Efficacy of nivolumab and ipilimumab in patients with malignant pleural mesothelioma is related to a subtype of effector memory cytotoxic T cells: Translational evidence from two clinical trials

Joanne M Mankor, Maria J Disselhorst, Myrthe Poncin, Paul Baas, Joachim G J V Aerts, Heleen Vroman, Joanne M Mankor, Maria J Disselhorst, Myrthe Poncin, Paul Baas, Joachim G J V Aerts, Heleen Vroman

Abstract

Background: Combined immune checkpoint inhibitor (ICI) treatment targeting PD-1 and CTLA-4 was suggested to yield clinical benefit over chemotherapy in malignant pleural mesothelioma (MPM), whereas aPD-1 monotherapy failed to provide benefit in phase-III trials. Success of ICI depends on the presence and activation of tumor-specific T cells. Therefore, we investigated whether T-cell characteristics are underlying clinical efficacy of ICI treatment in MPM.

Methods: Comprehensive immune cell profiling was performed on screening and on treatment peripheral blood samples of mesothelioma patients treated with nivolumab (aPD-1) monotherapy (NCT02497508), or a combination of nivolumab and ipilimumab (aCTLA-4) (NCT03048474).

Findings: aPD-1/aCTLA-4 combination treatment induced a profound increase in proliferation and activation of T cells, which was not observed upon aPD-1 monotherapy. Moreover, patients that responded to combination treatment had low frequencies of naive CD8 T cells and high frequencies of effector memory CD8 T cells that re-expressed RA (TEMRA) at screening. The frequency of Granzyme-B and Interferon-γ producing TEMRAs was also higher in responding patients.

Interpretation: High proportions of TEMRAs and cytokine production by TEMRAs before treatment, was associated with a better clinical outcome. TEMRAs, which likely comprise tumor-specific T cells, tend to require blockage of both aPD-1 and aCTLA-4 to be reactivated. In conclusion, peripheral blood TEMRAs can play a key role in explaining and predicting clinical benefit upon aPD-1/aCTLA-4 combination treatment.

Funding: Bristol-Myers Squibb sponsored NivoMes and INITIATE clinical trials and provided study drugs. No external funding was applicable for the flow cytometric analyses of peripheral blood samples described in this manuscript.

Keywords: Immune checkpoint inhibitors; Immune monitoring; Immunotherapy; Ipilimumab; Malignant pleural mesothelioma; Nivolumab.

Conflict of interest statement

Declaration of Competing Interest Prof. dr. Aerts reports personal fees from Eli Lilly, Roche, Boehringer Ingelheim, BMS, MSD, Amphera and AstraZeneca. Furthermore, prof. dr. Aerts has a patent pending on tumour lysate antigen (EP2938354A1) and is stock owner at Amphera B.V. Immunotherapy. Prof. dr. Baas reports grants from BMS, during the conduct of this study. Furthermore, he receives grants from MSD, outside the submitted work. All other authors do not have any financial relationships to disclose.

Copyright © 2020. Published by Elsevier B.V.

Figures

Fig. 1
Fig. 1
T- and NK-cell characteristics before and during aPD-1 monotherapy (a, c, d, f, h) Gating strategy for NK-cells (a), T-cells (c), CD4 T-cells subsets (d), CD8 T-cells subsets (f) and Treg subsets (h) respectively. (b, e, g, i) Percentage of T-and NK-cell subsets (b), CD4 T-cell subsets (e), CD8 T-cells subsets (g) and Treg subsets (I) respectively, at screening and on-treatment time points. (j, k, l) Percentage of Ki67+ CD 4 T-cell subsets (j), Tregs subsets (k) and CD8 T-cell subsets (l) respectively, at screening and on-treatment time points. (m, n, o). Paired samples are shown connected by black lines. Percentage of CD4 T-cell subsets (m), Treg subsets (n) and CD8 T-cell subsets (o) respectively, at the screening time point in responding and non-responding patients. Bars depict mean values with standard error of the mean.
Fig. 2
Fig. 2
T- and NK-cell characteristics before and during aPD-1/CTLA-4 combination therapy (a, b, c, d) Percentage of T-and NK-cell subsets (a), CD4 T-cell subsets (b), Treg subsets (c) and CD8 T-cells subsets (d) respectively, at screening and on-treatment time points. (e, f, g) Percentage of Ki67+ CD 4 T-cell subsets, (TCM p = 0.003, TEM p = 0.007, TEMRA p = 0.028) (e), Tregs subsets (f) and CD8 T-cell subsets (TN p = 0.036, TCM p = 0.03,) (g) respectively, at screening and on-treatment time points. (h, i, j) Comparison between responding (R) and non-responding (NR) patients for the percentage of Ki67+ CD 4 T-cell subsets (TCM R p = 0.01, TCM NR p = 0.04, TEM R p = 0.01) (h), Tregs subsets (i) and CD8 T-cell subsets (j) respectively, at screening and on-treatment time points. Paired samples are shown connected by black lines in each graph. Significance (Wilcoxon signed-rank test for paired analysis of screening and on-treatment samples and Mann–Whitney U test for comparison of response groups) is shown in each graph, with * p < 0.05 and ** p < 0.01. P values were corrected for multiple testing, using the Benjamini and Hochberg False Discovery Rate.
Fig. 3
Fig. 3
Percentage of ICOS+ T cell subsets before and during aPD-1/CTLA-4 combination therapy (a, b, c) Percentage of ICOS+ CD 4 T-cell subsets (TCM p = 0.002, TEM p = 0.003, TEMRA p = 0.004) (a), Tregs subsets (b) and CD8 T-cell subsets (TCM p =  0.003, TEM p = 0.012) (c) respectively, at screening and on-treatment time points. (d, e, f) Comparison between responding (R) and non-responding (NR) patients for the percentage of ICOS+ CD 4 T-cell subsets (TN NR p = 0.01, TCM NR p = 0.02, TEM NR p = 0.03, TEMRA NR p = 0.01 (d), Tregs subsets (nTreg NR p = 0.01) (e) and CD8 T-cell subsets (TCM R p = 0.03) (f) respectively, at screening and on-treatment time points. Paired samples are shown connected by black lines in each graph. Significance (Wilcoxon signed-rank test) is shown in each graph, with * p < 0.05 and ** p < 0.01. P values were corrected for multiple testing, using the Benjamini and Hochberg False Discovery Rate
Fig. 4
Fig. 4
Comparison of T-cell characteristics before aPD-1/CTLA-4 combination therapy in responding and non-responding patients (a, b, c) Percentage of CD4 T-cell subsets (a), Treg subsets (b) and CD8 T-cell subsets (TN p = 0.017, TCM p = 0.008, TEMRA p = 0.028) (c) respectively, at the screening time point in responding and non-responding patients. Bars depict mean values with standard error of the mean. Significance (Mann–Whitney U test) is shown in each graph, with * p < 0.05 and ** p < 0.01. P values were corrected for multiple testing, using the Benjamini and Hochberg False Discovery Rate. (d, e) EMRA CD8 T-cells proportions prior to treatment were used to stratify progression-free survival (PFS) (d) and overall survival (OS) (e). Median proportion of EMRA CD8 T cells was used as a cut off between the ‘high’ vs ‘low’ group. Statistical significance of the difference between the two Kaplan–Meier curves was tested by log rank test with p = 0.045 for PFS (median PFS of 3.5 vs 13.1 months) and p = 0.086 for OS (median OS of 10.2 vs 25.9 months).
Fig. 5
Fig. 5
Comparison of cytokine frequencies in CD8 T-cell subsets before aPD-1/CTLA-4 combination therapy in responding and non-responding patients (a, b) Percentage of IFNγ+ CD8 T-cell subsets (TEM p = 0.008, TEMRA p = 0.006) (a) and Granzyme-B+ CD8 T-cell subsets (TN p = 0.02, TCM p = 0.032, TEMRA p = 0.02) (b) respectively, at the screening time point in responding and non-responding patients. Bars depict mean values with standard error of the mean. Significance (Mann–Whitney U test) is shown in each graph, with * p < 0.05 and ** p < 0.01. P values were corrected for multiple testing, using the Benjamini and Hochberg False Discovery Rate. (c, d) Proportions of Granzyme-B+ EMRA CD8 T-cells prior to treatment were used to stratify progression-free survival (PFS) (d) and overall survival (OS) (e). Median proportion of Granzyme-B+ EMRA CD 8 T cells was used as a cut off between the ‘high’ vs ‘low’ group. Statistical significance of the difference between the two Kaplan–Meier curves was tested by log rank test with p = 0.14 for PFS (not significant, median PFS of 3.5 vs 10.8 months) and p = 0.051 for OS (not significant, median OS of 10.2 vs 32.6 months).

References

    1. Vogelzang NJ, Rusthoven JJ, Symanowski J, Denham C, Kaukel E, Ruffie P. Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol. 2003;21:2636–2644.
    1. Zalcman G, Mazieres J, Margery J, Greillier L, Audigier-Valette C, Moro-Sibilot D. Bevacizumab for newly diagnosed pleural mesothelioma in the Mesothelioma Avastin Cisplatin Pemetrexed Study (MAPS): a randomised, controlled, open-label, phase 3 trial. Lancet. 2016;387:1405–1414.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018
    1. Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018;553:7689. 446.
    1. Michielin O, Van Akkooi ACJ, Ascierto PA, Dummer R, Keilholz U. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2019;30:1884–1901.
    1. Maio M, Scherpereel A, Calabrò L, Aerts J, Perez SC, Bearz A. Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): a multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial. Lancet Oncol. 2017;18:1261–1273.
    1. Alley EW, Lopez J, Santoro A, Morosky A, Saraf S, Piperdi B. Clinical safety and activity of pembrolizumab in patients with malignant pleural mesothelioma (KEYNOTE-028): preliminary results from a non-randomised, open-label, phase 1b trial. Lancet Oncol. 2017;18:623–630.
    1. Quispel-Janssen J, van der Noort V, de Vries JF, Zimmerman M, Lalezari F, Thunnissen E. Programmed death 1 blockade with nivolumab in patients with recurrent malignant pleural mesothelioma. J Thorac Oncol. 2018;13:1569–1576.
    1. Popat S, Curioni-Fontecedro A, Polydoropoulou V, Shah R, O’Brien M, Pope A. A multicentre randomized phase III trial comparing pembrolizumab (P) vs single agent chemotherapy (CT) for advanced pre-treated malignant pleural mesothelioma (MPM): results from the European Thoracic Oncology Platform (ETOP 9-15) PROMISE-meso trial. Ann Oncol. 2019;30:v931.
    1. Marcq E, Siozopoulou V, de Waele J, Van Audenaerde J, Zwaenepoel K, Santermans E. Prognostic and predictive aspects of the tumor immune microenvironment and immune checkpoints in malignant pleural mesothelioma. OncoImmunology. 2017;6
    1. Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science. 2013;339:286–291.
    1. Minnema-Luiting J, Vroman H, Aerts J, Cornelissen R. Heterogeneity in immune cell content in malignant pleural mesothelioma. Int J Mol Sci. 2018;19
    1. Yap TA, Aerts JG, Popat S, Fennell DA. Novel insights into mesothelioma biology and implications for therapy. Nat Rev Cancer. 2017;17:475–488.
    1. Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194:950–959.
    1. Selby MJ, Engelhardt JJ, Johnston RJ, Lu LS, Han M, Thudium K. Preclinical development of ipilimumab and nivolumab combination immunotherapy: mouse tumor models, In vitro functional studies, and cynomolgus macaque toxicology. PLoS One. 2016;11
    1. Disselhorst MJ, Quispel-Janssen J, Lalezari F, Monkhorst K, de Vries JF, van der Noort V. Ipilimumab and nivolumab in the treatment of recurrent malignant pleural mesothelioma (INITIATE): results of a prospective, single-arm, phase 2 trial. Lancet Respir Med. 2019;7:260–270.
    1. Calabrò L, Morra A, Giannarelli D, Amato G, D’Incecco A, Covre A. Tremelimumab combined with durvalumab in patients with mesothelioma (NIBIT-MESO-1): an open-label, non-randomised, phase 2 study. Lancet Respir Med. 2018;6:451–460.
    1. Scherpereel A, Mazieres J, Greillier L, Lantuejoul S, Dô P, Bylicki O. Nivolumab or nivolumab plus ipilimumab in patients with relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): a multicentre, open-label, randomised, non-comparative, phase 2 trial. Lancet Oncol. 2019;20:239–253.
    1. Bratman SV, Yang SYC, Iafolla MAJ, Liu Z, Hansen AR, Bedard PL. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat Cancer. 2020
    1. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–571.
    1. Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci. 2017
    1. Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017
    1. Fundamental mechanisms of immune checkpoint blockade therapy, (2018).
    1. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang N-AAS, Andrews MC. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell. 2017;170:1120–1133. e17.
    1. Byrne MJ, Nowak AK. Modified RECIST criteria for assessment of response in malignant pleural mesothelioma. Ann Oncol. 2004;15:257–260.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing on JSTOR. J R Stat Soc Ser B. 1995;57:289–300.
    1. Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J. Distinct immune cell populations define response to anti-PD-1 monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell. 2019;35:238–255. e6.
    1. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang N-AAS, Andrews MC. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell. 2017;170:1120–1133. e17.
    1. Fairfax BP, Taylor CA, Watson RA, Nassiri I, Danielli S, Fang H. Peripheral CD8(+) T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat Med. 2020;26(2):193–199.
    1. Nakamura A, Kondo N, Nakamichi T, Kuroda A, Hashimoto M, Matsumoto S. Initial evaluation of nivolumab in patients with post-operative recurrence of malignant pleural mesothelioma. Jpn J Clin Oncol. 2020;50(8):920–925.
    1. Okada M, Kijima T, Aoe K, Kato T, Fujimoto N, Nakagawa K. Clinical efficacy and safety of nivolumab: results of a multicenter, open-label, single-arm, Japanese phase II study in malignant pleural mesothelioma (MERIT) Clin Cancer Res. 2019;25(18):5485–5492.
    1. Garg AD, Coulie PG, Van den Eynde BJ, Agostinis P. Integrating next-generation dendritic cell vaccines into the current cancer immunotherapy landscape. Trends Immunol. 2017;38(8):577–593.

Source: PubMed

3
구독하다