Avelumab in newly diagnosed glioblastoma

Francois H Jacques, Garth Nicholas, Ian A J Lorimer, Victorine Sikati Foko, Jasmine Prevost, Nathalie Dumais, Katy Milne, Brad H Nelson, John Woulfe, Gerard Jansen, B Erik Apedaile, Francois H Jacques, Garth Nicholas, Ian A J Lorimer, Victorine Sikati Foko, Jasmine Prevost, Nathalie Dumais, Katy Milne, Brad H Nelson, John Woulfe, Gerard Jansen, B Erik Apedaile

Abstract

Background: Glioblastoma (GBM) is known to use both local and systemic immunosuppressive strategies. One such strategy is the expression of the immune checkpoint protein programmed cell death ligand-1 (PD-L1) by both tumor cells and tumor-associated immune cells. Recent phase III trials using IgG4 antibodies targeting PD-1, the ligand for PD-L1, failed to show any benefit. Avelumab is an IgG1 monoclonal antibody targeting PD-L1. In contrast to the previously tested immune checkpoint inhibitors, it can directly bind tumor cells and immune cells expressing PD-L1 and can induce antibody-dependent cellular cytotoxicity.

Methods: We conducted a single center, open label, phase II study where avelumab 10 mg/kg IV Q2W was added concurrently to the first monthly temozolomide cycle in patients with newly diagnosed GBM. Immunohistochemical analyses were performed on surgery samples. The primary objective was safety. Secondary objectives were efficacy outcomes according to the immunotherapy Response Assessment in Neuro Oncology criteria, progression free survival (PFS), and overall survival (OS). Exploratory objectives aimed at determining prognostic biomarkers.

Results: Thirty patients were started on therapy and two were lost to follow-up. Median follow-up time (reverse Kaplan-Meier) was 41.7 months (IQR: 28.3-43.4). Three (10.0%) patients had a related or possibly related treatment emergent adverse event that lead to transient or permanent discontinuation of avelumab. Eight (26.7%) patients had one or more immune-related adverse events, and 8 (26.7%) patients had an infusion-related reaction. The overall response rate was 23.3%, median PFS was 9.7 months, and the median OS was 15.3 months. No pretreatment biomarkers showed any predictive value.

Conclusions: The addition of avelumab to standard therapy in patients with GBM was not associated with any new safety signal. There was no apparent improvement in OS.

Trial registration: NCT03047473 Registered February 9, 2017.

Keywords: PD-L1; avelumab; glioblastoma; immune checkpoint inhibitor; phase II.

© The Author(s) 2021. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.

Figures

Figure 1.
Figure 1.
Kaplan Meier plots for patients grouped according to steroid use. A. progression-free survival in patients who did not require the use of steroids at month 1 (red) and those who did (blue). B. Overall survival in patients who did not require the use of steroids at month 1 (red) and those who did (blue).
Figure 2.
Figure 2.
Biomarker analyses: A. Multicolour immunohistochemistry analyses of first surgery samples from trial patients. Top row of bar graphs show the results for the CD3/CD8/CD20 analysis. The bottom row of bar graphs show results for PD1/PDL1/CD68 analysis. Y axes show the number of positive cells per mm2 and x axes show results for individual patients. B. Single cell RNA-seq expression data from twenty-eight glioblastoma patients sequenced by Neftel et al. were analyzed for mRNA expression of immunohistochemistry markers used in this study, using Broad Institute Single Cell portal software https://singlecell.broadinstitute.org/single_cell. C. Comparison of immune status in first and second surgery samples from trial patients. Y axes show the number of positive cells per mm2 and x axes show paired bars for individual patient samples from first (grey bar) and second surgeries (black bar).

References

    1. Stupp R, Mason WP, van den Bent MJ, et al. ; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group . Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996.
    1. Preusser M, de Ribaupierre S, Wöhrer A, et al. . Current concepts and management of glioblastoma. Ann Neurol. 2011;70(1):9–21.
    1. Central Brain Tumor Registry of the United States. . Accessed December 8, 2020.
    1. Malmström A, Grønberg BH, Marosi C, et al. ; Nordic Clinical Brain Tumour Study Group (NCBTSG) . Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol. 2012;13(9):916–926.
    1. Hegi ME, Diserens AC, Godard S, et al. . Clinical trial substantiates the predictive value of O-6-methylguanine-DNA methyltransferase promoter methylation in glioblastoma patients treated with temozolomide. Clin Cancer Res. 2004;10(6):1871–1874.
    1. Sturm D, Witt H, Hovestadt V, et al. . Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012;22(4):425–437.
    1. Gilbert MR, Wang M, Aldape KD, et al. . Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J Clin Oncol. 2013;31(32):4085–4091.
    1. Stupp R, Hegi ME, Gorlia T, et al. ; European Organisation for Research and Treatment of Cancer (EORTC); Canadian Brain Tumor Consortium; CENTRIC Study Team . Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(10):1100–1108.
    1. Gilbert MR, Dignam JJ, Armstrong TS, et al. . A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):699–708.
    1. Reardon DA, Freeman G, Wu C, et al. . Immunotherapy advances for glioblastoma. Neuro Oncol. 2014;16(11):1441–1458.
    1. Avril T, Vauleon E, Tanguy-Royer S, Mosser J, Quillien V. Mechanisms of immunomodulation in human glioblastoma. Immunotherapy. 2011;3(4 Suppl):42–44.
    1. Qiu B, Zhang D, Wang C, et al. . IL-10 and TGF-β2 are overexpressed in tumor spheres cultured from human gliomas. Mol Biol Rep. 2011;38(5):3585–3591.
    1. Constam DB, Philipp J, Malipiero UV, ten Dijke P, Schachner M, Fontana A. Differential expression of transforming growth factor-beta 1, -beta 2, and -beta 3 by glioblastoma cells, astrocytes, and microglia. J Immunol. 1992;148(5):1404–1410.
    1. Albulescu R, Codrici E, Popescu ID, et al. . Cytokine patterns in brain tumour progression. Mediators Inflamm. 2013;2013:979748.
    1. Jacobs JF, Idema AJ, Bol KF, et al. . Regulatory T cells and the PD-L1/PD-1 pathway mediate immune suppression in malignant human brain tumors. Neuro Oncol. 2009;11(4):394–402.
    1. Wilmotte R, Burkhardt K, Kindler V, et al. . B7-homolog 1 expression by human glioma: a new mechanism of immune evasion. Neuroreport. 2005;16(10):1081–1085.
    1. Yao Y, Tao R, Wang X, Wang Y, Mao Y, Zhou LF. B7-H1 is correlated with malignancy-grade gliomas but is not expressed exclusively on tumor stem-like cells. Neuro Oncol. 2009;11(6):757–766.
    1. Reardon DA, Brandes AA, Omuro A, et al. . Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 phase 3 randomized clinical trial. JAMA Oncol. 2020;6(7):1003–1010.
    1. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, et al. . Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 2019;25(3):470–476.
    1. Cloughesy TF, Mochizuki AY, Orpilla JR, et al. . Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 2019;25(3):477–486.
    1. Wang C, Thudium KB, Han M, et al. . In vitro characterization of the anti-PD-1 antibody nivolumab, BMS-936558, and in vivo toxicology in non-human primates. Cancer Immunol Res. 2014;2(9):846–856.
    1. Hutter G, Theruvath J, Graef CM, et al. . Microglia are effector cells of CD47-SIRPα antiphagocytic axis disruption against glioblastoma. Proc Natl Acad Sci USA. 2019;116(3):997–1006.
    1. Wiesweg M. SurvivalAnalysis: High-Level Interface for Survival Analysis and Associated Plots; 2020. . Accessed December 8, 2020.
    1. R Core Team. R: A Language and Environment for Statistical Computing.Vienna, Austria: R Foundation for Statistical Computing; 2018. . Accessed December 8, 2020.
    1. Wickham H.Ggplot2: Elegant Graphics for Data Analysis. New York: Springer-Verlag; 2016.
    1. Altman DG, De Stavola BL, Love SB, Stepniewska KA. Review of survival analyses published in cancer journals. Br J Cancer. 1995;72(2):511–518.
    1. Lu S, Stein JE, Rimm DL, et al. . Comparison of biomarker modalities for predicting response to PD-1/PD-L1 checkpoint blockade: a systematic review and meta-analysis. JAMA Oncol. 2019;5(8):1195–1204.
    1. Grabowski MM, Sankey EW, Ryan KJ, et al. . Immune suppression in gliomas. J Neurooncol. 2021;151(1):3–12.
    1. Neftel C, Laffy J, Filbin MG, et al. . An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell. 2019;178(4):835–849.e21.
    1. Engler JR, Robinson AE, Smirnov I, et al. . Increased microglia/macrophage gene expression in a subset of adult and pediatric astrocytomas. PLoS One. 2012;7(8):e43339.
    1. Berghoff AS, Kiesel B, Widhalm G, et al. . Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. 2015;17(8):1064–1075.
    1. Nduom EK, Wei J, Yaghi NK, et al. . PD-L1 expression and prognostic impact in glioblastoma. Neuro Oncol. 2016;18(2):195–205.
    1. Zhao J, Chen AX, Gartrell RD, et al. . Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med. 2019;25(3):462–469.
    1. Heery CR, O’Sullivan-Coyne G, Madan RA, et al. . Avelumab for metastatic or locally advanced previously treated solid tumours (JAVELIN Solid Tumor): a phase 1a, multicohort, dose-escalation trial. Lancet Oncol. 2017;18(5):587–598.
    1. Marciscano AE, Gulley JL. Avelumab demonstrates promise in advanced NSCLC. Oncotarget. 2017;8(61):102767–102768.
    1. Lynes JP, Nwankwo AK, Sur HP, et al. . Biomarkers for immunotherapy for treatment of glioblastoma. J Immunother Cancer. 2020;8(1): e000348.
    1. Hepner A, Porter J, Hare F, et al. . Serum neurofilament light, glial fibrillary acidic protein and tau are possible serum biomarkers for activity of brain metastases and gliomas. World J Oncol. 2019;10(4–5):169–175.
    1. Zhang M, Hutter G, Kahn SA, et al. . Anti-CD47 treatment stimulates phagocytosis of glioblastoma by M1 and M2 polarized macrophages and promotes M1 polarized macrophages in vivo. PLoS One. 2016;11(4):e0153550.
    1. Jaiswal S, Jamieson CH, Pang WW, et al. . CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138(2):271–285.
    1. Weiskopf K. Cancer immunotherapy targeting the CD47/SIRPα axis. Eur J Cancer. 2017;76:100–109.
    1. Hardcastle J, Mills L, Malo CS, et al. . Immunovirotherapy with measles virus strains in combination with anti-PD-1 antibody blockade enhances antitumor activity in glioblastoma treatment. Neuro Oncol. 2017;19(4):493–502.
    1. Sharma P, Allison JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol. 2020;20(2):75–76.

Source: PubMed

3
Sottoscrivi