1
|
Turk OM, Woodall RC, Gutova M, Brown CE, Rockne RC, Munson JM. Delivery strategies for cell-based therapies in the brain: overcoming multiple barriers. Drug Deliv Transl Res 2021; 11:2448-2467. [PMID: 34718958 PMCID: PMC8987295 DOI: 10.1007/s13346-021-01079-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Cell-based therapies to the brain are promising for the treatment of multiple brain disorders including neurodegeneration and cancers. In order to access the brain parenchyma, there are multiple physiological barriers that must be overcome depending on the route of delivery. Specifically, the blood-brain barrier has been a major difficulty in drug delivery for decades, and it still presents a challenge for the delivery of therapeutic cells. Other barriers, including the blood-cerebrospinal fluid barrier and lymphatic-brain barrier, are less explored, but may offer specific challenges or opportunities for therapeutic delivery. Here we discuss the barriers to the brain and the strategies currently in place to deliver cell-based therapies, including engineered T cells, dendritic cells, and stem cells, to treat diseases. With a particular focus on cancers, we also highlight the current ongoing clinical trials that use cell-based therapies to treat disease, many of which show promise at treating some of the deadliest illnesses.
Collapse
Affiliation(s)
- Olivia M Turk
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Ryan C Woodall
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope, Duarte, CA, USA
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative Medicine, City of Hope, Duarte, CA, USA
| | - Christine E Brown
- Departments of Hematology & Hematopoietic Cell Transplantation and Immuno-Oncology, City of Hope, Duarte, CA, USA
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope, Duarte, CA, USA
| | - Jennifer M Munson
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, USA.
| |
Collapse
|
2
|
Immunotherapy Approaches for Pediatric CNS Tumors and Associated Neurotoxicity. Pediatr Neurol 2020; 107:7-15. [PMID: 32113728 DOI: 10.1016/j.pediatrneurol.2020.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/07/2019] [Accepted: 01/15/2020] [Indexed: 12/24/2022]
Abstract
Treatment for brain tumors has recently shifted to using the power of the immune system to destroy cancer cells with promising results. Many immunotherapeutic approaches that have been used in adults, including checkpoint inhibitors, vaccine therapy, adoptive immunotherapy, such as chimeric antigen receptor T cell therapy, and viral therapy, are now being evaluated in children. Although these treatments work through different mechanisms, they all activate the immune system and can result in inflammation at the site of disease. This can be especially problematic in the confined area of the brain causing potentially severe neurological side effects, which are of special concern in children with central nervous system malignancies. Steroids can be helpful in the management of neurological complications but carry the risk of making immunotherapeutic approaches less effective. Alternative therapeutic interventions to mitigate side effects are being evaluated. This review describes the most common immunotherapeutic modalities that are now under study for the treatment of pediatric brain tumors, their rationale, associated neurotoxicities, and current management.
Collapse
|
3
|
Bioengineered recombinant vault nanoparticles coupled with NY-ESO-1 glioma-associated antigens induce maturation of native dendritic cells. J Neurooncol 2020; 148:1-7. [DOI: 10.1007/s11060-020-03472-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/23/2020] [Indexed: 11/27/2022]
|
4
|
Kabir TF, Kunos CA, Villano JL, Chauhan A. Immunotherapy for Medulloblastoma: Current Perspectives. Immunotargets Ther 2020; 9:57-77. [PMID: 32368525 PMCID: PMC7182450 DOI: 10.2147/itt.s198162] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022] Open
Abstract
Background Immune-mediated therapies have transformed the treatment of metastatic melanoma and renal, bladder, and both small and non-small cell lung carcinomas. However, immunotherapy is yet to demonstrate dramatic results in brain tumors like medulloblastoma for a variety of reasons. Recent pre-clinical and early phase human trials provide encouraging results that may overcome the challenges of central nervous system (CNS) tumors, which include the intrinsic immunosuppressive properties of these cancers, a lack of antigen targets, antigenic variability, and the immune-restrictive site of the CNS. These studies highlight the growing potential of immunotherapy to treat patients with medulloblastoma, a disease that is a frequent cause of morbidity and mortality to children and young adults. Methods We conducted an inclusive review of the PubMed-indexed literature and studies listed in clinicaltrials.gov using combinations of the keywords medulloblastoma, immunotherapy, CNS tumors, brain tumors, vaccines, oncolytic virus, natural killer, and CAR T to identify trials evaluating immunotherapy in preclinical experiments or in patients with medulloblastoma. Given a limited number of investigations using immunotherapy to treat patients with medulloblastoma, 24 studies were selected for final analysis and manuscript citation. Results This review presents results from pre-clinical studies in medulloblastoma cell lines, animal models, and the limited trials involving human patients. Conclusion From our review, we suggest that cancer vaccines, oncolytic viral therapy, natural killer cells, and CAR T therapy hold promise against the innate immunosuppressive properties of medulloblastoma in order to prolong survival. There is an unmet need for immunotherapy regimens that target overexpressed antigens in medulloblastoma tumors. We advocate for more combination treatment clinical trials using conventional surgical and radiochemotherapy approaches in the near-term clinical development.
Collapse
Affiliation(s)
- Tanvir F Kabir
- Department of Internal Medicine, University of Louisville, Louisville, KY, USA
| | - Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - John L Villano
- Department of Internal Medicine-Medical Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Aman Chauhan
- Department of Internal Medicine-Medical Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
5
|
Leibold AT, Monaco GN, Dey M. The role of the immune system in brain metastasis. CURRENT NEUROBIOLOGY 2019; 10:33-48. [PMID: 31097897 PMCID: PMC6513348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Metastatic brain tumors are the most common brain tumors in adults. With numerous successful advancements in systemic treatment of most common cancer types, brain metastasis is becoming increasingly important in the overall prognosis of cancer patients. Brain metastasis of peripheral tumor is the result of complex interplay of primary tumor, immune system and central nervous system microenvironment. Once formed, brain metastases hide behind the blood brain barrier and become inaccessible to chemotherapies that are otherwise successful in targeting systemic cancer. The approval of immune checkpoint inhibitors for several common cancers such as advanced melanoma and lung cancers brings with it the opportunity and obligation to further understand the mechanisms of immunosuppression by tumors that spread to the brain as well as the interaction between the brain environment and tumor microenvironment. In this review paper we define the central role of the immune system in the development of brain metastases. We performed a comprehensive review of the literature to outline the molecular mechanisms of immunosuppression used by tumors and how the immune system interacts with the central nervous system to facilitate brain metastasis. In particular we discuss the tumor-type-specific mechanisms of metastasis of cancers that preferentially metastasize to the brain as well as the therapies that effectively modulate the immune response, such as immune checkpoint inhibitors and vaccines.
Collapse
Affiliation(s)
- Adam T Leibold
- Department of Neurosurgery, Indiana University School of Medicine, IU Simon Cancer Center, Indiana University, Purdue University Indianapolis, Indiana, USA
| | - Gina N Monaco
- Department of Neurosurgery, Indiana University School of Medicine, IU Simon Cancer Center, Indiana University, Purdue University Indianapolis, Indiana, USA
| | - Mahua Dey
- Department of Neurosurgery, Indiana University School of Medicine, IU Simon Cancer Center, Indiana University, Purdue University Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Abu Arja MH, Bouffet E, Finlay JL, AbdelBaki MS. Critical review of the management of primary central nervous nongerminomatous germ cell tumors. Pediatr Blood Cancer 2019; 66:e27658. [PMID: 30767415 DOI: 10.1002/pbc.27658] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/06/2019] [Accepted: 01/27/2019] [Indexed: 12/28/2022]
Abstract
Multimodal strategies have significantly improved the outcomes for patients with central nervous system nongerminomatous germ cell tumors. Two large cooperative group studies have recently reported much improved outcomes compared with historical series. However, a substantial proportion of patients still attain inadequate responses to initial chemotherapy prior to irradiation, with adverse impact upon survival; optimal induction chemotherapy regimens and radiotherapy strategies are as yet unidentified. Outcomes for patients with relapsed disease remain poor. There is an obvious need to incorporate molecular studies within prospective clinical trials that will likely lead to the incorporation of targeted, more effective future treatment strategies.
Collapse
Affiliation(s)
- Mohammad H Abu Arja
- The Division of Hematology, Oncology and Blood and Marrow Transplant, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, USA
| | - Eric Bouffet
- The Division of Hematology, Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jonathan L Finlay
- The Division of Hematology, Oncology and Blood and Marrow Transplant, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, USA
| | - Mohamed S AbdelBaki
- The Division of Hematology, Oncology and Blood and Marrow Transplant, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Chammas M, Saadeh F, Maaliki M, Assi H. Therapeutic Interventions in Adult Low-Grade Gliomas. J Clin Neurol 2018; 15:1-8. [PMID: 30198226 PMCID: PMC6325362 DOI: 10.3988/jcn.2019.15.1.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 01/05/2023] Open
Abstract
Treating adult low-grade gliomas (LGGs) is particularly challenging due to the highly infiltrative nature of this type of brain cancer. Although surgery, radiotherapy, and chemotherapy are the mainstay treatment modalities for LGGs, the optimal combination management plan for a particular patient based on individual symptoms and the risk of treatment-induced toxicity remains unclear. This review highlights the competency and limitations of standard treatment options while providing an essential therapeutic update regarding current clinical trials aimed at implementing targeted therapies with morbidity rates lower than those for current LGG treatments and also augmenting the killing of cancerous cells while maintaining an improved quality of life.
Collapse
Affiliation(s)
- Majid Chammas
- American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Fadi Saadeh
- American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Maya Maaliki
- American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Hazem Assi
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
8
|
Brezovakova V, Valachova B, Hanes J, Novak M, Jadhav S. Dendritic Cells as an Alternate Approach for Treatment of Neurodegenerative Disorders. Cell Mol Neurobiol 2018; 38:1207-1214. [PMID: 29948552 PMCID: PMC11481983 DOI: 10.1007/s10571-018-0598-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/11/2018] [Indexed: 12/17/2022]
Abstract
Despite years of research, Alzheimer's disease (AD) remains incurable and thus poses a major health challenge in coming years. This neurodegenerative disease belongs to a heterogeneous group of human tauopathies, characterized by the extracellular deposition of beta amyloid-Aβ and intracellular accumulation of tau protein in neuronal and glial cells, whereby tau pathology best correlates with disease progression. For decades, several disease-modifying agents were brought to clinical studies with promising efficacy in preclinical trials; however, all of the subsequent clinical trials failed. Therefore, the pursuit for therapeutic agents for the treatment of AD and other tauopathies still continue. Recent evidences show previously unidentified role of peripheral immune system in regulating the inflammatory status of the brain, mainly the dendritic cells. A decrease in functionality and count of dendritic cells has been observed in Alzheimer's disease. Here, we discuss a potential role of dendritic cell-based vaccines as therapeutic approach in ameliorating disease pathogenesis in AD and other tauopathies.
Collapse
Affiliation(s)
- Veronika Brezovakova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dúbravská cesta 9, 845 10, Bratislava, Slovak Republic
| | - Bernadeta Valachova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dúbravská cesta 9, 845 10, Bratislava, Slovak Republic
- Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dúbravská cesta 9, 845 10, Bratislava, Slovak Republic
- Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dúbravská cesta 9, 845 10, Bratislava, Slovak Republic
- Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dúbravská cesta 9, 845 10, Bratislava, Slovak Republic.
- Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic.
| |
Collapse
|
9
|
Su XW, Lu G, Leung CK, Liu Q, Li Y, Tsang KS, Zhao SD, Chan DTM, Kung HF, Poon WS. miR-181d regulates human dendritic cell maturation through NF-κB pathway. Cell Prolif 2017; 50. [PMID: 28731516 DOI: 10.1111/cpr.12358] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 05/15/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES MicroRNAs (miRNAs) are considered as the cellular regulators which post-transcriptionally modulate gene expression in diverse biological processes including cell development and immunity. In this study, we investigated functions of miR-181d in dendritic cells (DCs) maturation, and the underlying mechanisms were also explored. MATERIALS AND METHODS Here we did the miRNA screening in human DCs in response to lipopolysaccharides (LPS) by quantitative real-time PCR (qRT-PCR). The expressions of DCs maturation markers were measured after miRNA mimics transfections. The pharmacological inhibitors of signalling pathways were applied to examine miR-181d effect on DCs maturation by Western blot. Luciferase assay and mixed lymphocyte reaction (MLR) were also performed to reveal the target gene of miR-181d and test the viability of T cells treated with miR-181d transfected DCs. RESULTS Overexpression of miR-181d per se is sufficient to promote DCs maturation, and up-regulate CD80 and CD83 expressions without LPS. Besides, we showed that miR-181d activated NF-κB pathway and also promoted the expression of pro-inflammatory cytokine IL12 and TNF-α. Inhibition of NF-κB pathway suppressed DCs maturation. Luciferase reporter assay and target gene knockdown assay indicated that miR-181d targets regulator cylindromatosis (CYLD), a primary negative regulator of NF-κB pathway. MLR assay showed that miR-181d-transfected DCs could promote T-cell proliferation than iDCs in vitro. CONCLUSION Our study demonstrates that miR-181d is required for DCs maturation through the activation of NF-κB pathway by targeting CYLD.
Collapse
Affiliation(s)
- Xian Wei Su
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Lu
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Kwan Leung
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Qiang Liu
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Yi Li
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kam Sze Tsang
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Shi Dou Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Danny Tat Ming Chan
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Otto Wong Brain Tumour Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Hsiang Fu Kung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Otto Wong Brain Tumour Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
10
|
Abstract
Malignant brain tumors represent one of the most devastating forms of cancer with abject survival rates that have not changed in the past 60years. This is partly because the brain is a critical organ, and poses unique anatomical, physiological, and immunological barriers. The unique interplay of these barriers also provides an opportunity for creative engineering solutions. Cancer immunotherapy, a means of harnessing the host immune system for anti-tumor efficacy, is becoming a standard approach for treating many cancers. However, its use in brain tumors is not widespread. This review discusses the current approaches, and hurdles to these approaches in treating brain tumors, with a focus on immunotherapies. We identify critical barriers to immunoengineering brain tumor therapies and discuss possible solutions to these challenges.
Collapse
|
11
|
Gardeck AM, Sheehan J, Low WC. Immune and viral therapies for malignant primary brain tumors. Expert Opin Biol Ther 2017; 17:457-474. [DOI: 10.1080/14712598.2017.1296132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andrew M. Gardeck
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Sheehan
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
12
|
Dendritic Cell-Based Immunotherapy Treatment for Glioblastoma Multiforme. BIOMED RESEARCH INTERNATIONAL 2015; 2015:717530. [PMID: 26167495 PMCID: PMC4488155 DOI: 10.1155/2015/717530] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/08/2015] [Indexed: 12/23/2022]
Abstract
Glioblastoma multiforme (GBM) is the most malignant glioma and patients diagnosed with this disease had poor outcomes even treated with the combination of conventional treatment (surgery, chemotherapy, and radiation). Dendritic cells (DCs) are the most powerful antigen presenting cells and DC-based vaccination has the potential to target and eliminate GBM cells and enhance the responses of these cells to the existing therapies with minimal damage to the healthy tissues around them. It can enhance recognition of GBM cells by the patients' immune system and activate vast, potent, and long-lasting immune reactions to eliminate them. Therefore, this therapy can prolong the survival of GBM patients and has wide and bright future in the treatment of GBM. Also, the efficacy of this therapy can be strengthened in several ways at some degree: the manipulation of immune regulatory components or costimulatory molecules on DCs; the appropriate choices of antigens for loading to enhance the effectiveness of the therapy; regulation of positive regulators or negative regulators in GBM microenvironment.
Collapse
|
13
|
Ung N, Yang I. Nanotechnology to augment immunotherapy for the treatment of glioblastoma multiforme. J Neurooncol 2015; 123:473-81. [DOI: 10.1007/s11060-015-1814-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 05/20/2015] [Indexed: 12/30/2022]
|
14
|
Brandes AA, Bartolotti M, Marucci G, Ghimenton C, Agati R, Fioravanti A, Mascarin M, Volpin L, Ammannati F, Masotto B, Gardiman MP, De Biase D, Tallini G, Crisi G, Bartolini S, Franceschi E. New perspectives in the treatment of adult medulloblastoma in the era of molecular oncology. Crit Rev Oncol Hematol 2015; 94:348-359. [PMID: 25600839 DOI: 10.1016/j.critrevonc.2014.12.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 12/10/2014] [Accepted: 12/23/2014] [Indexed: 02/08/2023] Open
Abstract
Medulloblastoma is the most common central nervous system tumor in children, while it is extremely rare in adults. Multimodal treatment involving surgery, radiotherapy and chemotherapy can improve the prognosis of this disease, and recent advances in molecular biology have allowed the identification of molecular subgroups (WNT, SHH, Groups 3 and 4), each of which have different cytogenetic, mutational and gene expression signatures, demographics, histology and prognosis. The present review focuses on the state of the art for adult medulloblastoma treatment and on novel molecular advances and their future implications in the treatment of this disease.
Collapse
Affiliation(s)
- Alba A Brandes
- Department of Medical Oncology, Bellaria-Maggiore Hospitals, Azienda USL - IRCCS Institute of Neurological Sciences, Bologna, Italy.
| | - Marco Bartolotti
- Department of Medical Oncology, Bellaria-Maggiore Hospitals, Azienda USL - IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Gianluca Marucci
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), University, of Bologna, Section of Pathology, M. Malpighi, Bellaria Hospital, Bologna, Italy
| | | | - Raffaele Agati
- Department of Neuroradiology, Bellaria-Maggiore Hospitals, Azienda USL - IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Antonio Fioravanti
- Department of Neurosurgery, Bellaria Hospital - IRCCS Institute of Neurological Sciences, Azienda USL, Bologna, Italy
| | | | - Lorenzo Volpin
- Department of Neuroscience and Neurosurgery, San Bortolo Hospital, Vicenza, Italy
| | - Franco Ammannati
- Department of Neurosurgery I, Careggi University Hospital, Firenze, Italy
| | - Barbara Masotto
- Section of Neurosurgery, Department of Neuroscience, University of Verona, Verona, Italy
| | - Marina Paola Gardiman
- Surgical Pathology & Cytopathology Unit, Department of Medicine (DIMED), University Hospital, Padova, Italy
| | - Dario De Biase
- Department of Medicine (DIMES) - Anatomic Pathology Unit, Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Giovanni Tallini
- Department of Medicine (DIMES) - Anatomic Pathology Unit, Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Girolamo Crisi
- Department of Neuroradiology, Parma University Hospital, Parma, Italy
| | - Stefania Bartolini
- Department of Medical Oncology, Bellaria-Maggiore Hospitals, Azienda USL - IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Enrico Franceschi
- Department of Medical Oncology, Bellaria-Maggiore Hospitals, Azienda USL - IRCCS Institute of Neurological Sciences, Bologna, Italy
| |
Collapse
|
15
|
Baker GJ, Chockley P, Yadav VN, Doherty R, Ritt M, Sivaramakrishnan S, Castro MG, Lowenstein PR. Natural killer cells eradicate galectin-1-deficient glioma in the absence of adaptive immunity. Cancer Res 2014; 74:5079-90. [PMID: 25038230 DOI: 10.1158/0008-5472.can-14-1203] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Natural killer (NK) cells safeguard against early tumor formation by destroying transformed target cells in a process referred to as NK immune surveillance. However, the immune escape mechanisms used by malignant brain tumors to subvert this innate type of immune surveillance remain unclear. Here we show that malignant glioma cells suppress NK immune surveillance by overexpressing the β-galactoside-binding lectin galectin-1. Conversely, galectin-1-deficient glioma cells could be eradicated by host NK cells before the initiation of an antitumor T-cell response. In vitro experiments demonstrated that galectin-1-deficient GL26-Cit glioma cells are ∼3-fold more sensitive to NK-mediated tumor lysis than galectin-1-expressing cells. Our findings suggest that galectin-1 suppression in human glioma could improve patient survival by restoring NK immune surveillance that can eradicate glioma cells. Cancer Res; 74(18); 5079-90. ©2014 AACR.
Collapse
Affiliation(s)
- Gregory J Baker
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Peter Chockley
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Viveka Nand Yadav
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Robert Doherty
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Michael Ritt
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Sivaraj Sivaramakrishnan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan. Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
16
|
Chistiakov DA, Chekhonin VP. Extracellular vesicles shed by glioma cells: pathogenic role and clinical value. Tumour Biol 2014; 35:8425-38. [DOI: 10.1007/s13277-014-2262-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/18/2014] [Indexed: 02/03/2023] Open
|
17
|
Immunomodulatory effects of hemagglutinin- (HA-) modified A20 B-cell lymphoma expanded as a brain tumor on adoptively transferred HA-Specific CD4+ T cells. ScientificWorldJournal 2014; 2014:165265. [PMID: 24693228 PMCID: PMC3947776 DOI: 10.1155/2014/165265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 12/06/2013] [Indexed: 11/17/2022] Open
Abstract
Previously, the mouse A20 B-cell lymphoma engineered to express hemagglutinin (HA) antigen (A20HA) was used as a systemic tumor model. In this work, we used the A20HA cells as a brain tumor. HA-specific CD4(+) T cells were transferred intravenously in a tail vein 5 days after A20HA intracranial inoculation and analyzed on days 2, 9, and 16 after the adoptive transfer by different methods. The transferred cells demonstrated state of activation as early as day 2 after the adoptive transfer and most the of viable HA-specific cells became anergic on day 16. Additionally, symptoms of systemic immunosuppression were observed in mice with massive brain tumors at a late stage of the brain tumor progression (days 20-24 after the A20HA inoculation). Despite that, a deal of HA-specific CD4(+) T cells kept the functional activity even at the late stage of A20HA tumor growth. The activated HA-specific CD4(+) T cells were found also in the brain of brain-tumor-bearing mice. These cells were still responding to reactivation with HA-peptide in vitro. Our data support an idea about sufficient role of both the tumor-specific and -nonspecific mechanisms inducing immunosuppression in cancer patients.
Collapse
|
18
|
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
|
19
|
Zhang H, Tian M, Xiu C, Wang Y, Tang G. Enhancement of antitumor activity by combination of tumor lysate-pulsed dendritic cells and celecoxib in a rat glioma model. Oncol Res 2013; 20:447-55. [PMID: 24308155 DOI: 10.3727/096504013x13685487925176] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Using dendritic cell (DC)-based vaccines for treatment of gliomas has emerged as a meaningful and feasible treatment approach for inducing long-term survival, but this approach so far has failed to generate significant clinical responses. In the present study, we demonstrated that glioma lysate-pulsed DCs in combination with celecoxib, a selective cyclooxygenase 2 (COX-2) inhibitor, showed more significantly enhanced antitumor activity with increased apoptosis of tumor cells, reduced neovascularization, and developed a strong cytotoxic T lymphocyte (CTL) response in tumor-bearing rats. Celecoxib may reduce production of prostaglandin E2 and modulate the balance between T helper 1 (Th1) cytokines and T helper 2 (Th2) cytokines by increasing the pivotal Thl cytokine interleukin-12 and reducing Th2 cytokine interleukin-10. Taken together, our results demonstrated that selective inhibition of COX-2 using celecoxib combined with DC-based immunotherapy could act as an important novel strategy for improving future treatment of malignant gliomas.
Collapse
Affiliation(s)
- Hongtao Zhang
- Department of Neurosurgery, Yantai Yuhuangding Hospital Affiliated to Qingdao University School of Medicine, Yantai, Shandong, China
| | | | | | | | | |
Collapse
|
20
|
Sequential immunotherapy by vaccination with GM-CSF-expressing glioma cells and CTLA-4 blockade effectively treats established murine intracranial tumors. J Immunother 2013; 35:385-9. [PMID: 22576343 DOI: 10.1097/cji.0b013e3182562d59] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Malignant glioma is an incurable disease with a relatively short median survival. Several clinical trials have demonstrated that immunotherapy with vaccination is a safe and possibly effective way of prolonging survival. Antibody-based blockade of cytotoxic T-lymphocyte antigen 4 (CTLA-4) ligation on T lymphocytes is associated with enhanced antitumor immunity in animal models of cancer and in patients with advanced melanoma. We hypothesized that sequential therapy with granulocyte-macrophage-colony-stimulating factor (GM-CSF)-expressing whole-glioma-cell vaccination and CTLA-4 blockade is an effective strategy for treating established intracranial gliomas. GL261 glioma cells were injected into the right frontal lobes of syngeneic C57/BL6 mice. At days 3, 6, and 9 after tumor implantation, mice were treated with subcutaneous injection of irradiated GM-CSF-expressing GL261 cells. Mice were also treated with intraperitoneal injection of anti-CTLA-4 monoclonal antibodies (mAbs), either at days 3, 6, and 9 or days 12, 15, and 18. Animals were followed for survival. Splenocytes were harvested at day 22 for use in enzyme-linked immunosorbent spot assays. Early treatment of established intracranial gliomas with high-dose CTLA-4 blockade was associated with increased survival in GL261-bearing mice. Later treatment with anti-CTLA-4 monoclonal antibodies did not significantly improve survival compared with control-treated mice. Early vaccination followed by subsequent CTLA-4 blockade was associated with significantly improved survival versus either treatment alone and intensified tumor-specific immunity as measured by interferon-γ enzyme-linked immunosorbent spot assay. Sequential immunotherapy with GM-CSF-expressing irradiated glioma cells and CTLA-4 blockade synergistically prolongs survival in mice bearing established intracranial gliomas.
Collapse
|
21
|
Petrosiute A, Auletta JJ, Lazarus HM. Achieving graft-versus-tumor effect in brain tumor patients: from autologous progenitor cell transplant to active immunotherapy. Immunotherapy 2013. [PMID: 23194364 DOI: 10.2217/imt.12.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Success in treating aggressive brain tumors like glioblastoma multiforme and medulloblastoma remains challenging, in part because these malignancies overcome CNS immune surveillance. New insights into brain tumor immunology have led to a rational development of immunotherapeutic strategies, including cytotoxic Tlymphocyte therapies and dendritic cell vaccines. However, these therapies are most effective when applied in a setting of minimal residual disease, so require prior use of standard cytotoxic therapies or cytoreduction by surgery. Myeloablative chemotherapy with autologous hematopoietic cell transplantation (autoHCT) can offer a platform upon which different cellular therapies can be effectively instituted. Specifically, this approach provides an inherent 'chemical debulking' through high-dose chemotherapy and a graft-versus-tumor effect through an autologous T-cell replete graft. Furthermore, autoHCT may be beneficial in 'resetting' the body's immune system, potentially 'breaking' tumor tolerance, and in providing a 'boost' of immune effector cells (NK cells or cytotoxic T lymphocytes), which could augment desired anti-tumor effects. As literature on the use of autoHCT in brain tumors is scarce, aspects of immunotherapies applied in non-CNS malignancies are reviewed as potential therapies that could be used in conjunction with autoHCT to eradicate brain tumors.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Pediatrics, Hematology/Oncology, Rainbow Babies & Children's Hospital, Case Western Reserve University, 11100 Euclid Avenue, Mailstop 6054, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
22
|
Bovenberg MSS, Degeling MH, Tannous BA. Cell-based immunotherapy against gliomas: from bench to bedside. Mol Ther 2013; 21:1297-305. [PMID: 23648695 DOI: 10.1038/mt.2013.80] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/17/2013] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) comprises 51% of all gliomas and is the most malignant form of brain tumors with a median survival of 18-21 months. Standard-of-care treatment includes maximal surgical resection of the tumor mass in combination with radiation and chemotherapy. However, as the poor survival rate indicates, these treatments have not been effective in preventing disease progression. Cellular immunotherapy is currently being explored as therapeutic approach to treat malignant brain tumors. In this review, we discuss advances in active, passive, and vaccine-based immunotherapeutic strategies for gliomas both at the bench and in the clinic.
Collapse
Affiliation(s)
- M Sarah S Bovenberg
- Department of Neurology, Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | |
Collapse
|
23
|
Lasky JL, Panosyan EH, Plant A, Davidson T, Yong WH, Prins RM, Liau LM, Moore TB. Autologous tumor lysate-pulsed dendritic cell immunotherapy for pediatric patients with newly diagnosed or recurrent high-grade gliomas. Anticancer Res 2013; 33:2047-2056. [PMID: 23645755 PMCID: PMC4018463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Immunotherapy has the potential to improve clinical outcomes with little toxicity for pediatric patients with brain tumors. We conducted a pilot feasibility study of tumor lysate-pulsed dendritic cell (DC) vaccination in pediatric patients (1 to 18 years old) with newly diagnosed or recurrent high-grade glioma (HGG). A total of nine DC vaccine doses, each containing 1 × 10(6) cells per dose were administered to three out of the seven originally enrolled patients. Toxicities were limited to mild side-effects, except in one case of elevated alkaline phosphatase, which resolved without clinical consequences. Two patients with primary lesions amongst the three vaccinated were alive at the time of writing, both without evidence of disease. Pre- and post-vaccination tumor samples from a patient with an anaplastic oligoastrocytoma that recurred failed to demonstrate immune cell infiltration by immunohistochemistry. Peripheral cytokine levels were evaluated in one patient following DC vaccination and demonstrated some changes in relation to vaccination. DC vaccine is tolerable and feasible with some limitations for pediatric patients with HGG. Dendritic cell based immunotherapy may provide some clinical benefit in pediatric patients with glioma, especially for patients with minimal residual disease, but further investigation of this modality is required.
Collapse
Affiliation(s)
- Joseph L Lasky
- Department of Pediatrics, Division of Pediatric Hematology/ Oncology, Harbor-UCLA Medical Center/LA Biomed, David Geffen School of Medicine UCLA, Torrance, CA 90502, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Monocyte-derived cells of the brain in malignant gliomas. World Neurosurg 2013; 82:1012-4. [PMID: 23333988 DOI: 10.1016/j.wneu.2013.01.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 01/12/2013] [Indexed: 11/22/2022]
|
25
|
Barnard Z, Wakimoto H, Zaupa C, Patel AP, Klehm J, Martuza RL, Rabkin SD, Curry WT. Expression of FMS-like tyrosine kinase 3 ligand by oncolytic herpes simplex virus type I prolongs survival in mice bearing established syngeneic intracranial malignant glioma. Neurosurgery 2013; 71:741-8; discussion 748. [PMID: 22653387 DOI: 10.1227/neu.0b013e318260fd73] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Glioblastoma is a fatal brain tumor in needing urgent effective therapy. Treatments with both oncolytic viruses and immunotherapy have shown preclinical efficacy and clinical promise. We sought to exploit possible synergies between oncolytic herpes simplex virus type 1 (oHSV-1) infection of intracranial gliomas and delivery of immune-stimulating fms-like tyrosine kinase 3 ligand (Flt3L) by engineering a herpes vector to express the cytokine. OBJECTIVE To construct an oHSV-1 vector that expresses high levels of Flt3L and examine its antiglioma efficacy in an immunocompetent murine model. METHODS G47Δ and a bacterial artificial chromosome system were used to generate a novel oHSV-1, termed G47Δ-Flt3L, expressing Flt3L. Cytokine expression was confirmed, and G47Δ-Flt3L was injected intratumorally into established intracranial CT-2A gliomas in syngeneic C57/Bl6 mice. Animals were followed for survival and assessed by the Kaplan-Meier method. RESULTS G47Δ-Flt3L expressed high levels of Flt3L in culture. Expression of Flt3L affected neither viral replication nor had a cytotoxic effect on CT2A glioma cells. Direct inoculation into intracerebral CT2A glioma cells resulted in high levels of detectable Flt3L in mouse blood and was superior to parental G47Δ in prolonging survival in glioma-bearing animals. CONCLUSION Treatment with G47Δ-Flt3L improves survival of glioma-bearing mice.
Collapse
Affiliation(s)
- Zachary Barnard
- Simches Brain Tumor Research Laboratories, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
An optimized method for manufacturing a clinical scale dendritic cell-based vaccine for the treatment of glioblastoma. PLoS One 2012; 7:e52301. [PMID: 23284979 PMCID: PMC3527532 DOI: 10.1371/journal.pone.0052301] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 11/16/2012] [Indexed: 02/07/2023] Open
Abstract
Immune-based treatments represent a promising new class of therapy designed to boost the immune system to specifically eradicate malignant cells. Immunotherapy may generate specific anti-tumor immune responses, and dendritic cells (DC), professional antigen-presenting cells, are widely used in experimental cancer immunotherapy. Several reports describe methods for the generation of mature, antigen-pulsed DC for clinical use. Improved quality and standardization are desirable to obtain GMP-compliant protocols. In this study we describe the generation of DC from 31 Glioblastoma (GB) patients starting from their monocytes isolated by immunomagnetic CD14 selection using the CliniMACS® device. Upon differentiation of CD14+ with IL-4 and GM-CSF, DC were induced to maturation with TNF-α, PGE(2), IL-1β, and IL-6. Whole tumor lysate was obtained, for the first time, in a closed system using the semi-automated dissociator GentleMACS®. The yield of proteins improved by 130% compared to the manual dissociation method. Interestingly the Mean Fluorescence Intensity for CD83 increased significantly in DC pulsed with "new method" lysate compared to DC pulsed with "classical method" lysate. Our results indicate that immunomagnetic isolation of CD14(+) monocytes using the CliniMACS® device and their pulsing with whole tumor lysate proteins is a suitable method for clinical-scale generation of high quality, functional DC under GMP-grade conditions.
Collapse
|
27
|
Kushchayev SV, Kushchayeva YS, Wiener PC, Scheck AC, Badie B, Preul MC. Monocyte-derived cells of the brain and malignant gliomas: the double face of Janus. World Neurosurg 2012. [PMID: 23178919 DOI: 10.1016/j.wneu.2012.11.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Monocyte-derived cells of the brain (MDCB) are a diverse group of functional immune cells that are also highly abundant in gliomas. There is growing evidence that MDCB play essential roles in the pathogenesis of gliomas. The aim of this review was to collate and systematize contemporary knowledge about these cells as they relate to glioma progression and antiglioblastoma therapeutic modalities with a view toward improved effectiveness of therapy. METHODS We reviewed relevant studies to construct a summary of different MDCB subpopulations in steady state and in malignant gliomas and discuss their role in the development of malignant gliomas and potential future therapies. RESULTS Current studies suggest that MDCB subsets display different phenotypes and differentiation potentials depending on their milieu in the brain and exposure to tumoral influences. MDCB possess specific and unique functions, including those that are protumoral and those that are antitumoral. CONCLUSIONS Elucidating the role of mononuclear-derived cells associated with gliomas is crucial in designing novel immunotherapy strategies. Much progress is needed to characterize markers to identify cell subsets and their specific regulatory roles. Investigation of MDCB can be clinically relevant. Specific MDCB populations potentially can be used for glioma therapy as a target or as cell vehicles that might deliver cytotoxic substances or processes to the glioma microenvironment.
Collapse
Affiliation(s)
- Sergiy V Kushchayev
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Yevgeniya S Kushchayeva
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA; Department of Surgery, Medstar Washington Hospital Center, Washington, DC, USA
| | - Philip C Wiener
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Adrienne C Scheck
- Neuro-oncology Research Laboratory, Division of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Behnam Badie
- Division of Neurosurgery, Department of Surgery, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Mark C Preul
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.
| |
Collapse
|
28
|
Valle RD, Cerio ALDD, Inoges S, Tejada S, Pastor F, Villanueva H, Gallego J, Espinos J, Aristu J, Idoate MA, Andreu E, Bendandi M. Dendritic cell vaccination in glioblastoma after fluorescence-guided resection. World J Clin Oncol 2012; 3:142-149. [PMID: 23293753 PMCID: PMC3536842 DOI: 10.5306/wjco.v3.i11.142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 12/04/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To assess whether the addition of a customized, active immunotherapy to standard of care including fluorescence-guided surgery, may provide hints of an improved survival for patients with poor-prognosis, incurable glioblastoma multiform.
METHODS: Preliminary to our ongoing, phase-II clinical trial, we conducted a small pilot study enrolling five consecutive patients with resectable glioblastoma. In terms of Recursive Partitioning Analysis, four patients were class V and one was class IV. In all five cases, fluorescence-guided surgery was employed, followed by rapid steroid discontinuation. Patients were then treated with a combination of standard radio-chemotherapy with temozolomide and tumor lysate-pulsed, mature dendritic cell-based vaccinations.
RESULTS: Though all five patients ultimately progressed, with any further treatment left to the sole decision of the treating oncologist, active immunotherapy was very well tolerated and induced specific immune responses in all three patients for whom enough material was available for such an assessment. Median progression-free survival was 16.1 mo. Even more important, median and mean overall survival were 27 mo and 26 mo, respectively. Three patients have died with an overall survival of 9 mo, 27 mo and 27.4 mo, while the other two are still alive at 32 mo and 36 mo, the former receiving treatment with bevacizumab, while the latter has now been off therapy for 12 mo. Four of five patients were alive at two years.
CONCLUSION: Active immunotherapy with tumor lysate-pulsed, autologous dendritic cells is feasible, safe, well tolerated and biologically efficacious. A phase-II study is ongoing to possibly improve further on our very encouraging clinical results.
Collapse
|
29
|
Ge L, Cornforth AN, Hoa NT, Delgado C, Chiou SK, Zhou YH, Jadus MR. Differential glioma-associated tumor antigen expression profiles of human glioma cells grown in hypoxia. PLoS One 2012; 7:e42661. [PMID: 22957023 PMCID: PMC3434178 DOI: 10.1371/journal.pone.0042661] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 07/10/2012] [Indexed: 12/31/2022] Open
Abstract
Human U251 and D54 glioma cells were tested for expression of 25 glioma-associated tumor antigen precursor proteins (TAPP) under hypoxic (1% O2) or normoxic (21% O2) conditions. Hypoxic glioma cell lines increased their mRNA expression for nine TAPP (Aim2, Art-4, EphA2, EZH2, Fosl1, PTH-rP, Sox 11, Whsc2 and YKL-40), as assessed by quantitative reverse transcriptase real-time/polymerase chain reaction (qRT-PCR). Increased differences with three hypoxic-induced TAPP: EZH2, Whsc2 and YKL-40 were shown at the protein levels by fluorescent antibody staining and quantitative electrophoretic analysis. Two TAPP (MRP3 and Trp1) were down-regulated by hypoxia in glioma cell lines. Growing the glioma cells under hypoxia for 13 days, followed by returning them back to normoxic conditions for 7 days, and restored the original normoxic TAPP profile. Thus, hypoxia was an environmental factor that stimulated the transient expression of these antigens. Intracranial xenografts grown in nude mice derived from U251 cells that had been cultured under neurosphere stem cell conditions showed increased expression of Whsc2 or YKL-40, demonstrating that these in vitro properties of glioma also occur in vivo. Whsc2-specific cytotoxic T lymphocytes killed the hypoxic U251 glioma cells better than normoxic glioma cells. The antigens expressed by hypoxic tumor cells may be a better source of starting tumor material for loading dendritic cells for novel immunotherapy of glioma using tumor-associated antigens.
Collapse
Affiliation(s)
- Lisheng Ge
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | | | - Neil T. Hoa
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
- Research Service Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Christina Delgado
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
- Research Service Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Shiun Kwei Chiou
- Research Service Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Yi Hong Zhou
- Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of California Irvine, Orange, California, United States of America
- Department of Neurological Surgery, University of California Irvine, Irvine, California, United States of America
- Department of Biological Chemistry, University of California Irvine, Irvine, California, United States of America
| | - Martin R. Jadus
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
- Research Service Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
- Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of California Irvine, Orange, California, United States of America
- Pathology and Laboratory Medicine, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Medulloblasoma: challenges for effective immunotherapy. J Neurooncol 2011; 108:1-10. [PMID: 22173741 DOI: 10.1007/s11060-011-0776-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 11/29/2011] [Indexed: 01/21/2023]
Abstract
For medulloblastoma patients, the current therapeutic paradigm of surgery followed by radiation and chemotherapy can lead to long-term remission. However, the sequelae of treatment can be very debilitating, particularly in young children. Immunotherapy is an attractive treatment approach to optimize the targeting of tumor cells while sparing the vulnerable surrounding brain that is still developing in children. Understanding the relationship between medulloblastoma and the immune system is critical to develop effective immunologic-based treatment strategies for these patients. This review focuses on current knowledge of tumor immunology and the factors that contribute to the lack of immune system recognition of these tumors. The specificity of tumor antigens present in medulloblastoma is also discussed along with a summary of early clinical immunotherapy results.
Collapse
|
31
|
Hope and challenges for dendritic cell-based vaccine therapy for glioblastoma. World Neurosurg 2011; 77:633-5. [PMID: 22120228 DOI: 10.1016/j.wneu.2011.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 10/06/2011] [Indexed: 10/15/2022]
|
32
|
Immunotherapy of brain cancers: the past, the present, and future directions. Clin Dev Immunol 2011; 2010:296453. [PMID: 21437175 PMCID: PMC3061456 DOI: 10.1155/2010/296453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Treatment of brain cancers, especially high grade gliomas (WHO stage III and IV) is slowly making progress, but not as fast as medical researchers and the patients would like. Immunotherapy offers the opportunity to allow the patient's own immune system a chance to help eliminate the cancer. Immunotherapy's strength is that it efficiently treats relatively small tumors in experimental animal models. For some patients, immunotherapy has worked for them while not showing long-term toxicity. In this paper, we will trace the history of immunotherapy for brain cancers. We will also highlight some of the possible directions that this field may be taking in the immediate future for improving this therapeutic option.
Collapse
|
33
|
|
34
|
Antitumor cytotoxic T-cell response induced by a survivin peptide mimic. Cancer Immunol Immunother 2010; 59:1211-21. [PMID: 20422411 DOI: 10.1007/s00262-010-0845-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/01/2010] [Indexed: 12/23/2022]
Abstract
Survivin is a tumor-associated antigen with significant potential as a cancer vaccine target. We have identified a survivin peptide mimic containing human MHC class I epitopes and a potential class II ligand that induces a potent antitumor response in C57BL/6 mice with GL261 cerebral gliomas. This peptide is able to elicit both CD8+ CTL and T helper cell responses in C57BL/6 mice. The corresponding region of the human survivin molecule represented by peptide SVN53-67 is 100% homologous to the murine protein, but SVN53-67 is weakly immunogenic in man. We evaluated several amino acid substitutions in putative human MHC I anchor positions in SVN53-67 to identify potential peptide mimics that could provide an enhanced antitumor immune response against human glioma and primary central nervous system lymphoma (PCNSL) cells in culture. We evaluated survivin peptides with predicted binding to human HLA-A*0201 antigen using peptide-loaded dendritic cells from PBMC of patients with these malignancies. One alteration (M57) led to binding to HLA-A*0201 with significantly higher affinity. We compared the ability of autologous dendritic cells loaded with SVN53-67 peptide and SVN53-67/M57 in CTL assays against allomatched and autologous, survivin-expressing, human malignant glioma and PCNSL cells. Both SVN53-67 and SVN53-67/M57 produced CTL-mediated killing of malignant target cells; however, SVN53-67/M57 was significantly more effective than SVN53-67. Thus, SVN53-67/M57 may act as a peptide mimic to induce an enhanced antitumor CTL response in tumor patients. The use of SVN53-67/M57 as a cancer vaccine might have application for cancer vaccine therapy.
Collapse
|