We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Predictive markers of immune response in glioblastoma: hopes and facts

    Vincenzo Di Nunno

    *Author for correspondence: Tel.: +39 051 622 5101; Fax: +39 051 622 5057;

    E-mail Address: dinunnovincenzo88@gmail.com

    Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy

    ,
    Enrico Franceschi

    Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy

    ,
    Lidia Gatto

    Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy

    ,
    Stefania Bartolini

    Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy

    &
    Alba Ariela Brandes

    Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy

    Published Online:https://doi.org/10.2217/fon-2020-0047

    Immune-checkpoint inhibitors (ICI) represent a concrete hope for patients with advanced solid tumors. Indeed, patients responding to these agents may experience a long-lasting response. Recently, results of interventional clinical trials investigated the role of ICIs in patients with glioblastoma. Results of these studies suggested that only a small percentage of these patients could benefit from these agents. Research of predictive markers assumes a critical importance to adequately select patients likely to benefit from ICIs. Molecular and clinical variables associated to tumors and patients have been evaluated as potential predictive markers. Main aim of the current work is to summarize and critically evaluate current knowledge in this field.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Stupp R, Mason WP, van den Bent MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for GBM. N. Engl. J. Med. 352(10), 987–996 (2005). •• Randomized clinical trial evaluates concomitant and adjuvant temozolomide in patients with glioblastoma (GBM). Standard of care.
    • 2. Stupp R, Hegi ME, Mason WP et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in GBM in a randomised Phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10(5), 459–466 (2009). •• Randomized clinical trial evaluates concomitant and adjuvant temozolomide in patients with GBM. Standard of care. Long-term follow-up.
    • 3. Tosoni A, Franceschi E, Poggi R, Brandes AA. Relapsed GBM: treatment strategies for initial and subsequent recurrences. Curr. Treat. Options Oncol. 17(9), 49 (2016).
    • 4. Franceschi E, Lamberti G, Paccapelo A et al. Third-line therapy in recurrent GBM: is it another chance for bevacizumab? J. Neurooncol. 139(2), 383–388 (2018).
    • 5. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 348(6230), 56–61 (2015).
    • 6. Omuro A, Vlahovic G, Lim M et al. Nivolumab with or without ipilimumab in patients with recurrent GBM: results from exploratory Phase I cohorts of CheckMate 143. Neuro. Oncol. 20(5), 674–686 (2018). • A Phase I study evaluates combination between nivolumab and ipilimumab or nivolumab alone in patients with GBM.
    • 7. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R et al. Neoadjuvant nivolumab modifies the tumour immune microenvironment in resectable GBM. Nat. Med. 25(3), 470–476 (2019).
    • 8. Reardon DA, Omuro A, Brandes AA et al. Randomized Phase III study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent GBM: checkMate 143. Neuro Oncol. 19(3.1), iii21 (2017). • Preliminary results of a randomized Phase III trial evaluating the PD1 inhibitor nivolumab in patients with GBM.
    • 9. Lukas RV, Rodon J, Becker K et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J. Neurooncol. 140(2), 317–328 (2018).
    • 10. Anna C Filley, Mario Henriquez, Mahua Dey. Recurrent glioma clinical trial, CheckMate-143: the game is not over yet. Oncotarget 8(53), 91779–91794 (2017).
    • 11. Janning M, Kobus F, Babayan A et al. Determination of PD-L1 expression in circulating tumour cells of NSCLC patients and correlation with response to PD-1/PD-L1 inhibitors. Cancers (Basel) 11(6), E835 (2019).
    • 12. Kim KJ, Yang HK, Kim WH, Kang GH. Combined prognostic effect of PD-L1 expression and immunoscore in microsatellite-unstable advanced gastric cancers. Oncotarget 8(35), 58887–58902 (2017).
    • 13. Arora S, Velichinskii R, Lesh RW et al. Existing and emerging biomarkers for immune checkpoint immunotherapy in solid tumours. Adv. Ther. 36(10), 2638–2678 (2019).
    • 14. Reck M, Rodríguez-Abreu D, Robinson AG et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 375(19), 1823–1833 (2016).
    • 15. Ozaki Y, Muto S, Takagi H et al. Tumour mutation burden and immunological, genomic, and clinicopathological factors as biomarkers for checkpoint inhibitor treatment of patients with non-small-cell lung cancer. Cancer Immunol. Immunother. 69(1), 127–134 (2020).
    • 16. Goodman AM, Kato S, Bazhenova L et al. Tumour mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther. 16(11), 2598–2608 (2017).
    • 17. Chan TA, Yarchoan M, Jaffee E et al. Development of tumour mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann. Oncol. 30(1), 44–56 (2019).
    • 18. Park J, Kwon M, Kim KH et al. Immune checkpoint inhibitor-induced reinvigoration of tumour-infiltrating CD8(+) T cells is determined by their differentiation status in GBM. Clin Cancer Res. 25(8), 2549–2559 (2019).
    • 19. Louveau A, Smirnov I, Keyes TJ et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523(7560), 337–341 (2015).
    • 20. Aspelund A, Antila S, Proulx ST et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 212(7), 991–999 (2015).
    • 21. De Simone R, Giampaolo A, Giometto B et al. The costimulatory molecule B7 is expressed on human microglia in culture and in multiple sclerosis acute lesions. J. Neuropathol. Exp. Neurol. 54(2), 175–187 (1995).
    • 22. Aloisi F, Ria F, Penna G, Adorini L. Microglia are more efficient than astrocytes in antigen processing and in Th1 but not Th2 cell activation. J. Immunol. 160(10), 4671–4680 (1998).
    • 23. Carson MJ, Sutcliffe JG, Campbell IL. Microglia stimulate naive T-cell differentiation without stimulating T-cell proliferation. J. Neurosci. Res. 55(1), 127–134 (1999).
    • 24. Perng P, Lim M. Immunosuppressive mechanisms of malignant gliomas: parallels at non-CNS sites. Front. Oncol. 5, 153 (2015).
    • 25. Vitkovic L, Maeda S, Sternberg E. Anti- inflammatory cytokines: expression and action in the brain. Neuroimmunomodulation 9, 295–312 (2001).
    • 26. Gong D, Shi W, Yi SJ, Chen H, Groffen J, Heisterkamp N. TGFbeta signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol. 13, 31 (2012).
    • 27. Wainwright DA, Balyasnikova IV, Chang AL et al. IDO expression in brain tumours increases the recruitment of regulatory T cells and negatively impacts survival. Clin. Cancer Res. 18, 6110–6121 (2012).
    • 28. Uyttenhove C, Pilotte L, Théate I et al. Evidence for a tumoural immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 9, 1269–1274 (2003).
    • 29. Zhang I, Alizadeh D, Liang J et al. Characterization of arginase expression in glioma-associated microglia and macrophages. PLoS ONE 11, e0165118 (2016).
    • 30. Hawkins RA, O'Kane RL, Simpson IA, Vina JR. Structure of the blood–brain barrier and its role in the transport of amino acids. J. Nutr. 136, S218–S226 (2006).
    • 31. Okada H, Weller M, Huang R et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 16(15), e534–e542 (2015).
    • 32. Shi Y. Regulatory mechanisms of PD-L1 expression in cancer cells. Cancer Immunol. Immunother. 67(10), 1481–1489 (2018).
    • 33. Bristol-Myers Squibb (2019). https://news.bms.com/press-release/corporatefinancial-news/bristol-myers-squibb-provides-update-phase-3-opdivo-nivolumab-
    • 34. Bristol-Myers Squibb (2019). https://news.bms.com/press-release/corporatefinancial-news/bristol-myers-squibb-announces-phase-3-checkmate-498-study-did
    • 35. Wei R, Guo L, Wang Q, Miao J, Kwok HF, Lin Y. Targeting PD-L1 protein: translation, modification and transport. Curr. Protein Pept. Sci. 20(1), 82–91 (2019).
    • 36. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol. Cancer Ther. 14(4), 847–856 (2015).
    • 37. Nduom EK, Wei J, Yaghi NK et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro Oncol. 18(2), 195–205 (2016).
    • 38. Chen RQ, Liu F, Qiu XY, Chen XQ. The prognostic and therapeutic value of PD-L1 in glioma. Front. Pharmacol. 9, 1503 (2019).
    • 39. Garber ST, Hashimoto Y, Weathers SP et al. Immune checkpoint blockade as a potential therapeutic target: surveying CNS malignancies. Neuro Oncol. 18, 1357–1366 (2016).
    • 40. Hodges TR, Ott M, Xiu J et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro Oncol. 19, 1047–1057 (2017).
    • 41. Berghoff AS, Kiesel B, Widhalm G et al. Correlation of immune phenotype with IDH mutation in diffuse glioma. Neuro Oncol. 19, 1460–1468 (2017).
    • 42. Weller M, Reardon D, Brandes AA et al. Nivolumab vs bevacizumab in patients with recurrent glioblastoma: exploratory analysis of MGMT methylation status and baseline corticosteroid use. Neuro Oncol. 21(S6), vi12 (2019). • Subgroups analyses of a randomized Phase III trial evaluating nivolumab or bevacizumab in recurrent GBM.
    • 43. Hellmann MD, Ciuleanu TE, Pluzanski A et al. Nivolumab plus ipilimumab in lung cancer with a high tumour mutational burden. N. Engl. J. Med. 378(22), 2093–2104 (2018).
    • 44. Goodman AM, Kato S, Bazhenova L et al. Tumour mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther. 16(11), 2598–2608 (2017).
    • 45. Labriola M, Zhu J, Gupta R et al. Characterization of tumour mutational burden (TMB), PD-L1, and DNA repair genes to assess correlation with immune checkpoint inhibitors (ICIs) response in metastatic renal cell carcinoma (mRCC). J. Clin. Oncol. 37(S7), 589–589 (2019).
    • 46. Zehir A, Benayed R, Shah RH et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23(6), 703–713 (2017).
    • 47. McDonald KA, Kawaguchi T, Qi Q et al. Tumour heterogeneity correlates with less immune response and worse survival in breast cancer patients. Ann. Surg. Oncol. 26(7), 2191–2199 (2019).
    • 48. Johanns TM, Miller CA, Dorward IG et al. Immunogenomics of hypermutated glioblastoma: a patient with germline POLE deficiency treated with checkpoint blockade immunotherapy. Cancer Discov. 6(11), 1230–1236 (2016).
    • 49. Snyder A, Wolchok JD. Successful treatment of a patient with glioblastoma and a germline POLE mutation: where next? Cancer Discov. 6(11), 1210–1211 (2016).
    • 50. Chamberlain MC, Kim BT. Nivolumab for patients with recurrent glioblastoma progressing on bevacizumab: a retrospective case series. J. Neurooncol. 133(3), 561–569 (2017).
    • 51. Ranjan S, Quezado M, Garren N et al. Clinical decision making in the era of immunotherapy for high grade-glioma: report of four cases. BMC Cancer 18(1), 239 (2018).
    • 52. AlHarbi M, Ali Mobark N, AlMubarak L et al. Durable response to nivolumab in a pediatric patient with refractory glioblastoma and constitutional biallelic mismatch repair deficiency. Oncologist 23(12), 1401–1406 (2018).
    • 53. Kurz SC, Cabrera LP, Hastie D et al. PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology 91(14), e1355–e1359 (2018).
    • 54. Draaisma K, Wijnenga MM, Weenink B et al. PI3 kinase mutations and mutational load as poor prognostic markers in diffuse glioma patients. Acta Neuropathol. Commun. 3, 88 (2015).
    • 55. Hodges TR, Ott M, Xiu J et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro Oncol. 19(8), 1047–1057 (2017). • Review evaluates current knowledge about immunotherapy in patients with glioma.
    • 56. Le DT, Uram JN, Wang H et al. PD-1 blockade in tumours with mismatch-repair deficiency. N. Engl. J. Med. 372(26), 2509–2520 (2015).
    • 57. Le DT, Durham JN, Smith KN et al. Mismatch repair deficiency predicts response of solid tumours to PD-1 blockade. Science 357(6349), 409–413 (2017).
    • 58. Marabelle A, Le DT, Ascierto PA et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the Phase II KEYNOTE-158 study. J. Clin. Oncol. 38(1), 1–10 (2020).
    • 59. Gorlia T, Stupp R, Brandes AA et al. New prognostic factors and calculators for outcome prediction in patients with recurrent glioblastoma: a pooled analysis of EORTC Brain Tumour Group Phase I and II clinical trials. Eur. J. Cancer 48(8), 1176–1184 (2012).
    • 60. Franceschi E, Tosoni A, Minichillo S et al. PERNO study group. the prognostic roles of gender and O6-Methylguanine-DNA methyltransferase methylation status in glioblastoma patients: the female power. World Neurosurg. 112, e342–e347 (2018).
    • 61. Brandes AA, Franceschi E, Paccapelo A et al. Role of MGMT methylation status at time of diagnosis and recurrence for patients with glioblastoma: clinical implications. Oncologist 22(4), 432–437 (2017).
    • 62. Khan I, Waqas M, Shamim MS. Prognostic significance of IDH 1 mutation in patients with glioblastoma multiforme. J. Pak. Med. Assoc. 67(5), 816–817 (2017).
    • 63. Bady P, Kurscheid S, Delorenzi M et al. The DNA methylome of DDR genes and benefit from RT or TMZ in IDH mutant low-grade glioma treated in EORTC 22033. Acta Neuropathol. 135(4), 601–615 (2018).
    • 64. de Groot J, Penas-Prado M, Alfaro-Munoz KD et al. Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages. Neuro Oncol. pii:noz185 (2019).
    • 65. Pombo Antunes AR, Scheyltjens I, Duerinck J et al. Understanding the glioblastoma immune microenvironment as basis for the development of new immunotherapeutic strategies. Elife 9, pii: e52176 (2020).
    • 66. Hsu SPC, Chen YC, Chiang HC et al. Rapamycin and hydroxychloroquine combination alters macrophage polarization and sensitizes glioblastoma to immune checkpoint inhibitors. J. Neurooncol. 146(3), 417–426 (2020).
    • 67. Duan S, Yuan G, Liu X et al. PTEN deficiency reprogrammes human neural stem cells towards a glioblastoma stem cell-like phenotype. Nat. Commun. 6, 10068 (2015).
    • 68. Zhao J, Chen AX, Gartrell RD et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat. Med. 25(3), 462–469 (2019).