Nivolumab and sunitinib combination in advanced soft tissue sarcomas: a multicenter, single-arm, phase Ib/II trial

Javier Martin-Broto, Nadia Hindi, Giovanni Grignani, Javier Martinez-Trufero, Andres Redondo, Claudia Valverde, Silvia Stacchiotti, Antonio Lopez-Pousa, Lorenzo D'Ambrosio, Antonio Gutierrez, Herminia Perez-Vega, Victor Encinas-Tobajas, Enrique de Alava, Paola Collini, Maria Peña-Chilet, Joaquin Dopazo, Irene Carrasco-Garcia, Maria Lopez-Alvarez, David S Moura, Jose A Lopez-Martin, Javier Martin-Broto, Nadia Hindi, Giovanni Grignani, Javier Martinez-Trufero, Andres Redondo, Claudia Valverde, Silvia Stacchiotti, Antonio Lopez-Pousa, Lorenzo D'Ambrosio, Antonio Gutierrez, Herminia Perez-Vega, Victor Encinas-Tobajas, Enrique de Alava, Paola Collini, Maria Peña-Chilet, Joaquin Dopazo, Irene Carrasco-Garcia, Maria Lopez-Alvarez, David S Moura, Jose A Lopez-Martin

Abstract

Background: Sarcomas exhibit low expression of factors related to immune response, which could explain the modest activity of PD-1 inhibitors. A potential strategy to convert a cold into an inflamed microenvironment lies on a combination therapy. As tumor angiogenesis promotes immunosuppression, we designed a phase Ib/II trial to test the double inhibition of angiogenesis (sunitinib) and PD-1/PD-L1 axis (nivolumab).

Methods: This single-arm, phase Ib/II trial enrolled adult patients with selected subtypes of sarcoma. Phase Ib established two dose levels: level 0 with sunitinib 37.5 mg daily from day 1, plus nivolumab 3 mg/kg intravenously on day 15, and then every 2 weeks; and level -1 with sunitinib 37.5 mg on the first 14 days (induction) and then 25 mg per day plus nivolumab on the same schedule. The primary endpoint was to determine the recommended dose for phase II (phase I) and the 6-month progression-free survival rate, according to Response Evaluation Criteria in Solid Tumors 1.1 (phase II).

Results: From May 2017 to April 2019, 68 patients were enrolled: 16 in phase Ib and 52 in phase II. The recommended dose of sunitinib for phase II was 37.5 mg as induction and then 25 mg in combination with nivolumab. After a median follow-up of 17 months (4-26), the 6-month progression-free survival rate was 48% (95% CI 41% to 55%). The most common grade 3-4 adverse events included transaminitis (17.3%) and neutropenia (11.5%).

Conclusions: Sunitinib plus nivolumab is an active scheme with manageable toxicity in the treatment of selected patients with advanced soft tissue sarcoma, with almost half of patients free of progression at 6 months.Trial registration number NCT03277924.

Keywords: clinical trials as topic; immunotherapy; sarcoma; translational medical research.

Conflict of interest statement

Competing interests: JM-B reports research grants from PharmaMar, Eisai, Immix BioPharma and Novartis, outside the submitted work; honoraria for advisory board participation and expert testimony from PharmaMar, Eli Lilly and Company, Bayer and Eisai; and research funding for clinical studies (institutional) from PharmaMar, Eli Lilly and Company, AROG, Bayer, Eisai, Lixte, Karyopharm, Deciphera, GSK, Novartis, Blueprint, Nektar, Forma, Amgen and Daiichi Sankyo. NH reports grants, personal fees and non-financial support from PharmaMar, personal fees from Lilly, and grants from Eisai and Novartis, outside the submitted work, and research funding for clinical studies (institutional) from PharmaMar, Eli Lilly and Company, AROG, Bayer, Eisai, Lixte, Karyopharm, Deciphera, GSK, Novartis, Blueprint, Nektar, Forma, Amgen and Daiichi Sankyo. GG reports grants and personal fees from PharmaMar, grants from Novartis, and personal fees from Lilly, Pfizer, Bayer and Eisai, outside the submitted work. AR reports grants and personal fees from PharmaMar, personal fees from Lilly, Novartis, Amgen, AstraZeneca and Tesaro, grants and personal fees from Roche, and grants from Eisai, outside the submitted work. SS reports grants and personal fees from Bayer, Lilly and PharmaMar, and grants from GlaxoSmithKline, Novartis and Pfizer, outside the submitted work. EdA reports personal fees and non-financial support from Roche, BMS and PharmaMar, and personal fees from Bayer, outside the submitted work. ML-A declares institutional research grants from PharmaMar, Eisai, Immix BioPharma and Novartis, outside the submitted work. DSM reports institutional research grants from PharmaMar, Eisai, Immix BioPharma and Novartis, outside the submitted work, and travel support from PharmaMar, Eisai, Celgene, Bayer and Pfizer. JAL-M reports honoraria for advisory board participation and travel support from PharmaMar, Eli Lilly and Company, Bayer, Eisai, Novartis, BMS, MSD, Roche, Celgene, Pierre Fabre, Pfizer, GSK, Daiichi Sankyo, Amgen, and Chobani. All other authors declare no relevant relationship to disclose.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
CONSORT diagram. CONSORT, Consolidated Standards of Reporting Trials; ECOG, Eastern Cooperative Oncology Group; RECIST, Response Evaluation Criteria in Solid Tumors.
Figure 2
Figure 2
Response to treatment by patient according to RECIST. All evaluable patients (N=60) from phase Ib (n=14) and phase II (n=46) are shown. Tumor diameter was measured in millimeters. The dashed lines represent 20% increase in diameter and 30% decrease in diameter (RECIST progression and response cut-offs, respectively). *No size variation. RECIST, Response Evaluation Criteria in Solid Tumors.
Figure 3
Figure 3
Progression-free survival to treatment by patient, based on RECIST central radiological assessment. Each patient in the efficacy population in phase II is represented as bars (n=49). The vertical dashed line represents the median progression-free survival (5.6 months). The stars represent patients achieving RECIST objective responses. The arrows represent patients non-progressing in the last central radiological assessment. RECIST, Response Evaluation Criteria in Solid Tumors.

References

    1. Ryan CW, Merimsky O, Agulnik M, et al. . PICASSO III: a phase III, placebo-controlled study of doxorubicin with or without Palifosfamide in patients with metastatic soft tissue sarcoma. J Clin Oncol 2016;34:3898–905. 10.1200/JCO.2016.67.6684
    1. Tap WD, Papai Z, Van Tine BA, et al. . Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (th CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2017;18:1089–103. 10.1016/S1470-2045(17)30381-9
    1. Martin-Broto J, Pousa AL, de Las Peñas R, et al. . Randomized phase II study of trabectedin and doxorubicin compared with doxorubicin alone as first-line treatment in patients with advanced soft tissue sarcomas: a Spanish group for research on sarcoma study. J Clin Oncol 2016;34:2294–302. 10.1200/JCO.2015.65.3329
    1. Iglesia MD, Parker JS, Hoadley KA, et al. . Genomic analysis of immune cell infiltrates across 11 tumor types. J Natl Cancer Inst 2016;108:djw144. 10.1093/jnci/djw144
    1. Robert C, Long GV, Brady B, et al. . Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320–30. 10.1056/NEJMoa1412082
    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;9:34. 10.1186/s13073-017-0424-2
    1. Balch CM, Riley LB, Bae YJ, et al. . Patterns of human tumor-infiltrating lymphocytes in 120 human cancers. Arch Surg 1990;125:200–5. 10.1001/archsurg.1990.01410140078012
    1. Inaguma S, Wang Z, Lasota J, et al. . Comprehensive immunohistochemical study of programmed cell death ligand 1 (PD-L1): analysis in 5536 cases revealed consistent expression in trophoblastic tumors. Am J Surg Pathol 2016;40:1133–42. 10.1097/PAS.0000000000000653
    1. Pollack SM, He Q, Yearley JH, et al. . T-cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death-ligand 1 expression in patients with soft tissue sarcomas. Cancer 2017;123:3291–304. 10.1002/cncr.30726
    1. D'Angelo SP, Shoushtari AN, Agaram NP, et al. . Prevalence of tumor-infiltrating lymphocytes and PD-L1 expression in the soft tissue sarcoma microenvironment. Hum Pathol 2015;46:357–65. 10.1016/j.humpath.2014.11.001
    1. Torabi A, Amaya CN, Wians FH, et al. . Pd-1 and PD-L1 expression in bone and soft tissue sarcomas. Pathology 2017;49:506–13. 10.1016/j.pathol.2017.05.003
    1. Oike N, Kawashima H, Ogose A, et al. . Prognostic impact of the tumor immune microenvironment in synovial sarcoma. Cancer Sci 2018;109:3043–54. 10.1111/cas.13769
    1. Park HK, Kim M, Sung M, et al. . Status of programmed death-ligand 1 expression in sarcomas. J Transl Med 2018;16:303. 10.1186/s12967-018-1658-5
    1. Bertucci F, Finetti P, Perrot D, et al. . PDL1 expression is a poor-prognosis factor in soft-tissue sarcomas. Oncoimmunology 2017;6:e1278100. 10.1080/2162402X.2016.1278100
    1. Becht E, Giraldo NA, Lacroix L, et al. . Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol 2016;17:218. 10.1186/s13059-016-1070-5
    1. Petitprez F, de Reyniès A, Keung EZ, et al. . B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020;577:556–60. 10.1038/s41586-019-1906-8
    1. D'Angelo SP, Mahoney MR, Van Tine BA, et al. . Nivolumab with or without ipilimumab treatment for metastatic sarcoma (alliance A091401): two open-label, non-comparative, randomised, phase 2 trials. Lancet Oncol 2018;19:416–26. 10.1016/S1470-2045(18)30006-8
    1. Ben-Ami E, Barysauskas CM, Solomon S, et al. . Immunotherapy with single agent nivolumab for advanced leiomyosarcoma of the uterus: results of a phase 2 study. Cancer 2017;123:3285–90. 10.1002/cncr.30738
    1. Tawbi HA, Burgess M, Bolejack V, et al. . Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol 2017;18:1493–501. 10.1016/S1470-2045(17)30624-1
    1. Motz GT, Coukos G. The parallel lives of angiogenesis and immunosuppression: cancer and other tales. Nat Rev Immunol 2011;11:702–11. 10.1038/nri3064
    1. Brookmeyer R, Crowley J. A confidence interval for the median survival time. Biometrics 1982;38:29–41. 10.2307/2530286
    1. Van Glabbeke M, Verweij J, Judson I, et al. . Progression-free rate as the principal end-point for phase II trials in soft-tissue sarcomas. Eur J Cancer 2002;38:543–9. 10.1016/S0959-8049(01)00398-7
    1. van Erp NP, Mathijssen RHJ, van der Veldt AA, et al. . Myelosuppression by sunitinib is flt-3 genotype dependent. Br J Cancer 2010;103:757–8. 10.1038/sj.bjc.6605813
    1. George S, Merriam P, Maki RG, et al. . Multicenter phase II trial of sunitinib in the treatment of nongastrointestinal stromal tumor sarcomas. J Clin Oncol 2009;27:3154–60. 10.1200/JCO.2008.20.9890
    1. van der Graaf WTA, Blay J-Y, Chawla SP, et al. . Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2012;379:1879–86. 10.1016/S0140-6736(12)60651-5
    1. Schöffski P, Chawla S, Maki RG, et al. . Eribulin versus dacarbazine in previously treated patients with advanced liposarcoma or leiomyosarcoma: a randomised, open-label, multicentre, phase 3 trial. Lancet 2016;387:1629–37. 10.1016/S0140-6736(15)01283-0
    1. Demetri GD, von Mehren M, Jones RL, et al. . Efficacy and safety of trabectedin or dacarbazine for metastatic liposarcoma or leiomyosarcoma after failure of conventional chemotherapy: results of a phase III randomized multicenter clinical trial. J Clin Oncol 2016;34:786–93. 10.1200/JCO.2015.62.4734
    1. Penel N, Demetri GD, Blay JY, et al. . Growth modulation index as metric of clinical benefit assessment among advanced soft tissue sarcoma patients receiving trabectedin as a salvage therapy. Ann Oncol 2013;24:537–42. 10.1093/annonc/mds470
    1. Judson I, Morden JP, Kilburn L, et al. . Cediranib in patients with alveolar soft-part sarcoma (CASPS): a double-blind, placebo-controlled, randomised, phase 2 trial. Lancet Oncol 2019;20:1023–34. 10.1016/S1470-2045(19)30215-3
    1. Dancsok AR, Setsu N, Gao D, et al. . Expression of lymphocyte immunoregulatory biomarkers in bone and soft-tissue sarcomas. Mod Pathol 2019;32:1772–85. 10.1038/s41379-019-0312-y
    1. Lewin J, Davidson S, Anderson ND, et al. . Response to immune checkpoint inhibition in two patients with alveolar soft-part sarcoma. Cancer Immunol Res 2018;6:1001–7. 10.1158/2326-6066.CIR-18-0037
    1. Leone RD, Zhao L, Englert JM, et al. . Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 2019;366:1013–21. 10.1126/science.aav2588
    1. DeBerardinis RJ. Tumor microenvironment, metabolism, and immunotherapy. N Engl J Med 2020;382:869–71. 10.1056/NEJMcibr1914890
    1. Neubert NJ, Schmittnaegel M, Bordry N, et al. . T cell-induced CSF1 promotes melanoma resistance to PD1 blockade. Sci Transl Med 2018;10:eaan3311. 10.1126/scitranslmed.aan3311
    1. Zhu Y, Yang J, Xu D, et al. . Disruption of tumour-associated macrophage trafficking by the osteopontin-induced colony-stimulating factor-1 signalling sensitises hepatocellular carcinoma to anti-PD-L1 blockade. Gut 2019;68:1653–66. 10.1136/gutjnl-2019-318419

Source: PubMed

3
Subskrybuj