1
|
Sharma H, Narayanan KB, Ghosh S, Singh KK, Rehan P, Amist AD, Bhaskar R, Sinha JK. Nanotherapeutics for Meningitis: Enhancing Drug Delivery Across the Blood-Brain Barrier. Biomimetics (Basel) 2025; 10:25. [PMID: 39851741 PMCID: PMC11762342 DOI: 10.3390/biomimetics10010025] [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: 10/18/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Meningitis is the acute or chronic inflammation of the protective membranes, surrounding the brain and spinal cord, and this inflammatory process spreads throughout the subarachnoid space. The traditional drug delivery methods pose a disadvantage in limiting the capacity of crossing the blood-brain barrier (BBB) to reach the central nervous system (CNS). Hence, it is imperative to develop novel approaches that can overcome these constraints and offer efficient therapy for meningitis. Nanoparticle (NP)-based therapeutic approaches have the potential to address the limitations such as penetrating the BBB and achieving targeted drug release in specific cells and tissues. This review highlights recent advancements in nanotechnology-based approaches, such as functionalized polymeric nanoparticles, solid lipid nanoparticles (SLNs), nanostructured lipid carriers, nanoemulsions, liposomes, transferosomes, and metallic NPs for the treatment of meningitis. Recently, bionics has emerged as a next-generation technology in the development of novel ideas from biological principles, structures, and interactions for neurological and neuroinfectious diseases. Despite their potential, more studies are needed to ensure the safety and efficacy of NP-based drug delivery systems focusing on critical aspects such as toxicity, immunogenicity, and pharmacokinetics. Therefore, this review addresses current treatment strategies and innovative nanoparticle approaches, and it discusses future directions for efficient and targeted meningitis therapies.
Collapse
Affiliation(s)
- Hitaishi Sharma
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Symbiosis International (Deemed University), Hinjawadi, Pune 411057, Maharashtra, India
| | - Prarthana Rehan
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India
| | - Aparajita Dasgupta Amist
- Amity University Uttar Pradesh (AUUP), Sector 125, Gautam Buddha Nagar, Noida 201303, Uttar Pradesh, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | | |
Collapse
|
2
|
Jiménez-Cortegana C, Palomares F, Alba G, Santa-María C, de la Cruz-Merino L, Sánchez-Margalet V, López-Enríquez S. Dendritic cells: the yin and yang in disease progression. Front Immunol 2024; 14:1321051. [PMID: 38239364 PMCID: PMC10794555 DOI: 10.3389/fimmu.2023.1321051] [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: 10/16/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Dendritic cells (DCs) are antigen presenting cells that link innate and adaptive immunity. DCs have been historically considered as the most effective and potent cell population to capture, process and present antigens to activate naïve T cells and originate favorable immune responses in many diseases, such as cancer. However, in the last decades, it has been observed that DCs not only promote beneficial responses, but also drive the initiation and progression of some pathologies, including inflammatory bowel disease (IBD). In line with those notions, different therapeutic approaches have been tested to enhance or impair the concentration and role of the different DC subsets. The blockade of inhibitory pathways to promote DCs or DC-based vaccines have been successfully assessed in cancer, whereas the targeting of DCs to inhibit their functionality has proved to be favorable in IBD. In this review, we (a) described the general role of DCs, (b) explained the DC subsets and their role in immunogenicity, (c) analyzed the role of DCs in cancer and therapeutic approaches to promote immunogenic DCs and (d) analyzed the role of DCs in IBD and therapeutic approaches to reduced DC-induced inflammation. Therefore, we aimed to highlight the "yin-yang" role of DCs to improve the understand of this type of cells in disease progression.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Francisca Palomares
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Gonzalo Alba
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Consuelo Santa-María
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, Seville, Spain
| | - Luis de la Cruz-Merino
- Clinical Oncology Dept. Medicine Department, University of Seville, Virgen Macarena University Hospital, Seville, Spain
| | - Victor Sánchez-Margalet
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Soledad López-Enríquez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
3
|
Hwang EI, Sayour EJ, Flores CT, Grant G, Wechsler-Reya R, Hoang-Minh LB, Kieran MW, Salcido J, Prins RM, Figg JW, Platten M, Candelario KM, Hale PG, Blatt JE, Governale LS, Okada H, Mitchell DA, Pollack IF. The current landscape of immunotherapy for pediatric brain tumors. NATURE CANCER 2022; 3:11-24. [PMID: 35121998 DOI: 10.1038/s43018-021-00319-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Pediatric central nervous system tumors are the most common solid malignancies in childhood, and aggressive therapy often leads to long-term sequelae in survivors, making these tumors challenging to treat. Immunotherapy has revolutionized prospects for many cancer types in adults, but the intrinsic complexity of treating pediatric patients and the scarcity of clinical studies of children to inform effective approaches have hampered the development of effective immunotherapies in pediatric settings. Here, we review recent advances and ongoing challenges in pediatric brain cancer immunotherapy, as well as considerations for efficient clinical translation of efficacious immunotherapies into pediatric settings.
Collapse
Affiliation(s)
- Eugene I Hwang
- Division of Oncology, Brain Tumor Institute, Children's National Hospital, Washington, DC, USA.
| | - Elias J Sayour
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Catherine T Flores
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Gerald Grant
- Division of Pediatric Neurosurgery, Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA, USA
| | - Robert Wechsler-Reya
- Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lan B Hoang-Minh
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | | | | | - Robert M Prins
- Departments of Neurosurgery and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John W Figg
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University and CCU Brain Tumor Immunology, DKFZ, Heidelberg, Germany
| | - Kate M Candelario
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Paul G Hale
- Children's Brain Trust, Coral Springs, FL, USA
| | - Jason E Blatt
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Lance S Governale
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, CA, USA
| | - Duane A Mitchell
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Nonaka M, Suzuki-Anekoji M, Nakayama J, Mabashi-Asazuma H, Jarvis DL, Yeh JC, Yamasaki K, Akama TO, Huang CT, Campos AR, Nagaoka M, Sasai T, Kimura-Takagi I, Suwa Y, Yaegashi T, Shibata TK, Sugihara K, Nishizawa-Harada C, Fukuda M, Fukuda MN. Overcoming the blood-brain barrier by Annexin A1-binding peptide to target brain tumours. Br J Cancer 2020; 123:1633-1643. [PMID: 32921792 PMCID: PMC7686308 DOI: 10.1038/s41416-020-01066-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 11/09/2022] Open
Abstract
Background Annexin A1 is expressed specifically on the tumour vasculature surface. Intravenously injected IF7 targets tumour vasculature via annexin A1. We tested the hypothesis that IF7 overcomes the blood–brain barrier and that the intravenously injected IF7C(RR)-SN38 eradicates brain tumours in the mouse. Methods (1) A dual-tumour model was generated by inoculating luciferase-expressing melanoma B16 cell line, B16-Luc, into the brain and under the skin of syngeneic C57BL/6 mice. IF7C(RR)-SN38 was injected intravenously daily at 7.0 μmoles/kg and growth of tumours was assessed by chemiluminescence using an IVIS imager. A similar dual-tumour model was generated with the C6-Luc line in immunocompromised SCID mice. (2) IF7C(RR)-SN38 formulated with 10% Solutol HS15 was injected intravenously daily at 2.5 μmoles/kg into two brain tumour mouse models: B16-Luc cells in C57BL/6 mice, and C6-Luc cells in nude mice. Results (1) Daily IF7C(RR)-SN38 injection suppressed tumour growth regardless of cell lines or mouse strains. (2) Daily injection of Solutol-formulated IF7C(RR)-SN38 led into complete disappearance of B16-Luc brain tumour in C57BL/6 mice, whereas this did not occur in C6-Luc in nude mice. Conclusions IF7C(RR)-SN38 crosses the blood–brain barrier and suppresses growth of brain tumours in mouse models. Solutol HS15-formulated IF7C(RR)-SN38 may have promoted an antitumour immune response.
Collapse
Affiliation(s)
- Motohiro Nonaka
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.,Laboratory for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan.,Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Misa Suzuki-Anekoji
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | | | - Donald L Jarvis
- Department of Molecular Biology, University of Wyoming, Laramie, WY, 82071, USA
| | - Jiunn-Chern Yeh
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Kazuhiko Yamasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8566, Japan
| | - Tomoya O Akama
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Chun-Teng Huang
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Alexandre Rosa Campos
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Masato Nagaoka
- Yakult Central Institute, Kunitachi, Tokyo, 186-8650, Japan
| | - Toshio Sasai
- Yakult Central Institute, Kunitachi, Tokyo, 186-8650, Japan
| | | | - Yoichi Suwa
- Yakult Central Institute, Kunitachi, Tokyo, 186-8650, Japan
| | | | - Toshiaki K Shibata
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.,Department of Gynecology and Obstetrics, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Kazuhiro Sugihara
- Department of Gynecology and Obstetrics, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Chizuko Nishizawa-Harada
- Laboratory for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan
| | - Minoru Fukuda
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Michiko N Fukuda
- Cancer Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA. .,Laboratory for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan.
| |
Collapse
|
5
|
Chang CY, Jeon S, Yoon HJ, Choi B, Kim SS, Oshima M, Park EJ. Glial TLR2‐driven innate immune responses and CD8
+
T cell activation against brain tumor. Glia 2019; 67:1179-1195. [DOI: 10.1002/glia.23597] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/06/2018] [Accepted: 01/09/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Chi Young Chang
- Immunotherapeutics Branch National Cancer Center Goyang South Korea
| | - Sae‐Bom Jeon
- Immunotherapeutics Branch National Cancer Center Goyang South Korea
| | - Hee Jung Yoon
- Immunotherapeutics Branch National Cancer Center Goyang South Korea
| | - Bum‐Kyu Choi
- Immunotherapeutics Branch National Cancer Center Goyang South Korea
| | - Sang Soo Kim
- Particle Therapy Research Branch National Cancer Center Goyang South Korea
| | - Masanobu Oshima
- Division of Genetics Cancer Research Institute, Kanazawa University Kanazawa Japan
| | - Eun Jung Park
- Immunotherapeutics Branch National Cancer Center Goyang South Korea
- Department of Cancer Biomedical Science Graduate School of Cancer Science and Policy, National Cancer Center Goyang South Korea
| |
Collapse
|
6
|
da Fonseca ACC, Amaral R, Garcia C, Geraldo LH, Matias D, Lima FRS. Microglia in Cancer: For Good or for Bad? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:245-261. [PMID: 27714693 DOI: 10.1007/978-3-319-40764-7_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma is a malignant tumor of astrocytic origin that is highly invasive, proliferative and angiogenic. Despite current advances in multimodal therapies, such as surgery, radio- and chemotherapy, the outcome for patients with glioblastoma is nearly always fatal. The glioblastoma microenvironment has a tremendous influence over the tumor growth and spread. Microglia and macrophages are abundant cells in the tumor mass. Increasing evidence indicates that glioblastoma recruits these cell populations and signals in a way that microglia and macrophages are subverted to promote tumor progression. In this chapter, we discuss some aspects of the interaction between microglia and glioblastoma, consequences of this interaction for tumor progression and the possibility of microglial cells being used as therapeutic vectors, which opens up new alternatives for the development of GBM therapies targeting microglia.
Collapse
Affiliation(s)
- Anna Carolina Carvalho da Fonseca
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Rackele Amaral
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Celina Garcia
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Luiz Henrique Geraldo
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Diana Matias
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Flavia Regina Souza Lima
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil.
| |
Collapse
|
7
|
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: 3.7] [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
|
8
|
Fujita M, Matsui T, Ito A. Biomedical insights into cell adhesion and migration-from a viewpoint of central nervous system tumor immunology. Front Cell Dev Biol 2015; 3:55. [PMID: 26528477 PMCID: PMC4604325 DOI: 10.3389/fcell.2015.00055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/08/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mitsugu Fujita
- Department of Microbiology, Faculty of Medicine, Kindai University Osaka, Japan
| | - Takaaki Matsui
- Gene Regulation Research, Graduate School of Biological Sciences, Nara Institute of Science and Technology Nara, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University Osaka, Japan
| |
Collapse
|
9
|
Mahata B, Biswas S, Rayman P, Chahlavi A, Ko J, Bhattacharjee A, Li YT, Li Y, Das T, Sa G, Raychaudhuri B, Vogelbaum MA, Tannenbaum C, Finke JH, Biswas K. GBM Derived Gangliosides Induce T Cell Apoptosis through Activation of the Caspase Cascade Involving Both the Extrinsic and the Intrinsic Pathway. PLoS One 2015. [PMID: 26226135 PMCID: PMC4520498 DOI: 10.1371/journal.pone.0134425] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previously we demonstrated that human glioblastoma cell lines induce apoptosis in peripheral blood T cells through partial involvement of secreted gangliosides. Here we show that GBM-derived gangliosides induce apoptosis through involvement of the TNF receptor and activation of the caspase cascade. Culturing T lymphocytes with GBM cell line derived gangliosides (10-20μg/ml) demonstrated increased ROS production as early as 18 hrs as indicated by increased uptake of the dye H2DCFDA while western blotting demonstrated mitochondrial damage as evident by cleavage of Bid to t-Bid and by the release of cytochrome-c into the cytosol. Within 48-72 hrs apoptosis was evident by nuclear blebbing, trypan blue positivity and annexinV/7AAD staining. GBM-ganglioside induced activation of the effector caspase-3 along with both initiator caspases (-9 and -8) in T cells while both the caspase-8 and -9 inhibitors were equally effective in blocking apoptosis (60% protection) confirming the role of caspases in the apoptotic process. Ganglioside-induced T cell apoptosis did not involve production of TNF-α since anti-human TNFα antibody was unable to protect T cells from nuclear blebbing and subsequent cell death. However, confocal microscopy demonstrated co-localization of GM2 ganglioside with the TNF receptor and co-immunoprecipitation experiments showed recruitment of death domains FADD and TRADD with the TNF receptor post ganglioside treatment, suggesting direct interaction of gangliosides with the TNF receptor. Further confirmation of the interaction between GM2 and TNFR1 was obtained from confocal microscopy data with wild type and TNFR1 KO (TALEN mediated) Jurkat cells, which clearly demonstrated co-localization of GM2 and TNFR1 in the wild type cells but not in the TNFR1 KO clones. Thus, GBM-ganglioside can mediate T cell apoptosis by interacting with the TNF receptor followed by activation of both the extrinsic and the intrinsic pathway of caspases.
Collapse
Affiliation(s)
- Barun Mahata
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Soumika Biswas
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - Patricia Rayman
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - Ali Chahlavi
- Spine and Brain Institute, St. Vincent Medical Center, Jacksonville, Florida, United States of America
| | - Jennifer Ko
- Pathology Institute, Cleveland Clinic, Cleveland, United States of America
| | | | - Yu-Teh Li
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States of America
| | - Yuntao Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - Tanya Das
- Division of Molecular Medicine, Bose Institute, Kolkata, India
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - Baisakhi Raychaudhuri
- Brain Tumor and Neuro-oncology Center in the Neurological Institute, Cleveland Clinic, Cleveland, United States of America
| | - Michael A. Vogelbaum
- Brain Tumor and Neuro-oncology Center in the Neurological Institute, Cleveland Clinic, Cleveland, United States of America
| | - Charles Tannenbaum
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - James H. Finke
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States of America
| | - Kaushik Biswas
- Division of Molecular Medicine, Bose Institute, Kolkata, India
- * E-mail:
| |
Collapse
|
10
|
Zhang Y, Luo F, Li A, Qian J, Yao Z, Feng X, Chu Y. Systemic injection of TLR1/2 agonist improves adoptive antigen-specific T cell therapy in glioma-bearing mice. Clin Immunol 2014; 154:26-36. [PMID: 24928324 DOI: 10.1016/j.clim.2014.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/24/2014] [Accepted: 06/03/2014] [Indexed: 01/07/2023]
Abstract
Adoptive immunotherapy is an attractive strategy for glioma treatment. However, some obstacles still need be overcome. In this study, GL261-bearing mice treated with adoptively transferred antigen-specific T cells and systemic injection of bacterial lipoprotein (BLP), a TLR1/2 agonist, got a long-term survival and even immune protection. By analyzing adoptive T cells, it was found that BLP maintained T cell survival, proliferation and anti-tumor efficacy in the brains of tumor-bearing hosts. Moreover, tumor microenvironment was modified by up-regulating IFN-γ-secreting CD8+ T cells and down-regulating MDSC, which might be related with high CXCL10 and low CCL2 expression. In addition, TLR2 deficiency abrogated therapeutic effect with increased MDSC accumulation and decreased IFN-γ-secreting CD8+ T cells in the brains. Thus, the systemic injection of BLP could improve the adoptive T cell therapy by maintaining T cell persistence, modifying the tumor microenvironment and even inducing systemic anti-tumor immunity, which might offer a clinically promising immunotherapeutic strategy for glioma.
Collapse
Affiliation(s)
- Yufei Zhang
- Department of Immunology, School of Basic Medical Sciences and Biotherapy Research Center, Fudan University, Shanghai, China
| | - Feifei Luo
- Department of Immunology, School of Basic Medical Sciences and Biotherapy Research Center, Fudan University, Shanghai, China
| | - Anning Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiawen Qian
- Department of Immunology, School of Basic Medical Sciences and Biotherapy Research Center, Fudan University, Shanghai, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyuan Feng
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences and Biotherapy Research Center, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Mossoba ME, Medin JA. Cancer immunotherapy using virally transduced dendritic cells: animal studies and human clinical trials. Expert Rev Vaccines 2014; 5:717-32. [PMID: 17181444 DOI: 10.1586/14760584.5.5.717] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The immune system uses a process known as 'immunosurveillance' to help prevent the outgrowth of tumors. In cancer immunotherapy, a major goal is for immunity against tumor-associated antigens to be generated or strengthened in patients. To achieve this goal, several approaches have been tested, including the use of highly potent antigen-presenting cells called dendritic cells (DCs), which can activate T cells efficiently. Presentation of peptides derived from tumor antigens on the surface of DCs can stimulate strong antitumor immunity. Using recombinant viral vectors encoding tumor-associated antigens, DCs can be engineered efficiently to express sustained levels of tumor-antigen peptides. This review discusses the effectiveness of virally transduced DCs in treating tumors and generating antigen-specific T-cell responses. It covers mouse and nonhuman primate studies, preclinical in vitro human cell experiments and clinical trials.
Collapse
Affiliation(s)
- Miriam E Mossoba
- Department of Medical Biophysics, University of Toronto, 67 College Street, Room 426, Toronto, Ontario, M5G 2MI, Canada.
| | | |
Collapse
|
12
|
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: 2.5] [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
|
13
|
Pollack IF, Jakacki RI, Butterfield LH, Okada H. Ependymomas: development of immunotherapeutic strategies. Expert Rev Neurother 2013; 13:1089-98. [PMID: 24117271 PMCID: PMC3972122 DOI: 10.1586/14737175.2013.840420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ependymomas are among the most challenging childhood brain tumors. Although 50-70% of ependymomas are cured with surgery and irradiation, a significant percentage of tumors recur. Ependymomas that are not amenable to complete resection at diagnosis have a particularly poor prognosis, and the vast majority of affected children experience tumor recurrence. Although transient responses have been observed in recurrent tumors treated with re-irradiation and several chemotherapy regimens, long-term disease control is rarely achieved. Children with recurrent disease commonly experience cumulative neurological morbidity from repeated surgical and adjuvant therapy interventions and almost universally succumb to refractory tumor progression. Accordingly, conceptually new treatment approaches are needed, both to decrease the risk of tumor recurrence and to enhance disease control in those children who experience recurrent disease. This article reviews the current application of risk-based treatment stratification at diagnosis, the rationale for exploring the role of novel therapeutic strategies such as immunotherapy at recurrence and the concept behind a vaccine-based trial for these tumors.
Collapse
Affiliation(s)
- Ian F. Pollack
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Regina I. Jakacki
- Department of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa H Butterfield
- Department of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hideho Okada
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Pollack IF, Jakacki RI, Butterfield LH, Okada H. Peptide Vaccine Therapy for Childhood Gliomas. Neurosurgery 2013; 60 Suppl 1:113-9. [DOI: 10.1227/01.neu.0000430769.33467.68] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
15
|
Abstract
Although the immune system evolved to protect the host from foreign infection, it can sometimes recognize and attack host tissues, a phenomenon known as autoimmunity. In addition to genetic factors, environmental elements such as viruses and bacteria are thought to play a role in the development of autoimmune diseases. The major hypothesized mechanism by which infection with these agents can lead to autoimmunity is termed molecular mimicry. Here, immune responses initiated against foreign antigens are cross-reactive with self-antigens. This is thought to occur especially if the foreign antigen is similar in structure or amino acid sequence to the self-antigen. In this review, we explore evidence for the role of molecular mimicry in neurological diseases.
Collapse
Affiliation(s)
- Anne M Ercolini
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|
16
|
Khan-Farooqi HR, Prins RM, Liau LM. Tumor immunology, immunomics and targeted immunotherapy for central nervous system malignancies. Neurol Res 2013; 27:692-702. [PMID: 16197806 DOI: 10.1179/016164105x49490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Although the brain was traditionally considered as 'immunologically privileged', recent findings have implied an involvement of immune mechanisms in neurological disease and illness, including central nervous system (CNS) malignancies. In this review, we initially focus on aspects of the immune system critical for effective antitumor immunity, as an understanding of normal immunological functions and how they relate to tumor immunology will set a foundation for understanding the unique challenges facing the integration of neuro-oncology and neuroimmunology. We summarize current knowledge of immune responses in the 'immunologically quiescent' brain and its role in tumor immunology. We will then discuss the emerging field of 'immunomics' and recent advances in molecular technologies, such as DNA microarray, which are being applied to brain tumor antigen epitope discovery and patient stratification for brain cancer immunotherapy. This, in turn, should have significant importance for ultimately designing and developing efficient and focused strategies for anticancer immunotherapy. Finally, the current state of immune-based treatment paradigms and future directions will be discussed, paying particular attention to targeted antibody strategies, adoptive cellular immunotherapy, and tumor vaccine approaches that have been studied in clinical trials for CNS neoplasms.
Collapse
Affiliation(s)
- Haumith R Khan-Farooqi
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California 90095-6901, USA
| | | | | |
Collapse
|
17
|
Dunn GP, Fecci PE, Curry WT. Cancer immunoediting in malignant glioma. Neurosurgery 2013; 71:201-22; discussion 222-3. [PMID: 22353795 DOI: 10.1227/neu.0b013e31824f840d] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Significant work from many laboratories over the last decade in the study of cancer immunology has resulted in the development of the cancer immunoediting hypothesis. This contemporary framework of the naturally arising immune system-tumor interaction is thought to comprise 3 phases: elimination, wherein immunity subserves an extrinsic tumor suppressor function and destroys nascent tumor cells; equilibrium, wherein tumor cells are constrained in a period of latency under immune control; and escape, wherein tumor cells outpace immunity and progress clinically. In this review, we address in detail the relevance of the cancer immunoediting concept to neurosurgeons and neuro-oncologists treating and studying malignant glioma by exploring the de novo immune response to these tumors, how these tumors may persist in vivo, the mechanisms by which these cells may escape/attenuate immunity, and ultimately how this concept may influence our immunotherapeutic approaches.
Collapse
Affiliation(s)
- Gavin P Dunn
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
18
|
Syed ON, Mandigo CE, Killory BD, Canoll P, Bruce JN. Cancer-testis and melanocyte-differentiation antigen expression in malignant glioma and meningioma. J Clin Neurosci 2012; 19:1016-21. [DOI: 10.1016/j.jocn.2011.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 10/18/2011] [Indexed: 11/27/2022]
|
19
|
Wang M, Dong Q, Zhang D, Wang Y. Expression of delta-catenin is associated with progression of human astrocytoma. BMC Cancer 2011; 11:514. [PMID: 22151302 PMCID: PMC3262777 DOI: 10.1186/1471-2407-11-514] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 12/12/2011] [Indexed: 11/11/2022] Open
Abstract
Background δ-Catenin (CTNND2), which encodes a scaffold protein in humans, has been found in a few malignancies. However, the expression pattern and contribution of δ-catenin to astrocytoma progression are unclear. Methods We investigated δ-catenin expression in human astrocytoma samples and its function in astrocytoma cell lines using immunohistochemistry, siRNA knockdown, transfection, MTT, transwell migration and Rac1 pulldown techniques. Results δ-Catenin protein expression was detected in cytoplasm of astrocytoma cells by immunohistochemistry. Analysis showed that grade I astrocytoma (0%, 0/11) and glial cells from normal brain tissue exhibited negative staining. δ-Catenin expression was significantly higher in grade III-IV (35%, 29/84) compared to grade II astrocytoma cells (18%, 11/61); p < 0.01). In addition, CTNND2 overexpression promoted proliferation, invasion and Rac1 activity of U251 astrocytoma cells. Treatment of δ-catenin-transfected cells with a Rac1 inhibitor decreased Rac1 activity and invasion. δ-Catenin knockdown in U87 glioblastoma cell decreased cell proliferation, invasion and Rac1 activity. Conclusion The results suggest that δ-catenin expression is associated with the malignant progression of astrocytoma and promotes astrocytoma cell invasion through upregulation of Rac1 activity. δ-Catenin expression levels may serve as a useful marker of the biological behavior of astrocytoma cells.
Collapse
Affiliation(s)
- MingHao Wang
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang 110001, PR China.
| | | | | | | |
Collapse
|
20
|
TLR9 expression is associated with prognosis in patients with glioblastoma multiforme. J Clin Neurosci 2011; 19:75-80. [PMID: 22169598 DOI: 10.1016/j.jocn.2011.03.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/25/2011] [Accepted: 03/29/2011] [Indexed: 11/21/2022]
Abstract
The aim of this study was to determine if there was an association between expression of toll-like receptor 9 (TLR9) in glioblastoma tissue and patient outcome in glioblastoma multiforme. Further, we characterized the direct in vitro effects of the TLR9 agonist, CpG oligodeoxynucleotide (ODN), commonly used as a vaccine adjuvant in cancer immunotherapy, on glioma cells. TLR9 expression was assessed using immunohistochemical techniques, and enzyme-linked immunosorbent assays were used to investigate the expression of other proteins in glioma cells relevant to immunotherapy. There was a highly significant difference in both progression-free survival and overall survival between TLR9+ and TLR9- patients, with poorer outcome in TLR9+ patients. In in vitro glioma cells, there was a positive correlation between the protein levels of TLR9 and both matrix metalloproteinase (MMP)-2 and MMP-9 (p<0.05), but no relationship between TLR9 levels and levels of interleukin-6, transforming growth factor-β2 or signal transducer and activator of transcription (STAT)-3 (p>0.05). Our data indicate that expression of TLR9 correlates with shorter progression-free survival and overall survival in patients with glioblastoma multiforme. Our findings also indicate that caution is warranted when directly injecting the TLR9 agonist CpG ODN into glioma tissues as part of glioma immunotherapy. Because the CpG ODN is a TLR9 agonist, we recommend caution when using CpG ODN in immunotherapy.
Collapse
|
21
|
Combined Flt3L/TK gene therapy induces immunological surveillance which mediates an immune response against a surrogate brain tumor neoantigen. Mol Ther 2011; 19:1793-801. [PMID: 21505426 DOI: 10.1038/mt.2011.77] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor with a median survival of 14.6 months postdiagnosis. The infiltrative nature of GBM prevents complete resection and residual brain tumor cells give rise to recurrent GBM, a hallmark of this disease. Recurrent GBMs are known to harbor numerous mutations/gene rearrangements when compared to the primary tumor, which leads to the potential expression of novel proteins that could serve as tumor neoantigens. We have developed a combined immune-based gene therapeutic approach for GBM using adenoviral (Ads) mediated gene delivery of Herpes Simplex Virus Type 1-thymidine kinase (TK) into the tumor mass to induce tumor cells' death combined with an adenovirus expressing fms-like tyrosine kinase 3 ligand (Flt3L) to recruit dendritic cells (DCs) into the tumor microenvironment. This leads to the induction of specific anti-brain tumor immunity and immunological memory. In a model of GBM recurrence, we demonstrate that Flt3L/TK mediated immunological memory is capable of recognizing brain tumor neoantigens absent from the original treated tumor. These data demonstrate that the Flt3L/TK gene therapeutic approach can induce systemic immunological memory capable of recognizing a brain tumor neoantigen in a model of recurrent GBM.
Collapse
|
22
|
Shi H, Jiang X, Fu P, Zhou Y, Lu X. Use of dentritic cells pulsed with HLA-A2-restricted MAGE-A1 peptide to generate cytotoxic T lymphocytes against malignant glioma. ACTA ACUST UNITED AC 2010; 30:678-82. [PMID: 21063856 DOI: 10.1007/s11596-010-0564-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Indexed: 12/20/2022]
Abstract
This study developed a novel approach of targeting malignant glioma with pMAGE-A1(278-286)-specific cytotoxic T lymphocytes (CTLs) induced from the peripheral blood mononuclear cells of healthy donors by multiple stimulations with human leukocyte antigen (HLA)-A2-restricted pMAGE-A1(278-286) peptide-pulsed dentritic cells. Cytotoxic assays were performed by the colorimetric CytoTox 96 assay to analyze cytotoxic activity of the induced CTLs against various target cells. The induced CTLs showed approximately 45% specific lysis against T2pMAGE-A1(278-286) (pMAGE-A1(278-286) peptide pulsed T2 cells) and U251 (HLA-A2(+), MAGE-A1(+)) at an effector:target ratio of 40:1, and approximately 5% cytolysis against T2pHIV, A172 (HLA-A2(-), MAGE-A1(+)), K562 and T2 cells without being pulsed with peptide at any effector:target ratio. The specific killing activity of the induced CTLs against T2pMAGE-A1(278-286) and U251 was much more obvious than in any other control group (P<0.05). The cytotoxic activity against the T2pMAGE-A1(278-286) and U251 was significantly eliminated by anti-HLA class I mAb W6/32. These results suggest that pMAGE-A1(278-286) epitope may serve as a surrogate tumor antigen target of specific immunotherapy for treating HLA-A2 patients with malignant glioma.
Collapse
Affiliation(s)
- Haojun Shi
- Department of Radiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | |
Collapse
|
23
|
Saka M, Amano T, Kajiwara K, Yoshikawa K, Ideguchi M, Nomura S, Fujisawa H, Kato S, Fujii M, Ueno K, Hinoda Y, Suzuki M. Vaccine therapy with dendritic cells transfected with Il13ra2 mRNA for glioma in mice. J Neurosurg 2010; 113:270-9. [PMID: 19895199 DOI: 10.3171/2009.9.jns09708] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The Il13ra2 gene is often overexpressed in brain tumors, making Il13ra2 one of the vaccine targets for immunotherapy of glioma. In this study, using a mouse glioma model, the authors tested the hypothesis that vaccination using dendritic cells transfected with Il13ra2 mRNA induces strong immunological antitumor effects. METHODS A plasmid was constructed for transduction of the mRNAs transcribed in vitro into dendritic cells. This was done to transport the intracellular protein efficiently into major histocompatibility complex class II compartments by adding a late endosomal/lysosomal sorting signal to the Il13ra2 gene. The dendritic cells transfected with this Il13ra2 mRNA were injected intraperitoneally into the mouse glioma model at 3 and 10 days after tumor cell implantation. The antitumor effects were estimated based on the survival rate, results of histological analysis, and immunohistochemical findings for immune cells. RESULTS The group treated by vaccination therapy with dendritic cells transfected with Il13ra2 mRNA survived significantly longer than did the control groups. Immunohistochemical analysis revealed that greater numbers of T lymphocytes containing CD4+ and CD8+ T cells were found in the group vaccinated with dendritic cells transfected with Il13ra2 mRNA. CONCLUSIONS These results demonstrate the therapeutic potential of vaccination with dendritic cells transfected with Il13ra2 mRNA for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Makoto Saka
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wang C, Cao S, Yan Y, Ying Q, Jiang T, Xu K, Wu A. TLR9 expression in glioma tissues correlated to glioma progression and the prognosis of GBM patients. BMC Cancer 2010; 10:415. [PMID: 20696081 PMCID: PMC2924315 DOI: 10.1186/1471-2407-10-415] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2009] [Accepted: 08/10/2010] [Indexed: 11/10/2022] Open
Abstract
Background Our study aims to evaluate the expression of TLR9 in glioma tissues, examine the association between TLR9 expression, clinicopathological variables, and glioma patient outcome, we further characterized the direct effects of TLR9 agonist CpG ODN upon the proliferation and invasion of glioma cells in vitro. Methods RT-PCR and immunofluorescence were used to determine the expression of TLR9 in glioma cell lines and clinical glioma samples. Tissue microarry and immunohistochemistry were applied to evaluated TLR9 expression in 292 newly diagnosed glioma and 13 non-neoplastic brain tissues. We further investigated the effect of CpG ODN on the proliferation and invasion of glioma cells in vitro with MTT assays and matrigel transwell assay respectively. Results RT-PCR showed that TLR9 expressed in all the glioma samples and glioma cell lines we examined. The tissue array analysis indicated that TLR9 expression is correlated with malignancy of glioma (p < 0.01). Multivariate Cox regression analysis revealed that TLR9 expression is an independent prognostic factor for PFS of GBM patients(P = 0.026). TLR9 agonist CpG ODN has no significant effect on glioma proliferation, but matrigel transwell analysis showed that TLR9 agonist CpG ODN can significantly enhance glioma invasion in vitro. Conclusions Our data indicated that TLR9 expression increases according to the histopathological grade of glioma, and the TLR9 expression level is related to the PFS of GBM patients. In addition, our findings warrant caution in the directly injection of TLR9 agonist CpG ODN into glioma tissues for the glioma immunotherapy.
Collapse
Affiliation(s)
- Chao Wang
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Tran Thang NN, Derouazi M, Philippin G, Arcidiaco S, Di Berardino-Besson W, Masson F, Hoepner S, Riccadonna C, Burkhardt K, Guha A, Dietrich PY, Walker PR. Immune infiltration of spontaneous mouse astrocytomas is dominated by immunosuppressive cells from early stages of tumor development. Cancer Res 2010; 70:4829-39. [PMID: 20501837 DOI: 10.1158/0008-5472.can-09-3074] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immune infiltration of advanced human gliomas has been shown, but it is doubtful whether these immune cells affect tumor progression. It could be hypothesized that this infiltrate reflects recently recruited immune cells that are immediately overwhelmed by a high tumor burden. Alternatively, if there is earlier immune detection and infiltration of the tumor, the question arises as to when antitumor competency is lost. To address these issues, we analyzed a transgenic mouse model of spontaneous astrocytoma (GFAP-V(12)HA-ras mice), which allows the study of immune interactions with developing glioma, even at early asymptomatic stages. T cells, including a significant proportion of Tregs, are already present in the brain before symptoms develop, followed later by macrophages, natural killer cells, and dendritic cells. The effector potential of CD8 T-cells is defective, with the absence of granzyme B expression and low expression of IFN-gamma, tumor necrosis factor, and interleukin 2. Overall, our results show an early defective endogenous immune response to gliomas, and local accumulation of immunosuppressive cells at the tumor site. Thus, the antiglioma response is not simply overwhelmed at advanced stages of tumor growth, but is counterbalanced by an inhibitory microenvironment from the outset. Nevertheless, we determined that effector molecule expression (granzyme B, IFN-gamma) by brain-infiltrating CD8 T-cells could be enhanced, despite this unfavorable milieu, by strong immune stimuli. This potential to modulate the strong imbalance in local antiglioma immunity is encouraging for the development and optimization of future glioma immunotherapies.
Collapse
|
26
|
Grauer OM, Wesseling P, Adema GJ. Immunotherapy of diffuse gliomas: biological background, current status and future developments. Brain Pathol 2009; 19:674-93. [PMID: 19744040 DOI: 10.1111/j.1750-3639.2009.00315.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Despite aggressive multimodal treatment approaches, the prognosis for patients with diffuse gliomas remains disappointing. Glioma cells often extensively infiltrate in the surrounding brain parenchyma, a phenomenon that helps them to escape surgical removal, radiation exposure and chemotherapy. Moreover, conventional therapy is often associated with considerable local and systemic side effects. Therefore, the development of novel therapeutic approaches is essential to improve the outcome of these patients. Immunotherapy offers the opportunity to specifically target residual radio-and chemoresistant tumor cells without damaging healthy neighboring brain tissue. Significant progress has been made in recent years both in understanding the mechanisms of immune regulation in the central nervous system (CNS) as well as tumor-induced and host-mediated immunosuppression elicited by gliomas. In this review, after discussing the special requirements needed for the initiation and control of immune responses in the CNS, we focus on immunological phenomena observed in glioma patients, discuss different immunological approaches to attack glioma-associated target structures and touch on further strategies to improve the efficacy of immunotherapy of gliomas.
Collapse
Affiliation(s)
- Oliver M Grauer
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | | | | |
Collapse
|
27
|
Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B. Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice. Glia 2009; 57:1458-67. [PMID: 19306372 DOI: 10.1002/glia.20863] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
As the main effector-cell population of the central nervous system, microglia (MG) are considered to play an important immunoregulatory function in a number of pathological conditions such as inflammation, trauma, degenerative disease, and brain tumors. Recent studies, however, have suggested that the anti-neoplastic function of MG may be suppressed in malignant brain tumors. Considering the proposed suppressive role of signal transducers and activators of transcription 3 (Stat3) in antitumor immunity, we evaluated the role of Stat3 inhibition on MG and macrophage (MP) activation and tumor growth in a murine glioma model. N9 MG cells were exposed to GL261 glioma conditioned medium (GL261-CM) and evaluated for Stat3 activity and cytokine expression. Furthermore, the role of Stat3 inhibition on MG and MP activation was studied both in vitro and in vivo. Finally, the effect of Stat3 inhibition on tumor growth was assessed in intracranial GL261 gliomas. GL261-CM increased Stat3 activity in N9 cells in vitro and resulted in overexpression of IL-10 and IL-6, and downregulation of IL1-beta, a pro-inflammatory cytokine. Inhibition of Stat3 by CPA-7 or siRNA reversed glioma-induced cytokine expression profile in N9 cells. Furthermore, inactivation of Stat3 in intracranial GL261 tumors by siRNA resulted in MG/MP activation and tumor growth inhibition. Glioma-induced MG and MP suppression may be mediated thorough Stat3. Inhibition of Stat3 function in tumor MG/MP may result in their activation and can potentially be used as an adjunct immunotherapy approach for gliomas.
Collapse
Affiliation(s)
- Leying Zhang
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Although the safety of vaccine approaches for central nervous system (CNS) malignancies has been established in early phase clinical trials, the success of a vaccine strategy will depend critically on the ability of effector T cells to home in to CNS tumors and durably exert antitumor effects. Based on our recent studies, efficient CNS tumor homing is a characteristic of cytotoxic T lymphocytes (CTLs) with a type 1 phenotype (Tc1), and this appears to be related to the Tc1 response to the type 1 CXC chemokine ligand (CXCL) 10 [also known as interferon (IFN)-inducible protein (IP)-10] and expression of an integrin receptor very late antigen (VLA)-4 on Tc1. In addition, we have previously shown that direct intratumoral delivery of dendritic cells (DCs) ex vivo engineered to secrete IFN-alpha further enhances Tc1 homing via upregulation of CXCL10/IP-10 in the tumor microenvironment. As a means to induce IFN-alpha and CXCL10/IP-10 in the CNS tumor microenvironment in a clinically feasible manner, we used administration of polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose (poly-ICLC), a ligand for toll-like receptor 3 and melanoma differentiation-associated gene 5 (MDA5) in combination with vaccinations targeting CTL epitopes derived from glioma-associated antigens (GAAs). The combination of subcutaneous vaccination and i.m. poly-ICLC administration remarkably promoted systemic induction of antigen GAA-specific Tc1s expressing VLA-4 in the CNS tumors and improved the survival of tumor-bearing mice in the absence of detectable autoimmunity. Based on these data, we have implemented a phase I/II vaccination study using type 1 polarizing DCs loaded with GAA peptides in combination with poly-ICLC in patients with recurrent malignant glioma.
Collapse
Affiliation(s)
- Hideho Okada
- University of Pittsburgh School of Medicine, Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Reactive astrocytes form scar-like perivascular barriers to leukocytes during adaptive immune inflammation of the CNS. J Neurosci 2009; 29:11511-22. [PMID: 19759299 DOI: 10.1523/jneurosci.1514-09.2009] [Citation(s) in RCA: 338] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Factors that regulate leukocyte entry and spread through CNS parenchyma during different types of CNS insults are incompletely understood. Reactive astrocytes have been implicated in restricting the spread of leukocytes from damaged into healthy parenchyma during the acute and local innate inflammatory events that follow CNS trauma, but the roles of reactive astrocytes during the chronic and widespread CNS inflammation associated with adaptive or acquired immune responses are uncertain. Here, we investigated the effects of transgenically targeted ablation of proliferating, scar-forming reactive astrocytes on the acquired immune inflammation associated with experimental autoimmune encephalitis (EAE). In wild-type mice with EAE, we found that reactive astrocytes densely surrounded perivascular clusters of leukocytes in a manner reminiscent of astrocyte scar formation after CNS trauma. Transgenically targeted ablation of proliferating astrocytes disrupted formation of these perivascular scars and was associated with a pronounced and significant increase in leukocyte entry into CNS parenchyma, including immunohistochemically identified macrophages, T lymphocytes and neutrophils. This exacerbated inflammation was associated with a substantially more severe and rapidly fulminant clinical course. These findings provide experimental evidence that reactive astrocytes form scar-like perivascular barriers that restrict the influx of leukocytes into CNS parenchyma and protect CNS function during peripherally initiated, acquired immune inflammatory responses in the CNS. The findings suggest that loss or disruption of astrocyte functions may underlie or exacerbate the inflammation and pathologies associated with autoimmune diseases of the CNS, including multiple sclerosis.
Collapse
|
30
|
Miyazaki T, Moritake K, Yamada K, Hara N, Osago H, Shibata T, Akiyama Y, Tsuchiya M. Indoleamine 2,3-dioxygenase as a new target for malignant glioma therapy. Laboratory investigation. J Neurosurg 2009; 111:230-7. [PMID: 19199463 DOI: 10.3171/2008.10.jns081141] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Indoleamine 2,3-dioxygenase (IDO), a kynurenine pathway (KP) enzyme catalyzing oxidation of the essential amino acid tryptophan (Trp), is thought to be involved in the immune resistance of malignant tumors through T-cell inactivation caused by Trp depletion and metabolite accumulation. Human malignant gliomas may use this strategy to escape immune attack. The object of this study was to investigate the possibility of IDO-dependent Trp depletion by malignant gliomas and the practicability of using an IDO inhibitor together with anticancer drugs to reserve Trp without decreasing the cytotoxicity of the drugs. METHODS The authors studied expression of IDO and other KP enzymes and the effects of an IDO inhibitor, 1-methyl L-tryptophan (1MT), on Trp metabolism and cytotoxicity of anticancer drugs, together with direct measurement of KP metabolites, in cultured human malignant glioma cells. RESULTS Upon interferon-gamma (IFN-gamma) stimulation, the glioma cells greatly increased their IDO mRNA expression concomitant with depletion of Trp. The IDO inhibitor 1MT successfully prevented Trp consumption by the stimulated glioma cells. Combining 1MT with anticancer drugs (temozolomide, bischloroethylnitrosourea [BCNU], etoposide and cisplatin) did not interfere with the drugs' suppression of growth of LN229 glioma cells but rather increased their inhibitory effects on IDO activity. CONCLUSIONS These findings suggest that the robust IDO expression with rapid consumption of Trp in human glioma cells induced by IFN-gamma could lead to immune resistance in glioma cells. Indoleamine 2,3-dioxygenase inhibitors that prevent Trp depletion could be used with anticancer drugs to improve therapeutic effects.
Collapse
Affiliation(s)
- Takeshi Miyazaki
- Department of Neurosurgery, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Richter K, Hausmann J, Staeheli P. Interferon-gamma prevents death of bystander neurons during CD8 T cell responses in the brain. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1799-807. [PMID: 19359516 DOI: 10.2353/ajpath.2009.080897] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
T cells restricted to neurotropic viruses are potentially harmful as their activity may result in the destruction of neurons. In the Borna disease virus (BDV) model, antiviral CD8 T cells entering the brain of infected mice cause neurological disease but no substantial loss of neurons unless the animals lack interferon-gamma (IFN-gamma). We show here that glutamate receptor antagonists failed to prevent BDV-induced neuronal loss in IFN-gamma-deficient mice, suggesting that excitotoxicity resulting from glutamate receptor overstimulation is an unlikely explanation for the neuronal damage. Experiments with IFN-gamma-deficient mice lacking eosinophils indicated that these cells, which specifically accumulate in the infected brains of IFN-gamma-deficient mice, are not responsible for CA1 neuronal death. Interestingly, BDV-induced damage of CA1 neurons was reduced significantly in IFN-gamma-deficient mice lacking perforin, suggesting a key role for CD8 T cells in this pathological process. Specific death of hippocampal CA1 neurons could be triggered by adoptive transfer of BDV-specific CD8 T cells from IFN-gamma-deficient mice into uninfected mice that express transgene-encoded BDV antigen at high level in astrocytes. These results indicate that attack by CD8 T cells that cause the death of CA1 neurons might be directed toward regional astrocytes and that IFN-gamma protects vulnerable CA1 neurons from collateral damage resulting from exposure to potentially toxic substances generated as a result of CD8 T cell-mediated impairment of astrocyte function.
Collapse
Affiliation(s)
- Kirsten Richter
- Department of Virology, University of Freiburg, Freiburg, Germany
| | | | | |
Collapse
|
32
|
Abstract
The development of effective immunotherapy strategies for glioma requires adequate understanding of the unique immunological microenvironment in the central nervous system (CNS) and CNS tumors. Although the CNS is often considered to be an immunologically privileged site and poses unique challenges for the delivery of effector cells and molecules, recent advances in technology and discoveries in CNS immunology suggest novel mechanisms that may significantly improve the efficacy of immunotherapy against gliomas. In this review, we first summarize recent advances in the CNS and CNS tumor immunology. We address factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, with an emphasis on lessons learned from recent early-phase clinical trials. We also discuss novel immunotherapy strategies that have been recently tested in non-CNS tumors and show great potential for application to gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefit for patients with gliomas.
Collapse
Affiliation(s)
- Hideho Okada
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
T-cell mediated immunotherapy is a conceptually attractive treatment option to envisage for glioma, since T lymphocytes can actively seek out neoplastic cells in the brain, and they have the potential to safely and specifically eliminate tumor. Some antigenic targets on glioma cells are already defined, and we can be optimistic that more will be discovered from progress in T-cell epitope identification and gene expression profiling of brain tumors. In parallel, advances in immunology (regional immunology, neuroimmunology, tumor immunology) now equip us to build upon the results from current immunotherapy trials in which the safety and feasibility of brain tumor immunotherapy have already been confirmed. We can now look to the next phase of immunotherapy, in which we must harness the most promising basic science advances and existing clinical expertise, and apply these to randomized clinical trials to determine the real clinical impact and applicability of these approaches for treating patients with currently incurable malignant brain tumors.
Collapse
Affiliation(s)
- Erwin G. Meir
- School of Medicine, Emory University, Clifton Road 1365C, Atlanta, 30322 U.S.A
| |
Collapse
|
34
|
Grauer OM, Molling JW, Bennink E, Toonen LWJ, Sutmuller RPM, Nierkens S, Adema GJ. TLR ligands in the local treatment of established intracerebral murine gliomas. THE JOURNAL OF IMMUNOLOGY 2008; 181:6720-9. [PMID: 18981089 DOI: 10.4049/jimmunol.181.10.6720] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Local TLR stimulation is an attractive approach to induce antitumor immunity. In this study, we compared various TLR ligands for their ability to affect murine GL261 cells in vitro and to eradicate established intracerebral murine gliomas in vivo. Our data show that GL261 cells express TLR2, TLR3, and TLR4 and respond to the corresponding TLR ligands with increasing MHC class I expression and inducing IL-6 secretion in vitro, while TLR5, TLR7, and TLR9 are essentially absent. Remarkably, CpG-oligonucleotides (CpG-ODN, TLR9) appeared to inhibit GL261 cell proliferation in a cell-type specific, but CpG-motif and TLR9-independent manner. A single intratumoral injection of CpG-ODN most effectively inhibited glioma growth in vivo and cured 80% of glioma-bearing C57BL/6 mice. Intratumoral injection of Pam3Cys-SK4 (TLR1/2) or R848 (TLR7) also produced a significant survival benefit, whereas poly(I:C) (TLR3) or purified LPS (TLR4) stimulation alone was not effective. Additional studies using TLR9(+/+) wild-type and TLR9(-/-) knockout mice revealed that the efficacy of local CpG-ODN treatment in vivo required TLR9 expression on nontumor cells. Additional experiments demonstrated increased frequencies of tumor-infiltrating IFN-gamma producing CD4(+) and CD8(+) effector T cells and a marked increase in the ratio of CD4(+) effector T cells to CD4(+)FoxP3(+) regulatory T cells upon CpG-ODN treatment. Surviving CpG-ODN treated mice were also protected from a subsequent tumor challenge without further addition of CpG-ODN. In summary, this study underlines the potency of local TLR treatment in antiglioma therapy and demonstrates that local CpG-ODN treatment most effectively restores antitumor immunity in a therapeutic murine glioma model.
Collapse
Affiliation(s)
- Oliver M Grauer
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Calzascia T, Loh JMS, Di Berardino-Besson W, Masson F, Guillaume P, Burkhardt K, Herrera PL, Dietrich PY, Walker PR. Peripheral tolerance limits CNS accumulation of CD8 T cells specific for an antigen shared by tumor cells and normal astrocytes. Glia 2008; 56:1625-36. [DOI: 10.1002/glia.20715] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Waziri A, Killory B, Ogden AT, Canoll P, Anderson RCE, Kent SC, Anderson DE, Bruce JN. Preferential in situ CD4+CD56+ T cell activation and expansion within human glioblastoma. THE JOURNAL OF IMMUNOLOGY 2008; 180:7673-80. [PMID: 18490770 DOI: 10.4049/jimmunol.180.11.7673] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent evidence suggests that suppression of the cellular immune response is often attributable to populations of functionally distinct T cells that act to down-regulate Ag-specific effector T cells. Using flow cytometry, we evaluated tumor-infiltrating lymphocytes (TIL) from patients undergoing neurosurgical resection of glioblastoma multiforme (GBM), metastatic lung carcinoma, and meningioma for markers known to be expressed on immunoregulatory T cells. Ex vivo phenotypic characteristics, cellular proliferation, and cytokine expression patterns were compared between T cell subsets found in the PBMC and within TIL from fresh tumor samples. Interestingly, nearly half of all T cells infiltrating GBM specimens were CD56(+) T cells, while much smaller percentages of similar cells were identified within metastatic lung tumors and meningiomas. CD56(+) T cells identified within GBM were not canonical, or "invariant," NKT cells, as they demonstrated diverse TCR expression, a primarily CD4 single-positive phenotype, and lack of CD1d reactivity. The percentage of CD56(+) T cells exhibiting evidence of proliferation within GBM was 3- to 4-fold higher than the proportion of proliferating CD56(-) T cells from these lesions. In addition, direct ex vivo analysis of cytokine expression by TIL from GBM demonstrated significant numbers of IL-4/IL-13 positive cells, cytokines that are integral in the cell-mediated repression of tumor immunity in experimental models. We propose that GBM has a unique capacity to recruit and activate CD4(+)CD56(+) T cells, a population that has not been previously described within human tumors.
Collapse
Affiliation(s)
- Allen Waziri
- Department of Neurological Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Yao Y, Wang X, Jin K, Zhu J, Wang Y, Xiong S, Mao Y, Zhou L. B7-H4 is preferentially expressed in non-dividing brain tumor cells and in a subset of brain tumor stem-like cells. J Neurooncol 2008; 89:121-9. [DOI: 10.1007/s11060-008-9601-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Accepted: 04/15/2008] [Indexed: 01/09/2023]
|
38
|
Fujita M, Zhu X, Sasaki K, Ueda R, Low KL, Pollack IF, Okada H. Inhibition of STAT3 promotes the efficacy of adoptive transfer therapy using type-1 CTLs by modulation of the immunological microenvironment in a murine intracranial glioma. THE JOURNAL OF IMMUNOLOGY 2008; 180:2089-98. [PMID: 18250414 DOI: 10.4049/jimmunol.180.4.2089] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A variety of cancers, including malignant gliomas, show aberrant activation of STAT3, which plays a pivotal role in negative regulation of antitumor immunity. We hypothesized that inhibition of STAT3 signals would improve the efficacy of T cell adoptive transfer therapy by reversal of STAT3-induced immunosuppression in a murine GL261 intracranial glioma model. In vitro treatment of GL261 cells with JSI-124, a STAT3 inhibitor, reversed highly phosphorylated status of STAT3. Systemic i.p. administration of JSI-124 in glioma-bearing immunocompetent mice, but not athymic mice, resulted in prolonged survival, suggesting a role of adaptive immunity in the antitumor effect. Furthermore, JSI-124 promoted maturation of tumor-infiltrating CD11c(+) dendritic cells and activation of tumor-conditioned cytotoxic T cells, enhanced dendritic cells and GL261 production of CXCL-10, a critical chemokine for attraction of Tc1 cells. When i.p. JSI-124 administration was combined with i.v. transfer of Pmel-I mouse-derived type-1 CTLs (Tc1), glioma-bearing mice exhibited prolonged survival compared with i.p. JSI-124 or i.v. Tc1 therapy alone. Flow cytometric analyses of brain infiltrating lymphocytes revealed that JSI-124-treatment enhanced the tumor-homing of i.v. transferred Tc1 cells in a CXCL-10-dependent fashion. Systemic JSI-124 administration also up-regulated serum IL-15 levels, and promoted the persistence of transferred Tc1 in the host. These data suggest that systemic inhibition of STAT3 signaling can reverse the suppressive immunological environment of intracranial tumor bearing mice both systemically and locally, thereby promoting the efficacy of adoptive transfer therapy with Tc1.
Collapse
Affiliation(s)
- Mitsugu Fujita
- Department of Neurological Surgery and Brain Tumor Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Prins RM, Shu CJ, Radu CG, Vo DD, Khan-Farooqi H, Soto H, Yang MY, Lin MS, Shelly S, Witte ON, Ribas A, Liau LM. Anti-tumor activity and trafficking of self, tumor-specific T cells against tumors located in the brain. Cancer Immunol Immunother 2008; 57:1279-89. [PMID: 18253732 DOI: 10.1007/s00262-008-0461-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Accepted: 01/15/2008] [Indexed: 02/02/2023]
Abstract
It is commonly believed that T cells have difficulty reaching tumors located in the brain due to the presumed "immune privilege" of the central nervous system (CNS). Therefore, we studied the biodistribution and anti-tumor activity of adoptively transferred T cells specific for an endogenous tumor-associated antigen (TAA), gp100, expressed by tumors implanted in the brain. Mice with pre-established intracranial (i.c.) tumors underwent total body irradiation (TBI) to induce transient lymphopenia, followed by the adoptive transfer of gp100(25-33)-specific CD8+ T cells (Pmel-1). Pmel-1 cells were transduced to express the bioluminescent imaging (BLI) gene luciferase. Following adoptive transfer, recipient mice were vaccinated with hgp100(25-33) peptide-pulsed dendritic cells (hgp100(25-33)/DC) and systemic interleukin 2 (IL-2). This treatment regimen resulted in significant reduction in tumor size and extended survival. Imaging of T cell trafficking demonstrated early accumulation of transduced T cells in lymph nodes draining the hgp100(25-33)/DC vaccination sites, the spleen and the cervical lymph nodes draining the CNS tumor. Subsequently, transduced T cells accumulated in the bone marrow and brain tumor. BLI could also detect significant differences in the expansion of gp100-specific CD8+ T cells in the treatment group compared with mice that did not receive either DC vaccination or IL-2. These differences in BLI correlated with the differences seen both in survival and tumor infiltrating lymphocytes (TIL). These studies demonstrate that peripheral tolerance to endogenous TAA can be overcome to treat tumors in the brain and suggest a novel trafficking paradigm for the homing of tumor-specific T cells that target CNS tumors.
Collapse
Affiliation(s)
- Robert M Prins
- Department of Surgery, Division of Neurosurgery, David Geffen School of Medicine at UCLA, CHS 74-145, 10833 Le Conte Avenue, PO Box 956901, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ueda R, Low KL, Zhu X, Fujita M, Sasaki K, Whiteside TL, Butterfield LH, Okada H. Spontaneous immune responses against glioma-associated antigens in a long term survivor with malignant glioma. J Transl Med 2007; 5:68. [PMID: 18093336 PMCID: PMC2244605 DOI: 10.1186/1479-5876-5-68] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Accepted: 12/19/2007] [Indexed: 12/31/2022] Open
Abstract
Background In patients with high grade glioma, little is known regarding existence of naturally occurring adaptive T cell reactivity against glioma-associated antigens (GAAs). In this report, we characterized GAA-specific CD8+ T cells and innate immune cells in a patient who has survived with anaplastic astrocytoma (AA) for over 12 years without recurrence. Methods Peripheral blood mononuclear cells (PBMCs) derived from the long term survivor with AA were evaluated for the frequency, cytotoxic T lymphocyte (CTL) activity and differentiation status of CD8+ cells recognizing GAA-derived epitopes as well as relative numbers of other immune cell subsets. This patient's AA tissue was evaluated for expression of two GAAs EphA2 and interleukin-13 receptor α2 subunit (IL-13Rα2) by immunohistochemistry. Results The patient's tumor expressed both EphA2 and IL-13Rα2, and in vitro stimulated PBMC demonstrated superior EphA2883–891 and IL-13Rα2345–353-specific CTL reactivity compared to PBMC samples from two other patients with progressing malignant glioma. Unstimulated EphA2883–891-reactive CD8+ T cells contained high numbers of CD45RA-/CCR7- late effector and CD45RA-/CCR7+ central memory cells. Among other leukocyte subsets, elevated numbers of NK-T cells were found. Conclusion To our knowledge, the current study is one of the first demonstrating the presence of antigen-experienced, GAA-reactive CD8+ T cells in a patient who has survived with AA for over 12 years without recurrence. Further studies are warranted to determine whether the status of GAA-reactive CD8+ T cells dictates survival of patients and/or response to therapeutic vaccines.
Collapse
Affiliation(s)
- Ryo Ueda
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Okada H, Lieberman FS, Walter KA, Lunsford LD, Kondziolka DS, Bejjani GK, Hamilton RL, Torres-Trejo A, Kalinski P, Cai Q, Mabold JL, Edington HD, Butterfield LH, Whiteside TL, Potter DM, Schold SC, Pollack IF. Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of patients with malignant gliomas. J Transl Med 2007; 5:67. [PMID: 18093335 PMCID: PMC2254376 DOI: 10.1186/1479-5876-5-67] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 12/19/2007] [Indexed: 02/08/2023] Open
Abstract
Background The prognosis for malignant gliomas remains dismal. We addressed the safety, feasibility and preliminary clinical activity of the vaccinations using autologous glioma cells and interleukin (IL)-4 gene transfected fibroblasts. Methods In University of Pittsburgh Cancer Institute (UPCI) protocol 95-033, adult participants with recurrent glioblastoma multiforme (GBM) or anaplastic astrocytoma (AA) received gross total resection (GTR) of the recurrent tumors, followed by two vaccinations with autologous fibroblasts retrovirally transfected with TFG-IL4-Neo-TK vector admixed with irradiated autologous glioma cells. In UPCI 99-111, adult participants with newly diagnosed GBM or AA, following GTR and radiation therapy, received two intradermal vaccinations with the TFG-IL4-Neo-TK-transfected fibroblasts admixed with type-1 dendritic cells (DC) loaded with autologous tumor lysate. The participants were evaluated for occurrence of adverse events, immune response, and clinical response by radiological imaging. Results and Discussion In UPCI 95-033, only 2 of 6 participants received the vaccinations. Four other participants were withdrawn from the trial because of tumor progression prior to production of the cellular vaccine. However, both participants who received two vaccinations demonstrated encouraging immunological and clinical responses. Biopsies from the local vaccine sites from one participant displayed IL-4 dose-dependent infiltration of CD4+ as well as CD8+ T cells. Interferon (IFN)-γ Enzyme-Linked Immuno-SPOT (ELISPOT) assay in another human leukocyte antigen (HLA)-A2+ participant demonstrated systemic T-cell responses against an HLA-A2-restricted glioma-associated antigen (GAA) epitope EphA2883–891. Moreover, both participants demonstrated clinical and radiological improvement with no evidence of allergic encephalitis, although both participants eventually succumbed with the tumor recurrence. In 99-111, 5 of 6 enrolled participants received scheduled vaccinations with no incidence of major adverse events. Monocyte-derived DCs produced high levels of IL-12 p70. Treatment was well tolerated; however, we were unable to observe detectable IFN-γ post-vaccine responses or prolonged progression-free survival in these participants. Conclusion Feasibility challenges inherent in the generation of a patient-specific gene transfection-based vaccine strongly suggests the need for more practical formulations that would allow for the timely administration of vaccines. Nevertheless, successful generation of type-1 DCs and preliminary safety in the current study provide a strong rationale for further efforts to develop novel glioma vaccines.
Collapse
Affiliation(s)
- Hideho Okada
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Jiang X, Lu X, Liu R, Zhang F, Zhao H. HLA Tetramer Based Artificial Antigen-Presenting Cells Efficiently Stimulate CTLs Specific for Malignant Glioma. Clin Cancer Res 2007; 13:7329-34. [PMID: 18094414 DOI: 10.1158/1078-0432.ccr-07-1025] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The interleukin-13 receptor alpha 2 (IL-13R alpha 2) is a glioma-restricted cell-surface epitope not otherwise detected within the central nervous system. Here, we report a novel approach for targeting malignant glioma with IL-13R alpha 2-specific CTLs. EXPERIMENTAL DESIGN Artificial antigen-presenting cells (aAPC) were made by coating human leukocyte antigen (HLA)-A2/pIL-13R alpha 2(345-354) tetrameric complexes, anti-CD28 antibody, and CD83 molecules to cell-sized latex beads, and used to stimulate IL-13R alpha 2-specific CTLs from the peripheral blood mononuclear cells of HLA-A2+ healthy donors. After multiple stimulations, the induced CTLs were analyzed for tetramer staining, IFN-gamma production, and CTL reactivity. RESULTS Tetramer staining assay showed that the induced CTLs specifically bound HLA-A2/pIL-13R alpha 2(345-354) tetramers. The CTLs specifically produced IFN-gamma in response to the HLA-A2/pIL-13R alpha 2(345-354)-aAPCs and exhibited specific lysis against T2 cells pulsed with the peptide pIL-13R alpha 2(345-354) and HLA-A2+ glioma cells expressing IL-13R alpha 2(345-354), whereas HLA-A2(-) glioma cell lines that express IL-13R alpha 2(345-354) could not be recognized by the CTLs. The peptide-specific activity was inhibited by anti-HLA class I monoclonal antibody. CONCLUSION The induced CTLs specific for IL-13R alpha 2(345-354) peptide could be a potential target of specific immunotherapy for HLA-A2+ patients with malignant glioma.
Collapse
Affiliation(s)
- Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | |
Collapse
|
43
|
Xiaobing J, Xiaoling L, Ruen L, Fangcheng Z, Hongyang Z. Induction of cytotoxic T-lymphocytes specific for malignant glioma by HLA dimer-based artificial antigen-presenting cells. Cancer Biother Radiopharm 2007; 22:826-35. [PMID: 18158774 DOI: 10.1089/cbr.2007.0406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIM The aim of this study was to investigate the novel approach for targeting malignant glioma. METHODS Interleukin-13 receptor alpha2 (IL-13Ralpha2)-specific cytotoxic T-cells (CTLs) were induced from the peripheral blood lymphocytes (PBLs) of human leukocyte antigen (HLA)-A2 positive healthy donors by multiple stimulations with artificial antigen-presenting cells (aAPCs) made by coating HLA-A2-Ig/pIL-13Ralpha2(345-354) dimeric complexes, anti-CD28 antibody, and CD83 molecules to cell-sized latex beads. RESULTS The induced CTLs exhibited a specific lysis against T2 cells pulsed with the peptide pIL-13Ralpha2(345-354) and HLA-A2(+) glioma cells expressing IL-13Ralpha2(345-354), while HLA-A2(-) glioma cell lines that express IL-13Ralpha2(345-354) could not be recognized by the CTLs. The peptide-specific activity was inhibited by the anti-HLA class I monoclonal antibody. CONCLUSIONS The induced CTLs specific for the IL-13Ralpha2(345-354) peptide could be a potential target of specific immunotherapy for HLA-A2(+) patients with malignant glioma.
Collapse
MESH Headings
- Antibodies/chemistry
- Antibodies/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigen Presentation/immunology
- Antigens, CD/chemistry
- Antigens, CD/immunology
- CD28 Antigens/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Gene Expression
- Glioma/immunology
- Glioma/pathology
- Glioma/therapy
- HLA-A2 Antigen/immunology
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Testing
- Humans
- Immunoglobulins/chemistry
- Immunoglobulins/immunology
- Interferon-gamma/metabolism
- Interleukin-13 Receptor alpha2 Subunit/genetics
- Interleukin-13 Receptor alpha2 Subunit/immunology
- Leukocytes, Mononuclear/immunology
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/immunology
- Microspheres
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- CD83 Antigen
Collapse
Affiliation(s)
- Jiang Xiaobing
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | |
Collapse
|
44
|
Ghosh TK, Mickelson DJ, Lipson KE, Alkan SS. Inhibition of in vitro tumor cell proliferation by cytokines induced by combinations of TLR or TLR and TCR agonists. Int Immunopharmacol 2007; 7:1471-82. [PMID: 17761351 DOI: 10.1016/j.intimp.2007.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 06/03/2007] [Accepted: 06/04/2007] [Indexed: 01/12/2023]
Abstract
The objective of this study was to learn from in vitro studies how to better utilize Toll-like receptor (TLR) agonists in controlling tumor growth. One of the primary effects of TLR agonists is induction of cytokine and chemokine production. In order to identify combinations of cytokines or chemokines with optimal ability to inhibit in vitro tumor cell proliferation, a panel of 17 recombinant human or mouse cytokines that have minimal effect on primary cell survival, were tested individually or in combinations of 2, 3 or 4 on a panel of human and mouse chemotherapy sensitive and resistant tumor cell lines. A combination of high (>10 ng/ml) levels of IFNgamma with moderate concentrations of TNFalpha>IFNalpha>IL-6=IL-8 was most effective at inhibiting in vitro tumor cell viability and proliferation with minimal effect on primary cells. We also observed that similar cytokine profile could be induced in vitro PBMC culture by using certain combinations of TLR-TLR and TLR-TCR agonists. Thus, concomitant activation of TLR7/8 with TLR4 or TLR 7/8 with T cell receptor (TCR) in PBMC, amongst all possible paired TLR-TLR and TLR-TCR agonist combinations, produced cytokine mix high in IFNgamma, in combination with IFNalpha, IL-6, IL-8, TNFalpha. Such cytokine mix was equal or more effective tumor cell killing and inhibition of tumor cell proliferation than the best rec-cytokine mixture tested. These results suggest that, TLR and/or TCR agonists combinations generate an optimal mixture of cytokines and chemokines competent in regulating in vitro tumor growth, and imply that realizing such "right cytokine induction" in vivo might be more efficacious than that with individual cytokines or TLR agonists induced cytokine mix.
Collapse
Affiliation(s)
- Tarun K Ghosh
- 3M Pharmaceuticals, Department of Pharmacology, St. Paul, MN 55144 USA.
| | | | | | | |
Collapse
|
45
|
Masson F, Calzascia T, Di Berardino-Besson W, de Tribolet N, Dietrich PY, Walker PR. Brain microenvironment promotes the final functional maturation of tumor-specific effector CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:845-53. [PMID: 17617575 DOI: 10.4049/jimmunol.179.2.845] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
During the priming phase of an antitumor immune response, CD8(+) T cells undergo a program of differentiation driven by professional APCs in secondary lymphoid organs. This leads to clonal expansion and acquisition both of effector functions and a specific adhesion molecule pattern. Whether this program can be reshaped during the effector phase to adapt to the effector site microenvironment is unknown. We investigated this in murine brain tumor models using adoptive transfer of tumor-specific CD8(+) T cells, and in spontaneous immune responses of patients with malignant glioma. Our data show proliferation of Ag-experienced tumor-specific T cells within the brain parenchyma. Moreover, CD8(+) T cells further differentiated in the brain, exhibiting enhanced IFN-gamma and granzyme B expression and induction of alpha(E)(CD103)beta(7) integrin. This unexpected integrin expression identified a subpopulation of CD8(+) T cells conditioned by the brain microenvironment and also had functional consequences: alpha(E)(CD103)beta(7)-expressing CD8(+) T cells had enhanced retention in the brain. These findings were further investigated for CD8(+) T cells infiltrating human malignant glioma; CD8(+) T cells expressed alpha(E)(CD103)beta(7) integrin and granzyme B as in the murine models. Overall, our data indicate that the effector site plays an active role in shaping the effector phase of tumor immunity. The potential for local expansion and functional reprogramming should be considered when optimizing future immunotherapies for regional tumor control.
Collapse
Affiliation(s)
- Frédérick Masson
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
46
|
Jiang X, Lu X, Liu R, Zhang F, Zhao H. Induction of cytotoxic T lymphocytes specific to malignant glioma using T2 cells pulsed with HLA-A2-restricted interleukin-13 receptor alpha 2 peptide in vitro. Acta Biochim Biophys Sin (Shanghai) 2007; 39:641-8. [PMID: 17805458 DOI: 10.1111/j.1745-7270.2007.00331.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Interleukin-13 receptor alpha2 (IL-13Ralpha2) is a glioma-restricted cell-surface epitope not otherwise detected within the central nervous system. The present study is a report of a novel approach of targeting malignant glioma with IL-13Ralpha2-specific cytotoxic T lymphocyte (CTL) induced from the peripheral blood mononuclear cells of healthy donors by multiple stimulations with human leukocyte antigen (HLA)-A2-restricted IL-13Ralpha2(345-353) peptide-pulsed T2 cells. The induced CTL showed specific lysis against T2 cells pulsed with the peptide and HLA-A2+ glioma cells expressing IL-13R2(345-353), while HLA-A2 glioma cell lines that express IL-13Ralpha2(345-353) could not be recognized by CTL. The peptide-specific activity was inhibited by anti-HLA class I monoclonal antibody. These results suggest that the induced CTL specific for IL-13Ralpha2(345-353) peptide could be a potential target of specific immunotherapy for HLA-A2 patients with malignant glioma.
Collapse
Affiliation(s)
- Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | | | | | | | | |
Collapse
|
47
|
Sasaki K, Zhu X, Vasquez C, Nishimura F, Dusak JE, Huang J, Fujita M, Wesa A, Potter DM, Walker PR, Storkus WJ, Okada H. Preferential expression of very late antigen-4 on type 1 CTL cells plays a critical role in trafficking into central nervous system tumors. Cancer Res 2007; 67:6451-8. [PMID: 17616706 DOI: 10.1158/0008-5472.can-06-3280] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have previously shown preferential tumor-homing and therapeutic efficacy of adoptively transferred type 1 CTL (Tc1) when compared with type 2 CTL (Tc2) in mice bearing intracranial ovalbumin-transfected melanoma (M05). Further characterizing the expression of a panel of homing receptors on Tc1 and Tc2 cells, we found that very late antigen (VLA)-4 (a heterodimer of CD49d and CD29), but none of other receptors evaluated, was expressed at significantly higher levels on Tc1 cells than on Tc2 cells. Although CD49d (alpha(4) integrin) can form heterodimers with both beta(1) (CD29) and beta(7) integrins, alpha(4)beta(7) complexes were not expressed by either Tc1 or Tc2 cells, suggesting that CD49d is solely expressed in VLA-4 complexes. VLA-4 expression on Tc2 cells was down-regulated in an interleukin (IL)-4 dose-dependent manner but not by other type 2 cytokines, such as IL-10 and IL-13, suggesting that IL-4 uniquely down-regulates VLA-4 expression on these cells. In accordance with the differential expression of VLA-4 on Tc1 versus Tc2 cells, Tc1 cells alone were competent to adhere to plate-bound VCAM-1-Ig fusion protein. Finally, the efficient trafficking of Tc1 cells into intracranial M05 lesions in vivo was efficiently blocked by administration of monoclonal antibodies against CD49d or VCAM-1 or small interfering RNA-mediated silencing of CD49d on Tc1 cells. Collectively, these data support the critical role of VLA-4 in the effective intracranial tumor homing of adoptive-transferred, antigen-specific Tc1 cells and suggest that more effective vaccine and/or ex vivo T-cell activation regimens may be developed by promoting the generation of VLA-4(+) antitumor Tc1 cells.
Collapse
Affiliation(s)
- Kotaro Sasaki
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Grauer OM, Nierkens S, Bennink E, Toonen LWJ, Boon L, Wesseling P, Sutmuller RPM, Adema GJ. CD4+FoxP3+ regulatory T cells gradually accumulate in gliomas during tumor growth and efficiently suppress antiglioma immune responses in vivo. Int J Cancer 2007; 121:95-105. [PMID: 17315190 DOI: 10.1002/ijc.22607] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The suppressive activity of regulatory T cells (Treg) has been implicated as an important factor limiting immune mediated destruction of tumor cells. However, not much is known about the presence and function of Treg within tumors. Here we show in a syngeneic murine glioma model a time-dependent accumulation of CD4+FoxP3+ Treg in brain tumors. Further analysis revealed a time-dependent upregulation of CD25, CTLA-4, GITR and CXCR4 on intratumoral CD4+FoxP3+ Treg during tumor growth. Moreover, freshly isolated intratumoral Treg were highly suppressive when tested directly ex vivo. Treatment with anti-CD25 monoclonal antibodies (mAbs) significantly reduced the number of these highly suppressive CD4+FoxP3+ cells within the growing tumor and provoked a CD4 and CD8 T cell dependent destruction of the glioma cells. Combining Treg depletion with administration of blocking CTLA-4 mAbs further boosted glioma-specific CD4+ and CD8+ effector T cells as well as antiglioma IgG2a antibody titers resulting in complete tumor eradication without any signs of autoimmunity. These data illustrate that intratumoral accumulation and activation of CD4+FoxP3+ Treg act as a dominant immune escape mechanism for gliomas and underline the importance of controlling tumor-infiltrating Treg in glioma immunotherapy.
Collapse
Affiliation(s)
- Oliver M Grauer
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Hau P, Jachimczak P, Schlingensiepen R, Schulmeyer F, Jauch T, Steinbrecher A, Brawanski A, Proescholdt M, Schlaier J, Buchroithner J, Pichler J, Wurm G, Mehdorn M, Strege R, Schuierer G, Villarrubia V, Fellner F, Jansen O, Straube T, Nohria V, Goldbrunner M, Kunst M, Schmaus S, Stauder G, Bogdahn U, Schlingensiepen KH. Inhibition of TGF-β2 with AP 12009 in Recurrent Malignant Gliomas: From Preclinical to Phase I/II Studies. Oligonucleotides 2007; 17:201-12. [PMID: 17638524 DOI: 10.1089/oli.2006.0053] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transforming growth factor-beta2 (TGF-beta2) is known to suppress the immune response to cancer cells and plays a pivotal role in tumor progression by regulating key mechanisms including proliferation, metastasis, and angiogenesis. For targeted protein suppression the TGF-beta2-specific antisense oligodeoxynucleotide AP 12009 was developed. In vitro experiments have been performed to prove specificity and efficacy of the TGF-beta2 inhibitor AP 12009 employing patient-derived malignant glioma cells as well as peripheral blood mononuclear cells (PBMCs) from patients. Clinically, the antisense compound AP 12009 was assessed in three Phase I/II-studies for the treatment of patients with recurrent or refractory malignant (high-grade) glioma WHO grade III or IV. Although the study was not primarily designed as an efficacy evaluation, prolonged survival compared to literature data and response data were observed, which are very rarely seen in this tumor indication. Two patients experienced long-lasting complete tumor remissions. These results implicate targeted TGF-beta2-suppression using AP 12009 as a promising novel approach for malignant gliomas and other highly aggressive, TGF-beta-2-overexpressing tumors.
Collapse
Affiliation(s)
- Peter Hau
- Department of Neurology, University of Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Yang MY, Zetler PM, Prins RM, Khan-Farooqi H, Liau LM. Immunotherapy for patients with malignant glioma: from theoretical principles to clinical applications. Expert Rev Neurother 2007; 6:1481-94. [PMID: 17078788 DOI: 10.1586/14737175.6.10.1481] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malignant gliomas are the most common type of primary brain tumor and are in great need of novel therapeutic approaches. Advances in treatment have been very modest, significant improvement in survival has been lacking for many decades and prognosis remains dismal. Despite 'gross total' surgical resections and currently available radio-chemotherapy, malignant gliomas inevitably recur due to reservoirs of notoriously invasive tumor cells that infiltrate adjacent and nonadjacent areas of normal brain parenchyma. In principle, the immune system is uniquely qualified to recognize and target these infiltrative pockets of tumor cells, which have generally eluded conventional treatment approaches. In the span of the last 10 years, our understanding of the cancer-immune system relationship has increased exponentially, and yet, we are only beginning to tease apart the intricacies of the CNS and immune cell interactions. This article reviews the complex associations of the immune system with brain tumors. We provide an overview of currently available treatment options for malignant gliomas, existing gaps in our knowledge of brain tumor immunology, and molecular techniques and targets that might be exploited for improved patient stratification and design of 'custom immunotherapeutics'. We will also examine major new immunotherapy approaches that are being actively investigated to treat patients with malignant glioma, and identify some current and future research priorities in this area.
Collapse
Affiliation(s)
- Meng-Yin Yang
- Division of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-6901, USA.
| | | | | | | | | |
Collapse
|