Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
. 2015 Oct 29;17(Suppl 7):vii32–vii40. doi: 10.1093/neuonc/nov178

Immunotherapy for neuro-oncology: the critical rationale for combinatorial therapy

David A Reardon 1, Mark R Gilbert 1, Wolfgang Wick 1, Linda Liau 1
PMCID: PMC4625894  PMID: 26516225

Abstract

A successful therapeutic paradigm established historically in oncology involves combining agents with potentially complementary mechanisms of antitumor activity into rationally designed regimens. For example, cocktails of cytotoxic agents, which were carefully designed based on mechanisms of action, dose, and scheduling considerations, have led to dramatic improvements in survival including cures for childhood leukemia, Hodgkin's lymphoma, and several other complex cancers. Outcome for glioblastoma, the most common primary malignant CNS cancer, has been more modest, but nonetheless our current standard of care derives from confirmation that combination therapy surpasses single modality therapy. Immunotherapy has recently come of age for medical oncology with exciting therapeutic benefits achieved by several types of agents including vaccines, adoptive T cells, and immune checkpoint inhibitors against several types of cancers. Nonetheless, most benefits are relatively short, while others are durable but are limited to a minority of treated patients. Critical factors limiting efficacy of immunotherapeutics include insufficient immunogenicity and/or inadequate ability to overcome immunosuppressive factors exploited by tumors. The paradigm of rationally designed combinatorial regimens, originally established by cytotoxic therapy for oncology, may also prove relevant for immunotherapy. Realization of the true therapeutic potential of immunotherapy for medical oncology and neuro-oncology patients may require development of combinatorial regimens that optimize immunogenicity and target tumor adaptive immunosuppressive factors.

Keywords: glioblastoma, immunotherapy, immune checkpoint, programmed-death 1, vaccine


The past few years have seen remarkable outcomes achieved by a variety of immunotherapeutic strategies across a spectrum of cancers. Nonetheless, with the exception of chimeric antigen receptor (CAR) T cell therapy targeting CD19 for B lineage leukemia, where the majority of treated patients have achieved deep and durable antitumor benefit,1,2 outcomes achieved by single agent immunotherapeutics have been limited to either relatively short duration of benefit or benefit achieved by a relatively small percentage of treated patients. Sipuleucel-T, the first noninfectious, agent-based vaccine approved for cancer therapy, achieved approval for patients with metastatic, advanced prostate cancer based on a 4.1 month median overall survival (OS) improvement over placebo,3 while the CTLA-4 inhibitor ipilimumab has received approval for metastatic melanoma based on a 3.6 month median survival benefit.4 The humanized anti-PD-1 monoclonal antibodies nivolumab and pembrolizumab are associated with unprecedented > 5-year survival for metastatic melanoma, a disease previously regarded as treatment refractory and as deadly as glioblastoma, but only 10%–15% of patients achieve this success. These modest, yet encouraging, results provide proof-of-concept that immunotherapy can successfully battle cancer but, as with most important advances, additional questions arise. Are these successes a glimpse of the potential power of the immune system, or are they the best we can hope to achieve? Why are some cancers more responsive to immunotherapy than others? Are there bona fide immunotherapy refractory malignancies? For neuro-oncology, are CNS tumor patients different from other cancer patients? As detailed in a comprehensive, recent review, historical dogma regarding immunoprivilege of the CNS has been steadily replaced by data demonstrating a dynamic and effective interaction of the immune system between systemic and CNS compartments.5

Nonetheless, the answers to these important questions regarding the anticancer potential of immunotherapy lie in addressing 2 fundamental, yet paramount, considerations: (i) how robust is the tumor-specific immune response generated by a given immunotherapeutic, and (ii) is the antitumor immune response able to sufficiently overcome self-protective, immunosuppressive strategies generated by cancers? As detailed by Weller and Heimberger in this supplement, tumors—especially high-grade gliomas—exploit multiple mechanisms systemically as well as within the tumor microenvironment to abrogate antitumor immune responses. Although single- agent immunotherapy approaches may be able to address either immunogenicity or immunosuppression considerations, carefully designed combinatorial regimens will likely be required to address both issues effectively and ensure that a sufficiently potent antitumor immune response has been achieved and that the immunosuppressive barriers erected by tumors are sufficiently negated to allow ample opportunity for immune attack.

Immunotherapy Combined With Cytotoxic Therapy

Cytotoxic radiotherapy and chemotherapy, established therapeutic cornerstones for many malignancies including high-grade glioma, have historically been considered immunosuppressive and thus appear to be paradoxical partners for immunotherapy. Nonetheless, an intact immune system has been linked with enhanced antitumor activity for both radiation therapy and chemotherapy in preclinical cancer models, suggesting that immune responses contribute to the effectiveness of cytotoxic therapy.6,7 Furthermore, growing data link several mechanisms by which cytotoxic agents may lead to more robust antitumor immune attack.813 Prominent among these is the induction of immunogenic cell death, which is defined by 3 primary components: (i) translocation of calreticulin from endoplasmic reticulum to the cell surface, where it activates dendritic cells (DCs); (ii) ATP release, which recruits and matures DCs and upregulates CD40, CD80, and CD86 costimulatory molecules; and (iii) extracellular release of high-mobility group protein B1, which binds to pattern recognition receptor toll-like receptor 4 (TLR4) on DCs and enhances antigen cross-presentation and secretion of proinflammatory cytokines.7,14

Beyond induction of immunogenic cell death, radiation therapy and chemotherapy can enhance antitumor immune responses by myriad additional mechanisms as summarized in Table 1.1540 The abscopal effect, which manifests as a radiation therapy-induced tumor response outside the targeted field, represents a striking example of the ability of cytotoxic therapies to generate systemic antitumor immune responses.41 Temozolomide, an alkylating agent approved for the treatment of glioblastoma, has been shown to enhance antiglioma immune activity by different mechanisms that appear to be dose/schedule dependent including: (i) depletion of Tregs when administered in a metronomic dosing schedule22,23; (ii) generation of increased levels of cytotoxic T cells relative to Tregs during homeostatic proliferation following standard adjuvant dosing42; and (iii) marked expansion of antigen-specific CD8+ T cells following myeloablative dosing.43

Table 1.

Potential mechanisms cytotoxic radiotherapy and chemotherapy can enhance immune system activity

Mechanism Radiotherapy Chemotherapy Reference(s)
Immunogenic cell death 7,14
Chemokine induction (CXCL9/10/11) 24,35,40
Proinflammatory cytokine production 15,33
Increase MHC class I molecule expression 17
Increase co-stimulatory molecule expression 18,19
Decrease Tregs 22,23,28
Increased CD8 + effector cells (homeostatic proliferation) 32,40
Decrease inhibitory immune checkpoint molecules 36
Decrease myeloid suppressor cells 3739
Recruit/activate dendritic cells 25,27

Nonetheless, radiation and chemotherapy can also exert potent immunosuppressive actions including: (i) increased TGF-β activation44; (ii) fostering an M2 macrophage phenotype45; (iii) increased proportion of Tregs46; (iv) increased tumor cell PD-L1 expression induced by INFγ secreted by CD8+ T cells47; and (v) profound and sustained levels of lymphopenia.4850 Thus, the net effect of cytotoxic therapy on antitumor immune responses will likely reflect an imbalance of immune-enhancing versus immunosuppressive actions. Important variables that may impact this balance warrant further investigation and include dose, schedule and, for chemotherapy, agent of choice.

To date there has been limited preclinical investigation of cytotoxic agents combined with immunotherapeutics for glioblastoma. Temozolomide can enhance vaccine efficacy when administered via low, daily (metronomic), or myeloablative dosing schedules,43,51 while stereotactic radiation therapy can augment the antitumor activity of PD-1 blockade.52 Nonetheless, there is substantial effort underway to evaluate immunotherapy combined with cytotoxic therapy in the clinic. Table 2 presents a partial listing of ongoing clinical trials for newly diagnosed glioblastoma patients. Similarly, there is growing interest in evaluating immune checkpoint blockade with radiation therapy administered as either whole brain or stereotactic radiosurgery in patients with brain metastases. Trials to date focus on CTLA-4 blockade with radiotherapy for melanoma metastatic to the brain (NCT02115139, NCT02097732, NCT02107755, NCT01703507, and NCT01950195) while additional trials evaluating other immune checkpoint inhibitors as well as other types of brain metastases are in active planning.

Table 2.

Partial listing of ongoing clinical trials evaluating immunotherapy and cytotoxic therapy for newly diagnosed glioblastoma

Immunotherapy/Agent Phase # Patients Accrual Status Clinicaltrials.gov Comment
EGFRvIII peptide vaccine/Rindopepimut 3 700 Completed NCT01480479 Randomized; double-blind; placebo-controlled;
Tumor lysate loaded DCs/DCVax®-L 3 300 Ongoing NCT00045968 Randomized; double-blind; placebo-controlled;
Tumor lysate-loaded DCs 2 100 Ongoing NCT01567202 Randomized; double-blind; placebo-controlled;
Tumor-associated antigen vaccine/ICT107 2 124 Completed NCT01280552 Randomized; double-blind; placebo-controlled
Tumor-associated antigen vaccine/IMA950 1/2 16 Ongoing NCT01920191 IMA950
Autologous and allogeneic tumor-specific neoantigens/APVAC 1 30 Ongoing NCT02149225
Autologous tumor-specific neoantigens/NeoVax 1 16 Ongoing NCT02287428 MGMT-unmethylated patients only; excludes temozolomide
Autologous DCs + allogeneic stem cell lysate 1 40 Ongoing NCT02010606
Autologous DCs + allogeneic stem cell lysate 1 10 Ongoing NCT01957956
Autologous DCs loaded with CMV pp65-LAMP RNA 1 16 Ongoing NCT00639639
Autologous DCs loaded with CMV pp65-LAMP RNA + basliximab 1 18 Ongoing NCT00626483
PD-L1 mAb/MEDI4736 2 37 Ongoing NCT02336165 MGMT-unmethylated patients only; excludes temozolomide

Although many trials are underway, limited data are available from completed trials that evaluated immunotherapy integrated with cytotoxic therapy for glioblastoma. Encouraging outcomes have been reported for single-arm studies evaluating vaccines derived from autologous DCs pulsed with tumor lysate,5358 tumor-associated antigens,55,59 heat-shock protein peptide complex-96 (HSPPC-96),60 and EGFRvIII peptide.42,61,62 The only randomized, placebo-controlled clinical trial data to date derive from a preliminary outcome analysis of ICT-107, a vaccine consisting of autologous DCs pulsed with a cocktail of 6 tumor-associated antigens administered with standard radiation and temozolomide for newly diagnosed glioblastoma patients (NCT01280552). Although the impact of ICT-107 administration on OS did not reach statistical significance for the intent-to-treat population in this study, a subset analysis of HLA-A2 positive patients revealed that ICT-107 led to a 4-month improvement in both PFS and OS among MGMT-unmethylated patients and an improvement from 8.5 to 24.1 months in PFS for MGMT-methylated patients (HR: 0.259; P = .005), while the median survival for MGMT-methylated patients has not been reached.63

Immunotherapy Combined with Antiangiogenic Therapy

The precise impact of antiangiogenic agents on the pathophysiology of many cancers including glioblastoma is not clear. Nonetheless, growing data support the ability of antiangiogenic agents to shift the tumor microenvironment phenotype from immunosuppressive to immunosupportive.64,65 Vascular endothelial growth factor (VEGF), the primary proangiogenic growth factor in many cancers including malignant glioma, contributes significantly to the immunosuppressive ability of tumors.6668 Specifically, VEGF can inhibit DC maturation and antigen presentation, induce apoptosis of CD8+ T cells, enhance Treg activity, and diminish infiltration of T cells across tumor endothelium.6972 Of note, preclinical studies suggest that immunotherapeutics may be combined with VEGF inhibitors to generate enhanced antitumor benefit.6779 The rationale underlying the combination of immunotherapy approaches with antiangiogenic agents is based on the ability of VEGF inhibition to diminish immunosuppressive features of tumors6972,76,78,79 and enhance the antitumor activity of immunotherapies.73,75,76,78,79 Furthermore, preclinical strategies to normalize tumor vasculature, including administration of anti-VEGF therapy, can shift tumor-associated macrophages from an immune-inhibitory M2-like phenotype toward an immune-stimulatory M1-phenotype, increase tumor infiltrating CD8+ T cells, and enhance survival following whole tumor cell vaccination.74

Data from a recently published phase 1 study among metastatic melanoma patients revealed that administration of bevacizumab with ipilimumab, an inhibitor of the CTLA-4 immune checkpoint, led to improved OS relative to historical benchmarks of ipilimumab as well as evidence of increased intratumoral immune-cell trafficking.80 For glioblastoma, data have been reported from only one clinical trial evaluating immunotherapy plus antiangiogenic therapy. In the ReACT study, 70 patients with EGFRvIII-positive glioblastoma at first or second recurrence were randomized to receive bevacizumab plus placebo vaccine versus bevacizumab plus the EGFRvIII peptide vaccine, rindopepimut. In this double-blind, randomized, placebo-controlled study, rindopepimut recipients achieved a significantly longer OS (HR: 0.57; P = .0386). Administration of rindopepimut also conveyed a modest, yet not statistically significant, improvement in PFS (HR: 0.79; P = .3756) as well as a higher rate of durable (≥6 mo) radiographic responses.81 Importantly, these data represent the first randomized clinical trial to demonstrate a survival benefit associated with any type of immunotherapy for glioblastoma to date. Although the results of this trial indicate that rindopepimut improved outcome achieved by bevacizumab, it is not clear whether bevacizumab improved the outcome of rindopepimut because the trial lacked a rindopepimut-alone arm. Nonetheless, the overall results of this study support further clinical trials evaluating combinatorial regimens of immunotherapeutics plus antiangiogenic agents for glioblastoma. Currently, ongoing clinical trials evaluating this approach include trials that combine bevacizumab with: (i) PD-1 blockade (NCT02337491); (ii) PD-L1 blockade (NCT02336165); (iii) HSPPC-96 vaccine (NCT01814813); (iv) autologous tumor lysate vaccine (NCT02010606); or (v) a vaccine derived from combined autologous/allogeneic tumor lysates (NCT01903330).

Immunotherapy Plus Immunotherapy Combinatorial Strategies

Among possible combinatorial strategies for immunotherapy, the most exciting involves combining immunotherapeutics with complementary mechanisms of antitumor immune attack. As previously described, the efficacy of immunotherapeutics against cancer is ultimately dependent on 2 factors: (i) immunogenicity (ability to generate an immune response); and (ii) tumor self-protective immunosuppression strategies. A major contributing factor limiting the overall efficacy of most immunotherapeutics to date, which typically reflects single-agent therapy experience, is an inability to adequately address both of these factors.

One factor that may impact the immunogenicity of cancer vaccines is choice of antigen. Many vaccines target tumor-associated antigens. Immunoreactivity induced by these vaccines is predicted to be relatively low because tumor-associated antigens can also be expressed by normal tissues and may therefore evoke immunotolerance. In contrast, vaccines targeting tumor-specific antigens, which by definition are uniquely expressed by tumor cells and are not present on normal tissues, are expected to generate more potent immune responses that are not limited by normal self-tolerance mechanisms.

Another factor likely limiting the efficacy of cancer vaccines is that tumors can escape immunogenic immune responses induced by vaccines by downregulating target antigen expression or by expanding an existing subset of cells that lack target antigen expression. For example, among glioblastoma patients treated with the EGFRvIII-targeting peptide vaccine rindopepimut, expression of EGFRvIII was no longer detectable at the time of confirmed recurrence.62 This finding suggests that targeting multiple tumor-specific antigens may lessen the likelihood of immune escape and thereby generate more durable antitumor benefit compared with vaccines targeting a single antigen or a small number of antigens.

An insurmountable therapeutic hurdle for glioblastoma to date is the remarkable degree of heterogeneity within individual tumors.82,83 Given this challenge, it is not surprising that cytotoxic agents achieve modest benefit at best, while targeted molecular agents have essentially failed, even among genetically enriched patient populations.84,85 Exploiting the mutanome or constellation of tumor-specific mutations within a given tumor, which include both passenger and driver mutations, represents a challenging yet highly exciting opportunity for immunotherapy. Multiple studies point to the critical relationship between immune responses against tumor-specific mutations often referred to as neoantigens and effective tumor control.8692 In recent analyses, expression of a panel of tumor-specific neoantigens was demonstrated to be a critical predictor of long-term response following immune checkpoint therapy among patients with advanced melanoma93 or non–small cell lung cancer.94 The ability to target a spectrum of tumor-specific mutations, even if heterogeneously expressed within a given tumor, provides immunotherapy a unique opportunity to effectively exploit the challenge posed by intratumoral heterogeneity for therapeutic benefit.

On the other hand, immunosuppressive adaptations exploited by tumors can essentially neutralize antitumor immune responses regardless of the potency, specificity, and breadth. As detailed by Weller and Heimberger in this supplement, glioblastomas invoke a wide array of immunosuppressive strategies that include systemic factors such as lowered levels of T cell responsiveness, immunoglobulins, and monocyte/dendritic function as well as increased Tregs.9598 Even more remarkable are the host of immunosuppressive factors generated by tumors that function in the immediate microenvironment to act as a localized shield or cloaking device against antitumor immune attack including: (i) expression of multiple immunoinhibitory molecules including TGF-β, STAT-3, VEGF, IL-10, prostaglandin E2, tryptophan, and lectin-like transcript 199104; (ii) downregulation of MHC molecule expression101,105,106; (iii) enhanced infiltration of Tregs101,102,107111; (iv) polarization of microglia and tumor-associated macrophages toward an M2 phenotype112,113; (v) upregulation of Fas ligand114,115; (vi) impaired T cell function due to hostile factors including hypoxia116; and (vii) increased expression of inhibitory immune checkpoint molecules such as PD-L1, which was recently described to be prominently expressed by 80%–90% of glioblastoma tumors117 as well as the other B7H family molecule, B7H3104,118

Given the plethora of immunosuppressive mechanisms utilized by tumors including glioblastoma, combinatorial regimens will likely require one or more components that block immunosuppressive mechanisms. Certainly, the emergence of humanized mAbs able to block the CTLA-4, PD-1, and PD-L1 immune checkpoints offer much hope for oncology patients. Although the role of these agents for neuro-oncology patients is currently being defined in ongoing clinical trials, preclinical studies against orthotopic, immunocompetent glioblastoma models demonstrate exciting efficacy including evidence of enhanced benefit with combinatorial regimens.52,119121 Combinations of immune checkpoint inhibitors such as PD-1 plus CTLA-4 blockade offer potential synergy to expand effector T cells while decreasing regulatory T cells and myeloid suppressor cells.122 Furthermore, enhanced therapeutic benefit has been observed with such regimens in preclinical GBM models119,121 and in patients with advanced melanoma.123 However, such regimens may also be limited clinically by enhanced immune-mediated toxicity.123

Additional therapeutics against immunosuppressive mechanisms invoked by glioblastoma tumors are slowly evolving, but further research is critically needed. Initial results of a phase 1 clinical trial evaluating galunisertib (LY2157299;. an oral inhibitor against TGF-β) are encouraging,124 although phase 2 results in monotherapy or in combination with lomustine are negative.125 A clinical trial combining this agent with the PD-1 inhibitor nivolumab will begin in the near future (NCT02423343). Another ongoing study is evaluating the role of Treg targeting and incorporates basiliximab in combination with a vaccine (NCT00626483). In addition, VEGF inhibition plus immunotherapy warrants further investigation, as previously discussed.

Finally, a “ying-yang” combinatorial strategy of high appeal includes immunotherapy regimens integrating an agent capable of generating a robust antitumor immune response (eg, a vaccine against tumor specific antigens) with an agent capable of targeting tumor immunosuppression (eg, a checkpoint inhibitor). Although checkpoint blockade therapy alone may help relieve local immune suppression and overcome T cell exhaustion or anergy, it may be constrained by the size and specificity of the existing T cell population (T cells arising from the normal physiological presentation of the evolving tumor to the host immune system). This consideration may be particularly relevant for tumors that are considered relatively less immunogenic. The lack of coincident, focused immune stimulation may thus be a critical factor that limits maximal efficacy of checkpoint inhibition therapy, a deficit that may be overcome by an effective vaccine. Indeed, in many preclinical animal studies, the antitumor activity of immune-checkpoint blockade was dramatically enhanced when combined with a vaccine.126130 Although clinical data are only beginning to emerge, enhanced activity has also been observed among patients treated with combinatorial tumor vaccination plus immune-checkpoint blockade regimens.131133

Conclusion

The ability of immunotherapies to achieve their therapeutic potential for medical and neuro-oncology will likely depend on the development of rationally designed combinatorial regimens capable of optimizing potent antitumor immune responses while effectively negating self-protective immunosuppressive strategies exploited by many tumors. Glioblastoma tumors exhibit a profound repertoire of immunosuppressive adaptations. Nonetheless, multiple opportunities are emerging to incorporate immunotherapies into regimens that may enhance immunogenicity while targeting immunosuppression including combinations with cytotoxic agents, antiangiogenics, and other immunotherapeutics. Critical considerations for these regimens include evaluation of complementary mechanisms of action, dose, and scheduling. In addition, effective measurement of immunogenicity through incorporation of immunocorrelative assays, as well as investigation of changes in immunosuppressive factors, will also be of great value in ascertaining why or why not these agents succeed for our patients.

Disclosure Statement

David A. Reardon, MD, has served as a speakers' bureau member for Merck and Genentech/Roche and has received research support from Celldex Pharmaceuticals and Incyte, Inc. He has also served on advisory boards for Abbvie, Amgen, Cavion, EMD Serono, Genentech/Roche, Merck, Midatech, Momenta Pharmaceuticals, Novocure, Novartis, Regeneron, and Stemline Therapeutics.

Mark R. Gilbert, MD, has served on advisory boards for Abbvie, EMD Serono, Genentech/Roche, Merck, and Cell Medica. He has received research support from GlaxoSmithKline and Genentech.

Wolfgang Wick, MD, has received research grants from Apogenix, Boehringer Ingelheim, Eli Lilly, immatics, MSD, and Roche as well as honoraria for lectures or advisory board participation from MSD and Roche. He is or has been the coordinating investigator for sponsored clinical trials evaluating APG101 (Apogenix), bevacizumab (Roche), galunisertib (Eli Lilly), temozolomide (MSD), and temsirolimus (Pfizer).

Linda Liau, MD, PhD, has received research funding from Northwest Biotherapeutics, Inc.

References

  • 1.Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–422. [DOI] [PubMed] [Google Scholar]
  • 4.Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fecci PE, Heimberger AB, Sampson JH. Immunotherapy for primary brain tumors: no longer a matter of privilege. Clin Cancer Res. 2014;20(22):5620–5629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stone HB, Peters LJ, Milas L. Effect of host immune capability on radiocurability and subsequent transplantability of a murine fibrosarcoma. J Natl Cancer Inst. 1979;63(5):1229–1235. [PubMed] [Google Scholar]
  • 7.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. [DOI] [PubMed] [Google Scholar]
  • 8.Alizadeh D, Larmonier N. Chemotherapeutic targeting of cancer-induced immunosuppressive cells. Cancer Res. 2014;74(10):2663–2668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hato SV, Khong A, de Vries IJ, Lesterhuis WJ. Molecular pathways: the immunogenic effects of platinum-based chemotherapeutics. Clin Cancer Res. 2014;20(11):2831–2837. [DOI] [PubMed] [Google Scholar]
  • 10.Formenti SC, Demaria S. Combining radiotherapy and cancer immunotherapy: a paradigm shift. J Natl Cancer Inst. 2013;105(4):256–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tang C, Wang X, Soh H, et al. Combining radiation and immunotherapy: a new systemic therapy for solid tumors? Cancer Immunol Res. 2014;2(9):831–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Shurin MR. Dual role of immunomodulation by anticancer chemotherapy. Nat Med. 2013;19(1):20–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dalgleish AG. Rationale for combining immunotherapy with chemotherapy. Immunotherapy. 2015;7(3):309–316. [DOI] [PubMed] [Google Scholar]
  • 14.Kepp O, Senovilla L, Vitale I, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 2014;3(9):e955691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Burnette BC, Liang H, Lee Y, et al. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011;71(7):2488–2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ifeadi V, Garnett-Benson C. Sub-lethal irradiation of human colorectal tumor cells imparts enhanced and sustained susceptibility to multiple death receptor signaling pathways. PLoS One. 2012;7(2):e31762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203(5):1259–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bernstein MB, Garnett CT, Zhang H, et al. Radiation-induced modulation of costimulatory and coinhibitory T-cell signaling molecules on human prostate carcinoma cells promotes productive antitumor immune interactions. Cancer Biother Radiopharm. 2014;29(4):153–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kumari A, Cacan E, Greer SF, Garnett-Benson C. Turning T cells on: epigenetically enhanced expression of effector T-cell costimulatory molecules on irradiated human tumor cells. J Immunother Cancer. 2013;1:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Baluna RG, Eng TY, Thomas CR. Adhesion molecules in radiotherapy. Radiat Res. 2006;166(6):819–831. [DOI] [PubMed] [Google Scholar]
  • 21.Kim JY, Son YO, Park SW, et al. Increase of NKG2D ligands and sensitivity to NK cell-mediated cytotoxicity of tumor cells by heat shock and ionizing radiation. Exp Mol Med. 2006;38(5):474–484. [DOI] [PubMed] [Google Scholar]
  • 22.Ridolfi L, Petrini M, Granato AM, et al. Low-dose temozolomide before dendritic-cell vaccination reduces (specifically) CD4+CD25++Foxp3+ regulatory T-cells in advanced melanoma patients. J Transl Med. 2013;11:135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Banissi C, Ghiringhelli F, Chen L, Carpentier AF. Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model. Cancer Immunol Immunother. 2009;58(10):1627–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ma Y, Adjemian S, Galluzzi L, Zitvogel L, Kroemer G. Chemokines and chemokine receptors required for optimal responses to anticancer chemotherapy. Oncoimmunology. 2014;3(1):e27663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ma Y, Adjemian S, Mattarollo SR, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729–741. [DOI] [PubMed] [Google Scholar]
  • 26.Ma Y, Mattarollo SR, Adjemian S, et al. CCL2/CCR2-dependent recruitment of functional antigen-presenting cells into tumors upon chemotherapy. Cancer Res. 2014;74(2):436–445. [DOI] [PubMed] [Google Scholar]
  • 27.Shurin MR, Naiditch H, Gutkin DW, Umansky V, Shurin GV. ChemoImmunoModulation: immune regulation by the antineoplastic chemotherapeutic agents. Curr Med Chem. 2012;19(12):1792–1803. [DOI] [PubMed] [Google Scholar]
  • 28.Galustian C, Meyer B, Labarthe MC, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol Immunother. 2009;58(7):1033–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabzevari H. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood. 2005;105(7):2862–2868. [DOI] [PubMed] [Google Scholar]
  • 30.Zhang L, Dermawan K, Jin M, et al. Differential impairment of regulatory T cells rather than effector T cells by paclitaxel-based chemotherapy. Clin Immunol. 2008;129(2):219–229. [DOI] [PubMed] [Google Scholar]
  • 31.McCoy MJ, Lake RA, van der Most RG, Dick IM, Nowak AK. Post-chemotherapy T-cell recovery is a marker of improved survival in patients with advanced thoracic malignancies. Br J Cancer. 2012;107(7):1107–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ghiringhelli F, Larmonier N, Schmitt E, et al. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol. 2004;34(2):336–344. [DOI] [PubMed] [Google Scholar]
  • 33.Schiavoni G, Mattei F, Di Pucchio T, et al. Cyclophosphamide induces type I interferon and augments the number of CD44(hi) T lymphocytes in mice: implications for strategies of chemoimmunotherapy of cancer. Blood. 2000;95(6):2024–2030. [PubMed] [Google Scholar]
  • 34.Fine JH, Chen P, Mesci A, et al. Chemotherapy-induced genotoxic stress promotes sensitivity to natural killer cell cytotoxicity by enabling missing-self recognition. Cancer Res. 2010;70(18):7102–7113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hong M, Puaux AL, Huang C, et al. Chemotherapy induces intratumoral expression of chemokines in cutaneous melanoma, favoring T-cell infiltration and tumor control. Cancer Res. 2011;71(22):6997–7009. [DOI] [PubMed] [Google Scholar]
  • 36.Lesterhuis WJ, Punt CJ, Hato SV, et al. Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice. J Clin Invest. 2011;121(8):3100–3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Alizadeh D, Trad M, Hanke NT, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014;74(1):104–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bruchard M, Mignot G, Derangere V, et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19(1):57–64. [DOI] [PubMed] [Google Scholar]
  • 39.Kodumudi KN, Woan K, Gilvary DL, Sahakian E, Wei S, Djeu JY. A novel chemoimmunomodulating property of docetaxel: suppression of myeloid-derived suppressor cells in tumor bearers. Clin Cancer Res. 2010;16(18):4583–4594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Matsumura S, Demaria S. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat Res. 2010;173(4):418–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wersall PJ, Blomgren H, Pisa P, Lax I, Kalkner KM, Svedman C. Regression of non-irradiated metastases after extracranial stereotactic radiotherapy in metastatic renal cell carcinoma. Acta Oncol. 2006;45(4):493–497. [DOI] [PubMed] [Google Scholar]
  • 42.Sampson JH, Aldape KD, Archer GE, et al. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro Oncol. 2011;13(3):324–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sanchez-Perez LA, Choi BD, Archer GE, et al. Myeloablative temozolomide enhances CD8(+) T-cell responses to vaccine and is required for efficacy against brain tumors in mice. PLoS One. 2013;8(3):e59082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dancea HC, Shareef MM, Ahmed MM. Role of Radiation-induced TGF-beta Signaling in Cancer Therapy. Mol Cell Pharmacol. 2009;1(1):44–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tsai CS, Chen FH, Wang CC, et al. Macrophages from irradiated tumors express higher levels of iNOS, arginase-I and COX-2, and promote tumor growth. Int J Radiat Oncol Biol Phys. 2007;68(2):499–507. [DOI] [PubMed] [Google Scholar]
  • 46.Kachikwu EL, Iwamoto KS, Liao YP, et al. Radiation enhances regulatory T cell representation. Int J Radiat Oncol Biol Phys. 2011;81(4):1128–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Dovedi SJ, Adlard AL, Lipowska-Bhalla G, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74(19):5458–5468. [DOI] [PubMed] [Google Scholar]
  • 48.Ellsworth S, Balmanoukian A, Kos F, et al. Sustained CD4T cell-driven lymphopenia without a compensatory IL-7/IL-15 response among high-grade glioma patients treated with radiation and temozolomide. Oncoimmunology. 2014;3(1):e27357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Campian JL, Sarai G, Ye X, Marur S, Grossman SA. Association between severe treatment-related lymphopenia and progression-free survival in patients with newly diagnosed squamous cell head and neck cancer. Head Neck. 2014;36(12):1747–1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wild AT, Ye X, Ellsworth SG, et al. The Association Between Chemoradiation-related Lymphopenia and Clinical Outcomes in Patients With Locally Advanced Pancreatic Adenocarcinoma. Am J Clin Oncol. 2015;38(3):259–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kim CH, Woo SJ, Park JS, et al. Enhanced antitumour immunity by combined use of temozolomide and TAT-survivin pulsed dendritic cells in a murine glioma. Immunology. 2007;122(4):615–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zeng J, See AP, Phallen J, 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):343–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Prins RM, Soto H, Konkankit V, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17(6):1603–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liau LM, Prins RM, Kiertscher SM, et al. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res. 2005;11(15):5515–5525. [DOI] [PubMed] [Google Scholar]
  • 55.Fong B, Jin R, Wang X, et al. Monitoring of regulatory T cell frequencies and expression of CTLA-4 on T cells, before and after DC vaccination, can predict survival in GBM patients. PloS One. 2012;7(4):e32614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ardon H, Van Gool SW, Verschuere T, et al. Integration of autologous dendritic cell-based immunotherapy in the standard of care treatment for patients with newly diagnosed glioblastoma: results of the HGG-2006 phase I/II trial. Cancer Immunol Immunother. 2012;6111:2033–2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fadul CE, Fisher JL, Hampton TH, et al. Immune response in patients with newly diagnosed glioblastoma multiforme treated with intranodal autologous tumor lysate-dendritic cell vaccination after radiation chemotherapy. J Immunother. 2011;34(4):382–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res. 2004;64(14):4973–4979. [DOI] [PubMed] [Google Scholar]
  • 59.Phuphanich S, Wheeler CJ, Rudnick JD, et al. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother. 2013;62(1):125–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Crane CA, Han SJ, Ahn B, et al. Individual patient-specific immunity against high-grade glioma after vaccination with autologous tumor derived peptides bound to the 96 KD chaperone protein. Clin Cancer Res. 2013;19(1):205–214. [DOI] [PubMed] [Google Scholar]
  • 61.Sampson JH, Archer GE, Mitchell DA, et al. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol Cancer Ther. 2009;8(10):2773–2779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sampson JH, Heimberger AB, Archer GE, et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28(31):4722–4729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wen PY, Reardon DA, Phuphanich S, et al. A randomized, double-blind, placebo-controlled phase 2 trial of dendritic cell (DC) vaccination with ICT-107 in newly diagnosed glioblastoma (GBM) patients. Paper presented at: 2014 American Society of Clinical Oncology Annual Meeting 2014; June 2014;Chicago, IL. [Google Scholar]
  • 64.Huang Y, Goel S, Duda DG, Fukumura D, Jain RK. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res. 2013;73(10):2943–2948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tartour E, Pere H, Maillere B, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev. 2011;30(1):83–95. [DOI] [PubMed] [Google Scholar]
  • 66.Griffioen AW. Anti-angiogenesis: making the tumor vulnerable to the immune system. Cancer Immunol Immunother. 2008;57(10):1553–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Johnson BF, Clay TM, Hobeika AC, Lyerly HK, Morse MA. Vascular endothelial growth factor and immunosuppression in cancer: current knowledge and potential for new therapy. Expert Opin Biol Ther. 2007;7(4):449–460. [DOI] [PubMed] [Google Scholar]
  • 68.Ohm JE, Carbone DP. VEGF as a mediator of tumor-associated immunodeficiency. Immunol Res. 2001;23(2–3):263–272. [DOI] [PubMed] [Google Scholar]
  • 69.Gabrilovich D, Ishida T, Oyama T, et al. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92(11):4150–4166. [PubMed] [Google Scholar]
  • 70.Gabrilovich DI, Chen HL, Girgis KR, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2(10):1096–1103. [DOI] [PubMed] [Google Scholar]
  • 71.Mulligan JK, Day TA, Gillespie MB, Rosenzweig SA, Young MR. Secretion of vascular endothelial growth factor by oral squamous cell carcinoma cells skews endothelial cells to suppress T-cell functions. Hum Immunol. 2009;70(6):375–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ohm JE, Gabrilovich DI, Sempowski GD, et al. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood. 2003;101(12):4878–4886. [DOI] [PubMed] [Google Scholar]
  • 73.Huang X, Raskovalova T, Lokshin A, et al. Combined antiangiogenic and immune therapy of prostate cancer. Angiogenesis. 2005;8(1):13–23. [DOI] [PubMed] [Google Scholar]
  • 74.Huang Y, Yuan J, Righi E, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA. 2012;109(43):17561–17566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Miyazaki J, Tsuzuki Y, Matsuzaki K, et al. Combination therapy with tumor-lysate pulsed dendritic cells and antiangiogenic drug TNP-470 for mouse pancreatic cancer. Int J Cancer. 2005;117(3):499–505. [DOI] [PubMed] [Google Scholar]
  • 76.Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res. 2010;70(15):6171–6180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.U.S. Department of Health and Human Services CfDE, and Research FaDa: FDA Project on Cancer Drug Aproval Endpoint. Last updated April 20, 2007.
  • 78.Li B, Lalani AS, Harding TC, et al. Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy. Clin Cancer Res. 2006;12(22):6808–6816. [DOI] [PubMed] [Google Scholar]
  • 79.Manning EA, Ullman JG, Leatherman JM, et al. A vascular endothelial growth factor receptor-2 inhibitor enhances antitumor immunity through an immune-based mechanism. Clin Cancer Res. 2007;13(13):3951–3959. [DOI] [PubMed] [Google Scholar]
  • 80.Hodi FS, Lawrence D, Lezcano C, et al. Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma. Cancer Immunol Res. 2014;2(7):632–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Reardon D, Schuster J, Tran D, et al. ReACT: A phase II study of rindopepimut vaccine (CDX-110) plus bevacizumab in relapsed glioblastoma. Paper presented at: 19th Annual Meeting of the Society for Neuro-Oncology 2014; November 2014: Miami, FL. [Google Scholar]
  • 82.Brennan CW, Verhaak RG, McKenna A, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Johnson BE, Mazor T, Hong C, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science. 2014;343(6167):189–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wick W, Weller M, Weiler M, Batchelor T, Yung AW, Platten M. Pathway inhibition: emerging molecular targets for treating glioblastoma. Neuro Oncol. 2011;13(6):566–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wen PY, Yung WK, Mellinghoff IK, et al. Phase II trial of phosphatidylinositol-3 kinase (PI3K) inhibitor buparlisib (BKM120) in recurrent glioblastoma. Paper presented at: 2014 American Society of Clinical Oncology Annual Meeting 2014; June 2014; Chicago, IL. [Google Scholar]
  • 86.Fritsch EF, Rajasagi M, Ott PA, Brusic V, Hacohen N, Wu CJ. HLA-binding properties of tumor neoepitopes in humans. Cancer Immunol Res. 2014;2(6):522–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74. [DOI] [PubMed] [Google Scholar]
  • 88.Castle JC, Kreiter S, Diekmann J, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72(5):1081–1091. [DOI] [PubMed] [Google Scholar]
  • 89.Yadav M, Jhunjhunwala S, Phung QT, et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature. 2014;515(7528):572–576. [DOI] [PubMed] [Google Scholar]
  • 90.Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515(7528):577–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Tran E, Turcotte S, Gros A, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science. 2014;344(6184):641–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Brown SD, Warren RL, Gibb EA, et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014;24(5):743–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fecci PE, Mitchell DA, Whitesides JF, et al. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006;66(6):3294–3302. [DOI] [PubMed] [Google Scholar]
  • 96.Waziri A. Glioblastoma-derived mechanisms of systemic immunosuppression. Neurosurg Clin N Am. 2010;21(1):31–42. [DOI] [PubMed] [Google Scholar]
  • 97.Avril T, Vauleon E, Tanguy-Royer S, Mosser J, Quillien V. Mechanisms of immunomodulation in human glioblastoma. Immunotherapy. 2011;3(4 Suppl):42–44. [DOI] [PubMed] [Google Scholar]
  • 98.Dix AR, Brooks WH, Roszman TL, Morford LA. Immune defects observed in patients with primary malignant brain tumors. J Neuroimmunol. 1999;100(1–2):216–232. [DOI] [PubMed] [Google Scholar]
  • 99.Wei J, Barr J, Kong LY, et al. Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway. Mol Cancer Ther. 2010;9(1):67–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Roth P, Mittelbronn M, Wick W, Meyermann R, Tatagiba M, Weller M. Malignant glioma cells counteract antitumor immune responses through expression of lectin-like transcript-1. Cancer Res. 2007;67(8):3540–3544. [DOI] [PubMed] [Google Scholar]
  • 101.Hussain SF, Yang D, Suki D, Aldape K, Grimm E, Heimberger AB. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro Oncol. 2006;8(3):261–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Tran Thang NN, Derouazi M, Philippin G, et al. Immune infiltration of spontaneous mouse astrocytomas is dominated by immunosuppressive cells from early stages of tumor development. Cancer Res. 2010;70(12):4829–4839. [DOI] [PubMed] [Google Scholar]
  • 103.Hishii M, Nitta T, Ishida H, et al. Human glioma-derived interleukin-10 inhibits antitumor immune responses in vitro. Neurosurgery. 1995;37(6):1160–1166; discussion 1166–1167. [DOI] [PubMed] [Google Scholar]
  • 104.Opitz CA, Litzenburger UM, Sahm F, et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011;478(7368):197–203. [DOI] [PubMed] [Google Scholar]
  • 105.Schartner JM, Hagar AR, Van Handel M, Zhang L, Nadkarni N, Badie B. Impaired capacity for upregulation of MHC class II in tumor-associated microglia. Glia. 2005;51(4):279–285. [DOI] [PubMed] [Google Scholar]
  • 106.Facoetti A, Nano R, Zelini P, et al. Human leukocyte antigen and antigen processing machinery component defects in astrocytic tumors. Clin Cancer Res. 2005;11(23):8304–8311. [DOI] [PubMed] [Google Scholar]
  • 107.Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, et al. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. 2008;14(16):5166–5172. [DOI] [PubMed] [Google Scholar]
  • 108.El Andaloussi A, Lesniak MS. An increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro Oncol. 2006;8(3):234–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Crane CA, Ahn BJ, Han SJ, Parsa AT. Soluble factors secreted by glioblastoma cell lines facilitate recruitment, survival, and expansion of regulatory T cells: implications for immunotherapy. Neuro Oncol. 2012;14(5):584–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Jacobs JF, Idema AJ, Bol KF, et al. Prognostic significance and mechanism of Treg infiltration in human brain tumors. J Neuroimmunol. 2010;225(1–2):195–199. [DOI] [PubMed] [Google Scholar]
  • 111.Wainwright DA, Sengupta S, Han Y, Lesniak MS. Thymus-derived rather than tumor-induced regulatory T cells predominate in brain tumors. Neuro Oncol. 2011;13(12):1308–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Badie B, Schartner J. Role of microglia in glioma biology. Microsc Res Tech. 2001;54(2):106–113. [DOI] [PubMed] [Google Scholar]
  • 113.Li W, Graeber MB. The molecular profile of microglia under the influence of glioma. Neuro Oncol. 2012;14(8):958–978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Walker DG, Chuah T, Rist MJ, Pender MP. T-cell apoptosis in human glioblastoma multiforme: implications for immunotherapy. J Neuroimmunol. 2006;175(1–2):59–68. [DOI] [PubMed] [Google Scholar]
  • 115.Jansen T, Tyler B, Mankowski JL, et al. FasL gene knock-down therapy enhances the antiglioma immune response. Neuro Oncol. 2010;12(5):482–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wei J, Wu A, Kong LY, et al. Hypoxia potentiates glioma-mediated immunosuppression. PloS One. 2011;6(1):e16195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lemke D, Pfenning PN, Sahm F, et al. Costimulatory protein 4IgB7H3 drives the malignant phenotype of glioblastoma by mediating immune escape and invasiveness. Clin Cancer Res. 2012;18(1):105–117. [DOI] [PubMed] [Google Scholar]
  • 119.Wainwright DA, Chang AL, Dey M, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4 and PD-L1 in mice with brain tumors. Clin Cancer Res. 2014;20(20):5290–5301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Fecci PE, Ochiai H, Mitchell DA, et al. Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4+ T cell compartment without affecting regulatory T-cell function. Clin Cancer Res. 2007;13(7):2158–2167. [DOI] [PubMed] [Google Scholar]
  • 121.Reardon D, Gokhale P, Ligon K, et al. Immune checkpoint blockade for glioblastoma: preclinical activity of single agent and combinatorial therapy. Paper presented at: 19th Annual Meeting of the Society for Neuro-Oncology ; November 2014; Miami, FL. [Google Scholar]
  • 122.Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA. 2010;107(9):4275–4280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Rodon J, Carducci MA, Sepulveda-Sanchez JM, et al. First-in-human dose study of the novel transforming growth factor-beta receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma. Clin Cancer Res. 2015;21(3):553–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Brandes AA, Carpentier AF, Kesari S, et al. A phase II study of galunisertib monotherapy or galunisertib plus lomustine compared to lomustine monotherapy in recurrent glioblastoma. Paper presented at: 2015 American Society of Clinical Oncology Annual Meeting; June 2015; Chicago, IL. [Google Scholar]
  • 126.Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116(7):1935–1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210(9):1695–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Son CH, Bae JH, Shin DY, et al. Ctla-4 blockade enhances antitumor immunity of intratumoral injection of immature dendritic cells into irradiated tumor in a mouse colon cancer model. J Immunother. 2014;37(1):1–7. [DOI] [PubMed] [Google Scholar]
  • 129.Karyampudi L, Lamichhane P, Scheid AD, et al. Accumulation of Memory Precursor CD8T Cells in Regressing Tumors Following Combination Therapy with Vaccine and Anti-PD-1 Antibody. Cancer Res. 2014;74(11):2974–2985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Fu J, Malm IJ, Kadayakkara DK, Levitsky H, Pardoll D, Kim YJ. Preclinical evidence that PD-1 blockade cooperates with cancer vaccine TEGVAX to elicit regression of established tumors. Cancer Res. 2014;74(15):4042–4052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Hodi FS, Butler M, Oble DA, et al. Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients. Proc Natl Acad Sci USA. 2008;105(8):3005–3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Hodi FS, Mihm MC, Soiffer RJ, et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA. 2003;100(8):4712–4717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Le DT, Lutz E, Uram JN, et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J Immunother. 2013;36(7):382–389. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press

RESOURCES