Abstract
Recent advances in glioblastoma therapy have led to optimism that more effective therapies will improve outcomes. Immunotherapy is a promising approach that has demonstrated the potential to eradicate cancer cells with cellular-level accuracy while minimizing damage to surrounding healthy tissue. Several vaccination strategies have been evaluated for activity against glioblastoma in clinical trials. These include peptide vaccines, polyvalent dendritic cell vaccines, heat shock protein vaccines and adoptive immunotherapy. In this review, we highlight clinical trials representative of each of these approaches and discuss strategies for integrating these therapies into routine patient care.
Keywords: chemotherapy, glioblastoma, immunotherapy, radiation therapy, tumor-associated antigen, vaccine
Recent advances in surgical techniques and innovations in the development and delivery of adjuvant therapies have improved the prognosis for patients with glioblastoma multiforme (GBM). Nonetheless, the outlook remains poor with a median life expectancy of 20 months [1,2] and a 3-year survival rate of only 10% [3]. Conventional therapies have succeeded in reliably delaying disease progression; however, the potential of these therapies to eradicate GBM is constrained by the tumor’s invasiveness, location and resistance to radiation and chemotherapy. Although metastasis outside the CNS is rare [4], GBM infiltrates normal brain tissue well beyond the radiographic borders of the tumor, precluding cure with surgical resection alone [5–7]. Radiation and chemotherapy are valuable adjuvant therapies; however, specific cell populations are chemo- and radio-resistant, resulting in inevitable tumor recurrence [8]. Specifically, failure to eliminate cancer stem cells likely plays a critical role in recurrence as this population has the capacity to repopulate the tumor bulk as well as differentiate into critical supporting structures [9,10].
The observation that infection sometimes precedes tumor regression predates modern medicine; however, the first attempts at purposeful manipulation of the immune system to fight cancer is credited to William Coley, who noted regression of a sarcoma following a case of erysipelas (streptococcal infection). Based on this observation, Coley administered killed bacteria vaccines (Coley’s Toxins) to a series of cancer patients and reported occasionally impressive results [11].
It was well into the next century, however, before rigorous scientific study of the immune system provided the tools necessary to understand and improve upon Coley’s approach. These efforts were introduced into widespread clinical consciousness in 2006 with the US FDA approval of Gardasil® (Merck, NJ, USA), the first preventative anticancer vaccine for cervical cancer. The approval of sipuleucel-T (Provenge®; Dendreon, WA, USA), a dendritic cell vaccine for hormone-resistant metastatic prostate cancer, in 2010 [12] and ipilimumab (Yervoy™; Bristol-Myers Squibb, NY, USA), a monoclonal antibody directed against the immune checkpoint CTLA-4, for metastatic melanoma in 2011 [13]; however, ushered in a new era in cancer immunotherapy, as these are the first active immunologic agents with proven activity against solid tumors.
Bolstered by these recent advances, immunotherapy appears to have arrived as a weapon in the oncology armamentarium. The potential of immunotherapy to target tumors with cellular-level accuracy makes this approach uniquely appealing for eliminating GBM cells that have infiltrated healthy brain tissue. However, there are a number of challenges still facing successful use of immunotherapy against CNS tumors.
Patients with high-grade gliomas have long been known to exhibit alterations in local and systemic immune responses [14,15]. Although an in-depth discussion of the immunosuppressive microenvironment of GBM is beyond the scope of this review, we have reviewed this topic in detail elsewhere [16]. The addition of steroids, radiation therapy and lymphodepleting cytotoxic agents further suppresses immune function [17]. In addition, immunotherapeutic agents may produce delayed and variable radiographic responses, even in patients who exhibit significant clinical benefit [18], complicating evaluation of the effectiveness of an immunologically active agent, and potentially delaying critical clinical decision-making.
Despite these challenges, given the current limitations of GBM therapies and the emergence of immune-based therapies in other cancers, at this juncture it is prudent to take stock of the current state of immunotherapy and envision how this approach might be employed in routine clinical practice. Two distinct sets of challenges must be confronted in accomplishing this goal. First, preclinical research must elucidate and exploit immune targets that reliably and safely generate antitumor responses in the CNS. These therapies must then be implemented against a rapidly progressive disease in an immunologically fragile patient population. This review has two purposes. First, we will highlight select vaccines in clinical trials in order to evaluate the current status of GBM immunotherapy. We will then draw from preclinical studies and experiences in other tumors to consider how this burgeoning treatment modality might be integrated into the care of GBM patients.
Current approaches
Peptide vaccines
Peptide vaccines involve administration of tumor-associated antigens (TAAs) in a pro-inflammatory context (usually via coadministration of an adjuvant) as a means of priming the immune system against cancer cells. This strategy requires antigen cross-presentation, but is otherwise similar to vaccination strategies commonly implemented in infectious disease [19].
The first step in selectively targeting tumor cells is identifying appropriate TAAs. Ideally, the expression of a targeted TAA is restricted to malignant cells (or malignant cells and nonvital tissues) and plays a critical role in tumor progression. Several TAAs common in other tumors have also been targeted in GBM, including HER-2, TRP-2, gp100, MAGE-1, IL-13Rα2 and AIM-2 [20]. However, owing to many of these antigens also being expressed in normal tissues, T cells directed against these antigens are subject to negative selection in the thymus. Conversely, neoantigens are not subject to negative selection and may represent more ideal immunologic targets.
EGF receptors (EGFRs) have been shown to drive tumor progression in a variety of cancers by regulating cell proliferation, differentiation and survival, as well as modulating downstream signaling pathways involved in invasion and angiogenesis [21,22]. Furthermore, aberrant EGFR activity has been observed in the majority of solid tumors, including GBM [23–29]. In the late 1980s, Burt Vogelstein, along with his postdoctoral fellow Albert Wong, and Darell Bigner codiscovered a variant of EGFR commonly expressed in GBMs [30,31]. This EGFR variant (EGFRvIII) is rarely expressed in normal tissues but is the most common variant of the EGFR in GBM, being expressed in 27–67% of tumors [23,32].
The first trial to demonstrate that targeting EGFRvIII with a peptide vaccine was safe and potentially efficacious was ACTIVATE [33]. This trial utilized a 14-amino acid peptide from the EGFRvIII protein (PEPvIII) conjugated to keyhole limpet hemocyanin. In this study, 18 subjects with immunohistochemical confirmation of EGFRvIII positivity were enrolled and received the vaccine in combination with radiation therapy and temozolomide. Three vaccines were administered in 2-week intervals with the first administered 2 weeks following surgery. After initial dosing, the vaccine was administered once per month until radiographic progression was noted. Median time to progression (14.2 months) and median survival (26 months) in this study compared favorably with historical controls. In addition, immune-based assays performed on sera from patients who received the vaccine showed increased titers of anti-EGFRvIII antibodies, as well as an increase in CD8+, IFNγ-producing T cells. Notably, 82% of tumors that recurred did not express EGFRvIII. The authors attributed this to immunoediting under immunologic pressure and interpreted this finding as support for the immunologic activity of the vaccine. Other authors, however, have cautioned against this interpretation and noted that independent confirmation of this finding is warranted [34].
While the ACTIVATE trial was ongoing, Stupp et al. demonstrated a survival benefit with surgery, radiation and continuous daily temozolomide followed by six cycles of adjuvant temozolomide [3]. The Stupp protocol subsequently became standard of care and was implemented in the ACTIVATE II trial [35]. Twenty-one subjects with confirmation of EGFRvIII expression were enrolled and received the PEPvIII vaccine, CDX-110 (Celldex Therapeutics, MA, USA). The first dose was administered within 6 weeks following completion of chemotherapy and radiation with an additional two doses administered at 2-week intervals. Vaccination was continued at 1-month intervals thereafter until the time of tumor progression. The results of this study corroborated the findings of the ACTIVATE trial as median time to progression was 15.2 months and median overall survival was 23.6 months [35].
A larger Phase II study of CDX-110 is currently in progress [36]. Termed ACT III, this study is a randomized, multicenter clinical trial with 81 patients enrolled. The treatment group received the Stupp protocol in addition to CDX-110, while the control group received the Stupp protocol alone. Based on interim data from the first 40 patients, the authors reported that 70% of patients were progression-free at 5.5 months [36].
There are two clinical trials of CDX-110 that are currently enrolling [201]. ACT IV is a PhaseIII clinical trial of CDX-110 in patients with newly diagnosed GBM. This is a two-arm, double-blind, randomized study in which half of the patients will receive CDX-110, a control cohort will receive a KLH vaccine and both arms will receive standard treatment based on the Stupp protocol. All patients will be followed until death. ReACT is a clinical trial of CDX-110 in patients with recurrent GBM. ReACT is a two-arm, double-blind, Phase II study that will compare the CDX-110 vaccine in combination with bevacizumab with bevacizumab plus keyhole limpet hemocyanin. Patients will be treated until radiographic evidence of disease progression is noted. At the time of progression, treatment with CDX-110 will be terminated and patients will be permitted to receive other therapies. A second group of patients previously refractory to bevacizumab will also be enrolled and will receive bevacizumab plus CDX-110. Additional information on the ACT IV and ReACT trials is available on the Celldex website [201]. Rindopepimut (CDX-110) is being studied in combination with temozolomide in newly diagnosed patients in a European trial currently underway [202]. In this study, 140 total patients will be enrolled. A summary of completed EGFRvIII peptide vaccine trials is provided in Table 1 and ongoing EGFRvIII peptide vaccine trials are summarized in Table 2.
Table 1.
Study | Phase | Patients (n) | Antigen | Trial results | Ref. |
---|---|---|---|---|---|
Sampson et al. (2010) | II | 18 | EGFRvIII | TTP 14.2 months Median survival 26 months |
[33] |
Del Vecchio et al. (2012) (preliminary results only) | II | 21 | EGFRvIII | Median survival 23.6 months | [35] |
EGFR: EGF receptor; TTP: Time to progression.
Table 2.
Study | Phase | Estimated enrollment (n) | Status | Primary outcome | Ref. |
---|---|---|---|---|---|
Phase II study of CDX-110 in patients with glioblastoma multiforme (ACT III) | II | 82 | Ongoing | Progression-free survival | [202] |
Phase I rindopepimut after conventional radiation in children with diffuse intrinsic pontine gliomas | I | 12 | Recruiting | Safety | [202] |
A study of rindopepimut/GM-CSF in patients with relapsed EGFRvIII-positive glioblastoma (ReACT) | II | 95 | Recruiting | Progression-free survival | [202] |
Phase III study of rindopepimut/GM-CSF in patients with newly diagnosed glioblastoma (ACT IV) | III | 440 | Recruiting | Overall survival | [202] |
A trial investigating the IMA950 vaccine and GM-CSF for glioblastoma | I | 45 | – | Safety | [202] |
An international, randomized, double-blind, controlled Phase II study of rindopepimut/GM-CSF with adjuvant temozolomide in patients with newly diagnosed, surgically resected, EGFRvIII-positive glioblastoma (IRRADIANCE) | II | 140 | – | Progression-free survival | [203] |
EGFR: EGF receptor.
While considerable effort has been invested in peptide vaccines targeting EGFRvIII, it should be noted that many other antigens are also being targeted using peptide vaccines. One particularly interesting vaccine, IMA950, consists of 11 synthetic TAAs and is in early Phase I testing in Europe. This vaccine is unique because nine of the TAAs bind to the HLA class I allele A*02, while two of them bind to HLA class II alleles. The theoretical advantage of this strategy is that both cytotoxic T lymphocytes (CTLs) and T-helper cells are expected to have antitumor activity [37].
Dendritic cell vaccines
Dendritic cells (DCs) are a subset of leukocytes that are derived from CD34+ bone marrow progenitor cells and function primarily in immune surveillance and antigen presentation [38]. DCs are professional APCs, circulating throughout the body and surveying the local environment using receptors for pathogen-associated molecular patterns. Once the immature DC recognizes a protein structure that is unique to a pathogen and becomes activated, the foreign protein is endocytosed and the DC migrates to T-cell rich areas of lymph nodes, where the foreign peptide is presented on MHC class I or II [39]. Once activated, DCs express proinflammatory cytokines and are potent stimulators of both CD4 and CD8 T cells [40]. Clinical trials of DCs pulsed with TAAs have shown some promise, despite heterogeneity in dose, route, location of administration and adjuvant [20,41–50].
The first study to use DCs loaded with the EGFRvIII peptide was the VICTOR I study [51]. In this trial, DCs were preloaded with the EGFRvIII peptide conjugated to KLH. Fifteen patients were initially enrolled, although only 12 received the vaccine as three patients experienced progression during radiotherapy. Three doses were administered within the first 2 weeks following surgery and two additional doses were administered at 2-week intervals thereafter. The results of this study demonstrated that the vaccine was safe and suggested a potential survival benefit as the median time to progression of 18.7 months and median survival of 22.8 months compared favorably with historical controls [51].
The ICT-107 vaccine (Immunocellular Therapeutics, CA, USA) and DC-Vax Brain (Northwest Biotherapeutics, MD, USA) contain DCs loaded with multiple TAAs. The ICT-107 vaccine uses DCs loaded with TRP-2, GP100, HER2, MAGE-1, IL13Rα2 and AIM-2. A Phase I study of this vaccine in newly diagnosed GBM patients reported a progression-free survival of 16.9 months and a median overall survival of 38.4 months [20]. Vaccination with DC-Vax Brain (Northwest Biotherapeutics) included administration of a Toll-like receptor agonist and reported similar results in a Phase I study of DCs pulsed with autologous tumor lysate [44]. Of note, only minor toxicities have been reported in these DC vaccine trials. Despite these studies being underpowered to demonstrate a survival benefit, the results further support the safety and potential efficacy of DC tumor vaccines. A summary of select DC vaccine studies is provided in Table 3. Ongoing and actively recruiting clinical trials, as obtained through the NIH clinical trials database [203], is provided in Table 4.
Table 3.
Study | Phase | Patients (n) | Number of injected DCs | DC loading | Location of injection | Trial result(s) | Ref. |
---|---|---|---|---|---|---|---|
Phuphanich et al. (2012) | I | 17 | 1 × 107 | Peptide epitopes: HER2, TRP-2, gp100, MAGE-1, IL13Rα2 and AIM-2. HLA-A1 and/or HLA-A2 | ID | PFS: 16.9 months Median survival: 38.4 months |
[20] |
Iwami et al. (2012) | I | 8 | 1 × 107 | IL13Rα2 | ID | SD: 1 Regression: 1 |
[106] |
Prins et al. (2011) | I | 23 | – | Tumor cell lysate | ID | Median survival: 31.4 months | [44] |
Okada et al. (2011) | I/II | 13 | – | Peptide epitopes EphA2, IL-13Rα2, YKL-40 and gp100 | ID | SD: 6 CR: 1 PR: 2 Median survival for recurrent GBM: 12 months |
[107] |
Sampson et al. (2009) | I | 12 | 1 × 108 | EGFRvIII-specific peptide conjugated to keyhole limpet hemocyanin | ID | TTP: 10.2 months Median survival: 18.7 months |
[45] |
Wheeler et al. (2008) | II | 32 | 1 × 107 to 4 × 107 | Tumor cell lysate | ID | TTP: 208 vs 167 days for controls | [47] |
De Vleeschouwer et al. (2008) | I/II | 56 | 0.7 × 106 to 1 × 106 | Tumor cell lysate | ID | 12-month OS: 37.4% 24-month OS: 14.8% 36-month OS: 11.1% |
[41] |
Walker et al. (2008) | I | 13 | 1 × 106 | Tumor cell lysate | ID | 1-year survival: 38% | [46] |
Yamanaka et al. (2005) | I/II | 24 | 1 × 107 | Tumor cell lysate | ID or ID and IT | PR: 1 MR: 3 SD: 10 |
[49] |
Kikuchi et al. (2004) | I | 15 | 3.6 × 106 to 32.3 × 106 | Tumor cell lysate | ID | PR: 4 NC: 1 MR: 1 |
[42] |
Yamanaka et al. (2003) | I/II | 10 | 10 × 106 to 32 × 106 | Tumor cell lysate | ID and IT | MR: 2 NC: 4 |
[48] |
Kikuchi et al. (2001) | I | 8 | 2.4 × 106 to 8.7 × 106 | Tumor cell lysate | ID | SD: 6 | [43] |
Yu et al. (2001) | I | 9 | 1 × 106 | MHC class I peptides | ID | Median survival: 455 vs 257 days for controls | [50] |
CR: Complete response; DC: Dendritic cell; EGFR: EGF receptor; GBM: Glioblastoma multiforme; ID: Intradermal; IT: Intratumoral; MR: Minor response; NC: No change; OS: Overall survival; PFS: Progression-free survival; PR: Partial response; SD: Stable disease; TTP: Time to progression.
Table 4.
Study | Phase | Estimated enrollment (n) | Status | Primary outcome(s) | Ref. |
---|---|---|---|---|---|
Phase II feasibility study of DC vaccination for newly diagnosed GBM | II | 11 | Ongoing | Tumor-specific, cytotoxic T-cell response | [202] |
Vaccine therapy in treating patients with malignant glioma | I | 8 | Ongoing | Dose-limiting toxicity, survival, tumor progression and cellular immune response | [202] |
Vaccine therapy in treating patients with newly diagnosed GBM | I | 16 | Ongoing | Safety and feasibilty | [202] |
DC cancer vaccine for high-grade glioma (GBM-Vax) | II | 56 | Recruiting | PFS | [202] |
Safe study of DC-based therapy targeting tumor stem cells in glioblastoma | I/II | 20 | Recruiting | Adverse events | [202] |
Efficacy and safety of autologous DC vaccination in GBM after complete surgical resection | II | 37 | Recruiting | PFS | [202] |
Vaccine therapy in treating patients with malignant glioma | I | 18 | Ongoing | Dose-limiting toxicity | [202] |
DC vaccination for patients with solid tumors | I/II | 10 | Recruiting | Immunogenicity | [202] |
Study of a drug [DCVax®-L] to treat newly diagnosed GBM brain cancer | III | 300 | Recruiting | PFS | [202] |
A study of ICT-107 immunotherapy in GBM | II | 200 | Recruiting | OS | [202] |
Vaccine therapy in treating patients undergoing surgery for recurrent GBM | I | 50 | Recruiting | Safety and feasibility | [202] |
DC: Dendritic cell; GBM: Glioblastoma multiforme; OS: Overall survival; PFS: Progression-free survival.
Heat shock protein vaccines
Heat shock proteins (HSPs) aid in protein folding, regulate apoptosis and modulate immune responses [52–54]. HSPs are overexpressed in the cytoplasm and on the cell surface of GBM cells [55]. This overexpression is likely due to a combination of the hypoxic GBM microenvironment and the high metabolic demands of rapidly proliferating tumor cells [56]. HSPs have been shown to interact with many proteins known to drive tumorigenesis, including EGFR, PDGF receptor, FAK, AKT, p53 and PI3K [57–59].
HSP vaccines are a subclass of protein vaccines that are composed of HSPs bound to tumor peptides. In this setting, the HSP is believed to be a proinflammatory signal while the peptide provides a specific immunologic target. Evidence for this comes from preclinical studies that have demonstrated that antitumor immunity is specific to HSPs derived from tumor cells rather than normal tissue [60–62]. In addition, neither tumor-derived HSPs nor TAAs alone induced antitumor immunity. However, when TAAs were complexed to HSPs, antitumor immunity was restored. However, when carried by albumin in human serum, the same peptides did not result in antitumor activity [63]. These data suggest that the HSP–peptide complex is required to induce antitumor immunity.
APCs mediate the antitumor immune response in HSP vaccines via interaction with the cell surface receptors CD91, TLR-4 and CD14 [63–68]. Following recognition of the HSP–peptide complex by APCs, TAAs are endocytosed and presented in the context of MHC class I to prime CD8 T cells [69]. In this process, proinflammatory cytokines, including TNF-α, IL-1β, IL-12, IL-6 and GM-CSF, are released from the activated APC, supporting immune cell activation.
The only HSP vaccine with results reported in GBM is the ProPhage (Agenus, MA, USA) vaccine (formerly HSPPC-96), which is composed of the 96 kDa HSP, gp96, complexed to endogenous tumor peptides [70]. This vaccine is manufactured using multistep affinity and nonaffinity chromatography and is currently under clinical investigation for GBM along with a variety of other solid and blood-borne cancers [71–73]. Results of the first Phase I study in patients with recurrent GBM showed promising results (Table 5). In patients receiving four vaccines, 11/12 patients responded with a median survival of 47 weeks. Brain biopsy revealed CD4+, CD8+, CD56+ and IFN-γ-producing cells [74]. A summary of actively recruiting trials for HSP vaccines is also provided in Table 5.
Table 5.
Study | Phase | Estimated enrollment (n) | Trial results | Primary outcome(s) | Ref. |
---|---|---|---|---|---|
Completed | |||||
Crane et al. (2012) | I | 12 | Median survival: 47 weeks in immunologic responders | – | [74] |
Ongoing | |||||
HSPPC-96 vaccine with temozolomide in patients with newly diagnosed GBM (HeatShock) | II | 55 | Ongoing | Safety, survival | [202] |
GP96 HSP–peptide complex vaccine in treating patients with recurrent or progressive glioma | I/II | 50 | Ongoing | Safety, maximum tolerated dose, toxicity and PFS | [202] |
GBM: Glioblastoma multiforme; HSP: Heat shock protein; PFS: Progression-free survival.
Adoptive immunotherapy
Adoptive immunotherapy involves ex vivo activation of autologous immune cells and subsequent infusion back into the patient. This strategy has primarily used lymphocyte-activated killer cells and CTLs. Lymphocyte-activated killer cells are generated by culturing autologous peripheral lymphocytes with IL-2, generating T cells and NK cells. These cells are then injected intra-tumorally or -venously, where they can become activated by host APCs. Clinical trials using adoptive immunotherapy with LAK cells have reported variable toxicity and efficacy (Table 6).
Table 6.
Study | Phase | Patients (n) | Results | Ref. |
---|---|---|---|---|
Dillman et al. (2009) | I/II | 33 | Median survival: 20.5 months | [108] |
Dillman et al. (2004) | I/II | 40 | Median survival: 17.5 months | [109] |
Sankhla et al. (1996) | I | 10 | PR: 2 | [110] |
Hayes et al. (1995) | I | 15 | Median survival following reoperation: 53 weeks | [111] |
Boiardi et al. (1994) | I | 9 | CR: 1 PR: 2 SD: 4 |
[112] |
Jeffes et al. (1993) | I | 19 | Median survival: 37 weeks | [113] |
Blancher et al. (1993) | I | 13 | Tumor progression noted after 4–12 weeks | [114] |
Lillehei et al. (1991) | I | 20 | Median survival: 63 weeks | [115] |
Barba et al. (1989) | I | 10 | PR: 1 | [116] |
Jacobs et al. (1986) | I | 9 | No PR or SD | [117] |
CR: Complete response; PR: Partial response; SD: Stable disease.
Other studies have used tumor-infiltrating T lymphocytes or T cells from draining lymph nodes (Table 7). In comparison with studies with LAK cells, peripheral transfer of CTLs was associated with only minor toxicities. Conversely, more serious toxicities, including transient cerebral edema and hemorrhage, were reported in studies where CTLs were injected directly into the tumor cavity [75,76].
Table 7.
Study | Phase | Patients (n) | Results | Ref. |
---|---|---|---|---|
Tsuboi et al. (2003) | I | 10 | CR: 1 PR: 4 Median survival: 5 months Overall response rate: 50% |
[76] |
Wood et al. (2000) | I | 9 | PR: 3 Survival >4 years: 2 |
[118] |
Plautz et al. (2000) | I | 9 | PR: 3 (1 GBM and 2 grade III survival >4 years) |
[119] |
Sloan et al. (2000) | I | 19 | CR: 1 PR: 7 Median survival: 12 months |
[120] |
Quattrocchi et al. (1999) | I | 6 | CR: 1 PR: 2 |
[75] |
Tsurushima et al. (1999) | I | 4 | PR: 3 SD: 1 |
[121] |
Plautz et al. (1998) | I | 10 | SD: 1 4 patients alive after 1 year |
[122] |
Kruse et al. (1997) | I | 5 | Transient toxicities: Survival (AO) >30 months Survival (AA) >28 months |
[123] |
Holladay et al. (1996) | I | 15 | No PR or SD Disease-free survival >8 months: 7 |
[124] |
Kitahara et al. (1987) | I | 5 | PR: 2 1 patient alive >104 weeks |
[125] |
AA: Anaplastic astrocytoma; AO: Anaplastic oligodendroglioma; CR: Complete response; GBM: Glioblastoma multiforme; PR: Partial response; SD: Stable disease.
Implementing immunotherapy for GBM
Experience with conventional cancer therapies suggests that combination therapy is generally superior to monotherapy [3,2]. This principle likely holds true for immunotherapy as well. For example, data from a Phase I trial of combination immunotherapy with ipilimumab and bevacizumab in advanced melanoma suggest that combination therapy can be administered safely and may be superior to either therapy alone [77]. In addition, tumors may more readily develop resistance against single-agent immunotherapy. As previously mentioned, GBM has been reported to develop resistance in clinical trials of the EGFRvIII peptide vaccine by downregulating expression of the targeted antigen [33]. If this is the case, one strategy for combating immunoediting may be combining EGFRvIII vaccination with antineoplastic therapies with activity against non-EGFRvIII expressing cells. Finally, given the pace of GBM progression and the potential consequences of delaying treatment, upfront, multimodal combination therapy has obvious appeal.
Combination with chemotherapy
Lymphodepletion is a common side effect of many chemotherapeutic agents and is generally considered counterproductive to immunotherapy. However, other effects of chemotherapy include spilling of TAAs [78] and upregulation of pro-inflammatory molecules [79]. In addition, some agents, such as cyclophosphamide, may selectively deplete immunosuppressive T regs [80]. Locally harnessing these proinflammatory effects while avoiding systemic immune suppression has generally proven challenging. For example, combining sipuleucel-T with chemotherapy in patients with metastatic prostate cancer has not demonstrated a reliable benefit in survival or disease control [81].
Trials of some chemoimmunotherapy combinations, however, support the potential efficacy of this approach. For example, a recent trial of ipilimumab plus dacarbazine in 502 patients with metastatic melanoma found that the addition of ipilimumab improved overall survival compared with dacarabzine alone (11.2 vs 9.1 months) and carried a risk of adverse events comparable with ipilimumab alone [82]. A smaller Phase II study compared this combination with ipilimumab and reported a trend toward increased overall survival with combination therapy (11.4 vs 14.3 months) [83].
Preclinical data suggest that local drug delivery may be superior to systemic administration for the treatment of intracranial tumors in combinatorial chemoimmunotherapy regimens. Local delivery of chemotherapy has several theoretical advantages, including circumventing the blood–brain barrier and harnessing local proinflammatory effects while minimizing systemic immune toxicity. For example, a preclinical study found that local delivery of carmustine using biodegradable wafers in combination with local delivery of IL-2 demonstrated a synergistic survival benefit in a rat glioma model [84]. This effect does not seem to be limited to a specific chemotherapy, as biodegradable polymers loaded with a variety of chemotherapeutic agents have been shown to not only improve survival, but also augment the local inflammatory response in the setting of local IL-2 administration [85].
Local delivery of immunotherapy may also be advantageous for some agents. Immune responses in the brain require coordination of local and systemic immune activation [86]; thus, some therapies may be more immunologically active when delivered directly to the CNS than when administered systemically [87,88]. IL-2 provides the most striking example. Preclinical data suggest that intracranial implantation of nonreplicating tumor cells transduced with IL-2 affords protection against intracranial tumor challenge in a murine glioma model; however, this protective effect is not observed when the cells are implanted subcutaneously [89].
There have been no clinical trials of combinatorial chemoimmunotherapy for malignant gliomas to date. Trials of chemotherapy and systemically delivered IL-2 in other tumors, however, have been generally disappointing [90–92]. This is perhaps not surprising in light of the preclinical studies discussed above and the well-established role of autocrine IL-2 signaling in CD8 T-cell function [93]. Of course, in meta-static cancer, local drug delivery is difficult to achieve given the inherent difficulty of targeting occult metastases and circulating tumor cells. In brain tumors, however, a variety of technologies ranging from biodegradable polymers to micro-chips are capable of delivering agents directly to the tumor site [94–96]. Furthermore, retrospective data suggest that local delivery of bis-chloroethylnitrosourea combined with systemic temozolomide is safe and may produce favorable median survival in patients with GBM [1].
Combination with radiation therapy
Radiation therapy has long been a mainstay of GBM treatment for its direct cytotoxic effects, but the immunologic effects of ionizing radiation are only beginning to be understood [97]. The hypothesis that localized radiation therapy can produce systemic antitumor activity (the abscopal effect) dates back to the early 1950s [98]. Clinical documentation of this phenomenon, however, has been scarce. The most convincing evidence for an immune-mediated abscopal effect in humans comes from a recent case report of a patient with metastatic melanoma receiving ipilimumab [99]. This patient received focused radiation therapy to a paraspinal mass and experienced regression of several lesions outside the irradiated field. Interestingly, this response corresponded with serologic evidence of an immune response against the TAA NY-ESO-1.
The abscopal effect has not been documented in primary or metastatic brain tumors. Recently, however, the case of a patient with a previously observed abscopal effect who developed a brain metastasis and underwent focal radiation therapy to this lesion and treatment with ipilimumab was reported [100]. This patient exhibited regression of his extracranial metastases following intracranial radiation therapy with a corresponding increase in anti-MAGEA3 titer and evidence of a new serologic response to PASD1. In this prime-boost scenario, it is difficult to determine if the second response resulted from irradiation of the patient’s intracranial lesion, ipilimumab administration or both. Nevertheless, taken together, these reports suggest that the combination of radiation therapy and ipilimumab may promote offsite tumor regression and, furthermore, provide evidence that radiation therapy has immunologic activity in the CNS. It should be noted that radiation therapy has been associated with a fall in CD4 count in glioma patients [101]. More detailed immunologic evaluation may provide a putative mechanism for this phenomenon, allowing for identification of therapeutic targets.
Combination with surgery
Surgery is the cornerstone of therapy for high-grade gliomas and must be evaluated as an integral part of any prospective immunotherapy regimen. The use of perioperative corticosteroids is an obvious consideration. Further study is needed to delineate the deleterious effects of corticosteroids on specific immunotherapy regimens. This information may then be used to design treatment regimens that minimize these effects. In addition, the consequences of delaying adjuvant immunotherapy while a patient recovers from surgery must be evaluated. Neo-adjuvant immunotherapy may potentially obviate some of these adverse effects; however, in CNS tumors, the theoretical advantages of neo-adjuvant immunotherapy should be tempered with the potential for iatrogenic edema prior to surgical decompression.
While these are critical practical considerations, it is perhaps more interesting to consider the immunologic effects of surgery itself. For example, in preclinical cancer models, cytoreductive surgery has been demonstrated to restore anti-tumor immune activity to deactivated immune cells [102]. The mechanisms of this response are not well characterized, but may be secondary to increased T-cell trafficking or sudden reduction of antigen load. Surgery also affords direct access to the tumor, facilitating local delivery of immunologically active agents and affording an opportunity to tailor therapies to the immunologic microenvironment of the patient’s tumor. One such example is the use of autologous whole-cell lysate to load DCs or HSP vaccines [103]. Another example is the strategy of harvesting tumor-infiltrating lymphocytes, expanding and activating these cells ex vivo and infusing them for adoptive T-cell therapy. The latter approach has been evaluated in clinical trails for melanoma, with encouraging results [104].
Future perspective
Immunotherapy is a promising approach in GBM due to its potential to eradicate neoplastic cells while sparing normal tissue and generating durable antitumor activity. In addition, immunotherapy may effectively target specific cell populations [105], an approach that could be expanded to eliminate cells that are resistant to chemotherapy and ionizing radiation. Significant efforts are underway to develop immunologically active agents against GBM, including a number of completed and ongoing clinical trials [16]. The results of these trials will provide further insight into mechanisms of escape and guide the development of future therapies.
We believe that in addition to identifying new immunologic targets and developing therapies to exploit these targets, the next 5–10 years will see a significant effort in understanding the immunologic effects of conventional GBM therapies and developing safe, and potentially powerful combination immunotherapy regimens. Several classes of immunotherapeutic agents are currently in development. In addition to active immunotherapy, adoptive cell transfer and antibodies against immune checkpoints, such as CTLA-4 and PD-1, and other targets with immunologic activity such as VEGF, hold the potential to generate robust antitumor immune responses.
Executive summary.
Current approaches
The most extensively studied vaccine strategies have been peptide, dendritic cell and heat shock protein vaccines.
Passive immunotherapies, including adoptive immunotherapy have also been evaluated with variable results.
EGF receptor vIII is the most frequently targeted glioblastoma multiforme (GBM) neoantigen.
Other tumor-associated antigens, including TRP-2, GP100, HER2, MAGE-1, IL13Rα2 and AIM-2 have also been targeted in polyvalent vaccines.
Dendritic cell vaccines for GBM have generally been well tolerated with minimal side effects.
Although some of these agents have demonstrated some activity against GBM, to-date none have been shown to be superior to current standard of care.
Implementing immunotherapy for GBM
Combination immunotherapy is likely superior to monotherapy and is currently being tested in clinical trials for CNS and non-CNS malignancies.
Understanding the immunologic effects of traditional therapies for GBM, such as chemotherapy, ionizing radiation and surgery, will prove critical for integrating immunotherapy into routine clinical practice.
Local delivery of chemotherapy may be more immunologically advantageous than systemic delivery.
Radiation has been shown to have both immuno-suppressive and -stimulatory effects. A better understanding of these effects will allow for development of combination regimens in which ionizing radiation is used to augment antitumor immune responses.
Surgery will likely remain the mainstay of therapy for GBM; however, the immunologic consequences of surgery are poorly understood. Immunotherapies must be integrated into patient care in a manner that does not delay or complicate surgery and ideally takes advantage of postoperative inflammation and access to tumor tissue, including tumor-infiltrating immune cells.
Footnotes
For reprint orders, please contact: reprints@futuremedicine.com
Financial & competing interests disclosure
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as:
▪ of interest
▪▪ of considerable interest
- 1.McGirt MJ, Than KD, Weingart JD, et al. Gliadel (BCNU) wafer plus concomitant temozolomide therapy after primary resection of glioblastoma multiforme. J Neurosurg. 2009;110(3):583–588. doi: 10.3171/2008.5.17557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Miglierini P, Bouchekoua M, Rousseau B, Dam Hieu P, Malhaire JP, Pradier O. Impact of the per-operatory application of GLIADEL wafers (BCNU, carmustine) in combination with temozolomide and radiotherapy in patients with glioblastoma multiforme: efficacy and toxicity. Clin Neurol Neurosurg. 2012;114(9):1222–1225. doi: 10.1016/j.clineuro.2012.02.056. [DOI] [PubMed] [Google Scholar]
- 3▪▪.Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. doi: 10.1056/NEJMoa043330. This trial established the current standard of care for glioblastoma. [DOI] [PubMed] [Google Scholar]
- 4.Zhen L, Yufeng C, Zhenyu S, Lei X. Multiple extracranial metastases from secondary glioblastoma multiforme: a case report and review of the literature. J Neurooncol. 2010;97(3):451–457. doi: 10.1007/s11060-009-0044-9. [DOI] [PubMed] [Google Scholar]
- 5.Kallenberg K, Bock HC, Helms G, et al. Untreated glioblastoma multiforme: increased myo-inositol and glutamine levels in the contralateral cerebral hemisphere at proton MR spectroscopy. Radiology. 2009;253(3):805–812. doi: 10.1148/radiol.2533071654. [DOI] [PubMed] [Google Scholar]
- 6.Lu KV, Chang JP, Parachoniak CA, et al. VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell. 2012;22(1):21–35. doi: 10.1016/j.ccr.2012.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gordon VD, Valentine MT, Gardel ML, et al. Measuring the mechanical stress induced by an expanding multicellular tumor system: a case study. Exp Cell Res. 2003;289(1):58–66. doi: 10.1016/s0014-4827(03)00256-8. [DOI] [PubMed] [Google Scholar]
- 8.Chen J, Li Y, Yu TS, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488(7412):522–526. doi: 10.1038/nature11287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wang R, Chadalavada K, Wilshire J, et al. Glioblastoma stem-like cells give rise to tumour endothelium. Nature. 2010;468(7325):829–833. doi: 10.1038/nature09624. [DOI] [PubMed] [Google Scholar]
- 10.Ricci-Vitiani L, Pallini R, Biffoni M, et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature. 2010;468(7325):824–828. doi: 10.1038/nature09557. [DOI] [PubMed] [Google Scholar]
- 11.Wiemann B, Starnes CO. Coley’s toxins, tumor necrosis factor and cancer research: a historical perspective. Pharmacol Ther. 1994;64(3):529–564. doi: 10.1016/0163-7258(94)90023-x. [DOI] [PubMed] [Google Scholar]
- 12.Bot A. The landmark approval of Provenge, what it means to immunology and “in this issue”: the complex relation between vaccines and autoimmunity. Int Rev Immunol. 2010;29(3):235–238. doi: 10.3109/08830185.2010.490777. [DOI] [PubMed] [Google Scholar]
- 13.Ledford H. Melanoma drug wins US approval. Nature. 2011;471(7340):561. doi: 10.1038/471561a. [DOI] [PubMed] [Google Scholar]
- 14.Ullen H, Blom U, Blomgren H, von Holst H. Blood lymphocyte subsets in patients with primary intracranial tumours. Correlation to histological tumour type and anatomical site. Acta Neurochir (Wien) 1986;81(3–4):100–105. doi: 10.1007/BF01401229. [DOI] [PubMed] [Google Scholar]
- 15.Bodmer S, Strommer K, Frei K, et al. Immunosuppression and transforming growth factor-beta in glioblastoma. Preferential production of transforming growth factor-beta 2. J Immunol. 1989;143(10):3222–3229. [PubMed] [Google Scholar]
- 16.Jackson C, Ruzevick J, Phallen J, Belcaid Z, Lim M. Challenges in immunotherapy presented by the glioblastoma multiforme microenvironment. Clin Dev Immunol. 2011;2011:732413. doi: 10.1155/2011/732413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fadul CE, Fisher JL, Gui J, Hampton TH, Cote AL, Ernstoff MS. Immune modulation effects of concomitant temozolomide and radiation therapy on peripheral blood mononuclear cells in patients with glioblastoma multiforme. Neuro Oncol. 2011;13(4):393–400. doi: 10.1093/neuonc/noq204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.O’Regan KN, Jagannathan JP, Ramaiya N, Hodi FS. Radiologic aspects of immune-related tumor response criteria and patterns of immune-related adverse events in patients undergoing ipilimumab therapy. AJR Am J Roentgenol. 2011;197(2):W241–W246. doi: 10.2214/AJR.10.6032. [DOI] [PubMed] [Google Scholar]
- 19.Rock KL, Shen L. Cross-presentation: underlying mechanisms and role in immune surveillance. Immunol Rev. 2005;207:166–183. doi: 10.1111/j.0105-2896.2005.00301.x. [DOI] [PubMed] [Google Scholar]
- 20▪.Phuphanich S, Wheeler CJ, Rudnick JD, et al. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother. 2012 doi: 10.1007/s00262-012-1319-0. (Epub ahead of print). Phase I clinical trial of ICT-107, a dendritic cell vaccine targeting multiple class I peptides. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Engebraaten O, Bjerkvig R, Pedersen PH, Laerum OD. Effects of EGF, bFGF, NGF and PDGF(bb) on cell proliferative, migratory and invasive capacities of human brain-tumour biopsies in vitro. Int J Cancer. 1993;53(2):209–214. doi: 10.1002/ijc.2910530206. [DOI] [PubMed] [Google Scholar]
- 22.Goldman CK, Kim J, Wong WL, King V, Brock T, Gillespie GY. Epidermal growth factor stimulates vascular endothelial growth factor production by human malignant glioma cells: a model of glioblastoma multiforme pathophysiology. Mol Biol Cell. 1993;4(1):121–133. doi: 10.1091/mbc.4.1.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wong AJ, Ruppert JM, Bigner SH, et al. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc Natl Acad Sci USA. 1992;89(7):2965–2969. doi: 10.1073/pnas.89.7.2965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gorgoulis V, Aninos D, Mikou P, et al. Expression of EGF, TGF-alpha and EGFR in squamous cell lung carcinomas. Anticancer Res. 1992;12(4):1183–1187. [PubMed] [Google Scholar]
- 25.Irish JC, Bernstein A. Oncogenes in head and neck cancer. Laryngoscope. 1993;103(1 Pt 1):42–52. doi: 10.1288/00005537-199301000-00009. [DOI] [PubMed] [Google Scholar]
- 26.Korc M, Meltzer P, Trent J. Enhanced expression of epidermal growth factor receptor correlates with alterations of chromosome 7 in human pancreatic cancer. Proc Natl Acad Sci USA. 1986;83(14):5141–5144. doi: 10.1073/pnas.83.14.5141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Moorghen M, Ince P, Finney KJ, Watson AJ, Harris AL. Epidermal growth factor receptors in colorectal carcinoma. Anticancer Res. 1990;10(3):605–611. [PubMed] [Google Scholar]
- 28.Ishikawa J, Maeda S, Umezu K, Sugiyama T, Kamidono S. Amplification and overexpression of the epidermal growth factor receptor gene in human renal-cell carcinoma. Int J Cancer. 1990;45(6):1018–1021. doi: 10.1002/ijc.2910450606. [DOI] [PubMed] [Google Scholar]
- 29.Zajchowski D, Band V, Pauzie N, Tager A, Stampfer M, Sager R. Expression of growth factors and oncogenes in normal and tumor-derived human mammary epithelial cells. Cancer Res. 1988;48(24 Pt 1):7041–7047. [PubMed] [Google Scholar]
- 30▪.Humphrey PA, Wong AJ, Vogelstein B, et al. Amplification and expression of the epidermal growth factor receptor gene in human glioma xenografts. Cancer Res. 1988;48(8):2231–2238. This study, along with [31], describes the discovery of EGF receptor mutations in human gliomas. [PubMed] [Google Scholar]
- 31▪.Yamazaki H, Fukui Y, Ueyama Y, et al. Amplification of the structurally and functionally altered epidermal growth factor receptor gene (c-erbB) in human brain tumors. Mol Cell Biol. 1988;8(4):1816–1820. doi: 10.1128/mcb.8.4.1816. This study, along with [30], describes the discovery of EGF receptor mutations in human gliomas. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Humphrey PA, Wong AJ, Vogelstein B, et al. Anti-synthetic peptide antibody reacting at the fusion junction of deletion-mutant epidermal growth factor receptors in human glioblastoma. Proc Natl Acad Sci USA. 1990;87(11):4207–4211. doi: 10.1073/pnas.87.11.4207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33▪▪.Sampson JH, Heimberger AB, Archer GE, et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28(31):4722–4729. doi: 10.1200/JCO.2010.28.6963. The authors report on a Phase II trial of an EGFRvIII-targeted peptide vaccine and present evidence for immunologic escape via loss of the EGFRvIII antigen. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lesniak MS. Immunotherapy for glioblastoma: the devil is in the details. J Clin Oncol. 2011;29(22):3105. doi: 10.1200/JCO.2011.34.9019. author reply 3105–3106. [DOI] [PubMed] [Google Scholar]
- 35.Del Vecchio CA, Li G, Wong AJ. Targeting EGF receptor variant III: tumor-specific peptide vaccination for malignant gliomas. Expert Rev Vaccines. 2012;11(2):133–144. doi: 10.1586/erv.11.177. [DOI] [PubMed] [Google Scholar]
- 36.Lai R, Recht LD, Reardon DA, et al. Interim data for ACT III: Phase II trial of PF-04948568 (CDX-110) in combination with temozolomide (TMZ) in patients with glioblastoma (GBM) J Clin Oncol. 2010;28(Suppl 15):Abstract 2014. [Google Scholar]
- 37.Oliver Schoor NH, Dutoit V, Weinschenk T, et al. IMA950: a novel multi-peptide cancer vaccine for treatment of glioma. Presented at: American Association for Cancer Research 101st Annual Meeting; Washington, DC, USA. April 17–21 2010.2010. [Google Scholar]
- 38.Lipscomb MF, Masten BJ. Dendritic cells: immune regulators in health and disease. Physiol Rev. 2002;82(1):97–130. doi: 10.1152/physrev.00023.2001. [DOI] [PubMed] [Google Scholar]
- 39.Reis e Sousa C, Stahl PD, Austyn JM. Phagocytosis of antigens by Langerhans cells in vitro. J Exp Med. 1993;178(2):509–519. doi: 10.1084/jem.178.2.509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Steinman RM, Turley S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med. 2000;191(3):411–416. doi: 10.1084/jem.191.3.411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.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(10):3098–3104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
- 42.Kikuchi T, Akasaki Y, Abe T, et al. Vaccination of glioma patients with fusions of dendritic and glioma cells and recombinant human interleukin 12. J Immunother. 2004;27(6):452–459. doi: 10.1097/00002371-200411000-00005. [DOI] [PubMed] [Google Scholar]
- 43.Kikuchi T, Akasaki Y, Irie M, Homma S, Abe T, Ohno T. Results of a Phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunol Immunother. 2001;50(7):337–344. doi: 10.1007/s002620100205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Prins RM, Soto H, Konkankit V, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17(6):1603–1615. doi: 10.1158/1078-0432.CCR-10-2563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sampson JH, Archer GE, Mitchell DA, et al. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol Cancer Ther. 2009;8(10):2773–2779. doi: 10.1158/1535-7163.MCT-09-0124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Walker DG, Laherty R, Tomlinson FH, Chuah T, Schmidt C. Results of a Phase I dendritic cell vaccine trial for malignant astrocytoma: potential interaction with adjuvant chemotherapy. J Clin Neurosci. 2008;15(2):114–121. doi: 10.1016/j.jocn.2007.08.007. [DOI] [PubMed] [Google Scholar]
- 47.Wheeler CJ, Black KL, Liu G, et al. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Res. 2008;68(14):5955–5964. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
- 48.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(7):1172–1179. doi: 10.1038/sj.bjc.6601268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yamanaka R, Homma J, Yajima 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(11):4160–4167. doi: 10.1158/1078-0432.CCR-05-0120. [DOI] [PubMed] [Google Scholar]
- 50.Yu JS, Wheeler CJ, Zeltzer 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(3):842–847. [PubMed] [Google Scholar]
- 51.Sampson JH, Archer GE, Mitchell DA, Heimberger AB, Bigner DD. Tumor-specific immunotherapy targeting the EGFRvIII mutation in patients with malignant glioma. Semin Immunol. 2008;20(5):267–275. doi: 10.1016/j.smim.2008.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Borkovich KA, Farrelly FW, Finkelstein DB, Taulien J, Lindquist S. hsp82 is an essential protein that is required in higher concentrations for growth of cells at higher temperatures. Mol Cell Biol. 1989;9(9):3919–3930. doi: 10.1128/mcb.9.9.3919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Beckmann RP, Mizzen LE, Welch WJ. Interaction of Hsp 70 with newly synthesized proteins: implications for protein folding and assembly. Science. 1990;248(4957):850–854. doi: 10.1126/science.2188360. [DOI] [PubMed] [Google Scholar]
- 54.Gurbuxani S, Bruey JM, Fromentin A, et al. Selective depletion of inducible HSP70 enhances immunogenicity of rat colon cancer cells. Oncogene. 2001;20(51):7478–7485. doi: 10.1038/sj.onc.1204948. [DOI] [PubMed] [Google Scholar]
- 55.Graner MW, Cumming RI, Bigner DD. The heat shock response and chaperones/heat shock proteins in brain tumors: surface expression, release, and possible immune consequences. J Neurosci. 2007;27(42):11214–11227. doi: 10.1523/JNEUROSCI.3588-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hermisson M, Strik H, Rieger J, Dichgans J, Meyermann R, Weller M. Expression and functional activity of heat shock proteins in human glioblastoma multiforme. Neurology. 2000;54(6):1357–1365. doi: 10.1212/wnl.54.6.1357. [DOI] [PubMed] [Google Scholar]
- 57.Soo ET, Yip GW, Lwin ZM, Kumar SD, Bay BH. Heat shock proteins as novel therapeutic targets in cancer. In Vivo. 2008;22(3):311–315. [PubMed] [Google Scholar]
- 58.Rich JN, Bigner DD. Development of novel targeted therapies in the treatment of malignant glioma. Nat Rev Drug Discov. 2004;3(5):430–446. doi: 10.1038/nrd1380. [DOI] [PubMed] [Google Scholar]
- 59.Graner MW, Bigner DD. Chaperone proteins and brain tumors: potential targets and possible therapeutics. Neuro Oncol. 2005;7(3):260–278. doi: 10.1215/S1152851704001188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Udono H, Srivastava PK. Comparison of tumor-specific immunogenicities of stress-induced proteins gp96, hsp90, and hsp70. J Immunol. 1994;152(11):5398–5403. [PubMed] [Google Scholar]
- 61.Basu S, Srivastava PK. Calreticulin, a peptide-binding chaperone of the endoplasmic reticulum, elicits tumor- and peptide-specific immunity. J Exp Med. 1999;189(5):797–802. doi: 10.1084/jem.189.5.797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Srivastava PK, Chen YT, Old LJ. 5′-structural analysis of genes encoding polymorphic antigens of chemically induced tumors. Proc Natl Acad Sci USA. 1987;84(11):3807–3811. doi: 10.1073/pnas.84.11.3807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Blachere NE, Li Z, Chandawarkar RY, et al. Heat shock protein-peptide complexes, reconstituted in vitro, elicit peptide-specific cytotoxic T lymphocyte response and tumor immunity. J Exp Med. 1997;186(8):1315–1322. doi: 10.1084/jem.186.8.1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Blachere NE, Udono H, Janetzki S, Li Z, Heike M, Srivastava PK. Heat shock protein vaccines against cancer. J Immunother Emphasis Tumor Immunol. 1993;14(4):352–356. doi: 10.1097/00002371-199311000-00016. [DOI] [PubMed] [Google Scholar]
- 65.Binder RJ, Han DK, Srivastava PK. CD91: a receptor for heat shock protein gp96. Nat Immunol. 2000;1(2):151–155. doi: 10.1038/77835. [DOI] [PubMed] [Google Scholar]
- 66.Delneste Y, Magistrelli G, Gauchat J, et al. Involvement of LOX-1 in dendritic cell-mediated antigen cross-presentation. Immunity. 2002;17(3):353–362. doi: 10.1016/s1074-7613(02)00388-6. [DOI] [PubMed] [Google Scholar]
- 67.Srivastava PK, Udono H, Blachere NE, Li Z. Heat shock proteins transfer peptides during antigen processing and CTL priming. Immunogenetics. 1994;39(2):93–98. doi: 10.1007/BF00188611. [DOI] [PubMed] [Google Scholar]
- 68.Asea A, Rehli M, Kabingu E, et al. Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem. 2002;277(17):15028–15034. doi: 10.1074/jbc.M200497200. [DOI] [PubMed] [Google Scholar]
- 69.Suto R, Srivastava PK. A mechanism for the specific immunogenicity of heat shock protein-chaperoned peptides. Science. 1995;269(5230):1585–1588. doi: 10.1126/science.7545313. [DOI] [PubMed] [Google Scholar]
- 70.Parsa AT, Aghi M, Ahn B, et al. Autologous heat shock protein vaccine for patients with newly diagnosed and recurrent glioblastoma. Presented at: The 18th International Conference on Brain Tumor Research and Therapy; Travemünde, Germany. 18–20 May 2010. [Google Scholar]
- 71.Belli F, Testori A, Rivoltini L, et al. Vaccination of metastatic melanoma patients with autologous tumor-derived heat shock protein gp96-peptide complexes: clinical and immunologic findings. J Clin Oncol. 2002;20(20):4169–4180. doi: 10.1200/JCO.2002.09.134. [DOI] [PubMed] [Google Scholar]
- 72.Li Z, Qiao Y, Liu B, et al. Combination of imatinib mesylate with autologous leukocyte-derived heat shock protein and chronic myelogenous leukemia. Clin Cancer Res. 2005;11(12):4460–4468. doi: 10.1158/1078-0432.CCR-05-0250. [DOI] [PubMed] [Google Scholar]
- 73.Mazzaferro V, Coppa J, Carrabba MG, et al. Vaccination with autologous tumor-derived heat-shock protein gp96 after liver resection for metastatic colorectal cancer. Clin Cancer Res. 2003;9(9):3235–3245. [PubMed] [Google Scholar]
- 74.Crane CA, Han SJ, Ahn BJ, 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. 2012 doi: 10.1158/1078-0432.CCR-11-3358. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
- 75.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(2):141–157. doi: 10.1023/a:1006293606710. [DOI] [PubMed] [Google Scholar]
- 76.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(9):3294–3302. [PubMed] [Google Scholar]
- 77.Hodi FS, Friedlander PA, Atkins MB, et al. A Phase I trial of ipilimumab plus in patients with unresectable stage III or stage IV melanoma. Presented at: American Society of Clinical Oncology Annual Meeting; Chicago, IL, USA. 3–7 June 2011. [Google Scholar]
- 78.Akbulut H, Tang Y, Akbulut KG, Maynard J, Deisseroth A. Chemotherapy targeted to cancer tissue potentiates antigen-specific immune response induced by vaccine for in vivo antigen loading and activation of dendritic cells. Mol Ther. 2008;16(10):1753–1760. doi: 10.1038/mt.2008.158. [DOI] [PubMed] [Google Scholar]
- 79.Hong M, Puaux AL, Huang C, et al. Chemotherapy induces intratumoral expression of chemokines in cutaneous melanoma, favoring T-cell infiltration and tumor control. Cancer Res. 2011;71(22):6997–7009. doi: 10.1158/0008-5472.CAN-11-1466. [DOI] [PubMed] [Google Scholar]
- 80.Ercolini AM, Ladle BH, Manning EA, et al. Recruitment of latent pools of high-avidity CD8(+) T cells to the antitumor immune response. J Exp Med. 2005;201(10):1591–1602. doi: 10.1084/jem.20042167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Slovin S. Chemotherapy and immunotherapy combination in advanced prostate cancer. Clin Adv Hematol Oncol. 2012;10(2):90–100. [PubMed] [Google Scholar]
- 82.Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364(26):2517–2526. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
- 83.Hersh EM, O’Day SJ, Powderly J, et al. A Phase II multicenter study of ipilimumab with or without dacarbazine in chemotherapy-naive patients with advanced melanoma. Invest New Drugs. 2011;29(3):489–498. doi: 10.1007/s10637-009-9376-8. [DOI] [PubMed] [Google Scholar]
- 84.Rhines LD, Sampath P, DiMeco F, et al. Local immunotherapy with interleukin-2 delivered from biodegradable polymer microspheres combined with interstitial chemotherapy: a novel treatment for experimental malignant glioma. Neurosurgery. 2003;52(4):872–879. doi: 10.1227/01.neu.0000053211.39087.d1. discussion 879–880. [DOI] [PubMed] [Google Scholar]
- 85▪.Sampath P, Hanes J, DiMeco F, et al. Paracrine immunotherapy with interleukin-2 and local chemotherapy is synergistic in the treatment of experimental brain tumors. Cancer Res. 1999;59(9):2107–2114. This study provides support for local delivery of immunotherapy and chemotherapy in a preclinical model. The immunologic effects of local chemotherapy delivery in brain tumors are yet to be evaluated in clinical trials, but may prove advantageous for combination with immunotherapy. [PubMed] [Google Scholar]
- 86.Ransohoff RM, Brown MA. Innate immunity in the central nervous system. J Clin Invest. 2012;122(4):1164–1171. doi: 10.1172/JCI58644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.DiMeco F, Rhines LD, Hanes J, et al. Paracrine delivery of IL-12 against intracranial 9L gliosarcoma in rats. J Neurosurg. 2000;92(3):419–427. doi: 10.3171/jns.2000.92.3.0419. [DOI] [PubMed] [Google Scholar]
- 88.Hanes J, Sills A, Zhao Z, et al. Controlled local delivery of interleukin-2 by biodegradable polymers protects animals from experimental brain tumors and liver tumors. Pharm Res. 2001;18(7):899–906. doi: 10.1023/a:1010963307097. [DOI] [PubMed] [Google Scholar]
- 89.Thompson RC, Pardoll DM, Jaffee EM, et al. Systemic and local paracrine cytokine therapies using transduced tumor cells are synergistic in treating intracranial tumors. J Immunother Emphasis Tumor Immunol. 1996;19(6):405–413. doi: 10.1097/00002371-199611000-00004. [DOI] [PubMed] [Google Scholar]
- 90.Ridolfi R, Chiarion-Sileni V, Guida M, et al. Cisplatin, dacarbazine with or without subcutaneous interleukin-2, and interferon alpha-2b in advanced melanoma outpatients: results from an Italian multicenter Phase III randomized clinical trial. J Clin Oncol. 2002;20(6):1600–1607. doi: 10.1200/JCO.2002.20.6.1600. [DOI] [PubMed] [Google Scholar]
- 91.Atzpodien J, Neuber K, Kamanabrou D, et al. Combination chemotherapy with or without s. c IL-2 and IFN-alpha: results of a prospectively randomized trial of the Cooperative Advanced Malignant Melanoma Chemoimmunotherapy Group (ACIMM) Br J Cancer. 2002;86(2):179–184. doi: 10.1038/sj.bjc.6600043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Dillman RO, Soori G, Wiemann MC, et al. Phase II trial of subcutaneous interleukin-2, subcutaneous interferon-alpha, intravenous combination chemotherapy, and oral tamoxifen in the treatment of metastatic melanoma: final results of cancer biotherapy research group 94–11. Cancer Biother Radiopharm. 2000;15(5):487–494. doi: 10.1089/cbr.2000.15.487. [DOI] [PubMed] [Google Scholar]
- 93.Feau S, Arens R, Togher S, Schoenberger SP. Autocrine IL-2 is required for secondary population expansion of CD8(+) memory T cells. Nat Immunol. 2011;12(9):908–913. doi: 10.1038/ni.2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Masi BC, Tyler BM, Bow H, et al. Intracranial MEMS based temozolomide delivery in a 9L rat gliosarcoma model. Biomaterials. 2012;33(23):5768–5775. doi: 10.1016/j.biomaterials.2012.04.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Scott AW, Tyler BM, Masi BC, et al. Intracranial microcapsule drug delivery device for the treatment of an experimental gliosarcoma model. Biomaterials. 2011;32(10):2532–2539. doi: 10.1016/j.biomaterials.2010.12.020. [DOI] [PubMed] [Google Scholar]
- 96.Attenello FJ, Mukherjee D, Datoo G, et al. Use of gliadel (BCNU) wafer in the surgical treatment of malignant glioma: a 10-year institutional experience. Ann Surg Oncol. 2008;15(10):2887–2893. doi: 10.1245/s10434-008-0048-2. [DOI] [PubMed] [Google Scholar]
- 97.Drake CG. Radiation-induced immune modulation. In: DeWeese TL, editor. Molecular Determinants of Radiation Response. Springer; NY, USA: 2011. pp. 251–263. [Google Scholar]
- 98.Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26(305):234–241. doi: 10.1259/0007-1285-26-305-234. [DOI] [PubMed] [Google Scholar]
- 99.Postow MA, Callahan MK, Barker CA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–931. doi: 10.1056/NEJMoa1112824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100▪▪.Stamell EF, Wolchok JD, Gnjatic S, Lee NY, Brownell I. The abscopal effect associated with a systemic anti-melanoma immune response. Int J Radiat Oncol Biol Phys. 2012 doi: 10.1016/j.ijrobp.2012.03.017. (Epub ahead of print). This case report presents the first evidence in humans that the abscopal effect is immunologically mediated. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Grossman SA, Ye X, Lesser G, et al. Immunosuppression in patients with high-grade gliomas treated with radiation and temozolomide. Clin Cancer Res. 2011;17(16):5473–5480. doi: 10.1158/1078-0432.CCR-11-0774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102▪▪.Predina JD, Kapoor V, Judy BF, et al. Cytoreduction surgery reduces systemic myeloid suppressor cell populations and restores intratumoral immunotherapy effectiveness. J Hematol Oncol. 2012;5(1):34. doi: 10.1186/1756-8722-5-34. The authors report a drop in CD4 count in patients with glioblastoma treated with temozolomide and radiation therapy. These findings represent an important consideration for combination immunotherapy regimens that include radiation therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.See AP, Pradilla G, Yang I, Han S, Parsa AT, Lim M. Heat shock protein-peptide complex in the treatment of glioblastoma. Expert Rev Vaccines. 2011;10(6):721–731. doi: 10.1586/erv.11.49. [DOI] [PubMed] [Google Scholar]
- 104.Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17(13):4550–4557. doi: 10.1158/1078-0432.CCR-11-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Xu Q, Liu G, Yuan X, et al. Antigen-specific T-cell response from dendritic cell vaccination using cancer stem-like cell-associated antigens. Stem Cells. 2009;27(8):1734–1740. doi: 10.1002/stem.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.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(6):733–742. doi: 10.3109/14653249.2012.666633. [DOI] [PubMed] [Google Scholar]
- 107.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 plarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29(3):330–336. doi: 10.1200/JCO.2010.30.7744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Dillman RO, Duma CM, Ellis RA, et al. Intralesional lymphokine-activated killer cells as adjuvant therapy for primary glioblastoma. J Immunother. 2009;32(9):914–919. doi: 10.1097/CJI.0b013e3181b2910f. [DOI] [PubMed] [Google Scholar]
- 109.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(5):398–404. doi: 10.1097/00002371-200409000-00009. [DOI] [PubMed] [Google Scholar]
- 110.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(2):133–140. doi: 10.1007/BF00177476. [DOI] [PubMed] [Google Scholar]
- 111.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(5):840–852. doi: 10.1002/1097-0142(19950901)76:5<840::aid-cncr2820760519>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
- 112.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(3):193–197. doi: 10.1007/BF01533386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.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(2):141–155. doi: 10.1007/BF01053935. [DOI] [PubMed] [Google Scholar]
- 114.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(5):331–341. [PubMed] [Google Scholar]
- 115.Lillehei KO, Mitchell DH, Johnson SD, McCleary EL, Kruse CA. Long-term follow-up of patients with recurrent malignant gliomas treated with adjuvant adoptive immunotherapy. Neurosurgery. 1991;28(1):16–23. doi: 10.1097/00006123-199101000-00003. [DOI] [PubMed] [Google Scholar]
- 116.Barba D, Saris SC, Holder C, Rosenberg SA, Oldfield EH. Intratumoral LAK cell and interleukin-2 therapy of human gliomas. J Neurosurg. 1989;70(2):175–182. doi: 10.3171/jns.1989.70.2.0175. [DOI] [PubMed] [Google Scholar]
- 117.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(4 Pt 2):2101–2104. [PubMed] [Google Scholar]
- 118.Wood GW, Holladay FP, Turner T, Wang YY, Chiga M. 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(2):113–120. doi: 10.1023/a:1006456421177. [DOI] [PubMed] [Google Scholar]
- 119.Plautz GE, Miller DW, Barnett GH, et al. T cell adoptive immunotherapy of newly diagnosed gliomas. Clin Cancer Res. 2000;6(6):2209–2218. [PubMed] [Google Scholar]
- 120.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(6):e9. doi: 10.3171/foc.2000.9.6.10. [DOI] [PubMed] [Google Scholar]
- 121.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(5):536–545. doi: 10.1111/j.1349-7006.1999.tb00781.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Plautz GE, Barnett GH, Miller DW, et al. Systemic T cell adoptive immunotherapy of malignant gliomas. J Neurosurg. 1998;89(1):42–51. doi: 10.3171/jns.1998.89.1.0042. [DOI] [PubMed] [Google Scholar]
- 123.Kruse CA, Cepeda L, Owens B, Johnson SD, Stears J, Lillehei KO. Treatment of recurrent glioma with intracavitary alloreactive cytotoxic T lymphocytes and interleukin-2. Cancer Immunol Immunother. 1997;45(2):77–87. doi: 10.1007/s002620050405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.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(2):179–189. doi: 10.1007/BF00177482. [DOI] [PubMed] [Google Scholar]
- 125.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(4):329–336. doi: 10.1007/BF00195603. [DOI] [PubMed] [Google Scholar]
Websites
- 201.Celldex Therapeutics. www.celldextherapeutics.com.
- 202.US NIH clinical trials registry. www.clinicaltrials.gov.
- 203.EU clinical trials register. www.clinicaltrialsregister.eu.