1
|
Jackson LR, Erickson A, Camphausen K, Krauze AV. Understanding the Immune System and Biospecimen-Based Response in Glioblastoma: A Practical Guide to Utilizing Signal Redundancy for Biomarker and Immune Signature Discovery. Curr Oncol 2024; 32:16. [PMID: 39851932 PMCID: PMC11763554 DOI: 10.3390/curroncol32010016] [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: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 01/26/2025] Open
Abstract
Glioblastoma (GBM) is a primary central nervous system malignancy with a median survival of 15-20 months. The presence of both intra- and intertumoral heterogeneity limits understanding of biological mechanisms leading to tumor resistance, including immune escape. An attractive field of research to examine treatment resistance are immune signatures composed of cluster of differentiation (CD) markers and cytokines. CD markers are surface markers expressed on various cells throughout the body, often associated with immune cells. Cytokines are the effector molecules of the immune system. Together, CD markers and cytokines can serve as useful biomarkers to reflect immune status in patients with GBM. However, there are gaps in the understanding of the intricate interactions between GBM and the peripheral immune system and how these interactions change with standard and immune-modulating treatments. The key to understanding the true nature of these interactions is through multi-omic analysis of tumor progression and treatment response. This review aims to identify potential non-invasive blood-based biomarkers that can contribute to an immune signature through multi-omic approaches, leading to a better understanding of immune involvement in GBM.
Collapse
Affiliation(s)
| | | | | | - Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA; (L.R.J.); (A.E.); (K.C.)
| |
Collapse
|
2
|
Desgraves JF, Mendez Valdez MJ, Chandar J, Gurses ME, Henderson L, Castro JR, Seetheram D, Ivan ME, Komotar RJ, Shah AH. Antisense Oligonucleotides for Rapid Translation of Gene Therapy in Glioblastoma. Cancers (Basel) 2024; 16:1944. [PMID: 38792022 PMCID: PMC11119631 DOI: 10.3390/cancers16101944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/27/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
PURPOSE The limited efficacy of current treatments for malignant brain tumors necessitates novel therapeutic strategies. This study aimed to assess the potential of antisense oligonucleotides (ASOs) as adjuvant therapy for high-grade gliomas, focusing on their CNS penetration and clinical translation prospects. METHODS A comprehensive review of the existing literature was conducted to evaluate the implications of ASOs in neuro-oncology. Studies that investigated ASO therapy's efficacy, CNS penetration, and safety profile were analyzed to assess its potential as a therapeutic intervention for high-grade gliomas. RESULTS ASOs present a promising avenue for enhancing targeted gene therapies in malignant gliomas. Their potent CNS penetration, in vivo durability, and efficient transduction offer advantages over conventional treatments. Preliminary in vivo and in vitro studies suggest ASOs as a viable adjuvant therapy for high-grade gliomas, warranting further exploration in clinical trials. CONCLUSIONS ASOs hold significant promise as adjuvant therapy for high-grade gliomas, offering improved CNS penetration and durability compared with existing treatments. While preliminary studies are encouraging, additional research is needed to establish the safety and efficacy of ASO therapy in clinical settings. Further investigation and clinical trials are warranted to validate ASOs as a transformative approach in neuro-oncology.
Collapse
Affiliation(s)
- Jelisah F. Desgraves
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| | - Mynor J. Mendez Valdez
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| | - Jay Chandar
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| | - Muhammet Enes Gurses
- Department of Neurosurgery, Miller School of Medicine, University of Miami, 1095 NW 14th Terrace (D4-6), Miami, FL 33136, USA; (M.E.I.); (R.J.K.)
| | - Lisa Henderson
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| | - Jesus R. Castro
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| | - Deepa Seetheram
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| | - Michael E. Ivan
- Department of Neurosurgery, Miller School of Medicine, University of Miami, 1095 NW 14th Terrace (D4-6), Miami, FL 33136, USA; (M.E.I.); (R.J.K.)
| | - Ricardo J. Komotar
- Department of Neurosurgery, Miller School of Medicine, University of Miami, 1095 NW 14th Terrace (D4-6), Miami, FL 33136, USA; (M.E.I.); (R.J.K.)
| | - Ashish H. Shah
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.F.D.); (M.J.M.V.); (J.C.); (L.H.); (J.R.C.); (D.S.); (A.H.S.)
| |
Collapse
|
3
|
Duan W, Yang L, Liu J, Dai Z, Wang Z, Zhang H, Zhang X, Liang X, Luo P, Zhang J, Liu Z, Zhang N, Mo H, Qu C, Xia Z, Cheng Q. A TGF-β signaling-related lncRNA signature for prediction of glioma prognosis, immune microenvironment, and immunotherapy response. CNS Neurosci Ther 2024; 30:e14489. [PMID: 37850692 PMCID: PMC11017415 DOI: 10.1111/cns.14489] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 07/27/2023] [Accepted: 09/24/2023] [Indexed: 10/19/2023] Open
Abstract
AIMS The dysregulation of TGF-β signaling is a crucial pathophysiological process in tumorigenesis and progression. LncRNAs have diverse biological functions and are significant participants in the regulation of tumor signaling pathways. However, the clinical value of lncRNAs related to TGF-β signaling in glioma is currently unclear. METHODS Data on glioma's RNA-seq transcriptome, somatic mutation, DNA methylation data, and clinicopathological information were derived from the CGGA and TCGA databases. A prognostic lncRNA signature was constructed by Cox and LASSO regression analyses. TIMER2.0 database was utilized to deduce immune infiltration characteristics. "ELMER v.2" was used to reconstruct TF-methylation-gene regulatory network. Immunotherapy and chemotherapy response predictions were implemented by the TIDE algorithm and GDSC database, respectively. In vitro and in vivo experiments were conducted to verify the results and clarify the regulatory mechanism of lncRNA. RESULTS In glioma, a TGF-β signaling-related 15-lncRNA signature was constructed, including AC010173.1, HOXA-AS2, AC074286.1, AL592424.1, DRAIC, HOXC13-AS, AC007938.1, AC010729.1, AC013472.3, AC093895.1, AC131097.4, AL606970.4, HOXC-AS1, AGAP2-AS1, and AC002456.1. This signature proved to be a reliable prognostic tool, with high risk indicating an unfavorable prognosis and being linked to malignant clinicopathological and genomic mutation traits. Risk levels were associated with different immune infiltration landscapes, where high risk was indicative of high levels of macrophage infiltration. In addition, high risk also suggested better immunotherapy and chemotherapy response. cg05987823 was an important methylation site in glioma progression, and AP-1 transcription factor family participated in the regulation of signature lncRNA expression. AGAP2-AS1 knockdown in in vitro and in vivo experiments inhibited the proliferation, migration, and invasion of glioma cells, as well as the growth of glioma, by downregulating the expression levels of NF-κB and ERK 1/2 in the TGF-β signaling pathway. CONCLUSIONS A prognostic lncRNA signature of TGF-β signaling was established in glioma, which can be used for prognostic judgment, immune infiltration status inference, and immunotherapy response prediction. AGAP2-AS1 plays an important role in glioma progression.
Collapse
Affiliation(s)
- Wei‐Wei Duan
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Li‐Ting Yang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Jian Liu
- Experiment Center of Medical InnovationThe First Hospital of Hunan University of Chinese MedicineChangshaHunanChina
| | - Zi‐Yu Dai
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ze‐Yu Wang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- MRC Centre for Regenerative Medicine, Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Hao Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xun Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xi‐Song Liang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Peng Luo
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jian Zhang
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zao‐Qu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Nan Zhang
- One‐third Lab, College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinHei LongjiangChina
| | - Hao‐Yang Mo
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Chun‐Run Qu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zhi‐Wei Xia
- Department of NeurologyHunan Aerospace HospitalChangshaHunanChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
4
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
5
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Xu H, Zhang L, Gao J, Wang J, Wang Y, Xiao D, Chai S. Molecular and clinical features of a potential immunotherapy target ELK3 in glioma. Medicine (Baltimore) 2022; 101:e29544. [PMID: 35905257 PMCID: PMC9333475 DOI: 10.1097/md.0000000000029544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glioma represents the most prevalent malignant primary brain cancer, and its treatment remains a tremendous challenge. Novel and efficient molecular targets are therefore required for improving diagnosis, survival prediction, and treatment outcomes. Additionally, some studies have shown that immunity is highly associated with glioma progression. Our study aimed to investigate the clinicopathological features, prognostic significance, and immunotherapeutic targetability of ELK3, a member of the erythroblast transformation-specific transcription factor family, in glioma using bioinformatics analyses. ELK3 transcript levels in glioma tissues were evaluated using the Gene Expression Omnibus and The Cancer Genome Atlas databases. Clinical and transcriptomic data of The Cancer Genome Atlas glioma patients were analyzed to identify the molecular and clinical characterizations of ELK3. The prognostic significance of ELK3 was assessed using Cox regression and Kaplan-Meier analysis. The biological pathways related to ELK3 expression were identified by gene set enrichment analysis. The relationships between ELK3 and inflammatory responses, immune cell infiltration, and immune checkpoints were explored using canonical correlation analysis and gene set variation analysis. ELK3 was upregulated in gliomas, and its high expression was correlated with advanced clinicopathologic features and unfavorable prognosis. Gene set enrichment analysis revealed that several immune-related pathways were tightly linked to high ELK3 expression. gene set variation analysis and correlograms demonstrated that ELK3 was robustly associated with inflammatory and immune responses. Correlation analyses indicated that ELK3 was positively associated with infiltrating immune cells and synergistic with several immune checkpoints. ELK3 may serve as a novel marker of poor prognosis and a potential immunotherapeutic target in glioma.
Collapse
Affiliation(s)
- Hao Xu
- Department of Neurosurgery, Wuhan Changjiang Shipping General Hospital, Wuhan, China
| | - Li Zhang
- School of Information Management, Wuhan University, Wuhan, China
| | - Jin Gao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiajing Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongdong Xiao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songshan Chai
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Songshan Chai, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan, Hubei 430071, China (e-mail: )
| |
Collapse
|
7
|
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
|
8
|
Wu X, Yang L, Wang J, Hao Y, Wang C, Lu Z. The Involvement of Long Non-Coding RNAs in Glioma: From Early Detection to Immunotherapy. Front Immunol 2022; 13:897754. [PMID: 35619711 PMCID: PMC9127066 DOI: 10.3389/fimmu.2022.897754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Glioma is a brain tumor that arises in the central nervous system and is categorized according to histology and molecular genetic characteristics. Long non-coding RNAs (lncRNAs) are RNAs longer than 200 nucleotides in length. They have been reported to influence significant events such as carcinogenesis, progression, and increased treatment resistance on glioma cells. Long non-coding RNAs promote cell proliferation, migration, epithelial-to-mesenchymal transition and invasion in glioma cells. Various significant advancements in transcriptomic profiling studies have enabled the identification of immune-related long non-coding RNAs as immune cell-specific gene expression regulators that mediates both stimulatory and suppressive immune responses, implying lncRNAs as potential candidates for improving immunotherapy efficacy against tumors and due to the lack of different diagnostic and treatments for glioma, lncRNAs are potential candidates to be used as future diagnostic, prognostic biomarker and treatment tools for glioma. This review’s primary purpose is to concentrate on the role of long non-coding RNAs in early glioma identification, treatment, and immunotherapy.
Collapse
Affiliation(s)
- Xiaoben Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lei Yang
- Department of Medical Engineering, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yingying Hao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Changyin Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
9
|
Bone metastasis from glioblastoma: a systematic review. J Neurooncol 2022; 158:379-392. [PMID: 35578056 DOI: 10.1007/s11060-022-04025-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is a devastating disease with poor overall survival. Despite the common occurrence of GBM among primary brain tumors, metastatic disease is rare. Our goal was to perform a systematic literature review on GBM with osseous metastases and understand the rate of metastasis to the vertebral column as compared to the remainder of the skeleton, and how this histology would fit into our current paradigm of treatment for bone metastases. METHODS A Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)-compliant literature search was performed using the PubMed database from 1952 to 2021. Search terms included "GBM", "glioblastoma", "high-grade glioma", "bone metastasis", and "bone metastases". RESULTS Of 659 studies initially identified, 67 articles were included in the current review. From these 67 articles, a total of 92 distinct patient case presentations of metastatic glioblastoma to bone were identified. Of these cases, 58 (63%) involved the vertebral column while the remainder involved lesions within the skull, sternum, rib cage, and appendicular skeleton. CONCLUSION Metastatic dissemination of GBM to bone occurs. While the true incidence is unknown, workup for metastatic disease, especially involving the spinal column, is warranted in symptomatic patients. Lastly, management of patients with GBM vertebral column metastases can follow the International Spine Oncology Consortium two-step multidisciplinary algorithm for the management of spinal metastases.
Collapse
|
10
|
Zhong C, Tao B, Li X, Xiang W, Peng L, Peng T, Chen L, Xia X, You J, Yang X. HOXA-AS2 contributes to regulatory T cell proliferation and immune tolerance in glioma through the miR-302a/KDM2A/JAG1 axis. Cell Death Dis 2022; 13:160. [PMID: 35181676 PMCID: PMC8857186 DOI: 10.1038/s41419-021-04471-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/23/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
Long non-coding RNAs (lncRNAs) have been manifested to manipulate diverse biological processes, including tumor-induced immune tolerance. Thus, we aimed in this study to identify the expression pattern of lncRNA homeobox A cluster antisense RNA 2 (HOXA-AS2) in glioma and decipher its role in immune tolerance and glioma progression. We found aberrant upregulation of lncRNA HOXA-AS2, lysine demethylase 2A (KDM2A), and jagged 1 (JAG1) and a downregulation of microRNA-302a (miR-302a) in glioma specimens. Next, RNA immunoprecipitation, chromatin immunoprecipitation, and dual-luciferase reporter gene assay demonstrated that lncRNA HOXA-AS2 upregulated KDM2A expression by preventing miR-302a from binding to its 3′untranslated region. The functional experiments suggested that lncRNA HOXA-AS2 could promote regulatory T (Treg) cell proliferation and immune tolerance, which might be achieved through inhibition of miR-302a and activation of KDM2A/JAG1 axis. These findings were validated in a tumor xenograft mouse model. To conclude, lncRNA HOXA-AS2 facilitates KDM2A/JAG1 expression to promote Treg cell proliferation and immune tolerance in glioma by binding to miR-302a. These findings may aid in the development of novel antitumor targets.
Collapse
Affiliation(s)
- Chuanhong Zhong
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Bei Tao
- Rheumatism Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China
| | - Xianglong Li
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Wei Xiang
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Lilei Peng
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Tangming Peng
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Ligang Chen
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Xiangguo Xia
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China.,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China.,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China.,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China
| | - Jian You
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China. .,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China. .,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China. .,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China.
| | - Xiaobo Yang
- Neurosurgery Department, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, P. R. China. .,Sichuan Clinical Research Center for Neurosurgery, 646000, Luzhou, P. R. China. .,Academician (Expert) Workstation of Sichuan Province, 646000, Luzhou, P. R. China. .,Laboratory of Neurological Disease and Brain Function, 646000, Luzhou, P. R. China.
| |
Collapse
|
11
|
Shamshiripour P, Nikoobakht M, Mansourinejad Z, Ahmadvand D, Akbarpour M. A comprehensive update to DC therapy for glioma; a systematic review and meta-analysis. Expert Rev Vaccines 2022; 21:513-531. [DOI: 10.1080/14760584.2022.2027759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Parisa Shamshiripour
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of medical imaging technology and molecular imaging, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Nikoobakht
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - zahra Mansourinejad
- Department of systems biology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Davoud Ahmadvand
- Department of medical imaging technology and molecular imaging, Iran University of Medical Sciences, Tehran, Iran
| | - Mahzad Akbarpour
- Advanced Cellular Therapeutics Facility, David and Etta Jonas Center for Cellular Therapy, Hematopoietic Cellular Therapy Program, The University of Chicago Medical Center, Chicago 60637 IL, USA
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno-TACT), Universal Science and Education Research Network (USERN), Chicago, USA
| |
Collapse
|
12
|
Zhao N, Zhang J, Zhao Q, Chen C, Wang H. Mechanisms of Long Non-Coding RNAs in Biological Characteristics and Aerobic Glycolysis of Glioma. Int J Mol Sci 2021; 22:ijms222011197. [PMID: 34681857 PMCID: PMC8541290 DOI: 10.3390/ijms222011197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023] Open
Abstract
Glioma is the most common and aggressive tumor of the central nervous system. The uncontrolled proliferation, cellular heterogeneity, and diffusive capacity of glioma cells contribute to a very poor prognosis of patients with high grade glioma. Compared to normal cells, cancer cells exhibit a higher rate of glucose uptake, which is accompanied with the metabolic switch from oxidative phosphorylation to aerobic glycolysis. The metabolic reprogramming of cancer cell supports excessive cell proliferation, which are frequently mediated by the activation of oncogenes or the perturbations of tumor suppressor genes. Recently, a growing body of evidence has started to reveal that long noncoding RNAs (lncRNAs) are implicated in a wide spectrum of biological processes in glioma, including malignant phenotypes and aerobic glycolysis. However, the mechanisms of diverse lncRNAs in the initiation and progression of gliomas remain to be fully unveiled. In this review, we summarized the diverse roles of lncRNAs in shaping the biological features and aerobic glycolysis of glioma. The thorough understanding of lncRNAs in glioma biology provides opportunities for developing diagnostic biomarkers and novel therapeutic strategies targeting gliomas.
Collapse
|
13
|
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: 46] [Impact Index Per Article: 11.5] [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
|
14
|
Liu S, Zhao Q, Shi W, Zheng Z, Liu Z, Meng L, Dong L, Jiang X. Advances in radiotherapy and comprehensive treatment of high-grade glioma: immunotherapy and tumor-treating fields. J Cancer 2021; 12:1094-1104. [PMID: 33442407 PMCID: PMC7797642 DOI: 10.7150/jca.51107] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/21/2020] [Indexed: 12/18/2022] Open
Abstract
High-grade gliomas (HGGs) are the most common primary malignant brain tumors. They have a high degree of malignancy and show invasive growth. The personal treatment plan for HGG is based on the patient's age, performance status, and degree of tumor invasion. The basic treatment plan for HGG involves tumor resection, radiotherapy (RT) with concomitant temozolomide (TMZ), and adjuvant TMZ chemotherapy. The basic radiation technology includes conventional RT, three-dimensional conformal RT, intensity-modulated RT, and stereotactic RT. As our understanding of tumor pathogenesis has deepened, so-called comprehensive treatment schemes have attracted attention. These combine RT with chemotherapy, molecular targeted therapy, immunotherapy, or tumor-treating fields. These emerging treatments are expected to improve the prospects of patients with HGG. In the present article, we review the recent advances in RT and comprehensive treatment for patients with newly diagnosed and recurrent HGG.
Collapse
Affiliation(s)
- Shiyu Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Weiyan Shi
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zijing Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Lihua Dong
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| |
Collapse
|
15
|
Ayasoufi K, Pfaller CK, Evgin L, Khadka RH, Tritz ZP, Goddery EN, Fain CE, Yokanovich LT, Himes BT, Jin F, Zheng J, Schuelke MR, Hansen MJ, Tung W, Parney IF, Pease LR, Vile RG, Johnson AJ. Brain cancer induces systemic immunosuppression through release of non-steroid soluble mediators. Brain 2020; 143:3629-3652. [PMID: 33253355 PMCID: PMC7954397 DOI: 10.1093/brain/awaa343] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 01/09/2023] Open
Abstract
Immunosuppression of unknown aetiology is a hallmark feature of glioblastoma and is characterized by decreased CD4 T-cell counts and downregulation of major histocompatibility complex class II expression on peripheral blood monocytes in patients. This immunosuppression is a critical barrier to the successful development of immunotherapies for glioblastoma. We recapitulated the immunosuppression observed in glioblastoma patients in the C57BL/6 mouse and investigated the aetiology of low CD4 T-cell counts. We determined that thymic involution was a hallmark feature of immunosuppression in three distinct models of brain cancer, including mice harbouring GL261 glioma, B16 melanoma, and in a spontaneous model of diffuse intrinsic pontine glioma. In addition to thymic involution, we determined that tumour growth in the brain induced significant splenic involution, reductions in peripheral T cells, reduced MHC II expression on blood leucocytes, and a modest increase in bone marrow resident CD4 T cells. Using parabiosis we report that thymic involution, declines in peripheral T-cell counts, and reduced major histocompatibility complex class II expression levels were mediated through circulating blood-derived factors. Conversely, T-cell sequestration in the bone marrow was not governed through circulating factors. Serum isolated from glioma-bearing mice potently inhibited proliferation and functions of T cells both in vitro and in vivo. Interestingly, the factor responsible for immunosuppression in serum is non-steroidal and of high molecular weight. Through further analysis of neurological disease models, we determined that the immunosuppression was not unique to cancer itself, but rather occurs in response to brain injury. Non-cancerous acute neurological insults also induced significant thymic involution and rendered serum immunosuppressive. Both thymic involution and serum-derived immunosuppression were reversible upon clearance of brain insults. These findings demonstrate that brain cancers cause multifaceted immunosuppression and pinpoint circulating factors as a target of intervention to restore immunity.
Collapse
Affiliation(s)
| | - Christian K Pfaller
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Paul-Ehrlich-Institute, Division of Veterinary Medicine, Langen, Germany
| | - Laura Evgin
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
| | - Roman H Khadka
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Zachariah P Tritz
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Emma N Goddery
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Cori E Fain
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Lila T Yokanovich
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Benjamin T Himes
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN, USA
| | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Jiaying Zheng
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Matthew R Schuelke
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Immunology, Mayo Clinic Medical Scientist Training Program, Rochester, Minnesota, USA
| | | | - Wesley Tung
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Ian F Parney
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN, USA
| | - Larry R Pease
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Richard G Vile
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Department of Neurology, Rochester, MN, USA
| |
Collapse
|
16
|
Hsu JBK, Lee GA, Chang TH, Huang SW, Le NQK, Chen YC, Kuo DP, Li YT, Chen CY. Radiomic Immunophenotyping of GSEA-Assessed Immunophenotypes of Glioblastoma and Its Implications for Prognosis: A Feasibility Study. Cancers (Basel) 2020; 12:cancers12103039. [PMID: 33086550 PMCID: PMC7603270 DOI: 10.3390/cancers12103039] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/05/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Characterization of immunophenotypes in GBM is important for therapeutic stratification and helps predict treatment response and prognosis. However, identifying immunophenotypes of patients with GBM requires multiple laboratory experiments and is time consuming. We developed a non-invasive method to evaluate enrichment levels of CTL, aDC, Treg, and MDSC immune cells to classify immunophenotypes of GBM tumor microenvironment with radiomic features of MR imaging. Five immunophenotypes (G1–G5) of GBM can be classified with specific gene set enrichment analysis. G2 had the worst prognosis and comprised highly enriched MDSCs and lowly enriched CTLs. G3 had the best prognosis and comprised lowly enriched MDSCs and Tregs and highly enriched CTLs. Moreover, the developed radiomics models can successfully identified these two groups by immune cell subsets enriched levels prediction. Therefore, it is possible to characterize immunophenotypes of GBM and predict patient prognosis with radiomics methods. Abstract Characterization of immunophenotypes in glioblastoma (GBM) is important for therapeutic stratification and helps predict treatment response and prognosis. Radiomics can be used to predict molecular subtypes and gene expression levels. However, whether radiomics aids immunophenotyping prediction is still unknown. In this study, to classify immunophenotypes in patients with GBM, we developed machine learning-based magnetic resonance (MR) radiomic models to evaluate the enrichment levels of four immune subsets: Cytotoxic T lymphocytes (CTLs), activated dendritic cells, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs). Independent testing data and the leave-one-out cross-validation method were used to evaluate model effectiveness and model performance, respectively. We identified five immunophenotypes (G1 to G5) based on the enrichment level for the four immune subsets. G2 had the worst prognosis and comprised highly enriched MDSCs and lowly enriched CTLs. G3 had the best prognosis and comprised lowly enriched MDSCs and Tregs and highly enriched CTLs. The average accuracy of T1-weighted contrasted MR radiomics models of the enrichment level for the four immune subsets reached 79% and predicted G2, G3, and the “immune-cold” phenotype (G1) according to our radiomics models. Our radiomic immunophenotyping models feasibly characterize the immunophenotypes of GBM and can predict patient prognosis.
Collapse
Affiliation(s)
- Justin Bo-Kai Hsu
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (J.B.-K.H.); (G.A.L.); (S.-W.H.)
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (Y.-C.C.); (D.-P.K.); (Y.-T.L.)
| | - Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (J.B.-K.H.); (G.A.L.); (S.-W.H.)
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (Y.-C.C.); (D.-P.K.); (Y.-T.L.)
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 110, Taiwan;
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Shiu-Wen Huang
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (J.B.-K.H.); (G.A.L.); (S.-W.H.)
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yung-Chieh Chen
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (Y.-C.C.); (D.-P.K.); (Y.-T.L.)
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Duen-Pang Kuo
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (Y.-C.C.); (D.-P.K.); (Y.-T.L.)
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yi-Tien Li
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (Y.-C.C.); (D.-P.K.); (Y.-T.L.)
- Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Cheng-Yu Chen
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (Y.-C.C.); (D.-P.K.); (Y.-T.L.)
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: ; Tel.: +886-2-2737-2181
| |
Collapse
|
17
|
Pearson JRD, Cuzzubbo S, McArthur S, Durrant LG, Adhikaree J, Tinsley CJ, Pockley AG, McArdle SEB. Immune Escape in Glioblastoma Multiforme and the Adaptation of Immunotherapies for Treatment. Front Immunol 2020; 11:582106. [PMID: 33178210 PMCID: PMC7594513 DOI: 10.3389/fimmu.2020.582106] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequently occurring primary brain tumor and has a very poor prognosis, with only around 5% of patients surviving for a period of 5 years or more after diagnosis. Despite aggressive multimodal therapy, consisting mostly of a combination of surgery, radiotherapy, and temozolomide chemotherapy, tumors nearly always recur close to the site of resection. For the past 15 years, very little progress has been made with regards to improving patient survival. Although immunotherapy represents an attractive therapy modality due to the promising pre-clinical results observed, many of these potential immunotherapeutic approaches fail during clinical trials, and to date no immunotherapeutic treatments for GBM have been approved. As for many other difficult to treat cancers, GBM combines a lack of immunogenicity with few mutations and a highly immunosuppressive tumor microenvironment (TME). Unfortunately, both tumor and immune cells have been shown to contribute towards this immunosuppressive phenotype. In addition, current therapeutics also exacerbate this immunosuppression which might explain the failure of immunotherapy-based clinical trials in the GBM setting. Understanding how these mechanisms interact with one another, as well as how one can increase the anti-tumor immune response by addressing local immunosuppression will lead to better clinical results for immune-based therapeutics. Improving therapeutic delivery across the blood brain barrier also presents a challenge for immunotherapy and future therapies will need to consider this. This review highlights the immunosuppressive mechanisms employed by GBM cancers and examines potential immunotherapeutic treatments that can overcome these significant immunosuppressive hurdles.
Collapse
Affiliation(s)
- Joshua R. D. Pearson
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stefania Cuzzubbo
- Université de Paris, PARCC, INSERM U970, Paris, France
- Laboratoire de Recherches Biochirurgicales (Fondation Carpentier), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France
| | - Simon McArthur
- Institute of Dentistry, Barts & the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, London, United Kingdom
| | - Lindy G. Durrant
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Jason Adhikaree
- Academic Oncology, Nottingham University NHS Trusts, City Hospital Campus, Nottingham, United Kingdom
| | - Chris J. Tinsley
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - A. Graham Pockley
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephanie E. B. McArdle
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
18
|
Modulation of glioma-inflammation crosstalk profiles in human glioblastoma cells by indirubin-3'-(2,3 dihydroxypropyl)-oximether (E804) and 7-bromoindirubin-3'-oxime (7BIO). Chem Biol Interact 2019; 312:108816. [PMID: 31505164 DOI: 10.1016/j.cbi.2019.108816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/05/2019] [Indexed: 11/23/2022]
Abstract
Indirubins E804 (indirubin-3'-(2,3 dihydroxypropyl)-oximether) and 7BIO (7-Bromoindirubin-3'-oxime) are synthetic derivatives of natural indirubin, the active compound in Danggui Longhui Wan, a traditional Chinese remedy for cancer and inflammation. Herein, we explore E804 and 7BIO for their potential to modulate key pro-inflammatory genes and cytokines in LN-18 and T98G glioblastoma cells. High grade gliomas typically secrete large amounts of inflammatory cytokines and growth factors that promote tumor growth in an autocrine fashion. Inflammation is emerging as a key concern in the success of new treatment modalities for glioblastomas. Studies indicate that select indirubin derivatives bind and activate signaling of the AHR pathway, as well as inhibit cyclin-dependent kinases and STAT3 signaling. AHR signaling is involved in hematopoiesis, immune function, cell cycling, and inflammation, and thus may be a possible target for glioma treatment. To determine the significance of the AHR pathway in LN-18 and T98G glioma inflammatory profiles, and on the effects of E804 and 7BIO on these profiles, we used 6,2',4'-trimethoxyflavone (TMF), a putative selective AHR antagonist. It was confirmed that E804 and 7BIO activates the AHR leading to cyp1b1 expression, and that TMF antagonizes expression. We then employed a commercial cancer inflammation and immunity crosstalk qRT-PCR array to screen for anti-inflammatory related properties. TMF alone inhibited expression of ifng, ptsg2, il12b, tnfa, il10, il13, the balance between pd1 and pdl1, and even expression of mhc1a/b. E804 was very potent in suppressing many pro-inflammatory genes, including il1a, il1b, il12a, ptgs2, tlr4, and others. E804 also affected expression of il6, vegfa, and stat3. Conversely, 7BIO induced cox2, but suppressed a different selection of pro-inflammatory genes including nos2, tnfa, and igf1. Secretion of IL-6 protein, an iconic inflammatory cytokine, was decreased by E804. VEGF (vascular endothelial growth factor) protein secretion was upregulated by 7BIO, yet downregulated by E804 and E804 plus TMF. Thus, E804 is both an AHR ligand and regulator of important pro-inflammatory cytokines such as IL-6 and oncogene STAT3, among others. Our results point to the use of E804 and TMF in combination as a promising new treatment for glioblastoma.
Collapse
|
19
|
Schneider JR, Kwan K, Boockvar JA. Missing Naïve T Cells in the Setting of Glioblastoma Are Found to Be Sequestered in the Bone Marrow. Neurosurgery 2019; 85:E194-E195. [PMID: 31304546 DOI: 10.1093/neuros/nyz167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Julia R Schneider
- Department of Neurosurgery Lenox Hill Hospital Donald and Barbara Zucker School of Medicine at Hofstra/Northwell New York, New York.,Brain Tumor Biotech Center The Feinstein Institute for Medical Research Northwell Health Manhasset, New York
| | - Kevin Kwan
- Department of Neurosurgery Lenox Hill Hospital Donald and Barbara Zucker School of Medicine at Hofstra/Northwell New York, New York.,Brain Tumor Biotech Center The Feinstein Institute for Medical Research Northwell Health Manhasset, New York
| | - John A Boockvar
- Department of Neurosurgery Lenox Hill Hospital Donald and Barbara Zucker School of Medicine at Hofstra/Northwell New York, New York.,Brain Tumor Biotech Center The Feinstein Institute for Medical Research Northwell Health Manhasset, New York
| |
Collapse
|
20
|
Graner MW. Roles of Extracellular Vesicles in High-Grade Gliomas: Tiny Particles with Outsized Influence. Annu Rev Genomics Hum Genet 2019; 20:331-357. [PMID: 30978305 DOI: 10.1146/annurev-genom-083118-015324] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High-grade gliomas, particularly glioblastomas (grade IV), are devastating diseases with dismal prognoses; afflicted patients seldom live longer than 15 months, and their quality of life suffers immensely. Our current standard-of-care therapy has remained essentially unchanged for almost 15 years, with little new therapeutic progress. We desperately need a better biologic understanding of these complicated tumors in a complicated organ. One area of rejuvenated study relates to extracellular vesicles (EVs)-membrane-enclosed nano- or microsized particles that originate from the endosomal system or are shed from the plasma membrane. EVs contribute to tumor heterogeneity (including the maintenance of glioma stem cells or their differentiation), the impacts of hypoxia (angiogenesis and coagulopathies), interactions amid the tumor microenvironment (concerning the survival of astrocytes, neurons, endothelial cells, blood vessels, the blood-brain barrier, and the ensuing inflammation), and influences on the immune system (both stimulatory and suppressive). This article reviews glioma EVs and the ways that EVs manifest themselves as autocrine, paracrine, and endocrine factors in proximal and distal intra- and intercellular communications. The reader should note that there is much controversy, and indeed confusion, in the field over the exact roles for EVs in many biological processes, and we will engage some of these difficulties herein.
Collapse
Affiliation(s)
- Michael W Graner
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045, USA;
| |
Collapse
|
21
|
Rajani KR, Carlstrom LP, Parney IF, Johnson AJ, Warrington AE, Burns TC. Harnessing Radiation Biology to Augment Immunotherapy for Glioblastoma. Front Oncol 2019; 8:656. [PMID: 30854331 PMCID: PMC6395389 DOI: 10.3389/fonc.2018.00656] [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: 10/08/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most common adult primary brain tumor and carries a dismal prognosis. Radiation is a standard first-line therapy, typically deployed following maximal safe surgical debulking, when possible, in combination with cytotoxic chemotherapy. For other systemic cancers, standard of care is being transformed by immunotherapies, including checkpoint-blocking antibodies targeting CTLA-4 and PD-1/PD-L1, with potential for long-term remission. Ongoing studies are evaluating the role of immunotherapies for GBM. Despite dramatic responses in some cases, randomized trials to date have not met primary outcomes. Challenges have been attributed in part to the immunologically "cold" nature of glioblastoma relative to other malignancies successfully treated with immunotherapy. Radiation may serve as a mechanism to improve tumor immunogenicity. In this review, we critically evaluate current evidence regarding radiation as a synergistic facilitator of immunotherapies through modulation of both the innate and adaptive immune milieu. Although current preclinical data encourage efforts to harness synergistic biology between radiation and immunotherapy, several practical and scientific challenges remain. Moreover, insights from radiation biology may unveil additional novel opportunities to help mobilize immunity against GBM.
Collapse
Affiliation(s)
- Karishma R. Rajani
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Lucas P. Carlstrom
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Ian F. Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Terry C. Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
22
|
Abstract
High-grade glioma is the most common primary brain tumor, with glioblastoma multiforme (GBM) accounting for 52% of all brain tumors. The current standard of care (SOC) of GBM involves surgery followed by adjuvant fractionated radiotherapy and chemotherapy. However, little progress has been made in extending overall survival, progression-free survival, and quality of life. Attempts to characterize and customize treatment of GBM have led to mitigating the deleterious effects of radiotherapy using hypofractionated radiotherapy, as well as various immunotherapies as a promising strategy for the incurable disease. A combination of radiotherapy and immunotherapy may prove to be even more effective than either alone, and preclinical evidence suggests that hypofractionated radiotherapy can actually prime the immune system to make immunotherapy more effective. This review addresses the complications of the current radiotherapy regimen, various methods of immunotherapy, and preclinical and clinical data from combined radioimmunotherapy trials.
Collapse
|
23
|
Ma Q, Long W, Xing C, Chu J, Luo M, Wang HY, Liu Q, Wang RF. Cancer Stem Cells and Immunosuppressive Microenvironment in Glioma. Front Immunol 2018; 9:2924. [PMID: 30619286 PMCID: PMC6308128 DOI: 10.3389/fimmu.2018.02924] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
Glioma is one of the most common malignant tumors of the central nervous system and is characterized by extensive infiltrative growth, neovascularization, and resistance to various combined therapies. In addition to heterogenous populations of tumor cells, the glioma stem cells (GSCs) and other nontumor cells present in the glioma microenvironment serve as critical regulators of tumor progression and recurrence. In this review, we discuss the role of several resident or peripheral factors with distinct tumor-promoting features and their dynamic interactions in the development of glioma. Localized antitumor factors could be silenced or even converted to suppressive phenotypes, due to stemness-related cell reprogramming and immunosuppressive mediators in glioma-derived microenvironment. Furthermore, we summarize the latest knowledge on GSCs and key microenvironment components, and discuss the emerging immunotherapeutic strategies to cure this disease.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Wenyong Long
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Changsheng Xing
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Junjun Chu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Mei Luo
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Qing Liu
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, United States.,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, United States
| |
Collapse
|
24
|
Stefani FR, Eberstål S, Vergani S, Kristiansen TA, Bengzon J. Low-dose irradiated mesenchymal stromal cells break tumor defensive properties in vivo. Int J Cancer 2018; 143:2200-2212. [PMID: 29752716 PMCID: PMC6220775 DOI: 10.1002/ijc.31599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 12/16/2022]
Abstract
Solid tumors, including gliomas, still represent a challenge to clinicians and first line treatments often fail, calling for new paradigms in cancer therapy. Novel strategies to overcome tumor resistance are mainly represented by multi-targeted approaches, and cell vector-based therapy is one of the most promising treatment modalities under development. Here, we show that mouse bone marrow-derived mesenchymal stromal cells (MSCs), when primed with low-dose irradiation (irMSCs), undergo changes in their immunogenic and angiogenic capacity and acquire anti-tumoral properties in a mouse model of glioblastoma (GBM). Following grafting in GL261 glioblastoma, irMSCs migrate extensively and selectively within the tumor and infiltrate predominantly the peri-vascular niche, leading to rejection of established tumors and cure in 29% of animals. The therapeutic radiation dose window is narrow, with effects seen between 2 and 15 Gy, peaking at 5 Gy. A single low-dose radiation decreases MSCs inherent immune suppressive properties in vitro as well as shapes their immune regulatory ability in vivo. Intra-tumorally grafted irMSCs stimulate the immune system and decrease immune suppression. Additionally, irMSCs enhance peri-tumoral reactive astrocytosis and display anti-angiogenic properties. Hence, the present study provides strong evidence for a therapeutic potential of low-dose irMSCs in cancer as well as giving new insight into MSC biology and applications.
Collapse
Affiliation(s)
- Francesca Romana Stefani
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Clinical Sciences, Division of Neurosurgery, Lund University, Lund, Sweden
| | - Sofia Eberstål
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Clinical Sciences, Division of Neurosurgery, Lund University, Lund, Sweden
| | - Stefano Vergani
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Trine A Kristiansen
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Johan Bengzon
- Stem Cell Center, Lund University, Lund, Sweden.,Department of Clinical Sciences, Division of Neurosurgery, Lund University, Lund, Sweden.,Department of Neurosurgery, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
25
|
Immunohistochemical Characterization of Immune Cell Infiltration in Feline Glioma. J Comp Pathol 2018; 160:15-22. [DOI: 10.1016/j.jcpa.2018.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/16/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022]
|
26
|
Quezada C, Torres Á, Niechi I, Uribe D, Contreras-Duarte S, Toledo F, San Martín R, Gutiérrez J, Sobrevia L. Role of extracellular vesicles in glioma progression. Mol Aspects Med 2018; 60:38-51. [PMID: 29222067 DOI: 10.1016/j.mam.2017.12.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022]
Abstract
The role of extracellular vesicles in cancer biology has emerged as a focus of the study of great importance and has been shown to directly influence tumour development in several cancers including brain tumours, such as gliomas. Gliomas are the most aggressive brain tumours, and in the last time, a considerable effort has been made to understand their biology. Studies focus in the signalling pathways involved in the processes of angiogenesis, viability, drug resistance and immune response evasion, as well as gliomas ability to infiltrate healthy tissue, a phenomenon regulated by the migratory and invasive capacity of the cells within a tumour. In this review, we summarize the different types and classifications of extracellular vesicles, their intravesicular content, and their role in the regulation of tumour progression processes in glioma.
Collapse
Affiliation(s)
- Claudia Quezada
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile.
| | - Ángelo Torres
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Niechi
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Uribe
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Susana Contreras-Duarte
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile
| | - Rody San Martín
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Jaime Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastián, Santiago 7510157, Chile.
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia.
| |
Collapse
|
27
|
Sayal P, Zafar A, Highley R. A Rare Case of Concurrent Herpes Simplex Encephalitis and Glioblastoma Multiforme. Asian J Neurosurg 2018; 13:78-82. [PMID: 29492128 PMCID: PMC5820902 DOI: 10.4103/1793-5482.181141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Herpes encephalitis superimposed on an intracranial malignancy has previously been described mainly in the context of malignancy imitating infection or in the postoperative setting after neurosurgical intervention. We report a rare case of de novo presentation of concurrent herpes encephalitis and glioblastoma. A 63-year-old man presented with status epilepticus and subsequent magnetic resonance imaging (MRI) brain showed a right temporal enhancing lesion with mass effect. He underwent a craniotomy and debulking of this lesion, which on subsequent histology was positive for herpes simplex virus (HSV) antigens and HSV DNA was confirmed by polymerase chain reaction analysis. The sample however also had some hypercellular areas with atypical astrocytes. Our patient recovered well from surgery and was eventually commenced on acyclovir albeit with a delay of 3 weeks due to the initial diagnostic dilemma. However, he re-presented with lethargy and confusion a further 3 weeks later and an MRI scan showed recurrence of the temporal lesion with MR spectroscopy more suggestive of high-grade glioma. He, therefore, underwent a further debulking surgery and the histology revealed a WHO Grade 4 glioblastoma with some residual areas of inflammation. A diagnosis of 2 co-existing pathologies namely HSV encephalitis and glioblastoma was thus reached. Unfortunately, due to poor performance status, he could not undergo chemo-radiotherapy and died 8 months after presentation. Immuno-modulators, expressed locally and globally in glioma patients, are likely to render them susceptible to infections. There are an increasing number of reports of HSV encephalitis in the glioma setting postoperatively. However, we report a de novo presentation which has only been recognized once before in the 1970s. Recognition of HSV encephalitis in glioma patients in the de novo and also the postoperative context is important for commencing early treatment and preventing poor outcomes.
Collapse
Affiliation(s)
- Parag Sayal
- Department of Neurosurgery, The National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Arif Zafar
- Department of Neurosurgery, Hull Royal Infirmary, Hull, United Kingdom
| | - Robin Highley
- Department of Neuropathology, Hull Royal Infirmary, Hull, United Kingdom
| |
Collapse
|
28
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
29
|
Cui X, Morales RTT, Qian W, Wang H, Gagner JP, Dolgalev I, Placantonakis D, Zagzag D, Cimmino L, Snuderl M, Lam RHW, Chen W. Hacking macrophage-associated immunosuppression for regulating glioblastoma angiogenesis. Biomaterials 2018; 161:164-178. [PMID: 29421553 DOI: 10.1016/j.biomaterials.2018.01.053] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GBM) is the most lethal primary adult brain tumor and its pathology is hallmarked by distorted neovascularization, diffuse tumor-associated macrophage infiltration, and potent immunosuppression. Reconstituting organotypic tumor angiogenesis models with biomimetic cell heterogeneity and interactions, pro-/anti-inflammatory milieu and extracellular matrix (ECM) mechanics is critical for preclinical anti-angiogenic therapeutic screening. However, current in vitro systems do not accurately mirror in vivo human brain tumor microenvironment. Here, we engineered a three-dimensional (3D), microfluidic angiogenesis model with controllable and biomimetic immunosuppressive conditions, immune-vascular and cell-matrix interactions. We demonstrate in vitro, GL261 and CT-2A GBM-like tumors steer macrophage polarization towards a M2-like phenotype for fostering an immunosuppressive and proangiogenic niche, which is consistent with human brain tumors. We distinguished that GBM and M2-like immunosuppressive macrophages promote angiogenesis, while M1-like pro-inflammatory macrophages suppress angiogenesis, which we coin "inflammation-driven angiogenesis." We observed soluble immunosuppressive cytokines, predominantly TGF-β1, and surface integrin (αvβ3) endothelial-macrophage interactions are required in inflammation-driven angiogenesis. We demonstrated tuning cell-adhesion receptors using an integrin (αvβ3)-specific collagen hydrogel regulated inflammation-driven angiogenesis through Src-PI3K-YAP signaling, highlighting the importance of altered cell-ECM interactions in inflammation. To validate the preclinical applications of our 3D organoid model and mechanistic findings of inflammation-driven angiogenesis, we screened a novel dual integrin (αvβ3) and cytokine receptor (TGFβ-R1) blockade that suppresses GBM tumor neovascularization by simultaneously targeting macrophage-associated immunosuppression, endothelial-macrophage interactions, and altered ECM. Hence, we provide an interactive and controllable GBM tumor microenvironment and highlight the importance of macrophage-associated immunosuppression in GBM angiogenesis, paving a new direction of screening novel anti-angiogenic therapies.
Collapse
Affiliation(s)
- Xin Cui
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Renee-Tyler Tan Morales
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Weiyi Qian
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Haoyu Wang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Jean-Pierre Gagner
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Igor Dolgalev
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Dimitris Placantonakis
- Department of Neurosurgery, New York University School of Medicine, New York, NY 10016, USA
| | - David Zagzag
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Neurosurgery, New York University School of Medicine, New York, NY 10016, USA
| | - Luisa Cimmino
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Matija Snuderl
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Raymond H W Lam
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong.
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA.
| |
Collapse
|
30
|
Optimizing EphA2-CAR T Cells for the Adoptive Immunotherapy of Glioma. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 9:70-80. [PMID: 29552579 PMCID: PMC5852415 DOI: 10.1016/j.omtm.2018.01.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/16/2018] [Indexed: 11/23/2022]
Abstract
Glioblastoma is the most aggressive primary brain tumor in humans and is virtually incurable with conventional therapies. Chimeric antigen receptor (CAR) T cell therapy targeting the glioblastoma antigen EphA2 is an attractive approach to improve outcomes because EphA2 is expressed highly in glioblastoma but only at low levels in normal brain tissue. Building upon our previous findings in this area, we generated and evaluated a panel of EphA2-specific CARs. We demonstrate here that T cells expressing CD28.ζ and 41BB.ζ CARs with short spacers had similar effector function, resulting in potent antitumor activity. In addition, incorporating the 41BB signaling domain into CD28.ζ CARs did not improve CAR T cell function. While we could not determine functional differences between CD28.ζ, 41BB.ζ, and CD28.41BB.ζ CAR T cells, we selected CD28.ζ CAR T cells for further clinical development based on safety consideration.
Collapse
|
31
|
Kamran N, Alghamri MS, Nunez FJ, Shah D, Asad AS, Candolfi M, Altshuler D, Lowenstein PR, Castro MG. Current state and future prospects of immunotherapy for glioma. Immunotherapy 2018; 10:317-339. [PMID: 29421984 PMCID: PMC5810852 DOI: 10.2217/imt-2017-0122] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/30/2017] [Indexed: 12/14/2022] Open
Abstract
There is a large unmet need for effective therapeutic approaches for glioma, the most malignant brain tumor. Clinical and preclinical studies have enormously expanded our knowledge about the molecular aspects of this deadly disease and its interaction with the host immune system. In this review we highlight the wide array of immunotherapeutic interventions that are currently being tested in glioma patients. Given the molecular heterogeneity, tumor immunoediting and the profound immunosuppression that characterize glioma, it has become clear that combinatorial approaches targeting multiple pathways tailored to the genetic signature of the tumor will be required in order to achieve optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Kamran
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Felipe J Nunez
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Diana Shah
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Antonela S Asad
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - David Altshuler
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Maria G Castro
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| |
Collapse
|
32
|
Jacobs DI, Liu Y, Gabrusiewicz K, Tsavachidis S, Armstrong GN, Zhou R, Wei J, Ivan C, Calin G, Molinaro AM, Rice T, Bracci PM, Hansen HM, Wiencke JK, Wrensch MR, Heimberger AB, Bondy ML. Germline polymorphisms in myeloid-associated genes are not associated with survival in glioma patients. J Neurooncol 2018; 136:33-39. [PMID: 28965162 PMCID: PMC5756111 DOI: 10.1007/s11060-017-2622-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/08/2017] [Indexed: 01/07/2023]
Abstract
Immune cells of myeloid origin, including microglia, macrophages, and myeloid-derived suppressor cells adopt immunosuppressive phenotypes that support gliomagenesis. Here, we tested an a priori hypothesis that single nucleotide polymorphisms (SNPs) in genes related to glioma-associated myeloid cell regulation and function are also associated with patient survival after glioma diagnosis. Subjects for this study were 992 glioma patients treated at The University of Texas MD Anderson Cancer Center in Houston, Texas between 1992 and 2008. Haplotype-tagging SNPs in 91 myeloid-associated genes were analyzed for association with survival by Cox regression. Individual SNP- and gene-based tests were performed separately in glioblastoma (WHO grade IV, n = 511) and lower-grade glioma (WHO grade II-III, n = 481) groups. After adjustment for multiple testing, no myeloid-associated gene variants were significantly associated with survival in glioblastoma. Two SNPs, rs147960238 in CD163 (p = 2.2 × 10-5) and rs17138945 in MET (p = 5.6 × 10-5) were significantly associated with survival of patients with lower-grade glioma. However, these associations were not confirmed in an independent analysis of 563 lower-grade glioma cases from the University of California at San Francisco Adult Glioma Study (p = 0.65 and p = 0.41, respectively). The results of this study do not support a role for inherited polymorphisms in myeloid-associated genes in affecting survival of patients diagnosed with glioblastoma or lower-grade glioma.
Collapse
Affiliation(s)
- Daniel I Jacobs
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Yanhong Liu
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Konrad Gabrusiewicz
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Spiridon Tsavachidis
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Georgina N Armstrong
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Renke Zhou
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Jun Wei
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Annette M Molinaro
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Paige M Bracci
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret R Wrensch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Melissa L Bondy
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA.
| |
Collapse
|
33
|
Liu Q, Xue Y, Chen Q, Chen H, Zhang X, Wang L, Han C, Que S, Lou M, Lan J. PomGnT1 enhances temozolomide resistance by activating epithelial-mesenchymal transition signaling in glioblastoma. Oncol Rep 2017; 38:2911-2918. [PMID: 29048655 DOI: 10.3892/or.2017.5964] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/28/2017] [Indexed: 11/06/2022] Open
Abstract
Temozolomide (TMZ) is commonly used in glioblastoma (GBM) chemotherapy. However, a great challenge for TMZ treatment is the rapid development of resistance and subsequent tumor recurrence and poor outcome. In the present study we established TMZ-resistant GBM cells (U87-TR and U251-TR) and found that the expression of PomGnT1 was significantly upregulated in TMZ-resistant GBM cells compared with the TMZ-sensitive counterparts. Furthermore, overexpression of PomGnT1 in U87-MG and U251-MG cells led to increased IC50 values for TMZ and reduced apoptosis of cells. Knockdown of PomGnT1 in both U87-TR and U251-TR cells led to decreased IC50 values for TMZ and enhanced apoptosis. Biochemical analysis revealed that PomGnT1 regulates the expression of factors in epithelial-mesenchymal transition signaling including TCF8, vimentin, β-catenin and Slug in GBM cells. These findings demonstrate that PomGnT1 might be a new focus of GBM research for treatment of recurrent TMZ-resistant GBM.
Collapse
Affiliation(s)
- Qi Liu
- Department of Neurosurgery, Jingdezhen Second Hospital, Jingdezhen, Jiangxi, P.R. China
| | - Yajun Xue
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Qingshan Chen
- Department of Neurosurgery, The Second People's Hospital of Liaocheng City, Liaocheng, Shandong, P.R. China
| | - Huairui Chen
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Xiaofei Zhang
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Leiping Wang
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Cong Han
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Shuanglin Que
- Department of Neurosurgery, Longyan First Hospital, Fujian Medical University, Longyan, Fujian, P.R. China
| | - Meiqing Lou
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jin Lan
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
34
|
Miska J, Rashidi A, Chang AL, Muroski ME, Han Y, Zhang L, Lesniak MS. Anti-GITR therapy promotes immunity against malignant glioma in a murine model. Cancer Immunol Immunother 2016; 65:1555-1567. [PMID: 27734112 DOI: 10.1007/s00262-016-1912-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/30/2016] [Indexed: 01/06/2023]
Abstract
Regulatory T cells (Tregs) are potently immunosuppressive cells that accumulate within the glioma microenvironment. The reduction in their function and/or trafficking has been previously shown to enhance survival in preclinical models of glioma. Glucocorticoid-induced TNFR-related protein (GITR) is a tumor necrosis factor superfamily receptor enriched on Tregs that has shown promise as a target for immunotherapy. An agonistic antibody against GITR has been demonstrated to inhibit Tregs in a number of models and has only been recently addressed in glioma. In this study, we examined the modality of the antibody function at the tumor site as opposed to the periphery as the blood-brain barrier prevents efficient antibody delivery to brain tumors. Mice harboring established GL261 tumors were treated with anti-GITR monotherapy and were shown to have a significant increase in overall survival (p < 0.01) when antibodies were injected directly into the glioma core, whereas peripheral antibody treatment only had a modest effect. Peripheral treatment resulted in a significant decrease in granzyme B (GrB) expression by Tregs, whereas intratumoral treatment resulted in both a decrease in GrB expression by Tregs and their selective depletion, which was largely mediated by FcγR-mediated destruction. We also discovered that anti-GITR treatment results in the enhanced survival and functionality of dendritic cells (DCs)-a previously unreported effect of this immunotherapy. In effect, this study demonstrates that the targeting of GITR is a feasible and noteworthy treatment option for glioma, but is largely dependent on the anatomical location in which the antibodies are delivered.
Collapse
Affiliation(s)
- Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Alan L Chang
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Megan E Muroski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Yu Han
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Lingjiao Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| |
Collapse
|
35
|
Penner F, Brossa S, Barbui AM, Ducati A, Cavallo R, Zenga F. Caulobacter spp: A Rare Pathogen Responsible for Paucisintomatic Persisitant Meningitis in a Glioblastoma Patient. World Neurosurg 2016; 96:611.e11-611.e13. [PMID: 27650802 DOI: 10.1016/j.wneu.2016.09.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Caulobacter spp. are Gram-negative bacteria that have rarely been found to be pathogenic in humans. CASE DESCRIPTION This report describes the first case, to our knowledge, of meningitis in an adult patient caused by Caulobacter spp. A 75-year-old man was operated for a glioblastoma with no evident signs of primary infection in the wound site. Eight days after surgery, the patient developed signs and symptoms of meningitis. Caulobacter was then isolated on 3 separate occasions in the patient's cerebrospinal fluid. Thereafter, specific antibiotic therapy began. After 2 weeks of therapy, the patient was discharged with complete resolution of any related symptoms. CONCLUSIONS Caulobacter spp. can cause adult meningitis even where there is no evidence of surgical site infection.
Collapse
Affiliation(s)
- Federica Penner
- Division of Neurosurgery, Department of Neuroscience, University of Torino, Turin, Italy.
| | - Silvia Brossa
- Microbiology and Virology Laboratory, University of Turin, Turin, Italy
| | - Anna Maria Barbui
- Microbiology and Virology Laboratory, University of Turin, Turin, Italy
| | - Alessandro Ducati
- Division of Neurosurgery, Department of Neuroscience, University of Torino, Turin, Italy
| | - Rossana Cavallo
- Microbiology and Virology Laboratory, University of Turin, Turin, Italy
| | - Francesco Zenga
- Division of Neurosurgery, Department of Neuroscience, University of Torino, Turin, Italy
| |
Collapse
|
36
|
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: 3.9] [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
|
37
|
Parajuli P, Anand R, Mandalaparty C, Suryadevara R, Sriranga PU, Michelhaugh SK, Cazacu S, Finniss S, Thakur A, Lum LG, Schalk D, Brodie C, Mittal S. Preferential expression of functional IL-17R in glioma stem cells: potential role in self-renewal. Oncotarget 2016; 7:6121-35. [PMID: 26755664 PMCID: PMC4868744 DOI: 10.18632/oncotarget.6847] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/26/2015] [Indexed: 02/07/2023] Open
Abstract
Gliomas are the most common primary brain tumor and one of the most lethal solid tumors. Mechanistic studies into identification of novel biomarkers are needed to develop new therapeutic strategies for this deadly disease. The objective for this study was to explore the potential direct impact of IL-17-IL-17R interaction in gliomas. Immunohistochemistry and flow cytometry analysis of 12 tumor samples obtained from patients with high grade gliomas revealed that a considerable population (2-19%) of cells in all malignant gliomas expressed IL-17RA, with remarkable co-expression of the glioma stem cell (GSC) markers CD133, Nestin, and Sox2. IL-17 enhanced the self-renewal of GSCs as determined by proliferation and Matrigel® colony assays. IL-17 also induced cytokine/chemokine (IL-6, IL-8, interferon-γ-inducible protein [IP-10], and monocyte chemoattractant protein-1 [MCP-1]) secretion in GSCs, which were differentially blocked by antibodies against IL-17R and IL-6R. Western blot analysis showed that IL-17 modulated the activity of signal transducer and activator of transcription 3 (STAT3), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), glycogen synthase kinase-3β (GSK-3β) and β-catenin in GSCs. While IL-17R-mediated secretion of IL-6 and IL-8 were significantly blocked by inhibitors of NF-κB and STAT3; NF-κB inhibitor was more potent than STAT3 inhibitor in blocking IL-17-induced MCP-1 secretion. Overall, our results suggest that IL-17-IL-17R interaction in GSCs induces an autocrine/paracrine cytokine feedback loop, which may provide an important signaling component for maintenance/self-renewal of GSCs via constitutive activation of both NF-κB and STAT3. The results also strongly implicate IL-17R as an important functional biomarker for therapeutic targeting of GSCs.
Collapse
Affiliation(s)
- Prahlad Parajuli
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Rohit Anand
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | | | - Raviteja Suryadevara
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Preethi U. Sriranga
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Sharon K. Michelhaugh
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Simona Cazacu
- Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Susan Finniss
- Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Archana Thakur
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Lawrence G. Lum
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Internal Medicine, Immunology and Microbiology, and Pediatrics, Wayne State University, Detroit, MI, USA
| | - Dana Schalk
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| | - Chaya Brodie
- Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
38
|
The Dichotomy of Tumor Exosomes (TEX) in Cancer Immunity: Is It All in the ConTEXt? Vaccines (Basel) 2015; 3:1019-51. [PMID: 26694473 PMCID: PMC4693230 DOI: 10.3390/vaccines3041019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/24/2015] [Accepted: 12/05/2015] [Indexed: 02/06/2023] Open
Abstract
Exosomes are virus-sized nanoparticles (30–130 nm) formed intracellularly as intravesicular bodies/intralumenal vesicles within maturing endosomes (“multivesicular bodies”, MVBs). If MVBs fuse with the cell’s plasma membrane, the interior vesicles may be released extracellularly, and are termed “exosomes”. The protein cargo of exosomes consists of cytosolic, membrane, and extracellular proteins, along with membrane-derived lipids, and an extraordinary variety of nucleic acids. As such, exosomes reflect the status and identity of the parent cell, and are considered as tiny cellular surrogates. Because of this closely entwined relationship between exosome content and the source/status of the parental cell, conceivably exosomes could be used as vaccines against various pathologies, as they contain antigens associated with a given disease, e.g., cancer. Tumor-derived exosomes (TEX) have been shown to be potent anticancer vaccines in animal models, driving antigen-specific T and B cell responses, but much recent literature concerning TEX strongly places the vesicles as powerfully immunosuppressive. This dichotomy suggests that the context in which the immune system encounters TEX is critical in determining immune stimulation versus immunosuppression. Here, we review literature on both sides of this immune coin, and suggest that it may be time to revisit the concept of TEX as anticancer vaccines in clinical settings.
Collapse
|
39
|
Hellwinkel JE, Redzic JS, Harland TA, Gunaydin D, Anchordoquy TJ, Graner MW. Glioma-derived extracellular vesicles selectively suppress immune responses. Neuro Oncol 2015; 18:497-506. [PMID: 26385614 DOI: 10.1093/neuonc/nov170] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 07/23/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioma-related immunosuppression is well documented; however, the mechanisms of suppression are not fully understood. Here we explore a role for glioma extracellular vesicles (EVs) as a means of immune modulation. METHODS Healthy donor peripheral blood mononuclear cells (PBMCs) were incubated with mitogenic stimuli and various concentrations of glioma-derived EVs. Intracellular signaling and cytokine output were determined by protein microarrays, and phenotypic changes were assessed by flow cytometry. Recall antigen testing, mixed lymphocyte reactions, and migration assays analyzed PBMC functional capacity. RESULTS Protein microarray data revealed induction of an immunosuppressive phenotype and cytokine output at high tumor-vesicle concentrations but an activated phenotype at low concentrations. T cell activation antigen expression confirmed differential activation profiles. Functional analyses revealed decreased migratory capacity of PBMCs after incubation with EVs; however, recall antigen and mixed lymphocyte tests indicated that activation capacity is still retained in EV-treated cells. CONCLUSION The differential effects of high and low EV concentrations dictate modulatory effects on PBMCs. These data provide a role for EVs at high concentrations for inducing selective tolerance of an immune response in a tumor setting. This suggests that lymphocytes in patients' circulation are not irreparably impaired, as previously thought, but can be rescued to augment antitumor responses.
Collapse
Affiliation(s)
- Justin E Hellwinkel
- Dept of Neurosurgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.E.H, T.A.H, D.G., M.W.G); Skaggs School of Pharmacy, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.S.R, T.J.A)
| | - Jasmina S Redzic
- Dept of Neurosurgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.E.H, T.A.H, D.G., M.W.G); Skaggs School of Pharmacy, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.S.R, T.J.A)
| | - Tessa A Harland
- Dept of Neurosurgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.E.H, T.A.H, D.G., M.W.G); Skaggs School of Pharmacy, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.S.R, T.J.A)
| | - Dicle Gunaydin
- Dept of Neurosurgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.E.H, T.A.H, D.G., M.W.G); Skaggs School of Pharmacy, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.S.R, T.J.A)
| | - Thomas J Anchordoquy
- Dept of Neurosurgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.E.H, T.A.H, D.G., M.W.G); Skaggs School of Pharmacy, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.S.R, T.J.A)
| | - Michael W Graner
- Dept of Neurosurgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.E.H, T.A.H, D.G., M.W.G); Skaggs School of Pharmacy, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado (J.S.R, T.J.A)
| |
Collapse
|
40
|
Glass R, Synowitz M. CNS macrophages and peripheral myeloid cells in brain tumours. Acta Neuropathol 2014; 128:347-62. [PMID: 24722970 DOI: 10.1007/s00401-014-1274-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/10/2014] [Accepted: 03/25/2014] [Indexed: 12/15/2022]
Abstract
Primary brain tumours (gliomas) initiate a strong host response and can contain large amounts of immune cells (myeloid cells) such as microglia and tumour-infiltrating macrophages. In gliomas the course of pathology is not only controlled by the genetic make-up of the tumour cells, but also depends on the interplay with myeloid cells in the tumour microenvironment. Especially malignant gliomas such as glioblastoma multiforme (GBM) are notoriously immune-suppressive and it is now evident that GBM cells manipulate myeloid cells to support tumour expansion. The protumorigenic effects of glioma-associated myeloid cells comprise a support for angiogenesis as well as tumour cell invasion, proliferation and survival. Different strategies for inhibiting the pathological functions of myeloid cells in gliomas are explored, and blocking the tropism of microglia/macrophages to gliomas or manipulating the signal transduction pathways for immune cell activation has been successful in pre-clinical models. Hence, myeloid cells are now emerging as a promising target for new adjuvant therapies for gliomas. However, it is also becoming evident that some myeloid-directed glioma therapies may only be beneficial for distinct subclasses of gliomas and that a more cell-type-specific manipulation of either microglia or macrophages may improve therapeutic outcomes.
Collapse
|
41
|
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
|
42
|
Bhatia A, Kumar Y. Cellular and molecular mechanisms in cancer immune escape: a comprehensive review. Expert Rev Clin Immunol 2013; 10:41-62. [PMID: 24325346 DOI: 10.1586/1744666x.2014.865519] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immune escape is the final phase of cancer immunoediting process wherein cancer modulates our immune system to escape from being destroyed by it. Many cellular and molecular events govern the cancer's evasion of host immune response. The tumor undergoes continuous remodeling at the genetic, epigenetic and metabolic level to acquire resistance to apoptosis. At the same time, it effectively modifies all the components of the host's immunome so as to escape from its antitumor effects. Moreover, it induces accumulation of suppressive cells like Treg and myeloid derived suppressor cells and factors which also enable it to elude the immune system. Recent research in this area helps in defining the role of newer players like miRNAs and exosomes in immune escape. The immunotherapeutic approaches developed to target the escape phase appear quite promising; however, the quest for a perfect therapeutic agent that can achieve maximum cure with minimal toxicity continues.
Collapse
Affiliation(s)
- Alka Bhatia
- Department of Experimental Medicine & Biotechnology, PGIMER, Chandigarh-160012, India
| | | |
Collapse
|
43
|
Prosniak M, Harshyne LA, Andrews DW, Kenyon LC, Bedelbaeva K, Apanasovich TV, Heber-Katz E, Curtis MT, Cotzia P, Hooper DC. Glioma grade is associated with the accumulation and activity of cells bearing M2 monocyte markers. Clin Cancer Res 2013; 19:3776-86. [PMID: 23741072 DOI: 10.1158/1078-0432.ccr-12-1940] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE This study is directed at identifying the cell source(s) of immunomodulatory cytokines in high-grade gliomas and establishing whether the analysis of associated markers has implications for tumor grading. EXPERIMENTAL DESIGN Glioma specimens classified as WHO grade II-IV by histopathology were assessed by gene expression analysis and immunohistochemistry to identify the cells producing interleukin (IL)-10, which was confirmed by flow cytometry and factor secretion in culture. Finally, principal component analysis (PCA) and mixture discriminant analysis (MDA) were used to investigate associations between expressed genes and glioma grade. RESULTS The principle source of glioma-associated IL-10 is a cell type that bears phenotype markers consistent with M2 monocytes but does not express all M2-associated genes. Measures of expression of the M2 cell markers CD14, CD68, CD163, and CD204, which are elevated in high-grade gliomas, and the neutrophil/myeloid-derived suppressor cell (MDSC) subset marker CD15, which is reduced, provide the best index of glioma grade. CONCLUSIONS Grade II and IV astrocytomas can be clearly differentiated on the basis of the expression of certain M2 markers in tumor tissues, whereas grade III astrocytomas exhibit a range of expression between the lower and higher grade specimens. The content of CD163(+) cells distinguishes grade III astrocytoma subsets with different prognosis.
Collapse
Affiliation(s)
- Michael Prosniak
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
There is increasing evidence in the literature pointing to an important role of inflammation during initiation and progression of cancer. Glioblastoma is the most common malignant primary brain tumor with approximately 23,000 newly-diagnosed cases each year in the United States, and has a dismal median survival of only 15 months. Although the blood-brain barrier maintains an immune-privileged status of the brain under steady state, intracranial tumors including gliomas are invariably infiltrated with various types of immune cells. The T helper 17 (Th17) cells, a recently discovered interleukin (IL)-17-producing T cell subtype, have been reported in several extracranial and some intracranial tumors, where they have been implicated in either pro- or antitumor activity depending on the tumor type. Here, we present a succinct review of the current literature on the prevalence and potential role of IL-17 in malignant gliomas. Further mechanistic studies on IL-17 mediated inflammatory pathway in gliomas may provide with opportunities for novel immunotherapeutic interventions.
Collapse
Affiliation(s)
- Prahlad Parajuli
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, USA
| |
Collapse
|
45
|
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: 359] [Impact Index Per Article: 27.6] [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
|
46
|
Verschuere T, De Vleeschouwer S, Lefranc F, Kiss R, Van Gool SW. Galectin-1 and immunotherapy for brain cancer. Expert Rev Neurother 2011; 11:533-43. [PMID: 21469926 DOI: 10.1586/ern.11.40] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prognosis of patients diagnosed with high-grade glioma continues to be dismal in spite of multimodal treatment. Active specific immunotherapy by means of dendritic cell vaccination is considered to be a new promising concept that aims at generating an anti-tumoral immune response. However, it is now widely accepted that the success of immunotherapeutic strategies to promote tumor regression will rely not only on enhancing the effector arm of the immune response but also on downregulation of the counteracting tolerogenic signals. In this article, we summarize evidence that galectin-1, an evolutionarily conserved glycan-binding protein that is abundantly expressed in high-grade glioma, is an important player in glioma-mediated immune escape.
Collapse
Affiliation(s)
- Tina Verschuere
- Laboratory of Experimental Immunology, Catholic University Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
47
|
Scutellaria extract and wogonin inhibit tumor-mediated induction of T(reg) cells via inhibition of TGF-β1 activity. Cancer Immunol Immunother 2011; 61:701-11. [PMID: 22057676 DOI: 10.1007/s00262-011-1130-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 10/14/2011] [Indexed: 12/22/2022]
Abstract
A number of studies have implicated tumor-induced T(reg) cell activity in the sub-optimal response to therapeutic vaccines. Development of neo-adjuvant strategies targeting T(reg) cells is therefore imperative. Scutellaria extracts or constituent flavonoids have shown encouraging efficacy against various tumors, including gliomas, in both pre-clinical and clinical studies. We report here, for the first time, that Scutellaria ocmulgee leaf extract (SocL) and flavonoid wogonin could inhibit TGF-β1-induced T(reg) activity in malignant gliomas. F344 rats, subcutaneously transplanted with F98 gliomas, were treated with SocL. There was a significant inhibition of intra-tumoral TGF-β1 and T(reg) cell frequency as well as peripheral blood TGF-β1 levels in SocL-treated animals compared to the controls. SocL extract and wogonin also inhibited glioma-induced, TGF-β1-mediated T(reg) activity in vitro. SocL extract and wogonin also inhibited the secretion of IL-10 in T(reg) culture; whereas the level of IL-2 was either unchanged or marginally enhanced. We also observed an inhibition of Smad-3, GSK-3β and ERK1/2 signaling by SocL and wogonin in T(reg) cells, while phosphorylation of P38 MAPK was considerably enhanced, indicating that SocL or wogonin could inhibit the T cells' response to TGF-β1 via modulation of both Smad and non-Smad signaling pathways. Overall, this study suggests that Scutellaria can potentially reverse tumor-mediated immune suppression via inhibition of TGF-β1 secretion as well as via inhibition of T cells' response to TGF-β1. This may provide an opportunity for developing a novel adjuvant therapeutic strategy for malignant gliomas, combining Scutellaria with immunotherapy and chemo/radio-therapeutic regimen, which could potentially improve the disease outcome.
Collapse
|
48
|
Westphal M, Lamszus K. The neurobiology of gliomas: from cell biology to the development of therapeutic approaches. Nat Rev Neurosci 2011; 12:495-508. [PMID: 21811295 DOI: 10.1038/nrn3060] [Citation(s) in RCA: 218] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gliomas are the most common type of primary brain tumour and are often fast growing with a poor prognosis for the patient. Their complex cellular composition, diffuse invasiveness and capacity to escape therapies has challenged researchers for decades and hampered progress towards an effective treatment. Recent molecular characterization of tumour cells combined with new insights into cellular diversification that occurs during development, and the modelling of these processes in transgenic animals have enabled a more detailed understanding of the events that underlie gliomagenesis. Combining this enhanced understanding of the relationship between neural stem cell biology and the cell lineage relationships of tumour cells with model systems offers new opportunities to develop specific and effective therapies.
Collapse
Affiliation(s)
- Manfred Westphal
- Department of Neurosurgery, University Hospital Hamburg Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | |
Collapse
|
49
|
Gousias K, Kyritsis A. Comment on “Opportunistic cytomegalovirus infection in a patient receiving temozolomide for treatment of malignant glioma”. J Clin Neurosci 2010; 17:412. [DOI: 10.1016/j.jocn.2009.07.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 07/20/2009] [Indexed: 10/19/2022]
|
50
|
Candolfi M, Kroeger KM, Muhammad AKMG, Yagiz K, Farrokhi C, Pechnick RN, Lowenstein PR, Castro MG. Gene therapy for brain cancer: combination therapies provide enhanced efficacy and safety. Curr Gene Ther 2010; 9:409-21. [PMID: 19860655 DOI: 10.2174/156652309789753301] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer in adults. Despite significant advances in treatment and intensive research, the prognosis for patients with GBM remains poor. Therapeutic challenges for GBM include its invasive nature, the proximity of the tumor to vital brain structures often preventing total resection, and the resistance of recurrent GBM to conventional radiotherapy and chemotherapy. Gene therapy has been proposed as a useful adjuvant for GBM, to be used in conjunction with current treatment. Work from our laboratory has shown that combination of conditional cytotoxic with immunotherapeutic approaches for the treatment of GBM elicits regression of large intracranial tumor masses and anti-tumor immunological memory in syngeneic rodent models of GBM. In this review we examined the currently available animal models for GBM, including rodent transplantable models, endogenous rodent tumor models and spontaneous GBM in dogs. We discuss non-invasive surrogate end points to assess tumor progression and therapeutic efficacy, such as behavioral tests and circulating biomarkers. Growing preclinical and clinical data contradict the old dogma that cytotoxic anti-cancer therapy would lead to an immune-suppression that would impair the ability of the immune system to mount an anti-tumor response. The implications of the findings reviewed indicate that combination of cytotoxic therapy with immunotherapy will lead to synergistic antitumor efficacy with reduced neurotoxicity and supports the clinical implementation of combined cytotoxic-immunotherapeutic strategies for the treatment of patients with GBM.
Collapse
Affiliation(s)
- Marianela Candolfi
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | |
Collapse
|