1
|
Hou AJ, Shih RM, Uy BR, Shafer A, Chang ZL, Comin-Anduix B, Guemes M, Galic Z, Phyu S, Okada H, Grausam KB, Breunig JJ, Brown CE, Nathanson DA, Prins RM, Chen YY. IL-13Rα2/TGF-β bispecific CAR-T cells counter TGF-β-mediated immune suppression and potentiate anti-tumor responses in glioblastoma. Neuro Oncol 2024; 26:1850-1866. [PMID: 38982561 PMCID: PMC11449012 DOI: 10.1093/neuonc/noae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapies targeting glioblastoma (GBM)-associated antigens such as interleukin-13 receptor subunit alpha-2 (IL-13Rα2) have achieved limited clinical efficacy to date, in part due to an immunosuppressive tumor microenvironment (TME) characterized by inhibitory molecules such as transforming growth factor-beta (TGF-β). The aim of this study was to engineer more potent GBM-targeting CAR-T cells by countering TGF-β-mediated immune suppression in the TME. METHODS We engineered a single-chain, bispecific CAR targeting IL-13Rα2 and TGF-β, which programs tumor-specific T cells to convert TGF-β from an immunosuppressant to an immunostimulant. Bispecific IL-13Rα2/TGF-β CAR-T cells were evaluated for efficacy and safety against both patient-derived GBM xenografts and syngeneic models of murine glioma. RESULTS Treatment with IL-13Rα2/TGF-β CAR-T cells leads to greater T-cell infiltration and reduced suppressive myeloid cell presence in the tumor-bearing brain compared to treatment with conventional IL-13Rα2 CAR-T cells, resulting in improved survival in both patient-derived GBM xenografts and syngeneic models of murine glioma. CONCLUSIONS Our findings demonstrate that by reprogramming tumor-specific T-cell responses to TGF-β, bispecific IL-13Rα2/TGF-β CAR-T cells resist and remodel the immunosuppressive TME to drive potent anti-tumor responses in GBM.
Collapse
Affiliation(s)
- Andrew J Hou
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California, USA
| | - Ryan M Shih
- Department of Molecular Biology, University of California, Los Angeles, California, USA
| | - Benjamin R Uy
- Department of Neurosurgery, University of California, Los Angeles, California, USA
| | - Amanda Shafer
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - ZeNan L Chang
- Department of Molecular Biology, University of California, Los Angeles, California, USA
| | - Begonya Comin-Anduix
- Department of Surgery, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Miriam Guemes
- Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles, California, USA
| | - Zoran Galic
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles, California, USA
| | - Su Phyu
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, California, USA
- Parker Institute for Cancer Immunotherapy Center at UCSF, San Francisco, California, USA
| | - Katie B Grausam
- Board of Governor’s Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joshua J Breunig
- Board of Governor’s Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, California, USA
| | - David A Nathanson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| | - Robert M Prins
- Department of Neurosurgery, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, California, USA
| |
Collapse
|
2
|
Qian D, Liu Y, Zheng J, Cai J. Dendritic cell therapy for neurospoagioma: Immunomodulation mediated by tumor vaccine. Cell Death Discov 2024; 10:11. [PMID: 38184649 PMCID: PMC10771477 DOI: 10.1038/s41420-023-01782-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 01/08/2024] Open
Abstract
Neurospagioma, arising from different glial cells such as astrocytes, oligodendrocytes, and ependymal cells, stands as the prevalent intracranial tumor within the central nervous system. Among its variants, glioblastoma (GBM) represents the most aggressive form, characterized by a notably high occurrence rate and a discouragingly low survival prognosis. The formidable challenge posed by glioblastoma underscores its critical importance as a life-threatening ailment. Currently, clinical approaches often involve surgical excision along with a combination of radiotherapy and chemotherapy. However, these treatments frequently result in a notable recurrence rate, accompanied by substantial adverse effects that significantly compromise the overall prognosis. Hence, there is a crucial need to investigate novel and dependable treatment strategies. Dendritic cells (DCs), being specialized antigen-presenting cells (APCs), hold a significant position in both innate and adaptive immune responses. Presently, DC vaccines have gained widespread application in the treatment of various tumors, including neurospoagioma. In this review, we summarize the immunomodulatory effects and related mechanisms of DC vaccines in neurospoagioma as well as the progress of clinical trials to propose possible challenges of DC vaccines and new development directions.
Collapse
Affiliation(s)
- Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, 215500, Jiangsu Province, China.
| | - Yuxiang Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 150086, Harbin, China
| | - Jie Zheng
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, 215500, Jiangsu Province, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 150086, Harbin, China.
| |
Collapse
|
3
|
Li X, Xiao X, Wang Y, Gu G, Li T, Wang Y, Li C, Zhang P, Ji N, Zhang Y, Zhang L. Expression of Interleukin-13 Receptor Alpha 2 in Brainstem Gliomas. Cancers (Basel) 2024; 16:228. [PMID: 38201655 PMCID: PMC10777982 DOI: 10.3390/cancers16010228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
The objective of this study was to investigate IL13Ra2 expression in brainstem glioma (BSG) and its correlation with key markers, functions, and prognostic implications, evaluating its therapeutic potential. A total of 80 tumor samples from BSG patients were analyzed. Multiplex immunofluorescence was used to examine six markers-IL13Ra2, H3.3K27M, CD133, Ki67, HLA-1, and CD4-establishing relationships between IL13Ra2 and these markers. Survival analysis, employing Kaplan-Meier and Cox proportional hazard regression models, encompassed 66 patients with complete follow-up. RNA-Seq data from a previously published study involving 98 patients were analyzed using the DESeq2 library to determine differential gene expression between groups. Gene Ontology (GO) enrichment and single-sample gene set enrichment analysis (ssGSEA) via the clusterProfiler library were used to delineate the gene functions of differentially expressed genes (DEGs). Nearly all the BSG patients displayed varying IL13Ra2 expression, with 45.0% (36/80) exhibiting over a 20% increase. Elevated IL13Ra2 levels were notably observed in pontine gliomas, diffuse intrinsic pontine gliomas (DIPGs), H3F3A-mutant gliomas, and WHO IV gliomas. IL13Ra2 expression was strongly correlated with H3.3K27M mutant protein, Ki67, and CD133. Patients with IL13Ra2 expression >20% showed shorter overall survival compared to those with ≤20% IL13Ra2 expression. The Cox proportional hazard regression model identified H3F3A mutations, rather than IL13Ra2 expression, as an independent prognostic factor. Analysis of RNA-Seq data from our prior cohort confirmed IL13Ra2's correlation with H3.3, CD133, and Ki67 levels. Widespread IL13Ra2 expression in BSG, particularly elevated in the H3F3A mutant group, was strongly correlated with H3F3A mutations, increased proliferation, and heightened tumor stemness. IL13Ra2 represents a promising therapeutic target for BSGs, potentially benefiting patients with H3K27M mutations, DIPGs, WHO Grade IV, and pontine location-specific BSGs, particularly those with H3K27M mutations.
Collapse
Affiliation(s)
- Xiaoou Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xiong Xiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Tian Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Chunzhao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yang Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.L.); (X.X.); (Y.W.); (G.G.); (T.L.); (Y.W.); (C.L.); (P.Z.); (N.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
4
|
Van Gool SW, Van de Vliet P, Kampers LFC, Kosmal J, Sprenger T, Reich E, Schirrmacher V, Stuecker W. Methods behind oncolytic virus-based DC vaccines in cancer: Toward a multiphase combined treatment strategy for Glioblastoma (GBM) patients. Methods Cell Biol 2023; 183:51-113. [PMID: 38548421 DOI: 10.1016/bs.mcb.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Glioblastoma (GBM) remains an orphan cancer disease with poor outcome. Novel treatment strategies are needed. Immunotherapy has several modes of action. The addition of active specific immunotherapy with dendritic cell vaccines resulted in improved overall survival of patients. Integration of DC vaccination within the first-line combined treatment became a challenge, and immunogenic cell death immunotherapy during chemotherapy was introduced. We used a retrospective analysis using real world data to evaluate the complex combined treatment, which included individualized multimodal immunotherapy during and after standard of care, and which required adaptations during treatment, and found a further improvement of overall survival. We also discuss the use of real world data as evidence. Novel strategies to move the field of individualized multimodal immunotherapy forward for GBM patients are reviewed.
Collapse
Affiliation(s)
| | | | | | | | | | - Ella Reich
- Immun-onkologisches Zentrum Köln, Cologne, Germany
| | | | | |
Collapse
|
5
|
Hotchkiss KM, Batich KA, Mohan A, Rahman R, Piantadosi S, Khasraw M. Dendritic cell vaccine trials in gliomas: Untangling the lines. Neuro Oncol 2023; 25:1752-1762. [PMID: 37289203 PMCID: PMC10547519 DOI: 10.1093/neuonc/noad088] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Glioblastoma is a deadly brain tumor without any significantly successful treatments to date. Tumor antigen-targeted immunotherapy platforms including peptide and dendritic cell (DC) vaccines, have extended survival in hematologic malignancies. The relatively "cold" tumor immune microenvironment and heterogenous nature of glioblastoma have proven to be major limitations to translational application and efficacy of DC vaccines. Furthermore, many DC vaccine trials in glioblastoma are difficult to interpret due to a lack of contemporaneous controls, absence of any control comparison, or inconsistent patient populations. Here we review glioblastoma immunobiology aspects that are relevant to DC vaccines, review the clinical experience with DC vaccines targeting glioblastoma, discuss challenges in clinical trial design, and summarize conclusions and directions for future research for the development of effective DC vaccines for patients.
Collapse
Affiliation(s)
- Kelly M Hotchkiss
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina, USA
| | - Kristen A Batich
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina, USA
| | - Aditya Mohan
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina, USA
| | - Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven Piantadosi
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA(S.P.)
| | - Mustafa Khasraw
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
6
|
Zheng Y, Ma X, Feng S, Zhu H, Chen X, Yu X, Shu K, Zhang S. Dendritic cell vaccine of gliomas: challenges from bench to bed. Front Immunol 2023; 14:1259562. [PMID: 37781367 PMCID: PMC10536174 DOI: 10.3389/fimmu.2023.1259562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Gliomas account for the majority of brain malignant tumors. As the most malignant subtype of glioma, glioblastoma (GBM) is barely effectively treated by traditional therapies (surgery combined with radiochemotherapy), resulting in poor prognosis. Meanwhile, due to its "cold tumor" phenotype, GBM fails to respond to multiple immunotherapies. As its capacity to prime T cell response, dendritic cells (DCs) are essential to anti-tumor immunity. In recent years, as a therapeutic method, dendritic cell vaccine (DCV) has been immensely developed. However, there have long been obstacles that limit the use of DCV yet to be tackled. As is shown in the following review, the role of DCs in anti-tumor immunity and the inhibitory effects of tumor microenvironment (TME) on DCs are described, the previous clinical trials of DCV in the treatment of GBM are summarized, and the challenges and possible development directions of DCV are analyzed.
Collapse
Affiliation(s)
- Ye Zheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Ma
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shouchang Feng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjiang Yu
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Xiong Q, Zhu J, Zhang Y, Deng H. CAR-NK cell therapy for glioblastoma: what to do next? Front Oncol 2023; 13:1192128. [PMID: 37404752 PMCID: PMC10315652 DOI: 10.3389/fonc.2023.1192128] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/29/2023] [Indexed: 07/06/2023] Open
Abstract
Glioblastoma is a malignant tumor with the highest morbidity and mortality in the central nervous system. Conventional surgical resection combined with radiotherapy or chemotherapy has a high recurrence rate and poor prognosis. The 5-year survival rate of patients is less than 10%. In tumor immunotherapy, CAR-T cell therapy represented by chimeric antigen receptor-modified T cells has achieved great success in hematological tumors. However, the application of CAR-T cells in solid tumors such as glioblastoma still faces many challenges. CAR-NK cells are another potential adoptive cell therapy strategy after CAR-T cells. Compared with CAR-T cell therapy, CAR-NK cells have similar anti-tumor effects. CAR-NK cells can also avoid some deficiencies in CAR-T cell therapy, a research hotspot in tumor immunity. This article summarizes the preclinical research status of CAR-NK cells in glioblastoma and the problems and challenges faced by CAR-NK in glioblastoma.
Collapse
|
8
|
Zhang Y, Su J. Interleukin-2 family cytokines: An overview of genes, expression, signaling and functional roles in teleost. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 141:104645. [PMID: 36696924 DOI: 10.1016/j.dci.2023.104645] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 06/17/2023]
Abstract
The interleukin-2 (IL-2) family cytokines include IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, which share γ chain (γc) subunit in receptors. The IL-2 family cytokines have unique biological effects that regulate differentiation, survival and activation of multiple lymphocyte lineages. Deficiency of IL-2 family signaling pathway in mammals prevents CD4+ T cells from developing effector functions and CD8+ T cells from developing immunological memory. In the present review, we addressed available information from teleost IL-2 family cytokines and discussed implications in teleost immunity. Also, we described and discussed their expression profiles, receptors, signaling transductions and functions. In teleost, IL-2 family has 5 members (IL-2, IL-4/13, IL-7, IL-15, IL-21) without IL-9, and their receptors share a common γc subunit and include other 6 subunits (IL-2Rβ1/2, IL-4Rα1/2, IL-13Rα1/2, IL-7Rα, IL-15Rα, and IL-21Rα1/2). Some paralogues have changes in domain structure and show differential expression, modulation, functions. IL-2 family cytokines constitutively express in many immune associated tissues and are largely induced after pathogenic microbial stimulation. In general, there are relatively conserved functions in the IL-2 family throughout vertebrates, and many of the key IL-2 family members are important in lymphocyte proliferation and differentiation, development, inflammation from fishes to mammals. This review will give an update on the effective information of teleost IL-2 family cytokines. Thus, it will provide a source of reference for other researchers/readers and inspire further interest.
Collapse
Affiliation(s)
- Yanqi Zhang
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Jianguo Su
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
9
|
Gu A, Bai Y, Zhang C, Xu C, An Z, Zhang Y, Zhong SH, Hu Y, Zhong X. IL13Rα2-targeted third-generation CAR-T cells with CD28 transmembrane domain mediate the best anti-glioblastoma efficacy. Cancer Immunol Immunother 2023:10.1007/s00262-023-03423-5. [PMID: 36991262 DOI: 10.1007/s00262-023-03423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/12/2023] [Indexed: 03/30/2023]
Abstract
Chimeric antigen receptor (CAR)-modified T (CAR-T) cell therapy has been proven to be a powerful tool for the treatment of cancer, however, the limits are obvious, especially for solid tumors. Therefore, constantly optimizing the structure of CAR to improve its therapeutic effect is necessary. In this study, we generated three different third-generation CARs targeting IL13Rα2, with the same scFv, but different transmembrane domains (TMDs) from CD4, CD8 or CD28 (IL13-CD4TM-28.BB.ζ, IL13-CD8TM-28.BB.ζ and IL13-CD28TM-28.BB.ζ). CARs were transduced into primary T cells using retroviruses. The anti-GBM efficacy of CAR-T cells was monitored by flow cytometry and real-time cell analysis (RTCA) in vitro and examined in two xenograft mouse models. The differentially expressed genes related to different anti-GBM activity were screened by high throughput RNA sequencing. We observed that T cells transduced with these three CARs have similar anti-tumor activity when co-cultured with U373 cells which expressed higher IL13Rα2 but exhibited different anti-tumor activity when co-cultured with U251 cells that expressed lower IL13Rα2. All the three groups of CAR-T cells can be activated by U373 cells, but only IL13-CD28TM-28.BB.ζ CAR-T cells could be activated and expressed increased IFN-γ after co-culturing with U251 cells. IL13-CD28TM-28.BB.ζ CAR-T cells exhibited the best anti-tumor activity in xenograft mouse models which can infiltrate into the tumors. The superior anti-tumor efficacy of IL13-CD28TM-28.BB.ζ CAR-T cells was partially owing to differentially expressed extracellular assembly, extracellular matrix, cell migration and adhesion-related genes which contribute to the lower activation threshold, increased cell proliferation, and elevated migration capacity.
Collapse
|
10
|
Mantica M, Drappatz J. Immunotherapy associated central nervous system complications in primary brain tumors. Front Oncol 2023; 13:1124198. [PMID: 36874119 PMCID: PMC9981156 DOI: 10.3389/fonc.2023.1124198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
Advances clarifying the genetics and function of the immune system within the central nervous system (CNS) and brain tumor microenvironment have led to increasing momentum and number of clinical trials using immunotherapy for primary brain tumors. While neurological complications of immunotherapy in extra-cranial malignancies is well described, the CNS toxicities of immunotherapy in patients with primary brain tumors with their own unique physiology and challenges are burgeoning. This review highlights the emerging and unique CNS complications associated with immunotherapy including checkpoint inhibitors, oncolytic viruses, adoptive cell transfer/chimeric antigen receptor (CAR) T cell and vaccines for primary brain tumors, as well as reviews modalities that have been currently employed or are undergoing investigation for treatment of such toxicities.
Collapse
Affiliation(s)
- Megan Mantica
- Department of Neurology, University of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Jan Drappatz
- Department of Neurology, University of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| |
Collapse
|
11
|
Aggarwal P, Luo W, Pehlivan KC, Hoang H, Rajappa P, Cripe TP, Cassady KA, Lee DA, Cairo MS. Pediatric versus adult high grade glioma: Immunotherapeutic and genomic considerations. Front Immunol 2022; 13:1038096. [PMID: 36483545 PMCID: PMC9722734 DOI: 10.3389/fimmu.2022.1038096] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022] Open
Abstract
High grade gliomas are identified as malignant central nervous tumors that spread rapidly and have a universally poor prognosis. Historically high grade gliomas in the pediatric population have been treated similarly to adult high grade gliomas. For the first time, the most recent classification of central nervous system tumors by World Health Organization has divided adult from pediatric type diffuse high grade gliomas, underscoring the biologic differences between these tumors in different age groups. The objective of our review is to compare high grade gliomas in the adult versus pediatric patient populations, highlighting similarities and differences in epidemiology, etiology, pathogenesis and therapeutic approaches. High grade gliomas in adults versus children have varying clinical presentations, molecular biology background, and response to chemotherapy, as well as unique molecular targets. However, increasing evidence show that they both respond to recently developed immunotherapies. This review summarizes the distinctions and commonalities between the two in disease pathogenesis and response to therapeutic interventions with a focus on immunotherapy.
Collapse
Affiliation(s)
- Payal Aggarwal
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | | | - Hai Hoang
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Prajwal Rajappa
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Dean A. Lee
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
12
|
Bottlenecks and opportunities in immunotherapy for glioma: a narrative review. JOURNAL OF BIO-X RESEARCH 2022. [DOI: 10.1097/jbr.0000000000000135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
13
|
Jaén M, Martín-Regalado Á, Bartolomé RA, Robles J, Casal JI. Interleukin 13 receptor alpha 2 (IL13Rα2): Expression, signaling pathways and therapeutic applications in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188802. [PMID: 36152905 DOI: 10.1016/j.bbcan.2022.188802] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 10/14/2022]
Abstract
Interleukin 13 receptor alpha 2 (IL13Rα2) is increasingly recognized as a relevant player in cancer invasion and metastasis. Despite being initially considered a decoy receptor for dampening the levels of interleukin 13 (IL-13) in diverse inflammatory conditions, accumulating evidences in the last decades indicate the capacity of IL13Rα2 for mediating IL-13 signaling in cancer cells. The biological reasons behind the expression of this receptor with such extremely high affinity for IL-13 in cancer cells remain unclear. Elevated expression of IL13Rα2 is commonly associated with invasion, late stage and cancer metastasis that results in poor prognosis for glioblastoma, colorectal or breast cancer, among others. The discovery of new mediators and effectors of IL13Rα2 signaling has been critical for deciphering its underlying molecular mechanisms in cancer progression. Still, many questions about the effects of inflammation, the cancer type and the tumor degree in the expression of IL13Rα2 remain largely uncharacterized. Here, we review and discuss the current status of the IL13Rα2 biology in cancer, with particular emphasis in the role of inflammation-driven expression and the regulation of different signaling pathways. As IL13Rα2 implications in cancer continue to grow exponentially, we highlight new targeted therapies recently developed for glioblastoma, colorectal cancer and other IL13Rα2-positive tumors.
Collapse
Affiliation(s)
- Marta Jaén
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ángela Martín-Regalado
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Javier Robles
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain; Protein Alternatives SL, Tres Cantos, Madrid, Spain
| | - J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
14
|
In vivo DNA-launched bispecific T cell engager targeting IL-13Rα2 controls tumor growth in an animal model of glioblastoma multiforme. Mol Ther Oncolytics 2022; 26:289-301. [PMID: 36090479 PMCID: PMC9418050 DOI: 10.1016/j.omto.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/03/2022] [Indexed: 11/21/2022] Open
Abstract
Glioblastoma is an aggressive tumor with poor survival rates. Bispecific T cell engagers (BTEs) against different cancers are in various stages of clinical development. Toxicity resulting from cytokine release syndrome and the short half-life of BTEs, which necessitates continuous infusion, complicating delivery and increasing costs, are major challenges in the field. Here we describe the development of in vivo DNA-launched BTEs (dBTEs) with highly focused targeting of interleukin-13 receptor α2 (IL-13Rα2), a glioblastoma cell-surface target. We developed 4 BTEs targeting 2 epitopes of IL-13Rα2 and studied how heavy-light chain orientation affects BTE function. The dBTEs induced T cell activation, cytokine production, and tumor cytolysis in the presence of IL-13Rα2+ tumor cells, but we observed unique patterns of immune activation. We found a strong correlation between granzyme B secretion and dBTE-induced cytolysis of specific and nonspecific tumors. We down-selected dBTE PB01-forward based on lower cytokine induction profile and highest activation specificity. In vivo, dBTE PB01-forward demonstrated an improved half-life versus intravenous recombinant BTE delivery. In an orthotopic glioblastoma model, dBTE PB01-forward controlled tumor growth, improving animal survival, supporting the hypothesis that the blood-brain barrier does not affect the function of systemically delivered dBTE. Further study of PB01-forward for targeting glioblastoma and other IL-13Rα2+ cancers is warranted.
Collapse
|
15
|
Li L, Zhou J, Dong X, Liao Q, Zhou D, Zhou Y. Dendritic cell vaccines for glioblastoma fail to complete clinical translation: Bottlenecks and potential countermeasures. Int Immunopharmacol 2022; 109:108929. [PMID: 35700581 DOI: 10.1016/j.intimp.2022.108929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/20/2022] [Accepted: 06/05/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma (GBM) is a heterogeneous and invasive WHO grade IV brain tumor. Patients with GBM have a median overall survival (OS) of only 14 to 17 months when treated with surgical resection and chemoradiation. As one of the most promising anti-tumor immunotherapies, dendritic cell (DC) vaccines have demonstrated good efficacy, safety, and tolerability in many clinical trials. However, to date, no Phase III clinical trial has achieved positive endpoints and truly implement clinical development and transformation. Moreover, the survival benefits of DC vaccines for patients with GBM seem to have a delayed effect; therefore, we urgently require strategies to optimize DC vaccines to advance the time point of its survival benefits. Here, we discuss the latest clinical trial progress of DC vaccines in GBM and summarize the benefits and drawbacks of various vaccine design options, as well as the challenges faced in clinical translation. Moreover, we target future combination therapy strategies for DC vaccines in GBM, which provides a new perspective for comprehensively understanding the effectiveness, limitations, and new directions of the development of DC vaccines.
Collapse
Affiliation(s)
- Luohong Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Jing Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Xueting Dong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Dongbo Zhou
- Department of Geriatric, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China, Hunan 410008, China.
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
16
|
Knudson KM, Hwang S, McCann MS, Joshi BH, Husain SR, Puri RK. Recent Advances in IL-13Rα2-Directed Cancer Immunotherapy. Front Immunol 2022; 13:878365. [PMID: 35464460 PMCID: PMC9023787 DOI: 10.3389/fimmu.2022.878365] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/17/2022] [Indexed: 01/14/2023] Open
Abstract
Interleukin-13 receptor subunit alpha-2 (IL-13Rα2, CD213A), a high-affinity membrane receptor of the anti-inflammatory Th2 cytokine IL-13, is overexpressed in a variety of solid tumors and is correlated with poor prognosis in glioblastoma, colorectal cancer, adrenocortical carcinoma, pancreatic cancer, and breast cancer. While initially hypothesized as a decoy receptor for IL-13-mediated signaling, recent evidence demonstrates IL-13 can signal through IL-13Rα2 in human cells. In addition, expression of IL-13Rα2 and IL-13Rα2-mediated signaling has been shown to promote tumor proliferation, cell survival, tumor progression, invasion, and metastasis. Given its differential expression in tumor versus normal tissue, IL-13Rα2 is an attractive immunotherapy target, as both a targetable receptor and an immunogenic antigen. Multiple promising strategies, including immunotoxins, cancer vaccines, and chimeric antigen receptor (CAR) T cells, have been developed to target IL-13Rα2. In this mini-review, we discuss recent developments surrounding IL-13Rα2-targeted therapies in pre-clinical and clinical study, including potential strategies to improve IL-13Rα2-directed cancer treatment efficacy.
Collapse
|
17
|
Shamshiripour P, Nikoobakht M, Mansourinejad Z, Ahmadvand D, Akbarpour M. A comprehensive update to DC therapy for glioma; a systematic review and meta-analysis. Expert Rev Vaccines 2022; 21:513-531. [DOI: 10.1080/14760584.2022.2027759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Parisa Shamshiripour
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of medical imaging technology and molecular imaging, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Nikoobakht
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - zahra Mansourinejad
- Department of systems biology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Davoud Ahmadvand
- Department of medical imaging technology and molecular imaging, Iran University of Medical Sciences, Tehran, Iran
| | - Mahzad Akbarpour
- Advanced Cellular Therapeutics Facility, David and Etta Jonas Center for Cellular Therapy, Hematopoietic Cellular Therapy Program, The University of Chicago Medical Center, Chicago 60637 IL, USA
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno-TACT), Universal Science and Education Research Network (USERN), Chicago, USA
| |
Collapse
|
18
|
Datsi A, Sorg RV. Dendritic Cell Vaccination of Glioblastoma: Road to Success or Dead End. Front Immunol 2021; 12:770390. [PMID: 34795675 PMCID: PMC8592940 DOI: 10.3389/fimmu.2021.770390] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastomas (GBM) are the most frequent and aggressive malignant primary brain tumor and remains a therapeutic challenge: even after multimodal therapy, median survival of patients is only 15 months. Dendritic cell vaccination (DCV) is an active immunotherapy that aims at inducing an antitumoral immune response. Numerous DCV trials have been performed, vaccinating hundreds of GBM patients and confirming feasibility and safety. Many of these studies reported induction of an antitumoral immune response and indicated improved survival after DCV. However, two controlled randomized trials failed to detect a survival benefit. This raises the question of whether the promising concept of DCV may not hold true or whether we are not yet realizing the full potential of this therapeutic approach. Here, we discuss the results of recent vaccination trials, relevant parameters of the vaccines themselves and of their application, and possible synergies between DCV and other therapeutic approaches targeting the immunosuppressive microenvironment of GBM.
Collapse
Affiliation(s)
- Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University Hospital, Medical Faculty, Düsseldorf, Germany
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University Hospital, Medical Faculty, Düsseldorf, Germany
| |
Collapse
|
19
|
Kim K, Gwak HS, Han N, Hong EK, Choi BK, Lee S, Choi S, Park JH, Seok JH, Jeon Y, Cho H, Lee SJ, Lee Y, Nam KT, Song SW. Chimeric Antigen Receptor T Cells With Modified Interleukin-13 Preferentially Recognize IL13Rα2 and Suppress Malignant Glioma: A Preclinical Study. Front Immunol 2021; 12:715000. [PMID: 34819930 PMCID: PMC8606595 DOI: 10.3389/fimmu.2021.715000] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/15/2021] [Indexed: 12/05/2022] Open
Abstract
Background Interleukin-13 receptor α 2 (IL13Rα2) is a promising tumor-directed antigen of malignant glioma (MG). Here, we examine the efficacy and safety of T cells containing a YYB-103 chimeric antigen receptor (CAR) that can preferentially bind to IL13Rα2 on MG cells. Methods IL13 was modified on the extracellular domain by substitution of amino acids with E13K, R66D, S69D, and R109K and stably transfected into human T cells using a retroviral vector. The in vitro efficacy of YYB-103 CAR T cells was tested in cell lines with differing IL13Rα1 and IL13Rα2 expression. The in vivo efficacy of intracerebroventricular (i.c.v.) and intravenous (i.v.) routes of YYB-103 CAR T-cell administration were tested in orthotopic MG mouse models. Immunohistochemical staining of MG was performed using WHO grade 3/4 surgical specimens from 53 patients. IL13Rα2 expression was quantified by H-score calculated from staining intensity and percentage of positive cells. Results Binding affinity assay of YYB-103 verified apparently nil binding to IL13Rα1, which was more selective than previously reported IL13 modification (E13Y). YYB-103 CAR T cells showed selective toxicity toward co-cultured U87MG (IL13Rα1+/IL13Rα2+) cells but not A431 (IL13Rα1+/IL13Rα2-) cells. Consistently, YYB-103 CAR T cells suppressed tumor growth in nude mice receiving orthotopic injection of U87 MG cells. Both i.c.v. and i.v. injections of YYB-103 CAR T cells reduced tumor volume and prolonged overall survival of tumor-bearing mice. The median H-score for IL13Rα2 in patient-derived MG tissue was 5 (mean, 57.5; SD, 87.2; range, 0 to 300). Conclusion This preclinical study demonstrates the efficacy of IL13Rα2-targeted YYB-103 CAR T cells against MG cells. The use of modified IL13 to construct a CAR facilitated the selective targeting of IL13Rα2-expressing MG cells while sparing IL13Rα1-expressing cells. Notably, YYB-103 CAR T cells exhibited effective blood-brain barrier crossing, suggesting compatibility with i.v. administration rather than intracranial injection. Additionally, the high H-score for IL13Rα2 in glioblastoma, especially in conjunction with the poor prognostic markers of wild-type isocitrate dehydrogenase-1 (IDH-1) and unmethylated O6-methyl guanine methyl-transferase (MGMT), could be used to determine the eligibility of patients with recurrent glioblastoma for a future clinical trial of YYB-103 CAR T cells.
Collapse
Affiliation(s)
- Kiwan Kim
- Department of Drug Development I, CellabMED Inc., Seoul, South Korea
| | - Ho-Shin Gwak
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, South Korea
| | - Nayoung Han
- Department of Pathology, Program for Immunotherapy Research, National Cancer Center, Goyang, South Korea
| | - Eun Kyung Hong
- Department of Pathology, Program for Immunotherapy Research, National Cancer Center, Goyang, South Korea
| | - Beom K. Choi
- Biomedicine Production Branch, Program for Immunotherapy Research, National Cancer Center, Goyang, South Korea
| | - Sangeun Lee
- Department of Drug Development I, CellabMED Inc., Seoul, South Korea
| | - Soyoung Choi
- Department of Drug Development I, CellabMED Inc., Seoul, South Korea
| | - Ju-Hwang Park
- Department of Process Development, CellabMED Inc., Seoul, South Korea
| | - Ji-Hye Seok
- Department of Process Development, CellabMED Inc., Seoul, South Korea
| | - Yeongha Jeon
- Department of Drug Development II, CellabMED Inc., Seoul, South Korea
| | - Hyuntae Cho
- Department of Clinical Development, CellabMED Inc., Seoul, South Korea
| | - Song-Jae Lee
- Research Institute, CellabMED Inc., Seoul, South Korea
| | - Yura Lee
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | | |
Collapse
|
20
|
Frederico SC, Hancock JC, Brettschneider EES, Ratnam NM, Gilbert MR, Terabe M. Making a Cold Tumor Hot: The Role of Vaccines in the Treatment of Glioblastoma. Front Oncol 2021; 11:672508. [PMID: 34041034 PMCID: PMC8141615 DOI: 10.3389/fonc.2021.672508] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 12/28/2022] Open
Abstract
The use of immunotherapies for the treatment of brain tumors is a topic that has garnered considerable excitement in recent years. Discoveries such as the presence of a glymphatic system and immune surveillance in the central nervous system (CNS) have shattered the theory of immune privilege and opened up the possibility of treating CNS malignancies with immunotherapies. However, despite many immunotherapy clinical trials aimed at treating glioblastoma (GBM), very few have demonstrated a significant survival benefit. Several factors for this have been identified, one of which is that GBMs are immunologically "cold," implying that the cancer does not induce a strong T cell response. It is postulated that this is why clinical trials using an immune checkpoint inhibitor alone have not demonstrated efficacy. While it is well established that anti-cancer T cell responses can be facilitated by the presentation of tumor-specific antigens to the immune system, treatment-related death of GBM cells and subsequent release of molecules have not been shown to be sufficient to evoke an anti-tumor immune response effective enough to have a significant impact. To overcome this limitation, vaccines can be used to introduce exogenous antigens at higher concentrations to the immune system to induce strong tumor antigen-specific T cell responses. In this review, we will describe vaccination strategies that are under investigation to treat GBM; categorizing them based on their target antigens, form of antigens, vehicles used, and pairing with specific adjuvants. We will review the concept of vaccine therapy in combination with immune checkpoint inhibitors, as it is hypothesized that this approach may be more effective in overcoming the immunosuppressive milieu of GBM. Clinical trial design and the need for incorporating robust immune monitoring into future studies will also be discussed here. We believe that the integration of evolving technologies of vaccine development, delivery, and immune monitoring will further enhance the role of these therapies and will likely remain an important area of investigation for future treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Stephen C. Frederico
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, United States
| | - John C. Hancock
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, United States
| | - Emily E. S. Brettschneider
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, United States
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Nivedita M. Ratnam
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, United States
| | - Mark R. Gilbert
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Randomized Controlled Immunotherapy Clinical Trials for GBM Challenged. Cancers (Basel) 2020; 13:cancers13010032. [PMID: 33374196 PMCID: PMC7796083 DOI: 10.3390/cancers13010032] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Although multiple meta-analyses on active specific immunotherapy treatment for glioblastoma multiforme (GBM) have demonstrated a significant prolongation of overall survival, no single research group has succeeded in demonstrating the efficacy of this type of treatment in a prospective, double-blind, placebo-controlled, randomized clinical trial. In this paper, we explain how the complexity of the tumor biology and tumor–host interactions make proper stratification of a control group impossible. The individualized characteristics of advanced therapy medicinal products for immunotherapy contribute to heterogeneity within an experimental group. The dynamics of each tumor and in each patient aggravate comparative stable patient groups. Finally, combinations of immunotherapy strategies should be integrated with first-line treatment. We illustrate the complexity of a combined first-line treatment with individualized multimodal immunotherapy in a group of 70 adults with GBM and demonstrate that the integration of immunogenic cell death treatment within maintenance chemotherapy followed by dendritic cell vaccines and maintenance immunotherapy might provide a step towards improving the overall survival rate of GBM patients. Abstract Immunotherapies represent a promising strategy for glioblastoma multiforme (GBM) treatment. Different immunotherapies include the use of checkpoint inhibitors, adoptive cell therapies such as chimeric antigen receptor (CAR) T cells, and vaccines such as dendritic cell vaccines. Antibodies have also been used as toxin or radioactive particle delivery vehicles to eliminate target cells in the treatment of GBM. Oncolytic viral therapy and other immunogenic cell death-inducing treatments bridge the antitumor strategy with immunization and installation of immune control over the disease. These strategies should be included in the standard treatment protocol for GBM. Some immunotherapies are individualized in terms of the medicinal product, the immune target, and the immune tumor–host contact. Current individualized immunotherapy strategies focus on combinations of approaches. Standardization appears to be impossible in the face of complex controlled trial designs. To define appropriate control groups, stratification according to the Recursive Partitioning Analysis classification, MGMT promotor methylation, epigenetic GBM sub-typing, tumor microenvironment, systemic immune functioning before and after radiochemotherapy, and the need for/type of symptom-relieving drugs is required. Moreover, maintenance of a fixed treatment protocol for a dynamic, deadly cancer disease in a permanently changing tumor–host immune context might be inappropriate. This complexity is illustrated using our own data on individualized multimodal immunotherapies for GBM. Individualized medicines, including multimodal immunotherapies, are a rational and optimal yet also flexible approach to induce long-term tumor control. However, innovative methods are needed to assess the efficacy of complex individualized treatments and implement them more quickly into the general health system.
Collapse
|
22
|
Maggi E, Veneziani I, Moretta L, Cosmi L, Annunziato F. Group 2 Innate Lymphoid Cells: A Double-Edged Sword in Cancer? Cancers (Basel) 2020; 12:cancers12113452. [PMID: 33233582 PMCID: PMC7699723 DOI: 10.3390/cancers12113452] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Group 2 Innate Lymphoid Cells (ILC2s) belong to the family of helper ILCs which provide host defense against infectious agents, participate in inflammatory responses and mediate lymphoid organogenesis and tissue repair, mainly at the skin and mucosal level. Based on their transcriptional, phenotypic and functional profile, ILC2s mirror the features of the adaptive CD4+ Th2 cell subset, both contributing to the so-called type 2 immune response. Similar to other ILCs, ILC2s are rapidly activated by signals deriving from tissue and/or other tissue-resident immune cells. The biologic activity of ILCs needs to be tightly regulated in order to prevent them from contributing to severe inflammation and damage in several organs. Indeed, ILC2s display both enhancing and regulatory roles in several pathophysiological conditions, including tumors. In this review, we summarize the actual knowledge about ILC2s ability to induce or impair a protective immune response, their pro- or antitumor activity in murine models, human (children and adults) pathologies and the potential strategies to improve cancer immunotherapy by exploiting the features of ILC2s.
Collapse
Affiliation(s)
- Enrico Maggi
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
- Correspondence: ; Tel.: +39-06-6859-3617
| | - Irene Veneziani
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
| | - Lorenzo Moretta
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.C.); (F.A.)
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.C.); (F.A.)
| |
Collapse
|
23
|
Khodamoradi E, Hoseini-Ghahfarokhi M, Amini P, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. Targets for protection and mitigation of radiation injury. Cell Mol Life Sci 2020; 77:3129-3159. [PMID: 32072238 PMCID: PMC11104832 DOI: 10.1007/s00018-020-03479-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Abstract
Protection of normal tissues against toxic effects of ionizing radiation is a critical issue in clinical and environmental radiobiology. Investigations in recent decades have suggested potential targets that are involved in the protection against radiation-induced damages to normal tissues and can be proposed for mitigation of radiation injury. Emerging evidences have been shown to be in contrast to an old dogma in radiation biology; a major amount of reactive oxygen species (ROS) production and cell toxicity occur during some hours to years after exposure to ionizing radiation. This can be attributed to upregulation of inflammatory and fibrosis mediators, epigenetic changes and disruption of the normal metabolism of oxygen. In the current review, we explain the cellular and molecular changes following exposure of normal tissues to ionizing radiation. Furthermore, we review potential targets that can be proposed for protection and mitigation of radiation toxicity.
Collapse
Affiliation(s)
- Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Hoseini-Ghahfarokhi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
- Misan Radiotherapy Center, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
24
|
Guo J, Muse E, Christians AJ, Swanson SJ, Davila E. An Anticancer Drug Cocktail of Three Kinase Inhibitors Improved Response to a Dendritic Cell-Based Cancer Vaccine. Cancer Immunol Res 2019; 7:1523-1534. [PMID: 31266784 PMCID: PMC6726569 DOI: 10.1158/2326-6066.cir-18-0684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/22/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
Monocyte-derived dendritic cell (moDC)-based cancer therapies intended to elicit antitumor T-cell responses have limited efficacy in most clinical trials. However, potent and sustained antitumor activity in a limited number of patients highlights the therapeutic potential of moDCs. In vitro culture conditions used to generate moDCs can be inconsistent, and moDCs generated in vitro are less effective than natural DCs. On the basis of our study highlighting the ability for certain kinase inhibitors to enhance tumor antigenicity, we therefore screened kinase inhibitors for their ability to improve DC immunogenicity. We identified AKT inhibitor MK2206, DNA-PK inhibitor NU7441, and MEK inhibitor trametinib as the compounds most effective at modulating moDC immunogenicity. The combination of these drugs, referred to as MKNUTRA, enhanced moDC activity over treatment with individual drugs while exhibiting minimal toxicity. An evaluation of 335 activation and T-cell-suppressive surface proteins on moDCs revealed that MKNUTRA treatment more effectively matured cells and reduced the expression of tolerogenic proteins as compared with control moDCs. MKNUTRA treatment imparted to ICT107, a glioblastoma (GBM) DC-based vaccine that has completed phase II trials, an increased ability to stimulate patient-derived autologous CD8+ T cells against the brain tumor antigens IL13Rα2(345-354) and TRP2(180-188) In vivo, treating ICT107 with MKNUTRA, prior to injection into mice with an established GBM tumor, reduced tumor growth kinetics. This response was associated with an increased frequency of tumor-reactive lymphocytes within tumors and in peripheral tissues. These studies broaden the application of targeted anticancer drugs and highlight their ability to increase moDC immunogenicity.
Collapse
Affiliation(s)
- Jitao Guo
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Elena Muse
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Allison J Christians
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | | | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
- Human Immunology and Immunotherapy Initiative, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Comprehensive Cancer Center, Aurora, Colorado
| |
Collapse
|
25
|
A Characterization of Dendritic Cells and Their Role in Immunotherapy in Glioblastoma: From Preclinical Studies to Clinical Trials. Cancers (Basel) 2019; 11:cancers11040537. [PMID: 30991681 PMCID: PMC6521200 DOI: 10.3390/cancers11040537] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is the most common and fatal primary central nervous system malignancy in adults with a median survival of less than 15 months. Surgery, radiation, and chemotherapy are the standard of care and provide modest benefits in survival, but tumor recurrence is inevitable. The poor prognosis of GBM has made the development of novel therapies targeting GBM of paramount importance. Immunotherapy via dendritic cells (DCs) has garnered attention and research as a potential strategy to boost anti-tumor immunity in recent years. As the “professional” antigen processing and presenting cells, DCs play a key role in the initiation of anti-tumor immune responses. Pre-clinical studies in GBM have shown long-term tumor survival and immunological memory in murine models with stimulation of DC activity with various antigens and costimulatory molecules. Phase I and II clinical trials of DC vaccines in GBM have demonstrated some efficacy in improving the median overall survival with minimal to no toxicity with promising initial results from the first Phase III trial. However, there remains no standardization of vaccines in terms of which antigens are used to pulse DCs ex vivo, sites of DC injection, and optimal adjuvant therapies. Future work with DC vaccines aims to elucidate the efficacy of DC-based therapy alone or in combination with other immunotherapy adjuvants in additional Phase III trials.
Collapse
|
26
|
Rapp M, Grauer OM, Kamp M, Sevens N, Zotz N, Sabel M, Sorg RV. A randomized controlled phase II trial of vaccination with lysate-loaded, mature dendritic cells integrated into standard radiochemotherapy of newly diagnosed glioblastoma (GlioVax): study protocol for a randomized controlled trial. Trials 2018; 19:293. [PMID: 29801515 PMCID: PMC5970474 DOI: 10.1186/s13063-018-2659-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/02/2018] [Indexed: 01/06/2023] Open
Abstract
Background Despite the combination of surgical resection, radio- and chemotherapy, median survival of glioblastoma multiforme (GBM) patients only slightly increased in the last years. Disease recurrence is definite with no effective therapy existing after tumor removal. Dendritic cell (DC) vaccination is a promising active immunotherapeutic approach. There is clear evidence that it is feasible, results in immunological anti-tumoral responses, and appears to be beneficial for survival and quality of life of GBM patients. Moreover, combining it with the standard therapy of GBM may allow exploiting synergies between the treatment modalities. In this randomized controlled trial, we seek to confirm these promising initial results. Methods One hundred and thirty-six newly diagnosed, isocitrate dehydrogenase wildtype GBM patients will be randomly allocated (1:1 ratio, stratified by O6-methylguanine-DNA-methyltransferase promotor methylation status) after near-complete resection in a multicenter, prospective phase II trial into two groups: (1) patients receiving the current therapeutic “gold standard” of radio/temozolomide chemotherapy and (2) patients receiving DC vaccination as an add-on to the standard therapy. A recruitment period of 30 months is anticipated; follow-up will be 2 years. The primary objective of the study is to compare overall survival (OS) between the two groups. Secondary objectives are comparing progression-free survival (PFS) and 6-, 12- and 24-month OS and PFS rates, the safety profile, overall and neurological performance and quality of life. Discussion Until now, close to 500 GBM patients have been treated with DC vaccination in clinical trials or on a compassionate-use basis. Results have been encouraging, but cannot provide robust evidence of clinical efficacy because studies have been non-controlled or patient numbers have been low. Therefore, a prospective, randomized phase II trial with a sufficiently large number of patients is now mandatory for clear evidence regarding the impact of DC vaccination on PFS and OS in GBM. Trial registration Protocol code: GlioVax, date of registration: 17. February 2017. Trial identifier: EudraCT-Number 2017–000304-14. German Registry for Clinical Studies, ID: DRKS00013248 (approved primary register in the WHO network) and at ClinicalTrials.gov, ID: NCT03395587. Registered on 11 March 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2659-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marion Rapp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Department of Neurosurgery, Heinrich Heine University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - Oliver M Grauer
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Marcel Kamp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Natalie Sevens
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Nikola Zotz
- Coordination Center for Clinical Trials, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Michael Sabel
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| |
Collapse
|
27
|
Prinzing BL, Gottschalk SM, Krenciute G. CAR T-cell therapy for glioblastoma: ready for the next round of clinical testing? Expert Rev Anticancer Ther 2018; 18:451-461. [PMID: 29533108 PMCID: PMC6191291 DOI: 10.1080/14737140.2018.1451749] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The outcome for patients with glioblastoma (GBM) remains poor, and there is an urgent need to develop novel therapeutic approaches. T cells genetically modified with chimeric antigen receptors (CARs) hold the promise to improve outcomes since they recognize and kill cells through different mechanisms than conventional therapeutics. Areas covered: This article reviews CAR design, tumor associated antigens expressed by GBMs that can be targeted with CAR T cells, preclinical and clinical studies conducted with CAR T cells, and genetic approaches to enhance their effector function. Expert commentary: While preclinical studies have highlighted the potent anti-GBM activity of CAR T cells, the initial foray of CAR T-cell therapies into the clinic resulted only in limited benefits for GBM patients. Additional genetic modification of CAR T cells has resulted in a significant increase in their anti-GBM activity in preclinical models. We are optimistic that clinical testing of these enhanced CAR T cells will be safe and result in improved anti-glioma activity in GBM patients.
Collapse
Affiliation(s)
- Brooke L. Prinzing
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, Texas 77030
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Stephen M. Gottschalk
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Giedre Krenciute
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
28
|
Survival outcomes in pediatric recurrent high-grade glioma: results of a 20-year systematic review and meta-analysis. J Neurooncol 2017; 137:103-110. [PMID: 29204840 DOI: 10.1007/s11060-017-2701-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/24/2017] [Indexed: 10/18/2022]
Abstract
Recurrent pediatric high-grade glioma is a leading cause of cancer-related death in children. We report results of a systematic review and meta-analysis investigating survival outcome in pediatric patients with recurrent high-grade glioma over the last 20 years. MEDLINE/PubMed, EMBASE, Web of Science and Cochrane Review databases were searched for relevant studies reporting on survival outcomes for pediatric patients with recurrent high-grade glioma treated between 1996 and 2016. Progression-free survival (PFS) and overall survival (OS) were calculated cumulatively over all studies, by therapy subgroup, and by decade of treatment. Random effects models were used to control for heterogeneity as measured by the I2 statistic. A total of 17 studies across 4 treatment strategies were included. Eleven investigated traditional chemotherapy, 1 investigated targeted therapy, 3 investigated immunotherapy, and 2 investigated radiotherapy. A total of 129 patients were included with a median age of 10.0 years. Cumulative PFS was 3.5 months (95% CI 2.1-5.0). Cumulative OS was 5.6 months (95% CI 3.9-7.3). OS was 4.0 months (95% CI 1.9-6.1) using traditional chemotherapy, 9.3 months using targeted therapies (95% CI 5.4-13), 6.9 months using immunotherapy (95% CI 2.1-12), and 14 months using reirradiation (95% CI 2.8-25). OS between 1996 and 2006 was 4.2 months (95% CI 2.1-6.2) compared to 8.5 months (95% CI 5.6-11) after 2006. Pediatric patients with recurrent high-grade glioma suffer from poor PFS and OS, regardless of therapy. There may be a trend towards improved OS in the last decade.
Collapse
|
29
|
Sequeida A, Maisey K, Imarai M. Interleukin 4/13 receptors: An overview of genes, expression and functional role in teleost fish. Cytokine Growth Factor Rev 2017; 38:66-72. [PMID: 28988781 DOI: 10.1016/j.cytogfr.2017.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
Abstract
In superior vertebrates, Interleukin 4 (IL-4) and Interleukin 13 (IL-13) play key and diverse roles to support immune responses acting on cell surface receptors. When stimulated, receptors activate intracellular signalling cascades switching cell phenotypes according to stimuli. In teleost fish, Interleukin 4/13 (IL-4/13) is the ancestral family cytokine related to both IL-4 and IL-13. Every private and common receptor subunit for IL-4/13 have in fish at least two paralogues and, as in mammals, soluble forms are also part of the receptor system. Reports for findings of fish IL-4/13 receptors have covered comparative analysis, transcriptomic profiles and to a lesser extent, functional analysis regarding ligand-receptor interactions and their biological effects. This review addresses available information from fish IL-4/13 receptors and discusses overall implications on teleost immunity, summarized gene induction strategies and pathogen-induced gene modulation, which may be useful tools to enhance immune response. Additionally, we present novel coding sequences for Atlantic salmon (Salmo salar) common gamma chain receptor (γC), Interleukin 13 receptor alpha 1A chain (IL-13Rα1A) and Interleukin 13 receptor alpha 1B chain (IL-13Rα1B).
Collapse
Affiliation(s)
- A Sequeida
- Laboratory of Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363 Santiago, Chile
| | - K Maisey
- Laboratory of Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363 Santiago, Chile; Laboratory of Comparative Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile,Av. Bernardo O'Higgins, 3363 Santiago, Chile
| | - M Imarai
- Laboratory of Immunology, Center for Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363 Santiago, Chile.
| |
Collapse
|
30
|
Kuramitsu S, Yamamichi A, Ohka F, Motomura K, Hara M, Natsume A. Adoptive immunotherapy for the treatment of glioblastoma: progress and possibilities. Immunotherapy 2017; 8:1393-1404. [PMID: 28000534 DOI: 10.2217/imt-2016-0076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Patients with glioblastoma have a very poor prognosis. Adoptive cellular therapy (ACT) is defined as the collection of circulating or tumor-infiltrating lymphocytes, their selection, modification, expansion and activation, and their re-administration to patients in order to induce antitumor activity. Although various ACTs have been attempted, most failed to improve the outcome. Immune checkpoint blockade antibodies and T cell engineering with tumor-specific chimeric antigen receptors suggest the emergence of a new era of immunotherapy. Here, we summarize approaches with ACTs using genetically modified T cells, which have been improved by enhancing their antitumor activity, and discuss strategies to develop these therapies. The mechanisms by which gliomas modulate and evade the immune system are also discussed.
Collapse
Affiliation(s)
- Shunichiro Kuramitsu
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Akane Yamamichi
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Fumiharu Ohka
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Kazuya Motomura
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Masahito Hara
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
31
|
Tao Z, Li S, Ichim TE, Yang J, Riordan N, Yenugonda V, Babic I, Kesari S. Cellular immunotherapy of cancer: an overview and future directions. Immunotherapy 2017; 9:589-606. [DOI: 10.2217/imt-2016-0086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The clinical success of checkpoint inhibitors has led to a renaissance of interest in cancer immunotherapies. In particular, the possibility of ex vivo expanding autologous lymphocytes that specifically recognize tumor cells has attracted much research and clinical trial interest. In this review, we discuss the historical background of tumor immunotherapy using cell-based approaches, and provide some rationale for overcoming current barriers to success of autologous immunotherapy. An overview of adoptive transfer of lymphocytes, tumor infiltrating lymphocytes and dendritic cell therapies is provided. We conclude with discussing the possibility of gene-manipulating immune cells in order to augment therapeutic activity, including silencing of the immune-suppressive zinc finger orphan nuclear receptor, NR2F6, as an attractive means of overcoming tumor-associated immune suppression.
Collapse
Affiliation(s)
- Ziqi Tao
- The Affiliated XuZhou Center Hospital of Nanjing University of Chinese Medicine, The Affiliated XuZhou Hospital of Medical College of Southeast University, Jiangsu, China
| | - Shuang Li
- Department of Endocrinology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | | | - Junbao Yang
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Neil Riordan
- Medistem Panama, Inc., City of Knowledge, Clayton, Republic of Panama
| | - Venkata Yenugonda
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Ivan Babic
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
- John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA 90404, USA
| |
Collapse
|
32
|
Dutoit V, Migliorini D, Dietrich PY, Walker PR. Immunotherapy of Malignant Tumors in the Brain: How Different from Other Sites? Front Oncol 2016; 6:256. [PMID: 28003994 PMCID: PMC5141244 DOI: 10.3389/fonc.2016.00256] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/24/2016] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy is now advancing at remarkable pace for tumors located in various tissues, including the brain. Strategies launched decades ago, such as tumor antigen-specific therapeutic vaccines and adoptive transfer of tumor-infiltrating lymphocytes are being complemented by molecular engineering approaches allowing the development of tumor-specific TCR transgenic and chimeric antigen receptor T cells. In addition, the spectacular results obtained in the last years with immune checkpoint inhibitors are transfiguring immunotherapy, these agents being used both as single molecules, but also in combination with other immunotherapeutic modalities. Implementation of these various strategies is ongoing for more and more malignancies, including tumors located in the brain, raising the question of the immunological particularities of this site. This may necessitate cautious selection of tumor antigens, minimizing the immunosuppressive environment and promoting efficient T cell trafficking to the tumor. Once these aspects are taken into account, we might efficiently design immunotherapy for patients suffering from tumors located in the brain, with beneficial clinical outcome.
Collapse
Affiliation(s)
- Valérie Dutoit
- Laboratory of Tumor Immunology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Denis Migliorini
- Oncology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Pierre-Yves Dietrich
- Oncology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| | - Paul R Walker
- Laboratory of Tumor Immunology, Center of Oncology, Geneva University Hospitals and University of Geneva , Geneva , Switzerland
| |
Collapse
|
33
|
Artene SA, Turcu-Stiolica A, Hartley R, Ciurea ME, Daianu O, Brindusa C, Alexandru O, Tataranu LG, Purcaru SO, Dricu A. Dendritic cell immunotherapy versus bevacizumab plus irinotecan in recurrent malignant glioma patients: a survival gain analysis. Onco Targets Ther 2016; 9:6669-6677. [PMID: 27877052 PMCID: PMC5108618 DOI: 10.2147/ott.s112842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The bevacizumab and irinotecan protocol is considered a standard treatment regimen for recurrent malignant glioma. Recent advances in immunotherapy have hinted that vaccination with dendritic cells could become an alternative salvage therapy for the treatment of recurrent malignant glioma. Methods A search was performed on PubMed, Cochrane Library, Web of Science, ScienceDirect, and Embase in order to identify studies with patients receiving bevacizumab plus irinotecan or dendritic cell therapy for recurrent malignant gliomas. The data obtained from these studies were used to perform a systematic review and survival gain analysis. Results Fourteen clinical studies with patients receiving either bevacizumab plus irinotecan or dendritic cell vaccination were identified. Seven studies followed patients that received bevacizumab plus irinotecan (302 patients) and seven studies included patients that received dendritic cell immunotherapy (80 patients). For the patients who received bevacizumab plus irinotecan, the mean reported median overall survival was 7.5 (95% confidence interval [CI] 4.84–10.16) months. For the patients who received dendritic cell immunotherapy, the mean reported median overall survival was 17.9 (95% CI 11.34–24.46) months. For irinotecan + bevacizumab group, the mean survival gain was −0.02±2.00, while that for the dendritic cell immunotherapy group was −0.01±4.54. Conclusion For patients with recurrent malignant gliomas, dendritic cell immunotherapy treatment does not have a significantly different effect when compared with bevacizumab and irinotecan in terms of survival gain (P=0.535) and does not improve weighted survival gain (P=0.620).
Collapse
Affiliation(s)
| | | | | | | | | | | | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Craiova
| | | | | | | |
Collapse
|
34
|
Abstract
Vaccination against cancer-associated antigens has long held the promise of inducting potent antitumor immunity, targeted cytotoxicity while sparing normal tissues, and long-lasting immunologic memory that can provide surveillance against tumor recurrence. Evaluation of vaccination strategies in preclinical brain tumor models has borne out the capacity for the immune system to effectively and safely eradicate established tumors within the central nervous system. Early phase clinical trials have established the feasibility, safety, and immunogenicity of several vaccine platforms, predominantly in patients with glioblastoma. Definitive demonstration of clinical benefit awaits further study, but initial results have been encouraging. With increased understanding of the stimulatory and regulatory pathways that govern immunologic responses and the enhanced capacity to identify novel antigenic targets using genomic interrogation of tumor cells, vaccination platforms for patients with malignant brain tumors are advancing with increasing personalized complexity and integration into combinatorial treatment paradigms.
Collapse
Affiliation(s)
- John H Sampson
- Preston Robert Tisch Brain Tumor Center at Duke, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida (D.A.M.)
| | - Duane A Mitchell
- Preston Robert Tisch Brain Tumor Center at Duke, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida (D.A.M.)
| |
Collapse
|
35
|
Hodges TR, Ferguson SD, Caruso HG, Kohanbash G, Zhou S, Cloughesy TF, Berger MS, Poste GH, Khasraw M, Ba S, Jiang T, Mikkelson T, Yung WKA, de Groot JF, Fine H, Cantley LC, Mellinghoff IK, Mitchell DA, Okada H, Heimberger AB. Prioritization schema for immunotherapy clinical trials in glioblastoma. Oncoimmunology 2016; 5:e1145332. [PMID: 27471611 DOI: 10.1080/2162402x.2016.1145332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/12/2016] [Accepted: 01/16/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Emerging immunotherapeutic strategies for the treatment of glioblastoma (GBM) such as dendritic cell (DC) vaccines, heat shock proteins, peptide vaccines, and adoptive T-cell therapeutics, to name a few, have transitioned from the bench to clinical trials. With upcoming strategies and developing therapeutics, it is challenging to critically evaluate the practical, clinical potential of individual approaches and to advise patients on the most promising clinical trials. METHODS The authors propose a system to prioritize such therapies in an organized and data-driven fashion. This schema is based on four categories of factors: antigenic target robustness, immune-activation and -effector responses, preclinical vetting, and early evidence of clinical response. Each of these categories is subdivided to focus on the most salient elements for developing a successful immunotherapeutic approach for GBM, and a numerical score is generated. RESULTS The Score Card reveals therapeutics that have the most robust data to support their use, provides a reference prioritization score, and can be applied in a reiterative fashion with emerging data. CONCLUSIONS The authors hope that this schema will give physicians an evidence-based and rational framework to make the best referral decisions to better guide and serve this patient population.
Collapse
Affiliation(s)
- Tiffany R Hodges
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Hillary G Caruso
- The Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Gary Kohanbash
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Timothy F Cloughesy
- Department of Neuro-Oncology, the University of California at Los Angeles , Los Angeles, CA, USA
| | - Mitchel S Berger
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | | | | | - Sujuan Ba
- The National Foundation for Cancer Research, Bethesda, MD, USA, Asian Fund for Cancer Research , Hong Kong, People's Republic of China
| | - Tao Jiang
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University , Beijing, China
| | - Tom Mikkelson
- Department of Neurosurgery, Henry Ford Health System , Detroit, MI, USA
| | - W K Alfred Yung
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Howard Fine
- Division of Neuro-Oncology, Weill Cornell Medical College , New York, NY, USA
| | - Lewis C Cantley
- Department of Systems Biology, Harvard Medical School , Boston, MA, USA
| | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Duane A Mitchell
- Department of Neurosurgery, University of Florida , Gainesville, FL, USA
| | - Hideho Okada
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
36
|
Immunomonitoring in glioma immunotherapy: current status and future perspectives. J Neurooncol 2015; 127:1-13. [PMID: 26638171 DOI: 10.1007/s11060-015-2018-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 11/25/2015] [Indexed: 12/28/2022]
Abstract
Given the continued poor clinical outcomes and refractory nature of glioblastoma multiforme to traditional interventions, immunotherapy is gaining traction due to its potential for specific tumor-targeting and long-term antitumor protective surveillance. Currently, development of glioma immunotherapy relies on overall survival as an endpoint in clinical trials. However, the identification of surrogate immunologic biomarkers can accelerate the development of successful immunotherapeutic strategies. Immunomonitoring techniques possess the potential to elucidate immunological mechanisms of antitumor responses, monitor disease progression, evaluate therapeutic effect, identify candidates for immunotherapy, and serve as prognostic markers of clinical outcome. Current immunomonitoring assays assess delayed-type hypersensitivity, T cell proliferation, cytotoxic T-lymphocyte function, cytokine secretion profiles, antibody titers, and lymphocyte phenotypes. Yet, no single immunomonitoring technique can reliably predict outcomes, relegating immunological markers to exploratory endpoints. In response, the most recent immunomonitoring assays are incorporating emerging technologies and novel analysis techniques to approach the goal of identifying a competent immunological biomarker which predicts therapy responsiveness and clinical outcome. This review addresses the current status of immunomonitoring in glioma vaccine clinical trials with emphasis on correlations with clinical response.
Collapse
|
37
|
Suzuki A, Leland P, Joshi BH, Puri RK. Targeting of IL-4 and IL-13 receptors for cancer therapy. Cytokine 2015; 75:79-88. [DOI: 10.1016/j.cyto.2015.05.026] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 02/03/2023]
|
38
|
Kaye EC, Baker JN, Broniscer A. Management of diffuse intrinsic pontine glioma in children: current and future strategies for improving prognosis. CNS Oncol 2015; 3:421-31. [PMID: 25438813 DOI: 10.2217/cns.14.47] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is one of the deadliest pediatric central nervous system cancers in spite of treatment with radiation therapy, the current standard of care. The outcome of affected children remains dismal despite multiple clinical trials that investigated radiation therapy combined with chemotherapy. Recently, multiple genome-wide studies unveiled the distinct molecular characteristics of DIPGs and preclinical models of DIPG were developed to mimic the human disease. Both of these accomplishments have generated tremendous progress in the research of new therapies for children with DIPG. Here we review some of these promising new strategies.
Collapse
Affiliation(s)
- Erica C Kaye
- Department of Oncology, St Jude Children's Research Hospital; 262 Danny Thomas Place, Mail Stop 260, Memphis, TN 38105, USA
| | | | | |
Collapse
|
39
|
Van Gool SW. Brain Tumor Immunotherapy: What have We Learned so Far? Front Oncol 2015; 5:98. [PMID: 26137448 PMCID: PMC4470276 DOI: 10.3389/fonc.2015.00098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/13/2015] [Indexed: 12/17/2022] Open
Abstract
High grade glioma is a rare brain cancer, incurable in spite of modern neurosurgery, radiotherapy, and chemotherapy. Novel approaches are in research, and immunotherapy emerges as a promising strategy. Clinical experiences with active specific immunotherapy demonstrate feasibility, safety and most importantly, but incompletely understood, prolonged long-term survival in a fraction of the patients. In relapsed patients, we developed an immunotherapy schedule and we categorized patients into clinically defined risk profiles. We learned how to combine immunotherapy with standard multimodal treatment strategies for newly diagnosed glioblastoma multiforme patients. The developmental program allows further improvements related to newest scientific insights. Finally, we developed a mode of care within academic centers to organize cell-based therapies for experimental clinical trials in a large number of patients.
Collapse
|
40
|
Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, Naranjo A, Starr R, Wagner J, Wright C, Zhai Y, Bading JR, Ressler JA, Portnow J, D'Apuzzo M, Forman SJ, Jensen MC. Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin Cancer Res 2015; 21:4062-72. [PMID: 26059190 DOI: 10.1158/1078-0432.ccr-15-0428] [Citation(s) in RCA: 535] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/17/2015] [Indexed: 01/27/2023]
Abstract
PURPOSE A first-in-human pilot safety and feasibility trial evaluating chimeric antigen receptor (CAR)-engineered, autologous primary human CD8(+) cytotoxic T lymphocytes (CTL) targeting IL13Rα2 for the treatment of recurrent glioblastoma (GBM). EXPERIMENTAL DESIGN Three patients with recurrent GBM were treated with IL13(E13Y)-zetakine CD8(+) CTL targeting IL13Rα2. Patients received up to 12 local infusions at a maximum dose of 10(8) CAR-engineered T cells via a catheter/reservoir system. RESULTS We demonstrate the feasibility of manufacturing sufficient numbers of autologous CTL clones expressing an IL13(E13Y)-zetakine CAR for redirected HLA-independent IL13Rα2-specific effector function for a cohort of patients diagnosed with GBM. Intracranial delivery of the IL13-zetakine(+) CTL clones into the resection cavity of 3 patients with recurrent disease was well-tolerated, with manageable temporary brain inflammation. Following infusion of IL13-zetakine(+) CTLs, evidence for transient anti-glioma responses was observed in 2 of the patients. Analysis of tumor tissue from 1 patient before and after T-cell therapy suggested reduced overall IL13Rα2 expression within the tumor following treatment. MRI analysis of another patient indicated an increase in tumor necrotic volume at the site of IL13-zetakine(+) T-cell administration. CONCLUSIONS These findings provide promising first-in-human clinical experience for intracranial administration of IL13Rα2-specific CAR T cells for the treatment of GBM, establishing a foundation on which future refinements of adoptive CAR T-cell therapies can be applied.
Collapse
Affiliation(s)
- Christine E Brown
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California.
| | - Behnam Badie
- Department of Neurosurgery, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Michael E Barish
- Department of Neurosciences, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Lihong Weng
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Julie R Ostberg
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Wen-Chung Chang
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Araceli Naranjo
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Renate Starr
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Jamie Wagner
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Christine Wright
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Yubo Zhai
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - James R Bading
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Julie A Ressler
- Department of Diagnostic Radiology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Jana Portnow
- Department of Medical Oncology and Therapeutics Research, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Massimo D'Apuzzo
- Department of Pathology, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Stephen J Forman
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California. Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California
| | - Michael C Jensen
- Center for Childhood Cancer, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
41
|
Interleukin-13 receptor alpha 2-targeted glioblastoma immunotherapy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:952128. [PMID: 25247196 PMCID: PMC4163479 DOI: 10.1155/2014/952128] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/05/2014] [Indexed: 01/23/2023]
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor, and despite several refinements in its multimodal management, generally has very poor prognosis. Targeted immunotherapy is an emerging field of research that shows great promise in the treatment of GBM. One of the most extensively studied targets is the interleukin-13 receptor alpha chain variant 2 (IL13Rα2). Its selective expression on GBM, discovered almost two decades ago, has been a target for therapy ever since. Immunotherapeutic strategies have been developed targeting IL13Rα2, including monoclonal antibodies as well as cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-expressing T cells. Advanced therapeutic development has led to the completion of several clinical trials with promising outcomes. In this review, we will discuss the recent advances in the IL13Rα2-targeted immunotherapy and evaluate the most promising strategy for targeted GBM immunotherapy.
Collapse
|
42
|
van Gool S. Immunotherapy for high-grade glioma: how to go beyond Phase I/II clinical trials. Immunotherapy 2014; 5:1043-6. [PMID: 24138559 DOI: 10.2217/imt.13.86] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Evaluation of: Lasky JL 3rd, Panosyan EH, Plant A et al. Autologous tumor lysate-pulsed dendritic cell immunotherapy for pediatric patients with newly diagnosed or recurrent high-grade gliomas. Anticancer Res. 33, 2047-2056 (2013). Immunotherapy for children and adults with high-grade glioma (HGG) is an emerging innovative treatment approach, which aims at stimulating the body's own immune system against HGG by using autologous dendritic cells pulsed with autologous tumor lysate as a therapeutic vaccine. This is the third report on immunotherapy for HGG in children, bringing additional knowledge and experience to the scientific community. However, at the same time, this and other manuscripts urge for the next step in treatment development.
Collapse
Affiliation(s)
- Stefaan van Gool
- Laboratory of Pediatric Immunology, Pediatric Neuro-Oncology, University of Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
43
|
Lowenstein PR, Castro MG. The value of EGFRvIII as the target for glioma vaccines. Am Soc Clin Oncol Educ Book 2014:42-50. [PMID: 24857059 DOI: 10.14694/edbook_am.2014.34.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Malignant brain tumors continue to be rapidly progressive and resistant to most treatments. Even with state-of-the-art standard of care (surgery, chemotherapy, and radiotherapy) long-term survival in the last 80 years improved from 6 to 15 months. Improved imaging has also likely contributed to prolonged survival. Immunotherapy for cancer dates back to publications from 1742. The central idea is that the immune system can detect and eliminate foreign antigens, either from infectious agents or tumors, and thus could be therapeutic in brain tumors. Recent introduction of immune modulators of cytotoxic T-lymphocyte antigen (CTLA)-4 and programmed cell death 1/programmed cell death 1 ligand (PD-1/PDL1) add much excitement to this field. For brain tumors, there are several ongoing phase I and III trials to determine whether any of the current immunotherapy approaches can demonstrate activity in randomized, controlled double-blinded trials-with ongoing and historical trials presented in tables within the manuscript. Immunotherapy has explored the use of various types of antigens (obtained either from homogenates of patients' tumors or synthetically produced), and various immunization procedures and adjuvants. Glioma antigens have also been isolated from the patients' own tumor, then produced in vitro (for example the glioma antigen EGFRvIII), and used to immunize patients directly, or with carriers such as dendritic cells with or without additional adjuvants. Several of these practical approaches are currently in phase III trials. Remaining challenges are how to increase the percentage of complete responses and response duration, and the enigmatic absence of an almost total lack of adverse brain inflammation following immunization of brain tumor patients, as has been observed following immunization against brain antigens in other diseases, such as Alzheimer's Disease.
Collapse
Affiliation(s)
- Pedro R Lowenstein
- From the Department of Neurosurgery and Cell and Developmental Biology, Graduate Program in Immunology, and Graduate Program in Cancer Biology, The University of Michigan Comprehensive Cancer Center, The University of Michigan School of Medicine, Ann Arbor, MI
| | - Maria G Castro
- From the Department of Neurosurgery and Cell and Developmental Biology, Graduate Program in Immunology, and Graduate Program in Cancer Biology, The University of Michigan Comprehensive Cancer Center, The University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
44
|
T cells redirected to interleukin-13Rα2 with interleukin-13 mutein--chimeric antigen receptors have anti-glioma activity but also recognize interleukin-13Rα1. Cytotherapy 2014; 16:1121-31. [PMID: 24841514 DOI: 10.1016/j.jcyt.2014.02.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 02/18/2014] [Accepted: 02/21/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND AIMS Outcomes for patients with glioblastoma remain poor despite aggressive multimodal therapy. Immunotherapy with genetically modified T cells expressing chimeric antigen receptors (CARs) targeting interleukin (IL) 13Rα2, human epidermal growth factor receptor 2, epidermal growth factor variant III or erythropoietin-producing hepatocellular carcinoma A2 has shown promise for the treatment of glioma in preclinical models. On the basis of IL13Rα2 immunotoxins that contain IL13 molecules with one or two amino acid substitutions (IL13 muteins) to confer specificity to IL13Rα2, investigators have constructed CARS with IL13 muteins as antigen-binding domains. Whereas the specificity of IL13 muteins in the context of immunotoxins is well characterized, limited information is available for CAR T cells. METHODS We constructed four second-generation CARs with IL13 muteins with one or two amino acid substitutions, and evaluated the effector function of IL13-mutein CAR T cells in vitro and in vivo. RESULTS T cells expressing all four CARs recognized IL13Rα1 or IL13Rα2 recombinant protein in contrast to control protein (IL4R) as judged by interferon-γ production. IL13 protein produced significantly more IL2, indicating that IL13 mutein-CAR T cells have a higher affinity to IL13Rα2 than to IL13Rα1. In cytotoxicity assays, CAR T cells killed IL13Rα1- and/or IL13Rα2-positive cells in contrast to IL13Rα1- and IL13Rα2-negative controls. Although we observed no significant differences between IL13 mutein-CAR T cells in vitro, only T cells expressing IL13 mutein-CARs with an E13K amino acid substitution had anti-tumor activity in vivo that resulted in a survival advantage of treated animals. CONCLUSIONS Our study highlights that the specificity/avidity of ligands is context-dependent and that evaluating CAR T cells in preclinical animal model is critical to assess their potential benefit.
Collapse
|
45
|
Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro Oncol 2014; 16:1304-12. [PMID: 24723564 DOI: 10.1093/neuonc/nou045] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains one of the most lethal primary brain tumors despite surgical and therapeutic advancements. Targeted therapies of neoplastic diseases, including GBM, have received a great deal of interest in recent years. A highly studied target of GBM is interleukin-13 receptor α chain variant 2 (IL13Rα2). Targeted therapies against IL13Rα2 in GBM include fusion chimera proteins of IL-13 and bacterial toxins, nanoparticles, and oncolytic viruses. In addition, immunotherapies have been developed using monoclonal antibodies and cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-modified T cells. Advanced therapeutic development has led to the completion of phase I clinical trials for chimeric antigen receptor-modified T cells and phase III clinical trials for IL-13-conjugated bacterial toxin, with promising outcomes. Selective expression of IL13Rα2 on tumor cells, while absent in the surrounding normal brain tissue, has motivated continued study of IL13Rα2 as an important candidate for targeted glioma therapy. Here, we review the preclinical and clinical studies targeting IL13Rα2 in GBM and discuss new advances and promising applications.
Collapse
Affiliation(s)
- Bart Thaci
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Christine E Brown
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Emanuela Binello
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Katherine Werbaneth
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Prakash Sampath
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Sadhak Sengupta
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| |
Collapse
|
46
|
Dejaegher J, Van Gool S, De Vleeschouwer S. Dendritic cell vaccination for glioblastoma multiforme: review with focus on predictive factors for treatment response. Immunotargets Ther 2014; 3:55-66. [PMID: 27471700 PMCID: PMC4918234 DOI: 10.2147/itt.s40121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and most aggressive type of primary brain cancer. Since median overall survival with multimodal standard therapy is only 15 months, there is a clear need for additional effective and long-lasting treatments. Dendritic cell (DC) vaccination is an experimental immunotherapy being tested in several Phase I and Phase II clinical trials. In these trials, safety and feasibility have been proven, and promising clinical results have been reported. On the other hand, it is becoming clear that not every GBM patient will benefit from this highly personalized treatment. Defining the subgroup of patients likely to respond to DC vaccination will position this option correctly amongst other new GBM treatment modalities, and pave the way to incorporation in standard therapy. This review provides an overview of GBM treatment options and focuses on the currently known prognostic and predictive factors for response to DC vaccination. In this way, it will provide the clinician with the theoretical background to refer patients who might benefit from this treatment.
Collapse
Affiliation(s)
| | - Stefaan Van Gool
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
47
|
Chandramohan V, Mitchell DA, Johnson LA, Sampson JH, Bigner DD. Antibody, T-cell and dendritic cell immunotherapy for malignant brain tumors. Future Oncol 2014; 9:977-90. [PMID: 23837761 DOI: 10.2217/fon.13.47] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Modest improvement in brain tumor patient survival has been achieved through advances in surgical, adjuvant radiation and chemotherapeutic strategies. However, these traditional approaches have been unsuccessful in permanently controlling these aggressive tumors, with recurrence being quite common. Hence, there is a need for novel therapeutic approaches that specifically target the molecularly diverse brain tumor cell population. The ability of the immune system to recognize altered tumor cells while avoiding surrounding normal cells offers an enormous advantage over the nonspecific nature of the conventional treatment schemes. Therefore, immunotherapy represents a promising approach that may supplement the standard therapies in eliminating the residual brain tumor cells. This review summarizes different immunotherapeutic approaches currently being tested for malignant brain tumor treatment.
Collapse
|
48
|
Abstract
Just like any other effective immunization in medicine, cancer vaccines need to have antigens with particular specificity and immunostimulatory features, the immune responses to be elicited in the body, and therapeutic effect-regression or prevention of the cancer-must be meaningful and clinically observable. There are many choices for cancer antigens, such as tissue-specific proteins, cancer-specific proteins, class I- or class II-restricted peptides derived from those, or in situ and whole-cell-derived products are some examples. Another translational issue is that cancer patients are heterogeneous with respect to the extent to which the immune system is already activated with potential to impact the tumor growth or, conversely, the extent to which the immune system has been impaired through a prior and ongoing interaction with the tumor. Conventional or immunologic tests have potential to define a subset of patients with better chance or response, so that particular vaccines can be tested. Treatment of cancer patients is expensive, and trials are slow. To meet these challenges in practical terms will require not only careful scientific technical work for product development, coordination with clinicians to define patient subsets with diseases that can show responses, but also a comprehensive, practical implementation so that we can unlock the full potential of anticancer vaccines.
Collapse
|
49
|
Ahn BJ, Pollack IF, Okada H. Immune-checkpoint blockade and active immunotherapy for glioma. Cancers (Basel) 2013; 5:1379-412. [PMID: 24202450 PMCID: PMC3875944 DOI: 10.3390/cancers5041379] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 10/24/2013] [Accepted: 10/24/2013] [Indexed: 02/01/2023] Open
Abstract
Cancer immunotherapy has made tremendous progress, including promising results in patients with malignant gliomas. Nonetheless, the immunological microenvironment of the brain and tumors arising therein is still believed to be suboptimal for sufficient antitumor immune responses for a variety of reasons, including the operation of “immune-checkpoint” mechanisms. While these mechanisms prevent autoimmunity in physiological conditions, malignant tumors, including brain tumors, actively employ these mechanisms to evade from immunological attacks. Development of agents designed to unblock these checkpoint steps is currently one of the most active areas of cancer research. In this review, we summarize recent progresses in the field of brain tumor immunology with particular foci in the area of immune-checkpoint mechanisms and development of active immunotherapy strategies. In the last decade, a number of specific monoclonal antibodies designed to block immune-checkpoint mechanisms have been developed and show efficacy in other cancers, such as melanoma. On the other hand, active immunotherapy approaches, such as vaccines, have shown encouraging outcomes. We believe that development of effective immunotherapy approaches should ultimately integrate those checkpoint-blockade agents to enhance the efficacy of therapeutic approaches. With these agents available, it is going to be quite an exciting time in the field. The eventual success of immunotherapies for brain tumors will be dependent upon not only an in-depth understanding of immunology behind the brain and brain tumors, but also collaboration and teamwork for the development of novel trials that address multiple layers of immunological challenges in gliomas.
Collapse
Affiliation(s)
- Brian J. Ahn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; E-Mail:
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; E-Mail:
| | - Ian F. Pollack
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; E-Mail:
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hideho Okada
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; E-Mail:
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; E-Mail:
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-412-623-3111; Fax: +1-412-623-1415
| |
Collapse
|
50
|
Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2013; 2:e25771. [PMID: 24286020 PMCID: PMC3841205 DOI: 10.4161/onci.25771] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) occupy a privileged position at the interface between innate and adaptive immunity, orchestrating a large panel of responses to both physiological and pathological cues. In particular, whereas the presentation of antigens by immature DCs generally results in the development of immunological tolerance, mature DCs are capable of priming robust, and hence therapeutically relevant, adaptive immune responses. In line with this notion, functional defects in the DC compartment have been shown to etiologically contribute to pathological conditions including (but perhaps not limited to) infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. Thus, the possibility of harnessing the elevated immunological potential of DCs for anticancer therapy has attracted considerable interest from both researchers and clinicians over the last decade. Alongside, several methods have been developed not only to isolate DCs from cancer patients, expand them, load them with tumor-associated antigens and hence generate highly immunogenic clinical grade infusion products, but also to directly target DCs in vivo. This intense experimental effort has culminated in 2010 with the approval by the US FDA of a DC-based preparation (sipuleucel-T, Provenge®) for the treatment of asymptomatic or minimally symptomatic metastatic castration-refractory prostate cancer. As an update to the latest Trial Watch dealing with this exciting field of research (October 2012), here we summarize recent advances in DC-based anticancer regimens, covering both high-impact studies that have been published during the last 13 mo and clinical trials that have been launched in the same period to assess the antineoplastic potential of this variant of cellular immunotherapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|