Immunotherapy for High-Grade Gliomas


NIH clinical trial

Type

Phase

Eligibility criteria

Primary outcome

Dosing regimen

Treatment groups

NCT02311920

Checkpt

1

Newly diagnosed GBM

Maximum safe dose

In addition to TMZ, Arm I receives anti-CTLA4 Ab ipilimumab once every 4 weeks (4 doses) followed every 3 months (4 doses), Arm II receives anti-PD1 Ab nivolumab once every 2 weeks (32 doses), and Arm III receives both Abs

Arm I: TMZ + Ipi

Arm II: TMZ + Nivo

Arm III: TMZ + Ipi + Nivo

NCT02526017

mAb + checkpt

1

GBM

Safety, RD, efficacy (ORR)

Arm I will receive anti-CSF1R Ab FPA008 every 2 weeks. Arm II will receive 3 mg/kg nivolumab every 2 weeks in addition to dose-escalated FPA008 every 2 weeks. Arm III uses MTD determined in Arm II and expands patient group

Arm I: mAb

Arm II: mAB dose escalation + Nivo

Arm III: mAB + Nivo expansion group

NCT02521090

BiTE

1/2

Recurrent and refractory GBM

Toxicity (phase I), OS (phase II)

Phase I: patients receive anti-EGFR-CD3 BiTE armed T cells IT twice weekly for 4 weeks. Phase II: patients continue to receive BiTE armed T cells IV twice weekly for 2 weeks

Single-arm

NCT02423343

Small molecule + checkpt

1/2

GBM

MTD of galunisertib combined with Nivolumab

In Arm I escalating doses of TGFβR1 kinase small molecule inhibitor galunisertib are given daily for 14 days of each 4 week cycle in combination with nivolumab every 2 weeks. Arm II will use same treatment regimen with MTD galunisertib

Cohort A: dose escalation

Cohort B: expansion group

NCT02530502

Checkpt

1/2

Newly diagnosed GBM

Phase I: DLT

Phase II: PFS

Focal RT over 42 days followed by TMZ on days 1–42 and anti-PD1 antibody pembrolizumab on days 1, 22, and 43 for each course for up to 6 courses

Single-arm

NCT01952769

Checkpt

1/2

Diffuse intrinsic pontine glioma

Treatment-related toxicity

Anti-PD1 antibody given biweekly. Cohort A receives two doses with increasing concentration. Cohort B concentration varies between patients

Cohort A: 2 doses

Cohort B: doses of different concentration

NCT02337686

Checkpt

2

Recurrent GBM

PFS6 and immune effector:Treg ratio

Anti-PD-1 antibody pembrolizumab given once every 3 weeks (2 doses) before surgery and once every 3 weeks thereafter

Single-arm

NCT02337491

Checkpt + mAB

2

Recurrent GBM

PFS6 and MTD

Not described

Cohort A: pembrolizumab + bevacizumab

Cohort B: pembrolizumab

NCT01149850

mAb

2

Newly diagnosed GBM

Overall survival

Anti-VEGF antibody bevacizumab every 2 weeks

Single-arm

NCT02540161

mAb

2

Recurrent GBM

PFS6

Anti-EGFR antibody Sym004 given every 2 weeks at 18 mg/kg

Cohort A: non-bevacizumab failures

Cohort B: bevacizumab failures


Ab antibody; BiTE bispecific T cell engager; Checkpt immune checkpoint modulator; DLT dose-limiting toxicity; GBM glioblastoma; Ipi ipilimumab; IT intrathecal; mAb monoclonal antibody; Nivo nivolumab; OS overall survival; MTD maximum tolerated dose; PFS progression-free survival; PFS6 6-month progression-free survival; ORR objective response rate; RD recommended dose; TMZ temozolomide; Treg regulatory T cell





Bispecific Antibodies Redirect and Activate Effector Immune Cells


Various solid tumors show infiltration with T cells and increased T cell infiltration often correlates with a good clinical outcome [50]. T cell infiltration has also been shown in glioma and is increased in high-grade tumors [51]. Substantial evidence suggests that the redirection of these T cells to specifically recognize and kill tumor cells is able to eradicate well-established tumors [52, 53]. Furthermore, clinical data have shown that mABs suffer from major limitations in their mode of action, including alternative Fc glycosylation, leading to suboptimal effector cell interaction, competition with circulating IgG, and activation of inhibitory receptors [54].

Bispecific antibodies (bsABs) are capable of binding two distinct targets and can be used to link T cells to tumor cells. Bispecific T cell engagers (BiTEs) consist of two antibody-derived linked single-chain Fv fragments (scFv) that are translated in tandem. One arm of the BiTE recognizes, for instance, the CD3 epsilon subunit on the T cell and the other arm binds a tumor antigen (Fig. 12.1). Upon binding, the BiTE causes crosslinking between adjacent tumor cells and T cells, regardless of the T cell receptor recognition, leading to T cell activation, synapse formation, and tumor lysis via perforin and granzyme secretion. Following BiTE-mediated tumor cell lysis, the T cells proliferate, express surface activation markers, and undergo serial rounds of killing [53, 5557]. Furthermore, since crosslinking depends on binding to CD3 epsilon, T cell subsets implicated with tumor progression, such as Tregs, are also activated to lyse tumor cells [58, 59].

A371342_1_En_12_Fig1_HTML.gif


Fig. 12.1
BiTE mode of action. The anti-CD3-EGFRvIII bispecific T cell engager (BiTE) is able to bind the CD3 epsilon subunit of the T cell receptor with one of its single-chain Fv (scFv) fragments and EGFRvIII on the glioma cell with the other scFv fragment. This leads to spatially restricted crosslinking and activation of the T cell, resulting in T cell-mediated tumor cell cytotoxicity via synapse formation and the release of perforin and granzyme

Since T cell activation requires physical linking to a tumor antigen, the immune activation is spatially and temporally restricted and highly specific for the chosen antigen. Furthermore, the small size of the BiTE results in a short half-life that allows quick regulation of antibody-mediated toxicity [60].

A recent clinical trial aims to treat patients with recurrent or refractory glioblastoma with a bispecific antibody made by the heteroconjugation of anti-EGFR and anti-CD3 antibody. Autologous activated T cells are loaded with the anti-EGFR-CD3 BiTE and injected intravenously into the patient with the goal of increasing T cell-mediated cytotoxicity toward tumor expressing EGFR [61]. The aim in this trial will be to determine whether a therapeutic window exists that will allow cell killing of EGFR overexpressing tumor cells without afflicting normal tissue (Table 12.1).

Our laboratory recently developed a BiTE produced by the heteroconjugation of an anti-EGFRvIII and anti-CD3 antibody. Experiments in mice show that systemic administration of the BiTE activates T cells in mice, resulting in extended survival and durable complete cures at rates of up to 75% [62]. Given the tumor specificity of the EGFRvIII antigen, treatment of patients with this antibody may have fewer side effects and increased efficacy.


Immune Checkpoint Modulators


The growth of a tumor is marked by significant changes to the microenvironment, leading to cancer-associated immunosuppression. This means that despite the presence of tumor-specific endogenous T cells, tumors escape destruction by upregulating inhibitory ligands that bind to inhibitory receptors on T cells, secretion of inhibitory cytokines (including TGF-beta and IL-10), and other mechanisms. This immunosuppression is particularly pronounced in glioma patients and leads to T cell dysfunction and an increase in the regulatory T cell phenotype [6366].

Novel strategies for dealing with tumor-associated immunosuppression are the development of antagonistic mABs which block inhibitory ligands, such as CTLA-4, PD-1 and PD-L1, and agonistic mABs that stimulate the immune response by binding agonistic cell surface molecules, such as OX40 and 4-1BB (Fig. 12.2). Recent advances, in particular the FDA approval of the nivolumab–ipilimumab combination for the treatment of metastatic melanoma, highlight the powerful effect and curative potential of immune checkpoint modulators [67].

A371342_1_En_12_Fig2_HTML.gif


Fig. 12.2
Immune checkpoint modulators. Monoclonal antibodies directed against the immune checkpoint inhibitors CTLA4 and PD1/PD-L1 are used to prevent downregulation of T cell activity and show high potential in GBM. OX40 and 4-1BB are agonistic molecules that, when bound by an antibody, stimulate T cell activity. Both mechanisms lead to a broad upregulation of immune cell activity. APC, antigen-presenting cell

Using anti-CTLA4 antibodies, our laboratory was able to show that systemic CTLA-4 blockade leads to long-term survival in 80% of treated mice with established gliomas without eliciting experimental allergic encephalomyelitis. Furthermore, treatment resulted in the recovery of normal CD4+ T cell counts and proliferative capacity and also suppressed increases in CD4+ CD25+ Foxp3+ GITR+ regulatory T cell fractions [68].

The first clinical trials with anti-CTLA4 and anti-PD-1 antibodies have recently begun for the treatment of newly diagnosed and recurrent GBM and are being tested alone or in combination with other checkpoint modulators, small molecules, and mAbs (Table 12.1). In one study comparable to the recent approval of ipilimumab–nivolumab combination for melanoma, anti-CTLA4 and anti-PD-1 are being tested separately or in combination in a three-armed study in patients with newly diagnosed GBM (Table 12.1).

However, even though trials using checkpoint inhibitors and agonists or combinations thereof have shown unprecedented potential for treating various cancer types, only a certain percentage of patients respond and toxicities are significant [3, 69]. The reasons for this are still unclear but are likely to also occur in GBM, emphasizing the need for in-depth diagnosis and hinting at the future of personalized medicine where certain checkpoint modulators or combinations thereof are prescribed based on patient-specific cancer and genetic traits.



Vaccinations for Tumor Control


The goal of vaccination is to sensitize the immune system against a target antigen and thereby elicit a potent and specific immune response that includes a memory response to the target. While vaccination has been used to successfully prevent and eradicate numerous diseases such as polio, tetanus, and typhoid, anti-tumor vaccinations have not shown the same efficacy and a lot of research is currently ongoing in this field.


Peptides


The major determinant for peptide vaccine-mediated immunogenicity is antigen choice. TAAs, given their expression on normal cells, usually elicit a subdued immune response due to central tolerance. On the other hand, TSAs, given their exclusive presentation on tumor cells, generally elicit a robust immune response similar to the immune response seen against antigens of infectious diseases.

The advantage of TAAs is their high frequency of expression in gliomas, making it possible to give most patients off-the-shelf synthetic tumor antigen peptides. Furthermore, by giving patients a cocktail of peptides, a broader immune response targeting multiple tumor subsets can be elicited. In contrast, TSAs are unique to the tumor and thereby peptides from these antigens may result in a highly tumor-focused immune response.

The mutated protein EGFRvIII, as discussed previously, represents an ideal target for anti-tumor immunotherapy. Our laboratory constructed a 13-amino-acid peptide spanning the vIII mutation and conjugated it to keyhole limpet hemocyanin (KLH). A phase II clinical trial showed that patients with EGFRvIII-positive newly diagnosed GBM, when vaccinated with rindopepimut, the EGFRvIII peptide, had a median survival of 26 months compared with the control historical cohort, which had a median survival of 15 months [70]. These positive results led to the start of a currently ongoing phase III clinical trial with the EGFRvIII peptide vaccine (Table 12.2).


Table 12.2
Recent clinical trials employing vaccination immunotherapy for high-grade gliomas




































































































































NIH clinical trial

Type

Phase

Eligibility criteria

Primary outcome

Dosing regimen

Treatment groups

NCT02510950

Peptide (personalized)

0

Newly diagnosed glioblastoma

Safety and tolerability, feasibility of creating vaccine

Neoantigen-specific long peptide vaccine + poly-ICLC given on cycle 1 day 1 of maintenance TMZ, then on days 3, 5, 8, 15, 22, followed by maintenance on day 22 of subsequent cycles

Single-arm

NCT02454634

Peptide

1

IDH1R132H mutated grade III–IV gliomas

Safety and tolerability (RLT), immunogenicity of IDH1 peptide

20-mer peptide with IDH1(R132H) mutation given 8 times every 2 or 4 weeks

Single-arm

NCT02287428

Peptide

1

MGMT unmethylated, newly diagnosed glioblastoma

# of adverse events, # of patients with >10 actionable peptides

Injections with personalized peptide pool (NeoVax) with 5 priming and 2 boost doses over 7 months

Single-arm

NCT02149225

Peptide (personalized)

1

Newly diagnosed glioblastoma patients

Safety study (# of AEs and SEAs), frequency of CD8 T cell specific for peptides

5–10 peptides (individually assembled, APVAC1) plus Poly-ICLC and GM-CSF given 11 times over 22 weeks. GM-CSF given along first 6 vaccinations. 2 peptides synthesized de novo (APVAC2) and given 6 months after enrollment 8 times within 10 weeks

Single-arm

NCT02049489

DC loaded with peptide antigens

1

Recurrent GBM

Safety study

At least 4 doses of DCs loaded with CD133 peptides given followed by additional vaccines for maintenance

Single-arm

NCT02010606

DC loaded with lysate

1

Newly diagnosed or recurrent GBM

Safety, adverse events, treatment-related toxicities

Autologous DCs loaded with lysate from allogeneic GBM stem-like cell line given once every week for 4 weeks followed by once every 8 weeks

Cohort A: newly diagnosed GBM

Cohort B: recurrent GBM

NCT01491893

Virus

1

Recurrent supratentorial GBM

Maximum tolerated dose or optimal dose

Genetically recombinant, non-pathogenic poliovirus:rhinovirus chimera with tumor-specific conditional replication phenotype (PVSRIPO) given directly into tumor during biopsy

Single-arm

NCT01967758

Virus

1

Treated and recurrent WHO grade III/IV astrocytomas

Maximum tolerated dose

Live attenuated strain of L. monocytogenes expressing EGFRvII and NY = ESO-1 antigens (ADU-623) given on day 0, 21, 42, 63 (Arm I: 3E7 cfu, Arm II: 3E8 cfu, Arm III: 3E9 cfu)

Arm I: low dose

Arm II: medium dose

Arm III: high dose

NCT02649582

DC loaded with RNA

1/2

Newly diagnosed, histologically verified GBM

Overall survival

WT1 mRNA loaded DC vaccine given weekly for 3 weeks followed by maintenance vaccine on day 21 of every TMZ cycle

Single-arm

NCT01567202

DC loaded with lysate

2

Histologically confirmed GBM

Overall survival

8-10E6 DCs loaded with autogeneic glioma stem-like cell-associated antigens given once a week for 6 weeks

Triple-blind

Arm I: DCs

Arm II: placebo

NCT02366728

DC loaded with RNA

2

Newly diagnosed GBM

Overall survival

CMV pp65-loaded DC vaccines #1–3 given every two weeks followed by vaccine #4 (only Arms I–II). Td given to all during vaccine #1. Arm III patients receive basiliximab 1 week before vaccine #1–2. Before vaccine #4, Arm I receives unloaded DCs, Arm II–III Td dose

Arm I: unloaded DCs + loaded DCs

Arm II: Td + loaded DCs

Arm III: Td + loaded DCs + basiliximab

NCT02455557

Peptide

2

Survivin-positive GBM

Progression-free survival

Survivin-mimic peptide vaccine (SurVaxM) 1–2 weeks after chemoradiation followed by doses every 2 weeks (total of 4 doses) followed by doses every 12 weeks

Single-arm

NCT01480479

Peptide

3

Newly diagnosed EGFRvIII-positive GBM

Overall survival

EGFRvIII peptide rindopepimut or placebo given ID two times in month 1 followed by monthly injections

Arm I: rindopepimut

Arm II: placebo

NCT00045968

DC loaded with lysate

3

Newly diagnosed GBM

PFS

Autologous dendritic cells pulsed with tumor lysate antigen, called DCVax-L, or autologous PBMCs (placebo) given ID twice daily on days 0, 10, 20, and at weeks 8, 16, 32, 48, 72, 96, and 120

Arm I: DCVax-L

Arm II: placebo


AE adverse event; cfu colony-forming units; CMV cytomegalovirus; DC dendritic cell; GBM glioblastoma; ID intradermal; lysate tumor cell lysate; PBMC peripheral blood mononuclear cells; RLT regimen limiting toxicity; SAE serious adverse event; Td tetanus toxoid; TMZ temozolomide; PFS progression-free survival

However, given the heterogeneous nature of malignant brain tumor and peptide HLA restrictions, the drawback of single peptide vaccinations is that they may only be effective in a percentage of patients, and in the case of tumor-specific peptides only in the subset of patients expressing the mutated peptide. Trials are ongoing to determine whether combinations of multiple peptides will result in clinically effective peptide vaccination strategies (Table 12.2). Furthermore, increased research on neoantigens, antigens that spontaneously arise in individuals during the course of tumor progression, may lead to personalized solutions in which a patient’s tumor is sequenced after resection and peptide vaccinations are constructed based on the mutanome. Even though major challenges remain, such as locating immunogenic mutations and quickly constructing immunogenic peptides, clinical trials employing a personalized peptide pool approach have commenced (Table 12.2).


Whole Tumor Lysate


Whole tumor lysate can be used as a source of antigen and has the advantage of providing a tumor-specific repertoire of all potentially immunogenic epitopes. The rich repertoire of tumor-associated antigens contains epitopes for both CD8+ and CD4+ T cells, which is important as the parallel presentation of MHC Class I and II antigens could result in a stronger anti-tumor response and boost CD8+ T cell memory [71]. The use of tumor lysate and its encompassing antigen repertoire could also eliminate the time-consuming task of discovering strongly immunogenic antigens.

Tumor lysates can either be obtained from autologous tumor cells, which are taken from the patient, or from an allogenic cell line. Autologous tumor cells are only useful in patient-specific anti-tumor immunotherapies while allogenic tumor cells can be stored at cell banks and vaccines can be created en masse at GMP facilities [72]. Given alone, tumor lysates are administered with a strong adjuvant hapten to provoke a strong inflammatory response and increase their immunogenicity. In a murine glioma model, a CpG-tumor lysate vaccine given subcutaneously had a cure rate of up to 55% and showed significantly longer survival times than tumor lysate or CpG alone. Given their potential to be immunosuppressive, an alternative approach, discussed below, is to create dendritic cell vaccinations by pulsing dendritic cells with tumor lysate [73].


Dendritic Cells


Dendritic cells (DCs), with their powerful antigen-presenting function and unique ability to activate naïve T cells, form a crucial link between the innate and adaptive immune system. As sentinel members of the innate immune system, DCs scavenge for foreign antigens (PAMPs) and in response release cytokines. As members of the adaptive immune response, DCs take up pathogenic antigens, process them internally, and present them on their cell surface, thereby activating naïve, effector, and memory T cells and B cells, as well as maintaining tolerance against self-antigens [74]. In fact, DCs are described as the most potent endogenous activators of de novo T cell and B cell responses [75].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 17, 2017 | Posted by in NEUROLOGY | Comments Off on Immunotherapy for High-Grade Gliomas

Full access? Get Clinical Tree

Get Clinical Tree app for offline access