1
|
Uthanaphun T, Manochantr S, Tantrawatpan C, Tantikanlayaporn D, Kheolamai P. PL-hMSC and CH-hMSC derived soluble factors inhibit proliferation but improve hGBM cell migration by activating TGF-β and inhibiting Wnt signaling. Biosci Rep 2024; 44:BSR20231964. [PMID: 38687607 PMCID: PMC11130542 DOI: 10.1042/bsr20231964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/18/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive brain tumors. GBM resists most chemotherapeutic agents, resulting in a high mortality rate in patients. Human mesenchymal stem cells (hMSCs), which are parts of the cancer stroma, have been shown to be involved in the development and progression of GBM. However, different sources of hMSCs might affect GBM cells differently. In the present study, we established hMSCs from placenta (PL-hMSC) and chorion (CH-hMSC) to study the effects of their released soluble factors on the proliferation, migration, invasion, gene expression, and survival of human GBM cells, U251. We found that the soluble factors derived from CH-hMSCs and PL-hMSCs suppressed the proliferation of U251 cells in a dose-dependent manner. In contrast, soluble factors derived from both hMSC sources increased U251 migration without affecting their invasive property. The soluble factors derived from these hMSCs decreased the expression levels of CyclinD1, E2Fs and MYC genes that promote GBM cell proliferation but increased the expression level of TWIST gene, which promotes EMT and GBM cell migration. The functional study suggests that both hMSCs might exert their effects, at least in part, by activating TGF-β and suppressing Wnt/β-catenin signaling in U251 cells. Our study provides a better understanding of the interaction between GBM cells and gestational tissue-derived hMSCs. This knowledge might be used to develop safer and more effective stem cell therapy that improves the survival and quality of life of patients with GBM by manipulating the interaction between hMSCs and GBM cells.
Collapse
Affiliation(s)
- Tanawat Uthanaphun
- Master of Science Program in Stem Cell and Molecular Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Sirikul Manochantr
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Chairat Tantrawatpan
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pakpoom Kheolamai
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| |
Collapse
|
2
|
Alswailem R, Alqahtani FY, Aleanizy FS, Alrfaei BM, Badran M, Alqahtani QH, Abdelhady HG, Alsarra I. MicroRNA-219 loaded chitosan nanoparticles for treatment of glioblastoma. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:198-207. [PMID: 35762105 DOI: 10.1080/21691401.2022.2092123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recent evidence has implicated microRNA-219 (miR-219) in regulation of gene contributed in glioblastoma (GBM) pathogenesis. This study aimed to prepare miR-219 in chitosan (CS) nanoparticles (NPs), characterize and investigate their efficacy on human GBM cell line (U87 MG). NPs were prepared using ionic gelation method. The influence of process parameters on physicochemical characteristics of NPs was investigated. Apoptotic effect of miR-219 was examined on U87 MG cells. Formulated NPs showed particle size of 109 ± 2.18 nm, with poly dispersity index equal to 0.2 ± 0.05, and zeta potential of +20.5 ± 0.7 mV. Entrapment efficiency of miR-219 in loaded NP has reached 95%. The in vitro release study demonstrated sustained release pattern of miR-219 from CS-NPs. Gel retardation assay has confirmed the integrity of miR-219 after production process. The fabricated NPs reduced the survival of U87 MG cells to 78% after 24 h of post-transfection, and into 67.5% after 48 h. However, fibroblasts were not affected by the NPs, revealing their specificity for GBM cells. Given the tumour suppressing function of miR-219, and advantage of CS-NPs for gene delivery to the central nervous system, the presented NPs have a great potential for treatment of GBM.
Collapse
Affiliation(s)
- Rawan Alswailem
- Drug sector, Saudi Food and Drug Authority, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fulwah Yahya Alqahtani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fadilah Sfouq Aleanizy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bahauddeen M Alrfaei
- Department of Cellular Therapy and Cancer Research, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health, Riyadh, Saudi Arabia
| | - Mohammad Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Qamraa Hamad Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Ibrahim Alsarra
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Lee-Chang C. Understanding the relationship between gliomas and T cells: Paving the way for effective immunotherapy. Neuro Oncol 2022; 24:1658-1659. [PMID: 35660928 DOI: 10.1093/neuonc/noac152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL.,The Malnati Brain Tumor Institute, Chicago, IL
| |
Collapse
|
4
|
Chen X, Zhang Y, Zhao Q, Xiaoqun Chen LB, Zhou Z. Management of dermatologic adverse events associated with tumour treating fields in patients with glioblastoma multiforme: A 27-case series. Asia Pac J Oncol Nurs 2022; 9:100095. [PMID: 36060271 PMCID: PMC9428807 DOI: 10.1016/j.apjon.2022.100095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/26/2022] [Indexed: 11/25/2022] Open
|
5
|
Huang J, Chen Z, Park SW, Lai JHC, Chan KWY. Molecular Imaging of Brain Tumors and Drug Delivery Using CEST MRI: Promises and Challenges. Pharmaceutics 2022; 14:451. [PMID: 35214183 PMCID: PMC8880023 DOI: 10.3390/pharmaceutics14020451] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/10/2022] Open
Abstract
Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) detects molecules in their natural forms in a sensitive and non-invasive manner. This makes it a robust approach to assess brain tumors and related molecular alterations using endogenous molecules, such as proteins/peptides, and drugs approved for clinical use. In this review, we will discuss the promises of CEST MRI in the identification of tumors, tumor grading, detecting molecular alterations related to isocitrate dehydrogenase (IDH) and O-6-methylguanine-DNA methyltransferase (MGMT), assessment of treatment effects, and using multiple contrasts of CEST to develop theranostic approaches for cancer treatments. Promising applications include (i) using the CEST contrast of amide protons of proteins/peptides to detect brain tumors, such as glioblastoma multiforme (GBM) and low-grade gliomas; (ii) using multiple CEST contrasts for tumor stratification, and (iii) evaluation of the efficacy of drug delivery without the need of metallic or radioactive labels. These promising applications have raised enthusiasm, however, the use of CEST MRI is not trivial. CEST contrast depends on the pulse sequences, saturation parameters, methods used to analyze the CEST spectrum (i.e., Z-spectrum), and, importantly, how to interpret changes in CEST contrast and related molecular alterations in the brain. Emerging pulse sequence designs and data analysis approaches, including those assisted with deep learning, have enhanced the capability of CEST MRI in detecting molecules in brain tumors. CEST has become a specific marker for tumor grading and has the potential for prognosis and theranostics in brain tumors. With increasing understanding of the technical aspects and associated molecular alterations detected by CEST MRI, this young field is expected to have wide clinical applications in the near future.
Collapse
Affiliation(s)
- Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Se-Weon Park
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Joseph H. C. Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
- Tung Biomedical Science Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
6
|
Li Y, Sharma A, Maciaczyk J, Schmidt-Wolf IGH. Recent Development in NKT-Based Immunotherapy of Glioblastoma: From Bench to Bedside. Int J Mol Sci 2022; 23:ijms23031311. [PMID: 35163235 PMCID: PMC8835986 DOI: 10.3390/ijms23031311] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive and dismal disease with a median overall survival of around 15 months and a 5-year survival rate of 7.2%. Owing to genetic mutations, drug resistance, disruption to the blood–brain barrier (BBB)/blood–brain tumor barrier (BBTB), and the complexity of the immunosuppressive environment, the therapeutic approaches to GBM represent still major challenges. Conventional therapies, including surgery, radiotherapy, and standard chemotherapy with temozolomide, have not resulted in satisfactory improvements in the overall survival of GBM patients. Among cancer immunotherapeutic approaches, we propose that adjuvant NKT immunotherapy with invariant NKT (iNKT) and cytokine-induced killer (CIK) cells may improve the clinical scenario of this devastating disease. Considering this, herein, we discuss the current strategies of NKT therapy for GBM based primarily on in vitro/in vivo experiments, clinical trials, and the combinatorial approaches with future therapeutic potential.
Collapse
Affiliation(s)
- Yutao Li
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Amit Sharma
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
| | - Jarek Maciaczyk
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
- Department of Surgical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ingo G. H. Schmidt-Wolf
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-2871-7050
| |
Collapse
|
7
|
Lee-Chang C, Miska J, Hou D, Rashidi A, Zhang P, Burga RA, Jusué-Torres I, Xiao T, Arrieta VA, Zhang DY, Lopez-Rosas A, Han Y, Sonabend AM, Horbinski CM, Stupp R, Balyasnikova IV, Lesniak MS. Activation of 4-1BBL+ B cells with CD40 agonism and IFNγ elicits potent immunity against glioblastoma. J Exp Med 2021; 218:152130. [PMID: 32991668 PMCID: PMC7527974 DOI: 10.1084/jem.20200913] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/24/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has revolutionized the treatment of many tumors. However, most glioblastoma (GBM) patients have not, so far, benefited from such successes. With the goal of exploring ways to boost anti-GBM immunity, we developed a B cell-based vaccine (BVax) that consists of 4-1BBL+ B cells activated with CD40 agonism and IFNγ stimulation. BVax migrates to key secondary lymphoid organs and is proficient at antigen cross-presentation, which promotes both the survival and the functionality of CD8+ T cells. A combination of radiation, BVax, and PD-L1 blockade conferred tumor eradication in 80% of treated tumor-bearing animals. This treatment elicited immunological memory that prevented the growth of new tumors upon subsequent reinjection in cured mice. GBM patient-derived BVax was successful in activating autologous CD8+ T cells; these T cells showed a strong ability to kill autologous glioma cells. Our study provides an efficient alternative to current immunotherapeutic approaches that can be readily translated to the clinic.
Collapse
Affiliation(s)
- Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - David Hou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Aida Rashidi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Peng Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Rachel A Burga
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ignacio Jusué-Torres
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL.,Department of Neurological Surgery, Loyola University Chicago Stritch School of Medicine, Chicago, IL
| | - Ting Xiao
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Victor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL.,Plan de Estudios Combinados en Medicina, National Autonomous University of Mexico, Mexico City, Mexico
| | - Daniel Y Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Aurora Lopez-Rosas
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yu Han
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Craig M Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL.,Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
8
|
Godoi B, Gritzenco F, Kazmierczak JC, Anjos T, Sperança A, Peixoto MLB, Godoi M, Ledebuhr KNB, Brüning CA, Zamin LL. Base-Free Synthesis and Synthetic Applications of Novel 3-(Organochalcogenyl)prop-2-yn-1-yl Esters: Promising Anticancer Agents. SYNTHESIS-STUTTGART 2021. [DOI: 10.1055/a-1477-6470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AbstractThis manuscript portrays the CuI-catalyzed Csp-chalcogen bond formation through cross-coupling reactions of propynyl esters and diorganyl dichalcogenides by using DMSO as solvent, at room temperature, under base-free and open-to-air atmosphere conditions. Generally, the reactions have proceeded very smoothly, being tolerant to a range of substituents present in both substrates, affording the novel 3-(organochalcogenyl)prop-2-yn-1-yl esters in moderate to good yields. Noteworthy, the 3-(butylselanyl)prop-2-yn-1-yl benzoate proved to be useful as synthetic precursor in palladium-catalyzed Suzuki and Sonogashira type cross-coupling reactions by replacing the carbon–chalcogen bond by new carbon–carbon bond. Moreover, the 3-(phenylselanyl)prop-2-yn-1-yl benzoate has shown promising in vitro activity against glioblastoma cancer cells.
Collapse
Affiliation(s)
- Benhur Godoi
- Programa de Pós-Graduação em Ambiente e Tecnologias Sustentáveis, Núcleo de Síntese, Aplicação e Análise de Compostos Orgânicos e Inorgânicos, Federal University of Fronteira Sul
| | - Fabiane Gritzenco
- Programa de Pós-Graduação em Ambiente e Tecnologias Sustentáveis, Núcleo de Síntese, Aplicação e Análise de Compostos Orgânicos e Inorgânicos, Federal University of Fronteira Sul
| | | | - Thiago Anjos
- Departamento de Química, Universidade Federal de Santa Maria-UFSM
| | | | | | - Marcelo Godoi
- Escola de Química e Alimentos, Universidade Federal do Rio Grande
| | - Kauane N. B. Ledebuhr
- Laboratório de Bioquímica e Neurofarmacologia Molecular, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas
| | - César Augusto Brüning
- Laboratório de Bioquímica e Neurofarmacologia Molecular, Programa de Pós-Graduação em Bioquímica e Bioprospecção, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas
| | - Lauren L. Zamin
- Programa de Pós-Graduação em Ambiente e Tecnologias Sustentáveis, Núcleo de Síntese, Aplicação e Análise de Compostos Orgânicos e Inorgânicos, Federal University of Fronteira Sul
| |
Collapse
|
9
|
Yoon N, Kim HS, Lee JW, Lee EJ, Maeng LS, Yoon WS. Targeted Genomic Sequencing Reveals Different Evolutionary Patterns Between Locally and Distally Recurrent Glioblastomas. Cancer Genomics Proteomics 2021; 17:803-812. [PMID: 33099481 DOI: 10.21873/cgp.20234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/23/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/AIM Glioblastoma is the most malignant form of astrocytoma. The purpose of this study was to analyze the genetic characteristics of primary and recurrent glioblastomas using targeted sequencing and investigate the differences in mutational profiles between the locations of tumor recurrence. MATERIALS AND METHODS Fourteen glioblastoma patients who developed local (n=10) or distal (n=4) recurrence were included in the study. Targeted sequencing analysis was performed using the primary (n=14) and corresponding recurrent (n=14) tumor tissue samples. RESULTS The local and distal recurrence groups showed different genetic evolutionary patterns. Most of the locally recurrent glioblastomas demonstrated concordant mutational profiles between the primary and recurrent tumors, suggesting a linear evolution. In contrast, all cases of distally recurrent glioblastomas showed changes in mutational profiles with newly acquired mutations when compared to the corresponding primary tumors, suggesting a branching evolution. CONCLUSION Locally and distally recurrent glioblastomas exhibit different evolutionary patterns.
Collapse
Affiliation(s)
- Nara Yoon
- Department of Pathology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Hyun-Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Whee Lee
- Department of Radiology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Eui-Jin Lee
- Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Lee-So Maeng
- Department of Pathology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Wan Soo Yoon
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| |
Collapse
|
10
|
Hasbum A, Quintanilla J, Jr JA, Ding MH, Levy A, Chew SA. Strategies to better treat glioblastoma: antiangiogenic agents and endothelial cell targeting agents. Future Med Chem 2021; 13:393-418. [PMID: 33399488 PMCID: PMC7888526 DOI: 10.4155/fmc-2020-0289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive form of glioma, with poor prognosis and high mortality rates. As GBM is a highly vascularized cancer, antiangiogenic therapies to halt or minimize the rate of tumor growth are critical to improving treatment. In this review, antiangiogenic therapies, including small-molecule drugs, nucleic acids and proteins and peptides, are discussed. The authors further explore biomaterials that have been utilized to increase the bioavailability and bioactivity of antiangiogenic factors for better antitumor responses in GBM. Finally, the authors summarize the current status of biomaterial-based targeting moieties that target endothelial cells in GBM to more efficiently deliver therapeutics to these cells and avoid off-target cell or organ side effects.
Collapse
Affiliation(s)
- Asbiel Hasbum
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
| | - Jaqueline Quintanilla
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Juan A Amieva Jr
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - May-Hui Ding
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Arkene Levy
- Dr Kiran C Patel College of Allopathic Medicine, Nova Southeastern University, FL 33314, USA
| | - Sue Anne Chew
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| |
Collapse
|
11
|
Badr CE, Silver DJ, Siebzehnrubl FA, Deleyrolle LP. Metabolic heterogeneity and adaptability in brain tumors. Cell Mol Life Sci 2020; 77:5101-5119. [PMID: 32506168 PMCID: PMC8272080 DOI: 10.1007/s00018-020-03569-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022]
Abstract
The metabolic complexity and flexibility commonly observed in brain tumors, especially glioblastoma, is fundamental for their development and progression. The ability of tumor cells to modify their genetic landscape and adapt metabolically, subverts therapeutic efficacy, and inevitably instigates therapeutic resistance. To overcome these challenges and develop effective therapeutic strategies targeting essential metabolic processes, it is necessary to identify the mechanisms underlying heterogeneity and define metabolic preferences and liabilities of malignant cells. In this review, we will discuss metabolic diversity in brain cancer and highlight the role of cancer stem cells in regulating metabolic heterogeneity. We will also highlight potential therapeutic modalities targeting metabolic vulnerabilities and examine how intercellular metabolic signaling can shape the tumor microenvironment.
Collapse
Affiliation(s)
- Christian E Badr
- Neuro-Oncology Division, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, USA
| | - Daniel J Silver
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, CF24 4HQ, UK
| | - Loic P Deleyrolle
- Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Kim GB, Aragon-Sanabria V, Randolph L, Jiang H, Reynolds JA, Webb BS, Madhankumar A, Lian X, Connor JR, Yang J, Dong C. High-affinity mutant Interleukin-13 targeted CAR T cells enhance delivery of clickable biodegradable fluorescent nanoparticles to glioblastoma. Bioact Mater 2020; 5:624-635. [PMID: 32405577 PMCID: PMC7212185 DOI: 10.1016/j.bioactmat.2020.04.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma (GBM), the deadliest form of brain cancer, presents long-standing problems due to its localization. Chimeric antigen receptor (CAR) T cell immunotherapy has emerged as a powerful strategy to treat cancer. IL-13-receptor-α2 (IL13Rα2), present in over 75% of GBMs, has been recognized as an attractive candidate for anti-glioblastoma therapy. Here, we propose a novel multidisciplinary approach to target brain tumors using a combination of fluorescent, therapeutic nanoparticles and CAR T cells modified with a targeted-quadruple-mutant of IL13 (TQM-13) shown to have high binding affinity to IL13Rα2-expressing glioblastoma cells with low off-target toxicity. Azide-alkyne cycloaddition conjugation of nanoparticles to the surface of T cells allowed a facile, selective, and high-yielding clicking of the nanoparticles. Nanoparticles clicked onto T cells were retained for at least 8 days showing that the linkage is stable and promising a suitable time window for in vivo delivery. T cells clicked with doxorubicin-loaded nanoparticles showed a higher cytotoxic effect in vitro compared to bare T cells. In vitro and in vivo T cells expressing TQM-13 served as delivery shuttles for nanoparticles and significantly increased the number of nanoparticles reaching brain tumors compared to nanoparticles alone. This work represents a new platform to allow the delivery of therapeutic nanoparticles and T cells to solid tumors.
Collapse
Affiliation(s)
- Gloria B. Kim
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Virginia Aragon-Sanabria
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Lauren Randolph
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Hali Jiang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Joshua A. Reynolds
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Becky S. Webb
- Department of Neurosurgery, M. S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, 17033, United States
| | - Achuthamangalam Madhankumar
- Department of Neurosurgery, M. S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, 17033, United States
| | - Xiaojun Lian
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - James R. Connor
- Department of Neurosurgery, M. S. Hershey Medical Center, The Pennsylvania State University, Hershey, PA, 17033, United States
| | - Jian Yang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Cheng Dong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| |
Collapse
|
13
|
Ho YJ, Li JP, Fan CH, Liu HL, Yeh CK. Ultrasound in tumor immunotherapy: Current status and future developments. J Control Release 2020; 323:12-23. [PMID: 32302759 DOI: 10.1016/j.jconrel.2020.04.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022]
Abstract
Immunotherapy has considerable potential in eliminating cancers by activating the host's own immune system, while the thermal and mechanical effects of ultrasound have various applications in tumor therapy. Hyperthermia, ablation, histotripsy, and microbubble stable/inertial cavitation can alter the tumor microenvironment to enhance immunoactivation to inhibit tumor growth. Microbubble cavitation can increase vessel permeability and thereby improve the delivery of immune cells, cytokines, antigens, and antibodies to tumors. Violent microbubble cavitation can disrupt tumor cells and efficiently expose them to numerous antigens so as to promote the maturity of antigen-presenting cells and subsequent adaptive immune-cell activation. This review provides an overview and compares the mechanisms of ultrasound-induced immune modulation for peripheral and brain tumor therapy, even degenerative brain diseases therapy. The possibility of reversing tumors to an immunoactive microenvironment by utilizing the cavitation of microbubbles loaded with therapeutic gases is also proposed as another potential pathway for immunotherapy. Finally, we disuss the challenges and opportunities of ultrasound in immunotherapy for future development.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ju-Pi Li
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang-Gung University, Taoyuan 333, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
14
|
Cucchiara F, Pasqualetti F, Giorgi FS, Danesi R, Bocci G. Epileptogenesis and oncogenesis: An antineoplastic role for antiepileptic drugs in brain tumours? Pharmacol Res 2020; 156:104786. [PMID: 32278037 DOI: 10.1016/j.phrs.2020.104786] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
The first description of epileptic seizures due to brain tumours occurred in 19th century. Nevertheless, after over one hundred years, scientific literature is still lacking on how epilepsy and its treatment can affect tumour burden, progression and clinical outcomes. In patients with brain tumours, epilepsy dramatically impacts their quality of life (QoL). Even antiepileptic therapy seems to affect tumor lesion development. Numerous studies suggest that certain actors involved in epileptogenesis (inflammatory changes, glutamate and its ionotropic and metabotropic receptors, GABA-A and its GABA-AR receptor, as well as certain ligand- and voltage-gated ion channel) may also contribute to tumorigenesis. Although some antiepileptic drugs (AEDs) are known operating on such mechanisms underlying epilepsy and tumor development, few preclinical and clinical studies have tried to investigate them as targets of pharmacological tools acting to control both phenomena. The primary aim of this review is to summarize known determinants and pathophysiological mechanisms of seizures, as well as of cell growth and spread, in patients with brain tumors. Therefore, a special focus will be provided on the anticancer effects of commonly prescribed AEDs (including levetiracetam, valproic acid, oxcarbazepine and others), with an overview of both preclinical and clinical data. Potential clinical applications of this finding are discussed.
Collapse
Affiliation(s)
- Federico Cucchiara
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy; Scuola di Specializzazione in Farmacologia e Tossicologia Clinica, Università di Pisa, Pisa, Italy
| | - Francesco Pasqualetti
- U.O. Radioterapia, Azienda Ospedaliera Universitaria Pisana, Università di Pisa, Italy
| | - Filippo Sean Giorgi
- U.O. Neurologia, Azienda Ospedaliera Universitaria Pisana, Università di Pisa, Pisa, Italy; Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Romano Danesi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy; Scuola di Specializzazione in Farmacologia e Tossicologia Clinica, Università di Pisa, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy; Scuola di Specializzazione in Farmacologia e Tossicologia Clinica, Università di Pisa, Pisa, Italy.
| |
Collapse
|
15
|
Pellosi DS, Paula LB, de Melo MT, Tedesco AC. Targeted and Synergic Glioblastoma Treatment: Multifunctional Nanoparticles Delivering Verteporfin as Adjuvant Therapy for Temozolomide Chemotherapy. Mol Pharm 2019; 16:1009-1024. [PMID: 30698450 DOI: 10.1021/acs.molpharmaceut.8b01001] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite advances in cancer therapies, glioblastoma multiforme treatment remains inefficient due to the brain-blood barrier (BBB) inhibitory activity and to the low temozolomide (TMZ) chemotherapeutic selectivity. To improve therapeutic outcomes, in this work we propose two strategies, (i) photodynamic therapy (PDT) as adjuvant treatment and (ii) engineering of multifunctional theranostic/targeted nanoparticles ( m-NPs) that integrate biotin as a targeting moiety with rhodamine-B as a theranostic agent in pluronic P85/F127 copolymers. These smart m-NPs can surmount the BBB and coencapsulate multiple cargoes under optimized conditions. Overall, the present study conducts a rational m-NP design, characterization, and optimizes the formulation conditions. Confocal microscopy studies on T98-G, U87-MG, and U343 glioblastoma cells and on NIH-3T3 normal fibroblast cells show that the m-NPs and the encapsulated drugs are selectively taken up by tumor cells presenting a broad intracellular distribution. The formulations display no toxicity in the absence of light and are not toxic to healthy cells, but they exert a robust synergic action in cancer cells in the case of concomitant PDT/TMZ treatment, especially at low TMZ concentrations and higher light doses, as demonstrated by nonlinear dose-effect curves based on the Chou-Talalay method. The results evidenced different mechanisms of action related to the disjoint cell cycle phases at the optimal PDT/TMZ ratio. This effect favors synergism between the PDT and the chemotherapy with TMZ, enhances the antiproliferative effect, and overcomes cross-resistance mechanisms. These results point out that m-NP-based PDT adjuvant therapy is a promising strategy to improve TMZ-based glioblastoma multiforme treatments.
Collapse
Affiliation(s)
- Diogo S Pellosi
- Laboratory of Hybrid Materials, Department of Chemistry , Federal University of São Paulo , Diadema 04021-001 , Brazil.,Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Leonardo B Paula
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Maryanne T de Melo
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| | - Antonio C Tedesco
- Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Department of Chemistry FFCLRP , São Paulo University , Ribeirão Preto , Brazil
| |
Collapse
|
16
|
Wang Y, Li P, Wang B, Wang S, Liu P. Identification of myeloid-derived suppressor cells that have an immunosuppressive function in NF2 patients. J Cancer Res Clin Oncol 2019; 145:523-533. [PMID: 30603902 DOI: 10.1007/s00432-018-02825-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE There is no targeted drug therapy for NF2 patients, and surgery or radiosurgery is not always effective. Therefore, the exploration of new therapeutic pathways is urgently needed. METHODS We analyzed the expression of cytokines in the serum of NF2 patients and determined the percentage of HLA-DR-CD33+CD11b+ cells in blood and NF2-associated schwannomas. Furthermore, we analyzed the role of HLA-DR-CD33+CD11b+ cells in inhibiting T-cell proliferation, cytokine production, and transforming growth factor expression. RESULTS NF2 patients are in an immunosuppressed state with elevated IL-10 and TGF-β expression in plasma and the lymphocytes from NF2 patients secrete less IFN-γ and CD3+ T cells proliferate slower than normal healthy donors. HLA-DR-CD33+CD11b+ cells frequency significantly increased in the PBMCs and infiltrated in the tumor, these cells express higher iNOS, NOX2 and TGF-β, and induce TGF-β secretion to inhibit CD8+ T-cell proliferation, and induce T-cell transformation to a CD4+CD25+Foxp3+ regulatory T cells phenotype. NF2-associated schwannoma cells induced monocytes transformation into an HLA-DR-CD33+CD11b+ phenotype, and surgical removal of the tumor reduced the percentage of these cells. CONCLUSIONS HLA-DR-CD33+CD11b+ cells may represent a population of MDSCs in NF2 patients. Dissecting the mechanisms behind these suppressive mechanisms will be helpful for the design of effective immunotherapeutic protocols and likely provide a new effective treatment for NF2 patients.
Collapse
Affiliation(s)
- Ying Wang
- Beijing Neurosurgical Institute, Capital Medical University, No. 6, Tiantan Xili, Chongwen District, Beijing, 100050, China
| | - Peng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Wang
- Beijing Neurosurgical Institute, Capital Medical University, No. 6, Tiantan Xili, Chongwen District, Beijing, 100050, China
| | - Pinan Liu
- Beijing Neurosurgical Institute, Capital Medical University, No. 6, Tiantan Xili, Chongwen District, Beijing, 100050, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Scutti JAB. Importance of immune monitoring approaches and the use of immune checkpoints for the treatment of diffuse intrinsic pontine glioma: From bench to clinic and vice versa (Review). Int J Oncol 2018; 52:1041-1056. [PMID: 29484440 PMCID: PMC5843403 DOI: 10.3892/ijo.2018.4283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
On the basis of immunological results, it is not in doubt that the immune system is able to recognize and eliminate transformed cells. A plethora of studies have investigated the immune system of patients with cancer and how it is prone to immunosuppression, due in part to the decrease in lymphocyte proliferation and cytotoxic activity. The series of experiments published following the demonstration by Dr Allison's group of the potential effect of anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) paved the way for a new perception in cancer immunotherapy: Immune checkpoints. Several T cell-co-stimulatory molecules including cluster of differentiation (CD)28, inducible T cell co-stimulatory, 4-1BB, OX40, glucocorticoid-induced tumor necrosis factor receptor-related gene and CD27, and inhibitory molecules including T cell immunoglobulin and mucin domain-containing-3, programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1), V-domain immunoglobulin suppressor of T cells activation, T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain, and B and T lymphocyte attenuator have been described in regulating T cell functions, and have been demonstrated to be essential targets in immunotherapy. In preclinical studies, glioblastoma multiforme, a high-grade glioma, the monotherapy targeting PD-1/PD-L1 and CTLA-4 resulted in increased survival times. An improved understanding of the pharmacodynamics and immune monitoring on glioma cancers, particularly in diffuse intrinsic pontine glioma (DIPG), an orphan type of cancer, is expected to have a major contribution to the development of novel therapeutic approaches. On the basis of the recent preclinical and clinical studies of glioma, but not of DIPG, the present review makes a claim for the importance of investigating the tumor microenvironment, the immune response and the use of immune checkpoints (agonists or antagonists) in preclinical/clinical DIPG samples by immune monitoring approaches and high-dimensional analysis. Evaluating the potential predictive and correlative biomarkers in preclinical and clinical studies may assist in answering certain crucial questions that may be useful to improve the clinical response in patients with DIPG.
Collapse
|
18
|
Discovery of cell surface vimentin targeting mAb for direct disruption of GBM tumor initiating cells. Oncotarget 2018; 7:72021-72032. [PMID: 27713131 PMCID: PMC5342141 DOI: 10.18632/oncotarget.12458] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/29/2016] [Indexed: 01/10/2023] Open
Abstract
Intracellular vimentin overexpression has been associated with epithelial–mesenchymal transition, metastasis, invasion, and proliferation, but cell surface vimentin (CSV) is less understood. Furthermore, it remains unknown whether CSV can serve as a therapeutic target in CSV-expressing tumor cells. We found that CSV was present on glioblastoma multiforme (GBM) cancer stem cells and that CSV expression was associated with spheroid formation in those cells. A newly developed monoclonal antibody against CSV, 86C, specifically and significantly induced apoptosis and inhibited spheroid formation in GBM cells in vitro. The addition of 86C to GBM cells in vitro also led to rapid internalization of vimentin and decreased GBM cell viability. These findings were associated with an increase in caspase-3 activity, indicating activation of apoptosis. Finally, treatment with 86C inhibited GBM progression in vivo. In conclusion, CSV-expressing GBM cells have properties of tumor initiating cells, and targeting CSV with the monoclonal antibody 86C is a promising approach in the treatment of GBM.
Collapse
|
19
|
McFarland BC, Marks MP, Rowse AL, Fehling SC, Gerigk M, Qin H, Benveniste EN. Loss of SOCS3 in myeloid cells prolongs survival in a syngeneic model of glioma. Oncotarget 2018; 7:20621-35. [PMID: 26967393 PMCID: PMC4991480 DOI: 10.18632/oncotarget.7992] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/16/2016] [Indexed: 12/22/2022] Open
Abstract
In glioma, microglia and macrophages are the largest population of tumor-infiltrating cells, referred to as glioma associated macrophages (GAMs). Herein, we sought to determine the role of Suppressor of Cytokine Signaling 3 (SOCS3), a negative regulator of Signal Transducer and Activator of Transcription 3 (STAT3), in GAM functionality in glioma. We utilized a conditional model in which SOCS3 deletion is restricted to the myeloid cell population. We found that SOCS3-deficient bone marrow-derived macrophages display enhanced and prolonged expression of pro-inflammatory M1 cytokines when exposed to glioma tumor cell conditioned medium in vitro. Moreover, we found that deletion of SOCS3 in the myeloid cell population delays intracranial tumor growth and increases survival of mice bearing orthotopic glioma tumors in vivo. Although intracranial tumors from mice with SOCS3-deficient myeloid cells appear histologically similar to control mice, we observed that loss of SOCS3 in myeloid cells results in decreased M2 polarized macrophage infiltration in the tumors. Furthermore, loss of SOCS3 in myeloid cells results in increased CD8+ T-cell and decreased regulatory T-cell infiltration in the tumors. These findings demonstrate a beneficial effect of M1 polarized macrophages on suppressing glioma tumor growth, and highlight the importance of immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Braden C McFarland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Margaret P Marks
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amber L Rowse
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samuel C Fehling
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Magda Gerigk
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
20
|
Tarasova IA, Tereshkova AV, Lobas AA, Solovyeva EM, Sidorenko AS, Gorshkov V, Kjeldsen F, Bubis JA, Ivanov MV, Ilina IY, Moshkovskii SA, Chumakov PM, Gorshkov MV. Comparative proteomics as a tool for identifying specific alterations within interferon response pathways in human glioblastoma multiforme cells. Oncotarget 2018; 9:1785-1802. [PMID: 29416731 PMCID: PMC5788599 DOI: 10.18632/oncotarget.22751] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/27/2017] [Indexed: 12/13/2022] Open
Abstract
An acquisition of increased sensitivity of cancer cells to viruses is a common outcome of malignant progression that justifies the development of oncolytic viruses as anticancer therapeutics. Studying molecular changes that underlie the sensitivity to viruses would help to identify cases where oncolytic virus therapy would be most effective. We quantified changes in protein abundances in two glioblastoma multiforme (GBM) cell lines that differ in the ability to induce resistance to vesicular stomatitis virus (VSV) infection in response to type I interferon (IFN) treatment. In IFN-treated samples we observed an up-regulation of protein products of some IFN-regulated genes (IRGs). In total, the proteome analysis revealed up to 20% more proteins encoded by IRGs in the glioblastoma cell line, which develops resistance to VSV infection after pre-treatment with IFN. In both cell lines protein-protein interaction and signaling pathway analyses have revealed a significant stimulation of processes related to type I IFN signaling and defense responses to viruses. However, we observed a deficiency in STAT2 protein in the VSV-sensitive cell line that suggests a de-regulation of the JAK/STAT/IRF9 signaling. The study has shown that the up-regulation of IRG proteins induced by the IFNα treatment of GBM cells can be detected at the proteome level. Similar analyses could be applied for revealing functional alterations within the antiviral mechanisms in glioblastoma samples, accompanying by acquisition of sensitivity to oncolytic viruses. The approach can be useful for discovering the biomarkers that predict a potential sensitivity of individual glioblastoma tumors to oncolytic virus therapy.
Collapse
Affiliation(s)
- Irina A Tarasova
- Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alesya V Tereshkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Chumakov Institute of Poliomyelitis and Viral Encephalitides, Russian Academy of Sciences, 142782 Moscow, Russia
| | - Anna A Lobas
- Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Elizaveta M Solovyeva
- Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Alena S Sidorenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir Gorshkov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Frank Kjeldsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Julia A Bubis
- Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Mark V Ivanov
- Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Irina Y Ilina
- Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Sergei A Moshkovskii
- Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Peter M Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Chumakov Institute of Poliomyelitis and Viral Encephalitides, Russian Academy of Sciences, 142782 Moscow, Russia
| | - Mikhail V Gorshkov
- Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| |
Collapse
|
21
|
Intratumoral CD40 activation and checkpoint blockade induces T cell-mediated eradication of melanoma in the brain. Nat Commun 2017; 8:1447. [PMID: 29129918 PMCID: PMC5682289 DOI: 10.1038/s41467-017-01572-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 09/26/2017] [Indexed: 01/07/2023] Open
Abstract
CD40 agonists bind the CD40 molecule on antigen-presenting cells and activate them to prime tumor-specific CD8+ T cell responses. Here, we study the antitumor activity and mechanism of action of a nonreplicating adenovirus encoding a chimeric, membrane-bound CD40 ligand (ISF35). Intratumoral administration of ISF35 in subcutaneous B16 melanomas generates tumor-specific, CD8+ T cells that express PD-1 and suppress tumor growth. Combination therapy of ISF35 with systemic anti-PD-1 generates greater antitumor activity than each respective monotherapy. Triple combination of ISF35, anti-PD-1, and anti-CTLA-4 results in complete eradication of injected and noninjected subcutaneous tumors, as well as melanoma tumors in the brain. Therapeutic efficacy is associated with increases in the systemic level of tumor-specific CD8+ T cells, and an increased ratio of intratumoral CD8+ T cells to CD4+ Tregs. These results provide a proof of concept of systemic antitumor activity after intratumoral CD40 triggering with ISF35 in combination with checkpoint blockade for multifocal cancer, including the brain. Treatment options for metastatic melanoma are limited. Here the authors show that combining an immunostimulant adenovirus, currently in clinical trials for leukemia, with immune checkpoints blockade (ICB) results in systemic eradication of ICB resistant melanoma tumours from both skin and brain of mice.
Collapse
|
22
|
Ma B, Gao Z, Lou J, Zhang H, Yuan Z, Wu Q, Li X, Zhang B. Long non-coding RNA MEG3 contributes to cisplatin-induced apoptosis via inhibition of autophagy in human glioma cells. Mol Med Rep 2017; 16:2946-2952. [DOI: 10.3892/mmr.2017.6897] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 04/06/2017] [Indexed: 11/05/2022] Open
|
23
|
The Process and Regulatory Components of Inflammation in Brain Oncogenesis. Biomolecules 2017; 7:biom7020034. [PMID: 28346397 PMCID: PMC5485723 DOI: 10.3390/biom7020034] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/09/2017] [Accepted: 03/22/2017] [Indexed: 12/17/2022] Open
Abstract
Central nervous system tumors comprising the primary cancers and brain metastases remain the most lethal neoplasms and challenging to treat. Substantial evidence points to a paramount role for inflammation in the pathology leading to gliomagenesis, malignant progression and tumor aggressiveness in the central nervous system (CNS) microenvironment. This review summarizes the salient contributions of oxidative stress, interleukins, tumor necrosis factor-α(TNF-α), cyclooxygenases, and transcription factors such as signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) and the associated cross-talks to the inflammatory signaling in CNS cancers. The roles of reactive astrocytes, tumor associated microglia and macrophages, metabolic alterations, microsatellite instability, O6-methylguanine DNA methyltransferase (MGMT) DNA repair and epigenetic alterations mediated by the isocitrate dehydrogenase 1 (IDH1) mutations have been discussed. The inflammatory pathways with relevance to the brain cancer treatments have been highlighted.
Collapse
|
24
|
van Gool SW, Holm S, Rachor J, Adamson L, Technau A, Maass E, Frühwald MC, Schlegel PG, Eyrich M. Immunotherapy in atypical teratoid-rhabdoid tumors: Data from a survey of the HGG-Immuno Group. Cytotherapy 2016; 18:1178-86. [DOI: 10.1016/j.jcyt.2016.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/29/2016] [Accepted: 06/04/2016] [Indexed: 11/24/2022]
|
25
|
Ladomersky E, Zhai L, Gritsina G, Genet M, Lauing KL, Wu M, James CD, Wainwright DA. Advanced age negatively impacts survival in an experimental brain tumor model. Neurosci Lett 2016; 630:203-208. [PMID: 27493076 DOI: 10.1016/j.neulet.2016.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 07/28/2016] [Accepted: 08/01/2016] [Indexed: 12/23/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults, with an average age of 64 years at the time of diagnosis. To study GBM, a number of mouse brain tumor models have been utilized. In these animal models, subjects tend to range from 6 to 12 weeks of age, which is analogous to that of a human teenager. Here, we examined the impact of age on host immunity and the gene expression associated with immune evasion in immunocompetent mice engrafted with syngeneic intracranial GL261. The data indicate that, in mice with brain tumors, youth conveys an advantage to survival. While age did not affect the tumor-infiltrating T cell phenotype or quantity, we discovered that old mice express higher levels of the immunoevasion enzyme, IDO1, which was decreased by the presence of brain tumor. Interestingly, other genes associated with promoting immunosuppression including CTLA-4, PD-L1 and FoxP3, were unaffected by age. These data highlight the possibility that IDO1 contributes to faster GBM outgrowth with advanced age, providing rationale for future investigation into immunotherapeutic targeting in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | - C David James
- Department of Neurological Surgery, USA; Department of Biochemistry and Molecular Genetics, USA; Northwestern Brain Tumor Institute,USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, USA; Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Northwestern Brain Tumor Institute,USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
26
|
Ladomersky E, Genet M, Zhai L, Gritsina G, Lauing KL, Lulla RR, Fangusaro J, Lenzen A, Kumthekar P, Raizer JJ, Binder DC, James CD, Wainwright DA. Improving vaccine efficacy against malignant glioma. Oncoimmunology 2016; 5:e1196311. [PMID: 27622066 DOI: 10.1080/2162402x.2016.1196311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 12/19/2022] Open
Abstract
The effective treatment of adult and pediatric malignant glioma is a significant clinical challenge. In adults, glioblastoma (GBM) accounts for the majority of malignant glioma diagnoses with a median survival of 14.6 mo. In children, malignant glioma accounts for 20% of primary CNS tumors with a median survival of less than 1 y. Here, we discuss vaccine treatment for children diagnosed with malignant glioma, through targeting EphA2, IL-13Rα2 and/or histone H3 K27M, while in adults, treatments with RINTEGA, Prophage Series G-100 and dendritic cells are explored. We conclude by proposing new strategies that are built on current vaccine technologies and improved upon with novel combinatorial approaches.
Collapse
Affiliation(s)
- Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Matthew Genet
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Galina Gritsina
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Rishi R Lulla
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Ann & Robert Lurie Children's Hospital of Northwestern University, Chicago, IL, USA
| | - Jason Fangusaro
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Ann & Robert Lurie Children's Hospital of Northwestern University, Chicago, IL, USA
| | - Alicia Lenzen
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Ann & Robert Lurie Children's Hospital of Northwestern University, Chicago, IL, USA
| | - Priya Kumthekar
- Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jeffrey J Raizer
- Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David C Binder
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA; Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - C David James
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
27
|
Immunological Evasion in Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7487313. [PMID: 27294132 PMCID: PMC4884578 DOI: 10.1155/2016/7487313] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 04/19/2016] [Indexed: 12/25/2022]
Abstract
Glioblastoma is the most aggressive tumor in Central Nervous System in adults. Among its features, modulation of immune system stands out. Although immune system is capable of detecting and eliminating tumor cells mainly by cytotoxic T and NK cells, tumor microenvironment suppresses an effective response through recruitment of modulator cells such as regulatory T cells, monocyte-derived suppressor cells, M2 macrophages, and microglia as well as secretion of immunomodulators including IL-6, IL-10, CSF-1, TGF-β, and CCL2. Other mechanisms that induce immunosuppression include enzymes as indolamine 2,3-dioxygenase. For this reason it is important to develop new therapies that avoid this immune evasion to promote an effective response against glioblastoma.
Collapse
|
28
|
Razavi SM, Lee KE, Jin BE, Aujla PS, Gholamin S, Li G. Immune Evasion Strategies of Glioblastoma. Front Surg 2016; 3:11. [PMID: 26973839 PMCID: PMC4773586 DOI: 10.3389/fsurg.2016.00011] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/10/2016] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is the most devastating brain tumor, with associated poor prognosis. Despite advances in surgery and chemoradiation, the survival of afflicted patients has not improved significantly in the past three decades. Immunotherapy has been heralded as a promising approach in treatment of various cancers; however, the immune privileged environment of the brain usually curbs the optimal expected response in central nervous system malignancies. In addition, GBM cells create an immunosuppressive microenvironment and employ various methods to escape immune surveillance. The purpose of this review is to highlight the strategies by which GBM cells evade the host immune system. Further understanding of these strategies and the biology of this tumor will pave the way for developing novel immunotherapeutic approaches for treatment of GBM.
Collapse
Affiliation(s)
- Seyed-Mostafa Razavi
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Karen E Lee
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Benjamin E Jin
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Parvir S Aujla
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Sharareh Gholamin
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, CA , USA
| | - Gordon Li
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
29
|
Domingues P, González-Tablas M, Otero Á, Pascual D, Miranda D, Ruiz L, Sousa P, Ciudad J, Gonçalves JM, Lopes MC, Orfao A, Tabernero MD. Tumor infiltrating immune cells in gliomas and meningiomas. Brain Behav Immun 2016. [PMID: 26216710 DOI: 10.1016/j.bbi.2015.07.019] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tumor-infiltrating immune cells are part of a complex microenvironment that promotes and/or regulates tumor development and growth. Depending on the type of cells and their functional interactions, immune cells may play a key role in suppressing the tumor or in providing support for tumor growth, with relevant effects on patient behavior. In recent years, important advances have been achieved in the characterization of immune cell infiltrates in central nervous system (CNS) tumors, but their role in tumorigenesis and patient behavior still remain poorly understood. Overall, these studies have shown significant but variable levels of infiltration of CNS tumors by macrophage/microglial cells (TAM) and to a less extent also lymphocytes (particularly T-cells and NK cells, and less frequently also B-cells). Of note, TAM infiltrate gliomas at moderate numbers where they frequently show an immune suppressive phenotype and functional behavior; in contrast, infiltration by TAM may be very pronounced in meningiomas, particularly in cases that carry isolated monosomy 22, where the immune infiltrates also contain greater numbers of cytotoxic T and NK-cells associated with an enhanced anti-tumoral immune response. In line with this, the presence of regulatory T cells, is usually limited to a small fraction of all meningiomas, while frequently found in gliomas. Despite these differences between gliomas and meningiomas, both tumors show heterogeneous levels of infiltration by immune cells with variable functionality. In this review we summarize current knowledge about tumor-infiltrating immune cells in the two most common types of CNS tumors-gliomas and meningiomas-, as well as the role that such immune cells may play in the tumor microenvironment in controlling and/or promoting tumor development, growth and control.
Collapse
Affiliation(s)
- Patrícia Domingues
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María González-Tablas
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Álvaro Otero
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Daniel Pascual
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - David Miranda
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Laura Ruiz
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Pablo Sousa
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Juana Ciudad
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | | | - María Celeste Lopes
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Alberto Orfao
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María Dolores Tabernero
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain; Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain; Instituto de Estudios de Ciencias de la salud de Castilla y León (IECSCYL-IBSAL) and Research Unit of the University Hospital of Salamanca, Salamanca, Spain.
| |
Collapse
|
30
|
de Magalhães KCSF, Vaz JPM, Gontijo PAM, de Carvalho GTC, Christo PP, Simões RT, da Silva KR. Profile of patients with brain tumors and the role of nursing care. Rev Bras Enferm 2016; 69:138-43. [PMID: 26871228 DOI: 10.1590/0034-7167.2016690120i] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/01/2015] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE to describe the profile of 200 patients with central nervous system tumors (CNST), and the role of the nursing care. METHOD prospective, quantitative and descriptive analysis of medical records of 200 patients with TSNC. RESULTS a total of 61% of our patients had benign CNST and 39% had malignant tumors. The extent of patient dependence, according to the Karnofsky Performance Status scale, was significantly greater for patients with malignant CNST (p < .05), indicating that these patients needed more support with their activities of daily living. CONCLUSION patients with CNST need specialized care, with specific guidance regarding their disease and aspects of daily living after treatment. Thus, the nurse can function as a key element for the effectiveness of care provided to patients and family members with the aim of enhancing the quality of life of all those affected, directly or indirectly, by the disease.
Collapse
Affiliation(s)
| | | | | | | | - Paulo Pereira Christo
- Institute of Education and Research, Santa Casa de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Renata Toscano Simões
- Institute of Education and Research, Santa Casa de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Karla Rona da Silva
- Institute of Education and Research, Santa Casa de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
31
|
Binder DC, Davis AA, Wainwright DA. Immunotherapy for cancer in the central nervous system: Current and future directions. Oncoimmunology 2016; 5:e1082027. [PMID: 27057463 PMCID: PMC4801467 DOI: 10.1080/2162402x.2015.1082027] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/03/2015] [Accepted: 08/05/2015] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and still remains incurable. Although immunotherapeutic vaccination against GBM has demonstrated immune-stimulating activity with some promising survival benefits, tumor relapse is common, highlighting the need for additional and/or combinatorial approaches. Recently, antibodies targeting immune checkpoints were demonstrated to generate impressive clinical responses against advanced melanoma and other malignancies, in addition to showing potential for enhancing vaccination and radiotherapy (RT). Here, we summarize the current knowledge of central nervous system (CNS) immunosuppression, evaluate past and current immunotherapeutic trials and discuss promising future immunotherapeutic directions to treat CNS-localized malignancies.
Collapse
Affiliation(s)
- David C. Binder
- Commitee on Cancer Biology
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Andrew A. Davis
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Derek A. Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
32
|
Gopalakrishnan V, Tao RH, Dobson T, Brugmann W, Khatua S. Medulloblastoma development: tumor biology informs treatment decisions. CNS Oncol 2015; 4:79-89. [PMID: 25768332 DOI: 10.2217/cns.14.58] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Medulloblastoma is the most common malignant pediatric brain tumor. Current treatments including surgery, craniospinal radiation and high-dose chemotherapy have led to improvement in survival. However, the risk for recurrence as well as significant long-term neurocognitive and endocrine sequelae associated with current treatment modalities underscore the urgent need for novel tumor-specific, normal brain-sparing therapies. It has also provided the impetus for research focused on providing a better understanding of medulloblastoma biology. The expectation is that such studies will lead to the identification of new therapeutic targets and eventually to an increase in personalized treatment approaches.
Collapse
Affiliation(s)
- Vidya Gopalakrishnan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
33
|
Okada H, Weller M, Huang R, Finocchiaro G, Gilbert MR, Wick W, Ellingson BM, Hashimoto N, Pollack IF, Brandes AA, Franceschi E, Herold-Mende C, Nayak L, Panigrahy A, Pope WB, Prins R, Sampson JH, Wen PY, Reardon DA. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol 2015; 16:e534-e542. [PMID: 26545842 PMCID: PMC4638131 DOI: 10.1016/s1470-2045(15)00088-1] [Citation(s) in RCA: 502] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/13/2015] [Accepted: 06/15/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy is a promising area of therapy in patients with neuro-oncological malignancies. However, early-phase studies show unique challenges associated with the assessment of radiological changes in response to immunotherapy reflecting delayed responses or therapy-induced inflammation. Clinical benefit, including long-term survival and tumour regression, can still occur after initial disease progression or after the appearance of new lesions. Refinement of the response assessment criteria for patients with neuro-oncological malignancies undergoing immunotherapy is therefore warranted. Herein, a multinational and multidisciplinary panel of neuro-oncology immunotherapy experts describe immunotherapy Response Assessment for Neuro-Oncology (iRANO) criteria based on guidance for the determination of tumour progression outlined by the immune-related response criteria and the RANO working group. Among patients who demonstrate imaging findings meeting RANO criteria for progressive disease within 6 months of initiating immunotherapy, including the development of new lesions, confirmation of radiographic progression on follow-up imaging is recommended provided that the patient is not significantly worse clinically. The proposed criteria also include guidelines for the use of corticosteroids. We review the role of advanced imaging techniques and the role of measurement of clinical benefit endpoints including neurological and immunological functions. The iRANO guidelines put forth in this Review will evolve successively to improve their usefulness as further experience from immunotherapy trials in neuro-oncology accumulate.
Collapse
Affiliation(s)
- Hideho Okada
- Department of Neurological Surgery, University of California, San
Francisco, San Francisco, CA, USA
| | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich,
Switzerland
| | - Raymond Huang
- Department of Radiology, Brigham and Women's Hospital, Boston,
MA, USA
| | | | - Mark R. Gilbert
- Neuro-Oncology Branch, National Institutes of Health, Bethesda,
MD, USA
| | - Wolfgang Wick
- Department of Neurooncology, Heidelberg University Hospital,
Heidelberg, Germany
| | - Benjamin M. Ellingson
- Departments of Radiological Sciences, Bioengineering, Biomedical
Physics, and Psychiatry David Geffen School of Medicine University of California, Los
Angeles Los Angeles, CA, USA
| | - Naoya Hashimoto
- Department of Neurosurgery, Osaka University Graduate School of
Medicine, Suita, Osaka, Japan
| | - Ian F. Pollack
- Department of Neurological Surgery, University of Pittsburgh
School of Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Alba A. Brandes
- Department of Medical Oncology, Azienda USL–IRCCS
Institute of Neurological Science, Bologna, Italy
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda USL–IRCCS
Institute of Neurological Science, Bologna, Italy
| | - Christel Herold-Mende
- Department of Neurosurgery, Division of Experimental
Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute,
Boston, MA, USA
| | - Ashok Panigrahy
- Department of Radiology, University of Pittsburgh School of
Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Whitney B. Pope
- Department of Radiology, David Geffen School of Medicine at
University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert Prins
- Department of Neurosurgery, David Geffen School of Medicine at
University of California, Los Angeles, Los Angeles, CA, USA
| | - John H. Sampson
- Department of Neurosurgery, Duke University School of Medicine,
Durham, NC, USA
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute,
Boston, MA, USA
| | - David A. Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute,
Boston, MA, USA
| |
Collapse
|
34
|
Yan J, Kong LY, Hu J, Gabrusiewicz K, Dibra D, Xia X, Heimberger AB, Li S. FGL2 as a Multimodality Regulator of Tumor-Mediated Immune Suppression and Therapeutic Target in Gliomas. J Natl Cancer Inst 2015; 107:djv137. [PMID: 25971300 DOI: 10.1093/jnci/djv137] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fibrinogen-like protein 2 (FGL2) may promote glioblastoma multiforme (GBM) cancer development by inducing multiple immune-suppression mechanisms. METHODS The biological significance of FGL2 expression was assessed using the The Cancer Genome Atlast (TCGA) glioma database and tumor lysates analysis. The therapeutic effects of an anti-Fgl2 antibody and the role of immune suppression regulation by Fgl2 were determined in immune-competent, NOD-scid IL2Rgammanull (NSG), and FcɣRIIB-/- mice (n = 3-18 per group). Data were analyzed with two-way analysis of variance, log-rank survival analysis, and Pearson correlation. All statistical tests were two-sided. RESULTS In low-grade gliomas, 72.5% of patients maintained two copies of the FGL2 gene, whereas 83.8% of GBM patients had gene amplification or copy gain. Patients with high levels of FGL2 mRNA in glioma tissues had a lower overall survival (P = .009). Protein levels of FGL2 in GBM lysates were higher relative to low-grade glioma lysates (11.48±5.75ng/mg vs 3.96±1.01ng/mg, P = .003). In GL261 mice treated with an anti-FGL2 antibody, median survival was 27 days compared with only 17 days for mice treated with an isotype control antibody (P = .01). The anti-FGL2 antibody treatment reduced CD39(+) Tregs, M2 macrophages, programmed cell death protein 1 (PD-1), and myeloid-derived suppressor cells (MDSCs). FGL2-induced increases in M2, CD39, and PD-1 were ablated in FcɣRIIB-/- mice. CONCLUSIONS FGL2 augments glioma immunosuppression by increasing the expression levels of PD-1 and CD39, expanding the frequency of tumor-supportive M2 macrophages via the FcγRIIB pathway, and enhancing the number of MDSCs and CD39(+) regulatory T cells. Collectively, these results show that FGL2 functions as a key immune-suppressive modulator and has potential as an immunotherapeutic target for treating GBM.
Collapse
Affiliation(s)
- Jun Yan
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Ling-Yuan Kong
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jiemiao Hu
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Konrad Gabrusiewicz
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Denada Dibra
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Xueqing Xia
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Amy B Heimberger
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX.
| | - Shulin Li
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX.
| |
Collapse
|
35
|
Raychaudhuri B, Rayman P, Huang P, Grabowski M, Hambardzumyan D, Finke JH, Vogelbaum MA. Myeloid derived suppressor cell infiltration of murine and human gliomas is associated with reduction of tumor infiltrating lymphocytes. J Neurooncol 2015; 122:293-301. [DOI: 10.1007/s11060-015-1720-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 01/03/2015] [Indexed: 01/02/2023]
|
36
|
Abstract
Eph receptor tyrosine kinases and the corresponding ephrin ligands play a pivotal role in the glioma development and progression. Aberrant protein expression levels of the Eph receptors and ephrins are often associated with higher tumor grade and poor prognosis. Their function in tumorigenesis is complex due to the intricate network of possible co-occurring interactions between neighboring tumor cells and tumor microenvironment. Both Ephs and ephrins localize on the surface of tumor cells, tumor vasculature, glioma stem cells, tumor cells infiltrating brain, and immune cells infiltrating tumors. They can both promote and inhibit tumorigenicity depending on the downstream forward and reverse signalling generated. All the above-mentioned features make the Ephs/ephrins system an intriguing candidate for the development of new therapeutic strategies in glioma treatment. This review will give a general overview on the structure and the function of Ephs and ephrins, with a particular emphasis on the state of the knowledge of their role in malignant gliomas.
Collapse
Affiliation(s)
- Sara Ferluga
- Department of Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Waldemar Debinski
- Department of Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- To whom correspondence should be addressed: Waldemar Debinski, M.D., Ph.D., Director of Brain Tumor Center of Excellence, Thomas K. Hearn Jr. Brain Tumor Research Center, Professor of Neurosurgery, Radiation Oncology, and Cancer Biology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157, Phone: (336) 716-9712, Fax: (336) 713-7639,
| |
Collapse
|
37
|
Ordóñez R, Gallo-Oller G, Martínez-Soto S, Legarra S, Pata-Merci N, Guegan J, Danglot G, Bernheim A, Meléndez B, Rey JA, Castresana JS. Genome-wide microarray expression and genomic alterations by array-CGH analysis in neuroblastoma stem-like cells. PLoS One 2014; 9:e113105. [PMID: 25392930 PMCID: PMC4231109 DOI: 10.1371/journal.pone.0113105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/14/2014] [Indexed: 01/08/2023] Open
Abstract
Neuroblastoma has a very diverse clinical behaviour: from spontaneous regression to a very aggressive malignant progression and resistance to chemotherapy. This heterogeneous clinical behaviour might be due to the existence of Cancer Stem Cells (CSC), a subpopulation within the tumor with stem-like cell properties: a significant proliferation capacity, a unique self-renewal capacity, and therefore, a higher ability to form new tumors. We enriched the CSC-like cell population content of two commercial neuroblastoma cell lines by the use of conditioned cell culture media for neurospheres, and compared genomic gains and losses and genome expression by array-CGH and microarray analysis, respectively (in CSC-like versus standard tumor cells culture). Despite the array-CGH did not show significant differences between standard and CSC-like in both analyzed cell lines, the microarray expression analysis highlighted some of the most relevant biological processes and molecular functions that might be responsible for the CSC-like phenotype. Some signalling pathways detected seem to be involved in self-renewal of normal tissues (Wnt, Notch, Hh and TGF-β) and contribute to CSC phenotype. We focused on the aberrant activation of TGF-β and Hh signalling pathways, confirming the inhibition of repressors of TGF-β pathway, as SMAD6 and SMAD7 by RT-qPCR. The analysis of the Sonic Hedgehog pathway showed overexpression of PTCH1, GLI1 and SMO. We found overexpression of CD133 and CD15 in SIMA neurospheres, confirming that this cell line was particularly enriched in stem-like cells. This work shows a cross-talk among different pathways in neuroblastoma and its importance in CSC-like cells.
Collapse
Affiliation(s)
- Raquel Ordóñez
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, Pamplona, Spain
| | - Gabriel Gallo-Oller
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, Pamplona, Spain
| | - Soledad Martínez-Soto
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, Pamplona, Spain
| | - Sheila Legarra
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, Pamplona, Spain
| | | | | | | | | | - Bárbara Meléndez
- Molecular Pathology Research Unit, Department of Pathology, Virgen de la Salud Hospital, Toledo, Spain
| | - Juan A. Rey
- IdiPaz Research Unit, La Paz University Hospital, Madrid, Spain
| | - Javier S. Castresana
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, Pamplona, Spain
| |
Collapse
|
38
|
Rossmeisl JH. New treatment modalities for brain tumors in dogs and cats. Vet Clin North Am Small Anim Pract 2014; 44:1013-38. [PMID: 25441624 DOI: 10.1016/j.cvsm.2014.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite advancements in standard therapies, intracranial tumors remain a significant source of morbidity and mortality in veterinary and human medicine. Several newer approaches are gaining more widespread acceptance or are currently being prepared for translation from experimental to routine therapeutic use. Clinical trials in dogs with spontaneous brain tumors have contributed to the development and human translation of several novel therapeutic brain tumor approaches.
Collapse
Affiliation(s)
- John H Rossmeisl
- Neurology and Neurosurgery, Department of Small Animal Clinical Sciences, VA-MD Regional College of Veterinary Medicine, Virginia Tech, 215 Duckpond Drive, Mail Code 0442, Blacksburg, VA 24061, USA.
| |
Collapse
|
39
|
Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, Tobias A, Cheng Y, Kim JW, Qiao J, Zhang L, Han Y, Lesniak MS. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res 2014; 20:5290-301. [PMID: 24691018 PMCID: PMC4182350 DOI: 10.1158/1078-0432.ccr-14-0514] [Citation(s) in RCA: 439] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common form of malignant glioma in adults. Although protected by both the blood-brain and blood-tumor barriers, GBMs are actively infiltrated by T cells. Previous work has shown that IDO, CTLA-4, and PD-L1 are dominant molecular participants in the suppression of GBM immunity. This includes IDO-mediated regulatory T-cell (Treg; CD4(+)CD25(+)FoxP3(+)) accumulation, the interaction of T-cell-expressed, CTLA-4, with dendritic cell-expressed, CD80, as well as the interaction of tumor- and/or macrophage-expressed, PD-L1, with T-cell-expressed, PD-1. The individual inhibition of each pathway has been shown to increase survival in the context of experimental GBM. However, the impact of simultaneously targeting all three pathways in brain tumors has been left unanswered. EXPERIMENTAL DESIGN AND RESULTS In this report, we demonstrate that, when dually challenged, IDO-deficient tumors provide a selectively competitive survival advantage against IDO-competent tumors. Next, we provide novel observations regarding tryptophan catabolic enzyme expression, before showing that the therapeutic inhibition of IDO, CTLA-4, and PD-L1 in a mouse model of well-established glioma maximally decreases tumor-infiltrating Tregs, coincident with a significant increase in T-cell-mediated long-term survival. In fact, 100% of mice bearing intracranial tumors were long-term survivors following triple combination therapy. The expression and/or frequency of T cell expressed CD44, CTLA-4, PD-1, and IFN-γ depended on timing after immunotherapeutic administration. CONCLUSIONS Collectively, these data provide strong preclinical evidence that combinatorially targeting immunosuppression in malignant glioma is a strategy that has high potential value for future clinical trials in patients with GBM.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- Brain Neoplasms/drug therapy
- Brain Neoplasms/genetics
- Brain Neoplasms/immunology
- Brain Neoplasms/metabolism
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- CTLA-4 Antigen/antagonists & inhibitors
- Cell Line, Tumor
- Dacarbazine/administration & dosage
- Dacarbazine/analogs & derivatives
- Dacarbazine/pharmacology
- Disease Models, Animal
- Drug Therapy, Combination
- Glioma/drug therapy
- Glioma/genetics
- Glioma/immunology
- Glioma/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Mice
- Mice, Knockout
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Temozolomide
Collapse
Affiliation(s)
- Derek A Wainwright
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Alan L Chang
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Mahua Dey
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Irina V Balyasnikova
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Chung Kwon Kim
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Alex Tobias
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Yu Cheng
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Julius W Kim
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Jian Qiao
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Lingjiao Zhang
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Yu Han
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| |
Collapse
|
40
|
dePillis LG, Eladdadi A, Radunskaya AE. Modeling cancer-immune responses to therapy. J Pharmacokinet Pharmacodyn 2014; 41:461-78. [DOI: 10.1007/s10928-014-9386-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/17/2014] [Indexed: 12/26/2022]
|
41
|
Interleukin-13 receptor alpha 2-targeted glioblastoma immunotherapy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:952128. [PMID: 25247196 PMCID: PMC4163479 DOI: 10.1155/2014/952128] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 08/05/2014] [Indexed: 01/23/2023]
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor, and despite several refinements in its multimodal management, generally has very poor prognosis. Targeted immunotherapy is an emerging field of research that shows great promise in the treatment of GBM. One of the most extensively studied targets is the interleukin-13 receptor alpha chain variant 2 (IL13Rα2). Its selective expression on GBM, discovered almost two decades ago, has been a target for therapy ever since. Immunotherapeutic strategies have been developed targeting IL13Rα2, including monoclonal antibodies as well as cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-expressing T cells. Advanced therapeutic development has led to the completion of several clinical trials with promising outcomes. In this review, we will discuss the recent advances in the IL13Rα2-targeted immunotherapy and evaluate the most promising strategy for targeted GBM immunotherapy.
Collapse
|
42
|
Wilson TA, Karajannis MA, Harter DH. Glioblastoma multiforme: State of the art and future therapeutics. Surg Neurol Int 2014; 5:64. [PMID: 24991467 PMCID: PMC4078454 DOI: 10.4103/2152-7806.132138] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 03/13/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and lethal primary malignancy of the central nervous system (CNS). Despite the proven benefit of surgical resection and aggressive treatment with chemo- and radiotherapy, the prognosis remains very poor. Recent advances of our understanding of the biology and pathophysiology of GBM have allowed the development of a wide array of novel therapeutic approaches, which have been developed. These novel approaches include molecularly targeted therapies, immunotherapies, and gene therapy. METHODS We offer a brief review of the current standard of care, and a survey of novel therapeutic approaches for treatment of GBM. RESULTS Despite promising results in preclinical trials, many of these therapies have demonstrated limited therapeutic efficacy in human clinical trials. Thus, although survival of patients with GBM continues to slowly improve, treatment of GBM remains extremely challenging. CONCLUSION Continued research and development of targeted therapies, based on a detailed understanding of molecular pathogenesis can reasonably be expected to yield improved outcomes for patients with GBM.
Collapse
Affiliation(s)
- Taylor A Wilson
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| | - Matthias A Karajannis
- Department of Pediatrics, Division of Oncology, New York University School of Medicine, NY, USA
| | - David H Harter
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| |
Collapse
|
43
|
Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro Oncol 2014; 16:1304-12. [PMID: 24723564 DOI: 10.1093/neuonc/nou045] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains one of the most lethal primary brain tumors despite surgical and therapeutic advancements. Targeted therapies of neoplastic diseases, including GBM, have received a great deal of interest in recent years. A highly studied target of GBM is interleukin-13 receptor α chain variant 2 (IL13Rα2). Targeted therapies against IL13Rα2 in GBM include fusion chimera proteins of IL-13 and bacterial toxins, nanoparticles, and oncolytic viruses. In addition, immunotherapies have been developed using monoclonal antibodies and cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-modified T cells. Advanced therapeutic development has led to the completion of phase I clinical trials for chimeric antigen receptor-modified T cells and phase III clinical trials for IL-13-conjugated bacterial toxin, with promising outcomes. Selective expression of IL13Rα2 on tumor cells, while absent in the surrounding normal brain tissue, has motivated continued study of IL13Rα2 as an important candidate for targeted glioma therapy. Here, we review the preclinical and clinical studies targeting IL13Rα2 in GBM and discuss new advances and promising applications.
Collapse
Affiliation(s)
- Bart Thaci
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Christine E Brown
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Emanuela Binello
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Katherine Werbaneth
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Prakash Sampath
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| | - Sadhak Sengupta
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, Rhode Island (P.S., S.S.); Department of Neurosurgery, Boston University School of Medicine, Boston, Massachusetts (B.T., K.W., E.B., P.S., S.S.); Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Hospital, Duarte, California (C.E.B.)
| |
Collapse
|
44
|
Agrawal NS, Miller R, Lal R, Mahanti H, Dixon-Mah YN, DeCandio ML, Vandergrift WA, Varma AK, Patel SJ, Banik NL, Lindhorst SM, Giglio P, Das A. Current Studies of Immunotherapy on Glioblastoma. JOURNAL OF NEUROLOGY AND NEUROSURGERY 2014; 1:21000104. [PMID: 25346943 PMCID: PMC4208662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glioblastoma is a form of brain tumor with a very high morbidity and mortality. Despite decades of research, the best treatments currently in clinical practice only extend survival by a number of months. A promising alternative to conventional treatment for glioblastomas is immunotherapy. Although proposed over a century ago, the field of cancer immunotherapy has historically struggled to translate it into effective clinical treatments. Better understanding is needed of the various regulatory and co-stimulatory factors in the glioblastoma patient for more efficient immunotherapy treatments. The tumor microenvironment is anatomically shielded from normal immune-surveillance by the blood-brain barrier, irregular lymphatic drainage system, and it's in a potently immunosuppressive environment. Immunotherapy can potentially manipulate these forces effectively to enhance anti-tumor immune response and clinical benefit. New treatments utilizing the immune system show promise in terms of targeting and efficacy. This review article attempts to discuss current practices in glioblastoma treatment, the theory behind immunotherapy, and current research into various clinical trials.
Collapse
Affiliation(s)
- Neena Stephanie Agrawal
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rickey Miller
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Richa Lal
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Harshini Mahanti
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yaenette N. Dixon-Mah
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michele L. DeCandio
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - W Alex Vandergrift
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Abhay K. Varma
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sunil J. Patel
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L. Banik
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA,Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Scott M. Lindhorst
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA
| | - Pierre Giglio
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA,Corresponding Authors: Arabinda Das, Department of Neurosciences (Neurology and Neuro-oncology) and MUSC Brain & Spine Tumor Program, Medical University of South Carolina, Charleston, SC, 29425, USA, . Pierre Giglio, Department of Neurosciences (Neuro-oncology) and MUSC Brain & Spine Tumor Program, Medical University of South Carolina, Charleston, SC, 29425, USA,
| | - Arabinda Das
- Department of Neurosciences (Divisions of Neurology and Neurosurgery) & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC 29425, USA,Corresponding Authors: Arabinda Das, Department of Neurosciences (Neurology and Neuro-oncology) and MUSC Brain & Spine Tumor Program, Medical University of South Carolina, Charleston, SC, 29425, USA, . Pierre Giglio, Department of Neurosciences (Neuro-oncology) and MUSC Brain & Spine Tumor Program, Medical University of South Carolina, Charleston, SC, 29425, USA,
| |
Collapse
|
45
|
Thaci B, Ahmed AU, Ulasov IV, Wainwright DA, Nigam P, Auffinger B, Tobias AL, Han Y, Zhang L, Moon KS, Lesniak MS. Depletion of myeloid-derived suppressor cells during interleukin-12 immunogene therapy does not confer a survival advantage in experimental malignant glioma. Cancer Gene Ther 2014; 21:38-44. [PMID: 24434573 DOI: 10.1038/cgt.2013.81] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 11/09/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) accumulate in the glioma microenvironment during tumor progression and promote immunosuppression. Interleukin-12 (IL-12) immunogene therapy can alter MDSCs toward an antigen-presenting cell phenotype and these mature cells can have a central role in antigen presentation. It remains unclear, however, how MDSC depletion can affect glioma immunotherapy. In this study, we generated a replication-deficient adenoviral vector, Ad.5/3.cRGD-mIL12p70, that transduces the GL261-based murine glioma cell line, resulting in the induction of biologically active, murine IL12p70 expression. Ex vivo, IL-12 expressed by GL261 cells induced interferon-γ synthesis in CD8(+) T cells (P<0.001), CD4(+) T cells (P=0.009) and natural killer cells (P=0.036). When injected 1 week after tumor implantation, Ad.5/3.cRGD-mIL12p70 successfully prolonged the survival of glioma-bearing mice. Sixty percent of animals treated with IL-12 immunotherapy were long-term survivors over 175 days, whereas all the control group animals expired by 40 days after tumor implantation (P=0.026). Mice receiving Ad.5/3.cRGD-mIL12p70 also accumulated 50% less MDSCs in the brain than the control group (P=0.007). Moreover, in the IL-12 group, MDSCs significantly overexpressed CD80 and major histocompatibility complex class II molecules (P=0.041). Depletion of MDSCs with Gr1(+) antibody had no survival benefit induced by IL-12-mediated immunotherapy. Of note, IL-12 therapy increased the presence of myeloid dendritic cells (mDCs) in the glioma microenvironment (P=0.0069). Ultimately, the data show that in the context of IL-12 immunogene therapy, MDSCs are dispensable and mDCs may provide the majority of antigen presentation in the brain.
Collapse
Affiliation(s)
- B Thaci
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - A U Ahmed
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - I V Ulasov
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - D A Wainwright
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - P Nigam
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - B Auffinger
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - A L Tobias
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Y Han
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - L Zhang
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - K-S Moon
- 1] The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA [2] Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Jeollanam-do, Korea
| | - M S Lesniak
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| |
Collapse
|
46
|
Vasco C, Canazza A, Rizzo A, Mossa A, Corsini E, Silvani A, Fariselli L, Salmaggi A, Ciusani E. Circulating T regulatory cells migration and phenotype in glioblastoma patients: an in vitro study. J Neurooncol 2013; 115:353-63. [DOI: 10.1007/s11060-013-1236-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 08/25/2013] [Indexed: 12/20/2022]
|
47
|
Floyd RA, Castro Faria Neto HC, Zimmerman GA, Hensley K, Towner RA. Nitrone-based therapeutics for neurodegenerative diseases: their use alone or in combination with lanthionines. Free Radic Biol Med 2013; 62:145-156. [PMID: 23419732 PMCID: PMC3715559 DOI: 10.1016/j.freeradbiomed.2013.01.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 01/25/2013] [Accepted: 01/29/2013] [Indexed: 01/12/2023]
Abstract
The possibility of free radical reactions occurring in biological processes led to the development and employment of novel methods and techniques focused on determining their existence and importance in normal and pathological conditions. For this reason the use of nitrones for spin trapping free radicals became widespread in the 1970s and 1980s, when surprisingly the first evidence of their potent biological properties was noted. Since then widespread exploration and demonstration of the potent biological properties of phenyl-tert-butylnitrone (PBN) and its derivatives took place in preclinical models of septic shock and then in experimental stroke. The most extensive commercial effort made to capitalize on the potent properties of the PBN-nitrones was for acute ischemic stroke. This occurred during 1993-2006, when the 2,4-disulfonylphenyl PBN derivative, called NXY-059 in the stroke studies, was shown to be safe in humans and was taken all the way through clinical phase 3 trials and then was deemed to be ineffective. As summarized in this review, because of its excellent human safety profile, 2,4-disulfonylphenyl PBN, now called OKN-007 in the cancer studies, was tested as an anti-cancer agent in several preclinical glioma models and shown to be very effective. Based on these studies this compound is now scheduled to enter into early clinical trials for astrocytoma/glioblastoma multiforme this year. The potential use of OKN-007 in combination with neurotropic compounds such as the lanthionine ketamine esters is discussed for glioblastoma multiforme as well as for various other indications leading to dementia, such as aging, septic shock, and malaria infections. There is much more research and development activity ongoing for various indications with the nitrones, alone or in combination with other active compounds, as briefly noted in this review.
Collapse
Affiliation(s)
- Robert A Floyd
- Experimental Therapeutics, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | | | - Guy A Zimmerman
- Laboratorio de Immunofarmacologia, Instituto Oswaldo Cruz, IOC, Fiocruz, Rio de Janeiro, Brazil; Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Kenneth Hensley
- Department of Pathology and Department of Neurosciences, University of Toledo Medical Center, Toledo, OH
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
48
|
Joyard Y, Joncour VL, Castel H, Diouf CB, Bischoff L, Papamicaël C, Levacher V, Vera P, Bohn P. Synthesis and biological evaluation of a novel 99mTc labeled 2-nitroimidazole derivative as a potential agent for imaging tumor hypoxia. Bioorg Med Chem Lett 2013; 23:3704-8. [DOI: 10.1016/j.bmcl.2013.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 10/26/2022]
|
49
|
Impact of temozolomide on immune response during malignant glioma chemotherapy. Clin Dev Immunol 2012; 2012:831090. [PMID: 23133490 PMCID: PMC3486128 DOI: 10.1155/2012/831090] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/10/2012] [Accepted: 09/20/2012] [Indexed: 12/19/2022]
Abstract
Malignant glioma, or glioblastoma, is the most common and lethal form of brain tumor with a median survival time of 15 months. The established therapeutic regimen includes a tripartite therapy of surgical resection followed by radiation and temozolomide (TMZ) chemotherapy, concurrently with radiation and then as an adjuvant. TMZ, a DNA alkylating agent, is the most successful antiglioma drug and has added several months to the life expectancy of malignant glioma patients. However, TMZ is also responsible for inducing lymphopenia and myelosuppression in malignant glioma patients undergoing chemotherapy. Although TMZ-induced lymphopenia has been attributed to facilitate antitumor vaccination studies by inducing passive immune response, in general lymphopenic conditions have been associated with poor immune surveillance leading to opportunistic infections in glioma patients, as well as disrupting active antiglioma immune response by depleting both T and NK cells. Deletion of O6-methylguanine-DNA-methyltransferase (MGMT) activity, a DNA repair enzyme, by temozolomide has been determined to be the cause of lymphopenia. Drug-resistant mutation of the MGMT protein has been shown to render chemoprotection against TMZ. The immune modulating role of TMZ during glioma chemotherapy and possible mechanisms to establish a strong TMZ-resistant immune response have been discussed.
Collapse
|
50
|
Wainwright DA, Balyasnikova IV, Chang AL, Ahmed AU, Moon KS, Auffinger B, Tobias AL, Han Y, Lesniak MS. IDO expression in brain tumors increases the recruitment of regulatory T cells and negatively impacts survival. Clin Cancer Res 2012; 18:6110-21. [PMID: 22932670 DOI: 10.1158/1078-0432.ccr-12-2130] [Citation(s) in RCA: 326] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is an aggressive adult brain tumor with a poor prognosis. One hallmark of GBM is the accumulation of immunosuppressive and tumor-promoting CD4(+)FoxP3(+)GITR(+) regulatory T cells (Tregs). Here, we investigated the role of indoleamine 2,3 dioxygenase (IDO) in brain tumors and the impact on Treg recruitment. EXPERIMENTAL DESIGN To determine the clinical relevance of IDO expression in brain tumors, we first correlated patient survival to the level of IDO expression from resected glioma specimens. We also used novel orthotopic and transgenic models of glioma to study how IDO affects Tregs. The impact of tumor-derived and peripheral IDO expression on Treg recruitment, GITR expression, and long-term survival was determined. RESULTS Downregulated IDO expression in glioma predicted a significantly better prognosis in patients. Coincidently, both IDO-competent and deficient mice showed a survival advantage bearing IDO-deficient brain tumors, when compared with IDO-competent brain tumors. Moreover, IDO deficiency was associated with a significant decrease in brain-resident Tregs, both in orthotopic and transgenic mouse glioma models. IDO deficiency was also associated with lower GITR expression levels on Tregs. Interestingly, the long-term survival advantage conferred by IDO deficiency was lost in T-cell-deficient mice. CONCLUSIONS These clinical and preclinical data confirm that IDO expression increases the recruitment of immunosuppressive Tregs that lead to tumor outgrowth. In contrast, IDO deficiency decreases Treg recruitment and enhances T-cell-mediated tumor rejection. Thus, the data suggest a critical role for IDO-mediated immunosuppression in glioma and support the continued investigation of IDO-Treg interactions in the context of brain tumors.
Collapse
Affiliation(s)
- Derek A Wainwright
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|