1,425
Views
7
CrossRef citations to date
0
Altmetric
Review

The progress of immunotherapy for glioblastoma

, &
Pages 2654-2658 | Received 24 Jul 2015, Accepted 06 Aug 2015, Published online: 09 Nov 2015

Abstract

Glioblastoma is the most common primary brain tumor in adults, accounting for about half of all primary brain tumors. Despite multiple therapeutic interventions such as surgical resection, radiotherapy, and systemic chemotherapy, the prognosis for glioblastoma remains poor. Due to the scientific community's enhanced understanding of the CNS immune system and significant achievements in tumor immunotherapy in recent years, immunotherapy has become a promising GBM treatment. In vaccine therapy, a number of clinical trials have achieved encouraging results. In antibody therapy, antibodies are used to target immune checkpoints such as ipilimumab and nivolumab. Bioengineering technology has also lead to a new field of tumor immunotherapy, whereby genetically modified tumor-specific T cells are reintroduced into a patient's body.

Introduction

Glioblastoma (glioblastoma multiforme, GBM) is the most common primary brain tumors in adults, accounting for about half of primary brain tumor.Citation1 Despite surgical resection, radiotherapy, and systemic chemotherapy, the median survival is only 14.6 months, and the 5-year survival rate is less than 10%.Citation2-3 As current glioblastoma treatment standard is not tumor specific and tumor cells are prone to resist radiotherapy and alkylating agent, treatment commonly results in inevitable recurrence and poor prognosis.

The rapid development of tumor immunotherapy in recent years brings out encouraging results in related clinical trials. Targeted vaccine of prostatic acid phosphatase Provenge was approved for the treatment of prostate cancer,Citation4 and the human CTLA-4 monoclonal antibody Ipilimumab approved for the treatment of a progressive melanoma in 2011.Citation5 Showing high efficacy in clinical and preclinical studies, immunotherapy is expected to become a new therapy beyond surgery, radiotherapy, and chemotherapy to treat gliomas.

Basis of Cns Immunotherapy

CNS has traditionally been considered to be immune-privileged organ.Citation6 Recently, however, a variety of immune cells were found in normal CNS. First of all, microglia is considered currently to have the function of the APCs, can migrate to the CNS inflammation area and be activated, then secrete a variety of cytokines and chemokine.Citation7 In addition, mononuclear cells can differentiate into macrophages and dendritic cells (DCs), which exist in perivascular, choroid and meningeal. And T lymphocytes can be transferred to the CNS through the blood-brain barrier after activation in the cervical lymph node.Citation8 All these facts above suggest the existence of immune function in the CNS.

However, gliomas have much immune escape mechanism so that the tumor can escape the immune system destruction: (1) tumor factors: glioma cells can secrete a variety of immunosuppressive cytokines, including TGFβ, PGE2, IL-10 and VEGF; (2) external factors: such as age, taking hormones and radiation and chemotherapy that reduce body immunity; (3) immune cells: the increase of regulatory T cells (Tregs), myeloid-derived suppressor cell (MODC) in the tumor microenvironment will suppress immune system.Citation9 It is because of these reasons that our body can hardly produce normal immune response to glioma cells and that enhancing the targeting of glioma cells of the immune system by various means has thus become an important anti-tumor method.

Tumor Vaccination Therapy

Current immunotherapy can divide into active immunotherapy and passive immunotherapy.Citation10 Active immunotherapy refers to the use of foreign antigens to activate the body's tumor-specific immune system. We can either insert antigen directly into the body to activate DCs thus activating T lymphocyte cells, or insert DCs after they have been sensitized with antigen in vitro. Passive immunotherapy refers to the infusion of exogenous immune substances that surpass directly the tumor into the body, which don't require the activation of body's specific immune system. Passive immunotherapy includes antibody immunotherapy, adoptive immunotherapy, and other immune-modulatory therapy.

EGFRvIII vaccine

As the mutant of epidermal growth factor receptor (EGFR), EGFRvIII is only expressed in cancer cells but not in normal tissue, and it can directly lead to the growth of cancer cell. About 20-25% of patients with glioblastoma suffer from EGFRvIII excessive expression.Citation11 Earlier study of Heimburger et al. suggested that EGFRvIII is a negative prognostic factor for GBM patients.Citation12 Recent studies have shown that EGFRvIII expression does not have a significant relationship with patients' prognosis with the current standard of care.Citation11 The company Celldex in the United States has created the experimental immunotherapeutic vaccine targeting specifically EGFRvIII molecular against tumor called Rindopepimut, which is a kind of complex of polypeptide and immunogenic carrier protein. Clinical trials of Phase I and Phase II show that Rindopepimut can stimulate efficiently anti-tumor immunity in those EGFRvIII overexpression-caused GBM patients, thus prolonging their survival. In the 3 clinical trials of Phase II, the newly diagnosed GBM patients with EGFRvIII overexpression were given an intradermal injection of 500 μg Rindopepimut and 150 μg colony stimulating factor GM-CSF respectively 0, 14, 28 days after concurrent chemoradiotherapy. These patients were injected the same dose of Rindopepimut and GM-CSF on the 21st day in each standard adjuvant chemotherapy cycle until the tumor get further development or the patient cannot tolerate. No serious adverse reactions were found and the survival in this 3 Phase-II clinical trials is consistent, which show that Rindopepimut can significantly increase the median overall survival (OS) and progression-free survival (PFS) of EGFRvIII positive adult patients with GBM, as shown in .Citation13

Table 1. Survival of EGFRvIII positive adult patient with GBM in 3 Phase II Rindopepimut trials

A randomized, double-blind Phase II clinical trial named “ReACT” (2011-2015, NCT01498328) concerning relapsed glioblastoma is being carried out by Celldex in order to clarify the influence of Rindopepimut & Bevacizumab combination on the OS and PFS of the recurrent glioblastoma patient with EGFRvIII overexpression. The preliminary result of the study is satisfactory, and it found that EGFRvIII antibody titer associated with survival. The median OS of patients with lower antibody titer is 10.6 months while it reaches more than 20 months for those with high EGFRvIII antibody titer, that's why EGFRvIII antibody titer is recommended to be checked as a predictive marker in early examination.Citation14 Currently an ongoing Phase III clinical trial called “ACT IV Study” (2011-2016, NCT01480479) for newly diagnosed glioblastoma may make Rindopepimut be approved by FDA as the first kind of immunotherapeutic drug for primary brain tumors if the study gets positive result.

DCs vaccines

As the most important antigen-presenting-specific cells, DCs can start body's immune response by expressing high level of the MHC molecules and stimulating molecules such as B7-1 (CD80), B7-2 (CD86), CIM0. The ability to absorb all kinds of antigen and to secrete IL-2 enables DCs to activate naive T cells and initiate immune response. By mediating effectively the anti-tumor immune response of CD8+ and CD4+ T lymphocyte, DCs play a key role in the induction of an efficient and specific T cells immune response against the related antigens. Researchers cultured DCs and sensitized them with various types of tumor antigens both in vitro, and then injected them into the body to induce antigen-specific immune responses. The antigens used to sensitize DCs are mainly from surgically resected tumor cell lysates as well as the specific synthetic polypeptide, other special antigens include tumor stem cell antigen and cytomegalovirus antigen. Being currently hot in tumor immunotherapy research, DCs vaccine Phase I, II even III clinical trials are underway both at home and abroad, as shown in .Citation15

Table 2. Main DCs-based Phase II, III clinical trials

ICT-107 is the autologous peripheral blood mononuclear cells (PBMC) sourced DCs which were sensitized by 6 kinds of synthetic peptide (tumor stem cell antigen MAGE-1, her-2, AIM-2, TRP-2, gp100, and IL-13Rα2). A Phase I clinical trial of ICT-107 on 16 newly diagnosed GBM patients resulted in 16.9 months as median PFS and 38.4 months as median OS.Citation16 Professor Patrick Y. Wen has released some preliminary test results (Abstract No. 2005) of a Phase II clinical trial at the 2014 ASCO meeting. Without significant difference in side effects between the treatment group and the control group, the result suggests that ICT-107 is rather safe and well tolerated. Median PFS has increased by 3 months (bilateral test p = 0.01, HR = 0.53), and OS has also increased by 3 months (bilateral test p = 0.40, HR = 0.79). That is the first randomized, controlled trial of immunotherapy for GBM treatment with positive results (Increase in PFS has statistical significance).

At the moment, another Phase III multi-center, randomized, controlled clinical research aiming to observe DCVax's therapeutic effect for newly diagnosed GBM is being carried out by the company Northwest Biotherapeutics. 300 adult patients with newly diagnosed GBM are planned to be recruited for this research and to be randomly divided into 2 groups after standard of care, including surgery, concurrent chemoradiation and Temodar therapy, 2 out of 3 will additionally receive DCVax with the remaining one-third receiving a placebo. The primary endpoint of the study was PFS. (ClinicalTrials.gov identifier: NCT00045968).

Heat-shock protein-based vaccines

Using DCs as the vaccine carrier demands the culture of DCs in vitro and their sensitization with tumor antigen, which requires very specialized cell culture techniques. While the application of tumor-derived heat shock proteins (HSPs) as tumor antigen carrier can make use of DCs' antigen-presenting function without requiring the culture of DCs in vitro.Citation16 Belonging to a protein family whose main physiological function is to promote the mechanism of peptide folding by interacting with polypeptides, HSPs express at a higher level in case of fever, infection, hypoxia, cancer and other conditions. In addition, HSPs can be combined with the DCs and transfer the bound polypeptide antigens into them, thus activating CD4+ and CD8+ T cell response in tumor immunotherapy.

Bloch et al. has reported an open-label, single-arm, Phase II clinical trial of using autologous tumor-derived heat-shock protein peptide complex-96 (HPSC-96) vaccine in the treatment of recurrent GBM.Citation17 Between 3rd Oct. 2007 and 24th Oct. 2011, there are 41 patients with recurrent GBM that received an average of 6 doses of HPSC-96 vaccine intradermal injection, among which only one has encountered 3-degree adverse reaction related to vaccine. The 6-month survival rate of the patients is 90.2% (95% CI: 75.9-96.8), 12-month survival rate is 29.3% (95% CI: 16.6-45.7) and the median OS is 42.6 weeks (95% CI: 34.7-50.5).

In July 2014 Agenus Inc.. published a randomized, single-arm, Phase II clinical trial of using HPSC-96 vaccine in the treatment of newly diagnosed GBM patients. The addition of HPSC-96 vaccine to the standard of care in this research resulted in an increase of 9.6 months in median OS and 26% in 2-year survival rate. By contrast, the median survival was 14.6 months and 2-year survival rate was 26% in the case of standard treatment.Citation2-3 It was also found in this study that patients with low-expression of PD-L1 protein react significantly to HPSC-96 vaccine, indicating that the joint use of vaccine and other Anti-PD-L1 immunotherapeutic drug may improve the anti-tumor efficiency in the future.

Antibody-Based Immunotherapy

Antibody-based immunotherapy activates the immune system to eliminate tumor cells through the specific interaction between antibodies and antigens. Some suitable immunotherapy targets for glioma have been identified with the clarification of the pathological mechanisms of glioma, especially the mechanism of immunosuppression. Additionally, advances in bioengineering make the preparation of monoclonal antibodies increasingly simple. Current antibody-based immunotherapy mainly consists of the redirection of immune effector cells to interact with tumor mutations by means of bispecific monoclonal antibody-redirected, the antibody drug targeting negative immune regulatory molecules and the activation of anti-tumor immune by providing costimulatory signals,Citation18 among which the most promising is the immune checkpoint blockade. The immune checkpoint refers to some inhibitory regulatory molecules in the immune system that make tumor escape immune destruction by inhibiting T cell activity. Ipilimumab, the antibody of Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), is the first immune checkpoint-targeted drug approved by FDA. Other immune checkpoints, such as programmed death protein-1 (PD-1) and its ligands have also been proven to be able to induce the remission of a wide variety of tumors. Targeting immune checkpoints has a broad application prospect in anti-tumor immunotherapy.

Anti-PD1/PD-L1 therapies

Programmed cell death factor-1 (PD-1) is a type I transmembrane glycoprotein belonging to the immunoglobulin superfamily, which exists on the cell surface in the form of a monomer. It can, after binding to the ligand (PD-L1), phosphorylate the downstream molecules, transduce negative signals, inhibit T cell proliferation and cytokine production and induce T cell apoptosis. Some preclinical trials on animal tumor model have demonstrated that tumor microenvironment can promote tumor's expression of PD-L1 which induces the apoptosis of T lymphocytes, while PD-1/PD-L1 antibody can save T cells by blocking PD-1 /PD-L1 pathway thus enhancing the anti-tumor immunity.Citation19 Recent clinical trials of anti-PD-1, such as MDX-1106, show its good tolerance and anti-tumor activity for the patients with solid tumors.Citation20 In Sep. 2014, the anti-PD-1 drugs Keytruda was approved to treat advanced melanoma. As to the clinical applications in GBM treatment, a related study was reported at the ASCO 2014 annual meeting. A randomized, open-label, Phase - clinical trial (Abstract number: TPS2101) is about to begin for the treatment of recurrent glioblastoma with monoclonal antibody of PD-1, aiming to evaluate the efficacy and safety of treating recurrent glioblastoma with nivolumab (Human PD-1 monoclonal antibody) alone or together with Ipilimumab.

Anti-CTLA-4 therapy

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), also known as CD152, is a kind of leukocyte differentiation antigen. As a transmembrane receptor on T cells, CTLA-4 can inhibit the immune reaction of effector T cells by competing with CD28 on the same cell for the B7 family immune molecules on the surface of APC. Pre-clinical trials demonstrated that CTLA-4 can inhibit tumor immunity by activating Tregs and that CTLA-4 antibodies inhibit tumor growth. Being a human-sourced monoclonal CTLA-4 antibody, Ipilimumab can promote the activation and proliferation of T cells by blocking the interaction of CTLA-4 with its ligand CD28 after binding to it, thereby promoting the anti-tumor effect. Despite the fact that Ipilimumab has been proved to be able to improve significantly the prognosis in melanoma treatment and have good anti-tumor effect in animal glioma models, its effect in the treatment of GBM has not been verified.Citation21-22 Two studies on treating recurrent GBM with Ipilimumab (Abstract number e13026 and e13010), reported at the ASCO 2014 annual meeting, resulted in significant curative effect on some patients and none serious side effect. Clinical trials on the treatment of GBM with Ipilimumab will begin shortly.

Adoptive Immunotherapy

Adoptive immunotherapy refers to the infusion of activated immune effector cell into tumor patients so as to make use of its anti-tumor immunity directly. Immune effector cells include lymphokine-activated killer (LAK) cells, natural killer (NK) cells, T cells, tumor-infiltrating lymphocytes (TILs), cytotoxic T lymphocyte (CTLs), tumor antigen-specific TCR-transgenic T cells and chimeric antigen receptors-modified T cells (CART). The first 3 kinds of cells are tumor antigen-specific and may attack normal cells with high nerve toxicity. TILs are tumor-specific, but they need to be extracted from the tumor tissues, and their proliferation is difficult. Currently, the most used in adoptive immunotherapy are CTLs, including CD4+ and CD8+ T lymphocytes. TCR transgenic T cells, as well as CART, are still mainly at the laboratory stage. By such means of genetically modifying T cells and prolonging their survival time, the effect of specific T cells could be enhanced.Citation23-24 Additionally, the reinfusion of the T cells proliferated in vitro back into the body can correct lymphopenia and improve immunosuppressive environment,Citation25-26 thereby enhancing tumor immunity.

Internationally, TCR transgenic T cells and CART are currently hot spots for adoptive immunotherapy. In Feb. 2014, researchers from US Sloan-Kettering Cancer Institute,Citation27 reported on “Science Translational Medicine” that they first removed T cells from the patient's body, and then transformed them with genetic engineering by making them be able to identify CD19 proteins contained in leukemia cells. The cells amplified in vitro can help to destroy leukemia cells in vivo after their reinfusion back into the body. In this experiment, a total of 16 relapsed or refractory B-cell acute lymphoblastic leukemia patients were treated, resulting in complete remission of 14 people. In the trials of animal glioblastomas, EGFRvIII-specific CART also showed good therapeutic effect.Citation28-29 At present, a number of clinical trials concerning the adoptive immunotherapy of central nervous system tumors are underway, as seen in .Citation30

Table 3. Clinical trials of adoptive immunotherapy of CNS tumor

Outlook

Despite surgical resection, radiotherapy, and systemic chemotherapy, the prognosis for GBM remains poor. Therefore it's urgent to find a new alternative or adjuvant therapy. With the re-understanding of the CNS immune system and the significant achievements in the tumor immunotherapy in recent years, immunotherapy has become a kind of GBM treatment with big potential. Specificity of immunotherapy and memory characteristics of immune cells may have great significance in inhibiting tumor recurrence. In the vaccine therapy, a number of Phase II clinical trials have achieved encouraging results. Particularly, 2 Phase III clinical trials of the vaccines DCVax-Brain and Rindopepimut are very expected. With respect to antibody therapy, Ipilimumab has been proven to be able to improve significantly the survival and has been authorized for clinical use. The antibodies of CTLA-4 and PD-1 /PD-L1, such as Ipilimumab, nivolumab, etc. are being tested in clinical trials for their application in the treatment of GBM. By means of bioengineering technology, the reinfusion of genetically modified tumor-specific T cells back into the body has a unique advantage comparing with active immunotherapy, and the reinfusion of adoptive T cell has created another new field in tumor immunotherapy. There is no doubt that, with the development of the immunotherapy, new breakthroughs will take place in glioblastoma treatment shortly.

Disclosure of Potential Conflicts of Interest

None of the authors has any conflicts of interest to disclose.

References

  • Chakrabarti I, Cockburn M, Cozen W, Wang YP, Preston-Martin S. A population-based description of glioblastoma multiforme in Los Angeles County, 1+74-1999. Cancer 2005; 104(12):2798-806; PMID:16288487; http://dx.doi.org/10.1002/cncr.21539
  • Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New Engl J Med 2005; 352(10):987-96; PMID:15758009; http://dx.doi.org/10.1056/NEJMoa043330
  • Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009; 10(5):459-66; PMID:19269895; http://dx.doi.org/10.1016/S1470-2045(09)70025-7
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New Engl J Med 2010; 363(5):411-22; PMID:20818862; http://dx.doi.org/10.1056/NEJMoa1001294
  • Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. New Engl J Med. 2010; 363(8):711-23; PMID:20525992; http://dx.doi.org/10.1056/NEJMoa1003466
  • Sehgal A, Berger MS. Basic concepts of immunology and neuroimmunology. Neurosurgical FOCUS 2000; 9(6):1-6; http://dx.doi.org/10.3171/foc.2000.9.6.2
  • Tambuyzer BR, Ponsaerts P, Nouwen EJ. Microglia: gatekeepers of central nervous system immunology. J Leukocyte Biol 2008;85(3):352-70; PMID:19028958; http://dx.doi.org/10.1189/jlb.0608385
  • Hickey WF, Hsu BL, Kimura H. T-lymphocyte entry into the central nervous system. J Neurosci Res 1991; 28(2):254-60; PMID:2033653; http://dx.doi.org/10.1002/jnr.490280213
  • Fecci PE. Increased regulatory T-Cell fraction amidst a diminished cd4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res 2006; 66(6):3294-302; PMID:16540683; http://dx.doi.org/10.1158/0008-5472.CAN-05-3773
  • Vauleon E, Avril T, Collet B, Mosser J, Quillien V. Overview of cellular immunotherapy for patients with glioblastoma. Clin Dev Immunol 2010; 2010:1-18; PMID:20953324; http://dx.doi.org/10.1155/2010/689171
  • Weller M, Kaulich K, Hentschel B, Felsberg J, Gramatzki D, Pietsch T, Simon M, Westphal M, Schackert G, Tonn JC, et al. Assessment and prognostic significance of the epidermal growth factor receptor vIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy. Int J Cancer 2014; 134(10):2437-47; PMID:24614983; http://dx.doi.org/10.1002/ijc.28576
  • Heimberger AB, Hlatky R, Suki D, Yang D, Weinberg J, Gilbert M, Sawaya R, Aldape K. Prognostic effect of epidermal growth factor receptor and EGFRvIII in glioblastoma multiforme patients. Clin Cancer Res 2005; 11(4):1462-6; PMID:15746047; http://dx.doi.org/10.1158/1078-0432.CCR-04-1737
  • Babu R, Adamson DC. Rindopepimut: an evidence-based review of its therapeutic potential in the treatment of EGFRvIII-positive glioblastoma. Core Evid 2012; 7:93-103; PMID:23055947
  • Dixit S. Immunotherapy for high-grade glioma. Future Oncol 2014; 10(6):911-5; PMID:24941977; http://dx.doi.org/10.2217/fon.14.20
  • Xu LW, Chow KKH, Lim M, Li G. Current vaccine trials in glioblastoma: A review. J Immunol Res 2014; 2014:1-10; PMID:24804271; http://dx.doi.org/10.1155/2014/796856
  • McNulty S, Colaco CA, Blandford LE, Bailey CR, Baschieri S, Todryk S. Heat-shock proteins as dendritic cell-targeting vaccines–getting warmer. Immunology 2013; 139(4):407-15; PMID:23551234; http://dx.doi.org/10.1111/imm.12104
  • Bloch O, Crane CA, Fuks Y, Kaur R, Aghi MK, Berger MS, Butowski NA, Chang SM, Clarke JL, McDermott MW, et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro Oncol 2014; 16(2):274-9; PMID:24335700; http://dx.doi.org/10.1093/neuonc/not203
  • Gedeon PC, Riccione KA, Fecci PE, Sampson JH. Antibody-based immunotherapy for malignant glioma. Semin Oncol 2014; 41(4):496-510; PMID:25173142; http://dx.doi.org/10.1053/j.seminoncol.2014.06.004
  • Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 2012; 24(2):207-12; PMID:22236695; http://dx.doi.org/10.1016/j.coi.2011.12.009
  • Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010; 28(19):3167-75; PMID:20516446; http://dx.doi.org/10.1200/JCO.2009.26.7609
  • Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, et al. Anti-PD-1 Blockade and Stereotactic Radiation Produce Long-Term Survival in Mice With Intracranial Gliomas. Int J Radiat Oncol 2013; 86(2):343-9; PMID:23462419; http://dx.doi.org/10.1016/j.ijrobp.2012.12.025
  • Prieto PA, Yang JC, Sherry RM, Hughes MS, Kammula US, White DE, Levy CL, Rosenberg SA, Phan GQ. CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clin Cancer Res 2012; 18(7):2039-47; PMID:22271879; http://dx.doi.org/10.1158/1078-0432.CCR-11-1823
  • Peng W, Ye Y, Rabinovich BA, Liu C, Lou Y, Zhang M, Whittington M, Yang Y, Overwijk WW, Lizée G, et al. Transduction of tumor-specific T cells with CXCR2 chemokine receptor improves migration to tumor and antitumor immune responses. Clin Cancer Res 2010; 16(22):5458-68; PMID:20889916; http://dx.doi.org/10.1158/1078-0432.CCR-10-0712
  • Charo J, Finkelstein SE, Grewal N, Restifo NP, Robbins PF, Rosenberg SA. Bcl-2 overexpression enhances tumor-specific T-cell survival. Cancer Res 2005; 65(5):2001-8; PMID:15753400; http://dx.doi.org/10.1158/0008-5472.CAN-04-2006
  • Chung DS, Shin HJ, Hong YK. A new hope in immunotherapy for malignant gliomas: adoptive T cell transfer therapy. J Immunol Res 2014; 2014:326545; PMID:25009822; http://dx.doi.org/10.1155/2014/326545
  • Crough T, Beagley L, Smith C, Jones L, Walker DG, Khanna R. Ex vivo functional analysis, expansion and adoptive transfer of cytomegalovirus-specific T-cells in patients with glioblastoma multiforme. Immunol Cell Biol 2012; 90(9):872-80; PMID:22508289; http://dx.doi.org/10.1038/icb.2012.19
  • Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-Ojeda O, Olszewska M, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014; 6(224):224r-5r; PMID:24553386; http://dx.doi.org/10.1126/scitranslmed.3008226
  • Sampson JH, Choi BD, Sanchez-Perez L, Suryadevara CM, Snyder DJ, Flores CT, Schmittling RJ, Nair SK, Reap EA, Norberg PK, et al. EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clin Cancer Res 2014; 20(4):972-84; PMID:24352643; http://dx.doi.org/10.1158/1078-0432.CCR-13-0709
  • Choi BD, Suryadevara CM, Gedeon PC, Herndon JE 2nd, Sanchez-Perez L, Bigner DD, Sampson JH. Intracerebral delivery of a third generation EGFRvIII-specific chimeric antigen receptor is efficacious against human glioma. J Clin Neurosci 2014; 21(1):189-90; PMID:24054399; http://dx.doi.org/10.1016/j.jocn.2013.03.012
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: adoptive cell transfer immunotherapy. OncoImmunology 2012; 1(3):306-15; PMID:22737606; http://dx.doi.org/10.4161/onci.19549

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.