3,395
Views
43
CrossRef citations to date
0
Altmetric
Review

PD-1/PD-L1 immune checkpoint inhibitors in glioblastoma: clinical studies, challenges and potential

, &
Pages 546-553 | Received 25 Feb 2020, Accepted 10 Jun 2020, Published online: 09 Jul 2020

ABSTRACT

Immune checkpoint inhibitors (CIs) have changed the landscape of tumor immunotherapy. Glioblastoma (GBM) remains the most common primary malignant brain tumor in adults and has a very poor prognosis. Due to the high invasiveness and aggressiveness of GBM, there is considerable interest in programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) treatment. However, the immunosuppressive and immune-privileged characteristics of GBM limit the efficacy of CIs. While clinical studies of CI monotherapies have shown unsatisfactory survival benefits, new treatment strategies have received attention. Multiple clinical studies have focused on combination of standard therapy (temozolomide, radiotherapy), targeted therapy and other immunotherapies, and some have reported results. Here, we reviewed recent clinical trials of anti-PD-1/PD-L1 monotherapy, studies with neoadjuvant strategies, and preclinical and clinical studies of combination immunotherapies for GBM. The preliminary clinical reports in certain subsets of patients with hypermutated or mismatch repair system deficiency GBM are also discussed.

Introduction

Glioblastoma (GBM) is the most common primary brain tumor in adults and exhibits high aggressiveness, with a median overall survival (OS) time of less than 2 years and a 5-year OS rate of less than 10%Citation1. GBM represents one of the greatest therapeutic challenges of the modern era, with a low cure rate, high recurrence rate, and limited survival time. Therapeutic options are extremely limited at both the time of new diagnosis and relapse.

Studies on GBM therapies have been performed, and immune checkpoint inhibitors (CIs) have received considerable attention. CIs activate the anti-tumor response by inhibiting negative immune regulatory pathways, providing new therapeutic approaches for GBM.Citation2 The main category of CIs is monoclonal antibodies that target the programmed cell death protein-1 (PD-1) or its ligand (PD-L1); this category includes pembrolizumab, nivolumab, durvalumab, and atezolizumab. These anti-PD-1/PD-L1 CIs have shown significant efficacy for some tumors (such as melanoma,Citation3-5 non-small cell lung cancer (NSCLC),Citation6,Citation7 and small cell lung cancer (SCLC)Citation8) in clinical trials and have been approved for multiple tumors. For GBM, although preclinical studies have shown some potential for treatment, anti-PD-1/PD-L1 monotherapy has shown few satisfactory results in clinical studies. Most trials revealed a low tumor response, and the treatment did not prolong patient survival, which limited its therapeutic application. We summarized the existing clinical studies of anti-PD-1/PD-L1 antibodies and explored the directions for CI treatment, including the treatment of specific hypermutated tumors, neoadjuvant therapy, and combination therapies, to investigate the potential of anti-PD-1/PD-L1 treatment for GBM.

Anti-PD-1/PD-L1 monotherapy

Most clinical studies of GBM anti-PD-1/PD-L1 monotherapy have shown limited efficacy. showed identified clinical trials of anti PD-1/PD-L1 monotherapies. Pembrolizumab, which is FDA approved for NSCLC, melanoma, hepatocellular carcinoma and several other cancers, did not prolong the survival of patients with GBM when used as a single agent. The Keynote-028Citation9 clinical trial investigated pembrolizumab monotherapy in 26 patients with recurrent GBM, and reported limited survival benefits: median stable disease (SD) of 39.4 weeks (7.1–85.9 weeks), median progression free survival (PFS) of 2.8 months (1.9–9.1 months), and median OS of 14.4 months (10.3-not reached). Several studies of high grade gliomas (WHO grade 3 and 4) also showed that pembrolizumab monotherapy has limited efficacy compared with control groups.Citation10,Citation11 Nivolumab is another approved, widely used CI. Checkmate 143, the first large-scale phase 3 clinical trial involving CI treatment of GBM, initially evaluated ipilimumab + nivolumab. However, the major results were regarding nivolumab monotherapy due to increased adverse effects with ipilimumab.Citation12 Compared with the bevacizumab-treated control group, nivolumab did not significantly improve survival; the 12-month OS was 42% in both arms, while the median OS was 9.8 months in the nivolumab group and 10.0 months in the bevacizumab group; the median PFS was 1.5 months and 3.5 months in the nivolumab and bevacizumab groups, respectively. The anti-PD-L1 antibodiesCitation13 atezolizumabCitation8 and durvalumabCitation14 have been approved for several cancers. A phase 1 clinical trial of atezolizumab monotherapy involving 16 patients with recurrent GBMCitation15 demonstrated unextended survival, except for 3 patients with IDH or POLE mutations who survived longer than 16 months. A phase 2 trialCitation16,Citation17 evaluating durvalumab in 5 GBM cohorts published preliminary results. Data from subgroups indicated partially clinical benefits. In the arm involving 30 patients with recurrent GBM who received durvalumab monotherapy, the overall disease control rate was 60.0% (4 with a partial response, 14 with SD), the median OS was 28.9 weeks (22.9-not reached), and the median PFS was 13.9 weeks (8.1–24.0 weeks). In conclusion, the efficacy of CI monotherapy in treating GBM is not satisfactory. It is necessary to further analyze the underlying reasons and develop new treatment strategies.

Table 1. Clinical trials investigating PD-1/PD-L1 immune checkpoint blockade with glioblastoma

The efficacy of CIs may be affected by several factors, including tumor cell PD-L1 expression,Citation18 tumor-infiltrating lymphocytes (TILs),Citation19 tumor-infiltrating myeloid cells (TIMs),Citation20 tumor mutation burden (TMB), new antigen burden, microsatellite instability (MSI), mismatch repair (MMR) system status, and POLE mutation status.Citation21 As high PD-1 expression may be correlated with a worse prognosis in patients with GBM,Citation22 the results that 88% of patients with newly diagnosed GBM and 72.2% of patients with recurrent GBM have high PD-1 expressionCitation18 may be a challenge for further studies. In addition, since the existence of TILs is the basis of its effectiveness, the ‘cold’ microenvironment and the depletion of TILs in GBM also causes challenges for efficacy.Citation19

Patients with specific GBM subtypes may benefit more from CI therapies. A higher TMB, higher MSI, neoantigen, MMR system deficiency (MMRD), and germline POLE mutation usually suggest better efficacy, which has been confirmed in clinical practice.Citation23-26 In patients with melanoma,Citation27 NSCLC,Citation28 colorectal cancer,Citation29 and several other tumors,Citation30 hypermutated subtypes are often associated with better CI efficacy. In one case report about a GBM patient with hypermutation and POLE germline mutation,Citation23 radiographic response and increased lymphocyte infiltration were observed, suggesting preliminary effectiveness. However, the definition and classification of hypermutation are inconclusive. The determination of hypermutation is mainly based on TMB, which quantitatively reflects the quantity of tumor mutations. Different thresholds have been reported for pan-cancer analysis, and there is no consensus yet for the critical definition of ‘hypermutation’.Citation31,Citation32 For GBM, several different important criteria were used in published studies. Hodges et al. used TMB> 20 mutations/1.4 Mb as the criterion for glioma hypermutation.Citation33 Johnson et al. chose TMB> 20 mutations/Mb in pediatric brain gliomas.Citation34 Campbell et al. classified tumors with TMB> 9 mutations/Mb as hypermutated and tumors with TMB> 100 mutations/Mb as ultra-high mutated in GBM.Citation24,Citation31 Bouffet et al. defined hypermutation as >100 mutations per exome in GBM.Citation26 In follow-up studies, the appropriate definition of ‘hyper’ should be further classified and unified.

Another type of GBM associated with better CI efficacy is MMRD. The four MMR genes (PMS2, MLH1, MSH2, and MSH6) are responsible for correcting DNA mismatches during replication, maintaining DNA stability.Citation25 MMRD GBM demonstrated high TMB and predicted high neoantigen load, even higher than those of melanoma, lung cancer, and colon cancer, which are usually considered ‘immuno-responsive’ tumors.Citation26 In an MMRD pedigree study,Citation26 2 pediatric siblings received nivolumab after GBM recurrence (newly diagnosed at 3.5 years and 6.5 years and recurrent at 4 and 7 years, respectively), resulting in a significant radiologic and clinical response. Follow-up MRI showed tumor disappearance, which was stably maintained for a long time. An abstractCitation35 from the 2019 American Society of Clinical Oncology (ASCO) conference also reported that after treating 12 high grade glioma patients (with weak and absent MMR expression, according to immunochemistry) with pembrolizumab, 4 patients developed SD. In the weak MMR and absent MMR subgroups, the median OS was 2.8 months and 5.6 months, respectively, and the PFS was 1.8 months and 3.1 months, respectively. Although the tumors progressed in all patients, survival data showed that patients without MMR might benefit from pembrolizumab monotherapy.

Neoadjuvant therapy

As a new dosing regimen for current CI monotherapy, neoadjuvant treatment has been reported to reverse the immunosuppressive properties of the GBM microenvironment, enhance local and systemic antitumor immune responses, improve chemokine expression, and increase immune cell infiltration in tumors.Citation36 A single-arm phase 2 clinical trial treated 30 patients with GBM (27 recurrent and 3 newly diagnosed) with neoadjuvant nivolumabCitation37 and found a median OS of 7.3 months, and a median PFS of 4.1 months. Furthermore, 2 patients had PFS of 28.5 and 33.3 months. Multiple molecular and cellular analyzes showed that after neoadjuvant therapy, the expression of chemokines in tumor tissues was enhanced, and immune cell infiltration was increased. Another randomized trial involving 35 patients with recurrent GBMCitation38showed that survival was longer in the neoadjuvant pembrolizumab group than adjuvant pembrolizumab group (median PFS of 2.4 months and 3.3 months, respectively, and median OS of 13.7 months and 7.5 months, respectively). Tumor gene expression profiles also converted in the experimental group, with enhanced interferon-γ-related gene expression, increased T cell clonality, and downregulated PD-1 signal. These two clinical trials suggest therapeutic potential for neoadjuvant therapy. Furthermore, a retrospective study investigated 66 CI-treated patients with recurrent GBM to specifically identify molecular changes caused by neoadjuvant therapy and to predict its efficacy.Citation39 In responders, the expression of mitogen-activated protein kinase (MAPK) pathway-related genes (PTPN1, BRAF) increased, accompanied by an increase in T cell clone diversity and tumor microenvironment heterogeneity. However, in non-responders, enriched PTEN mutations indicated an immunosuppressive microenvironment, together with a failure of selective recruitment of lymphocytes. Such molecular alterations may act as response predictors and should be considered in further clinical investigations of neoadjuvant CI therapy.

Preclinical studies of combination therapy

As the efficacy of CI monotherapy has not meet expectations, combination therapies with CIs and other treatments are being pursued. Preclinical animal studies have indicated partial potential of the combination of standard therapy, targeted therapy, or other immunotherapies.

Addition of the standard medication temozolomide (TMZ) may enhance antitumor efficacy in rodent models.Citation40,Citation41 Local TMZ can increase the number of CD3 + T cells in the peripheral blood. Local TMZ + anti-PD1 treatment produced longer survival and higher circulating lymphocyte numbers than in monotherapy.Citation40 However, systemic TMZ chemotherapy may result in lymphodepletion, abrogate part of the immune response induced by CIs, and decrease immune memories,Citation42-44 suggesting that TMZ administration strategies should be considered.Citation45 For radiotherapy (RT) rodents treatedwith anti-PD1combined with RT had a longer survival time with increased TILs and decreased local immunosuppression, indicating therapeutic potential.Citation46

The targeted monoclonal antibody bevacizumab has been regularly used in recurrent GBM as salvage therapy since it was FDA approved in 2009.Citation47,Citation48 PD-L1 expression is upregulated in bevacizumab refractory tumors, and CIs may restore sensitivity to bevacizumab.Citation50 Bevacizumab can also promote the efficacy of CIs by inducing local high endothelial venules (HEVs),Citation50 regulating the immune microenvironment, and promoting the antitumor immune response.Citation50 However, the specific effects of GBM treatment still need to be further explored.

Combinations with other immunotherapies are also considerable strategies. TIM-3 and LAG-3 are co-inhibitory receptors expressed in TILs and can lead to immune exhaustion in the GBM microenvironment.Citation51,Citation52 The combination of anti-TIM3 + anti-PD1 + focal radiation and anti-LAG3 + anti-PD1 resulted in prolonged survival in murine gliomas,Citation53,Citation54 and follow-up examinations also revealed immune cell infiltration and immune memory cell formation. The combined measles virus (MV) + anti-PD-1-treated mouse group showed an extended survival time with an increased number of TILs.Citation55 The combination of dendritic cell (DC) vaccines + anti-PD1 extended survival time and increased integrin homing and immunologic memory markers on TILs,Citation56 which indicated a potential for clinical translation.

Pre-clinical research, especially the integration of pre-clinical models, may assist in optimizing combination strategies and match them with appropriate patient subsets.Citation57 However, the translation of preclinical murine models into clinical studies is an inevitable challenge.

Clinical studies of combination therapy

Clinical studies have provided valuable information about combination therapies. also includes essential information from combinational clinical trials. The phase 3 clinical trial Checkmate Citation498Citation58 focused on newly diagnosed, MGMT unmethylated GBM. Preliminary published results showed that compared to TMZ + RT therapy (without TMZ), nivolumab + RT treatment did not meet the main endpoint of prolonged OS. The phase 2 trial Checkmate 548Citation59 focused on newly diagnosed, MGMT methylated GBM and reported that compared to TMZ + RT treatment, the PFS of patients treated with nivolumab + TMZ + RT was not extended, and the OS has not been reported. At the 2019 ASCO conference, optimistic data were released. Patients with newly diagnosed MGMT unmethylated GBM were treated with durvalumab + RT after surgery followed by durvalumab monotherapy. Among 40 patients, the median OS was 15.1 months (12.0–18.4 months), and 8 (20%) patients remained alive when the abstract was published, with ongoing survival ranging from 15.7 to 34.9 months.Citation60 Initial trials did not demonstrate significantly improved efficacy after combination with standard therapy, and further studies have been pursued. On the other hand, due to optimistic data reported from small-scale studies, and several ongoing clinical trials, the combination with standard therapy should not be totally negated. The failure of combination studies to date should not detract from future attempts, but that the ideal drug pairings and patient population require careful consideration.

Several clinical trials of bevacizumab combination therapy have published preliminary results. A phase 2 clinical trial involving bevacizumab naïve patients with recurrent GBM reported primary data at the 2018 ASCO conference: compared with pembrolizumab monotherapy, combination with bevacizumab did not show superiority with regard to survival time (median OS of 10.3 months vs 8.8 months, 6-month PFS of 6.7% vs 26%).Citation61 Two phase 2 trials reported that the combination of pembrolizumab and bevacizumab could benefit tumor control and prolong survival time.Citation62,Citation63 Another phase 2 clinical trial treated bevacizumab refractory recurrent GBM with durvalumab + continuing bevacizumab.Citation64 Eight patients (36%) exhibited an OS ≥22 weeks, and 11 patients (50%) had a PFS ≥8 weeks. The results suggested that durvalumab + bevacizumab exhibited preliminary activity. The results of these studies indicate that bevacizumab combination therapy may be effective, and further investigations are warranted.

The results of clinical studies in combination with other immunotherapies are limited, but multiple clinical trials are in progress (shown in ). Nivolumab + ipilimumab combination treatment for melanoma showed improved PFS compared with monotherapy,Citation65 but due to differences in tumor conditions, the efficacy of this combination for GBM is limited by safety and tolerability. In the Checkmate 143 trial, in the nivolumab + ipilimumab group, 40% of patients were forced to discontinue treatment due to intolerance.Citation66 Several trials of combinations with vaccines, oncolytic viruses, and CAR-T therapies are in progress, and further results are anticipated.

Safety and tolerability

PD-1 inhibitors are generally well tolerated.Citation67,Citation68 According to the National Comprehensive Cancer Network (NCCN)Citation69,Citation70 and European Society for Medical Oncology (ESMO)Citation71 guidelines, the overall incidence of adverse events (AEs) caused by PD-1 antibodies was 26.8%, the incidence of high-level AEs was 6.1%, and no significant differences in AEs were observed between the PD-1 and PD-L1 antibody groups.Citation72The Checkmate 143Citation66 trial showed that the most common AEs associated with nivolumab included fatigue, diarrhea, headache, elevated lipase, and nausea. Patients had a greater tolerance for nivolumab than ipilimumab, and the incidence of AEs increased with the ipilimumab dosage. For neurological AEs, the overall incidence was 6.1%,Citation72 with a highly heterogeneous clinical spectrum. The most commonly reported AEs were grade 1–2, included headaches, encephalopathies and meningitis.Citation72 In Checkmate 143, headaches and seizures occurred most frequently among neurological AEs, which were also well tolerated.Citation73 There was also brain specific CI-related pseudo-progression reported. Pseudo-progression is due to a locally stimulated immune response, which can mimic the radiologic features of progression including enhancement and edema.Citation74 Therefore, imaging features of tumor deterioration at the initial stage of immunotherapy cannot exclude subsequent clinical benefit according to the Immunotherapy Response Assessment in Neuro-Oncology (iRANO) criteria.Citation75 In clinical practice, it is necessary to correctly distinguish pseudo-progression from true progression and determine treatment failure.

CI-related AEs need to be discovered in a timely manner and closely monitored. With prompt intervention, most can be effectively managed.Citation76 Glucocorticoids can reverse nearly all immune-related AEs and are suggested for grade 3–4 or prolonged grade 2 AEs.Citation77 Prolonged use of glucocorticoids may have benefits in preventing the recurrence of AEs, especially pneumonia and hepatitis. Glucocorticoids are widely used in GBM patients to reduce tumor-related cerebral edema and radiation necrosis.Citation78,Citation79 Nonetheless, the impact of glucocorticoids on patients with GBM treated with CIs has not been fully investigated. Evidence has shown that glucocorticoids do not reduce the efficacy of immunotherapy, but pre-administration before treatment may be associated with immunosuppression.Citation80,Citation81 The impact of glucocorticoids on OS also remains conflicting.Citation66,Citation79,Citation82 Thus, in clinical practice with patients with GBM, the clinical beneficial effect, immune suppressing effect, CI-related AEs reversing effect, and survival effect of steroids must be balanced. Additionally, oral mycophenolate, intravenous infliximab (anti-TNFa mAb), plasma exchange, and infusion of anti-thymocyte immunoglobulin can be considered for severe AEs.Citation69 Resuming immunotherapy after severe AEs requires cautiousness, and follow-up monitoring is necessary.Citation76

Conclusion

At present, the use of anti-PD-1/PD-L1 CIs for GBM has received attention, but the overall efficacy is still unsatisfactory. Due to the unique properties of GBM, many challenges still need to be overcome before reaching a ‘breakthrough’. Some studies have supported that screening specific patient subgroups may result in the possibility of greater benefits. Neoadjuvant administration, as a new treatment strategy, needs more clinical studies to assess its safety and efficacy. Data from large-scale clinical trials focusing on CIs combination therapies are limited, and most of the published results were negative, demonstrating no significant survival benefits. However, some small-scale studies and subgroups from large-scale studies reported varying degrees of benefits, indicating that detracting certain combination strategies still needs consideration. Furthermore, multiple clinical trials are under investigation, and the results are highly anticipated. Further studies surmounting challenges, such as investigating immunotherapy endpoints, identifying biomarkers for personalized treatment, and developing useful imagination evaluation criteria, are still being pursued.Citation57 In conclusion, CIs for GBM have multiple challenges and potential, and more in-depth studies and clinical trials are needed.

Disclosure of Potential Conflicts of Interest

The authors declare that there is no conflict of interest. The authors have no personal, financial, or institutional interest in any of the drugs, materials, or devices described in this article.

Additional information

Funding

This work was supported by Beijing Municipal Natural Science Foundation (7202150), Beijing Municipal Natural Science Foundation (19JCZDJC64200(Z)), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (2016-I2M-2-001), and Tsinghua University-Peking Union Medical College Hospital Initiative Scientific Research Program (2019ZLH101). No funding has been received from industry.

References

  • Jiang TM, Ma Y, Mao W, Mao Q, You Y, Yang X, Jiang C, Kang C, Li X, Chen L, et al. CGCG clinical practice guidelines for the management of adult diffuse gliomas. Cancer Lett. 2016;375(2):263–73. doi:10.1016/j.canlet.2016.01.024.
  • Garber ST, Hashimoto Y, Weathers SP, Xiu J, Gatalica Z, Verhaak RGW, Zhou S, Fuller GN, Khasraw M, de Groot J, et al. Immune checkpoint blockade as a potential therapeutic target: surveying CNS malignancies. Neuro Oncol. 2016;18(10):1357–66. doi:10.1093/neuonc/now132.
  • Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–32. doi:10.1056/NEJMoa1503093.
  • Schachter J, Ribas A, Long GV, Arance A, Grob -J-J, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390(10105):1853–62. doi:10.1016/S0140-6736(17)31601-X.
  • Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob -J-J, Cowey CL, Lao CD, Wagstaff J, Schadendorf D, Ferrucci PF, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–56. doi:10.1056/NEJMoa1709684.
  • Herbst RS, Baas P, Kim D-W, Felip E, Pérez-Gracia JL, Han J-Y, Molina J, Kim J-H, Arvis CD, Ahn M-J, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–50. doi:10.1016/S0140-6736(15)01281-7.
  • Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823–33. doi:10.1056/NEJMoa1606774.
  • Horn L, Mansfield AS, Szczesna A, Havel L, Krzakowski M, Hochmair MJ, Huemer F, Losonczy G, Johnson ML, Nishio M, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-CELL LUNG CANCER. N Engl J Med. 2018;379(23):2220–29. doi:10.1056/NEJMoa1809064.
  • Ott PA, Bang YJ, Piha-Paul SA, et al. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol. 2019;37(4):318-327. doi: 10.1200/JCO.2018.78.2276
  • Reiss SN, Yerram P, Modelevsky L, Grommes C. Retrospective review of safety and efficacy of programmed cell death-1 inhibitors in refractory high grade gliomas. J Immunother Cancer. 2017;5(1):99. doi:10.1186/s40425-017-0302-x.
  • Kurz SC, Cabrera LP, Hastie D, Huang R, Unadkat P, Rinne M, Nayak L, Lee EQ, Reardon DA, Wen PY, et al. PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology. 2018;91(14):e1355–e1359. doi:10.1212/WNL.0000000000006283.
  • Reardon DA, Brandes AA, Brandes AA, Rieger J, Wick A, Sepulveda J, Phuphanich S, de Souza P, Ahluwalia MS, Lim M, et al. Randomized phase 3 study evaluating the efficacy and safety of nivolumab Vs bevacizumab in patients with recurrent glioblastoma: checkmate 143. neuro-oncology. 2017;19(suppl_3):iii21. doi:10.1093/neuonc/nox036.071.
  • Chen RQ, Liu F, Qiu XY, Chen XQ. The prognostic and therapeutic value of PD-L1 in glioma. Front Pharmacol. 2018;9:1503. doi:10.3389/fphar.2018.01503.
  • Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KH, de Wit M, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2017;377(20):1919–29. doi:10.1056/NEJMoa1709937.
  • Lukas RV, Rodon J, Becker K, Wong ET, Shih K, Touat M, Fassò M, Osborne S, Molinero L, O’Hear C, et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J Neurooncol. 2018;140(2):317–28. doi:10.1007/s11060-018-2955-9.
  • Caccese M, Indraccolo S, Zagonel V, Lombardi G. PD-1/PD-L1 immune-checkpoint inhibitors in glioblastoma: A concise review. Crit Rev Oncol Hematol. 2019;135:128–34. doi:10.1016/j.critrevonc.2018.12.002.
  • Reardon DA, Kaley TJ, Dietrich J, Clarke JL, Dunn GP, Lim M, Cloughesy TF, Gan HK, Park AJ, Schwarzenberger P, et al. Phase 2 study to evaluate safety and efficacy of MEDI4736 (durvalumab DUR]) in glioblastoma (GBM) patients: an update. JCO. 2017;35(15):2042–2042. doi:10.1200/JCO.2017.35.15_suppl.2042.
  • Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, Wöhrer A, Dieckmann K, Filipits M, Brandstetter A, Weller M, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. 2015;17(8):1064–75. doi:10.1093/neuonc/nou307.
  • Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019;18(3):197–218. doi:10.1038/s41573-018-0007-y.
  • Antonios JP, Soto H, Everson RG, Moughon D, Orpilla JR, Shin NP, Sedighim S, Treger J, Odesa S, Tucker A, et al. Immunosuppressive tumor-infiltrating myeloid cells mediate adaptive immune resistance via a PD-1/PD-L1 mechanism in glioblastoma. Neuro Oncol. 2017;19(6):796–807. doi:10.1093/neuonc/now287.
  • Maxwell R, Jackson CM, Lim M. Clinical trials investigating immune checkpoint blockade in glioblastoma. Curr Treat Options Oncol. 2017;18(8):51. doi:10.1007/s11864-017-0492-y.
  • Nduom EK, Wei J, Yaghi NK, Huang N, Kong L-Y, Gabrusiewicz K, Ling X, Zhou S, Ivan C, Chen JQ, et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro Oncol. 2016;18(2):195–205. doi:10.1093/neuonc/nov172.
  • Johanns TM, Miller CA, Dorward IG, Tsien C, Chang E, Perry A, Uppaluri R, Ferguson C, Schmidt RE, Dahiya S, et al. Immunogenomics of hypermutated glioblastoma: a patient with germline POLE deficiency treated with checkpoint blockade immunotherapy. Cancer Discov. 2016;6(11):1230–36. doi:10.1158/2159-8290.CD-16-0575.
  • Barresi V, Simbolo M, Mafficini A, et al. Ultra-mutation in IDH wild-type glioblastomas of patients younger than 55 years is associated with defective mismatch repair, microsatellite instability, and giant cell enrichment. Cancers (Basel). 2019;11(9):1279. doi:10.3390/cancers11091279.
  • Jiricny J. The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol. 2006;7(5):335–46. doi:10.1038/nrm1907.
  • Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M, Durno C, Krueger J, Cabric V, Ramaswamy V, et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol. 2016;34(19):2206–11. doi:10.1200/JCO.2016.66.6552.
  • Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35–44. doi:10.1016/j.cell.2016.02.065.
  • Costantini A, Takam Kamga P, Dumenil C, Chinet T, Emile J-F, Giroux Leprieur E. Plasma biomarkers and immune checkpoint inhibitors in non-small cell lung cancer: new tools for better patient selection? Cancers (Basel). 2019;11(9):1269. doi:10.3390/cancers11091269.
  • Dawood S. The evolving role of immune oncology in colorectal cancer. Chin Clin Oncol. 2018;7(2):17–17. doi:10.21037/cco.2018.04.07.
  • Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542–e551. doi:10.1016/S1470-2045(16)30406-5.
  • Campbell BB, Light N, Fabrizio D, Zatzman M, Fuligni F, de Borja R, Davidson S, Edwards M, Elvin JA, Hodel KP, et al. Comprehensive analysis of hypermutation in human cancer. Cell. 2017;171(5):1042–1056 e10.
  • Baretti M, Le DT. DNA mismatch repair in cancer. Pharmacol Ther. 2018;189:45–62. doi:10.1016/j.pharmthera.2018.04.004.
  • Tiffany R, Hodges MO, Xiu J, Gatalica Z, Swensen J, Zhou S, Huse JT, de Groot J, Li S, Overwijk WW, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro-Oncology. 2017;19:047–1057.
  • Adrienne Johnson ES, Gay L, Vergilio J-A, Elvin J, Suh J, Daniel S, Covert M, Frampton GM, Ali S, Miller V, et al. Comprehensive genomic profiling of 282 pediatric low- and high-grade gliomas reveals genomic drivers, tumormutational burden, and hypermutation signatures. Oncologist. 2017;22:1478–90. doi:10.1634/theoncologist.2017-0242.
  • Giuseppe Lombardi MC, Simonelli M, Fassan M, Padovan M, Persico P, Bellu L, Dipasquale A, PaolaGardiman M, Indraccolo S, Zagonel V. Pembrolizumab (Pem) in recurrent high-grade glioma (HGG) patients (PTS) with mismatch repair deficiency (MMRd): an observational study. Am Soc Clin Oncol. 2019;2043. doi:10.1200/JCO.2019.37.15_suppl.2043.
  • Berghoff ASPM. Does neoadjuvant anti-PD1 therapy improve glioblastoma outcome? Nat Rev Neurol. 2019;15(6):314–15. doi:10.1038/s41582-019-0178-0.
  • Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, López-Janeiro A, Porciuncula A, Idoate MA, Inogés S, de Andrea C, López-diaz de Cerio A, Tejada S, et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 2019;25(3):470–76. doi:10.1038/s41591-018-0339-5.
  • Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB, Wang AC, Ellingson BM, Rytlewski JA, Sanders CM, 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):477–86. doi:10.1038/s41591-018-0337-7.
  • Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T, Bordbar D, Shan D, Samanamud J, Mahajan A, et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med. 2019;25(3):462–69. doi:10.1038/s41591-019-0349-y.
  • Dai B, Qi N, Li J, Zhang G. Temozolomide combined with PD-1 antibody therapy for mouse orthotopic glioma model. Biochem Biophys Res Commun. 2018;501(4):871–76. doi:10.1016/j.bbrc.2018.05.064.
  • Park J, Kim CG, Shim J-K, Kim JH, Lee H, Lee JE, Kim MH, Haam K, Jung I, Park S-H, et al. Effect of combined anti-PD-1 and temozolomide therapy in glioblastoma. Oncoimmunology. 2019;8(1):e1525243. doi:10.1080/2162402X.2018.1525243.
  • Silu Wang FY, Lu X, Li Q, Su Z, Lee J-H, Wang C, Linyong D. Efficacy of systemic temozolomide-activated phage-targeted gene therapy in human glioblastoma. EMBO Mol Med. 2019;11:e8492.
  • Karachi A, Dastmalchi F, Mitchell DA, Rahman M. Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro Oncol. 2018;20(12):1566–72. doi:10.1093/neuonc/noy072.
  • Heynckes S, Daka K, Franco P, Gaebelein A, Frenking JH, Doria-Medina R, Mader I, Delev D, Schnell O, Heiland DH, et al. Crosslink between temozolomide and PD-L1 immune-checkpoint inhibition in glioblastoma multiforme. BMC Cancer. 2019;19(1):117. doi:10.1186/s12885-019-5308-y.
  • Mathios D, Kim JE, Mangraviti A, Phallen J, Park C-K, Jackson CM, Garzon-Muvdi T, Kim E, Theodros D, Polanczyk M, et al. Anti-PD-1 antitumor immunity is enhanced by local and abrogated by systemic chemotherapy in GBM. Sci Transl Med. 2016;8(370):370ra180. doi:10.1126/scitranslmed.aag2942.
  • Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 2013;86(2):343–49. doi:10.1016/j.ijrobp.2012.12.025.
  • Field KM, Jordan JT, Wen PY, Rosenthal MA, Reardon DA. Bevacizumab and glioblastoma: scientific review, newly reported updates, and ongoing controversies. Cancer. 2015;121(7):997–1007. doi:10.1002/cncr.28935.
  • Diaz RJ, Ali S, Qadir MG, De La Fuente MI, Ivan ME, Komotar RJ. The role of bevacizumab in the treatment of glioblastoma. J Neurooncol. 2017;133(3):455–67. doi:10.1007/s11060-017-2477-x.
  • Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V, Feyen K, Tawney J, Hanahan D, Michael IP, et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med. 2017;9(385):eaak9679. doi:10.1126/scitranslmed.aak9679.
  • Tamura R, Tanaka T, Ohara K, Miyake K, Morimoto Y, Yamamoto Y, Kanai R, Akasaki Y, Murayama Y, Tamiya T, et al. Persistent restoration to the immunosupportive tumor microenvironment in glioblastoma by bevacizumab. Cancer Sci. 2019;110(2):499–508. doi:10.1111/cas.13889.
  • Joller N, Kuchroo VK. Tim-3, Lag-3, and TIGIT. Curr Top Microbiol Immunol. 2017;410:127–56. doi:10.1007/82_2017_62.
  • Hung AL, Garzon-Muvdi T, Lim M. Biomarkers and immunotherapeutic targets in glioblastoma. World Neurosurg. 2017;102:494–506. doi:10.1016/j.wneu.2017.03.011.
  • Kim JE, Patel MA, Mangraviti A, Kim ES, Theodros D, Velarde E, Liu A, Sankey EW, Tam A, Xu H, et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin Cancer Res. 2017;23(1):124–36. doi:10.1158/1078-0432.CCR-15-1535.
  • Harris-Bookman S, Mathios D, Martin AM, Xia Y, Kim E, Xu H, Belcaid Z, Polanczyk M, Barberi T, Theodros D, et al. Expression of LAG-3 and efficacy of combination treatment with anti-LAG-3 and anti-PD-1 monoclonal antibodies in glioblastoma. Int J Cancer. 2018;143(12):3201–08. doi:10.1002/ijc.31661.
  • Hardcastle J, Mills L, Malo CS, Jin F, Kurokawa C, Geekiyanage H, Schroeder M, Sarkaria J, Johnson AJ, Galanis E, 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. doi:10.1093/neuonc/now179.
  • Antonios JP, Soto H, Everson RG, Orpilla J, Moughon D, Shin N, Sedighim S, Yong WH, Li G, Cloughesy TF, et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight. 2016;1(10). doi:10.1172/jci.insight.87059.
  • Hegde PS, Chen DS. Top 10 challenges in cancer immunotherapy. Immunity. 2020;52(1):17–35. doi:10.1016/j.immuni.2019.12.011.
  • Squibb B-M, Bristol-myers squibb announces phase 3 CheckMate 498 study did not meet primary endpoint of overall survival with opdivo (nivolumab) plus radiation in patients with newly diagnosed MGMT-unmethylated glioblastoma multiforme. 2019 May 9. Accessed 2019 Nov 15. https://news.bms.com/press-release/corporatefinancial-news/bristol-myers-squibb-announces-phase-3-checkmate-498-study-did
  • Squibb B-M Bristol-myers squibb provides update on phase 3 opdivo (nivolumab) CheckMate −548 trial in patients with newly diagnosed MGMT-methylated glioblastoma multiforme. 2019 Sep 5. Accessed 2018 Nov 9. https://news.bms.com/press-release/corporatefinancial-news/bristol-myers-squibb-provides-update-phase-3-opdivo-nivolumab-]
  • A, Reardon TJK, Dietrich J, Clarke JL, Dunn G, Lim M, Cloughesy TF, Gan HK, Park AJ, Schwarzenberger P, et al. Phase II study to evaluate safety and efficacy of MEDI4736 (durvalumab) + radiotherapy in patients with newly diagnosed unmethylated MGMT glioblastoma (new unmeth GBM). J Clin Oncol 37, 2019 (suppl; abstr 2032) DOI: 10.1200/JCO.2019.37.15_suppl.2032
  • Reardon DANL, Peters KB, Peters KB, Clarke JL, Jordan JT, De Groot JF, Nghiemphu PL, Kaley TJ, Colman H, Gaffey SC, et al. Phase II study of pembrolizumab or pembrolizumab plus bevacizumab for recurrent glioblastoma (rGBM) patients. J Clin Oncol. 2018;36:2006. doi:10.1200/JCO.2018.36.15_suppl.2006.
  • Reardon DAGJ, Colman H, Jordan JT, Daras M, Clarke JL, Nghiemphu PL, Gaffey SC, Peters KB. Safety of pembrolizumab in combination with bevacizumab in recurrent glioblastoma (rGBM). J Clin Oncol. 2016;34. doi:10.1200/JCO.2016.34.15_suppl.2010.
  • Sahebjam SJP, Forsyth PAJ, Arrington J, Vrionis FD, Etame AB, Tran ND, Dalvi PH, Kim S, Macaulay RJ, Chinnaiyan P, et al. Safety and antitumor activity of hypofractionated stereotactic irradiation (HFSRT) with pembrolizumab (Pembro) and bevacizumab (Bev) in patients (pts) with recurrent high grade gliomas: preliminary results from phase I study. J Clin Oncol. 2016;34. doi:10.1200/JCO.2016.34.15_suppl.2041.
  • David Reardon TK, Dietrich J, Dietrich J, Clarke JL, Dunn GP, Lim M, Cloughesy T, Gan HK, Park A, Schwarzenberger P, et al. Atim-12. Phase 2 study to evaluate the clinical efficacy and safety of Medi4736 (Durvalumab [DUR]) in patients with Bevacizumab (BEV)-refractory recurrent Glioblastoma (GBM). neuro-oncology. 2017;19(suppl_6):vi28. doi:10.1093/neuonc/nox168.108.
  • Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34. doi:10.1056/NEJMoa1504030.
  • Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J, Cloughesy T, Voloschin A, Ramkissoon SH, Ligon KL, Latek R, et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol. 2018;20(5):674–86. doi:10.1093/neuonc/nox208.
  • Cousin S, Seneschal J, Italiano A. Toxicity profiles of immunotherapy. Pharmacol Ther. 2018;181:91–100. doi:10.1016/j.pharmthera.2017.07.005.
  • Farber SH, Elsamadicy AA, Atik AF, Suryadevara CM, Chongsathidkiet P, Fecci PE, Sampson JH. The safety of available immunotherapy for the treatment of glioblastoma. Expert Opin Drug Saf. 2017;16(3):277–87. doi:10.1080/14740338.2017.1273898.
  • Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, Budde LE, Costa L, Davies M, Dunnington D, et al. Management of immunotherapy-related toxicities, Version 1.2019. J Natl Compr Canc Netw. 2019;17(3):255–89. doi:10.6004/jnccn.2019.0013.
  • Thompson JA. New NCCN guidelines: recognition and management of immunotherapy-related toxicity. J Natl Compr Canc Netw. 2018;16(5S):594–96. doi:10.6004/jnccn.2018.0047.
  • Haanen J, Carbonnel F, Robert C, Kerr KM, Peters S, Larkin J, Jordan K. Management of toxicities from immunotherapy: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(suppl_4):iv119–iv142. doi:10.1093/annonc/mdx225.
  • Cuzzubbo S, Javeri F, Tissier M, Roumi A, Barlog C, Doridam J, Lebbe C, Belin C, Ursu R, Carpentier AF, et al. Neurological adverse events associated with immune checkpoint inhibitors: review of the literature. Eur J Cancer. 2017;73:1–8. doi:10.1016/j.ejca.2016.12.001.
  • Lim MOA, Vlahovic G, Reardon DA, Sahebjam S, Cloughesy T, Cloughesy T, Baehring J, Butowski N, Potter V, Zwirtes R, et al. Nivolumab (nivo) in combination with radiotherapy (RT) ± temozolomide (TMZ): updated safety results from CheckMate 143 in pts with methylated or unmethylated newly diagnosed glioblastoma (GBM). Ann Oncol. 2017;28:v109–21. doi:10.1093/annonc/mdx366.
  • Nayak L, DeAngelis LM, Brandes AA, Peereboom DM, Galanis E, Lin NU, Soffietti R, Macdonald DR, Chamberlain M, Perry J, et al. The Neurologic Assessment in Neuro-Oncology (NANO) scale: a tool to assess neurologic function for integration into the Response Assessment in Neuro-Oncology (RANO) criteria. Neuro Oncol. 2017;19(5):625–35. doi:10.1093/neuonc/nox029.
  • Okada H, Weller M, Huang R, Finocchiaro G, Gilbert MR, Wick W, Ellingson BM, Hashimoto N, Pollack IF, Brandes AA, et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 2015;16(15):e534–e542. doi:10.1016/S1470-2045(15)00088-1.
  • Puzanov I, Diab A, Abdallah K, Bingham CO, Brogdon C, Dadu R, Hamad L, Kim S, Lacouture ME, LeBoeuf NR, et al. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) toxicity management working group. J Immunother Cancer. 2017;5(1):95. doi:10.1186/s40425-017-0300-z.
  • Eigentler TK, Hassel JC, Berking C, Aberle J, Bachmann O, Grünwald V, Kähler KC, Loquai C, Reinmuth N, Steins M, et al. Diagnosis, monitoring and management of immune-related adverse drug reactions of anti-PD-1 antibody therapy. Cancer Treat Rev. 2016;45:7–18. doi:10.1016/j.ctrv.2016.02.003.
  • Weller M, van den Bent M, Hopkins K, Tonn JC, Stupp R, Falini A, Cohen-Jonathan-Moyal E, Frappaz D, Henriksson R, Balana C, et al. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol. 2014;15(9):e395–e403. doi:10.1016/S1470-2045(14)70011-7.
  • Kelly WJ, Gilbert MR. Glucocorticoids and immune checkpoint inhibitors in glioblastoma. J Neurooncol. 2020. doi:10.1007/s11060-020-03439-2.
  • Williams KJ, Grauer DW, Henry DW, Rockey ML. Corticosteroids for the management of immune-related adverse events in patients receiving checkpoint inhibitors. J Oncol Pharm Pract. 2019;25(3):544–50. doi:10.1177/1078155217744872.
  • Giles AJ, Hutchinson MND, Sonnemann HM, Jung J, Fecci PE, Ratnam NM, Zhang W, Song H, Bailey R, Davis D, et al. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer. 2018;6(1):51. doi:10.1186/s40425-018-0371-5.
  • Pitter KL, Tamagno I, Alikhanyan K, Hosni-Ahmed A, Pattwell SS, Donnola S, Dai C, Ozawa T, Chang M, Chan TA, et al. Corticosteroids compromise survival in glioblastoma. Brain. 2016;139(Pt 5):1458–71. doi:10.1093/brain/aww046.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.