Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 May 20.
Published in final edited form as: Am Soc Clin Oncol Educ Book. 2014:42–50. doi: 10.14694/EdBook_AM.2014.34.42

The Value of EGFRvIII as the Target for Glioma Vaccines

Pedro R Lowenstein 1, Maria G Castro 1
PMCID: PMC4438702  NIHMSID: NIHMS662804  PMID: 24857059

OVERVIEW

Malignant brain tumors continue to be rapidly progressive and resistant to most treatments. Even with state-of-the-art standard of care (surgery, chemotherapy, and radiotherapy) long-term survival in the last 80 years improved from 6 to 15 months. Improved imaging has also likely contributed to prolonged survival. Immunotherapy for cancer dates back to publications from 1742. The central idea is that the immune system can detect and eliminate foreign antigens, either from infectious agents or tumors, and thus could be therapeutic in brain tumors. Recent introduction of immune modulators of cytotoxic T-lymphocyte antigen (CTLA)-4 and programed cell death 1/programmed cell death 1 ligand (PD-1/PDL1) add much excitement to this field. For brain tumors, there are several ongoing phase I and III trials to determine whether any of the current immunotherapy approaches can demonstrate activity in randomized, controlled double-blinded trials—with ongoing and historical trials presented in tables within the manuscript. Immunotherapy has explored the use of various types of antigens (obtained either from homogenates of patients’ tumors or synthetically produced), and various immunization procedures and adjuvants. Glioma antigens have also been isolated from the patients’ own tumor, then produced in vitro (for example the glioma antigen EGFRvIII), and used to immunize patients directly, or with carriers such as dendritic cells with or without additional adjuvants. Several of these practical approaches are currently in phase III trials. Remaining challenges are how to increase the percentage of complete responses and response duration, and the enigmatic absence of an almost total lack of adverse brain inflammation following immunization of brain tumor patients, as has been observed following immunization against brain antigens in other diseases, such as Alzheimer’s Disease.


Recent improvements in the treatment of high-grade glioma tumors (glioblastoma multiforme, WHO grade 4) have been mostly credited to improvements in standard therapies. These improvements include surgery with electro-physiological monitoring that allows resection of the tumor with reduced negative neurological outcomes, optimization of radiotherapy, and the timing of delivery of temozolomide, which has allowed an increase in patient survival, although that still depends on the particular institution providing care. In the highest complexity centers with therapy optimization a median survival of 16 months is now considered standard, with patients occasionally surviving longer than 2 years. Nevertheless, even with optimized use of temozolomide, gross total resection assessed by MRI, and radiotherapy, routine long-term survival of several years remains elusive in the majority of cases.1

There are several new therapies waiting in the wings. Gene therapy25 has been moving new therapeutic approaches into large phase III trials, myriads of new chemotherapeutics targeted to potentially driving mutations in glioblastoma are progressing at breakneck speed, and new drugs that stimulate the immune response such as antibodies CTLA-4 and PD1 are being tested.68 Despite the frantic translational activity no new treatments, with the possible exception of one anti-angiogenic medication (bevacizumab) integrated into standard of care for patients with recurrent glioblastoma, have yet been incorporated as standard adjuncts to the standard of care triad.911

Different immunotherapeutic approaches have been tested preclinically and are currently in evaluation in early phase I to phase III trials. Approaches vary from the use of autologous dendritic cells loaded with tumor peptides removed from the patients’ own tumors, to autologous dendritic cells loaded with synthetized tumor antigenic peptides, to the use of autologous and/or allogeneic T cells engineered or selected to recognize glioma antigens. Dendritic cells are usually given as intradermal or subcutaneous vaccines, where as T cells are either delivered systemically or into the postsurgical resection cavity.

Overall increased survival benefit obtained so far has not been universal.12,13 Even if individual trials routinely report significant extensions of life, none of these novel therapies has yet been incorporated to the standard of care armamentarium. Importantly, although there is a potential to stimulate brain autoimmune responses as normal brain antigens are likely to be present among the peptides used to load dendritic cells, no such autoimmune responses or brain inflammation has been detected in most trials. An absence of autoimmune side effects has usually been taken to indicate the safety of the different kinds of dendritic cell vaccination approaches. On the other hand, there is a possibility that the almost complete lack of side effects could actually be a reflection of inefficient immunization in patients suffering from high-grade glioma, a population of patients already known to display pre-existing systemic immune-suppression. In comparison, when patients with Alzheimer’s were immunized systemically against beta-amyloid peptides, 10% to 15% developed clinically significant brain inflammation, which led to a hold on ongoing trials. As systemic immunization against brain antigens ought to lead to significant brain autoimmunity in at least a percentage of patients, the absence of such responses from glioma immunotherapy trials requires further examination. Of interest, there is a single report of brain inflammation after immunization with dendritic cell vaccinations.14 In that report of two subjects (pet dogs with high-grade glioma and treated experimentally at the University of Minnesota Veterinary School), the vaccines’ potency was attempted to be increased through concomitant delivery of interferon-gamma. The life-threatening brain inflammation may indicate that dendritic cell vaccines require additional boosts with other immune-stimulators. Similar brain inflammation has been reported in some patients with melanoma metastasis to the brain and treated with inhibitors of CTLA4-Ig.1517 Taken together these results indicate that brain inflammation that followed may be a side effect that could indicate potency of immune responses in attacking antigenic cells in the brain. Additionally, we have recently shown that Ad-TK and Ad-Flt3L gene therapy18 and dendritic cell vaccination potentiate each other.19 In a rat glioma model, when dendritic cell vaccination was administered at 10 to 14 days post-tumor implantation there were no effects on glioma growth, whereas administration of Ad-TK and Ad-Flt3L gene therapy provided a 50% rate of long-term survival. When administered jointly, animal survival was close to 90%.19 We interpret these data as indicating that dendritic cell vaccination may need immunologic adjuvants to exert its full effects. Which mechanisms normally inhibit the full efficiency of dendritic cell vaccination and which can be overcome by Ad-TK and Ad-Flt3L gene therapy, or delivery of interferon-gamma, deserves further study.

A variety of antigens specifically enriched in brain tumors have been proposed to serve as targets for immunotherapies (Tables 1 and 2). In addition, 25% to 30% of high-grade glioma tumors express a truncated variant of EGFR, known as EGFRvIII, which promotes ligand independent signaling. In-frame deletion of exons 2 through 7 from EGFR results in constitutively active kinase activity, and the presence of unique aminoacid sequences at novel mutation induced junctions. The extracelllar portion of EGFRvIII would make the tumor antigen more accessible to activated cytotoxic T cells. If EGFRvIII is indeed a driver of glioma tumors, directly attacking EGFRvIII and destroying glioma cells expressing EGFRvIII ought to provide a therapeutic benefit, as it would eliminate a powerful oncogenic stimulus. Paracrine interactions may also help multiply the effect of the mutation, or of the efforts to eliminate such cells from the tumors. Sampson et al have used vaccination against a 14-mer peptide, LEU-GLU-GLU-LYS-LYS-GLY-ASN-TYR-VAL-VAL-THR-ASP-HIS-CYS, derived from the mutated EGFR as antigen linked to KLH. Sampson et al have pioneered a number of early phase I and II trials. In earlier trials authors reported improved 6-month progression-free survival (PFS), and improved overall survival (OS); the median OS was 15 months in the contemporaneous control cohort compared with 26 months in the immunized cohort. The positive results have been used to support the progress to a randomized controlled double-blind phase III multicenter trial currently in progress.

TABLE 1.

Vaccine Trials for Glioblastoma Multiforme

Study Phase Patients (n) Number of Injected DCs DC Loading/Treatment Trial Result(s) Ref
Phuphanich et al (2013) I 17 1 × 107 Peptide epitopes: HER2, TRP-2, gp100, MAGE-1, IL13alpha2 and AIM-2. HLA-A1 and/or HLA-A2 PFS: 16.9 months Median survival: 38.4 months [32]
Wheeler et al (2008) II 32 1 × 107 to 4 × 107 Tumor cell lysate TTP: 208 vs. 167 days for controls [33]
Yu et al (2001) I 9 1 × 106 MHC class I peptides Median survival: 455 vs. 257 days for controls [34]
Iwami et al (2012) I 8 1 × 107 IL13Rα2 SD: 1 Regression: 1 [35]
Prins et al (2011) I 23 1 × 106 to 5 × 106 to 10 × 106 Tumor cell lysate Median survival: 31.4 months [36]
Liau et al (2005) I 12 1 × 106 to 5 × 105 to 1 × 107 DCs loaded with acid-eluted peptides 8 patients re-operated: T-cell infiltration inversely correlated with TGF-β2 and positively correlated with OS [37]
Okada et al (2011) I/II 13 1 or 3 × 107 Peptide epitopes EphA2, IL13alpha2, YKL-40 and gp100 SD: 6, CR: 1, PR: 2 Median survival for recurrent GBM: 12 months [38]
Sampson et al (2010) I 12 1 × 108 EGFRvIII-specific peptide conjugated to KLH [20]
Sampson et al (2009) I 12 1 × 108 EGFRvIII-specific peptide conjugated to KLH TTP: 10.2 months Median survival: 18.7 months [39]
De Vleeschouwer et al (2008) I/II 56 0.7 × 106 to 1 × 106 Tumor cell lysate 12-month OS: 37.4%, 24-month OS: 14.8%, 36-month OS: 11.1% [40]
Walker et al (2008) I 13 1 × 106 Tumor cell lysate 1-year survival: 38% [41]
Yamanaka et al (2005)* I/II 24 1 × 107 Tumor cell lysate PR: 1, MR: 3, SD: 10 [42]
Yamanaka et al (2003) I/II 10 10 × 106 to 32 × 106 Tumor cell lysate MR: 2, NC: 4 [43]
Kikuchi et al (2001) I 8 2.4 × 106 to 8.7 × 106 Tumor cell lysate SD: 6 [44]
Steiner et al (2004) Pilot 27 1 × 107 Active immunotherapy GBM cells infected with the Newcastle disease virus OS: 100 weeks [45]
Dillman et al (2009) I/II 36 1.75 ± 0.82 × 109 Adoptive immunotherapy Intratumoral LAKs in GBM after standard therapy Median survival: 20.5 months [46]
Dillman et al (2004) I/II 40 2.0 ± 1.0 × 109 or 2.7 ± 0.9 × 109 or 1.8 ± 0.9 × 109 Intralesional LAK cell therapy Median survival: 17.5 months [47]
Sankhla et al (1996) I 10 2.0 to 10.0 × 106 LAK and IL-2 PR: 2 [48]
Hayes et al (1995) I 15 1.2 MIU LAK and IL-2 Median survival: 53 weeks [49]
Boiardi et al (1994) I 9 1.5 × 108 Recombinant IL-2 and LAK CR: 1, PR: 2, SD: 4 [50]
Jeffes et al (1993) I 19 2.2 × 109 to 58 × 109 LAK Median survival: 37 weeks [51]
Blancher et al (1993) I 13 18 × 106 or 24 × 106 or 54 × 106 rIL-2 with and without LAK Tumor progression noted after 4–12 weeks [52]
Lillehei et al (1991) I 20 1.9–27.5 × 109 LAK and ASL with rIL-2 Median survival: 63 weeks [53]
Barba et al (1989) I 10 0.9–21.0 × 109 IL-2 or LAK PR: 1 [54]
Jacobs et al (1986) I 9 1 × 108 IL-2 or LAK No PR or SD [55]
Tsuboi et al (2003) I 10 3–247 × 107 ATTL cocultured in medium containing interleukin-1, –2, –4, and –6 CR: 1; PR: 4; Median survival: 5 months; Overall response rate: 50% [56]
Wood et al (2000) I 9 2 × 107 Lymphocyte stimulation with anti-CD3 and IL-2 PR: 3; Survival >4 years: 2 [57]
Holladay et al (1996) I 15 8.8 × 106 or 3 × 106 Tumor reactive T cells and IL-2 No PR or SD; Disease-free survival >8 months: 7 [58]
Plautz et al (2000) I 9 0.6–5.5 × 1010 Adoptive transfer ex vivo activated T lymphocytes PR: 3; (1 GBM and 2 grade III survival >4 years) [59]
Plautz et al (1998) I 10 17 to 56 × 106 Adoptive transfer ex vivo activated T lymphocytes SD: 1; 4 patients alive after 1 year [60]
Sloan et al (2000) I 19 2 × 107 Adoptive transfer ex vivo activated T lymphocytes CR: 1; PR: 7; Median survival: 12 months [61]
Quattrocchi et al (1999) I 6 1 × 109 TILs + 1 × 105 rIL-2 or 1 × 109 TILs + 4 × 105 rIL-2 Autologus intralesional TILs and rIL-2 in recurrent MGs CR: 1, PR: 2 [62]
Tsurushima et al (1999) I 4 0.07–11 × 107 Cytotoxic T lymphocytes injected via Ommaya tubes CR: 1; SD: 1 [63]
Kruse et al (1997) I 5 1.1–51.5 × 108 Alloreactive cytotoxic T lymphocytes and IL-2 Transient toxicities: Survival (AO) >30 months; Survival (AA) >28 months [64]
Kitahara et al (1987) I 5 5.0–10.4 × 108 Adoptive immunotherapy IL-2 dependent cytotoxic T lymphocytes PR: 2; 1 patient alive >104 weeks [65]
Crane et al (2012) I 12 N/A Heat shock protein peptide bound to 96 kD chaperone protein (HSP-96) Median survival: 47 week in immunologic responders [66]
*

ID or ID and IT

Abbreviations: CR, complete response; DC, dendritic cell; EGFR, EGF receptor; GBM, glioblastoma multiforme; ID, intradermal; IT, intratumoral; LAK, lymphokine activated killers; MR, minor response; NC, no change; OS, overall survival; PFS, progression-free survival; PR, partial response; SD, stable disease; TTP, time to progression

TABLE 2.

Ongoing Clinical Trials*

Phase Trial ID Estimated Completion Date/Enrollment Sponsor Protocol
Phase I/II NCT00293423 12/2013; n = 50 University of California, San Francisco Trial of Heat Shock Protein Peptide Complex-96 (HSPPC-96) vaccine for patients with recurrent high-grade glioma
Phase I NCT01403285 03/2014; n = 25 Immatics Biotechnologies GmbH Trial of peptide-based glioma vaccine IMA950 in patients with glioblastoma IMA950: multi-peptide vaccine containing 11 tumor-associated peptides (TUMAPs) found in a majority of glioblastomas
Phase I NCT01250470 09/2014; n = 9 Roswell Park Cancer Institute Study of safety, tolerability and immunological effects of SVN53-67/M57-KLH in patients with Survivin-positive malignant gliomas
Phase I NCT00626015 11/2013; n = 20 John Sampson, Duke University Medical Center Chemotherapy, radiation therapy, and vaccine (PEP-3-KLH) therapy with basiliximab in treating patients with glioblastoma multiforme that has been removed by surgery
Phase I/II NCT01920191 08/2014; n = 16 University Hospital, Geneva Study of intradermal IMA950 peptide-based vaccine adjuvanted with intramuscular Poly-ICLC in combination with temozolomide in newly diagnosed HLA-A2 glioblastoma patients
Phase II NCT01814813 04/2016; n = 222 Alliance for Clinical Trials in Oncology Randomized trial comparing the efficacy of Heat Shock Protein-Peptide Complex-96 (HSPPC-96) vaccine given with bevacizumab versus bevacizumab alone in the treatment of surgically resectable recurrent glioblastoma
Arm 1, HSPPC-96, concomitant bevacizumab
Arm 2, HSPPC-96 with bevacizumab at progression
Active comparator: Arm 3, bevacizumab
Phase II NCT00905060 01/2014; n = 555 University of California, San Francisco Multi-center, single arm investigation of HSPPC-96 vaccine with temozolomide in patients with newly diagnosed glioblastoma
Phase II NCT00643097 06/2016; n = 48 John Sampson, Duke University Medical Center A complementary trial of an immunotherapy vaccine against tumor-specific EGFRvIII
Arm I: Patients receive PEP-3-KLH conjugate vaccine and sargramostim (GM-CSF) intradermally on days 1, 15, and 29 and then monthly in the absence of disease progression or unacceptable toxicity
Arm II: Patients receive placebo vaccine intradermally on days 1, 15, and 29. Patients then receive PEP-3-KLH conjugate vaccine and GM-CSF monthly in the absence of disease progression or unacceptable toxicity
Phase II NCT01498328 06/2015; n = 168 Celldex Therapeutics Study of rindopepimut/GM-CSF in patients with relapsed EGFRvIII-positive glioblastoma (ReACT)
Group 1a: bevacizumab naive with bevacizumab + rindopepimut
Group 1b: bevacizumab naive with bevacizumab + KLH control
Group 2 and 2C: refractory to bevacizumab
Phase III NCT01498328 11/2016; n = 440 Celldex Therapeutics International, randomized, double-blind, controlled study of rindopepimut/GM-CSF with adjuvant temozolomide in newly diagnosed, surgically resected, EGFRvIII-positive glioblastoma (ACT IV)
Comparator: KLH plus temozolomide
2 id injections 2 wk apart, followed by monthly injections until tumor progression or intolerance
Phase II NCT00458601 12/2013; n = 82 Celldex Therapeutics CDX-110 in patients with glioblastoma multiforme (ACT III)
Phase I NCT01058850 9/2013; n = 3 Paul Graham Fisher, Stanford University Rindopepimut after conventional radiation in children with diffuse intrinsic pontine gliomas
Phase II NCT00323115 4/2008; n = 11 Dartmouth-Hitchcock Medical Center Phase II feasibility study of DC vaccination for newly diagnosed GBM
Phase I NCT00612001 1/2011; n = 8 UCLA; Jonsson Compr. Cancer Center Vaccine therapy for patients with malignant glioma
Phase I NCT00639639 6/2016; n = 16 J. Sampson, Duke Univ. Medical Center Vaccine therapy in treating patients with newly diagnosed glioblastoma multiforme
Phase II NCT01213407 3/2011; n = 56 Trimed Biotech GmbH DC cancer vaccine for high-grade glioma (GBM-Vax)
Phase I/II NCT00846456 12/2013; n = 20 Oslo University Hospital Safe study of dendritic cell (DC) based therapy targeting tumor stem cells in glioblastoma
Phase II NCT01006044 10/2013; n = 26 Clinica Universidad de Navarra, Univ. de Navarra Efficacy & safety of autologous dendritic cell vaccination in glioblastoma multiforme after complete surgical resection
Phase I NCT00068510 9/2013; n = 28 UCLA; Jonsson Compr. Cancer Center Vaccine therapy in treating patients with malignant glioma
Phase I/II NCT01291420 2/2014; n = 10 University Hospital, Antwerp DC vaccination for patients with solid tumors
Phase III NCT00045968 9/2014; n = 300 Northwest Biotherapeutics Study of a drug [DCVax-L] to treat newly diagnosed GBM brain cancer
Phase II NCT01280552 10/2014; n = 200 ImmunoCellular Therapeutics, Ltd. A study of ICT-107 immunotherapy in glioblastoma multiforme (GBM)
Phase I NCT00890032 9/2015; n = 50 J. Sampson, Duke Univ. Medical Center Vaccine therapy in treating patients undergoing surgery for recurrent glioblastoma multiforme
Phase II NCT00905060 1/2014; n = 55 UCSF HSPPC-96 vaccine with temozolomide in patients with newly diagnosed GBM (HeatShock)
Phase I/II NCT00293423 12/2013; n = 50 UCSF GP96 heat shock protein-peptide complex caccine in treating patients with recurrent or progressive glioma

Abbreviations: DC, dendritic cells; GBM, glioblastoma multiforme

*

Information from www.clinicaltrials.gov

In previous trials Sampson et al demonstrated increase in immunologic function specifically related to the recognition of the brain tumor antigen.20 In patients with EGFRvIII-positive tumors at initial resection, immune responses could be stimulated in about 50% of patients. In trials of 17 immunized patients, six developed EGFRvIII antibodies and three developed evidence of a T cell response against EGFRvIII. Of potential clinical relevance, the development of either a humoral or a cellular response to mutated EGFRvIII appeared to provide increased OS, though the small numbers challenged in depth statistical analysis.2123

One of the most interesting pathophysiological studies performed by the authors was the study of EGFRvIII expression at tumor recurrence.20 In 11 patients who could have tumor samples evaluated both before and after vaccination, 82% lacked EGFRvIII immunoreactivity at recurrence. These studies are a true tour de force as the difficulties involved are truly challenging. The authors favor the interpretation that this indicates tumor escape, although at this stage it is impossible to differentiate tumor escape from loss of immunoreactivity because of the blocking of endogenous EGFRvIII immunoreactivity by the induced anti-EGFRvIII antibodies, or downregulation of EGFRvIII expression. Although the clinical progression of the disease would favor an interpretation of effective tumor control by the immunization followed by tumor mutation and escape, the absence of more detailed molecular studies and the low numbers involved preclude drawing definitive conclusions. In the future, analysis of comprehensive results from large-scale phase III, randomized, double-blind trials of patients with brain tumors immunoreactive for EGFRvIII will allow us to address the potential mechanisms of action of the EGFRvIII vaccine.

Even given the shortcomings of the data at this time, a discussion of potential mechanisms of action is warranted. An accurate understanding of the mechanisms of tumor response (and apparent escape from immunologic control) will be crucial in devising the future pathways of this therapeutic approach. Tumor heterogeneity,24,25 already classically detected in studies of tumor pathology—the classical name of the tumor is glioblastoma multiforme—has now been detected at the molecular level. Thus, even though 30% of patients may express mutated EGFRvIII, it is not yet known whether the mutation is present in all tumor cells, or only in a subset of cancer cells. If only present on a subpopulation of glioma cells, challenges in tumor sampling during biopsy could possibly explain changes in detected EGFRvIII expression; that is, even without changes in the population of tumor cells, unwitting biases in biopsy sampling may erroneously indicate changes in the expression profile of gliomas. Equally, selection could be at the level of expression, an outcome which will also indicate selection bias in the case of expression going below the limit of antigenic detection. Recently, it has also been described that amplified EGFR resides within abnormal mini-chromosomes that also participate in the selection of expressing cells when these are grown under selection.26

If the observed selection does represent true elimination of the proportion of tumor cells expressing EGFRvIII, this would represent a strong indication that vaccination with multiple antigens may represent the way forward, as this would reduce the capacity of the tumors to escape from immune control. Therefore, detailed pathophysiological studies are necessary to understand both the power and the current challenges of immunization regimes against single antigens. A similar challenge was faced during the initial trials for the treatment of AIDS in patients infected with the rapidly mutating HIV virus. Initial treatment with only a single drug, rapidly selected mutant clones not expressing the drug target or cells with a mutated target now resistant to the drug inhibitor. However, as more anti-HIV drugs came onto the market it was realized that successful treatment necessitates simultaneous administration of multiple inhibitors of HIV’s life cycle. Once three to five inhibitors are used simultaneously, viral replication is rapidly suppressed as the HIV genome is unable to find simultaneous selected mutants to three to five inhibitors. Thus, the treatment of HIV infection with multi-pronged attacks has led to the current increase in patient survival.27

If we assume a similar pathophysiological process of selection and immune escape occurs on treatment of brain gliomas expressing EGFRvIII with specific vaccination paradigms, existing knowledge suggests that the combination of multiple inhibitors (or multiple vaccines directed against different antigens; or combination of immune-stimulators—e.g., antibodies to CTLA or PD-1-) would be predicted to eventually result in more effective results.

In vaccination against EGFRvIII it has been demonstrated that 33% of immunized patients developed humoral responses and 16% developed cellular responses.20 Given that responses appear to involve the humoral and cellular arms of the adaptive immune response, it would be of importance to be able to determine the mechanism of action of each arm, as potentially both could have a positive therapeutic influence of disease outcome. Although in theory immunization aims to eliminate tumor growth by inducing the generation of antitumor cytotoxic T cells, antibodies against individual antigens could also provide therapeutic benefits. For example, antibodies binding to EGRFvIII on glioma cells may serve to kill tumor cells by antibody-dependent cell-mediated cytotoxicity, or by binding to EGFRvIII and inhibiting its signaling capacity by restricting its ability to diffuse across the plasma membrane. Eventual isolation of induced antibodies and a dissection in vitro of their mechanisms of action will be central to the future directions of glioma immunotherapy. Equally, it will be important to isolate antitumor T cells from those patients that mount such responses to study their function, pathogenicity, and capacity to kill glioma tumor cells in vitro. Potentially such T cells could also serve as sources to isolate antitumor T-cell receptor genes that could, at a later stage, be engineered into the bone marrow of patients within the context of the Chimeric Antigen Receptor (CAR) approach to engineering effective antitumor cytotoxic T cells.28,29

Of course, outstanding questions to be resolved by future experiments remain: Why did only 50% of patients respond? Could this be a result of the general immune-suppression of glioma patients, or technical details of immunization procedures? Are the strongest results from this trial the increase in OS in a nonrandomized trial or the induction of immune responses in 50% of patients? It would be important to understand the mechanisms underlying a significant increase in OS even if immune responses were only detected in 50% of patients. Alternatively, the assessment of immune responses could be delayed until a treatment is found that provides a significant OS benefit in a randomized phase III trial, such as the sipuleucel-T trial for castration-resistant prostate cancer.30 How are we to interpret that increased OS in the sipuleucel-T trial was seen in the absence of effects on tumor progression, or even without stimulation of immune responses? In the case of brain tumors, would it be worth repeating a small trial, but attempting to obtain direct evidence of intracranial antitumor immune reactivity, possibly utilizing PET imaging? Which should be the way forward for immunotherapies to cross the bar of generalized clinical implementation?

Sampson et al reported an increased OS of 9 months compared with contemporaneous controls.20 If a comparable difference would be maintained in a larger randomized trial, the use of the EGFRvIII vaccine in suitable patients would be amply justified. The challenge remains how to determine the rational use of EGFRvIII vaccine in the absence of a large randomized, double blind, controlled, phase III trial. Thus, it is important to explore alternatives to study in detail small clinical trials. For example, it would be feasible to perform further small clinical trials that would compare various immunotherapeutic approaches, and analyze these trials using either adaptive trial designs, or Bayesian designs that simultaneously evaluate efficacy and toxicity.

Clinical statistical challenges remain in how best to assess survival benefits seen in small clinical trials. If we could demonstrate beyond reasonable doubt that those increases are indeed clinically significant it would go a long way to reassure all stakeholders that the benefit is not because of inadvertent bias. In other cases (i.e., at the early stages of treatment for childhood leukemias) it was possible to determine whether, even in the absence of long-term remissions, anticancer agents displayed temporary reductions in the proliferation of neoplastic cells. Such small, yet clinically significant effects led to further improvements, which, during years of continued research led to the ultimate reduction in the morbidity and mortality of diseases such as childhood leukemias. It remains the responsibility of clinicians, oncologists, neurosurgeons, neuroradiologists, as well as basic scientists involved in glioma research, to advance our statistical, oncological, and clinical assessment of small clinical trials, to achieve large therapeutic effect sizes, to benefit the patient community as a whole by being able to extract as much clinically relevant information as possible, and to separate true treatment effects from inadvertent experimental bias and artifacts. Unfortunately for the clinical translational study of glioma therapies, MRI studies have evidenced shortcomings. For example, it is sometimes difficult to determine if MRI images reflect true tumor recurrence or inflammation surrounding the original resection cavity, a phenomenon described as pseudoprogression.31 In response to these challenges various groups are now pursuing improved imaging modalities, in some cases accompanied by parallel histologic sampling to improve the accuracy of glioma imaging in the context of translation clinical trials.

In view of evidence in favor of therapeutic activity of anti-EGFRvIII immunotherapy, serious consideration should be given to the combination of this approach with vaccination against other antigens that are also now reporting partial yet significant therapeutic benefits. Just as HIV replication can only be blocked by simultaneously inhibiting three to five central physiological processes needed by the virus to replicate, a similar multipronged approach to the treatment of glioma tumors is predicted to carry stronger potential therapeutic benefits. Indeed, an analysis of current anticancer therapeutics indicates that nonspecific therapeutic agents, such as alkylating agents, antimetabolites, microtubule inhibitors, cytotoxic drugs, and topoisomerase inhibitors, among others continue to be the mainstay of cancer treatment and in many cases particular combinations have been proven to be able to even cure certain cancers.

The remaining clinical challenge is how to develop stable algorithms to determine the level of survival benefit that is indeed clinically significant to reassure all patients, clinicians, and researchers that the benefit is not because of inadvertent noncontrolled experimental biases.

We also need to take into consideration that many nonrandomized immunotherapy trials select for a favorable prognostic group of patients (e.g., only patients with gross total resection, good performance status, etc.) or compare trial data with historical controls. These issues further complicate interpretation of survival time in those studies in comparison with ‘standard-of-care’ data. Treatment-related immuno-suppression (i.e., lymphopenia) is a potential additional hurdle regarding the efficacy of immunotherapy. There are now clear data that profound lymphopenia is common, and that it can be prolonged in patients with glioma who have received radiation or chemoradiation. Nevertheless, the finding that specific immune responses can still be induced in subjects treated with standard of care indicates that the clinical consequences of the relationship between lymphopenia and responses to immunization need to be studied further. Clinical trials of any type ought to evaluate these challenges as part of trial design.

KEY POINTS.

  • Brain tumor immune therapies are now being tested in large, double-blind, randomized clinical trials.

  • The use of glioma-derived antigens, such as EGFRvIII, has provided novel approaches to immunotherapy against brain tumors.

  • The engineering of T cells recognizing specific antigens by using chimeric antigen receptors is the latest approach to brain immunotherapy; this approach is not being tested in clinical trials in human patients yet.

  • The use of modulators of CTLA-4 and PD1/PDL1 is a further immune modulator that may be of benefit for patients with glioma, and needs to be tested in clinical trials.

  • In the future it is likely that the combination of immune therapies acting through complementary mechanisms, i.e., vaccination against whole patients’ tumors, vaccination against individual or multiple specific tumor antigens, the use of anti-glioma engineered T cells, together with immune modulatory gene therapy (i.e., Ad-TK/Ad-Flt3L), and powerful blockers of negative regulators of T cell function such as CTLA4-Ig, and antibodies to PD1/PDL1, are likely to achieve the best possible outcomes for patients.

Footnotes

Disclosures of Potential Conflicts of Interest

The author(s) indicated no potential conflicts of interest.

References

  • 1.Grossman SA, Ye X, Piantadosi S, et al. Survival of patients with newly diagnosed glioblastoma treated with radiation and temozolomide in research studies in the United States. Clin Cancer Res. 2010;16:2443–2449. doi: 10.1158/1078-0432.CCR-09-3106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Simonato M, Bennett J, Boulis NM, et al. Progress in gene therapy for neurological disorders. Nat Rev Neurol. 2013;9:277–291. doi: 10.1038/nrneurol.2013.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Castro MG, Lowenstein PR. Gene therapy continues to make progress: clinical and regulatory perspectives. Curr Gene Ther. 2009;9:327–328. doi: 10.2174/156652309789753374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Candolfi M, Kroeger KM, Muhammad AK, et al. Gene therapy for brain cancer: combination therapies provide enhanced efficacy and safety. Curr Gene Ther. 2009;9:409–421. doi: 10.2174/156652309789753301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Jacobs AH, Winkler A, Castro MG, Lowenstein P. Human gene therapy and imaging in neurological diseases. Eur J Nucl Med Mol Imaging. 2005;32 (Suppl 2):S358–S383. doi: 10.1007/s00259-005-1960-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Blank CU. The perspective of immunotherapy: new molecules and new mechanisms of action in immune modulation. Curr Opin Oncol. 2014;26:204–214. doi: 10.1097/CCO.0000000000000054. [DOI] [PubMed] [Google Scholar]
  • 7.Konstantinou MP, Dutriaux C, Gaudy-Marqueste C, et al. Ipilimumab in melanoma patients with brain metastasis: a retro-spective multicentre evaluation of thirty-eight patients. Acta Derm Venereol. 2014;94:45–49. doi: 10.2340/00015555-1654. [DOI] [PubMed] [Google Scholar]
  • 8.Nieder C. Ipilimumab in patients with melanoma and brain metastases. Lancet Oncol. 2012;13:e277–e278. doi: 10.1016/S1470-2045(12)70303-0. [DOI] [PubMed] [Google Scholar]
  • 9.de Groot JF, Mandel JJ. Update on anti-angiogenic treatment for malignant gliomas. Curr Oncol Rep. 2014;16:380. doi: 10.1007/s11912-014-0380-6. [DOI] [PubMed] [Google Scholar]
  • 10.Flieger M, Ganswindt U, Schwarz SB, et al. Re-irradiation and bevacizumab in recurrent high-grade glioma: an effective treatment option. J Neurooncol. doi: 10.1007/s11060-014-1394-5. Epub 2014 Feb 7. [DOI] [PubMed] [Google Scholar]
  • 11.Soffietti R, Trevisan E, Rudá R. What have we learned from trials on antiangiogenic agents in glioblastoma? Expert Rev Neurother. 2014;14:1–3. doi: 10.1586/14737175.2014.873277. [DOI] [PubMed] [Google Scholar]
  • 12.Omuro A, DeAngelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA. 2013;310:1842–1850. doi: 10.1001/jama.2013.280319. [DOI] [PubMed] [Google Scholar]
  • 13.Weller M, Pfister SM, Wick W, et al. Molecular neuro-oncology in clinical practice: a new horizon. Lancet Oncol. 2013;14:e370–e379. doi: 10.1016/S1470-2045(13)70168-2. [DOI] [PubMed] [Google Scholar]
  • 14.Pluhar GE, Grogan PT, Seiler C, et al. Anti-tumor immune response correlates with neurological symptoms in a dog with spontaneous astrocytoma treated by gene and vaccine therapy. Vaccine. 2010;28:3371–3378. doi: 10.1016/j.vaccine.2010.02.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Assi H, Wilson KS. Immune toxicities and long remission duration after ipilimumab therapy for metastatic melanoma: two illustrative cases. Curr Oncol. 2013;20:e165–e169. doi: 10.3747/co.20.1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kaehler KC, Egberts F, Lorigan P, Hauschild A. Anti-CTLA-4 therapy-related autoimmune hypophysitis in a melanoma patient. Melanoma Res. 2009;19:333–334. doi: 10.1097/CMR.0b013e32832e0bff. [DOI] [PubMed] [Google Scholar]
  • 17.Hodi FS, Oble DA, Drappatz J, et al. CTLA-4 blockade with ipilimumab induces significant clinical benefit in a female with melanoma metastases to the CNS. Nat Clin Pract Oncol. 2008;5:557–561. doi: 10.1038/ncponc1183. [DOI] [PubMed] [Google Scholar]
  • 18.Curtin JF, Liu N, Candolfi M, et al. HMGB1 mediates endogenous TLR2 activation and brain tumor regression. PLoS Med. 2009;6:e10. doi: 10.1371/journal.pmed.1000010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mineharu Y, King GD, Muhammad AK, et al. Engineering the brain tumor microenvironment enhances the efficacy of dendritic cell vaccination: implications for clinical trial design. Clin Cancer Res. 2011;17:4705–4718. doi: 10.1158/1078-0432.CCR-11-0915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sampson JH, Heimberger AB, Archer GE, et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28:4722–4729. doi: 10.1200/JCO.2010.28.6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Heimberger AB, Sampson JH. The PEPvIII-KLH (CDX-110) vaccine in glioblastoma multiforme patients. Expert Opin Biol Ther. 2009;9:1087–1098. doi: 10.1517/14712590903124346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schmittling RJ, Archer GE, Mitchell DA, et al. Detection of humoral response in patients with glioblastoma receiving EGFRvIII-KLH vaccines. J Immunol Methods. 2008;339:74–81. doi: 10.1016/j.jim.2008.08.004. [DOI] [PubMed] [Google Scholar]
  • 23.Sampson JH, Archer GE, Mitchell DA, et al. Tumor-specific immunotherapy targeting the EGFRvIII mutation in patients with malignant glioma. Semin Immunol. 2008;20:267–275. doi: 10.1016/j.smim.2008.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bonavia R, Inda MM, Cavenee WK, Furnari FB. Heterogeneity maintenance in glioblastoma: a social network. Cancer Res. 2011;71:4055–4060. doi: 10.1158/0008-5472.CAN-11-0153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sottoriva A, Spiteri I, Piccirillo SG, et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U S A. 2013;110:4009–4014. doi: 10.1073/pnas.1219747110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nathanson DA, Gini B, Mottahedeh J, et al. Targeted therapy resistance mediated by dynamic regulation of extrachromosomal mutant EGFR DNA. Science. 2014;343:72–76. doi: 10.1126/science.1241328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Domingo E, Sheldon J, Perales C. Viral quasispecies evolution. Microbiol Mol Biol Rev. 2012;76:159–216. doi: 10.1128/MMBR.05023-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sadelain M, Brentjens R, Riviére I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3:388–398. doi: 10.1158/2159-8290.CD-12-0548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Davila ML, Brentjens R, Wang X, et al. How do CARs work?: early insights from recent clinical studies targeting CD19. Oncoimmunology. 2012;1:1577–1583. doi: 10.4161/onci.22524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Geary SM, Lemke CD, Lubaroff DM, Salem AK. Proposed mechanisms of action for prostate cancer vaccines. Nat Rev Urol. 2013;10:149–160. doi: 10.1038/nrurol.2013.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Butowski N, Chang SM. Endpoints for clinical trials and revised assessment in neuro-oncology. Curr Opin Neurol. 2012;25:780–785. doi: 10.1097/WCO.0b013e328359b45e. [DOI] [PubMed] [Google Scholar]
  • 32.Phuphanich S, Wheller CJ, Rudnick JD, et al. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother. 2013;62:125–135. doi: 10.1007/s00262-012-1319-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wheeler CJ, Black KL, Liu G, et al. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Res. 2008;68:5955–5964. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
  • 34.Yu JS, Wheeler CJ, Zelter PM, et al. Vaccination of malignant glioma patients with peptide-pulsed dendritic cells elicits systemic cytotoxicity and intracranial T-cell infiltration. Cancer Res. 2001;61:842–847. [PubMed] [Google Scholar]
  • 35.Iwami K, Shimato S, Ohno M, et al. Peptide-pulsed dendritic cell vaccination targeting interleukin-13 receptor alpha2 chain in recurrent malignant glioma patients with HLA-A*24/A*02 allele. Cytotherapy. 2012;14:733–742. doi: 10.3109/14653249.2012.666633. [DOI] [PubMed] [Google Scholar]
  • 36.Prins RM, Soto H, Konkankit V, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17:1603–1615. doi: 10.1158/1078-0432.CCR-10-2563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Liau LM, Prins RM, Kiertscher SM, et al. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res. 2005;11:5515–5525. doi: 10.1158/1078-0432.CCR-05-0464. [DOI] [PubMed] [Google Scholar]
  • 38.Okada H, Kalinski P, Ueda R, 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 carboxymethyl-cellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29:330–336. doi: 10.1200/JCO.2010.30.7744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sampson JH, Archer GE, Mitchell DA, et al. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol Cancer Ther. 2009;8:2773–2779. doi: 10.1158/1535-7163.MCT-09-0124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.De Vleeschouwer S, Fieuws S, Rutkowski S, et al. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res. 2008;14:3098–3104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
  • 41.Walker DG, Laherty R, Tomlinson FH, et al. Results of a phase I dendritic cell vaccine trial for malignant astrocytoma: potential interaction with adjuvant chemotherapy. J Clin Neurosci. 2008;15:114–121. doi: 10.1016/j.jocn.2007.08.007. [DOI] [PubMed] [Google Scholar]
  • 42.Yamanaka R, Homma J, Yakima N, et al. Clinical evaluation of dendritic cell vaccination for patients with recurrent glioma: results of a clinical phase I/II trial. Clin Cancer Res. 2005;11:4160–4167. doi: 10.1158/1078-0432.CCR-05-0120. [DOI] [PubMed] [Google Scholar]
  • 43.Yamanaka R, Abe T, Yajima N, et al. Vaccination of recurrent glioma patients with tumour lysate-pulsed dendritic cells elicits immune responses: results of a clinical phase I/II trial. Br J Cancer. 2003;89:1172–1179. doi: 10.1038/sj.bjc.6601268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kikuchi T, Akasaki Y, Irie M, et al. Results of a phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunol Immunother. 2001;50:337–344. doi: 10.1007/s002620100205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Steiner HH, Bonsanto MM, Beckhove P, et al. Antitumor vaccination of patients with glioblastoma multiforme: a pilot study to assess feasibility, safety, and clinical benefit. J Clin Oncol. 2004;22:4272–4281. doi: 10.1200/JCO.2004.09.038. [DOI] [PubMed] [Google Scholar]
  • 46.Dillman RO, Duma CM, Ellis RA, et al. Intralesional lymphokine-activated killer cells as adjuvant therapy for primary glioblastoma. J Immunother. 2009;32:914–919. doi: 10.1097/CJI.0b013e3181b2910f. [DOI] [PubMed] [Google Scholar]
  • 47.Dillman RO, Duma CM, Schiltz PM, et al. Intracavitary placement of autologous lymphokine-activated killer (LAK) cells after resection of recurrent glioblastoma. J Immunother. 2004;27:398–404. doi: 10.1097/00002371-200409000-00009. [DOI] [PubMed] [Google Scholar]
  • 48.Sankhla SK, Nadkarni JS, Bhagwati SN. Adoptive immunotherapy using lymphokine-activated killer (LAK) cells and interleukin-2 for recurrent malignant primary brain tumors. J Neurooncol. 1996;27:133–140. doi: 10.1007/BF00177476. [DOI] [PubMed] [Google Scholar]
  • 49.Hayes RL, Koslow M, Hiesiger EM, et al. Improved long term survival after intracavitary interleukin-2 and lymphokine-activated killer cells for adults with recurrent malignant glioma. Cancer. 1995;76:840–852. doi: 10.1002/1097-0142(19950901)76:5<840::aid-cncr2820760519>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 50.Boiardi A, Silvani A, Ruffini PA, et al. Loco-regional immunotherapy with recombinant interleukin-2 and adherent lymphokine-activated killer cells (A-LAK) in recurrent glioblastoma patients. Cancer Immunol Immunother. 1994;39:193–197. doi: 10.1007/BF01533386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jeffes EW, 3rd, Beamer YB, Jacques S, et al. Therapy of recurrent high grade gliomas with surgery, and autologous mitogen activated IL-2 stimulated killer (MAK) lymphocytes: I. enhancement of MAK lytic activity and cytokine production by PHA and clinical use of PHA. J Neurooncol. 1993;15:141–155. doi: 10.1007/BF01053935. [DOI] [PubMed] [Google Scholar]
  • 52.Blancher A, Roubinet F, Grancher AS, et al. Local immunotherapy of recurrent glioblastoma multiforme by intracerebral perfusion of interleukin-2 and LAK cells. Eur Cytokine Netw. 1993;4:331–341. [PubMed] [Google Scholar]
  • 53.Lillehei KO, Mitchell DH, Johnson SD, et al. Long-term follow-up of patients with recurrent malignant gliomas treated with adjuvant adoptive immunotherapy. Neurosurgery. 1991;28:16–23. doi: 10.1097/00006123-199101000-00003. [DOI] [PubMed] [Google Scholar]
  • 54.Barba D, Saris SC, Holder C, et al. Intratumoral LAK cell and interleukin-2 therapy of human gliomas. J Neurosurg. 1989;70:175–182. doi: 10.3171/jns.1989.70.2.0175. [DOI] [PubMed] [Google Scholar]
  • 55.Jacobs SK, Wilson DJ, Kornblith PL, Grimm EA. Interleukin-2 or autologous lymphokine-activated killer cell treatment of malignant glioma: Phase I trial. Cancer Res. 1986;46:2101–2104. [PubMed] [Google Scholar]
  • 56.Tsuboi K, Saijo K, Ishikawa E, et al. Effects of local injection of ex vivo expanded autologous tumor-specific T lymphocytes in cases with recurrent malignant gliomas. Clin Cancer Res. 2003;9:3294–3302. [PubMed] [Google Scholar]
  • 57.Wood GW, Holladay FP, Turner T, et al. A pilot study of autologous cancer cell vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in patients with recurrent grade III/IV astrocytoma. J Neurooncol. 2000;48:113–120. doi: 10.1023/a:1006456421177. [DOI] [PubMed] [Google Scholar]
  • 58.Holladay FP, Heitz-Turner T, Bayer WL, Wood GW. Autologous tumor cell vaccination combined with adoptive cellular immunotherapy in patients with grade III/IV astrocytoma. J Neurooncol. 1996;27:179–189. doi: 10.1007/BF00177482. [DOI] [PubMed] [Google Scholar]
  • 59.Plautz GE, Miller DW, Barnett GH, et al. T cell adoptive immunotherapy of newly diagnosed gliomas. Clin Cancer Res. 2000;6:2209–2218. [PubMed] [Google Scholar]
  • 60.Plautz GE, Barnett GH, Miller DW, et al. Systemic T cell adoptive immunotherapy of malignant gliomas. J Neurosurg. 1998;89:42–51. doi: 10.3171/jns.1998.89.1.0042. [DOI] [PubMed] [Google Scholar]
  • 61.Sloan AE, Dansey R, Zamorano L, et al. Adoptive immunotherapy in patients with recurrent malignant glioma: preliminary results of using autologous whole-tumor vaccine plus granulocyte-macrophage colony-stimulating factor and adoptive transfer of anti-CD3-activated lymphocytes. Neurosurg Focus. 2000;9:e9. doi: 10.3171/foc.2000.9.6.10. [DOI] [PubMed] [Google Scholar]
  • 62.Quattrocchi KB, Miller CH, Cush S, et al. Pilot study of local autologous tumor infiltrating lymphocytes for the treatment of recurrent malignant gliomas. J Neurooncol. 1999;45:141–157. doi: 10.1023/a:1006293606710. [DOI] [PubMed] [Google Scholar]
  • 63.Tsurushima H, Liu SQ, Tuboi K, et al. Reduction of end-stage malignant glioma by injection with autologous cytotoxic T lymphocytes. Jpn J Cancer Res. 1999;90:536–545. doi: 10.1111/j.1349-7006.1999.tb00781.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kruse CA, Cepeda L, Owens B, et al. Treatment of recurrent glioma with intracavitary alloreactive cytotoxic T lymphocytes and interleukin-2. Cancer Immunol Immunother. 1997;45:77–87. doi: 10.1007/s002620050405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kitahara T, Watanabe O, Yamaura A, et al. Establishment of interleukin 2 dependent cytotoxic T lymphocyte cell line specific for autologous brain tumor and its intracranial administration for therapy of the tumor. J Neurooncol. 1987;4:329–336. doi: 10.1007/BF00195603. [DOI] [PubMed] [Google Scholar]
  • 66.Crane CA, Han SJ, Ahn B, et al. Individual patient-specific immunity against high-grade glioma after vaccination with autologous tumor derived peptides bound to the 96 KD chaperone protein. Clin Cancer Res. 2013;19:205–214. doi: 10.1158/1078-0432.CCR-11-3358. [DOI] [PubMed] [Google Scholar]

RESOURCES