Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2016) Volume 7, Issue 5

Immunotherapy for Glioblastoma

Debebe Theodros, Dane Moran, Tomas Garzon-Muvdi and Michael Lim*
Department of Neurosurgery, Johns Hopkins University School of Medicine, The Johns Hopkins Hospital, The Johns Hopkins University, Baltimore, Maryland, USA
*Corresponding Author: Michael Lim, Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Phipps Suite 123, Baltimore, MD 21287, USA, Tel: 410-502-1627, Fax: 410-502-4954 Email:

Abstract

Glioblastoma (GBM) is the most common primary malignant brain cancer with a dismal prognosis in spite of aggressive treatment options. Although once thought to be an “immune-privileged” site, recent advances have begun to highlight the complex interaction between the immune system and the central nervous system. Thus, great interest has emerged in the ability of immunotherapy to potentially prolong the survival of patients suffering from GBM. Indeed, numerous clinical trials have demonstrated durable responses in late stage disease, as well as, among patients with brain metastasis. A variety of approaches to modulating the immune system exist and their efficacy are currently being investigated in various clinical trials. Here we provide a brief overview of neuroimmunology and explore the various approaches towards priming the immune system against GBM.

Keywords: Glioblastoma; Immunotherapy; Microglia; Tumor; Vaccine

Introduction

Recent scientific advances have solidified the role of the immune system in maintaining central nervous system (CNS) homeostasis. New insight into the dynamic interrogation of the CNS by the immune system reveals a dynamic interaction contrary to previously held notions that the brain is an immune sanctuary [1-4]. Significant advances using preclinical models of CNS autoimmune disease or infection have revealed clues as to the extent of immune surveillance occurring within the CNS. As such, new efforts are currently underway to better understand the immune response to primary and metastatic malignancy of the CNS. Indeed, a number of preclinical models suggest immunotherapy represents a potentially promising treatment modality for patients suffering from primary brain cancer [5-8]. Immunotherapeutic strategies to overcoming immunosuppression within the tumor microenvironment (TME) and restoring cytotoxic CD8+ T-cell responses include vaccine therapies, adoptive cell therapy, and immune checkpoint blockade among others. Here, we present a brief over of CNS immunology, strategies to implementing immunotherapy as a treatment modality for GBM and future directions.

Glioblastoma

Glioblastoma is the most prevalent adult malignant brain tumor with a median survival of less than two years and a 5-year overall survival of less than 10% [9-11]. Current standard of care (SOC) includes maximal-safe resection, chemotherapy and radiation therapy [11]. Furthermore, GBM is an inherently heterogeneous disease associated with extensive infiltration making complete cure challenging as patients ultimately succumb to recurrence [12]. However, the limits of conventional therapies may be overcome by modulating the host immune response to cancer. Great strides have been made towards re-purposing the immune system to eliminate CNS malignancy.

Central Nervous System Immunology

Immune cells of the CNS

The healthy CNS parenchyma is home to only one immune cell population, the microglia, which are highly specialized macrophages [13]. Microglia are distinct from peripheral monocytes or macrophages as they originate from a yolk sac progenitor and are maintained via local proliferation without reconstitution from the bone marrow [14,15]. However, myeloid cells are present within the CNS as well, specifically within the meninges, choroid plexus (CP), and perivascular spaces and are maintained by peripheral blood monocytes [14-16]. Despite the lack of resident T cells within the CNS parenchyma, the cellular composition of CSF is overwhelmingly lymphocytic, with ~90% of cells within circulating CSF being T cells. Moreover, the CD4+ to CD8+ ratio is 3.5 to 1 with the vast majority of CD4+ cells being central or effector memory T cells [17-19].

“Immune-privilege”

Nearly a century of work suggested the CNS is a site of “immune privilege,” a term first coined by Billingham and Boswell, which was a concept based up the observation that direct administration of antigens does not elicit an adaptive immune response [20,21]. However, the precise definition of “immune privilege” decayed with time and was recently re-defined [21]. CNS immune privilege is compartmentalized to the parenchyma, as intracerebroventricular (ICV) injection of various antigens results in generation of both humoral and cytotoxic T-cell responses [22]. Similarly, innate immune responses in the CNS are limited to the ventricles as well as the CP, and meninges [23]. Drainage of interstitial fluid to the cerebrospinal fluid (CSF) provides meningeal, perivascular and choroid plexus macrophages the ability to constantly survey potential antigens present within the parenchyma [24]. Furthermore, recent work clearly demonstrates direct connections between the CNS and deep cervical lymph nodes via lymphatic drainage creating the ability to generate immune responses peripherally [1,2]. Thus, the CNS is an immunologically active organ displaying the necessary anatomical structures to undergo immunosurveillance and potentially benefit from immunotherapy.

Immune Evasion

Despite the clear role of immunosurveillance in maintaining and preserving normal brain architecture and function, multiple mechanisms exist within the tumor microenvironment (TME) to stifle an effective immune response. These mechanisms include the hypoxic microenvironment itself, the ability of tumor cells to secrete highly immunosuppressive factors, decreased expression of major histocompatibility complex (MHC) upon various APC subsets, inhibition of lymphocyte activity through increased surface expression of co-inhibitory immune checkpoint molecules, and recruitment of immunosuppressive cells to the TME. Here, we briefly review the known mechanisms of immunosuppression within the GBM TME.

The relative importance of immunosuppressive cells within GBMs is becoming rapidly apparent. One such population includes regulatory T cells (Tregs) commonly defined as CD4+FoxP3+CD25+ T cells, which are crucial under homeostatic conditions for maintaining tolerance; however, have been readily identified in human GBM samples [25]. These Tregs seem to be thymic-derived; however, the blockade of the CC chemokine receptor 4 (CCR4), a major chemoattractant receptor, does not completely deplete Treg infiltration within the TME, suggesting other mechanisms of Treg chemoattraction to the TME [26,27]. Furthermore, abundance of Tregs within the TME has been shown to be associated with a poor prognosis [28-30]. Another cellular subset playing a role in maintaining a highly immunosuppressive TME are innate immune cells constituting tumor-associated macrophages (TAMs) and microglia. Factors such as colony-stimulating factor 1 (CSF-1), transforming growth factor-β (TGF-β), macrophage inhibitory cytokine-1 (MIC-1) and IL-10 recruit macrophages to the TME and shift polarization of recruited macrophages towards an M2 phenotype, decreasing phagocytosis while inhibiting cytotoxic T cell activity and enhancing Treg immunosuppression [31-35]. Additionally, TAMs and microglia influence GBM angiogenesis, growth, and invasion via secretion of endothelial growth factor (EGF), TGF-β, IL-6, CSF-1 and matrix metalloproteinases [32,36-39]

The TME itself is a highly immunosuppressive environment capable of inhibiting anti-tumor immune mediated responses through a variety of mechanisms. One such mechanism is the production of immunosuppressive cytokines, which induce immunosuppressive responses within the TME. One potent cytokine produced by GBM cells is IL-10, which enhances tumor growth while decreasing interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and MHC II expression, stifling anti-tumor immune responses [40-44]. Additionally, intense neovascularization, abnormal blood flow, and preferential oxygen consumption by rapidly proliferating tumor cells results in a hypoxic TME and activation of the STAT-3 inhibitory pathway within immune cells. Hypoxia induces numerous changes within the TME including the expansion of M2 TAMs and Tregs, which induce further vascularization and tumor cell invasion in a feed-forward manner as a result of STAT-3 mediated hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) expression [45]. Epigenetic alterations resulting in gain-of-function mutations promote natural killer (NK) cell-mediated lysis [46].

Immunotherapy Approaches

The SOC, dubbed the “Stupp Protocol,” involves radiotherapy plus concomitant daily Temozolomide (TMZ) at 75 mg/BSA2/day for 7 days a week throughout radiation, followed by six cycles of adjuvant TMZ dosed 150-200 mg/m2 for 5 days during each 28-day cycle based upon the landmark study by Stupp et al. [11]. This study demonstrated a significant increase in median survival from 12.1 months to 14.6 months with the addition of temozolomide to radiation therapy. Additionally, the two-year survival rate following radiation with temozolomide versus radiation alone was 26.5% vs. 10.4%, respectively. However, the vast majority of patients ultimately succumb to disease. Neoplastic invasion of glioma stem cells beyond radiographically defined tumor margins and present after gross total resection undergo selection for alkylating/radiation-resistant clones following SOC [47,48]. Furthermore, the immense heterogeneity of glioma stem cells as illustrated by the capability to differentiate into various cell types, as well as, unique molecular profiles such as presence of mutations to isocitrate dehydrogenase (IDH), O6-methylguanine-DNA methyltransferase (MGMT), and EGFR status further dictate response to treatment and prognosis. Thus, there is growing interest in novel treatments for GBM. Immunotherapy represents a potentially promising modality as early success has been demonstrated in a variety of solid malignancies [49,50].

Vaccine Therapy

GBM heterogeneity necessitates the need for patient-specific, antitumor immunotherapies with minimal toxicity. Strategies involving vaccination against tumor-associated antigens (TAA) have yielded success, as demonstrated by the FDA approved Gardasil® (Merck, NJ, USA) for cervical cancer and sipuleucel-T (Provenge®; Dendreon, WA, USA) for hormone-resistant metastatic prostate cancer [51]. Extensive efforts are underway to understand the potential role of vaccine therapy for GBMs are currently underway (Table 1). Here we discuss the various types of vaccines and their efficacy for GBMs.

NCT number Title Agent Phase Outcome measures
NCT00643097 Vaccine Therapy in Treating Patients With Newly Diagnosed Glioblastoma Multiforme PEP-3 vaccine + sargramostim + Temozolomide Phase 2 Humoral and Cellular Immune Response|Clinical Efficacy of Vaccination, in Terms of Progression-free Survival (PFS)|Response to Vaccination|Toxicity to PEP-3 Vaccine Immunization
NCT00639639 Vaccine Therapy in Treating Patients With Newly Diagnosed Glioblastoma Multiforme tetanus toxoid + therapeutic autologous dendritic cells and therapeutic autologous lymphocytes Phase 1 Feasibility and safety of vaccination with cytomegalovirus pp65-LAMP mRNA-loaded dendritic cells (DCs) with or without autologous lymphocyte transfer|Humoral and cellular immune responses|Time to progression|Differential ability of indium In-111-labeled DCs to track to the inguinal lymph nodes under different skin preparative conditions|Differential ability of indium In-111-labeled DCs to track to lymph nodes on the tumor bearing and non-tumor bearing side of the cervical lymph nodes|Immunologic cell infiltrate in recurrent tumors|Evidence of antigen-escape outgrowth in recurrent or progressive tumors
NCT02772094 Dendritic Cell-Based Tumor Vaccine Adjuvant Immunotherapy of Human Glioblastoma Multiforme (WHO Grade IV Gliomas) Dendritic Cell Vaccine Phase 2 Overall survival with measures of medium survival period (in days) and annual survival rates (in %)|Adverse effects, acute and chronic, assessed according to NCI CTCAE Version 3|Disease progression-free period
NCT00626483 Basiliximab in Treating Patients With Newly Diagnosed Glioblastoma Multiforme Undergoing Targeted Immunotherapy and Temozolomide-Caused Lymphopenia RNA-loaded dendritic cell vaccine + basiliximab Phase 1 Functional capacity of CD4+,CD25+, CD127- T-regulatory cells|Safety
NCT00890032 Vaccine Therapy in Treating Patients Undergoing Surgery for Recurrent Glioblastoma Multiforme BTSC mRNA-loaded DCs Phase 1 Feasibility and safety|Humoral and cellular immune responses
NCT01290692 Study To Test the Safety and Efficacy of TVI-Brain-1 As A Treatment for Recurrent Grade IV Glioma TVI-Brain-1 Phase 2 Progression Free Survival|Overall Survival|Quality of life|Toxicity|Time to progression|Objective response rate|Cancer immunogenicity
NCT01204684 Dendritic Cell Vaccine for Patients With Brain Tumors autologous tumor lysate-pulsed DC vaccination ± 0.2% resiquimod ± adjuvant polyICLC Phase 2 Most effective combination of DC vaccine components|Time to tumor progression and overall survival
NCT01522820 Vaccine Therapy With or Without Sirolimus in Treating Patients With NY-ESO-1 Expressing Solid Tumors DEC-205/NY-ESO-1 Fusion Protein CDX-1401+ Sirolimus Phase 1 Incidence of adverse events in patients receiving the DEC-205/NY-ESO-1 fusion protein CDX-1401 with and without sirolimus, as evaluated according to the NCI CTCAE scale version 4.0|NY-ESO-1 specific cellular immunity|NY-ESO-1 specific humoral immunity
NCT00458601 Phase II Study of Rindopepimut (CDX-110) in Patients With Glioblastoma Multiforme CDX-110 with GM-CSF + temozolomide Phase 2 Progression-free survival status|Safety and tolerability characterized by adverse events (term, grade, frequency).|Safety and tolerability characterized by physical examinations.|Safety and tolerability characterized by hematologic and metabolic panel (including CBC with differential, electrolytes, BUN, Cr, liver associated enzymes).|Safety and tolerability characterized by urinalysis.|Safety and tolerability characterized by vital signs.|Immune response; T-cell response to vaccine.|Immune response; antibody response to vaccine.|Immune response; HLA typing.|Overall survival.
NCT00045968 Study of a Drug [DCVax®-L] to Treat Newly Diagnosed GBM Brain Cancer Dendritic cell immunotherapy Phase 3 The primary objective of this study is to compare progression free survival from time of randomization between patients treated with DCVax-L and control patients.|The secondary objective is to compare overall survival and time to disease progression between DCVax-L treated and control patients.
NCT01400672 Imiquimod/Brain Tumor Initiating Cell (BTIC) Vaccine in Brain Stem Glioma Tumor Lysate Vaccine + Imiquimod + Radiation therapy Phase 1 Dose-limiting toxicity|Time to Tumor Progression|Drop-out rate
NCT01498328 A Study of Rindopepimut/GM-CSF in Patients With Relapsed EGFRvIII-Positive Glioblastoma Bevacizumab+ Rindopepimut (CDX-110) with GM-CSF Phase 2 Groups 1 and 2: Progression-free survival rate|Group 2C: Objective Response Rate|Safety and Tolerability|Anti-tumor activity|EGFRvIII-specific immune response
NCT01480479 Phase III Study of Rindopepimut/GM-CSF in Patients With Newly Diagnosed Glioblastoma Rindopepimut (CDX-110) with GM-CSF|Drug: Temozolomide|Drug: KLH Phase 3 Overall Survival|Progression-free survival|Safety and Tolerability
NCT01222221 Vaccine Therapy, Temozolomide, and Radiation Therapy in Treating Patients With Newly Diagnosed Glioblastoma Multiforme Glioblastoma multiforme multipeptide vaccine IMA950 + sargramostim + temozolomide + radiation therapy Phase 1 Causality of each adverse event (AE) to glioblastoma multiform multi-antigen vaccine IMA950 and GM-CSF and AE severity according to NCI CTCAE Version 4.0|Total number of patients showing patient-individual T-cell responses against a single or multiple tumor-associated peptides (TUMAP) contained in the study vaccine IMA950 at one or more post-vaccination time points by HLA multimer analysis|Progression-free survival (PSF) at 6 and 9 months post-surgery as assessed by the Macdonald criteria from conventional gadolinium-enhanced MRI and clinical assessment|Correlation between steroid levels and observed T-cell responses|Correlation between O6-methyl-DNA-methyltransferase (MGMT) promoter methylation status in tumor tissue using methylation-specific polymerase chain reaction and clinical benefit (PFS at 6 months and 9 months)|Kinetics of vaccine-induced TUMAP responses including summary descriptions of the time of onset, sustainability, and magnitude of the observed response
NCT00323115 Phase II Feasibility Study of Dendritic Cell Vaccination for Newly Diagnosed Glioblastoma Multiforme Autologous Dendritic Cell vaccine + Temozolomide + Radiotherapy Phase 2 Tumor-specific Cytotoxic T-cell Response|Feasibility and Toxicity Profile of Intra-nodal DC/Tumor Lysate Vaccination|Progression Free Survival (PFS)and Overall Survival (OS) Comparison to Prognostic Matched Historical Controls|Immunological Parameters With PFS vs Overall Survival|Radiological Response When There is Residual Enhancing Tumor at Baseline MRI
NCT01006044 Efficacy & Safety of Autologous Dendritic Cell Vaccination in Glioblastoma Multiforme After Complete Surgical Resection Autologous dendritic cells Phase 2 Evaluation of the treatment impact on progression-free survival|Safety evaluation|Evaluation of impact on other efficiency clinical parameters|Study of specific immune response and correlates with clinical outcome|Cell line characterization and correlate the final product with clinical efficacy
NCT00626015 Chemotherapy, Radiation Therapy, and Vaccine Therapy With Basiliximab in Treating Patients With Glioblastoma Multiforme That Has Been Removed by Surgery PEP-3-KLH conjugate vaccine + daclizumab + temozolomide Phase 1 Functional suppressive capacity of CD4+CD25+CD127- T-regulatory cells|Comparison of proliferative T-cell response to phytohemagglutinin (PHA) among treatment groups (with versus without daclizumab/basiliximab)
NCT00576537 Tumor Lysate Pulsed Dendritic Cell Immunotherapy for Patients With Brain Tumors Dendritic Cell Vaccine Immunotherapy Phase 2 Evaluate the safety/toxicity of subcutaneous injections of autologous dendritic cells
NCT00905060 HSPPC-96 Vaccine With Temozolomide in Patients With Newly Diagnosed GBM HSPPC-96 Phase 2 To evaluate the safety profile of HSPPC-96 administered concurrently temozolomide in patients with newly diagnosed GBM.|Survival Time|To evaluate the immunologic response to vaccine treatment|Progression free survival from date of surgical resection
NCT01081223 Phase I/II Study To Test The Safety and Efficacy of TVI-Brain-1 As A Treatment For Recurrent Grade IV Glioma Cancer vaccine plus immune adjuvant, plus activated white blood cells Phase 1|Phase 2 Relative toxicity|Progression free survival|Immunogenicity|Overall survival
NCT00846456 Safe Study of Dendritic Cell (DC) Based Therapy Targeting Tumor Stem Cells in Glioblastoma Dendritic cell vaccine with mRNA from tumor stem cells Phase 1|Phase 2 Adverse events|Evaluation of immunological response, time to disease progression and survival time
NCT00576641 Immunotherapy for Patients With Brain Stem Glioma and Glioblastoma autologous dendritic cells Phase 1 Evaluate safety/toxicity of Dendritic cell vaccine, Monitor survival and time to progression and monitor the cellular immune responses.
NCT01213407 Dendritic Cell Cancer Vaccine for High-grade Glioma Trivax, Temozolomide, Surgery, Radiotherapy Phase 2 Progression free survival|Quality of Life|Progression free survival at 18 and 24 months|Overall survival
NCT00612001 Vaccine Therapy in Treating Patients With Malignant Glioma Glioma-associated antigen peptide-pulsed autologous dendritic cell vaccine Phase 1 Dose-limiting toxicity and maximum tolerated dose of autologous dendritic cells pulsed with synthetic glioma-associated antigen (GAA) peptides|Survival|Tumor progression
NCT00069940 Vaccine Therapy and Sargramostim in Treating Patients With Sarcoma or Brain Tumor Sargramostim + telomerase + 540-548 peptide vaccine Phase 1  
NCT00003185 Biological Therapy in Treating Patients With Glioblastoma Multiforme Autologous tumor cell vaccine + sargramostim +  tumor-draining lymph node lymphocyte therapy + cyclophosphamide + conventional surgery Phase 2  
NCT01171469 Vaccination With Dendritic Cells Loaded With Brain Tumor Stem Cells for Progressive Malignant Brain Tumor Dendritic Cells + Imiquimod Phase 1 Maximum Tolerated Dose|Time to Tumor Progression
NCT00014573 Chemotherapy and Vaccine Therapy Followed by Bone Marrow or Peripheral Stem Cell Transplantation and Interleukin-2 in Treating Patients With Recurrent or Refractory Brain Cancer aldesleukin + autologous tumor cell vaccine + filgrastim + sargramostim + therapeutic autologous lymphocytes + cisplatin + cyclophosphamide + paclitaxel + autologous bone marrow transplantation + conventional surgery + peripheral blood stem cell transplantation Phase 2  
NCT00293423 GP96 Heat Shock Protein-Peptide Complex Vaccine in Treating Patients With Recurrent or Progressive Glioma HSPPC-96 Phase 1|Phase 2 Safety and maximum tolerated dose|Frequency of gp96 heat shock protein-peptide complex vaccine (Phase I [closed to accrual as of 7/25/2007])|Toxicity (Phase I [closed to accrual as of 7/25/2007])|Progression-free survival at 6 months (Phase II)|Immunological response (Phase I [closed to accrual as of 7/25/2007])|Safety (Phase II)|Tumor response as measured by neuro-imaging and neurologic exam (Phase II)|Survival (Phase II)|Immunological response (Phase II)
NCT00068510 Vaccine Therapy in Treating Patients With Malignant Glioma Therapeutic autologous dendritic cells Phase 1 Dose Limiting Toxicity|Time to tumor progression, overall survival and cellular immune responses in brain tumor patients injected with tumor lysate pulsed dendritic cells
NCT00004024 Biological Therapy Following Surgery and Radiation Therapy in Treating Patients With Primary or Recurrent Astrocytoma or Oligodendroglioma Aldesleukin + autologous tumor cell vaccine + muromonab-CD3 + sargramostim + therapeutic autologous lymphocytes + surgical procedure + radiation therapy Phase 2  

Table 1: Vaccine-based clinical trials for GBM. Source: clinicaltrials.gov.

Peptide vaccines

Peptide vaccines represent a platform of immunotherapy consisting of TAAs in combination with an adjuvant to prime T cells to mount an anti-tumor immune-mediated response. TAAs are uptaken by antigen-presenting cells (APCs), internally processed and mounted on MHC I or II and ultimately recognized by the cognate T cell receptor on CD8+ or CD4+ T cells, respectively [52]. Thus, identification of unique TAA and not over-expressed endogenous peptides predicts the success of potential peptide vaccines. Despite the identification of multiple TAAs such as, HER-2, gp-100, MAGE-1, AIM-2, and IL-13Rα2 in a variety of tumors, endogenous expression of these targets explains the presence of non-reactive T cells in patients [53]. One promising target, aberrant EGF receptors (EGFR) has been shown to regulate cell proliferation, differentiation, survival and invasiveness in multiple tumor types, including GBM [54-59]. One such variant, EGFRvIII, is selectively expressed on 27-67% of GBMs, representing a potential target for peptide vaccine therapy [59,60].

Based upon the EGFRvIII discovery, a Phase II multicenter trial termed the ACTIVATE trial was initiated. The ACTIVATE trial involved use of the PEPvIII-KLH peptide in combination with granulocyte macrophage-colony stimulating factor (GM-CSF) without pulsed autologous DCs. The ACTIVATE trial enrolled 19 patients with newly-diagnosed, EGFRvIII positive GBMs who underwent gross total resection and standard of care radiation and TMZ treatment. Patients underwent three biweekly intradermal injections at the upper thigh followed by monthly injections until radiographic progression or death. The median time-to-progression (TTP) was 12 months vs. a TTP of 7.1 months for historical controls (p=0.0058). Furthermore, ex vivo analysis demonstrated humoral responses as well as antigen-specific responses to PEPvIII and EGFRvIII which predicted median OS. The median time-to-progression (TTP) was 12 months, (p =0.0058). Pathological samples obtained from recurrent tumors were negative for EGFRvIII via immunohistochemical staining (IHC) in 82% of samples, which the authors attributed to immunoediting following vaccination [61].

Following the adoption of the Stupp protocol as SOC, the ACTIVATE (ACT) II trial was initiated. The ACT II trial enrolled 21 patients with EGFRvIII positive GBMs to receive CDX-110 (rindopepimut and GM-CSF) within 6 weeks of completion of SOC radiation and chemotherapy, followed by an additional two doses at two week intervals, then monthly vaccination until disease progression. Despite Grade 2 TMZ-related lymphopenia, similar clinical benefits were observed with a median TTP of 15.2 months and an OS of 23.6 months [62,63]. The ACT III trial, a multicenter, singlearm, phase II study, sought to confirm the results in a large, multicenter study. A total of 65 patients were enrolled and received Rindopepimut following SOC Stupp protocol. The median OS was 21.8 months with a 36-month OS of 26%, confirming the results of the previous trials [64]. With encouraging results, the ACT IV trial was initiated. This randomized, double-blind phase IV study enrolled 745 patients to either SOC and rindopepimut with GM-CSF versus SOC and KLH injection alone. Despite promising results in previous trials, the ACT IV trial was discontinued in March, 2016 based upon preliminary results revealing the control arm significantly outperforming the vaccine arm (hazard ratio=0.99, median OS: Rindopepimut 20.4 months vs. control 21.1 months). The ReACT trial, is a Phase II, randomized, double-blind trial currently underway evaluating Rindopepimut/GM-CSF vaccine therapy and bevacizumab treatment in currently 125 EGFRvIII positive recurrent GBM patients (NCT01498328). Results revealed in November 2015 demonstrated a significant benefit in OS with 25% of patients in the rindopepimut arm alive at 2 years versus 0% in the control arm.

Dendritic cell (DC) vaccines

Dendritic cells, termed “professional” APCs function as critical mediators of immune surveillance, antigen presentation, and cross talk between the innate and adaptive immune system. Recognition of pathogen-associated molecular patterns (PAMPs) results in internalization of foreign proteins/peptides, internal processing and extracellular presentation in the context of MHC I or II and migration/ activation of DCs to local draining lymph nodes and initiation of an adaptive immune response. Thus, enhanced priming of CD4+ and CD8+ T cells using DC vaccine platforms represent another interesting avenue of cancer immunotherapy.

The VICTOR I trial was a Phase I study with 12 patients vaccinated with autologous DCs pulsed with Rindopepimut (CDX-110; Celldex Therapeutics, MA, USA), a PEPvIII peptide conjugated to keyhole limet hemocyanin (KLH). Of note, expression of EGFRvIII expression was not an inclusion criterion; yet, twelve patients received three equal intradermal doses every two weeks. No patient suffered any serious adverse event greater than Grade II, with ex vivo analysis demonstrating evidence of antigen-specificity and a humoral response. The median progression free survival (PFS) was 10.2 months with an overall survival (OS) of 22.8 months. Despite a statistically insignificant increase in survival, the results of the VICTOR I trial provided evidence that a peptide-based vaccine may prove beneficial in patients with GBMs [61].

The ICT-107 vaccine, developed by Immunocellular Therapeutics Ltd. (CA, USA) is an autologous DC vaccine with activity against six antigens including AIM-2, GP100, IL13Rα2, HER2, MAGE-1 and TRP-2 and demonstrated clinical activity in a Phase I trial. The phase I trial consisted of 21 patients (17 newly-diagnosed GBM patients) with a PFS of 16.9 months and median OS of 38.4 months [53]. A Phase II, randomized, double-blind study of ICT-107 failed to meet the primary OS survival of 2-3 years among the ICT-107 group but did meet the secondary PFS outcome of 2-3 years. Based upon this work, a Phase III trial is currently underway actively recruiting patients (NCT02546102). Additionally, a Phase I trial investigating the therapeutic potential of ICT-121 (Immunocellular Therapeutics, Ltd.) in recurrent GBM is also underway actively recruiting patients (NCT02049489).

The DCVax platform (Northwest Biotherapeutics, Inc. MD, USA) is a DC-based vaccine platform currently in numerous trials for a variety of malignancies including GBM. Three different DCVax platforms exist, two involve purifying autologous DCs and in vitro differentiation by antigen pulsation. The third platform, DCVax-Direct, is derived from monocyte purification from leukopheresis followed by DC differentiation and in vitro stimulation with Calmette-Geurin to induce DC activation. The DCVax-Direct platform is used in cases of inadequate tumor sample/unresectable cases and is injected directly into tumors [65].

Phase I/II trials conducted out of the University of California, Los Angeles enrolled 39 patients (20 newly-diagnosed GBMs) revealed 33% of patients met or exceeded a median OS of 48.0 months and 27% exceeded a median OS of 72.0 months, with 2 patients alive greater than 10.0 years. Currently, a Phase III randomized, double-blind, multi-center trial investigating DCVax in newly diagnosed GBM patients is currently ongoing (NCT00045968) [66].

Heat shock protein (HSP) vaccines

Heat shock proteins (HSPs) represent a broad group of constitutively expressed proteins that function as intracellular molecular chaperones or proteases whose concentrations can rise dramatically in the setting of protein misfolding, unfolding, or aggregation [67-70]. These stress response proteins maintain protein architecture by responding to varying temperature, oxidative stresses, metabolic disturbances, exogenous chemical activity, viral infection, hypoxic conditions, and malignant transformations. Furthermore, soluble HSPs are capable of binding CD91 upon DCs leading to enhanced priming of CD4+ and CD8+ T cell responses.

Interestingly, immune responses are generated against peptide sequences associated with HSPs, while HSPs serve as adjuvants [71-73]. Furthermore, only HSP-peptide complexes are able to generate antitumor immune response [74]. One HSP of interest, GP96, released following cell death, has been shown to interact with Toll-like receptor 2 (TLR-2) and receptor 4 (TLR-4) on dendritic cells and macrophages. Binding of GP96 to TLR-2 or TLR-4 upon these cells increases expression of co-stimulatory molecules CD80, CD86, and CD40 as well as MHC II, IL-12, and TNF-α expression [75-77].

Crane et al. investigated the efficacy of HSP-96 for recurrent GBM in a phase I study involving 12 patients with autologous tumor-derived HSP peptide complex (HSPPC, Aegenus Incorporated). Eleven patients demonstrated specific peripheral immune responses and seven demonstrated increased immune cell infiltrate in post-vaccine tumor resection samples as well [77]. Bloch et al. conducted a phase II study evaluating tumor antigenic peptides in the context of HSP-96 for recurrent GBMs. The study enrolled 41 patients with a median PFS of 19.1 weeks and median, 6-month and 12-month OS were 42.7 weeks, 90.2% and 29.3%, respectively. Lastly, a higher absolute lymphocyte count (ALC) was found to correlate with improved survival [78]. Currently, multi-center, single arm Phase II trials evaluating the efficacy of HSPPC-96 in newly diagnosed GBMs (NCT00905060) as well as recurrent/progressive GBMs (NCT00293423) have completed accrual and are currently in follow-up phase with another phase II trial evaluating HSPPC-96 with or without bevacizumab in recurrent GBMs (NCT0181413) currently recruiting patients.

Adoptive Cell Therapy (ACT)

The elucidation of the function of T lymphocytes in the 1960’s followed by the discovery of IL-2 in 1976 represented the foundation through which adoptive cell therapy (ACT) could thrive [79,80]. Furthermore, success using IL-2 for patients with metastatic melanoma and renal cell carcinoma revealed the ability to induce an endogenous host immune response against cancer [81]. The observation that tumor specimens were heavily infiltrated by lymphocytes and that ex vivo expansion and adoptive transfers in murine models could establish regression of established tumors provided proof of principle followed by human studies resulted in objective responses, albeit for short durations [81-83].

Cytotoxic T lymphocytes (CTLs) represent an important component of host immune responses to cancer. Indeed, infiltrative tumor-reactive CTLs recognize non-self epitopes with specificity via the interaction of the T-cell receptor (TCR) with peptide in the context of MHC resulting in robust activation, proliferation and effector molecule/cytokine production. Autologous CTLs from tumor samples can be expanded in vitro in the presence of IL-2 and stimulated with antibodies specific to the TCR and passively transferred into host recipients.

Adoptive cell therapy (ACT) involves ex vivo autologous culture of tumor infiltrating lymphocytes in the presence of IL-2 and passive transfer following selection for lymphocytes with high-avidity for tumor epitopes. ACT is associated with numerous advantages relative to other cancer immunotherapies. These include the ability to expand large quantities of TILs in vitro, bypassing immunosuppressive environments seen in vivo. Lastly, host TME manipulation prior to ACT affords the ability to optimize the efficacy of transferred cells [84]. Here, we discuss ACT in the context of glioma treatment.

Lymphokine-activated killer (LAK) cells

Lymphokine-activated killer (LAK) cells represent a population of peripherally derived CD8+ cells activated in vitro in the presence of IL-2 with non-specific tumoricidal activity. Furthermore, these cells are capable of lysing fresh, non-cultured, natural killer (NK) cell-resistant tumor cells. Adoptive transfer of LAK cells with recombinant IL-2 mediated regression of a variety of metastatic tumors in numerous murine models [85-88]. Hayes et al. reported their results treating 19 adult patients with recurrent malignant glioma with intra-cavitary autologous LAK cells plus IL-2 following re-operation. Of note, one patient with anaplastic astrocytoma experienced a complete response and one patient with GBM experienced a delayed complete response with two other patients with GBM experiencing partial responses. Furthermore, the median survival was 53 weeks following re-operation compared to 25.5 weeks for contemporary patients with GBM who underwent re-operation and chemotherapy. Interestingly, aspiration from the Ommaya reservoir revealed regional eosinophilia and an extensive lymphocytic infiltrate [89].

Natural killer (NK) cells

NK cells, identified as CD56+ lymphocytes, represent a subset of cytotoxic lymphocytes capable of non-specific anti-viral and antitumor activity. Ligation of killer inhibitory receptors (KIRs) on NK cells with MHC I molecules inhibits the tyrosine- kinase-based cytolytic activity of NK cells [90]. Advantages to NK ACT include the short period of time needed to undergo NK cell expansion. Additionally, because NK cells kill in a non-specific manner, tumor specimens are not needed. However, the immunosuppressive glioma TME results in decreased IL-2 and IFN-γ production, which is critical for NK activity, representing a potential challenge to NK ACT.

Ishikawa et al. performed adoptive transfers of autologous NK cells derived from peripheral blood mononuclear cells (PBMCs) with IFN-β for patients suffering from recurrent high-grade gliomas. A total of 9 patients underwent 16 courses of ACT. Of those 9 patients, 3 experienced partial responses, 2 experienced a minor response, 4 experience no change in disease, and 7 experienced progressive disease with no signs of severe neurological toxicity [91]. This study highlighted the feasibility and safety of NK ACT for malignant gliomas.

Chimeric Antigen Receptor (CAR) T Cells

Significant advances over the past few decades have revolutionized the use of adoptive T-cell transfer and demonstrated clear durable responses in a variety of aggressive and metastatic diseases [92,93]. However, formidable challenges still abound regarding adoptive T-cell transfer, including technical challenges related to isolation of T cells from tumor specimens, large scale production and financial challenges/burdens. Many of these challenges are being overcome by the development of genetically engineered T cells derived from patients with transgenic T cell receptors (TCRs) or chimeric antigen receptors (CARs) derived from high-affinity antibodies capable of being designed with specificity to a variety of antigens. Indeed, these CAR T-cells have resulted in impressive clinical responses in hematological malignancies [94,95]. To date, the majority of CAR based studies have focused upon B-cell malignancies where CD19 or CD20 CARs have consistently demonstrated significant clinical responses [94-97]. Based on these successes, CAR therapies with specificity to the EGFRvIII protein are currently under active investigation for GBM. Indeed, the therapeutic potential of CAR therapy for GBM has been demonstrated [98-102].

Immune Checkpoint Therapy

Among the most exciting immunotherapeutic modalities, immune checkpoint blockade has garnered FDA approval for a variety of malignancies including melanoma, squamous and non-squamous non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC) and classical Hodgkin lymphoma (CHL). The amplitude and quality of T cell responses is initiated by TCR engagement and fine-tuned by co-stimulatory and co-inhibitory immune checkpoints. These co-stimulatory and co-inhibitory molecules maintain self-tolerance under normal conditions; however, a variety of malignancies expression checkpoint molecules in an effort to induce tolerance [103]. As a result, intense efforts focus upon the utilization of co-stimulatory agonist and co-inhibitory antagonist monoclonal antibodies as an additional approach to restore anti-tumor immune function for a variety of malignancies including GBMs (Table 2).

NCT number Title Agent Phase Outcome measures
NCT02798406 Combination Adenovirus + Pembrolizumab to Trigger Immune Virus Effects DNX-2401 + pembrolizumab Phase 2 Objective response rate (ORR) | Overall survival (OS) | Time to tumor response | Duration of response
NCT02852655 A Pilot Surgical Trial To Evaluate Early Immunologic Pharmacodynamic Parameters For The PD-1 Checkpoint Inhibitor, Pembrolizumab (MK-3475), In Patients With Surgically Accessible Recurrent/Progressive Glioblastoma Drug: MK-3475 Tumor Infiltrating T Lymphocyte (TIL) Density | Incidence of Treatment-Emergent Adverse Events | Progression Free Survival
NCT02667587 Study of Temozolomide Plus Radiation Therapy With Nivolumab or Placebo, for Newly Diagnosed Patients With Glioblastoma (GBM, a Malignant Brain Cancer). Nivolumab + Temozolomide + Radiotherapy Phase 2 Overall survival defined as time from the date of randomization to the date of death. | Progression free survival, defined as the time from randomization to the date of the first documented tumor progression or death to any cause.
NCT02617589 Study of Nivolumab Compared to Temozolomide, Given With Radiation Therapy, for Newly-diagnosed Patients With Glioblastoma (GBM, a Malignant Brain Cancer) Nivolumab + Temozolomide + Radiotherapy Phase 3 Overall survival (OS) | Progression free survival (PFS) | Overall survival
NCT02431572 A Pilot Study to Evaluate PBR PET in Brain Tumor Patients Treated With Chemoradiation or Immunotherapy PBR PET + Cancer Immunotherapy + Radiation and chemotherapy Change in PBR uptake (changes in PBR uptake by PET)
NCT02502708 Study of the IDO Pathway Inhibitor, Indoximod, and Temozolomide for Pediatric Patients With Progressive Primary Malignant Brain Tumors Indoximod + Temozolomide + Conformal Radiation Phase 1 Incidence of regimen limiting toxicities (RLTs) | Objective Response Rate | Pharmacokinetics: Serum concentrations (Cmax/Steady State) | Safety and Tolerability of Indoximod combined with Temozolomide as assessed by incidence and severity of adverse events, dose interruptions and dose reductions. | Progression Free Survival (PFS) | Time to Progression | Overall Survival | Safety and Feasibility of Indoximod combined with conformal radiation as assessed by incidence and severity of adverse events, dose interruptions and dose reductions.

Table 2: Checkpoint blockade-based clinical trials for GBM. Source: clinicaltrials.gov

Cytotoxic T lymphocyte antigen-4 (CTLA-4), an inhibitory checkpoint and member of the B7 family, was the first clinically targeted inhibitory checkpoint. While CTLA-4 binds B7-1 or B7-2 and serves as an inhibitory signal following TCR ligation with cognate antigen in the context of MHC, CD28 also binds B7-1 or B7-2 providing co-stimulation following TCR ligation [104-108]. Despite expression on CD8+, the role of CTLA-4 expression on CD4+ Th helper cells and Tregs appear to play the dominant physiological role. Moreover, CTLA-4 serves to dampen CD4+ Th helper cells while engagement on Tregs enhances suppressive activity [109-111]. The biological significance of CTLA-4 is highlighted by the lethal intense autoimmune phenotype demonstrated by Ctla-4-/- mice [112,113].

Despite initial concern over the potentially lethal ramifications of CTLA-4 blockade, Allison and colleagues revealed blockade of CTLA-4 did not result in overt immune toxicity in preclinical models and could enhance endogenous anti-tumor responses [114,115]. By the early 2000s, two fully humanized antagonist CTLA-4 antibodies; ipilimumab (Bristol Meyer-Squibb) and tremelimumab (Pfizer) began clinical testing. Ipilimumab would ultimately go on to become the first therapy resulting in a survival benefit and increased overall survival for patients with metastatic melanoma and was ultimately approved by the Food and Drug Administration (FDA) in 2010 [49]. Efforts are underway to investigate the safety and dosage of ipilimumab with temozolomide in newly diagnosed GBM (NCT02311920) with another Phase II/III study of standard of care (SOC) temozolomide in combination with ipilimumab for newly diagnosed glioblastoma (RTOG 1125) [116].

Similar to CTLA-4, the programmed death 1 (PD-1) inhibitory immune checkpoint receptor represents another promising target. The major biologic role of PD-1 appears to be in limiting peripheral immune responses during inflammatory insults [117-121]. Following T cell activation, PD-1 surface expression increases and engagement of PD-1 with either programmed death ligand 1 (PD-L1, B7-H1 or CD274) or programmed death ligand 2 (PD-L2, B7-DC, CD273) inhibits TCR-mediated T cell activation [117,118,122,123]. Persistently high levels of PD-1 expression occur during chronic antigen exposure resulting in T cell exhaustion. Interestingly, the PD-1:PD-L1/L2 interaction upon T cell infiltrating lymphocytes (TILs), myeloid cells and tumor cells appears to be a major mechanism of immune evasion in cancer [124-131]. PD-L1 expression on GBM tumor cells increases with loss of phosphatase and tensin homolog (PTEN) and activation of the phosphatidylinositol-3-OH kinase (PI3K) pathway [89].

Mounting evidence suggests the PD-1:PD-L1 pathway may play a role in mediating immune evasion in high-grade glioma [132-134]. A number of therapeutic human antibodies targeting the PD-1 receptor have been developed including Pembrolizumab (Merck) and Nivolumab (BMS) to name a few. Despite initial concerns, antibodies targeting the PD-1 pathway may not result in unique CNS toxicity [135]. The majority of clinical data available regarding CNS malignancy has primarily focused upon investigating the efficacy of anti-PD-1 therapy for brain metastasis. A non-randomized Phase II trial investigated the efficacy of Pembrolizumab for patients with untreated melanoma or non-small cell lung cancer (NSCLC) brain metastasis revealed durable responses in 4 of 18 patients with melanoma and 6 of 18 patients with NSCLC [136]. Given recent data demonstrating PD-1 expression upon tumor-infiltrating lymphocytes, recent clinical trials determining the efficacy of anti-PD-1 or anti-PDL1 therapy in primary brain tumors are under investigation [137,138]. A phase III trial comparing Nivolumab with bevacizumab and Nivolumab with or without Ipilimumab is currently recruiting patients although a small safety lead-in revealed an overall survival at 6 months of 70% (NCT02017717; Checkmate 143). A number of clinical trials involving anti-PD-1/L1 therapy for newly diagnosed or recurrent glioblastoma are currently underway (NCT02617589, NCT02667587, NCT02550249, NCT02311920, NCT02337491, NCT02337686, NCT02658279, NCT02336165).

Conclusions & Future Directions

Significant advances in the fields of neuro- and cancer immunology provide a compelling argument for the use of immunotherapy for CNS malignancies. Despite the devastating prognosis associated with GBM, immunotherapy represents a novel anticancer modality with the ability to result in drastic responses in otherwise incurable diseases. A greater understanding of the mechanisms through which GBMs evade the immune system will aid in the development of strategic immunotherapy regimens tailored to each person’s disease. Questions remain regarding the efficacy of immunotherapy in the context of the current SOC and how best to utilize immunotherapy. Future studies are necessary to explore the aforementioned questions; however, significant hope remains for the role of immunotherapy in the treatment of GBM.

References

  1. Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, et al. (2015) A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med 212: 991-999.
  2. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, et al. (2015) Structural and functional features of central nervous system lymphatic vessels. Nature 523: 337-334.
  3. Russo MV, McGavern DB (2015) Immune Surveillance of the CNS following Infection and Injury.  Trends Immunol 36: 637-650.
  4. Smolders J, Remmerswaal EB, Schuurman KG, Melief J, van Eden CG, et al. (2013) Characteristics of differentiated CD8(+) and CD4 (+) T cells present in the human brain. Acta Neuropathol 126: 525-535.
  5. Belcaid Z, Phallen JA, Zeng J, See AP, Mathios D, et al. (2014) Focal radiation therapy combined with 4-1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model. PLoS One 9: e101764.
  6. Kim JE, Patel MA, Mangraviti A, Kim ES, Theodros D, et al. (2016) Combination therapy with anti-PD-, anti-TIM-3, and focal radiation results in regression of murine gliomas.  Clin Cancer Res.
  7. Patel MA, Kim JE, Theodros D, Tam A, Velarde E, et al. (2016) Agonist anti-GITR monoclonal antibody and stereotactic radiation induce immune-mediated survival advantage in murine intracranial glioma. J Immunother Cancer 4: 28.
  8. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, et al. (2013) Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys 86: 343-349.
  9. Buckner JC (2003) Factors influencing survival in high-grade gliomas.  Semin Oncol 30: 10-14.
  10. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, et al. (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma.  N Engl J Med 352: 987-996.
  11. Liu Q, Nguyen DH, Dong Q, Shitaku P, Chung K, et al. (2009) Molecular properties of CD133+ glioblastoma stem cells derived from treatment-refractory recurrent brain tumors. J Neurooncol 94: 1-19.
  12. Ransohoff RM, Engelhardt B (2012) The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol 12: 623-635.
  13. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM (2011) Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool.  Nat Neurosci 14: 1142-1149.
  14. Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM (2007) Local self-renewal can sustain CNS microglia maintenance and function throughout adult life.  Nat Neurosci 10: 1538-1543.
  15. Ransohoff RM (2011) Microglia and monocytes: 'tis plain the twain meet in the brain.  Nat Neurosci 14: 1098-1100.
  16. Giunti D, Borsellino G, Benelli R, Marchese M, Capello E, et al. (2003) Phenotypic and functional analysis of T cells homing into the CSF of subjects with inflammatory diseases of the CNS.  J Leukoc Biol 73: 584-590.
  17. Kivisäkk P, Mahad DJ, Callahan MK, Trebst C, Tucky B, et al. (2003) Human cerebrospinal fluid central memory CD4+ T cells: evidence for trafficking through choroid plexus and meninges via P-selectin.  Proc Natl Acad Sci U S A 100: 8389-8394.
  18. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A (2014) Pillars article: two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999. 401: 708-712. J Immunol 192: 840-844.
  19. Billingham RE, Boswell T (1953) Studies on the problem of corneal homografts.  Proc R Soc Lond B Biol Sci 141: 392-406.
  20. Galea I, Bechmann I, Perry VH (2007) What is immune privilege (not)?  Trends Immunol 28: 12-18.
  21. Stevenson PG, Hawke S, Sloan DJ, Bangham CR (1997) The immunogenicity of intracerebral virus infection depends on anatomical site.  J Virol 71: 145-151.
  22. Blond D, Campbell SJ, Butchart AG, Perry VH, Anthony DC (2002) Differential induction of interleukin-1beta and tumour necrosis factor-alpha may account for specific patterns of leukocyte recruitment in the brain.  Brain Res 958: 89-99.
  23. Nayak D, Zinselmeyer BH, Corps KN, McGavern DB (2012) In vivo dynamics of innate immune sentinels in the CNS.  Intravital 1: 95-106.
  24. Elliott LH, Brooks WH, Roszman TL (1987) Activation of immunoregulatory lymphocytes obtained from patients with malignant gliomas.  J Neurosurg 67: 231-236.
  25. Crane CA, Ahn BJ, Han SJ, Parsa AT (2012) Soluble factors secreted by glioblastoma cell lines facilitate recruitment, survival, and expansion of regulatory T cells: implications for immunotherapy. Neuro Oncol 14: 584-595.
  26. Jacobs JF, Idema AJ, Bol KF, Grotenhuis JA, de Vries IJ, et al. (2010) Prognostic significance and mechanism of Treg infiltration in human brain tumors. J Neuroimmunol 225: 195-199.
  27. Ebert LM, Tan BS, Browning J, Svobodova S, Russell SE, et al. (2008) The regulatory T cell-associated transcription factor FoxP3 is expressed by tumor cells.  Cancer Res 68: 3001-3009.
  28. Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, Yang DS, Sun W, et al. (2008) Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas.  Clin Cancer Res 14: 5166-5172.
  29. Waziri A, Killory B, Ogden AT 3rd, Canoll P, Anderson RC, et al. (2008) Preferential in situ CD4+CD56+ T cell activation and expansion within human glioblastoma.  J Immunol 180: 7673-7680.
  30. Badie B, Schartner J, Klaver J, Vorpahl J (1999) In vitro modulation of microglia motility by glioma cells is mediated by hepatocyte growth factor/scatter factor. Neurosurgery 44: 1077-1082; discussion 1082-1073.
  31. Coniglio SJ, Eugenin E, Dobrenis K, Stanley ER, West BL, et al. (2012) Microglial stimulation of glioblastoma invasion involves epidermal growth factor receptor (EGFR) and colony stimulating factor 1 receptor (CSF-1R) signaling. Mol Med 18: 519-527.
  32. Desbaillets I, Tada M, de Tribolet N, Diserens AC, Hamou MF, et al. (1994) Human astrocytomas and glioblastomas express monocyte chemoattractant protein-1 (MCP-1) in vivo and in vitro. Int J Cancer 58: 240-247.
  33. Held-Feindt J, Hattermann K, Müerköster SS, Wedderkopp H, Knerlich-Lukoschus F, et al. (2010) CX3CR1 promotes recruitment of human glioma-infiltrating microglia/macrophages (GIMs).  Exp Cell Res 316: 1553-1566.
  34. Wu A, Wei J, Kong LY, Wang Y, Priebe W, et al. (2010) Glioma cancer stem cells induce immunosuppressive macrophages/microglia.  Neuro Oncol 12: 1113-1125.
  35. Lin HC, Song TY, Hu ML (2009) S-Adenosylhomocysteine promotes the invasion of C6 glioma cells via increased secretion of matrix metalloproteinase-2 in murine microglial BV2 cells. Toxicol Sci 112: 322-330.
  36. Markovic DS, Vinnakota K, Chirasani S, Synowitz M, Raguet H, et al. (2009) Gliomas induce and exploit microglial MT1-MMP expression for tumor expansion. Proc Natl Acad Sci U S A 106: 12530-12535.
  37. Wesolowska A, Kwiatkowska A, Slomnicki L, Dembinski M, Master A, et al. (2008) Microglia-derived TGF-beta as an important regulator of glioblastoma invasion--an inhibition of TGF-beta-dependent effects by shRNA against human TGF-beta type II receptor. Oncogene 27: 918-930.
  38. Zhang J, Sarkar S, Cua R, Zhou Y, Hader W, et al. (2012) A dialog between glioma and microglia that promotes tumor invasiveness through the CCL2/CCR2/interleukin-6 axis.  Carcinogenesis 33: 312-319.
  39. Huettner C, Czub S, Kerkau S, Roggendorf W, Tonn JC (1997) Interleukin 10 is expressed in human gliomas in vivo and increases glioma cell proliferation and motility in vitro.  Anticancer Res 17: 3217-3224.
  40. Huettner C, Paulus W, Roggendorf W (1995) Messenger RNA expression of the immunosuppressive cytokine IL-10 in human gliomas.  Am J Pathol 146: 317-322.
  41. Strle K, Zhou JH, Shen WH, Broussard SR, Johnson RW, et al. (2001) Interleukin-10 in the brain.  Crit Rev Immunol 21: 427-449.
  42. Trifunovi? J, Miller L, Debeljak ?, Horvat V (2015) Pathologic patterns of interleukin 10 expression--a review.  Biochem Med (Zagreb) 25: 36-48.
  43. Van Meir EG1 (1995) Cytokines and tumors of the central nervous system.  Glia 15: 264-288.
  44. Wei J, Wu A, Kong LY, Wang Y, Fuller G, et al. (2011) Hypoxia potentiates glioma-mediated immunosuppression.  PLoS One 6: e16195.
  45. Zhang X, Rao A, Sette P, Deibert C, Pomerantz A, et al. (2016) IDH mutant gliomas escape natural killer cell immune surveillance by downregulation of NKG2D ligand expression. Neuro Oncol 18: 1402-1412.
  46. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, et al. (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response.  Nature 444: 756-760.
  47. Liu G, Yuan X, Zeng Z, Tunici P, Ng H, et al. (2006) Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma.  Mol Cancer 5: 67.
  48. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma.  N Engl J Med 363: 711-723.
  49. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer.  N Engl J Med 366: 2443-2454.
  50. Bot A (2010) The landmark approval of Provenge, what it means to immunology and "in this issue": the complex relation between vaccines and autoimmunity.  Int Rev Immunol 29: 235-238.
  51. Rock KL, Shen L (2005) Cross-presentation: underlying mechanisms and role in immune surveillance. Immunol Rev 207: 166-183.
  52. Phuphanich S, Wheeler CJ, Rudnick JD, Mazer M, Wang H, et al. (2013) Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother 62: 125-135.
  53. Engebraaten O, Bjerkvig R, Pedersen PH, Laerum OD (1993) Effects of EGF, bFGF, NGF and PDGF(bb) on cell proliferative, migratory and invasive capacities of human brain-tumour biopsies in vitro.  Int J Cancer 53: 209-214.
  54. Goldman CK, Kim J, Wong WL, King V, Brock T, et al. (1993) Epidermal growth factor stimulates vascular endothelial growth factor production by human malignant glioma cells: a model of glioblastoma multiforme pathophysiology. Mol Biol Cell 4: 121-133.
  55. Gorgoulis V, Aninos D, Mikou P, Kanavaros P, Karameris A, et al. (1992) Expression of EGF, TGF-alpha and EGFR in squamous cell lung carcinomas.  Anticancer Res 12: 1183-1187.
  56. Irish JC, Bernstein A (1993) Oncogenes in head and neck cancer.  Laryngoscope 103: 42-52.
  57. Korc M, Meltzer P, Trent J (1986) Enhanced expression of epidermal growth factor receptor correlates with alterations of chromosome 7 in human pancreatic cancer. Proc Natl Acad Sci U S A 83: 5141-5144.
  58. Wong AJ, Ruppert JM, Bigner SH, Grzeschik CH, Humphrey PA, et al. (1992) Structural alterations of the epidermal growth factor receptor gene in human gliomas.  Proc Natl Acad Sci U S A 89: 2965-2969.
  59. Humphrey PA, Wong AJ, Vogelstein B, Zalutsky MR, Fuller GN, et al. (1990) Anti-synthetic peptide antibody reacting at the fusion junction of deletion-mutant epidermal growth factor receptors in human glioblastoma. Proc Natl Acad Sci U S A 87: 4207-421.
  60. Sampson JH, Archer GE, Mitchell DA, Heimberger AB, Herndon JE 2nd, et al. (2009) An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme.  Mol Cancer Ther 8: 2773-2779.
  61. Choi BD, Archer GE, Mitchell DA, Heimberger AB, McLendon RE, et al. (2009) EGFRvIII-targeted vaccination therapy of malignant glioma.  Brain Pathol 19: 713-723.
  62. Del Vecchio CA, Li G, Wong AJ (2012) Targeting EGF receptor variant III: tumor-specific peptide vaccination for malignant gliomas.  Expert Rev Vaccines 11: 133-144.
  63. Schuster J, Lai RK, Recht LD, Reardon DA, Paleologos NA, et al. (2015) A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: the ACT III study.  Neuro Oncol 17: 854-861.
  64. Hdeib A, Sloan AE (2015) Dendritic cell immunotherapy for solid tumors: evaluation of the DCVax® platform in the treatment of glioblastoma multiforme.  CNS Oncol 4: 63-69.
  65. Polyzoidis S, Ashkan K (2014) DCVax®-L--developed by Northwest Biotherapeutics.  Hum Vaccin Immunother 10: 3139-3145.
  66. Nover L, Hightower L (1991) Heat shock and development. Introduction. Results Probl Cell Differ 17: 1-4.
  67. Parsell DA, Lindquist S (1993) The function of heat-shock proteins in stress tolerance: degradation and reactivation of damaged proteins.  Annu Rev Genet 27: 437-496.
  68. Strbo N, Garcia-Soto A, Schreiber TH, Podack ER (2013) Secreted heat shock protein gp96-Ig: next-generation vaccines for cancer and infectious diseases.  Immunol Res 57: 311-325.
  69. Basu S, Srivastava PK (1999) Calreticulin, a peptide-binding chaperone of the endoplasmic reticulum, elicits tumor- and peptide-specific immunity.  J Exp Med 189: 797-802.
  70. Srivastava PK, Chen YT, Old LJ (1987) 5'-structural analysis of genes encoding polymorphic antigens of chemically induced tumors. Proc Natl Acad Sci U S A 84: 3807-381.
  71. Udono H, Srivastava PK (1994) Comparison of tumor-specific immunogenicities of stress-induced proteins gp96, hsp90, and hsp70.  J Immunol 152: 5398-5403.
  72. Blachere NE, Li Z, Chandawarkar RY, Suto R, Jaikaria NS, et al. (1997) Heat shock protein-peptide complexes, reconstituted in vitro, elicit peptide-specific cytotoxic T lymphocyte response and tumor immunity. J Exp Med 186: 1315-1322.
  73. Chen W, Syldath U, Bellmann K, Burkart V, Kolb H (1999) Human 60-kDa heat-shock protein: a danger signal to the innate immune system.  J Immunol 162: 3212-3219.
  74. Singh-Jasuja H, Scherer HU, Hilf N, Arnold-Schild D, Rammensee HG, et al. (2000) The heat shock protein gp96 induces maturation of dendritic cells and down-regulation of its receptor. Eur J Immunol 30:2211-2215.
  75. Crane CA, Han SJ, Ahn B, Oehlke J, Kivett V, et al. (2013) 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 19: 205-214.
  76. Bloch O, Crane CA, Fuks Y, Kaur R, Aghi MK, et al. (2014) Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro Oncol 16: 274-279.
  77. Delorme EJ, Alexander P (1964) Treatment of Primary Fibrosarcoma in the Rat with Immune Lymphocytes.  Lancet 2: 117-120.
  78. Fefer A (1969) Immunotherapy and chemotherapy of Moloney sarcoma virus-induced tumors in mice. Cancer Res 29: 2177-2183.
  79. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, et al. (1985) Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med 313: 1485-1492.
  80. Muul LM, Spiess PJ, Director EP, Rosenberg SA (1987) Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma. J Immunol 138: 989-995.
  81. Rosenberg SA, Spiess P, Lafreniere R (1986) A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233: 1318-132.
  82. Rosenberg SA, Restifo NP (2015) Adoptive cell transfer as personalized immunotherapy for human cancer.  Science 348: 62-68.
  83. Lafreniere R, Rosenberg SA (1985) Successful immunotherapy of murine experimental hepatic metastases with lymphokine-activated killer cells and recombinant interleukin 2. Cancer Res 45: 3735-374.
  84. Mazumder A, Rosenberg SA (1984) Successful immunotherapy of natural killer-resistant established pulmonary melanoma metastases by the intravenous adoptive transfer of syngeneic lymphocytes activated in vitro by interleukin 2. J Exp Med 159:495-507.
  85. Mule JJ, Shu S, Rosenberg SA (1985) The anti-tumor efficacy of lymphokine-activated killer cells and recombinant interleukin 2 in vivo. J Immunol 135: 646-652.
  86. Rosenberg S (1985) Lymphokine-activated killer cells: a new approach to immunotherapy of cancer.  J Natl Cancer Inst 75: 595-603.
  87. Hayes RL, Koslow M, Hiesiger EM, Hymes KB, Hochster HS, et al. (1995) Improved long term survival after intracavitary interleukin-2 and lymphokine-activated killer cells for adults with recurrent malignant glioma. Cancer 76: 840-852.
  88. Campbell KS, Hasegawa J (2013) Natural killer cell biology: an update and future directions.  J Allergy Clin Immunol 132: 536-544.
  89. Ishikawa E, Tsuboi K, Saijo K, Harada H, Takano S, et al. (2004) Autologous natural killer cell therapy for human recurrent malignant glioma. Anticancer Res 24: 1861-187.
  90. Hong JJ, Rosenberg SA, Dudley ME, Yang JC, White DE, et al. (2010) Successful treatment of melanoma brain metastases with adoptive cell therapy.  Clin Cancer Res 16: 4892-4898.
  91. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, et al. (2011) Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 17: 4550-4557.
  92. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, et al. (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia.  N Engl J Med 371: 1507-1517.
  93. Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365: 725-733.
  94. Kochenderfer JN, Rosenberg SA (2013) Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors.  Nat Rev Clin Oncol 10: 267-276.
  95. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, et al. (2012) CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. Blood 119: 3940-3950.
  96. Bullain SS, Sahin A, Szentirmai O, Sanchez C, Lin N, et al. (2009) Genetically engineered T cells to target EGFRvIII expressing glioblastoma. J Neurooncol 94: 373-382.
  97. Kahlon KS, Brown C, Cooper LJ, Raubitschek A, Forman SJ, et al. (2004) Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells.  Cancer Res 64: 9160-9166.
  98. Kong S, Sengupta S, Tyler B, Bais AJ, Ma Q, et al. (2012) Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells.  Clin Cancer Res 18: 5949-5960.
  99. Morgan RA, Johnson LA, Davis JL, Zheng Z, Woolard KD, et al. (2012) Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma.  Hum Gene Ther 23: 1043-1053.
  100. Sampson JH, Choi BD, Sanchez-Perez L, Suryadevara CM, Snyder DJ, et al. (2014) EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clin Cancer Res 20: 972-984.
  101. Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy.  Nat Rev Cancer 12: 252-264.
  102. Azuma M, Ito D, Yagita H, Okumura K, Phillips JH, et al. (1993) B70 antigen is a second ligand for CTLA-4 and CD28.  Nature 366: 76-79.
  103. Freeman GJ, Gribben JG, Boussiotis VA, Ng JW, Restivo VA Jr, et al. (1993) Cloning of B7-2: a CTLA-4 counter-receptor that costimulates human T cell proliferation.  Science 262: 909-911.
  104. Hathcock KS, Laszlo G, Dickler HB, Bradshaw J, Linsley P, et al. (1993) Identification of an alternative CTLA-4 ligand costimulatory for T cell activation.  Science 262: 905-907.
  105. Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle NK, et al. (1991) CTLA-4 is a second receptor for the B cell activation antigen B7.  J Exp Med 174: 561-569.
  106. Linsley PS, Clark EA, Ledbetter JA (1990) T-cell antigen CD28 mediates adhesion with B cells by interacting with activation antigen B7/BB-1. Proc Natl Acad Sci U S A 87:5031-5035.
  107. Lenschow DJ, Walunas TL, Bluestone JA (1996) CD28/B7 system of T cell costimulation.  Annu Rev Immunol 14: 233-258.
  108. Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP (2009) Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med 206:1717-1725.
  109. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, et al. (2008) CTLA-4 control over Foxp3+ regulatory T cell function.  Science 322: 271-275.
  110. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, et al. (1995) Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3:541-547.
  111. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, et al. (1995) Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270:985-988.
  112. Leach DR, Krummel MF, Allison JP (1996) Enhancement of antitumor immunity by CTLA-4 blockade.  Science 271: 1734-1736.
  113. van Elsas A, Hurwitz AA, Allison JP (1999) Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med 190:355-366.
  114. Tanaka S, Louis DN, Curry WT, Batchelor TT, Dietrich J (2013) Diagnostic and therapeutic avenues for glioblastoma: no longer a dead end? Nat Rev Clin Oncol 10: 14-26.
  115. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, et al. (2000) Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192:1027-1034.
  116. Ishida Y, Agata Y, Shibahara K, Honjo T (1992) Induced expression of PD-, a novel member of the immunoglobulin gene superfamily, upon programmed cell death.  EMBO J 11: 3887-3895.
  117. Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, et al. (2006) Tissue expression of PD-L1 mediates peripheral T cell tolerance.  J Exp Med 203: 883-895.
  118. Nishimura H, Nose M, Hiai H, Minato N, Honjo T (1999) Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor.  Immunity 11: 141-151.
  119. Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, et al. (2001) Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice.  Science 291: 319-322.
  120. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, et al. (2001) PD-L2 is a second ligand for PD-1 and inhibits T cell activation.  Nat Immunol 2: 261-268.
  121. Tseng SY, Otsuji M, Gorski K, Huang X, Slansky JE, et al. (2001) B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells. J Exp Med 193:839-846.
  122. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, et al. (2009) Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114:1537-1544.
  123. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, et al. (2006) Restoring function in exhausted CD8 T cells during chronic viral infection.  Nature 439: 682-687.
  124. Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, et al. (2004) PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells.  Cancer Res 64: 1140-1145.
  125. Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, et al. (2003) Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 9: 562-567.
  126. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, et al. (2002) Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion.  Nat Med 8: 793-800.
  127. Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, et al. (2009) Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med 206:1327-1337.
  128. Liu Y, Zeng B, Zhang Z, Zhang Y, Yang R (2008) B7-H1 on myeloid-derived suppressor cells in immune suppression by a mouse model of ovarian cancer. Clin Immunol 129: 471-48.
  129. Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Isaacs WB, et al. (2009) Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+.  Prostate 69: 1694-1703.
  130. Avril T, Saikali S, Vauleon E, Jary A, Hamlat A, et al. (2010) Distinct effects of human glioblastoma immunoregulatory molecules programmed cell death ligand-1 (PDL-1) and indoleamine 2,3-dioxygenase (IDO) on tumour-specific T cell functions. J Neuroimmunol 225: 22-33.
  131. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, et al. (2007) Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma.  Nat Med 13: 84-88.
  132. Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, et al. (2003) Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res 63:7462-7467.
  133. Rothermundt C, Hader C, Gillessen S (2016) Successful treatment with an anti-PD-1 antibody for progressing brain metastases in renal cell cancer.  Ann Oncol 27: 544-545.
  134. Goldberg SB, Gettinger SN, Mahajan A, Chiang AC, Herbst RS, et al. (2016) Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial.  Lancet Oncol 17: 976-983.
  135. Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, et al. (2015) Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma.  Neuro Oncol 17: 1064-1075.
  136. Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, et al. (2016) PD-L1 expression and prognostic impact in glioblastoma.  Neuro Oncol 18: 195-205.
Citation: Theodros D, Moran D, Garzon-Muvdi T, Lim M (2016) Immunotherapy for Glioblastoma. J Clin Cell Immunol 7:464.

Copyright: © 2016 Theodros D, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top