Skip to main content
Erschienen in: Current Treatment Options in Oncology 3/2019

Open Access 01.03.2019 | Neuro-oncology (GJ Lesser, Section Editor)

Current State of Immunotherapy for Treatment of Glioblastoma

verfasst von: Tresa McGranahan, MD PhD, Kate Elizabeth Therkelsen, MD, Sarah Ahmad, MD, Seema Nagpal, MD

Erschienen in: Current Treatment Options in Oncology | Ausgabe 3/2019

Opinion statement

At this time, there are no FDA-approved immune therapies for glioblastoma (GBM) despite many unique therapies currently in clinical trials. GBM is a highly immunosuppressive tumor and there are limitations to a safe immune response in the central nervous system. To date, there have been several failures of phase 3 immune therapy clinical trials in GBM. These trials have targeted single components of an antitumor immune response. Learning from these failures, the future of immunotherapy for GBM appears most hopeful for combination of immune therapies to overcome the profound immunosuppression of this disease. Understanding biomarkers for appropriate patient selection as well as tumor progression are necessary for implementation of immunotherapy for GBM
Hinweise
This article is part of the Topical Collection on Neuro-oncology

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Glioblastoma (GBM) is the most common and most aggressive primary malignant brain tumor in adults. Clinical trials in GBM have led to incremental improved median overall survival (mOS) that have improved survival in the general GBM population [1•]. While the most recent positive phase 3 clinical trial of tumor treating fields (TTF) had a mOS of 20.5 months [2••], it is the tail of long-term survivors that continues to provide hope to patients and neuro-oncologists. TTF gained acceptance not just because of a significant improvement in survival, but an increase in 5-year survival to 13% compared to 5% in the standard of care (SOC) arm [2••]. Durable responses to immunotherapy, seen in many cancers, have re-invigorated the study of immunotherapy in GBM.
The brain is not as immuno-privileged as once thought, yet obstacles remain for immunotherapy in treating GBM. Malignant gliomas are one of the most immunosuppressive solid tumors due in part to lymphopenia driven by bone marrow suppression [3]. GBM is also an immunologically quiet tumor, with low tumor mutational burden (TMB), few tumor infiltrating T cells (TILS), and low PD-1/PD-L1 expression, especially when compared to other cancers where immunotherapy has been the most successful [4, 5]. GBM is also a profoundly heterogeneous tumor which facilitates immune evasion [6]. The SOC for newly diagnosed GBM, the combination of radiation therapy (RT) and alkylating chemotherapy, confounds this immunosuppression. Often steroids are necessary for management of peritumoral edema but they decrease the efficacy of immunotherapies [7, 8]. In addition to hurdles for stimulating an effective immune response against GBM, robust immune activation within the intracranial space poses clinical safety risks including complications of cytokine release syndrome and autoimmune encephalitis [911]. Despite these challenges, long-term responders to immunotherapy have been reported in clinical trials of GBM, but no predictive biomarkers exist at this time [12•].
Immunotherapy in GBM has a long history, including immune stimulation, antibody-mediated immunotherapies, adoptive cellular immunotherapies, and vaccines [13]. At this time, phase 3 clinical trials have not demonstrated efficacy for immunotherapy in GBM and no FDA-approved immunotherapy for GBM exists. Given the challenges of immunotherapy for GBM, a combination approach will likely be required. Additionally, identification of biomarkers for patient selection and disease surveillance are essential. Given the risks of recurrent tumor sampling, there is a search for serum, CSF, and imaging biomarkers for GBM.
Here, we discuss recent findings and ongoing clinical research into immunotherapy for GBM, including checkpoint inhibitors, vaccines, CAR-T therapy, and viral therapy. We will also discuss our current understanding of biomarkers in GBM that influence candidacy for clinical trials and may explain the response and failures to different immune therapies.

Checkpoints

Checkpoint inhibition has revolutionized treatment of several advanced malignancies providing hope for cancer treatment and resulting in a well-deserved Nobel Prize. Inhibitors of immune checkpoints such as PD-1, PD-L1, and CTLA-4 promote a shift from the normal balance of the adaptive immune system to increased immune activation [14]. In preclinical data, checkpoint inhibitors showed promise for treatment of GBM; however, translation of this preclinical work to patients was complicated by CNS toxicity when used to treat other malignancies [11, 15].
An early phase 1 study of nivolumab (nivo) alone or in combination with ipilimumab (ipi) for treatment of recurrent GBM (rGBM) found higher toxicity with the combination PD-1 and CTLA-4 inhibition, but comparable mOS (12-month mOS 40% v. 30%) [16]. Based on the tolerance of nivo monotherapy and promising mOS compared to historical controls, a large phase 3 trial was developed. CheckMate-143 randomized patients with rGBM to treatment with nivo or bevacizumab (BEV). The genotoxic stress of radiation and chemotherapy used in SOC treatment for newly diagnosed GBM were predicted to increase the TMB in rGBM and favor response to checkpoint inhibition. Unfortunately, at interim analysis of 369 patients, nivo did not demonstrate a mOS benefit over BEV (9.8 m nivo v. 10 m BEV) [17]. It should be noted however that for patients who did respond, those responses were more durable in the nivo arm (11.1 m nivo v 5.3 BEV), again raising enthusiasm for a tail of long term survivors [16]. A single center retrospective of the use of pembrolizumab (pembro) in rGBM also found no benefit [18]. Randomized controlled clinical trials examining the use of nivo in the setting of newly diagnosed GBM (CheckMate 498 and 548) are ongoing. Checkpoint inhibition for GBM was recently reviewed in this journal [19].
Promising data from NSCLC, head and neck cancer, and bladder cancer has stimulated interest in neoadjuvant checkpoint inhibition in GBM [20, 21]. Reported at Society for NeuroOnclogy 2018, patients receiving two doses of pembro prior to re-resection of rGBM had improved mOS (13.7 m neoadjuvant v. 7.5 m adjuvant only) compared to those starting pembro after re-resection [22]. While a small study, this work raises hope that the combination of pembro with surgery may help increase antigen exposure as well as TIL infiltration into rGBM.
Several other phase 1 and 2 clinical trials are currently examining the role of checkpoint inhibitors in combination with other therapies (Table 1). There is great interest in the combination of radiation with checkpoint inhibition, based partly on the idea that radiation may increase antigen presentation as well as promote the abscopal effect. These clinical trials are examining use in both newly diagnosed GBM with standard fractionated chemoradiation, as well as in the rGBM with radiosurgery and hypo-fractioned radiation. There is also a phase 1 study examining use of pembro with MRI-guided laser ablation for rGBM. Addition of pembro to the SOC including TTF is also being studied in the 2-THE-TOP phase 2 clinical trial (NCT03405792). Checkpoint inhibition with combination vaccines and oncolytic viruses will be discussed below.
Table 1
Combination Checkpoint Inhibition Trials in GBM
  
Checkpoint inhibitor
Additional therapy
Phase
Clinical trials
Dual Checkpoint blockade
New diagnosis GBM
CTLA-4 (ipilimumab)
PD-1 (nivolumab)
I
NCT02311920
rGBM
PD-1 (nivolumab)
Anti-LAG-3(BMS 986016) or anti CD137(urelumab)
I
NCT02658981
refractory solid tumors
PD-1 (nivolumab)
anti-CD-27 (varlilumab)
I/II
NCT02335918
Advanced refractory cancers
PD-1 (nivolumab)
Intratumoral IDO1 inhibitor (INT230-6)
I/II
NCT03058289
Advanced cancers
PD-1 (nivolumab)
IDO1 inhibitor (epacadostat)
I/II
NCT02327078
rHGG
PD-L1 (durvalumab)
CTLA-4 (tremelimumab)
II
NCT02794883
Vaccines
New diagnosis GBM
PD-1 (pembrolizumab)
HSPPC-96
II
NCT03018288
AVeRT
rHGG
PD-1 (nivolumab)
pp65 DC
I
NCT02529072
rGBM
PD-1 (nivolumab)
DCVAX-L
II
NCT03014804
Oncolytic virus
rGBM
PD-1 (pembrolizumab)
DNX-2401
II
NCT02798406
Radiation
rHGG
pembro
hypofractionated stereotactic irradiation
I
NCT02313272
rGBM
nivo
SRS + Valproic acid
I
NCT02648633
rHGG
nivo
hypofractionated stereotactic irradiation
I
NCT02829931
rGBM
PD-L1 (durvalumab)
hypofractionated stereotactic irradiation
I/II
STERIMGLI - NCT02866747
Laser ablation
rHGG
MK-3475
MRI-guided laser ablation
I/II
NCT02311582
CSF-1R inibition
rHGG
nivo
CSF-1r inhibitor (BLZ945)
I/II
NCT02526017
rGBM
PD-1 (PDR001)
CSF-1r inhibitor (FPA008)
I
NCT02829723
TGF-beta
advanced solid tumors
nivo
TGF-beta inhibiotr (Galunisertib)
I/II
NCT02423343
rHGG recurrent high grade glioma, rGBM recurrent glioblastoma, SRS stereotactic radiosurgery
Preclinical research suggested that low-dose VEGF inhibition promotes the switch in the tumor microenvironment from immunosuppressive (M2-like) to a more immune supportive (M1-like) tumor microenvironment [23]. In melanoma, the combination of CTLA-4 inhibition (ipi) with VEGF inhibition of BEV found disease control rate of 67.4% in 46 patients treated [24]. Aside from the change in the tumor microenvironment, BEV may play another important role in GBM patients as a steroid substitute [25, 26]. A recent retrospective study of non-small cell lung cancer (NSCLC) patients demonstrated a greater than 10% reduction in overall response rate (complete and partial responses) in patients who were on any dose of steroid greater than 10 mg of prednisone a day (= 1.6 mg of dexamethasone) prior to starting treatment with a checkpoint inhibitor. This baseline steroid use was significantly associated with decreased PFS and mOS [8]. This raises concern that necessary management of peritumoral edema in GBM patients, even with the minimal effective dose of dexamethasone needed to control symptoms, may be sufficient to dampen response to checkpoint inhibition and possibly other immunotherapy. In GBM, BEV can be used as a steroid substitute and the safety of the combination of BEV with checkpoint inhibition may provide an opportunity to treat peritumoral edema without the immunosuppressive effects of steroids. For rGBM, there are several ongoing clinical trials examining PD-1 and PD-L1 inhibitors in combination with BEV. Preliminary results from these studies support the safety of this combination; however, among rGBM patients, the combination of pembro with BEV does not improve survival [27].
At this point, there is no role for checkpoint inhibition monotherapy in the treatment of most patients with GBM; however, the combination of checkpoint inhibition with other immune stimulating therapies may be considered. Serious, and even fatal, CNS immune adverse events have been reported with checkpoint inhibition [11]. Given this risk with checkpoint inhibitor monotherapy, as therapies seek to increase immune activation against GBM, there remains concern for complications for over activation of the immune system within the brain.

Checkpoint biomarkers

In parallel with these therapeutic trials, there are several ongoing studies to help better understand biomarkers to predict response to checkpoint inhibition. A number of biomarkers are thought to predict response to PD-1 and PD-L1 inhibitors in other malignancies. Specifically in NSCLC, it has become increasingly clear that response to PD-1 inhibitors correlates with the level of PD-L1 expression in tumor. In Keynote-042, a study of pembro compared to platinum-based chemotherapy in first-line metastatic NSCLC, patients with high expression (> 50%) receiving pembro had a 20.0-month mOS compared to 12.2 months in the chemotherapy group (HR 0.69). In contrast, patients with expression between 1 and 49% receiving pembro had a mOS 13.4 months versus 12.1 months with chemotherapy (HR 0.92) [28]. The CheckMate-057 study of nivo monotherapy versus docetaxel demonstrated no benefit for checkpoint inhibition in tumors with < 1% PD-L1 expression [29]. A study of 94 patients with GBM found median PD-L1 expressional 2.77% and that PD-L1 expression correlated with worse outcome [30] while an earlier study did not find PD-L1 to be a negative prognostic factor [31]. The role of PD-L1 expression on GBM tumor cell in response to checkpoint inhibition is unclear.
To better understand changes in the tumor microenvironment with PD-1 inhibition, pembro was given prior to re-resection in patients with GBM (NCT02337686). Analysis of the resected tumor demonstrated low T cell infiltrate that was not modulated by PD-1 inhibition [32]. Of note, while use of pembro did not improve survival, all patients required steroids after pembro. Studies of neoadjuvant checkpoint inhibition have found a trend toward increased TIL fractions as well as changes in several immune markers [22].
Two tests which reflect the overall genetic stability of tumors, TMB and microsatellite instability (MSI), also play a role in predicting which patients will have meaningful responses to PD-1 axis drugs. In 2017, pembro was approved for patients with MSI or mismatch repair deficiencies for all solid tumors regardless of histology. Higher TMB and MSI correlate with longer mOS [33]. In glioma patients, favorable status across all three of these biomarkers (PD-L1, MSI, TMB) appears to be rare and suggest that only a minority of patients will respond to checkpoint monotherapy [34]. Low frequency of these markers in GBM may be contributing to the disappointing results of PD-1/PD-L1 monotherapies to date [35].

Vaccines

Cancer vaccine therapy in GBM is not preventative, but rather is designed to induce an immune response against the tumor. For GBM, vaccines encompass a range of therapies including direct exposure to antigens (peptide or DNA) in combination with immune-stimulating molecules as well as stimulated patient-derived antigen presenting cells (dendritic cells (DC)). GBM antigen targets are most often tumor-associated antigens given GBM-specific antigens are rare. Some of these antigens are restricted by HLA types, limiting the patient population in which these vaccines may be considered. Use of whole tumor lysate as an antigen was lethal when studied in animal models [36]; however, modifications to GBM tumor lysate, such as heat shock proteins (HSP) and DC vaccines, have been well tolerated with promising early results. Tumor antigen vaccines and customized vaccines will be discussed separately below.

Tumor antigen vaccines

Cancer tumor antigens may be tumor-specific or tumor-associated antigens. At this time, the best studied tumor-specific antigen is a constitutively activated mutation of epidermal growth factor (EGFR), EGFRvIII. The EGFRvIII mutation is reported in 25–30% of GBM and was thought to be an independent negative prognostic factor [37]. Several forms of EGFRvIII vaccines were studied in phase 1 and 2 clinical trials with promising results. This leads to the development of rindopepimut, a conjugated EGFRvIII-specific peptide (also known as CDX-110 and PEPvIII), by Celldex therapeutics. Phase 1 and 2 clinical trials demonstrated promising mOS compared to historical controls leading to an international phase 3, ACT IV, clinical trial in newly diagnosed GBM patients with EGFRvIII mutation. This randomized study of rindopepimut versus control (KLH), added to the SOC adjuvant temozolomide, showed an impressive mOS of 20.1 months in the vaccine arm. However, the control of KLH alone had 20-month mOS, far exceeding the historical controls of 15.2 months. This called into question EGFRvIII as a negative prognostic biomarker. Interestingly, loss of antigen was seen in both the treatment and control arms [38••]. A phase 2 study of rindopepimut in rGBM in combination with BEV does favor treatment with mOS of 12 months in the vaccine group compared to 8.8 months in the KLH group. This finding suggests that combination of vaccine with anti-VEGF therapy may be necessary for a single antigen vaccine to demonstrate survival benefit. At this point, the future of studies of rindopepimut is unclear; however, there are ongoing CAR-T studies targeting this tumor-specific antigen (discussed below).
A number of tumor-associated antigens are being studied in GBM, and while not specific to tumor cells, limited expression elsewhere makes these safe targets for study [39]. Another single antigen vaccine with early promise is SurVaxM, a peptide mimic of survivin conjugated to KLH. While survivin is expressed in the majority of GBM, the vaccine is HLA-restricted, limiting patient inclusion [40]. Phase 2 results of this vaccine added to the SOC adjuvant temozolomide in newly diagnosed GBM were presented at SNO 2018. While immature, mOS in this single arm phase 2 study was a promising 26 months [41]. A randomized phase 2 for new diagnosis GBM of the combination of SurVaxM with PD1 blockade is planned.
There are several other peptide vaccines targeting multiple antigens. Another recently presented vaccine, SL701, consists of short synthetic peptides targeting IL-13Ra2, ephrin A2, and survivin. This is also an HLA-A2-restricted vaccine that has been studied in the first recurrence of GBM, and was found to have a mOS of 12 months in the second phase of this study. Target-specific CD8 response seen in 8/28 patients was associated with longer survival [42].
A six synthetic peptide stimulated DC vaccine with initial promise was ICT-107. In a phase 1 trial, it was found to be safe with a suggestion of benefit to patients who were HLA-A2 positive. STING (NCT 02546102) was a phase 3 clinical trial that opened in 2016, but later suspended in 2017 due to funding. There are many other ongoing studies of vaccines targeting tumor-associated antigens in GBM using peptide, DNA with immune stimulants as well as stimulated DC cells. A review of completed vaccine trials in GBM was recently published this year by Michael Lim [39].

Customized vaccines

For patients who have surgically accessible disease, custom vaccines are a promising area of clinical research. These vaccines require a minimum volume of resectable tumor to generate a custom vaccine, which limits the population of eligible patients.
DC-Vax-L uses whole tumor lysate to pulse patient-derived DCs. Currently over 10 years from diagnosis, some of the patients enrolled in the original phase 1 study of this vaccine are still alive [43]. The phase 3 of DC-Vax-L in newly diagnosed GBM results is still blinded; however, recent reports described a mOS of 23.1 months for all participants (90% of whom received the DC-Vax-L treatment due to crossover design). While the data remains blinded, there are concerns that this may only be interpreted as a single-arm study of 331 patients due to cross over as a result of pseudo-progression and not true progression [44•]. Again promising are reports of durable responders in the phase 3 with survival exceeding 7 years. This vaccine is also being studied in a phase 2 clinical trial in combination with the PD-1 inhibitor, nivo (NCT03014804).
Another promising custom vaccine being studied for new diagnosis GBM is HSPPC-96 (Prophage). In a single-arm phase 2 trial, mOS was 23.8 months; however, when patients were separated by PD-L1 expression on myeloid cells, mOS for those with low expression was an impressive 44.7 months [45]. HSPPC-96 is currently being studied in combination with pembro for newly diagnosed GBM. HSPPC-96 was also studied in 41 patients with rGBM with a mOS of 42.6 weeks [46]. A summary of key vaccine clinical trials in GBM is provided in Table 2.
Table 2
Key Vaccine Clinical Trials in GBM
Vaccine
Vaccine Category
NCT
Disease
Additional therapy
Control
Phase
n
Status
Results (treatment v. control)
Rindopepimut
Peptide
NCT01480479 (ACT IV)
new diagnosis GBM
GM-CSF + TMZ
KLH + TMZ
3
745
Terminated early (11/2016)
mOS 20.1 months v. 20.0 months
NCT00458601 ACT III
new diagnosis GBM
GM-CSF + TMZ
none
2
82
Completed 5/2016
mOS 21.8m
NCT01498328 (ReACT)
recurrent GBM
GM-CSF + BEV
KLH + BEV
2
127
Completed 2015
mOS 12.0m v. 8.8m
DC-Vax
DC vaccine autologus tumor pulsed
NCT00045968
new diagnosis GBM
 
unpulsed PBMC
3
348
Active, not recruiting
 
NCT03014804
recurrent GBM
Nivolumab
vaccine alone
2
30
planned to open 1/2019
 
NCT02146066
expanded access for patients who sceen-failed
      
HSPPC-96
Custom Peptide
NCT03018288
new diagnosis GBM (MGMT -, IDH wt)
pembrolizumab + TMZ
pembrolizumab + TMZ
2
108
Ongoing
 
NCT00905060
new diagnosis GBM
TMZ
none
2
46
completed
mOS 23.8m
NCT00293423
GTR recurrent GBM
TMZ
none
2
41
completed
mOS 10.65m
NCT01814813
recurrent GBM
BEV
BEV
2
90
not recruiting
 
NCT02722512
new and recurrent HGG and recurrent ependymoma 3-21 years
  
1
20
  
pp65 DC
DC - peptide pulsed CMV antigens
NCT02465268 (ATTAC-II)
new diagnosis GBM
GM-CSF + Td +TMZ
unpulsed PBMC
2
150
recruiting planned to open
 
NCT03688178 (DERIVc)
new diagnosis GBM
variliumab
vaccine alone
2
112
1/2019
 
NCT03615404
new diagnosis GBM <18 years old
GM-CSF + Td +TMZ
none
1
10
recruiting
 
NCT00639639
new diagnosis GBM
Td + TMZ
none
1
16
completed
 
NCT02529072 (AVERT)
recurrent HGG
nivolumab
 
1
7
not recruiting
 
ICT-107
DC -peptide pulsed
NCT01280552
new diagnosis GBM
TMZ
unpulsed DC cells
2
124
completed
mOS 18.3 v. 16.7m
NCT 02546102
new diagnosis GBM
TMZ
Control injection
3
414
suspended due to funding
 
TVI-Brain-1
DC stimulated killer T cells
NCT01290692
recurrent GBM
GM-CSF
 
2
86
completed 2/2014
Company website notes patient 5 years survivor and mOS 50% greater than historical controls
GTR gross total resection, rGBM recurrent GBM, TMZ temozolomide, BEV bevacizumab, nivo nivolumab, pembro pembrolizumab, mOS median overall survival, Td Tetanus-Diphtheria Toxoid, DC dendritic cell, KLH Keyhole limpet hemocyanin, HGG high grade glioma

Vaccine biomarkers

At this time, tumor-associated and tumor-specific antigen vaccines require confirmation that the tumor expresses the targeted antigen. Additionally, many tumor antigens are restricted to specific HLA types (class I restricted cytotoxic T cell or class II restricted helper T cell epitopes), for example HER-2, IL13Ra2, MAGE-1, and survivin. For these tumor-associated antigen vaccines, effective vaccines require tumor antigens that are presented on these restricted HLA alleles to generate an immune response [47]. This narrows the generalizability of these vaccines and clinical trials restrict enrollment to patients with the specific HLA alleles of interest, or stratify results based on HLA type.
Outside of antigen expression and HLA subtyping, there are not prospective biomarkers for response to GBM vaccines. All vaccine clinical trials are looking for correlates to predict immune response and hopefully survival response. Some of those that have been examined include IL-12 production [48], myeloid PD-L1 expression [45], T-reg CTLA-4 expression [49], circulating exomes [50, 51], and antibody production and CD8 response [52].
Additionally, as the DC-Vax-L clinical trial has demonstrated, our current imaging markers have undergone dramatic revolution in the past 10 years limiting the use of MRI for determining progression-free survival as a clinical trial outcome [53•].

CAR-T

CAR-T cell therapy is a newer therapy in oncology, currently approved in B cell lymphoma and leukemia [54]. In brief, CAR-T cells are autologous or allogeneic T cells modified such that the extracellular domains recognizes a unique tumor-associated antigen and the intracellular domain contains a T cell activation signal [55•]. These modified T cells are then administered into the patient, where they can initiate targeted lysis of cells bearing the associated tumor antigen [56]. CAR-T cell therapy has the advantage of bypassing the need for MHC presentation of antigen and development of adaptive immune response as well as bypassing the need for co-stimulatory signals.
Given the success with B cell lymphomas and leukemias, the potential of CAR-T cell therapy for solid tumors, including GBM, has been undergoing investigation [57]. To date, GBM patients treated with CAR-T cell therapy have not had unmanageable CNS effects, a concern given the side effect of elevated intracranial pressure and associated encephalopathy seen in CAR-T cell therapy in B cell lymphoma [58]. The tumor antigens that have been most investigated for CAR targets in GBM to date are IL-13Ra2, EGFRvIII, and Her2.
Interleukin 13 receptor alpha 2 (IL-13Ra2) modulates activation along the rapamycin pathway, and is typically associated with worsened prognosis in GBM [59, 60]. A safety and efficacy trial of CAR T cell therapy in GBM with IL-13Ra2 as the tumor marker was performed on a group of three patients who had a post-resection intracranial infusion, followed by a trial of post-resection direct intratumoral infusion followed intraventricular infusion [61, 62•]. One patient had a dramatic response with clinical and radiographic response that lasted for 7.5 months; however, his disease ultimately did recur [62•]. This CAR-T target is continuing to be studied in intratumoral, intraventricular, and dual delivery systems.
EGFRvIII, as described above in the vaccine section, has also been used a CAR-T cell therapy target. A study regarding tumor infiltration of CAR-T cell performed on ten patients who were given a single peripheral CAR-T cell infusion [63]. Brain specimens in the two patients who underwent post-infusion resection showed increased intratumoral EGFRvIIICAR-T DNA compared to peripheral blood after 2 weeks, and most patients showed decreased expression of EGFRvIII in tumors resected after infusion, suggesting infiltration of the CAR-T cells into the tumor. Patients did not experience tumor regression, although it is notable that this was a particularly poor prognostic group (MGMT negative and most with multifocal disease).
Human epidermal growth factor receptor 2 (HER-2), a tyrosine kinase receptor with high expression in some forms of GBM, has also been used as a potential target [64, 65]. Peripheral infusion of virus-specific (CMV seropositive) HER-2 CAR-T cells demonstrated relative safety of this method as well as persistence of HER-2 CAR-T cells over time (measured for 1 year) [66•]. Seven of the 17 patients did have a period of 8 weeks to 29 months of stable disease and one patient had partial response [66•].
There are currently several active trials investigating the range of intracavitary, intraventricular, and intravenous modes of administration of CAR-T cells for the antigens above as well as EphA2 (NCT02575261). Current studies suggest encouraging results regarding safety and penetrance of CAR-T cells into GBM, although findings regarding effect on tumor growth and recurrence are less conclusive.
No clear biomarkers for treatment response have been established for CAR-T in GBM. In other malignancies, mechanisms of response and resistance have been studied. It is reasonable to consider that similar biomarkers will be found for CAR-T for GBM [6769]. Given at this time, CAR-T cell therapy is targeting single antigens, antigen escape will likely limit effectiveness of CAR-T monotherapy.

Viral therapy

Viral therapy, while initially designed as a mechanism of gene delivery to provide tumor cells with susceptibility to chemotherapy, is now recognized as a form of immunotherapy. Infection of tumor cells with virus attracts the innate immune system leading to cytokine release and tumor cell lysis. This promotes generation of an adaptive immune response to new tumor antigens and potentially development of a long-term immunotherapy effect [70]. While no proven survival benefit has been shown, the excitement about this therapy is largely driven by the population of long-term survivors which was recently reviewed [12•].
Two of these therapies have made it to phase 3 clinical trials, ASPECT and Toca5. The first, ASPECT, studied a replication defective adenovirus, sitimagene ceradenovec, in newly diagnosed GBM [71]. This clinical trial enrolled during early 2000s when the neuro-oncology community was transitioning to the current SOC of adjuvant TMZ. As a result, not all patients within the trial were treated with what is now considered SOC. While there was prolonged time to death or re-intervention in the patients treated with virus, there was no difference in mOS.
The Toca5 clinical trial is comparing Toca-511 (a non-lytic retrovirus expressing cytosine deaminase) to standard therapies for recurrent high grade gliomas. This trial recently completed enrollment; however, phase 3 data is not yet available. If the patients in the phase 1 study were narrowed to patients who would have been eligible for the phase 3, there were 5 patients out of 23 who had durable responses (defined as greater than 24 weeks) and as of August 2017, all of those patients were still alive, one over 4 years [72].
Several other viral therapies have reported GBM patients with durable responses [12•]. These include replication competent HSV1 (G207), parvovirus (ParvOryx01), adenovirus (DNX-2401), and poliovirus (PVSRIPO). Long-term survivors are also reported in another adenovirus that functions as means of gene delivery to allow local delivery of IL-12 [73].
Ongoing study of most of these viruses is now including safety for the combination of viral delivery with checkpoint inhibition.

Conclusion

The success of immunotherapy in GBM faces several obstacles including the highly immunosuppressive nature of GBM and the limitations of the immune response in the central nervous system. Learning from phase 3 clinical trial failures, the future of immunotherapy for GBM appears most hopeful for combination therapies driven by biomarkers for appropriate patient selection. Given the extreme need for improved survival in GBM, current clinical trials are evaluating checkpoint inhibition in combination with novel therapies including vaccines, CAR-T cell therapy, and viral therapy.

Compliance with Ethical Standards

Conflict of Interest

Tresa McGranahan declares that she has no conflict of interest. Kate Elizabeth Therkelsen declares that she has no conflict of interest. Sarah Ahmad declares that she has no conflict of interest. Seema Nagpal has received clinical trial funding from Inovio/Regeneron and Tocagen, has received research support from Nektar Therapeutics, and has received compensation from Nektar Therapeutics for service as a consultant.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat • Zhu P, Du XL, Lu G, Zhu J-J. Survival benefit of glioblastoma patients after FDA approval of temozolomide concomitant with radiation and bevacizumab: a population-based study. Oncotarget. 2017;8:44015–31 Use of SEER and Texas Cancer Registry databases to confirm population-based increased survival for patients with GBM since advances in care of adjuvant temozolomide.PubMedPubMedCentral • Zhu P, Du XL, Lu G, Zhu J-J. Survival benefit of glioblastoma patients after FDA approval of temozolomide concomitant with radiation and bevacizumab: a population-based study. Oncotarget. 2017;8:44015–31 Use of SEER and Texas Cancer Registry databases to confirm population-based increased survival for patients with GBM since advances in care of adjuvant temozolomide.PubMedPubMedCentral
2.
Zurück zum Zitat •• Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318:2306–16 Positive phase 3 clinical trial with use of TTF resulted in increase in overall survival as well as 13% 5-year survival for patients with new diagnosis GBM.CrossRef •• Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318:2306–16 Positive phase 3 clinical trial with use of TTF resulted in increase in overall survival as well as 13% 5-year survival for patients with new diagnosis GBM.CrossRef
3.
Zurück zum Zitat Chongsathidkiet P, Jackson C, Koyama S, Loebel F, Cui X, Farber SH, et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat Med. 2018;24:1459–68.CrossRef Chongsathidkiet P, Jackson C, Koyama S, Loebel F, Cui X, Farber SH, et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat Med. 2018;24:1459–68.CrossRef
4.
Zurück zum Zitat Cristescu R, Mogg R, Ayers M, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science (New York, NY). 2018;362. Cristescu R, Mogg R, Ayers M, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science (New York, NY). 2018;362.
5.
Zurück zum Zitat Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018;48:812–30.e14.CrossRef Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018;48:812–30.e14.CrossRef
6.
Zurück zum Zitat Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science (New York, NY). 2014;344:1396–401.CrossRef Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science (New York, NY). 2014;344:1396–401.CrossRef
7.
Zurück zum Zitat Reardon D, Neuberg D, Keskin D, et al. Effect of dexamethasone in glioblastoma (GBM) patients on systemic and intratumoral T-cell responses induced by personalized neoantigen-targeting vaccine. Chicago: ASCO; 2018.CrossRef Reardon D, Neuberg D, Keskin D, et al. Effect of dexamethasone in glioblastoma (GBM) patients on systemic and intratumoral T-cell responses induced by personalized neoantigen-targeting vaccine. Chicago: ASCO; 2018.CrossRef
8.
Zurück zum Zitat Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, Ni A, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol. 2018;36:2872–8.CrossRef Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, Ni A, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol. 2018;36:2872–8.CrossRef
9.
Zurück zum Zitat Gust J, Hay KA, Hanafi LA, Li D, Myerson D, Gonzalez-Cuyar LF, et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;7:1404–19.CrossRef Gust J, Hay KA, Hanafi LA, Li D, Myerson D, Gonzalez-Cuyar LF, et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;7:1404–19.CrossRef
10.
Zurück zum Zitat Schneider S, Potthast S, Komminoth P, Schwegler G, Böhm S. PD-1 checkpoint inhibitor associated autoimmune encephalitis. Case Rep Oncol. 2017;10:473–8.CrossRef Schneider S, Potthast S, Komminoth P, Schwegler G, Böhm S. PD-1 checkpoint inhibitor associated autoimmune encephalitis. Case Rep Oncol. 2017;10:473–8.CrossRef
11.
Zurück zum Zitat Leitinger M, Varosanec MV, Pikija S, Wass RE, Bandke D, Weis S, et al. Fatal necrotizing encephalopathy after treatment with nivolumab for squamous non-small cell lung cancer: case report and review of the literature. Front Immunol. 2018;9:108.CrossRef Leitinger M, Varosanec MV, Pikija S, Wass RE, Bandke D, Weis S, et al. Fatal necrotizing encephalopathy after treatment with nivolumab for squamous non-small cell lung cancer: case report and review of the literature. Front Immunol. 2018;9:108.CrossRef
12.•
Zurück zum Zitat Chiocca EA, Nassiri F, Wang J, Peruzzi P, Zadeh G. Viral and other therapies for recurrent GBM: is a 24-month durable response unusual? Neuro Oncol. 2018. Meta-analysis examining long-term survivors in prior GBM clinical trials and viral therapy for GBM. Chiocca EA, Nassiri F, Wang J, Peruzzi P, Zadeh G. Viral and other therapies for recurrent GBM: is a 24-month durable response unusual? Neuro Oncol. 2018. Meta-analysis examining long-term survivors in prior GBM clinical trials and viral therapy for GBM.
13.
Zurück zum Zitat McGranahan T, Li G, Nagpal S. History and current state of immunotherapy in glioma and brain metastasis. Ther Adva Med Oncol. 2017;9:347–68.CrossRef McGranahan T, Li G, Nagpal S. History and current state of immunotherapy in glioma and brain metastasis. Ther Adva Med Oncol. 2017;9:347–68.CrossRef
14.
Zurück zum Zitat Preusser M, Lim M, Hafler DA, Reardon DA, Sampson JH. Prospects of immune checkpoint modulators in the treatment of glioblastoma. Nat Rev Neurol. 2015;11:504–14.CrossRef Preusser M, Lim M, Hafler DA, Reardon DA, Sampson JH. Prospects of immune checkpoint modulators in the treatment of glioblastoma. Nat Rev Neurol. 2015;11:504–14.CrossRef
15.
Zurück zum Zitat Cuzzubbo S, Javeri F, Tissier M, Roumi A, Barlog C, Doridam J, et al. Neurological adverse events associated with immune checkpoint inhibitors: review of the literature. Eur J Cancer. 2017;73:1–8.CrossRef Cuzzubbo S, Javeri F, Tissier M, Roumi A, Barlog C, Doridam J, et al. Neurological adverse events associated with immune checkpoint inhibitors: review of the literature. Eur J Cancer. 2017;73:1–8.CrossRef
16.
Zurück zum Zitat Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J, Cloughesy T, et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro-Oncology. 2018;20:674–86.CrossRef Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J, Cloughesy T, et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro-Oncology. 2018;20:674–86.CrossRef
17.
Zurück zum Zitat Reardon D, Omuro A, Brandes A, et al. OS10.3 randomized phase 3 study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent glioblastoma: CheckMate 143. Pheonix: Society for Neuro Oncology; 2017. Reardon D, Omuro A, Brandes A, et al. OS10.3 randomized phase 3 study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent glioblastoma: CheckMate 143. Pheonix: Society for Neuro Oncology; 2017.
18.
Zurück zum Zitat Reiss SN, Yerram P, Modelevsky L, Grommes C. Retrospective review of safety and efficacy of programmed cell death-1 inhibitors in refractory high grade gliomas. J Immunother Cancer. 2017;5:99.CrossRef Reiss SN, Yerram P, Modelevsky L, Grommes C. Retrospective review of safety and efficacy of programmed cell death-1 inhibitors in refractory high grade gliomas. J Immunother Cancer. 2017;5:99.CrossRef
19.
Zurück zum Zitat Maxwell R, Jackson CM, Lim M. Clinical trials investigating immune checkpoint blockade in glioblastoma. Curr Treat Options in Oncol. 2017;18:51.CrossRef Maxwell R, Jackson CM, Lim M. Clinical trials investigating immune checkpoint blockade in glioblastoma. Curr Treat Options in Oncol. 2017;18:51.CrossRef
20.
Zurück zum Zitat Necchi A, Anichini A, Raggi D, et al. Pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase ii study. J Clin Oncol. 2018; 0:JCO.18.01148. Necchi A, Anichini A, Raggi D, et al. Pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase ii study. J Clin Oncol. 2018; 0:JCO.18.01148.
21.
Zurück zum Zitat Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 2018;378:1976–86.CrossRef Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 2018;378:1976–86.CrossRef
22.
Zurück zum Zitat Prins RMA, Orpilla J, Lee A, Davidson T, Gaffey S, Sanders C, et al. Neoadjuvant anti-PD-1 immunotherapy promotes intratumoral and systemic immune responses in recurrent glioblastoma: An Ivy Consortium trial. New Orleans: Society for NeuroOncology; 2018. Prins RMA, Orpilla J, Lee A, Davidson T, Gaffey S, Sanders C, et al. Neoadjuvant anti-PD-1 immunotherapy promotes intratumoral and systemic immune responses in recurrent glioblastoma: An Ivy Consortium trial. New Orleans: Society for NeuroOncology; 2018.
23.
Zurück zum Zitat Huang Y, Goel S, Duda DG, Fukumura D, Jain RK. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res. 2013;73:2943–8.CrossRef Huang Y, Goel S, Duda DG, Fukumura D, Jain RK. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res. 2013;73:2943–8.CrossRef
24.
Zurück zum Zitat Hodi FS, Lawrence D, Lezcano C, Wu X, Zhou J, Sasada T, et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014;2:632–42.CrossRef Hodi FS, Lawrence D, Lezcano C, Wu X, Zhou J, Sasada T, et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014;2:632–42.CrossRef
25.
Zurück zum Zitat Pitter KL, Tamagno I, Alikhanyan K, Hosni-Ahmed A, Pattwell SS, Donnola S, et al. Corticosteroids compromise survival in glioblastoma. Brain J Neurol. 2016;139:1458–71.CrossRef Pitter KL, Tamagno I, Alikhanyan K, Hosni-Ahmed A, Pattwell SS, Donnola S, et al. Corticosteroids compromise survival in glioblastoma. Brain J Neurol. 2016;139:1458–71.CrossRef
26.
Zurück zum Zitat Ajlan A, Thomas P, Albakr A, Nagpal S, Recht L. Optimizing bevacizumab dosing in glioblastoma: less is more. J Neuro-Oncol. 2017;135:99–105.CrossRef Ajlan A, Thomas P, Albakr A, Nagpal S, Recht L. Optimizing bevacizumab dosing in glioblastoma: less is more. J Neuro-Oncol. 2017;135:99–105.CrossRef
27.
Zurück zum Zitat Reardon DA, Groot JFD, Colman H, et al. Safety of pembrolizumab in combination with bevacizumab in recurrent glioblastoma (rGBM). J Clin Oncol. 2016;34:2010.CrossRef Reardon DA, Groot JFD, Colman H, et al. Safety of pembrolizumab in combination with bevacizumab in recurrent glioblastoma (rGBM). J Clin Oncol. 2016;34:2010.CrossRef
28.
Zurück zum Zitat Lopes G, Wu Y, Kudaba I, et al. Pembrolizumab (pembro) versus platinum-based chemotherapy (chemo) as first-line therapy for advanced/metastatic NSCLC with a PD-L1 tumor proportion score (TPS) ≥ 1%: open-label, phase 3 KEYNOTE-042 study. Chicago: ASCO; 2018. Lopes G, Wu Y, Kudaba I, et al. Pembrolizumab (pembro) versus platinum-based chemotherapy (chemo) as first-line therapy for advanced/metastatic NSCLC with a PD-L1 tumor proportion score (TPS) ≥ 1%: open-label, phase 3 KEYNOTE-042 study. Chicago: ASCO; 2018.
29.
Zurück zum Zitat Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–39.CrossRef Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–39.CrossRef
30.
Zurück zum Zitat Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, Gabrusiewicz K, et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncology. 2016;18:195–205.CrossRef Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, Gabrusiewicz K, et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncology. 2016;18:195–205.CrossRef
31.
Zurück zum Zitat Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, Wöhrer A, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro-Oncology. 2015;17:1064–75.CrossRef Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, Wöhrer A, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro-Oncology. 2015;17:1064–75.CrossRef
32.
Zurück zum Zitat De Groot JF, Penas-Prado M, Mandel JJ, et al. Window-of-opportunity clinical trial of a PD-1 inhibitor in patients with recurrent glioblastoma. Chicago: ASCO; 2017. De Groot JF, Penas-Prado M, Mandel JJ, et al. Window-of-opportunity clinical trial of a PD-1 inhibitor in patients with recurrent glioblastoma. Chicago: ASCO; 2017.
33.
Zurück zum Zitat Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16:2598–608.CrossRef Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16:2598–608.CrossRef
34.
Zurück zum Zitat Hodges TR, Ott M, Xiu J, Gatalica Z, Swensen J, Zhou S, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro-Oncology. 2017;19:1047–57.CrossRef Hodges TR, Ott M, Xiu J, Gatalica Z, Swensen J, Zhou S, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro-Oncology. 2017;19:1047–57.CrossRef
35.
Zurück zum Zitat Martinez R, Schackert HK, Appelt H, Plaschke J, Baretton G, Schackert G. Low-level microsatellite instability phenotype in sporadic glioblastoma multiforme. J Cancer Res Clin Oncol. 2005;131:87–93.CrossRef Martinez R, Schackert HK, Appelt H, Plaschke J, Baretton G, Schackert G. Low-level microsatellite instability phenotype in sporadic glioblastoma multiforme. J Cancer Res Clin Oncol. 2005;131:87–93.CrossRef
36.
Zurück zum Zitat Bigner DD, Pitts CM, Wikstrand CJ. Induction of lethal experimental allergic encephalomyelitis in nonhuman primates and guinea pigs with human glioblastoma multiforme tissue. J Neurosurg. 1981;55:32–42.CrossRef Bigner DD, Pitts CM, Wikstrand CJ. Induction of lethal experimental allergic encephalomyelitis in nonhuman primates and guinea pigs with human glioblastoma multiforme tissue. J Neurosurg. 1981;55:32–42.CrossRef
37.
Zurück zum Zitat Weller M, Kaulich K, Hentschel B, Felsberg J, Gramatzki D, Pietsch T, et al. Assessment and prognostic significance of the epidermal growth factor receptor VIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy. Int J Cancer. 2014;134:2437–47.CrossRef Weller M, Kaulich K, Hentschel B, Felsberg J, Gramatzki D, Pietsch T, et al. Assessment and prognostic significance of the epidermal growth factor receptor VIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy. Int J Cancer. 2014;134:2437–47.CrossRef
38.
Zurück zum Zitat •• Weller M, Butowski N, Tran DD, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017;18:1373–85 Phase III trial of EGFRvIII vaccine, terminated early due to lack of difference in overall survival, despite earlier phase II trial showing a robust anti-EGFRvIII antibody response (suggesting that a humoral response may not be an adequate biomarker of outcome).CrossRef •• Weller M, Butowski N, Tran DD, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017;18:1373–85 Phase III trial of EGFRvIII vaccine, terminated early due to lack of difference in overall survival, despite earlier phase II trial showing a robust anti-EGFRvIII antibody response (suggesting that a humoral response may not be an adequate biomarker of outcome).CrossRef
39.
Zurück zum Zitat Lim M, Xia Y, Bettegowda C, Weller M. Current state of immunotherapy for glioblastoma. Nat Rev Clin Oncol. 2018;15:422–42.CrossRef Lim M, Xia Y, Bettegowda C, Weller M. Current state of immunotherapy for glioblastoma. Nat Rev Clin Oncol. 2018;15:422–42.CrossRef
40.
Zurück zum Zitat Ellis JM, Henson V, Slack R, Ng J, Hartzman RJ, Hurley CK. Frequencies of HLA-A2 Alleles in Five U.S. Population Groups. Predominance of A*02011 and Identification of HLA-A*0231. Ellis JM, Henson V, Slack R, Ng J, Hartzman RJ, Hurley CK. Frequencies of HLA-A2 Alleles in Five U.S. Population Groups. Predominance of A*02011 and Identification of HLA-A*0231.
41.
Zurück zum Zitat Ahluwalia MRD, Abad A, Curry W, Wong E, Peereboom D, Belal A, et al. Fenstermaker R. Phase II trial of a survivin vaccine (SurVaxM) for newly diagnosed glioblastoma. New Orleans: Society for Neuro Oncology; 2018. Ahluwalia MRD, Abad A, Curry W, Wong E, Peereboom D, Belal A, et al. Fenstermaker R. Phase II trial of a survivin vaccine (SurVaxM) for newly diagnosed glioblastoma. New Orleans: Society for Neuro Oncology; 2018.
42.
Zurück zum Zitat Peereboom DNL, Kumthekar P, Badruddoja M, Fink K, Lieberman F, Phuphanich S, et al. Phase 2 trial of SL-701 + bevacizumab in patients with previously treated glioblastoma (GBM) meets primary endpoint of OS-12, with preliminary correlation between long-term survival and target-specific CD8+ T cell immune response. New Orleans: Society for Neuro Oncology; 2018.CrossRef Peereboom DNL, Kumthekar P, Badruddoja M, Fink K, Lieberman F, Phuphanich S, et al. Phase 2 trial of SL-701 + bevacizumab in patients with previously treated glioblastoma (GBM) meets primary endpoint of OS-12, with preliminary correlation between long-term survival and target-specific CD8+ T cell immune response. New Orleans: Society for Neuro Oncology; 2018.CrossRef
43.
Zurück zum Zitat Liau LM. Brain tumor immunotherapy: lessons learned and future directions. New Orleans: Society for Neuro Oncology; 2018. Liau LM. Brain tumor immunotherapy: lessons learned and future directions. New Orleans: Society for Neuro Oncology; 2018.
44.
Zurück zum Zitat • Liau LM, Ashkan K, Tran DD, et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. 2018;16:142 Unblinded survival data from phase 3 clinical trial of DC-VAX-L tumor lysate vaccine.CrossRef • Liau LM, Ashkan K, Tran DD, et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. 2018;16:142 Unblinded survival data from phase 3 clinical trial of DC-VAX-L tumor lysate vaccine.CrossRef
45.
Zurück zum Zitat Bloch O, Lim M, Sughrue ME, Komotar RJ, Abrahams JM, O'Rourke DM, et al. Autologous heat shock protein peptide vaccination for newly diagnosed glioblastoma: impact of peripheral PD-L1 expression on response to therapy. Clin Cancer Res. 2017;23:3575–84.CrossRef Bloch O, Lim M, Sughrue ME, Komotar RJ, Abrahams JM, O'Rourke DM, et al. Autologous heat shock protein peptide vaccination for newly diagnosed glioblastoma: impact of peripheral PD-L1 expression on response to therapy. Clin Cancer Res. 2017;23:3575–84.CrossRef
46.
Zurück zum Zitat Bloch O, Crane CA, Fuks Y, Kaur R, Aghi MK, Berger MS, et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro-Oncology. 2014;16:274–9.CrossRef Bloch O, Crane CA, Fuks Y, Kaur R, Aghi MK, Berger MS, et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro-Oncology. 2014;16:274–9.CrossRef
47.
Zurück zum Zitat Zhang JG, Eguchi J, Kruse CA, Gomez GG, Fakhrai H, Schroter S, et al. Antigenic profiling of glioma cells to generate allogeneic vaccines or dendritic cell-based therapeutics. Clin Cancer Res. 2007;13:566–75.CrossRef Zhang JG, Eguchi J, Kruse CA, Gomez GG, Fakhrai H, Schroter S, et al. Antigenic profiling of glioma cells to generate allogeneic vaccines or dendritic cell-based therapeutics. Clin Cancer Res. 2007;13:566–75.CrossRef
48.
Zurück zum Zitat Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29:330–6.CrossRef Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29:330–6.CrossRef
49.
Zurück zum Zitat Fong B, Jin R, Wang X, Safaee M, Lisiero DN, Yang I, 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:e32614.CrossRef Fong B, Jin R, Wang X, Safaee M, Lisiero DN, Yang I, 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:e32614.CrossRef
50.
Zurück zum Zitat Galbo PM Jr, Ciesielski MJ, Figel S, et al. Circulating CD9+/GFAP+/survivin+ exosomes in malignant glioma patients following survivin vaccination. Oncotarget. 2017;8:114722–35.CrossRef Galbo PM Jr, Ciesielski MJ, Figel S, et al. Circulating CD9+/GFAP+/survivin+ exosomes in malignant glioma patients following survivin vaccination. Oncotarget. 2017;8:114722–35.CrossRef
51.
Zurück zum Zitat Manterola L, Guruceaga E, Gallego Perez-Larraya J, et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-Oncology. 2014;16:520–7.CrossRef Manterola L, Guruceaga E, Gallego Perez-Larraya J, et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-Oncology. 2014;16:520–7.CrossRef
52.
Zurück zum Zitat Schaller TH, Sampson JH. Advances and challenges: dendritic cell vaccination strategies for glioblastoma. Expert Rev Vaccines. 2017;16:27–36.CrossRef Schaller TH, Sampson JH. Advances and challenges: dendritic cell vaccination strategies for glioblastoma. Expert Rev Vaccines. 2017;16:27–36.CrossRef
53.
Zurück zum Zitat • Okada H, Weller M, Huang R, et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 2015;16:e534–e42 RANO criteria for evaluate response to immunotherapy.CrossRef • Okada H, Weller M, Huang R, et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 2015;16:e534–e42 RANO criteria for evaluate response to immunotherapy.CrossRef
54.
Zurück zum Zitat Commissioner Oot. Press announcements - FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma. Commissioner Oot. Press announcements - FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma.
55.•
Zurück zum Zitat Bagley SJ, Desai AS, Linette GP, June CH, O’Rourke DM. CAR T Cell therapy for glioblastoma: recent clinical advances and future challenges. Neuro-Oncology. 2018. An in-depth overview regarding CAR-T cell perceived benefits of CAR T cell therapy for GBM specifically with a comprehensive analysis of published study outcomes as well as discussion of active studies and future considerations. Bagley SJ, Desai AS, Linette GP, June CH, O’Rourke DM. CAR T Cell therapy for glioblastoma: recent clinical advances and future challenges. Neuro-Oncology. 2018. An in-depth overview regarding CAR-T cell perceived benefits of CAR T cell therapy for GBM specifically with a comprehensive analysis of published study outcomes as well as discussion of active studies and future considerations.
56.
Zurück zum Zitat Chen D, Yang J. Development of novel antigen receptors for CAR T-cell therapy directed toward solid malignancies. Transl Res. 2017;187:11–21.CrossRef Chen D, Yang J. Development of novel antigen receptors for CAR T-cell therapy directed toward solid malignancies. Transl Res. 2017;187:11–21.CrossRef
57.
Zurück zum Zitat • Rodriguez A, Brown C, Badie B. Chimeric antigen receptor T-cell therapy for glioblastoma. Transl Res. 2017;187:93–102 A prior review of CAR-T cell therapy in GBM studies which includes a comprehensive and concise discussion of CAR T cell engineering.CrossRef • Rodriguez A, Brown C, Badie B. Chimeric antigen receptor T-cell therapy for glioblastoma. Transl Res. 2017;187:93–102 A prior review of CAR-T cell therapy in GBM studies which includes a comprehensive and concise discussion of CAR T cell engineering.CrossRef
58.
Zurück zum Zitat Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T-cell therapy — assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15:47–62.CrossRef Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T-cell therapy — assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15:47–62.CrossRef
59.
Zurück zum Zitat Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro-Oncology. 2014;16:1304–12.CrossRef Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro-Oncology. 2014;16:1304–12.CrossRef
60.
Zurück zum Zitat Brown CE, Starr R, Aguilar B, Shami AF, Martinez C, D’Apuzzo M, et al. Stem-like tumor-initiating cells isolated from IL13Rα2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T cells. Clin Cancer Res. 2012;18:2199–209.CrossRef Brown CE, Starr R, Aguilar B, Shami AF, Martinez C, D’Apuzzo M, et al. Stem-like tumor-initiating cells isolated from IL13Rα2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T cells. Clin Cancer Res. 2012;18:2199–209.CrossRef
61.
Zurück zum Zitat Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, et al. Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin Cancer Res. 2015;21:4062–72.CrossRef Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, et al. Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin Cancer Res. 2015;21:4062–72.CrossRef
62.
Zurück zum Zitat • Brown CE, Alizadeh D, Starr R, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375:2561–9 Case report of a patient with multifocal GBM with a dramatic radiographic response to CAR-T cell therapy.CrossRef • Brown CE, Alizadeh D, Starr R, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375:2561–9 Case report of a patient with multifocal GBM with a dramatic radiographic response to CAR-T cell therapy.CrossRef
63.
Zurück zum Zitat O'Rourke DM, Nasrallah MP, Desai A, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9. O'Rourke DM, Nasrallah MP, Desai A, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9.
64.
Zurück zum Zitat Ahmed N, Salsman VS, Kew Y, Shaffer D, Powell S, Zhang YJ, et al. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res. 2010;16:474–85.CrossRef Ahmed N, Salsman VS, Kew Y, Shaffer D, Powell S, Zhang YJ, et al. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res. 2010;16:474–85.CrossRef
65.
Zurück zum Zitat Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, et al. ErbB2/HER2-specific NK cells for targeted therapy of glioblastoma. J Natl Cancer Inst. 2016;108. Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, et al. ErbB2/HER2-specific NK cells for targeted therapy of glioblastoma. J Natl Cancer Inst. 2016;108.
66.
Zurück zum Zitat • Ahmed N, Brawley V, Hegde M, et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 2017;3:1094–101 This was a phase I trial of 17 patients who were administered peripheral HER-2 virus specific CAR-T cell therapy that demonstrated evidence for relative safety, persistence of HER-2 CAR-T cells in peripheral blood for up to one year, and suggestion of a period of disease stability.CrossRef • Ahmed N, Brawley V, Hegde M, et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 2017;3:1094–101 This was a phase I trial of 17 patients who were administered peripheral HER-2 virus specific CAR-T cell therapy that demonstrated evidence for relative safety, persistence of HER-2 CAR-T cells in peripheral blood for up to one year, and suggestion of a period of disease stability.CrossRef
67.
Zurück zum Zitat Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018;24:563–71.CrossRef Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018;24:563–71.CrossRef
68.
Zurück zum Zitat Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–44.CrossRef Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–44.CrossRef
69.
Zurück zum Zitat Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6.CrossRef Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6.CrossRef
70.
Zurück zum Zitat Peruzzi P, Chiocca EA. Viruses in cancer therapy - from benchwarmers to quarterbacks. Nat Rev Clin Oncol. 2018;15:657–8.CrossRef Peruzzi P, Chiocca EA. Viruses in cancer therapy - from benchwarmers to quarterbacks. Nat Rev Clin Oncol. 2018;15:657–8.CrossRef
71.
Zurück zum Zitat Westphal M, Ylä-Herttuala S, Martin J, Warnke P, Menei P, Eckland D, et al. Adenovirus-mediated gene therapy with sitimagene ceradenovec followed by intravenous ganciclovir for patients with operable high-grade glioma (ASPECT): a randomised, open-label, phase 3 trial. Lancet Oncol. 2013;14:823–33.CrossRef Westphal M, Ylä-Herttuala S, Martin J, Warnke P, Menei P, Eckland D, et al. Adenovirus-mediated gene therapy with sitimagene ceradenovec followed by intravenous ganciclovir for patients with operable high-grade glioma (ASPECT): a randomised, open-label, phase 3 trial. Lancet Oncol. 2013;14:823–33.CrossRef
72.
Zurück zum Zitat Cloughesy TF, Landolfi J, Vogelbaum MA, Ostertag D, Elder JB, Bloomfield S, et al. Durable complete responses in some recurrent high-grade glioma patients treated with Toca 511 + Toca FC. Neuro-Oncology. 2018;20:1383–92.CrossRef Cloughesy TF, Landolfi J, Vogelbaum MA, Ostertag D, Elder JB, Bloomfield S, et al. Durable complete responses in some recurrent high-grade glioma patients treated with Toca 511 + Toca FC. Neuro-Oncology. 2018;20:1383–92.CrossRef
73.
Zurück zum Zitat Barrett JA, Cai H, Miao J, Khare PD, Gonzalez P, Dalsing-Hernandez J, et al. Regulated intratumoral expression of IL-12 using a RheoSwitch therapeutic system((R)) (RTS((R))) gene switch as gene therapy for the treatment of glioma. Cancer Gene Ther. 2018;25:106–16.CrossRef Barrett JA, Cai H, Miao J, Khare PD, Gonzalez P, Dalsing-Hernandez J, et al. Regulated intratumoral expression of IL-12 using a RheoSwitch therapeutic system((R)) (RTS((R))) gene switch as gene therapy for the treatment of glioma. Cancer Gene Ther. 2018;25:106–16.CrossRef
Metadaten
Titel
Current State of Immunotherapy for Treatment of Glioblastoma
verfasst von
Tresa McGranahan, MD PhD
Kate Elizabeth Therkelsen, MD
Sarah Ahmad, MD
Seema Nagpal, MD
Publikationsdatum
01.03.2019
Verlag
Springer US
Erschienen in
Current Treatment Options in Oncology / Ausgabe 3/2019
Print ISSN: 1527-2729
Elektronische ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-019-0619-4

Weitere Artikel der Ausgabe 3/2019

Current Treatment Options in Oncology 3/2019 Zur Ausgabe

Neuro-oncology (GJ Lesser, Section Editor)

Current Treatment Options for Breast Cancer Brain Metastases

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.