1
|
Peng P, Cheng F, Dong Y, Chen Z, Zhang X, Guo D, Yu X, Lu Y, Ke Y, Zhang B, He X, Wan F. High expression of TXNDC11 indicated unfavorable prognosis of glioma. Transl Cancer Res 2022; 10:5040-5051. [PMID: 35116356 PMCID: PMC8799221 DOI: 10.21037/tcr-21-1326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022]
Abstract
Background Thioredoxin domain containing 11 (TXNDC11) has been implicated in numerous cancers. Nevertheless, the function of TXNDC11 in glioma is not well described. This study aimed to assess clinical significance of TXNDC11 in glioma based on bioinformatics analysis and immunohistochemical (IHC) staining. Methods GEPIA2, The Cancer Genome Atlas (TCGA), and Gene Expression Omnibus (GEO) databases were employed to detect the levels of TXNDC11 transcript in glioma. Gene expression profiles and data from the methylation chip with clinical details from TCGA and Chinese Glioma Genome Atlas (CGGA) of glioma samples were examined. The methylation of TXNDC11 in glioma was evaluated by 450K methylation chip data analysis. The pathways involved in TXNDC11 expression were screened by gene set enrichment analysis (GSEA). The correlation between TXNDC11 and immune cells was analyzed. Protein level of TXNDC11 was detected by IHC staining in glioma specimens. Results TXNDC11 was highly expressed in glioma, and high TXNDC11 expression was associated with poor overall survival (OS) and worse clinical prognostic variables. The methylation of cg04399632 was statistically different between glioma samples and normal samples, and was negatively correlated with TXNDC11 expression in glioma patients. Survival analysis demonstrated a poorer prognosis in glioma patients with cg04399632 hypomethylation. TXNDC11-high phenotype was associated with certain immune-related pathways and other signaling pathways in glioma. The expression of TXNDC11 was correlated positively with M2 macrophage infiltration and negatively with M0 and M1 macrophage infiltration. IHC staining confirmed that TXNDC11 expression increased in higher-grade glioma. Conclusions High expression of TXNDC11 may predict unfavorable prognosis of glioma patients.
Collapse
Affiliation(s)
- Peng Peng
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangling Cheng
- Department of Surgery, Hepatic Surgery Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuting Dong
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Zirong Chen
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolin Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjiang Yu
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyang Lu
- School of Data Science, Chinese University of Hong Kong, Shenzhen, China
| | - Yuyong Ke
- Department of Neurosurgery, Renmin Hospital of Yangxin County, Huangshi, China
| | - Bin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ximiao He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wan
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Audi ZF, Saker Z, Rizk M, Harati H, Fares Y, Bahmad HF, Nabha SM. Immunosuppression in Medulloblastoma: Insights into Cancer Immunity and Immunotherapy. Curr Treat Options Oncol 2021; 22:83. [PMID: 34328587 DOI: 10.1007/s11864-021-00874-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT Medulloblastoma (MB) is the most common pediatric brain malignancy, with a 5-year overall survival (OS) rate of around 65%. The conventional MB treatment, comprising surgical resection followed by irradiation and adjuvant chemotherapy, often leads to impairment in normal body functions and poor quality of life, especially with the increased risk of recurrence and subsequent development of secondary malignancies. The development and progression of MB are facilitated by a variety of immune-evading mechanisms such as the secretion of immunosuppressive molecules, activation of immunosuppressive cells, inhibition of immune checkpoint molecules, impairment of adhesive molecules, downregulation of the major histocompatibility complex (MHC) molecules, protection against apoptosis, and activation of immunosuppressive pathways. Understanding the tumor-immune relationship in MB is crucial for effective development of immune-based therapeutic strategies. In this comprehensive review, we discuss the immunological aspect of the brain, focusing on the current knowledge tackling the mechanisms of MB immune suppression and evasion. We also highlight several key immunotherapeutic approaches developed to date for the treatment of MB.
Collapse
Affiliation(s)
- Zahraa F Audi
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.,Department of Neurosurgery, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, USA.
| | - Sanaa M Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
3
|
Grabowski MM, Sankey EW, Ryan KJ, Chongsathidkiet P, Lorrey SJ, Wilkinson DS, Fecci PE. Immune suppression in gliomas. J Neurooncol 2021; 151:3-12. [PMID: 32542437 PMCID: PMC7843555 DOI: 10.1007/s11060-020-03483-y] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The overall survival in patients with gliomas has not significantly increased in the modern era, despite advances such as immunotherapy. This is in part due to their notorious ability to suppress local and systemic immune responses, severely restricting treatment efficacy. METHODS We have reviewed the preclinical and clinical evidence for immunosuppression seen throughout the disease process in gliomas. This review aims to discuss the various ways that brain tumors, and gliomas in particular, co-opt the body's immune system to evade detection and ensure tumor survival and proliferation. RESULTS A multitude of mechanisms are discussed by which neoplastic cells evade detection and destruction by the immune system. These include tumor-induced T-cell and NK cell dysfunction, regulatory T-cell and myeloid-derived suppressor cell expansion, M2 phenotypic transformation in glioma-associated macrophages/microglia, upregulation of immunosuppressive glioma cell surface factors and cytokines, tumor microenvironment hypoxia, and iatrogenic sequelae of immunosuppressive treatments. CONCLUSIONS Gliomas create a profoundly immunosuppressive environment, both locally within the tumor and systemically. Future research should aim to address these immunosuppressive mechanisms in the effort to generate treatment options with meaningful survival benefits for this patient population.
Collapse
Affiliation(s)
- Matthew M Grabowski
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA
| | - Eric W Sankey
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA
| | - Katherine J Ryan
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA
| | - Pakawat Chongsathidkiet
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA
| | - Selena J Lorrey
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA
| | - Daniel S Wilkinson
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, 303 Research Drive, 220 Sands Bldg, Durham, NC, 27710, USA.
| |
Collapse
|
4
|
Blessing MM, Blackburn PR, Krishnan C, Harrod VL, Barr Fritcher EG, Zysk CD, Jackson RA, Milosevic D, Nair AA, Davila JI, Balcom JR, Jenkins RB, Halling KC, Kipp BR, Nageswara Rao AA, Laack NN, Daniels DJ, Macon WR, Ida CM. Desmoplastic Infantile Ganglioglioma: A MAPK Pathway-Driven and Microglia/Macrophage-Rich Neuroepithelial Tumor. J Neuropathol Exp Neurol 2019; 78:1011-1021. [DOI: 10.1093/jnen/nlz086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract
MAPK pathway activation has been recurrently observed in desmoplastic infantile ganglioglioma/astrocytoma (DIG/DIA) with reported disproportionally low mutation allele frequencies relative to the apparent high tumor content, suggesting that MAPK pathway alterations may be subclonal. We sought to expand the number of molecularly profiled cases and investigate if tumor cell composition could account for the observed low mutation allele frequencies. Molecular (targeted neuro-oncology next-generation sequencing/RNA sequencing and OncoScan microarray) and immunohistochemical (CD68-PGM1/CD163/CD14/CD11c/lysozyme/CD3/CD20/CD34/PD-L1) studies were performed in 7 DIG. Activating MAPK pathway alterations were identified in 4 (57%) cases: 3 had a BRAF mutation (V600E/V600D/V600_W604delinsDQTDG, at 8%–27% variant allele frequency) and 1 showed a TPM3-NTRK1 fusion. Copy number changes were infrequent and nonrecurrent. All tumors had at least 30% of cells morphologically and immunophenotypically consistent with microglial/macrophage lineage. Two subtotally resected tumors regrew; 1 was re-excised and received adjuvant treatment (chemotherapy/targeted therapy), with clinical response to targeted therapy only. Even with residual tumor, all patients are alive (median follow-up, 83 months; 19–139). This study further supports DIG as another MAPK pathway-driven neuroepithelial tumor, thus expanding potential treatment options for tumors not amenable to surgical cure, and suggests that DIG is a microglia/macrophage-rich neuroepithelial tumor with frequent low driver mutation allele frequencies.
Collapse
Affiliation(s)
- Melissa M Blessing
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Patrick R Blackburn
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Chandra Krishnan
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Virginia L Harrod
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Emily G Barr Fritcher
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Christopher D Zysk
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Rory A Jackson
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Dragana Milosevic
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Asha A Nair
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Jaime I Davila
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Jessica R Balcom
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Robert B Jenkins
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Kevin C Halling
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Benjamin R Kipp
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Amulya A Nageswara Rao
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Nadia N Laack
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - David J Daniels
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - William R Macon
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| | - Cristiane M Ida
- Departments of Laboratory Medicine and Pathology, Health Sciences Research, Pediatrics, Radiation Oncology, and Neurologic Surgery, Mayo Clinic, Rochester, Minnesota; and Departments of Pathology and Neuro-Oncology, Dell Children’s Medical Center, Austin, Texas
| |
Collapse
|
5
|
Matsko DE, Zrelov AA, Ulitin AY, Matsko MV, Skliar SS, Baksheeva AO, Imyanitov EN. [Gemistocytic astrocytomas]. Arkh Patol 2019; 80:27-38. [PMID: 30059069 DOI: 10.17116/patol201880427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Gemistocytic astrocytomas (GA) are a variant of diffuse astrocytomas GII (WHO, 2016). Like all diffuse astrocytomas, GA recur with time, which is often accompanied by malignant degeneretion into the anaplastic astrocytoma GIII or to the secondary glioblastoma GIV. However, the progression-free survival and overall survival in patients with GA is less than in patients with diffuse astrocytomas. Given that this group of patients, according to the WHO classification (2016), is classified as GII, patients with GA usually do not receive comprehensive treatment. We have conducted a thorough analysis of research on this problem for the period from 1956 to 2017. Differences in the histological pattern, immunohistochemical and molecular-genetic profiles, survival of patients with GA and diffuse astrocytomas GII are shown there. A clinical case of a patient with transformation of a diffuse astrocytoma in GA (GIII) and then into a secondary glioblastoma is presented.
Collapse
Affiliation(s)
- D E Matsko
- Polenov's neurosurgery institute - the branch of V.A. Almazov National Medical Research Centre, Saint-Petersburg, Russia; Saint-Petersburg Clinical Research Center Specialized Types of Medical Care, Saint-Petersburg, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia; Saint-Petersburg Medico-Social Institute, Saint-Petersburg, Russia
| | - A A Zrelov
- Polenov's neurosurgery institute - the branch of V.A. Almazov National Medical Research Centre, Saint-Petersburg, Russia; North-Western State Medical University n. a. I.I. Mechnikov, Saint-Petersburg, Russia
| | - A Yu Ulitin
- Polenov's neurosurgery institute - the branch of V.A. Almazov National Medical Research Centre, Saint-Petersburg, Russia; North-Western State Medical University n. a. I.I. Mechnikov, Saint-Petersburg, Russia
| | - M V Matsko
- Polenov's neurosurgery institute - the branch of V.A. Almazov National Medical Research Centre, Saint-Petersburg, Russia; Saint-Petersburg Clinical Research Center Specialized Types of Medical Care, Saint-Petersburg, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia; Saint-Petersburg Medico-Social Institute, Saint-Petersburg, Russia
| | - S S Skliar
- Polenov's neurosurgery institute - the branch of V.A. Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - A O Baksheeva
- Polenov's neurosurgery institute - the branch of V.A. Almazov National Medical Research Centre, Saint-Petersburg, Russia; North-Western State Medical University n. a. I.I. Mechnikov, Saint-Petersburg, Russia
| | - E N Imyanitov
- Saint-Petersburg Clinical Research Center Specialized Types of Medical Care, Saint-Petersburg, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia; North-Western State Medical University n. a. I.I. Mechnikov, Saint-Petersburg, Russia; N.N. Petrov National Medical Research Centre of Oncology, Saint-Petersburg, Russia
| |
Collapse
|
6
|
Ayanlaja AA, Zhang B, Ji G, Gao Y, Wang J, Kanwore K, Gao D. The reversible effects of glial cell line-derived neurotrophic factor (GDNF) in the human brain. Semin Cancer Biol 2018; 53:212-222. [PMID: 30059726 DOI: 10.1016/j.semcancer.2018.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent survival factor, and a member of the transforming growth factor β (TGF-β) superfamily acting on different neuronal activities. GDNF was originally identified as a neurotrophic factor crucially involved in the survival of dopaminergic neurons of the nigrostriatal pathway and is currently an established therapeutic target in Parkinson's disease. However, GDNF was later reported to be highly expressed in gliomas, especially in glioblastomas, and was demonstrated as a potent proliferation factor involved in the development and migration of gliomas. Here, we review our current understanding and progress made so far by researchers in our laboratories with references to relevant articles to support our discoveries. We present past and recent discoveries on the mechanisms involved in the protection of neurons by GDNF and examine its emerging roles in gliomas, as well as reasons for the abnormal expression in Glioblastoma Multiforme (GBM). Collectively, our work establishes a paradigm by which the ability of GDNF to protect dopaminergic neurons from degradation and its corresponding effects on glioma cells points to an underlying biological vulnerability in the effects of GDNF in the normal brain which can be subverted for use by cancer cells. Hence, presenting novel opportunities for intervention in glioma therapies.
Collapse
Affiliation(s)
- Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Baole Zhang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - GuangQuan Ji
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yue Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jie Wang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - DianShuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
7
|
Boussiotis VA, Charest A. Immunotherapies for malignant glioma. Oncogene 2018; 37:1121-1141. [PMID: 29242608 PMCID: PMC5828703 DOI: 10.1038/s41388-017-0024-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/31/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant primary brain cancer with a dreadful overall survival and for which treatment options are limited. Recent breakthroughs in novel immune-related treatment strategies for cancer have spurred interests in usurping the power of the patient's immune system to recognize and eliminate GBM. Here, we discuss the unique properties of GBM's tumor microenvironment, the effects of GBM standard on care therapy on tumor-associated immune cells, and review several approaches aimed at therapeutically targeting the immune system for GBM treatment. We believe that a comprehensive understanding of the intricate micro-environmental landscape of GBM will abound into the development of novel immunotherapy strategies for GBM patients.
Collapse
Affiliation(s)
- Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Alain Charest
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA.
- Division of Genetics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Sørensen MD, Dahlrot RH, Boldt HB, Hansen S, Kristensen BW. Tumour-associated microglia/macrophages predict poor prognosis in high-grade gliomas and correlate with an aggressive tumour subtype. Neuropathol Appl Neurobiol 2017; 44:185-206. [PMID: 28767130 DOI: 10.1111/nan.12428] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 07/27/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023]
Abstract
AIMS Glioblastomas are highly aggressive and treatment resistant. Increasing evidence suggests that tumour-associated macrophages/microglia (TAMs) facilitate tumour progression by acquiring a M2-like phenotype. Our objective was to investigate the prognostic value of TAMs in gliomas using automated quantitative double immunofluorescence. METHODS Samples from 240 patients with primary glioma were stained with antibodies against ionized calcium-binding adaptor molecule-1 (IBA-1) and cluster of differentiation 204 (CD204) to detect TAMs and M2-like TAMs. The expression levels were quantified by software-based classifiers. The associations between TAMs, gemistocytic cells and glioblastoma subtype were examined with immuno- and haematoxylin-eosin stainings. Three tissue arrays containing glioblastoma specimens were included to study IBA-1/CD204 levels in central tumour and tumour periphery and to characterize CD204+ cells. RESULTS Our data revealed that the amount of especially CD204+ TAMs increases with malignancy grade. In grade III-IV, high CD204 expression was associated with shorter survival, while high IBA-1 intensity correlated with a longer survival. In grade IV, CD204 showed independent prognostic value when adjusting for clinical data and the methylation status of O6-methylguanine-DNA methyltransferase. Our findings were confirmed in two bioinformatics databases. TAMs were more abundant in central tumour tissue, mesenchymal glioblastomas and gliomas with many gemistocytic cells. CD204+ TAMs co-expressed proteins related to tumour aggressiveness including matrix metallopeptidase-14 and hypoxia-inducible factor-1α. CONCLUSIONS This is the first study to use automated quantitative immunofluorescence to determine the prognostic impact of TAMs. Our results suggest that M2-like TAMs hold an unfavourable prognostic value in high-grade gliomas and may contribute to a pro-tumourigenic microenvironment.
Collapse
Affiliation(s)
- M D Sørensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - R H Dahlrot
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - H B Boldt
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - S Hansen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - B W Kristensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
9
|
Immune microenvironment of gliomas. J Transl Med 2017; 97:498-518. [PMID: 28287634 DOI: 10.1038/labinvest.2017.19] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/13/2022] Open
Abstract
High-grade gliomas are rapidly progressing tumors of the central nervous system (CNS) with a very poor prognosis despite extensive resection combined with radiation and/or chemotherapy. Histopathological and flow cytometry analyses of human and rodent experimental gliomas revealed heterogeneity of a tumor and its niche, composed of reactive astrocytes, endothelial cells, and numerous immune cells. Infiltrating immune cells consist of CNS resident (microglia) and peripheral macrophages, granulocytes, myeloid-derived suppressor cells (MDSCs), and T lymphocytes. Intratumoral density of glioma-associated microglia/macrophages (GAMs) and MDSCs is the highest in malignant gliomas and inversely correlates with patient survival. Although GAMs have a few innate immune functions intact, their ability to be stimulated via TLRs, secrete cytokines, and upregulate co-stimulatory molecules is not sufficient to initiate antitumor immune responses. Moreover, tumor-reprogrammed GAMs release immunosuppressive cytokines and chemokines shaping antitumor responses. Both GAMs and MDSCs have ability to attract T regulatory lymphocytes to the tumor, but MDSCs inhibit cytotoxic responses mediated by natural killer cells, and block the activation of tumor-reactive CD4+ T helper cells and cytotoxic CD8+ T cells. The presence of regulatory T cells may further contribute to the lack of effective immune activation against malignant gliomas. We review the immunological aspects of glioma microenvironment, in particular composition and various roles of the immune cells infiltrating malignant human gliomas and experimental rodent gliomas. We describe tumor-derived signals and mechanisms driving myeloid cell accumulation and reprogramming. Although, understanding the complexity of cell-cell interactions in glioma microenvironment is far from being achieved, recent studies demonstrated several glioma-derived factors that trigger migration, accumulation, and reprogramming of immune cells. Identification of these factors may facilitate development of immunotherapy for gliomas as immunomodulatory and immune evasion mechanisms employed by malignant gliomas pose an appalling challenge to brain tumor immunotherapy.
Collapse
|
10
|
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of early myeloid progenitors and precursors at different stages of differentiation into granulocytes, macrophages, and dendritic cells. Blockade of their differentiation into mature myeloid cells in cancer results in an expansion of this population. High-grade gliomas are the most common malignant tumours of the central nervous system (CNS), with a poor prognosis despite intensive radiation and chemotherapy. Histopathological and flow cytometry analyses of human and rodent experimental gliomas revealed the extensive heterogeneity of immune cells infiltrating gliomas and their microenvironment. Immune cell infiltrates consist of: resident (microglia) and peripheral macrophages, granulocytes, myeloid-derived suppressor cells, and T lymphocytes. Intratumoural density of glioma-associated MDSCs correlates positively with the histological grade of gliomas and patient’s survival. MDSCs have the ability to attract T regulatory lymphocytes to the tumour, but block the activation of tumour-reactive CD4+ T helper cells and cytotoxic CD8+ T cells. Immunomodulatory mechanisms employed by malignant gliomas pose an appalling challenge to brain tumour immunotherapy. In this mini-review we describe phenotypic and functional characteristics of MDSCs in humans and rodents, and their occurrence and potential roles in glioma progression. While understanding the complexity of immune cell interactions in the glioma microenvironment is far from being accomplished, there is significant progress that may lead to the development of immunotherapy for gliomas.
Collapse
|
11
|
Petrosky KY, Knight HL, Westmoreland SV, Miller AD. Atypical nodular astrocytosis in simian immunodeficiency virus-infected rhesus macaques (Macaca mulatta). J Med Primatol 2014; 43:468-76. [PMID: 25082291 DOI: 10.1111/jmp.12138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Simian immunodeficiency virus (SIV), a model for HIV pathogenesis, is associated with neuropathology. METHODS Five SIV-infected animals were selected following a database search of 1206 SIV-infected animals for nodular or astrocytic lesions. Two of five had neurologic dysfunction, and 3 of 5 were incidental findings. RESULTS Histologic examination revealed multifocal nodular foci in the gray and white matter formed by interlacing astrocytes with abundant cytoplasm and large, reactive nuclei. Nodules were often enmeshed with small capillaries. Immunohistochemistry revealed variable immunoreactivity for a panel of markers: GFAP (4/5), vimentin (5/5), Glut-1 (1/5), CNPase (0/5), S100 (5/5), Iba1 (0/5), Ki67 (0/5), and p53 (4/4). In situ hybridization failed to detect any SIV RNA (0/5). Immunohistochemistry for simian virus 40, rhesus cytomegalovirus, and rhesus lymphocryptovirus failed to detect any antigen within the lesions. CONCLUSION The immunoreactivity of p53 in the lesions compared with adjacent tissue suggests a local derangement in astrocyte proliferation and function.
Collapse
Affiliation(s)
- Keiko Y Petrosky
- Division of Comparative Pathology, New England Primate Research Center, Harvard Medical School, Southborough, MA, USA
| | | | | | | |
Collapse
|
12
|
Clinical outcome of surgically treated low-grade gliomas: A retrospective analysis of a single institute. Clin Neurol Neurosurg 2013; 115:2508-13. [DOI: 10.1016/j.clineuro.2013.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/26/2013] [Accepted: 10/13/2013] [Indexed: 11/22/2022]
|
13
|
Babu R, Bagley JH, Park JG, Friedman AH, Adamson C. Low-grade astrocytomas: the prognostic value of fibrillary, gemistocytic, and protoplasmic tumor histology. J Neurosurg 2013; 119:434-41. [PMID: 23662821 DOI: 10.3171/2013.4.jns122329] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Low-grade astrocytomas are slow-growing, infiltrative gliomas that over time may progress into more malignant tumors. Various factors have been shown to affect the time to progression and overall survival including age, performance status, tumor size, and the extent of resection. However, more recently it has been suggested that histological subtypes (fibrillary, protoplasmic, and gemistocytic) may impact patient outcome. In this study the authors have performed a large comparative population-based analysis to examine the characteristics and survival of patients with the various subtypes of WHO Grade II astrocytomas. METHODS Patients diagnosed with fibrillary, protoplasmic, and gemistocytic astrocytomas were identified through the Surveillance, Epidemiology, and End Results (SEER) database. The chi-square test and Student t-test were used to evaluate differences in patient and treatment characteristics between astrocytoma subtypes. Kaplan-Meier analysis was used to assess overall survival, and the log-rank test was used to evaluate the differences between survival curves. Univariate and multivariate analyses were also performed to determine the effect of various patient, tumor, and treatment variables on overall survival. RESULTS A total of 500 cases were included in the analysis, consisting of 326 fibrillary (65.2%), 29 protoplasmic (5.8%), and 145 gemistocytic (29%) variants. Gemistocytic astrocytomas presented at a significantly older age than the fibrillary variant (46.8 vs 37.7 years, p < 0.0001), with protoplasmic and fibrillary subtypes having a similar age. Although protoplasmic and fibrillary variants underwent radiotherapy at similar rates, gemistocytic tumors more frequently received radiotherapy (p = 0.0001). Univariate analysis revealed older age, larger tumor size, and the use of radiotherapy to be poor prognostic factors, with resection being associated with improved survival. The gemistocytic subtype (hazard ratio [HR] 1.62 [95% CI 1.27-2.07], p = 0.0001) also resulted in significantly worse survival than fibrillary tumors. Bivariate analyses demonstrated that older age, the use of radiotherapy, and resection significantly influenced median survival. Tumor subtype also affected median survival; patients who harbored gemistocytic tumors experienced less than half the median survival of fibrillary and protoplasmic tumors (38 vs 82 months, p = 0.0003). Multivariate analysis revealed increasing age (HR 1.05 [95% CI 1.04-1.05], p < 0.0001), larger tumor size (HR 1.02 [95% CI 1.01-1.03], p = 0.0002), and the use of resection (HR 0.70 [95% CI 0.52-0.94], p = 0.018) to be independent predictors of survival. Examination of tumor subtype revealed that the gemistocytic variant (HR 1.30 [95% CI 0.98-1.74], p = 0.074) was associated with worse patient survival than fibrillary tumors, although this only approached significance. The protoplasmic subtype did not affect overall survival (p = 0.33). CONCLUSIONS Gemistocytic tumor histology was associated with worse survival than fibrillary and protoplasmic astrocytomas. As protoplasmic astrocytomas have a survival similar to fibrillary tumors, there may be limited utility to the identification of this rare variant. However, increased attention should be paid to the presence of gemistocytes in low-grade gliomas as this is associated with shorter time to progression, increased malignant transformation, and reduced overall survival.
Collapse
Affiliation(s)
- Ranjith Babu
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | |
Collapse
|
14
|
Ku MC, Wolf SA, Respondek D, Matyash V, Pohlmann A, Waiczies S, Waiczies H, Niendorf T, Synowitz M, Glass R, Kettenmann H. GDNF mediates glioblastoma-induced microglia attraction but not astrogliosis. Acta Neuropathol 2013; 125:609-20. [PMID: 23344256 DOI: 10.1007/s00401-013-1079-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/09/2013] [Indexed: 12/14/2022]
Abstract
High-grade gliomas are the most common primary brain tumors. Their malignancy is promoted by the complex crosstalk between different cell types in the central nervous system. Microglia/brain macrophages infiltrate high-grade gliomas and contribute to their progression. To identify factors that mediate the attraction of microglia/macrophages to malignant brain tumors, we established a glioma cell encapsulation model that was applied in vivo. Mouse GL261 glioma cell line and human high-grade glioma cells were seeded into hollow fibers (HF) that allow the passage of soluble molecules but not cells. The glioma cell containing HF were implanted into one brain hemisphere and simultaneously HF with non-transformed fibroblasts (controls) were introduced into the contralateral hemisphere. Implanted mouse and human glioma- but not fibroblast-containing HF attracted microglia and up-regulated immunoreactivity for GFAP, which is a marker of astrogliosis. In this study, we identified GDNF as an important factor for microglial attraction: (1) GL261 and human glioma cells secret GDNF, (2) reduced GDNF production by siRNA in GL261 in mouse glioma cells diminished attraction of microglia, (3) over-expression of GDNF in fibroblasts promoted microglia attraction in our HF assay. In vitro migration assays also showed that GDNF is a strong chemoattractant for microglia. While GDNF release from human or mouse glioma had a profound effect on microglial attraction, the glioma-induced astrogliosis was not affected. Finally, we could show that injection of GL261 mouse glioma cells with GDNF knockdown by shRNA into mouse brains resulted in reduced tumor expansion and improved survival as compared to injection of control cells.
Collapse
Affiliation(s)
- Min-Chi Ku
- Department of Cellular Neuroscience, Max Delbrück Center for Molecular Medicine (MDC), Robert Rössle Str. 10, 13125 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Early prognostic factors related to progression and malignant transformation of low-grade gliomas. Clin Neurol Neurosurg 2011; 113:752-7. [DOI: 10.1016/j.clineuro.2011.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 07/10/2011] [Accepted: 08/05/2011] [Indexed: 11/17/2022]
|
16
|
Kateb B, Yamamoto V, Alizadeh D, Zhang L, Manohara HM, Bronikowski MJ, Badie B. Multi-walled carbon nanotube (MWCNT) synthesis, preparation, labeling, and functionalization. Methods Mol Biol 2010; 651:307-317. [PMID: 20686974 DOI: 10.1007/978-1-60761-786-0_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Nanomedicine is a growing field with a great potential for introducing new generation of targeted and personalized drug. Amongst new generation of nano-vectors are carbon nanotubes (CNTs), which can be produced as single or multi-walled. Multi-walled carbon nanotubes (MWCNTs) can be fabricated as biocompatible nanostructures (cylindrical bulky tubes). These structures are currently under investigation for their application in nanomedicine as viable and safe nanovectors for gene and drug delivery. In this chapter, we will provide you with the necessary information to understand the synthesis of MWCNTs, functionalization, PKH26 labeling, RNAi, and DNA loading for in vitro experimentation and in vivo implantation of labeled MWCNT in mice as well as materials used in this experimentation. We used this technique to manipulate microglia as part of a novel application for the brain cancer immunotherapy. Our published data show this is a promising technique for labeling, and gene and drug delivery into microglia.
Collapse
Affiliation(s)
- Babak Kateb
- Brain Mapping Foundation and International Brain Mapping & Intraoperative Surgical Planning Society (IBMISPS), West Hollywood, CA, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Graeber MB, Streit WJ. Microglia: biology and pathology. Acta Neuropathol 2010; 119:89-105. [PMID: 20012873 DOI: 10.1007/s00401-009-0622-0] [Citation(s) in RCA: 511] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 12/02/2009] [Indexed: 12/15/2022]
Abstract
The past 20 years have seen a gain in knowledge on microglia biology and microglia functions in disease that exceeds the expectations formulated when the microglia "immune network" was introduced. More than 10,000 articles have been published during this time. Important new research avenues of clinical importance have opened up such as the role of microglia in pain and in brain tumors. New controversies have also emerged such as the question of whether microglia are active or reactive players in neurodegenerative disease conditions, or whether they may be victims themselves. Premature commercial interests may be responsible for some of the confusion that currently surrounds microglia in both the Alzheimer and Parkinson's disease research fields. A critical review of the literature shows that the concept of "(micro)glial inflammation" is still open to interpretation, despite a prevailing slant towards a negative meaning. Perhaps the most exciting foreseeable development concerns research on the role of microglia in synaptic plasticity, which is expected to yield an answer to the question whether microglia are the brain's electricians. This review provides an analysis of the latest developments in the microglia field.
Collapse
Affiliation(s)
- Manuel B Graeber
- Division of Neuropathology, Department of Pathology and Clinical Laboratory Medicine, Faculty of Medicine, Neurosciences Center, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia.
| | | |
Collapse
|
18
|
Davenport CM, Sevastou IG, Hooper C, Pocock JM. Inhibiting p53 pathways in microglia attenuates microglial-evoked neurotoxicity following exposure to Alzheimer peptides. J Neurochem 2009; 112:552-63. [PMID: 19895660 DOI: 10.1111/j.1471-4159.2009.06485.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microglial activation can lead to microglial apoptosis, which may serve to remove highly reactive and possibly neurotoxic microglia. However the loss of microglia may have consequences for future recovery, protection and repair. P53, a nuclear phosphoprotein transcription factor, is critical for activating the expression of genes involved in cell-cycle arrest and stress-induced apoptosis. In neurodegenerative diseases the expression of p53 is significantly increased in glial cells, and microglial numbers fall. Following activation with chromogranin A (100 nM), or beta-amyloid(25-35), (10 microM), microglia became apoptotic. Furthermore, p53 expression was enhanced, peaking at 4-6 h after exposure to activators. The p53 transcription inhibitor, pifithrin-alpha, (10 microM) significantly reduced the expression of p53 in microglia and significantly modulated the levels of microglial apoptosis induced by activation. Lithium chloride (5 mM), which can modulate p53-mediated pathways, also reduced p53 expression and reduced microglial apoptosis suggesting glycogen synthase kinase-3 plays a role. Regulating p53 pathways modulated microglial inducible nitric oxide synthase expression and tumour necrosis factor alpha secretion. Inhibiting p53 mediated microglial apoptosis prevented microglial neurotoxicity suggesting targeting of p53-mediated pathways in microglia may have therapeutic benefit in Alzheimer's disease.
Collapse
Affiliation(s)
- Christopher M Davenport
- Cell Signalling Laboratory, Department of Neuroinflammation, UCL Institute of Neurology, London, UK
| | | | | | | |
Collapse
|
19
|
McGirt MJ, Chaichana KL, Attenello FJ, Weingart JD, Than K, Burger PC, Olivi A, Brem H, Quinoñes-Hinojosa A. EXTENT OF SURGICAL RESECTION IS INDEPENDENTLY ASSOCIATED WITH SURVIVAL IN PATIENTS WITH HEMISPHERIC INFILTRATING LOW-GRADE GLIOMAS. Neurosurgery 2008; 63:700-7; author reply 707-8. [PMID: 18981880 DOI: 10.1227/01.neu.0000325729.41085.73] [Citation(s) in RCA: 377] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
ABSTRACT
OBJECTIVE
It remains unknown whether the extent of surgical resection affects survival or disease progression in patients with supratentorial low-grade gliomas.
METHODS
We conducted a retrospective cohort study (n = 170) between 1996 and 2007 at a single institution to determine whether increasing extent of surgical resection was associated with improved progression-free survival (PFS) and overall survival (OS). Surgical resection of gliomas defined as gross total resection (GTR) (complete resection of the preoperative fluid-attenuated inversion recovery signal abnormality), near total resection (NTR) (<3-mm thin residual fluid-attenuated inversion recovery signal abnormality around the rim of the resection cavity only), or subtotal resection (STR) (residual nodular fluid-attenuated inversion recovery signal abnormality) based on magnetic resonance imaging performed less than 48 hours after surgery. Our main outcome measures were OS, PFS, and malignant degeneration-free survival (conversion to high-grade glioma).
RESULTS
One hundred thirty-two primary and 38 revision resections were performed for low-grade astrocytomas (n = 93) or oligodendrogliomas (n = 77). GTR, NTR, and STR were achieved in 65 (38%), 39 (23%), and 66 (39%) cases, respectively. GTR versus STR was independently associated with increased OS (hazard ratio, 0.36; 95% confidence interval, 0.16–0.84; P = 0.017) and PFS (HR, 0.56; 95% confidence interval, 0.32–0.98; P = 0.043) and a trend of increased malignant degeneration-free survival (hazard ratio, 0.46; 95% confidence interval, 0.20–1.03; P = 0.060). NTR versus STR was not independently associated with improved OS, PFS, or malignant degeneration-free survival. Five-year OS after GTR, NTR, and STR was 95, 80, 70%, respectively, and 10-year OS was 76, 57, and 49%, respectively. After GTR, NTR, and STR, median time to tumor progression was 7.0, 4.0, and 3.5 years, respectively. Median time to malignant degeneration after GTR, NTR, and STR was 12.5, 5.8, and 7 years, respectively.
CONCLUSION
GTR was associated with a delay in tumor progression and malignant degeneration as well as improved OS independent of age, degree of disability, histological subtype, or revision versus primary resection. GTR should be safely attempted when not limited by eloquent cortex.
Collapse
Affiliation(s)
- Matthew J. McGirt
- Department of Neurosurgery, The Johns Hopkins School of Medicine, and Neuro-oncology Surgical Outcomes Research Laboratory, Baltimore, Maryland
| | - Kaisorn L. Chaichana
- Department of Neurosurgery, The Johns Hopkins School of Medicine, and Neuro-oncology Surgical Outcomes Research Laboratory, Baltimore, Maryland
| | - Frank J. Attenello
- Department of Neurosurgery, The Johns Hopkins School of Medicine, and Neuro-oncology Surgical Outcomes Research Laboratory, Baltimore, Maryland
| | - Jon D. Weingart
- Departments of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Khoi Than
- Department of Neurosurgery, The Johns Hopkins School of Medicine, and Neuro-oncology Surgical Outcomes Research Laboratory, Baltimore, Maryland
| | - Peter C. Burger
- Departments of Neurosurgery, Oncology, and Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Alessandro Olivi
- Departments of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Henry Brem
- Departments of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Alfredo Quinoñes-Hinojosa
- Departments of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, and Neuro-oncology Surgical Outcomes Research Laboratory, Baltimore, Maryland
| |
Collapse
|