239–1251.
    1. Jansen YJL, Rozeman EA, Mason R, Goldinger SM, Geukes Foppen MH, Hoejberg L, et al. . Discontinuation of anti-PD-1 antibody therapy in the absence of disease progression or treatment limiting toxicity: clinical outcomes in advanced melanoma. Ann Oncol 2019; 30:1154–1161.
    1. Regan MM, Werner L, Rao S, Gupte-Singh K, Hodi FS, Kirkwood JM, et al. . Treatment-free survival: a novel outcome measure of the effects of immune checkpoint inhibition-A pooled analysis of patients with advanced melanoma. J Clin Oncol 2019; 37:3350–3358.
    1. Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR, et al. . Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 2009; 27:6199–6206.
    1. Manola J, Atkins M, Ibrahim J, Kirkwood J. Prognostic factors in metastatic melanoma: a pooled analysis of Eastern Cooperative Oncology Group trials. J Clin Oncol 2000; 18:3782–3793.
    1. Nakamura Y, Kitano S, Takahashi A, Tsutsumida A, Namikawa K, Tanese K, et al. . Nivolumab for advanced melanoma: pretreatment prognostic factors and early outcome markers during therapy. Oncotarget 2016; 7:77404–77415.
    1. Sirott MN, Bajorin DF, Wong GY, Tao Y, Chapman PB, Templeton MA, Houghton AN. Prognostic factors in patients with metastatic malignant melanoma. A multivariate analysis. Cancer 1993; 72:3091–3098.
    1. Agarwala SS, Keilholz U, Gilles E, Bedikian AY, Wu J, Kay R, et al. . LDH correlation with survival in advanced melanoma from two large, randomised trials (Oblimersen GM301 and EORTC 18951). Eur J Cancer 2009; 45:1807–1814.
    1. Hodi FS, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Cowey CL, et al. . Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol 2018; 19:1480–1492.
    1. Kumar R, Angelini S, Czene K, Sauroja I, Hahka-Kemppinen M, Pyrhönen S, Hemminki K. BRAF mutations in metastatic melanoma: a possible association with clinical outcome. Clin Cancer Res 2003; 9:3362–3368.
    1. Long GV, Menzies AM, Nagrial AM, Haydu LE, Hamilton AL, Mann GJ, et al. . Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 2011; 29:1239–1246.
    1. von Moos R, Seifert B, Simcock M, Goldinger SM, Gillessen S, Ochsenbein A, et al. Swiss Group for Clinical Cancer Research (SAKK). First-line temozolomide combined with bevacizumab in metastatic melanoma: a multicentre phase II trial (SAKK 50/07). Ann Oncol 2012; 23:531–536.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. . Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2019; 381:1535–1546.
    1. Christiansen SA, Swoboda D, Gardner K, et al. . Off treatment survival (OTS) in patients (pts) with advanced melanoma after anti-PD1 therapy. J Clin Oncol 2018; 36 (15 Suppl):9554. Abstract 9554.
    1. Tan AC, Emmett L, Lo S, Liu V, Kapoor R, Carlino MS, et al. . FDG-PET response and outcome from anti-PD-1 therapy in metastatic melanoma. Ann Oncol 2018; 29:2115–2120.
    1. Palmer SR, Erickson LA, Ichetovkin I, Knauer DJ, Markovic SN. Circulating serologic and molecular biomarkers in malignant melanoma. Mayo Clin Proc 2011; 86:981–990.
    1. Ny L, Hernberg M, Nyakas M, Koivunen J, Oddershede L, Yoon M, et al. . BRAF mutational status as a prognostic marker for survival in malignant melanoma: a systematic review and meta-analysis. Acta Oncol 2020; 59:833–844.

Source: PubMed

3
Abonner