1
|
Bhardwaj JS, Paliwal S, Singhvi G, Taliyan R. Immunological challenges and opportunities in glioblastoma multiforme: A comprehensive view from immune system lens. Life Sci 2024; 357:123089. [PMID: 39362586 DOI: 10.1016/j.lfs.2024.123089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Glioblastoma multiforme (GBM), also known as grade IV astrocytoma, is the most common and deadly brain tumour. It has a poor prognosis and a low survival rate. GBM cells' immunological escape mechanism helps them resist advanced multimodal therapy. In physiological homeostasis, brain astrocytes and microglia suppress infections and clear the potential pathogen from the system. However, in severe pathological conditions like cancer, the immune response fails to eliminate mutated and rapidly over-proliferating GBM cells. The malignant cells' interactions with immune cells and the neoplasm's immunosuppressive environment enable the avoidance and their clearance. Immunotherapy efficiently addresses these difficulties, as shown by sufficient evidence. This review discusses how GBM cells inhibit and elude the immune system. These include MHC molecule expression alteration and PD-L1 and CTLA-4 immune checkpoint overexpression. Without co-stimulation, these changes induce effector T-cell tolerance and anergy. The review also covers how MDSCs, TAMs, Herpes Virus Entry Mediators, and Human cytomegalovirus protein decrease the effector immune response against glioblastoma. The latter part discusses various therapies that are available in the market or under clinical trials which revolves around combating resistance against the available multimodal therapies. The recent trends indicate that there are various monoclonal antibodies and peptide-based vaccines that can be utilized to overcome the immune evasion technique harbored by GBM cells. A strategic development of Immunotherapy considering these hallmarks of immune evasion may help in designing a therapy that may prove to be effective in killing the GBM cells thereby, improving the overall survival of GBM-affected patients.
Collapse
Affiliation(s)
- Jayant Singh Bhardwaj
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Shivangi Paliwal
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India.
| |
Collapse
|
2
|
Zhang H, Grippin A, Sun M, Ma Y, Kim BYS, Teng L, Jiang W, Yang Z. New avenues for cancer immunotherapy: Cell-mediated drug delivery systems. J Control Release 2024; 375:712-732. [PMID: 39326499 DOI: 10.1016/j.jconrel.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Cancer research has become increasingly complex over the past few decades as knowledge of the heterogeneity of cancer cells, their proliferative ability, and their tumor microenvironments has become available. Although conventional therapies remain the most compelling option for cancer treatment to date, immunotherapy is a promising way to harness natural immune defenses to target and kill cancer cells. Cell-mediated drug delivery systems (CDDSs) have been an active line of research for enhancing the therapeutic efficacy and specificity of cancer immunotherapy. These systems can be tailored to different types of immune cells, allowing immune evasion and accumulation in the tumor microenvironment. By enabling the targeted delivery of therapeutic agents such as immune stimulants, cytokines, antibodies, and antigens, CDDSs have improved the survival of some patients with cancer. This review summarizes the research status of CDDSs, with a focus on their underlying mechanisms of action, biology, and clinical applications. We also discuss opportunities and challenges for implementation of CDDSs into mainstream cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Adam Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
3
|
Mercado NB, Real JN, Kaiserman J, Panagioti E, Cook CH, Lawler SE. Clinical implications of cytomegalovirus in glioblastoma progression and therapy. NPJ Precis Oncol 2024; 8:213. [PMID: 39343770 PMCID: PMC11439950 DOI: 10.1038/s41698-024-00709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Glioblastoma (GBM) is one of the deadliest brain cancers with a median survival of only 15 months. This poor prognosis has prompted exploration of novel therapeutic targets for GBM patients. Human cytomegalovirus (HCMV) has been implicated in GBM; however, its impact remains poorly defined, and there is conflicting data over the presence of HCMV in tumors. Nonetheless, clinical trials targeting HCMV have shown promising initial data, and evidence suggests that HCMV may negatively impact GBM patient survival by multiple mechanisms including changes in GBM cell behavior and the tumor microenvironment (TME) that potentiate tumor progression as well as therapy-induced virus reactivation. Moreover, HCMV has many effects on host immunity that could impact tumor behavior by altering the TME, which are largely unexplored. The goal of this review is to describe these potential interactions between HCMV and GBM. Better understanding of these processes may allow the development of new therapeutic modalities to improve GBM patient outcomes.
Collapse
Affiliation(s)
- Noe B Mercado
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US
- The Warren Alpert Medical School, Brown University, Providence, RI, US
| | - Jacqueline N Real
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US
- The Warren Alpert Medical School, Brown University, Providence, RI, US
| | - Jacob Kaiserman
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US
- The Warren Alpert Medical School, Brown University, Providence, RI, US
| | - Eleni Panagioti
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Charles H Cook
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Sean E Lawler
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, US.
- The Warren Alpert Medical School, Brown University, Providence, RI, US.
| |
Collapse
|
4
|
Hawly J, Murcar MG, Schcolnik-Cabrera A, Issa ME. Glioblastoma stem cell metabolism and immunity. Cancer Metastasis Rev 2024; 43:1015-1035. [PMID: 38530545 DOI: 10.1007/s10555-024-10183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Despite enormous efforts being invested in the development of novel therapies for brain malignancies, there remains a dire need for effective treatments, particularly for pediatric glioblastomas. Their poor prognosis has been attributed to the fact that conventional therapies target tumoral cells, but not glioblastoma stem cells (GSCs). GSCs are characterized by self-renewal, tumorigenicity, poor differentiation, and resistance to therapy. These characteristics represent the fundamental tools needed to recapitulate the tumor and result in a relapse. The mechanisms by which GSCs alter metabolic cues and escape elimination by immune cells are discussed in this article, along with potential strategies to harness effector immune cells against GSCs. As cellular immunotherapy is making significant advances in a variety of cancers, leveraging this underexplored reservoir may result in significant improvements in the treatment options for brain malignancies.
Collapse
Affiliation(s)
- Joseph Hawly
- Faculty of Medicine and Medical Sciences, University of Balamand, Dekouaneh, Lebanon
| | - Micaela G Murcar
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Mark E Issa
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
5
|
An W, Ren C, Yuan L, Qiu Z, Wang P, Cheng Y, He Z, Han X, Li S, An Y. High expression of SIGLEC7 may promote M2-type macrophage polarization leading to adverse prognosis in glioma patients. Front Immunol 2024; 15:1411072. [PMID: 39211050 PMCID: PMC11357930 DOI: 10.3389/fimmu.2024.1411072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Gliomas are the most common primary intracranial tumors, known for their high invasiveness and destructiveness. Sialic acid-binding immunoglobulin-like lectin 7 (SIGLEC7) is present in various immune cells, especially macrophages, and significantly affects immune homeostasis and cancer cell response. However, research on the role and prognostic impact of SIGLEC7 in glioma patients is currently limited. Methods We utilized transcriptomic data from 702 glioma patients in The Cancer Genome Atlas (TCGA) and 693 glioma patients in the Chinese Glioma Genome Atlas (CGGA), along with clinical samples we collected, to comprehensively investigate the impact of SIGLEC7 on glioma expression patterns, biological functions, and prognostic value. We focused on its role in glioma-related immune responses and immune cell infiltration and analyzed its expression at the single-cell level. Finally, we validated the role of SIGLEC7 in gliomas through tissue and cell experiments. Results SIGLEC7 expression was significantly increased in glioma patients with malignant characteristics. Survival analysis indicated that glioma patients with high SIGLEC7 expression had significantly lower survival rates. Gene function analysis revealed that SIGLEC7 is primarily involved in immune and inflammatory responses and is strongly negatively correlated with tumor-associated immune regulation. Additionally, the expression of most immune checkpoints was positively correlated with SIGLEC7, and immune cell infiltration analysis clearly demonstrated a significant positive correlation between SIGLEC7 expression and M2 macrophage infiltration levels. Single-cell analysis, along with tissue and cell experiments, confirmed that SIGLEC7 enhances macrophage polarization towards the M2 phenotype, thereby promoting glioma invasiveness through the immunosuppressive effects of M2 macrophages. Cox regression analysis and the establishment of survival prediction models indicated that high SIGLEC7 expression is an unfavorable prognostic factor for glioma patients. Discussion High SIGLEC7 expression predicts poor prognosis in glioma patients and is closely associated with M2 macrophages in the tumor environment. In the future, SIGLEC7 may become a promising target for glioma immunotherapy.
Collapse
Affiliation(s)
- Wenhao An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Changyuan Ren
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Lei Yuan
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Zhiqiang Qiu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, United States
| | - Peishen Wang
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Yanwen Cheng
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Zi He
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Xinye Han
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yihua An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Joshi DC, Sharma A, Prasad S, Singh K, Kumar M, Sherawat K, Tuli HS, Gupta M. Novel therapeutic agents in clinical trials: emerging approaches in cancer therapy. Discov Oncol 2024; 15:342. [PMID: 39127974 PMCID: PMC11317456 DOI: 10.1007/s12672-024-01195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host's immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, "precision medicine" customizes therapies to each patient's unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Anurag Sharma
- Invertis Institute of Pharmacy, Invertis University Bareilly Uttar Pradesh, Bareilly, India
| | - Sonima Prasad
- Chandigarh University, Ludhiana-Chandigarh State Highway, Gharuan, Mohali, Punjab, 140413, India
| | - Karishma Singh
- Institute of Pharmaceutical Sciences, Faculty of Engineering and Technology, University of Lucknow, Lucknow, India
| | - Mayank Kumar
- Himalayan Institute of Pharmacy, Road, Near Suketi Fossil Park, Kala Amb, Hamidpur, Himachal Pradesh, India
| | - Kajal Sherawat
- Meerut Institute of Technology, Meerut, Uttar Pradesh, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India.
| |
Collapse
|
7
|
Stergiopoulos GM, Concilio SC, Galanis E. An Update on the Clinical Status, Challenges, and Future Directions of Oncolytic Virotherapy for Malignant Gliomas. Curr Treat Options Oncol 2024; 25:952-991. [PMID: 38896326 DOI: 10.1007/s11864-024-01211-6] [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] [Accepted: 04/24/2024] [Indexed: 06/21/2024]
Abstract
OPINION STATEMENT Malignant gliomas are common central nervous system tumors that pose a significant clinical challenge due to the lack of effective treatments. Glioblastoma (GBM), a grade 4 malignant glioma, is the most prevalent primary malignant brain tumor and is associated with poor prognosis. Current clinical trials are exploring various strategies to combat GBM, with oncolytic viruses (OVs) appearing particularly promising. In addition to ongoing and recently completed clinical trials, one OV (Teserpaturev, Delytact®) received provisional approval for GBM treatment in Japan. OVs are designed to selectively target and eliminate cancer cells while promoting changes in the tumor microenvironment that can trigger and support long-lasting anti-tumor immunity. OVs offer the potential to remodel the tumor microenvironment and reverse systemic immune exhaustion. Additionally, an increasing number of OVs are armed with immunomodulatory payloads or combined with immunotherapy approaches in an effort to promote anti-tumor responses in a tumor-targeted manner. Recently completed oncolytic virotherapy trials can guide the way for future treatment individualization through patient preselection, enhancing the likelihood of achieving the highest possible clinical success. These trials also offer valuable insight into the numerous challenges inherent in malignant glioma treatment, some of which OVs can help overcome.
Collapse
Affiliation(s)
| | | | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Guyon J, Haidar Ahmad S, El Baba R, Le Quang M, Bikfalvi A, Daubon T, Herbein G. Generation of glioblastoma in mice engrafted with human cytomegalovirus-infected astrocytes. Cancer Gene Ther 2024; 31:1070-1080. [PMID: 38553638 PMCID: PMC11257955 DOI: 10.1038/s41417-024-00767-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 07/20/2024]
Abstract
Mounting evidence is identifying human cytomegalovirus (HCMV) as a potential oncogenic virus. HCMV has been detected in glioblastoma multiforme (GB). Herewith, we present the first experimental evidence for the generation of CMV-Elicited Glioblastoma Cells (CEGBCs) possessing glioblastoma-like traits that lead to the formation of glioblastoma in orthotopically xenografted mice. In addition to the already reported oncogenic HCMV-DB strain, we isolated three HCMV clinical strains from GB tissues that transformed HAs toward CEGBCs and generated spheroids from CEGBCs that resulted in the appearance of glioblastoma-like tumors in xenografted mice. These tumors were nestin-positive mostly in the invasive part surrounded by GFAP-positive reactive astrocytes. The glioblastoma immunohistochemistry phenotype was confirmed by EGFR and cMet gene amplification in the tumor parallel to the detection of HCMV IE and UL69 genes and proteins. Our results fit with an HCMV-induced glioblastoma model of oncogenesis in vivo which will open the door to new therapeutic approaches and assess the anti-HCMV treatment as well as immunotherapy in fighting GB which is characterized by poor prognosis.
Collapse
Affiliation(s)
- Joris Guyon
- University of Bordeaux, INSERM U1312, BRIC, Bordeaux, France
- CHU Bordeaux, Department of Medical Pharmacology, Bordeaux, France
| | - Sandy Haidar Ahmad
- University of Franche-Comté, Pathogens & Inflammation/EPILAB Laboratory, EA 4266, Besançon, France
| | - Ranim El Baba
- University of Franche-Comté, Pathogens & Inflammation/EPILAB Laboratory, EA 4266, Besançon, France
| | - Mégane Le Quang
- Pathology Department, University Hospital of Bordeaux, Bordeaux, France
| | | | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC UMR5095, Bordeaux, France
| | - Georges Herbein
- University of Franche-Comté, Pathogens & Inflammation/EPILAB Laboratory, EA 4266, Besançon, France.
- CHU Besançon, Department of Virology, Besançon, France.
| |
Collapse
|
9
|
Aghajani M, Jalilzadeh N, Aghebati-Maleki A, Yari A, Tabnak P, Mardi A, Saeedi H, Aghebati-Maleki L, Baradaran B. Current approaches in glioblastoma multiforme immunotherapy. Clin Transl Oncol 2024; 26:1584-1612. [PMID: 38512448 DOI: 10.1007/s12094-024-03395-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/08/2024] [Indexed: 03/23/2024]
Abstract
Glioblastoma multiform (GBM) is the most prevalent CNS (central nervous system) tumor in adults, with an average survival length shorter than 2 years and rare metastasis to organs other than CNS. Despite extensive attempts at surgical resecting, the inherently permeable nature of this disease has rendered relapse nearly unavoidable. Thus, immunotherapy is a feasible alternative, as stimulated immune cells can enter into the remote and inaccessible tumor cells. Immunotherapy has revolutionized patient upshots in various malignancies and might introduce different effective ways for GBM patients. Currently, researchers are exploring various immunotherapeutic strategies in patients with GBM to target both the innate and acquired immune responses. These approaches include reprogrammed tumor-associated macrophages, the use of specific antibodies to inhibit tumor progression and metastasis, modifying tumor-associated macrophages with antibodies, vaccines that utilize tumor-specific dendritic cells to activate anti-tumor T cells, immune checkpoint inhibitors, and enhanced T cells that function against tumor cells. Despite these findings, there is still room for improving the response faults of the many currently tested immunotherapies. This study aims to review the currently used immunotherapy approaches with their molecular mechanisms and clinical application in GBM.
Collapse
Affiliation(s)
- Marjan Aghajani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Jalilzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Molecular Medicine Department, Faculty of Modern Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Peyman Tabnak
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Liang T, Zhou X, Wang Y, Ma W. Glioma hexokinase 3 positively correlates with malignancy and macrophage infiltration. Metab Brain Dis 2024; 39:719-729. [PMID: 38687460 DOI: 10.1007/s11011-023-01333-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/01/2023] [Indexed: 05/02/2024]
Abstract
BACKGROUND Glioma is the main subtype of primary central nervous system (CNS) tumor with high malignancy and poor prognosis under current therapeutic approaches. Glycolysis and suppressive tumor microenvironment (TME) are key markers of glioma with great importance for aggressive features of glioma and inferior clinical outcomes. Hexokinase 3 (HK3) is an important rate-limiting enzyme in glycolysis, but its function in glioma remains unknown. METHODS This study comprehensively assessed the expression distribution and immunological effect of HK3 via pan-cancer analysis based on datasets from Genotype Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), and The Cancer Genome Atlas (TCGA). Furthermore, it explored the malignant phenotype and genomic landscape between low-HK3 and high-HK3 expression groups in gliomas from Chinese Glioma Genome Atlas (CGGA) and TCGA. Moreover, data from the TIMER website predicted the relationship between macrophage infiltration and HK3 expression. Also, single-cell sequencing data were used to validate the relationship. RESULTS For pan-cancer patients, HK3 was expressed in various cancers. The results showed that HK3 was highly expressed in gliomas and positively correlated with tumor-infiltrating immune cells (TIICs), immune checkpoints, immunomodulators, and chemokines. Meanwhile, HK3 expression was highest in normal immune cells and tissues. In gliomas, the expression of HK3 was found to be closely correlated with the malignant clinical characteristics and the infiltration of macrophages. Also, HK3 was proven to be positively associated with macrophage through single-cell sequencing data and immunohistochemistry techniques. Finally, it is predicted that samples with high HK3 expression are often malignant entities and also significant genomic aberrations of driver oncogenes. CONCLUSIONS This is the first comprehensive research to figure out the relationship between HK3 and TME characteristics in gliomas. HK3 is positively associated with macrophage infiltration and can induce the immunosuppressive TME and malignant phenotype of gliomas.
Collapse
Affiliation(s)
- Tingyu Liang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingang Zhou
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Lan J, Liu Y, Chen J, Liu H, Feng Y, Liu J, Chen L. Advanced tumor electric fields therapy: A review of innovative research and development and prospect of application in glioblastoma. CNS Neurosci Ther 2024; 30:e14720. [PMID: 38715344 PMCID: PMC11077002 DOI: 10.1111/cns.14720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive malignant tumor with a high mortality rate and is the most prevalent primary intracranial tumor that remains incurable. The current standard treatment, which involves surgery along with concurrent radiotherapy and chemotherapy, only yields a survival time of 14-16 months. However, the introduction of tumor electric fields therapy (TEFT) has provided a glimmer of hope for patients with newly diagnosed and recurrent GBM, as it has been shown to extend the median survival time to 20 months. The combination of TEFT and other advanced therapies is a promising trend in the field of GBM, facilitated by advancements in medical technology. AIMS In this review, we provide a concise overview of the mechanism and efficacy of TEFT. In addition, we mainly discussed the innovation of TEFT and our proposed blueprint for TEFT implementation. CONCLUSION Tumor electric fields therapy is an effective and highly promising treatment modality for GBM. The full therapeutic potential of TEFT can be exploited by combined with other innovative technologies and treatments.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| | - Yuyang Liu
- Medical School of Chinese PLABeijingChina
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Junyi Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalHainanChina
| | - Yaping Feng
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jialin Liu
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Ling Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| |
Collapse
|
12
|
Huang R, Lu X, Sun X, Wu H. Prognostic significance of alterations in fibrinogen level and fibrinogen-to-lymphocyte ratio after radiotherapy on survival outcomes in glioblastoma. Transl Cancer Res 2024; 13:1887-1903. [PMID: 38737673 PMCID: PMC11082672 DOI: 10.21037/tcr-23-2271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/19/2024] [Indexed: 05/14/2024]
Abstract
Background Fibrinogen (FIB) plays an important role in tumor initiation, progression, and metastasis, but its clinical significance in glioblastoma has not been studied. We intend to explore the prognostic value by retrospectively analyzing the changes in FIB and fibrinogen-to-lymphocyte ratio (FLR) in glioblastoma patients before and after radiotherapy, and study the impact of radiotherapy on them. Methods This study retrospectively included 104 patients who were newly diagnosed with glioblastoma between February 2017 and February 2022 and analysed their clinical data from before to after radiotherapy. The cut-off values for FLR and FIB were calculated using a receiver operating characteristic curve. For inter-group comparisons, the Mann-Whitney U or t-test was applied. The prognostic importance of FIB and FLR was evaluated using the Kaplan-Meier curve and the Cox regression model. Spearman correlation coefficients were calculated to evaluate the association of FIB and FLR with radiotherapy-related dose-volume parameters. Results The mean progression-free survival (PFS) and overall survival (OS) of the high FIB and high FLR groups were significantly lower than those of the low FIB and low FLR groups (P<0.05). Larger planning target volume (PTV), mean brain dose, and mean brainstem dose were independent prognostic factors for poor PFS and OS in patients with glioblastoma. Conclusions FLR was a unique and very accurate predictor for the prognosis of glioblastoma, and FIB rise after radiation was a predictive sign of poor survival. Both PTV volume and dose volume for involved organs could significantly affect the FIB and FLR values in patients with glioblastoma.
Collapse
Affiliation(s)
- Rong Huang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoxu Lu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xueming Sun
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Hui Wu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
13
|
Wu JW, Liu Y, Dai XJ, Liu HM, Zheng YC, Liu HM. CD155 as an emerging target in tumor immunotherapy. Int Immunopharmacol 2024; 131:111896. [PMID: 38518596 DOI: 10.1016/j.intimp.2024.111896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/24/2024]
Abstract
CD155 is an immunoglobulin-like protein overexpressed in almost all the tumor cells, which not only promotes proliferation, adhesion, invasion, and migration of tumor cells, but also regulates immune responses by interacting with TIGIT, CD226 or CD96 receptors expressed on several immune cells, thereby modulating the functionality of these cellular subsets. As a novel immune checkpoint, the inhibition of CD155/TIGIT, either as a standalone treatment or in conjunction with other immune checkpoint inhibitors, has demonstrated efficacy in managing advanced solid malignancies. In this review, we summarize the intricate relationship between on tumor surface CD155 and its receptors, with further discussion on how they regulate the occurrence of tumor immune escape. In addition, novel therapeutic strategies and clinical trials targeting CD155 and its receptors are summarized, providing a strong rationale and way forward for the development of next-generation immunotherapies.
Collapse
Affiliation(s)
- Jiang-Wan Wu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Xing-Jie Dai
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Hui-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
14
|
Lv Q, Zhang Z, Fu H, Li D, Liu Y, Sun Y, Wu M. Predictive Panel for Immunotherapy in Low-Grade Glioma. World Neurosurg 2024; 183:e825-e837. [PMID: 38216032 DOI: 10.1016/j.wneu.2024.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND The main treatment of low-grade glioma (LGG) is still surgical resection followed by radiotherapy and/or chemotherapy, which has certain limitations, including side effects and drug resistance. Immunotherapy is a promising treatment for LGG, but it is generally hindered by the tumor microenvironment with the limited expression of tumor antigens. METHODS We integrated RNA sequencing data sets and clinical information and conducted consistent cluster analysis to explore the most suitable patients for immune checkpoint therapy. Gene set enrichment analysis, UMAP analysis, mutation correlation analysis, TIMER analysis, and TIDE analysis were used to identify the immune characteristics of 3 immune subtypes and the feasibility of 5 antigens as immune checkpoint markers. RESULTS We analyzed the isolation and mutation of homologous recombination repair genes (HRR) of the 3 immune subtypes, and the HRR genes of the 3 subtypes were obviously segregated. Among them, the IS2 subtype has a large number of HRR gene mutations, which increases the immunogenicity of tumors-this is consistent with the results of tumor mutation load analysis of 3 immune subtypes. Then we evaluated the immune cell infiltration of immune subtypes and found that IS2 and IS3 subtypes were rich in immune cells. It is worth noting that there are many Treg cells and NK cells in the IS1 subtype. In addition, when analyzing the immune checkpoint gene expression of the 3 subtypes, we found that they were upregulated most in IS2 subtypes compared with other subtypes. Then when we further confirmed the role of immune-related genes in LGG; through TIDE analysis and TISIDB analysis, we obtained 5 markers that can predict the efficacy of ICB in patients with LGG. In addition, we confirmed that they were associated with poor prognosis through survival analysis. CONCLUSIONS We obtained 3 reliable immune subtypes, and patients with the IS2 subtype are suitable for immunotherapy, in which NAMPT, SLC11A1, TNC, VIM, and SPP1 are predictive panel markers for ICB in the LGG group. Our findings provide a rationale for immunotherapy selection and prediction of patient prognosis in LGG patients.
Collapse
Affiliation(s)
- Qingqing Lv
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyu Zhang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Haijuan Fu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Danyang Li
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yihao Liu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingnan Sun
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
15
|
Kim HD, Yeh CY, Chang YC, Kim CH. Dawn era for revisited cancer therapy by innate immune system and immune checkpoint inhibitors. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167019. [PMID: 38211726 DOI: 10.1016/j.bbadis.2024.167019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
Immunotherapy is a promising therapeutic strategy for cancer. However, it shows limited efficacy against certain tumor types. The activation of innate immunity can suppress tumors by mitigating inflammatory and malignant behaviors through immune surveillance. The tumor microenvironment, which is composed of immune cells and cancer cells, plays a crucial role in determining the outcomes of immunotherapy. Relying solely on immune checkpoint inhibitors is not an optimal approach. Instead, there is a need to consider the use of a combination of immune checkpoint inhibitors with other modulators of the innate immune system to improve the tumor microenvironment. This can be achieved through methods such as immune cell antigen presentation and recognition. In this review, we delve into the significance of innate immune cells in tumor regression, as well as the role of the interaction of tumor cells with innate immune cells in evading host immune surveillance. These findings pave the way for the next chapter in the field of immunotherapy.
Collapse
Affiliation(s)
- Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do 16419, Republic of Korea
| | - Chia-Ying Yeh
- Department of Biomedicine Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chan Chang
- Department of Biomedicine Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do 16419, Republic of Korea; Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea.
| |
Collapse
|
16
|
Zhang Z, Xu X, Du J, Chen X, Xue Y, Zhang J, Yang X, Chen X, Xie J, Ju S. Redox-responsive polymer micelles co-encapsulating immune checkpoint inhibitors and chemotherapeutic agents for glioblastoma therapy. Nat Commun 2024; 15:1118. [PMID: 38320994 PMCID: PMC10847518 DOI: 10.1038/s41467-024-44963-3] [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: 05/03/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Immunotherapy with immune checkpoint blockade (ICB) for glioblastoma (GBM) is promising but its clinical efficacy is seriously challenged by the blood-tumor barrier (BTB) and immunosuppressive tumor microenvironment. Here, anti-programmed death-ligand 1 antibodies (aPD-L1) are loaded into a redox-responsive micelle and the ICB efficacy is further amplified by paclitaxel (PTX)-induced immunogenic cell death (ICD) via a co-encapsulation approach for the reinvigoration of local anti-GBM immune responses. Consequently, the micelles cross the BTB and are retained in the reductive tumor microenvironment without altering the bioactivity of aPD-L1. The ICB efficacy is enhanced by the aPD-L1 and PTX combination with suppression of primary and recurrent GBM, accumulation of cytotoxic T lymphocytes, and induction of long-lasting immunological memory in the orthotopic GBM-bearing mice. The co-encapsulation approach facilitating efficient antibody delivery and combining with chemotherapeutic agent-induced ICD demonstrate that the chemo-immunotherapy might reprogram local immunity to empower immunotherapy against GBM.
Collapse
Affiliation(s)
- Zhiqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xiaoxuan Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jiawei Du
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xin Chen
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, 210009, China
| | - Yonger Xue
- Center for BioDelivery Sciences, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jianqiong Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, 210009, China
| | - Xue Yang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Jinbing Xie
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China.
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
17
|
Ding J, Chen Z, Ding W, Xiang Y, Yang J. DNA polymerase ζ suppresses the radiosensitivity of glioma cells by regulating the PI3K/AKT/mTOR pathway. Autoimmunity 2023; 56:2234101. [PMID: 37448296 DOI: 10.1080/08916934.2023.2234101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
Glioblastoma is the most common glioma with high mortality and poor prognosis. Radiation resistance is one of the large challenges in the treatment of glioma. The study aimed to identify whether DNA polymerase ζ affects glioma cell radiosensitivity. The mRNA and protein levels of REV3L and REV7 were examined using quantitative real-time PCR and western blot. After REV3L and REV7 knockdown in a GBM cell line (A172), we assessed cell viability, colonies, apoptosis, and immune escape. The underlying mechanisms were evaluated using western blot and were confirmed using rescue experiments. The results showed that REV3L and REV7 levels were increased in glioma and related to poor survival. γ-ray treatment inhibited cell viability, survival fraction, and immune escape, and induced apoptosis of glioma cells from a GBM cell line, whereas knockdown of REV3L or REV7 enhanced these effects. Mechanically, silencing of REV3L or REV7 inactivated the PI3K/AKT/mTOR pathway. IGF-1 treatment abrogated the effects on cell viability, colonies, and apoptosis induced by REV3L or REV7 knocking down. Taken together, silencing of REV3L and REV7 inhibited radiation resistance via inactivating the PI3K/AKT/mTOR pathway, suggesting that targeting DNA polymerase ζ may be a new strategy to reduce the radiotherapy resistance of glioma.
Collapse
Affiliation(s)
- Jiqiang Ding
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhisheng Chen
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Weilong Ding
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yongsheng Xiang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Junbao Yang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Senrung A, Tripathi T, Yadav J, Janjua D, Chaudhary A, Chhokar A, Aggarwal N, Joshi U, Goswami N, Bharti AC. In vivo antiangiogenic effect of nimbolide, trans-chalcone and piperine for use against glioblastoma. BMC Cancer 2023; 23:1173. [PMID: 38036978 PMCID: PMC10691152 DOI: 10.1186/s12885-023-11625-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Angiogenesis is an important hallmark of Glioblastoma (GBM) marked by elevated vascular endothelial growth factor-A (VEGF-A) and its receptor 2 (VEGFR-2). As previously reported nimbolide (NBL), trans-chalcone (TC) and piperine (PPR) possess promising antiangiogenic activity in several cancers however, their comparative efficacy and mechanism of antiangiogenic activity in GBM against VEGFR-2 has not been elucidated. METHODS 2D and 3D spheroids cultures of U87 (Uppsala 87 Malignant Glioma) were used for evaluation of non-cytotxoic dose for anti-angiogenic activity. The antiangiogenic effect was investigated by the GBM U87 cell line bearing chick CAM model. Excised U87 xenografts were histologically examined for blood vascular density by histochemistry. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to detect the presence of avian and human VEGF-A and VEGFR-2 mRNA transcripts. RESULTS Using 2D and 3D spheroid models, the non-cytotoxic dose of NBL, TC and PPR was ≤ 11 µM. We found NBL, TC and PPR inhibit U87-induced neoangiogenesis in a dose-dependent manner in the CAM stand-alone model as well as in CAM U87 xenograft model. The results also indicate that these natural compounds inhibit the expression of notable angiogenic factors, VEGF-A and VEGFR-2. A positive correlation was found between blood vascular density and VEGF-A as well as VEGFR-2 transcripts. CONCLUSION Taken together, NBL, TC and PPR can suppress U87-induced neoangiogenesis via a reduction in VEGF-A and its receptor VEGFR-2 transcript expression at noncytotoxic concentrations. These phytochemicals showed their utility as adjuvants to GBM therapy, with Piperine demonstrating superior effectiveness among them all.
Collapse
Affiliation(s)
- Anna Senrung
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Deshbandhu College, University of Delhi, Delhi, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Udit Joshi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Nidhi Goswami
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India.
| |
Collapse
|
19
|
Liang K, Guo Z, Zhang S, Chen D, Zou R, Weng Y, Peng C, Xu Z, Zhang J, Liu X, Pang X, Ji Y, Liao D, Lai M, Peng H, Ke Y, Wang Z, Wang Y. GPR37 expression as a prognostic marker in gliomas: a bioinformatics-based analysis. Aging (Albany NY) 2023; 15:10146-10167. [PMID: 37837549 PMCID: PMC10599758 DOI: 10.18632/aging.205063] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Gliomas are the most frequently diagnosed primary brain tumors, and are associated with multiple molecular aberrations during their development and progression. GPR37 is an orphan G protein-coupled receptor (GPCR) that is implicated in different physiological pathways in the brain, and has been linked to various malignancies. The aim of this study was to explore the relationship between GPR37 gene expression and the clinicopathological factors, patient prognosis, tumor-infiltrating immune cell signature GSEA and methylation levels in glioma. METHODS We explored the diagnostic value, clinical relevance, and molecular function of GPR37 in glioma using TCGA, STRING, cBioPortal, Tumor Immunity Estimation Resource (TIMER) database and MethSurv databases. Besides, the "ssGSEA" algorithm was conducted to estimate immune cells infiltration abundance, with 'ggplot2' package visualizing the results. Immunohistochemical staining of clinical samples were used to verify the speculations of bioinformatics analysis. RESULTS GPR37 expression was significantly higher in the glioma tissues compared to the normal brain tissues, and was linked to poor prognosis. Functional annotation of GPR37 showed enrichment of ether lipid metabolism, fat digestion and absorption, and histidine metabolism. In addition, GSEA showed that GPR37 was positively correlated to the positive regulation of macrophage derived foam cell differentiation, negative regulation of T cell receptor signaling pathway, neuroactive ligand receptor interaction, calcium signaling pathway, and negatively associated with immunoglobulin complex, immunoglobulin complex circulating, ribosome and spliceosome mediated by circulating immunoglobulin etc. TIMER2.0 and ssGSEA showed that GPR37 expression was significantly associated with the infiltration of T cells, CD8 T cell, eosinophils, macrophages, neutrophils, NK CD56dim cells, NK cells, plasmacytoid DCs (pDCs), T helper cells and T effector memory (Tem) cells. In addition, high GPR37 expression was positively correlated with increased infiltration of M2 macrophages, which in turn was associated with poor prognosis. Furthermore, GPR37 was positively correlated with various immune checkpoints (ICPs). Finally, hypomethylation of the GPR37 promoter was associated with its high expression levels and poor prognosis in glioma. CONCLUSION GPR37 had diagnostic and prognostic value in glioma. The possible biological mechanisms of GPR37 provide novel insights into the clinical diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Kairong Liang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhaoxiong Guo
- Science and Technology Innovation Center, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shizhen Zhang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Danmin Chen
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Renheng Zou
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yuhao Weng
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Chengxiang Peng
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhichao Xu
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jingbai Zhang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Xiaorui Liu
- Department of Pharmacy, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Xiao Pang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yunxiang Ji
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Degui Liao
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Miaoling Lai
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Huaidong Peng
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yanbin Ke
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhaotao Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
20
|
Xia Z, Tu R, Liu F, Zhang H, Dai Z, Wang Z, Luo P, He S, Xiao G, Feng J, Cheng Q. PD-L1-related IncRNAs are associated with malignant characteristics and immune microenvironment in glioma. Aging (Albany NY) 2023; 15:10785-10810. [PMID: 37837543 PMCID: PMC10599717 DOI: 10.18632/aging.205120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/21/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND The expression of long non-coding RNA (lncRNA) can function as diagnostic and therapeutic biomarker for tumors. This research explores the role of PD-L1-related lncRNAs in affecting malignant characteristics and the immune microenvironment of glioma. METHODS Downloading gene expression profiles and clinicopathological information of glioma from TCGA and CGGA databases, 6 PD-L1-related lncRNAs were identified through correlation analysis, Cox and LASSO regression analysis, establishing the risk score model based on them. Bioinformatics analysis and cell experiments in vitro were adopted to verify the effects of LINC01271 on glioma. RESULTS Risk scores based on 6 PD-L1-related lncRNAs (AL355974.3, LINC01271, AC011899.3, MIR4500HG, LINC02594, AL357055.3) can reflect malignant characteristics and immunotherapy response of glioma. Patients with high LINC01271 expression had a worse prognosis, a higher abundance of M1 subtype macrophages in the immune microenvironment, and a higher degree of tumor malignancy. Experiments in vitro confirmed its positive regulatory effect on the proliferation and migration of glioma cells. CONCLUSIONS The risk score model based on 6 PD-L1-related lncRNAs can reflect the malignant characteristics and prognosis of glioma. LINC01271 can independently be used as a new target for prognosis evaluation and therapy.
Collapse
Affiliation(s)
- Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Changsha 410205, Hunan, P.R. China
| | - Ruxin Tu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
- MRC Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Little France, Edinburgh, EH16 4UU, UK
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, P.R. China
| | - Shiqing He
- Department of Neurosurgery, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang 421001, Hunan, P.R. China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Jie Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
- Hunan Clinical Research Center for Cerebrovascular Disease, Changsha 410008, Hunan Province, P.R. China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| |
Collapse
|
21
|
Qin Z, Huang Y, Li Z, Pan G, Zheng L, Xiao X, Wang F, Chen J, Chen X, Lin X, Li K, Yan G, Zhang H, Xing F. Glioblastoma Vascular Plasticity Limits Effector T-cell Infiltration and Is Blocked by cAMP Activation. Cancer Immunol Res 2023; 11:1351-1366. [PMID: 37540804 DOI: 10.1158/2326-6066.cir-22-0872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/20/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Glioblastoma (GBM) is the deadliest form of brain cancer. It is a highly angiogenic and immunosuppressive malignancy. Although immune checkpoint blockade therapies have revolutionized treatment for many types of cancer, their therapeutic efficacy in GBM has been far less than expected or even ineffective. In this study, we found that the genomic signature of glioma-derived endothelial cells (GdEC) correlates with an immunosuppressive state and poor prognosis of patients with glioma. We established an in vitro model of GdEC differentiation for drug screening and used this to determine that cyclic adenosine monophosphate (cAMP) activators could effectively block GdEC formation by inducing oxidative stress. Furthermore, cAMP activators impaired GdEC differentiation in vivo, normalized the tumor vessels, and altered the tumor immune profile, especially increasing the influx and function of CD8+ effector T cells. Dual blockade of GdECs and PD-1 induced tumor regression and established antitumor immune memory. Thus, our study reveals that endothelial transdifferentiation of GBM shapes an endothelial immune cell barrier and supports the clinical development of combining GdEC blockade and immunotherapy for GBM. See related Spotlight by Lee et al., p. 1300.
Collapse
Affiliation(s)
- Zixi Qin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Youwei Huang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, P.R. China
| | - Zeying Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Guopeng Pan
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Liangying Zheng
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, P.R. China
| | - Xiao Xiao
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, P.R. China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Fang Wang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Jiahong Chen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Xueqin Chen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Xi Lin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Kai Li
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P.R. China
| | - Haipeng Zhang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Fan Xing
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, P.R. China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
22
|
Hajikarimloo B, Fahim F, Sabbagh Alvani M, Oveisi S, Zali A, Anvari H, Oraee-Yazdani S. Management of Glioblastoma Multiforme During the Severe Acute Respiratory Syndrome Coronavirus 2 Pandemic: A Review of the Literature. World Neurosurg 2023; 178:87-92. [PMID: 37429378 DOI: 10.1016/j.wneu.2023.05.094] [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: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND Patients with glioblastoma multiforme (GBM) and other patients with cancer are at a greater risk of developing severe complications as a result of coronavirus disease 2019 (COVID-19) infection. Therefore, it is crucial to adjust therapeutic approaches to reduce exposure and complications and achieve the most appropriate treatment outcomes. OBJECTIVE Our goal was to help physicians to make decisions based on the latest data in the literature. METHOD We provide a comprehensive review of the literature on the current issues of GBM and COVID-19 infection. RESULTS The mortality of patients with diffuse glioma as a result of COVID-19 infection was 39%, which is higher than in the general population. The statistics showed that 84.5% of patients with diagnosed brain cancer (mostly GBM) and 89.9% of their caregivers received COVID-19 vaccines. The decision to apply different therapeutic approaches must be made individually based on age, tumor grade, molecular profile, and performance status. The advantages and disadvantages of adjuvant radiotherapy and chemotherapy after the surgery should be evaluated carefully. In the setting of the follow-up period, special considerations must be considered to minimize COVID-19 exposure. CONCLUSIONS The pandemic altered medical approaches worldwide, and the management of patients in an immunocompromised state, such as patients with GBM, is challenging; therefore, special considerations must be considered.
Collapse
Affiliation(s)
- Bardia Hajikarimloo
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzan Fahim
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadamin Sabbagh Alvani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayeh Oveisi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Anvari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Kendirli A, de la Rosa C, Lämmle KF, Eglseer K, Bauer IJ, Kavaka V, Winklmeier S, Zhuo L, Wichmann C, Gerdes LA, Kümpfel T, Dornmair K, Beltrán E, Kerschensteiner M, Kawakami N. A genome-wide in vivo CRISPR screen identifies essential regulators of T cell migration to the CNS in a multiple sclerosis model. Nat Neurosci 2023; 26:1713-1725. [PMID: 37709997 PMCID: PMC10545543 DOI: 10.1038/s41593-023-01432-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Multiple sclerosis (MS) involves the infiltration of autoreactive T cells into the CNS, yet we lack a comprehensive understanding of the signaling pathways that regulate this process. Here, we conducted a genome-wide in vivo CRISPR screen in a rat MS model and identified 5 essential brakes and 18 essential facilitators of T cell migration to the CNS. While the transcription factor ETS1 limits entry to the CNS by controlling T cell responsiveness, three functional modules, centered around the adhesion molecule α4-integrin, the chemokine receptor CXCR3 and the GRK2 kinase, are required for CNS migration of autoreactive CD4+ T cells. Single-cell analysis of T cells from individuals with MS confirmed that the expression of these essential regulators correlates with the propensity of CD4+ T cells to reach the CNS. Our data thus reveal key regulators of the fundamental step in the induction of MS lesions.
Collapse
Affiliation(s)
- Arek Kendirli
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Clara de la Rosa
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katrin F Lämmle
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Klara Eglseer
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Isabel J Bauer
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Vladyslav Kavaka
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Stephan Winklmeier
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - La Zhuo
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
- Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried, Germany.
| |
Collapse
|
24
|
Dabkowski TR, Varkhedi M, Song JJ, Gozlan EC, Blanck G. Neuroblastoma and Glioblastoma Cases With Amplified Oncogenes Have Reduced Numbers of Tumor-Resident Adaptive Immune Receptor Recombinations. JCO Precis Oncol 2023; 7:e2300057. [PMID: 38085056 DOI: 10.1200/po.23.00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 12/18/2023] Open
Abstract
PURPOSE In certain cancers, oncogene amplification is correlated with an immunologically cold or noninflamed, tumor immune microenvironment (TIME) and a worse prognosis, for example, in the case of MYCN-amplified neuroblastoma (NBL). However, for other cancer types, the relationship between oncogene amplification and immune response is more complicated or unresolved. One such cancer is glioblastoma multiforme (GBM), in which the epidermal growth factor receptor (EGFR) oncogene is commonly amplified. Unlike MYCN-amplified NBL, EGFR-amplified GBM has not been shown to correlate with a distinct survival probability. METHODS Given this contrasting state for NBL and GBM, we sought to apply a genomics approach to evaluating the immune response for cases with gene amplification. RESULTS Our results confirmed and added further specificity to the cold TIME of MYCN-amplified NBL. Moreover, we demonstrated a novel state of immunologically cold EGFR-amplified GBM tumors. CONCLUSION This approach to using copy number variation and immune receptor recombination read recovery levels to assess gene amplification and TIME, respectively, may be particularly efficient for the rapid evaluation of many other cancer types.
Collapse
Affiliation(s)
- Toriana R Dabkowski
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Mallika Varkhedi
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Joanna J Song
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Etienne C Gozlan
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
25
|
Jain P, Vashist S, Panjiyar BK. Navigating the Immune Challenge in Glioblastoma: Exploring Immunotherapeutic Avenues for Overcoming Immune Suppression. Cureus 2023; 15:e46089. [PMID: 37900496 PMCID: PMC10611557 DOI: 10.7759/cureus.46089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor known for its short survival time, typically 14-18 months from diagnosis to fatality. Managing GBM poses significant challenges due to factors like the formidable blood-brain barrier, the immunosuppressive conditions within GBM, and the intricacies of surgical procedures. Currently, the typical treatment for GBM combines surgical procedures, radiation therapy, and chemotherapy using temozolomide. Unfortunately, this conventional approach has not proven effective in substantially extending the lives of GBM patients. Consequently, researchers are exploring alternative methods for GBM management. One promising avenue receiving attention in recent years is immunotherapy. This approach has successfully treated cancer types like non-small cell lung cancer and blood-related malignancies. Various immunotherapeutic strategies are currently under investigation for GBM treatment, including checkpoint inhibitors, vaccines, chimeric antigen receptor (CAR) T-cell therapy, and oncolytic viruses. A comprehensive review of 26 high-quality studies conducted over the past decade, involving thorough searches of databases such as PubMed and Google Scholar, has been conducted. The findings from this review suggest that while immunotherapeutic strategies show promise, they face significant limitations and challenges in practical application for GBM treatment. The study emphasizes the importance of combining diverse approaches, customizing treatments for individual patients, and ongoing research efforts to improve GBM patients' outlook.
Collapse
Affiliation(s)
- Prateek Jain
- Internal Medicine, Maulana Azad Medical College, Delhi, IND
| | | | - Binay K Panjiyar
- Medicine, Harvard Medical School, Boston, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
26
|
Yao ZG, Hua F, Yin ZH, Xue YJ, Hou YH, Nie YC, Zheng ZM, Zhao MQ, Guo XH, Ma C, Li XK, Wang Z, Liu GC, Zhang GH. Characteristics of glioblastomas and immune microenvironment in a Chinese family with Lynch syndrome and concurrent porokeratosis. Front Oncol 2023; 13:1194232. [PMID: 37529690 PMCID: PMC10388537 DOI: 10.3389/fonc.2023.1194232] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Background Lynch syndrome (LS)-associated glioblastoma (GBM) is rare in clinical practice, and simultaneous occurrence with cutaneous porokeratosis is even rarer. In this study, we analyzed the clinicopathological and genetic characteristics of LS-associated GBMs and concurrent porokeratosis, as well as evaluated the tumor immune microenvironment (TIME) of LS-associated GBMs. Methods Immunohistochemical staining was used to confirm the histopathological diagnosis, assess MMR and PD-1/PD-L1 status, and identify immune cell subsets. FISH was used to detect amplification of EGFR and PDGFRA, and deletion of 1p/19q and CDKN2A. Targeted NGS assay analyzed somatic variants, MSI, and TMB status, while whole-exome sequencing and Sanger sequencing were carried out to analyze the germline mutations. Results In the LS family, three members (I:1, II:1 and II:4) were affected by GBM. GBMs with loss of MSH2 and MSH6 expression displayed giant and multinucleated bizarre cells, along with mutations in ARID1A, TP53, ATM, and NF1 genes. All GBMs had TMB-H but not MSI-H. CD8+ T cells and CD163+ macrophages were abundant in each GBM tissue. The primary and recurrent GBMs of II:1 showed mesenchymal characteristics with high PD-L1 expression. The family members harbored a novel heterozygous germline mutation in MSH2 and FDPS genes, confirming the diagnosis of LS and disseminated superficial actinic porokeratosis. Conclusion LS-associated GBM exhibits heterogeneity in clinicopathologic and molecular genetic features, as well as a suppressive TIME. The presence of MMR deficiency and TMB-H may serve as predictive factors for the response to immune checkpoint inhibitor therapy in GBMs. The identification of LS-associated GBM can provide significant benefits to both patients and their family members, including accurate diagnosis, genetic counseling, and appropriate screening or surveillance protocols. Our study serves as a reminder to clinicians and pathologists to consider the possibility of concurrent genetic syndromes in individuals or families.
Collapse
Affiliation(s)
- Zhi-Gang Yao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fang Hua
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, United States
| | - Zuo-Hua Yin
- Department of Pathology, The People’s Hospital of Huaiyin, Jinan, Shandong, China
| | - Ying-Jie Xue
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yang-Hao Hou
- Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Yi-Cong Nie
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhi-Ming Zheng
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Miao-Qing Zhao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao-Hong Guo
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Chao Ma
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Xiao-Kang Li
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhou Wang
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guang-Cun Liu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Gui-Hui Zhang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
27
|
Braun FK, Rothhammer-Hampl T, Lorenz J, Pohl S, Menevse AN, Vollmann-Zwerenz A, Bumes E, Büttner M, Zoubaa S, Proescholdt M, Schmidt NO, Hau P, Beckhove P, Winner B, Riemenschneider MJ. Scaffold-Based (Matrigel™) 3D Culture Technique of Glioblastoma Recovers a Patient-like Immunosuppressive Phenotype. Cells 2023; 12:1856. [PMID: 37508520 PMCID: PMC10378658 DOI: 10.3390/cells12141856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Conventional 2D cultures are commonly used in cancer research though they come with limitations such as the lack of microenvironment or reduced cell heterogeneity. In this study, we investigated in what respect a scaffold-based (Matrigel™) 3D culture technique can ameliorate the limitations of 2D cultures. NGS-based bulk and single-cell sequencing of matched pairs of 2D and 3D models showed an altered transcription of key immune regulatory genes in around 36% of 3D models, indicating the reoccurrence of an immune suppressive phenotype. Changes included the presentation of different HLA surface molecules as well as cellular stressors. We also investigated the 3D tumor organoids in a co-culture setting with tumor-infiltrating lymphocytes (TILs). Of note, lymphocyte-mediated cell killing appeared less effective in clearing 3D models than their 2D counterparts. IFN-γ release, as well as live cell staining and proliferation analysis, pointed toward an elevated resistance of 3D models. In conclusion, we found that the scaffold-based (Matrigel™) 3D culture technique affects the transcriptional profile in a subset of GBM models. Thus, these models allow for depicting clinically relevant aspects of tumor-immune interaction, with the potential to explore immunotherapeutic approaches in an easily accessible in vitro system.
Collapse
Affiliation(s)
- Frank K Braun
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany
| | | | - Julia Lorenz
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Sandra Pohl
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Ayse-Nur Menevse
- Division of Interventional Immunology, Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Arabel Vollmann-Zwerenz
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Elisabeth Bumes
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Maren Büttner
- Institute of Computational Biology, Helmholtz Center Munich, 85764 Munich, Germany
| | - Saida Zoubaa
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Nils O Schmidt
- Department of Neurosurgery, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Philipp Beckhove
- Division of Interventional Immunology, Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- IZKF Junior Research Group 3 and BMBF Research Group Neuroscience, Interdisciplinary Center for Clinical Research, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | |
Collapse
|
28
|
Abstract
Niacin (vitamin B3) is an essential nutrient that treats pellagra, and prior to the advent of statins, niacin was commonly used to counter dyslipidemia. Recent evidence has posited niacin as a promising therapeutic for several neurological disorders. In this review, we discuss the biochemistry of niacin, including its homeostatic roles in NAD+ supplementation and metabolism. Niacin also has roles outside of metabolism, largely through engaging hydroxycarboxylic acid receptor 2 (Hcar2). These receptor-mediated activities of niacin include regulation of immune responses, phagocytosis of myelin debris after demyelination or of amyloid beta in models of Alzheimer's disease, and cholesterol efflux from cells. We describe the neurological disorders in which niacin has been investigated or has been proposed as a candidate medication. These are multiple sclerosis, Alzheimer's disease, Parkinson's disease, glioblastoma and amyotrophic lateral sclerosis. Finally, we explore the proposed mechanisms through which niacin may ameliorate neuropathology. While several questions remain, the prospect of niacin as a therapeutic to alleviate neurological impairment is promising.
Collapse
Affiliation(s)
- Emily Wuerch
- Hotchkiss Brain Institute, 3330 Hospital Drive NW, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Gloria Roldan Urgoiti
- Hotchkiss Brain Institute, 3330 Hospital Drive NW, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Calgary, AB, Canada
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute, 3330 Hospital Drive NW, Calgary, AB, Canada.
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
29
|
Yao G, Deng L, Long X, Zhou Y, Zhou X. An integrated bioinformatic investigation of focal adhesion-related genes in glioma followed by preliminary validation of COL1A2 in tumorigenesis. Aging (Albany NY) 2023; 15:6225-6254. [PMID: 37354488 PMCID: PMC10373961 DOI: 10.18632/aging.204834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/10/2023] [Indexed: 06/26/2023]
Abstract
Focal adhesions (FAs) allow cells to contact the extracellular matrix, helping to maintain tension and enabling signal transmission in cell migration, differentiation, and apoptosis. In addition, FAs are associated with changes in the tumor microenvironment (TME) that lead to malignant progression and drug resistance in tumors. However, there are still few studies on the comprehensive analysis of focal adhesion-related genes (FARGs) in glioma. Expression data and clinical information of glioma samples were downloaded from public databases. Two distinct molecular subtypes were identified based on FARGs using an unsupervised consensus clustering algorithm. A scoring system consisting of nine FARGs was constructed using integrated LASSO regression and multivariate Cox regression. It not only has outstanding prognostic value but also can guide immunotherapy of glioma patients, which was verified in TCGA, CGGA, GSE16011, and IMvigor210 cohorts. The results of bioinformatics analysis, immunohistochemistry staining, and western blotting all revealed that the expression of COL1A2 was up-regulated in glioblastoma and related to poor prognosis outcomes in patients from public datasets. COL1A2 promotes the proliferation, migration, and invasion of glioblastoma cells. A positive correlation between COL1A2 and CD8 was determined in GBM specimens from eight patients. Moreover, the results of cell co-cultured assay showed that COL1A2 participated in the killing of GBM cells by Jurkat cells. Our study indicates that the FARGs have prominent application value in the identification of molecular subtypes and prediction of survival outcomes in glioma patients. Bioinformatics analysis and experimental verification provide a direction for further research on FARGs.
Collapse
Affiliation(s)
- Guojun Yao
- Department of Neurosurgery, The First People’s Hospital of Fuzhou City, Fuzhou 344099, Jiangxi, P.R. China
| | - Ling Deng
- College of Nursing and Rehabilitation, Fuzhou Medical College of Nanchang University, Fuzhou 344099, Jiangxi, P.R. China
| | - Xinquan Long
- Department of Neurosurgery, The First People’s Hospital of Fuzhou City, Fuzhou 344099, Jiangxi, P.R. China
| | - Yufan Zhou
- Department of Neurosurgery, The First People’s Hospital of Fuzhou City, Fuzhou 344099, Jiangxi, P.R. China
| | - Xiang Zhou
- Department of Neurosurgery, The First People’s Hospital of Fuzhou City, Fuzhou 344099, Jiangxi, P.R. China
| |
Collapse
|
30
|
Xiong Q, Zhu J, Zhang Y, Deng H. CAR-NK cell therapy for glioblastoma: what to do next? Front Oncol 2023; 13:1192128. [PMID: 37404752 PMCID: PMC10315652 DOI: 10.3389/fonc.2023.1192128] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/29/2023] [Indexed: 07/06/2023] Open
Abstract
Glioblastoma is a malignant tumor with the highest morbidity and mortality in the central nervous system. Conventional surgical resection combined with radiotherapy or chemotherapy has a high recurrence rate and poor prognosis. The 5-year survival rate of patients is less than 10%. In tumor immunotherapy, CAR-T cell therapy represented by chimeric antigen receptor-modified T cells has achieved great success in hematological tumors. However, the application of CAR-T cells in solid tumors such as glioblastoma still faces many challenges. CAR-NK cells are another potential adoptive cell therapy strategy after CAR-T cells. Compared with CAR-T cell therapy, CAR-NK cells have similar anti-tumor effects. CAR-NK cells can also avoid some deficiencies in CAR-T cell therapy, a research hotspot in tumor immunity. This article summarizes the preclinical research status of CAR-NK cells in glioblastoma and the problems and challenges faced by CAR-NK in glioblastoma.
Collapse
|
31
|
Liu X, Chen J, Chen W, Xu Y, Shen Y, Xu X. Targeting IGF2BP3 in Cancer. Int J Mol Sci 2023; 24:ijms24119423. [PMID: 37298373 DOI: 10.3390/ijms24119423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
RNA-binding proteins (RBPs) can regulate multiple pathways by binding to RNAs, playing a variety of functions, such as localization, stability, and immunity. In recent years, with the development of technology, researchers have discovered that RBPs play a key role in the N6-methyladenosine (m6A) modification process. M6A methylation is the most abundant form of RNA modification in eukaryotes, which is defined as methylation on the sixth N atom of adenine in RNA. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) is one of the components of m6A binding proteins, which plays an important role in decoding m6A marks and performing various biological functions. IGF2BP3 is abnormally expressed in many human cancers, often associated with poor prognosis. Here, we summarize the physiological role of IGF2BP3 in organisms and describe its role and mechanism in tumors. These data suggest that IGF2BP3 may be a valuable therapeutic target and prognostic marker in the future.
Collapse
Affiliation(s)
- Xin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiayu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenliang Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yangtao Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yang Shen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
32
|
El Baba R, Pasquereau S, Haidar Ahmad S, Monnien F, Abad M, Bibeau F, Herbein G. EZH2-Myc driven glioblastoma elicited by cytomegalovirus infection of human astrocytes. Oncogene 2023:10.1038/s41388-023-02709-3. [PMID: 37147437 DOI: 10.1038/s41388-023-02709-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
Mounting evidence is identifying human cytomegalovirus (HCMV) as a potential oncogenic virus. HCMV has been detected in malignant gliomas. EZH2 and Myc play a potential oncogenic role, correlating with the glioma grade. Herewith, we present the first experimental evidence for HCMV as a reprogramming vector, straight through the dedifferentiation of mature human astrocytes, and generation of CMV-Elicited Glioblastoma Cells (CEGBCs) possessing glioblastoma-like traits. HCMV counterparts the progression of the perceived cellular and molecular mechanisms succeeding the transformation and invasion processes with CEGBCs involved in spheroid formation and invasiveness. Glioblastoma multiforme (GBM) biopsies were characterized by an elevated EZH2 and Myc expression, possessing a strong positive correlation between the aforementioned markers in the presence of HCMV. From GBM tissues, we isolated HCMV clinical strains that transformed HAs toward CEGBCs exhibiting upregulated EZH2 and Myc. Spheroids generated from CEGBCs possessed invasion potential and were sensitive to EZH2 inhibitor, ganciclovir, and temozolomide triple therapy. HCMV clinical strains transform HAs and fit with an HCMV-induced glioblastoma model of oncogenesis, and supports the tumorigenic properties of Myc and EZH2 which might be highly pertinent in the pathophysiology of astrocytic brain tumors and thereby paving the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Ranim El Baba
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | - Sébastien Pasquereau
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | - Sandy Haidar Ahmad
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France
| | | | - Marine Abad
- Department of Pathology, CHU Besançon, Besançon, France
| | | | - Georges Herbein
- Department of Pathogens & Inflammation-EPILAB Laboratory EA4266, University of Franche-Comté, Besançon, France.
- Department of Virology, CHU Besançon, Besançon, France.
| |
Collapse
|
33
|
Montemurro N, Pahwa B, Tayal A, Shukla A, De Jesus Encarnacion M, Ramirez I, Nurmukhametov R, Chavda V, De Carlo A. Macrophages in Recurrent Glioblastoma as a Prognostic Factor in the Synergistic System of the Tumor Microenvironment. Neurol Int 2023; 15:595-608. [PMID: 37218976 DOI: 10.3390/neurolint15020037] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is a common and highly malignant primary tumor of the central nervous system in adults. Ever more recent papers are focusing on understanding the role of the tumor microenvironment (TME) in affecting tumorigenesis and the subsequent prognosis. We assessed the impact of macrophages in the TME on the prognosis in patients with recurrent GBM. A PubMed, MEDLINE and Scopus review was conducted to identify all studies dealing with macrophages in the GBM microenvironment from January 2016 to December 2022. Glioma-associated macrophages (GAMs) act critically in enhancing tumor progression and can alter drug resistance, promoting resistance to radiotherapy and establishing an immunosuppressive environment. M1 macrophages are characterized by increased secretion of proinflammatory cytokines, such as IL-1ß, tumor necrosis factor (TNF), IL-27, matrix metalloproteinase (MMPs), CCL2, and VEGF (vascular endothelial growth factor), IGF1, that can lead to the destruction of the tissue. In contrast, M2 is supposed to participate in immunosuppression and tumor progression, which is formed after being exposed to the macrophage M-CSF, IL-10, IL-35 and the transforming growth factor-ß (TGF-β). Because there is currently no standard of care in recurrent GBM, novel identified targeted therapies based on the complex signaling and interactions between the glioma stem cells (GSCs) and the TME, especially resident microglia and bone-marrow-derived macrophages, may be helpful in improving the overall survival of these patients in the near future.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| | - Bhavya Pahwa
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anish Tayal
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anushruti Shukla
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | | | - Issael Ramirez
- Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Renat Nurmukhametov
- Department of Spinal Surgery, Central Clinical Hospital of the Russian Academy of Sciences, 121359 Moscow, Russia
| | - Vishal Chavda
- Department of Pathology, Stanford of School of Medicine, Stanford University Medical Centre, Palo Alto, CA 94305, USA
| | - Antonella De Carlo
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
34
|
Wang Y, Li S, Peng Y, Ma W, Wang Y, Li W. Progress in phase III clinical trials of molecular targeted therapy and immunotherapy for glioblastoma. CANCER INNOVATION 2023; 2:114-130. [PMID: 38090060 PMCID: PMC10686181 DOI: 10.1002/cai2.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 10/15/2024]
Abstract
Glioblastoma (GBM) is the most common primary central nervous system tumor, whose prognosis remains poor under the sequential standard of care, such as neurosurgery followed by concurrent temozolomide radiochemotherapy and adjuvant temozolomide chemotherapy in the presence or absence of tumor treating fields. Accordingly, the advent of molecular targeted therapy and immunotherapy has opened a new era of tumor management. A diverse range of targeted drugs have been tested in patients with GBM in phase III clinical trials. However, these drugs are ineffective for all patients, as evidenced by the fact that only a minority of patients in these trials showed prolonged survival. Furthermore, there are several published phase III clinical trials that involve immune checkpoint inhibitors, peptide vaccines, dendritic cell vaccines, and virotherapy. Accordingly, this review comprehensively overviews existing studies of targeted drugs and immunotherapy for glioma and discusses the challenge and perspective of targeted drugs and immunotherapy for glioma to clarify future directions.
Collapse
Affiliation(s)
- Yuekun Wang
- Department of Neurosurgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shenglan Li
- Department of Neuro‐oncology, Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yichen Peng
- Department of Neuro‐oncology, Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wenbin Li
- Department of Neuro‐oncology, Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
35
|
Hypocretin-1 suppresses malignant progression of glioblastoma cells through Notch1 signaling pathway. Brain Res Bull 2023; 196:46-58. [PMID: 36925051 DOI: 10.1016/j.brainresbull.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Hypocretin-1 is a multifunctional neuropeptide that has been identified as a potential antitumor agent for its role in inhibiting tumor growth, including in colon cancer, neuroendocrine tumor, and prostate cancer. However, the role and mechanism of hypocretin-1 in the occurrence and development of malignant glioma have not been well studied. Therefore, we investigated the effect of hypocretin-1 on glioblastoma proliferation, apoptosis, migration and invasion and its mechanism. We found that the hypocretin-1 receptor was expressed in both glioma cell lines and glioma tissues. Hypocretin-1 treatment can inhibit glioblastoma cell proliferation, migration and invasion, and induce cell apoptosis. Meanwhile, hypocretin-1 treatment significantly reduces tumor growth rate and tumor weight. In addition, mechanistic studies have found that hypocretin-1 exerts antitumor effects by inhibiting NOTCH signaling pathway. Overexpression of NICD significantly reversed the antitumor effect of hypocretin on glioblastoma. Taken together, these findings suggest that hypocretin-1 inhibits glioblastoma proliferation, migration and invasion and induces apoptosis in vitro and in vivo through NOTCH signaling pathway.
Collapse
|
36
|
Zhu X, Fang Y, Chen Y, Chen Y, Hong W, Wei W, Tu J. Interaction of tumor-associated microglia/macrophages and cancer stem cells in glioma. Life Sci 2023; 320:121558. [PMID: 36889666 DOI: 10.1016/j.lfs.2023.121558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
Glioma is the most common tumor of the primary central nervous system, and its malignant phenotype has been shown to be closely related to glioma stem cells (GSCs). Although temozolomide has significantly improved the therapeutic outcome of glioma with a high penetration rate of the blood-brain barrier, resistance is often present in patients. Moreover, evidence has shown that the crosstalk between GSCs and tumor-associated microglia/macrophages (TAMs) affect the clinical occurrence, growth, and multi-tolerance of chemoradiotherapy in gliomas. Here, we highlight its vital roles in the maintenance of the stemness of GSCs and the ability of GSCs to recruit TAMs to the tumor microenvironment and promote their polarization into tumor-promoting macrophages, hence providing groundwork for future research into new treatment strategies of cancer.
Collapse
Affiliation(s)
- Xiangling Zhu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yilong Fang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yizhao Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Chen
- Department of Gynecology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wenming Hong
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| |
Collapse
|
37
|
Jin X, Zhao X. A new immune checkpoint-associated nine-gene signature for prognostic prediction of glioblastoma. Medicine (Baltimore) 2023; 102:e33150. [PMID: 36862886 PMCID: PMC9981394 DOI: 10.1097/md.0000000000033150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023] Open
Abstract
Glioblastoma (GBM) is a highly malignant neurological tumor that has a poor prognosis. While pyroptosis affects cancer cell proliferation, invasion and migration, function of pyroptosis-related genes (PRGs) in GBM as well as the prognostic significance of PRGs remain obscure. By analyzing the mechanisms involved in the association between pyroptosis and GBM, our study hopes to provide new insights into the treatment of GBM. Here, 32 out of 52 PRGs were identified as the differentially expressed genes between GBM tumor versus normal tissues. And all GBM cases were assigned to 2 groups according to the expression of the differentially expressed genes using comprehensive bioinformatics analysis. The least absolute shrinkage and selection operator analysis led to the construction of a 9-gene signature, and the cancer genome atlas cohort of GBM patients were categorized into high risk and low risk subgroups. A significant increase in the survival possibility was found in low risk patients in comparison with the high risk ones. Consistently, low risk patients of a gene expression omnibus cohort displayed a markedly longer overall survival than the high risk counterparts. The risk score calculated using the gene signature was found to be an independent predictor of survival of GBM cases. Besides, we observed significant differences in the expression levels of immune checkpoints between the high risk versus low risk GBM cases, providing instructive suggestions for immunotherapy of GBM. Overall, the present study developed a new multigene signature for prognostic prediction of GBM.
Collapse
Affiliation(s)
- Xiao Jin
- The Personnel Department, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Fengtai District, Beijing, China
| | - Xiang Zhao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
38
|
Pineda E, Domenech M, Hernández A, Comas S, Balaña C. Recurrent Glioblastoma: Ongoing Clinical Challenges and Future Prospects. Onco Targets Ther 2023; 16:71-86. [PMID: 36721854 PMCID: PMC9884437 DOI: 10.2147/ott.s366371] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Virtually all glioblastomas treated in the first-line setting will recur in a short period of time, and the search for alternative effective treatments has so far been unsuccessful. Various obstacles remain unresolved, and no effective salvage therapy for recurrent glioblastoma can be envisaged in the short term. One of the main impediments to progress is the low incidence of the disease itself in comparison with other pathologies, which will be made even lower by the recent WHO classification of gliomas, which includes molecular alterations. This new classification helps refine patient prognosis but does not clarify the most appropriate treatment. Other impediments are related to clinical trials: glioblastoma patients are often excluded from trials due to their advanced age and limiting neurological symptoms; there is also the question of how best to measure treatment efficacy, which conditions the design of trials and can affect the acceptance of results by oncologists and medicine agencies. Other obstacles are related to the drugs themselves: most treatments cannot cross the blood-brain-barrier or the brain-to-tumor barrier to reach therapeutic drug levels in the tumor without producing toxicity; the drugs under study may have adverse metabolic interactions with those required for symptom control; identifying the target of the drug can be a complex issue. Additionally, the optimal method of treatment - local vs systemic therapy, the choice of chemotherapy, irradiation, targeted therapy, immunotherapy, or a combination thereof - is not yet clear in glioblastoma in comparison with other cancers. Finally, in addition to curing or stabilizing the disease, glioblastoma therapy should aim at maintaining the neurological status of the patients to enable them to return to their previous lifestyle. Here we review currently available treatments, obstacles in the search for new treatments, and novel lines of research that show promise for the future.
Collapse
Affiliation(s)
- Estela Pineda
- Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Marta Domenech
- Medical Oncology, Institut Catala d’Oncologia (ICO) Badalona, Barcelona, Spain
| | - Ainhoa Hernández
- Medical Oncology, Institut Catala d’Oncologia (ICO) Badalona, Barcelona, Spain
| | - Silvia Comas
- Radiation Oncology, Institut Catala d’Oncologia (ICO) Badalona, Badalona, Spain
| | - Carmen Balaña
- Medical Oncology, Institut Catala d’Oncologia (ICO) Badalona, Barcelona, Spain,Correspondence: Carmen Balaña, Institut Catala d’Oncologia (ICO) Badalona, Carretera Canyet s/n, Badalona, 08916, Spain, Tel +34 497 89 25, Fax +34 497 89 50, Email
| |
Collapse
|
39
|
Zhang Z, Shen X, Tan Z, Mei Y, Lu T, Ji Y, Cheng S, Xu Y, Wang Z, Liu X, He W, Chen Z, Chen S, Lv Q. Interferon gamma-related gene signature based on anti-tumor immunity predicts glioma patient prognosis. Front Genet 2023; 13:1053263. [PMID: 36712869 PMCID: PMC9880184 DOI: 10.3389/fgene.2022.1053263] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Background: Glioma is the most common primary tumor of the central nervous system. The conventional glioma treatment strategies include surgical excision and chemo- and radiation-therapy. Interferon Gamma (IFN-γ) is a soluble dimer cytokine involved in immune escape of gliomas. In this study, we sought to identify IFN-γ-related genes to construct a glioma prognostic model to guide its clinical treatment. Methods: RNA sequences and clinicopathological data were downloaded from The Cancer Genome Atlas (TCGA) and the China Glioma Genome Atlas (CGGA). Using univariate Cox analysis and the Least Absolute Shrinkage and Selection Operator (LASSO) regression algorithm, IFN-γ-related prognostic genes were selected to construct a risk scoring model, and analyze its correlation with the clinical features. A high-precision nomogram was drawn to predict prognosis, and its performance was evaluated using calibration curve. Finally, immune cell infiltration and immune checkpoint molecule expression were analyzed to explore the tumor microenvironment characteristics associated with the risk scoring model. Results: Four out of 198 IFN-γ-related genes were selected to construct a risk score model with good predictive performance. The expression of four IFN-γ-related genes in glioma tissues was significantly increased compared to normal brain tissue (p < 0.001). Based on ROC analysis, the risk score model accurately predicted the overall survival rate of glioma patients at 1 year (AUC: The Cancer Genome Atlas 0.89, CGGA 0.59), 3 years (AUC: TCGA 0.89, CGGA 0.68), and 5 years (AUC: TCGA 0.88, CGGA 0.70). Kaplan-Meier analysis showed that the overall survival rate of the high-risk group was significantly lower than that of the low-risk group (p < 0.0001). Moreover, high-risk scores were associated with wild-type IDH1, wild-type ATRX, and 1P/19Q non-co-deletion. The nomogram predicted the survival rate of glioma patients based on the risk score and multiple clinicopathological factors such as age, sex, pathological grade, and IDH Status, among others. Risk score and infiltrating immune cells including CD8 T-cell, resting CD4 memory T-cell, regulatory T-cell (Tregs), M2 macrophages, resting NK cells, activated mast cells, and neutrophils were positively correlated (p < 0.05). In addition, risk scores closely associated with expression of immune checkpoint molecules such as PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, TIGIT, CD48, CD226, and CD96. Conclusion: Our risk score model reveals that IFN-γ -associated genes are an independent prognostic factor for predicting overall survival in glioma, which is closely associated with immune cell infiltration and immune checkpoint molecule expression. This model will be helpful in predicting the effectiveness of immunotherapy and survival rate in patients with glioma.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China,Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Xiaoli Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zilong Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yuran Mei
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Tianzhu Lu
- Department of Radiation Oncology and Head and Neck Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Yulong Ji
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Sida Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yu Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zekun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinxian Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei He
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhen Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shuhui Chen
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China,Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qiaoli Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China,*Correspondence: Qiaoli Lv,
| |
Collapse
|
40
|
Laurent PA, Morel D, Meziani L, Depil S, Deutsch E. Radiotherapy as a means to increase the efficacy of T-cell therapy in solid tumors. Oncoimmunology 2022; 12:2158013. [PMID: 36567802 PMCID: PMC9788698 DOI: 10.1080/2162402x.2022.2158013] [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] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have demonstrated significant improvements in the treatment of refractory B-cell malignancies that previously showed limited survival. In contrast, early-phase clinical studies targeting solid tumors have been disappointing. This may be due to both a lack of specific and homogeneously expressed targets at the surface of tumor cells, as well as intrinsic properties of the solid tumor microenvironment that limit homing and activation of adoptive T cells. Faced with these antagonistic conditions, radiotherapy (RT) has the potential to change the overall tumor landscape, from depleting tumor cells to reshaping the tumor microenvironment. In this article, we describe the current landscape and discuss how RT may play a pivotal role for enhancing the efficacy of adoptive T-cell therapies in solid tumors. Indeed, by improving homing, expansion and activation of infused T cells while reducing tumor volume and heterogeneity, the use of RT could help the implementation of engineered T cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Pierre-Antoine Laurent
- Department of Radiation Oncology, Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| | - Daphne Morel
- Drug Development Department (D.I.T.E.P), Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
| | - Lydia Meziani
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| | | | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus; UNICANCER, Villejuif, France
- INSERM U1030, Molecular Radiation Therapy and Therapeutic Innovation, Gustave Roussy Cancer Campus, University of Paris-Saclay, SIRIC SOCRATE, Villejuif, France
| |
Collapse
|
41
|
Protein Kinase Inhibitors as a New Target for Immune System Modulation and Brain Cancer Management. Int J Mol Sci 2022; 23:ijms232415693. [PMID: 36555334 PMCID: PMC9778944 DOI: 10.3390/ijms232415693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
High-grade brain tumors are malignant tumors with poor survival and remain the most difficult tumors to treat. An important contributing factor to the development and progression of brain tumors is their ability to evade the immune system. Several immunotherapeutic strategies including vaccines and checkpoint inhibitors have been studied to improve the effectiveness of the immune system in destroying cancer cells. Recent studies have shown that kinase inhibitors, capable of inhibiting signal transduction cascades that affect cell proliferation, migration, and angiogenesis, have additional immunological effects. In this review, we explain the beneficial therapeutic effects of novel small-molecule kinase inhibitors and explore how, through different mechanisms, they increase the protective antitumor immune response in high-grade brain tumors.
Collapse
|
42
|
Zhou J, Li L, Jia M, Liao Q, Peng G, Luo G, Zhou Y. Dendritic cell vaccines improve the glioma microenvironment: Influence, challenges, and future directions. Cancer Med 2022; 12:7207-7221. [PMID: 36464889 PMCID: PMC10067114 DOI: 10.1002/cam4.5511] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Gliomas, especially the glioblastomas, are one of the most aggressive intracranial tumors with poor prognosis. This might be explained by the heterogeneity of tumor cells and the inhibitory immunological microenvironment. Dendritic cells (DCs), as the most potent in vivo functional antigen-presenting cells, link innate immunity with adaptive immunity. However, their function is suppressed in gliomas. Therefore, overcoming the dysfunction of DCs in the TME might be critical to treat gliomas. METHOD In this paper we proposed the specificity of the glioma microenvironment, analyzed the pathways leading to the dysfunction of DCs in tumor microenvironment of patients with glioma, summarized influence of DC-based immunotherapy on the tumor microenvironment and proposed new development directions and possible challenges of DC vaccines. RESULT DC vaccines can improve the immunosuppressive microenvironment of glioma patients. It will bring good treatment prospects to patients. We also proposed new development directions and possible challenges of DC vaccines, thus providing an integrated understanding of efficacy on DC vaccines for glioma treatment.
Collapse
Affiliation(s)
- Jing Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
- Cancer Research Institute, Basic School of Medicine Central South University Changsha Hunan China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| | - Luohong Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
- Cancer Research Institute, Basic School of Medicine Central South University Changsha Hunan China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| | - Minqi Jia
- Department of Radiation Oncology Peking University Cancer Hospital & Institute Beijing China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| | - Guiping Peng
- Xiangya School of Medicine Central South University Changsha China
| | - Gengqiu Luo
- Department of Pathology, Xiangya Hospital, Basic School of Medicine Central South University Changsha Hunan China
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
- Cancer Research Institute, Basic School of Medicine Central South University Changsha Hunan China
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine Central South University Changsha Hunan China
| |
Collapse
|
43
|
Li H, He J, Li M, Li K, Pu X, Guo Y. Immune landscape-based machine-learning-assisted subclassification, prognosis, and immunotherapy prediction for glioblastoma. Front Immunol 2022; 13:1027631. [PMID: 36532035 PMCID: PMC9751405 DOI: 10.3389/fimmu.2022.1027631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction As a malignant brain tumor, glioblastoma (GBM) is characterized by intratumor heterogeneity, a worse prognosis, and highly invasive, lethal, and refractory natures. Immunotherapy has been becoming a promising strategy to treat diverse cancers. It has been known that there are highly heterogeneous immunosuppressive microenvironments among different GBM molecular subtypes that mainly include classical (CL), mesenchymal (MES), and proneural (PN), respectively. Therefore, an in-depth understanding of immune landscapes among them is essential for identifying novel immune markers of GBM. Methods and results In the present study, based on collecting the largest number of 109 immune signatures, we aim to achieve a precise diagnosis, prognosis, and immunotherapy prediction for GBM by performing a comprehensive immunogenomic analysis. Firstly, machine-learning (ML) methods were proposed to evaluate the diagnostic values of these immune signatures, and the optimal classifier was constructed for accurate recognition of three GBM subtypes with robust and promising performance. The prognostic values of these signatures were then confirmed, and a risk score was established to divide all GBM patients into high-, medium-, and low-risk groups with a high predictive accuracy for overall survival (OS). Therefore, complete differential analysis across GBM subtypes was performed in terms of the immune characteristics along with clinicopathological and molecular features, which indicates that MES shows much higher immune heterogeneity compared to CL and PN but has significantly better immunotherapy responses, although MES patients may have an immunosuppressive microenvironment and be more proinflammatory and invasive. Finally, the MES subtype is proved to be more sensitive to 17-AAG, docetaxel, and erlotinib using drug sensitivity analysis and three compounds of AS-703026, PD-0325901, and MEK1-2-inhibitor might be potential therapeutic agents. Conclusion Overall, the findings of this research could help enhance our understanding of the tumor immune microenvironment and provide new insights for improving the prognosis and immunotherapy of GBM patients.
Collapse
|
44
|
Boot J, Rosser G, Kancheva D, Vinel C, Lim YM, Pomella N, Zhang X, Guglielmi L, Sheer D, Barnes M, Brandner S, Nelander S, Movahedi K, Marino S. Global hypo-methylation in a proportion of glioblastoma enriched for an astrocytic signature is associated with increased invasion and altered immune landscape. eLife 2022; 11:e77335. [PMID: 36412091 PMCID: PMC9681209 DOI: 10.7554/elife.77335] [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: 01/25/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
We describe a subset of glioblastoma, the most prevalent malignant adult brain tumour, harbouring a bias towards hypomethylation at defined differentially methylated regions. This epigenetic signature correlates with an enrichment for an astrocytic gene signature, which together with the identification of enriched predicted binding sites of transcription factors known to cause demethylation and to be involved in astrocytic/glial lineage specification, point to a shared ontogeny between these glioblastomas and astroglial progenitors. At functional level, increased invasiveness, at least in part mediated by SRPX2, and macrophage infiltration characterise this subset of glioblastoma.
Collapse
Affiliation(s)
- James Boot
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Gabriel Rosser
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Dailya Kancheva
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit BrusselBrusselsBelgium
| | - Claire Vinel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Yau Mun Lim
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, and Department of Neurodegenerative Disease, Queen Square, Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Nicola Pomella
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Xinyu Zhang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Loredana Guglielmi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Denise Sheer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Michael Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Sebastian Brandner
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, and Department of Neurodegenerative Disease, Queen Square, Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Sven Nelander
- Department of Immunology Genetics and Pathology, Uppsala UniversityUppsalaSweden
| | - Kiavash Movahedi
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit BrusselBrusselsBelgium
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| |
Collapse
|
45
|
Robilliard LD, Yu J, Anchan A, Finlay G, Angel CE, Graham ES. Comprehensive Assessment of Secreted Immuno-Modulatory Cytokines by Serum-Differentiated and Stem-like Glioblastoma Cells Reveals Distinct Differences between Glioblastoma Phenotypes. Int J Mol Sci 2022; 23:ijms232214164. [PMID: 36430641 PMCID: PMC9692434 DOI: 10.3390/ijms232214164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/04/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is refractory to therapy and presents a significant oncological challenge. Promising immunotherapies have not shown the promise observed in other aggressive cancers. The reasons for this include the highly immuno-suppressive tumour microenvironment controlled by the glioblastoma cells and heterogeneous phenotype of the glioblastoma cells. Here, we wanted to better understand which glioblastoma phenotypes produced the regulatory cytokines, particularly those that are implicated in shaping the immune microenvironment. In this study, we employed nanoString analysis of the glioblastoma transcriptome, and proteomic analysis (proteome profiler arrays and cytokine profiling) of secreted cytokines by different glioblastoma phenotypes. These phenotypes were cultured to reflect a spectrum of glioblastoma cells present in tumours, by culturing an enhanced stem-like phenotype of glioblastoma cells or a more differentiated phenotype following culture with serum. Extensive secretome profiling reveals that there is considerable heterogeneity in secretion patterns between serum-derived and glioblastoma stem-like cells, as well as between individuals. Generally, however, the serum-derived phenotypes appear to be the primary producers of cytokines associated with immune cell recruitment into the tumour microenvironment. Therefore, these glioblastoma cells have considerable importance in shaping the immune landscape in glioblastoma and represent a valuable therapeutic target that should not be ignored.
Collapse
Affiliation(s)
- Laverne D. Robilliard
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
| | - Jane Yu
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
| | - Akshata Anchan
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
| | - Graeme Finlay
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Catherine E. Angel
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1010, New Zealand
| | - E Scott Graham
- School of Medical Sciences, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Centre for Brain Research, University of Auckland, Auckland 1023, New Zealand
- Correspondence:
| |
Collapse
|
46
|
Chen X, Tian F, Wu Z. A Genomic Instability-Associated Prognostic Signature for Glioblastoma Patients. World Neurosurg 2022; 167:e515-e526. [PMID: 35977679 DOI: 10.1016/j.wneu.2022.08.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Genomic instability and aberrant tumor mutation burden are widely accepted hallmarks of cancer. Glioblastoma (GBM) is a common brain tumor in adults, and survival of patients with GBM is poor. This study aimed to investigate the prognostic value of genomic instability-derived genes in GBM. METHODS GBM data were downloaded from The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. Differential expression analysis of all samples with different tumor mutation burden was performed. Univariate Cox and LASSO Cox regression analyses were integrated to determine the optimal genes for constructing a risk score model. Multivariate Cox regression analysis and survival analysis determined independent prognostic indicators. Immune cell infiltration was analyzed by CIBERSORT algorithm. RESULTS In GMB patients with high and low tumor mutation burden, we identified 154 differentially expressed genes, which were significantly enriched in 47 Gene Ontology terms and 6 Kyoto Encyclopedia of Genes and Genomes pathways. To establish a risk score, 9 genes were further screened, including SDC1, CXCL1, CXCL6, RGS4, PCDHGB2, CA9, ZAR1, CHRM3, and SLN. High-risk patients had worse prognosis in two databases. The performance of a nomogram including prognostic factors (risk score and age) was good. Moreover, mast cells resting was significantly differentially infiltrated between high- and low-risk GBM samples. CONCLUSIONS The risk score constructed by 9 genomic instability-derived genes could reliably predict prognosis of GBM patients. The nomogram based on age and risk score also had a good prognostic predictive value.
Collapse
Affiliation(s)
- Xiaodong Chen
- Neurosurgery Department, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Fen Tian
- Nephrology Department, The Affiliated Hospital of Qingdao University, Shandong, China.
| | - Zeyu Wu
- Neurosurgery Department, The Affiliated Hospital of Qingdao University, Shandong, China
| |
Collapse
|
47
|
Di Castro MA, Garofalo S, De Felice E, Meneghetti N, Di Pietro E, Mormino A, Mazzoni A, Caleo M, Maggi L, Limatola C. Environmental enrichment counteracts the effects of glioma in primary visual cortex. Neurobiol Dis 2022; 174:105894. [DOI: 10.1016/j.nbd.2022.105894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
|
48
|
Zhao W, Wu Y, Zhao F, Xue Z, Liu W, Cao Z, Zhao Z, Huang B, Han M, Li X. Scoring model based on the signature of non-m6A-related neoantigen-coding lncRNAs assists in immune microenvironment analysis and TCR-neoantigen pair selection in gliomas. J Transl Med 2022; 20:494. [DOI: 10.1186/s12967-022-03713-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/20/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Small peptides encoded by long non-coding RNAs (lncRNAs) have attracted attention for their various functions. Recent studies indicate that these small peptides participate in immune responses and antigen presentation. However, the significance of RNA modifications remains unclear.
Methods
Thirteen non-m6A-related neoantigen-coding lncRNAs were selected for analysis from the TransLnc database. Next, a neoantigen activation score (NAS) model was established based on the characteristics of the lncRNAs. Machine learning was employed to expand the model to two additional RNA-seq and two single-cell sequencing datasets for further validation. The DLpTCR algorithm was used to predict T cell receptor (TCR)-peptide binding probability.
Results
The non-m6A-related NAS model predicted patients’ overall survival outcomes more precisely than the m6A-related NAS model. Furthermore, the non-m6A-related NAS was positively correlated with tumor cells’ evolutionary level, immune infiltration, and antigen presentation. However, high NAS gliomas also showed more PD-L1 expression and high mutation frequencies of T-cell positive regulators. Interestingly, results of intercellular communication analysis suggest that T cell-high neoplastic cell interaction is weaker in both of the NAS groups which might arise from decreased IFNGR1 expression. Moreover, we identified unique TCR-peptide pairs present in all glioma samples based on peptides encoded by the 13 selected lncRNAs. And increased levels of neoantigen-active TCR patterns were found in high NAS gliomas.
Conclusions
Our work suggests that non-m6A-related neoantigen-coding lncRNAs play an essential role in glioma progression and that screened TCR clonotypes might provide potential avenues for chimeric antigen receptor T cell (CAR-T) therapy for gliomas.
Collapse
|
49
|
Zheng J, Qiu Y, Wu Z, Wang X, Jiang X. Exploring the multidimensional heterogeneities of glioblastoma multiforme based on sample-specific edge perturbation in gene interaction network. Front Immunol 2022; 13:944030. [PMID: 36105808 PMCID: PMC9464945 DOI: 10.3389/fimmu.2022.944030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/12/2022] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain cancer with great heterogeneities in many aspects, such as prognosis, clinicopathological features, immune landscapes, and immunotherapeutic responses. Considering that gene interaction network is relatively stable in a healthy state but widely perturbed in cancers, we sought to explore the multidimensional heterogeneities of GBM through evaluating the degree of network perturbations. The gene interaction network perturbations of GBM samples (TCGA cohort) and normal samples (GTEx database) were characterized by edge perturbations, which were quantized through evaluating the change in relative gene expression value. An unsupervised consensus clustering analysis was performed to identify edge perturbation-based clusters of GBM samples. Results revealed that the edge perturbation of GBM samples was stronger than that of normal samples. Four edge perturbation-based clusters of GBM samples were identified and showed prominent heterogeneities in prognosis, clinicopathological features, somatic genomic alterations, immune landscapes, and immunotherapeutic responses. In addition, a sample-specific perturbation of gene interaction score (SPGIScore) was constructed based on the differently expressed genes (DEGs) among four clusters, and exhibited a robust ability to predict prognosis. In conclusion, the bioinformatics approach based on sample-specific edge perturbation in gene interaction network provided a new perspective to understanding the multidimensional heterogeneities of GBM.
Collapse
Affiliation(s)
- Jianglin Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Qiu
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhipeng Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xuan Wang, ; Xiaobing Jiang,
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xuan Wang, ; Xiaobing Jiang,
| |
Collapse
|
50
|
Mews EA, Beckmann P, Patchava M, Wang Y, Largaespada DA, Wagner CR. Multivalent, Bispecific αB7-H3-αCD3 Chemically Self-Assembled Nanorings Direct Potent T Cell Responses against Medulloblastoma. ACS NANO 2022; 16:12185-12201. [PMID: 35876221 PMCID: PMC9885520 DOI: 10.1021/acsnano.2c02850] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Few therapeutic options have been made available for treating central nervous system tumors, especially upon recurrence. Recurrent medulloblastoma is uniformly lethal with no approved therapies. Recent preclinical studies have shown promising results for eradicating various solid tumors by targeting the overexpressed immune checkpoint molecule, B7-H3. However, due to several therapy-related toxicities and reports of tumor escape, the full potential of targeting this pan-cancer antigen has yet to be realized. Here, we designed and characterized bispecific chemically self-assembling nanorings (CSANs) that target the T cell receptor, CD3ε, and tumor associated antigen, B7-H3, derived from the humanized 8H9 single chain variable fragment. We show that the αB7-H3-αCD3 CSANs increase T cell infiltration and facilitate selective cytotoxicity of B7-H3+ medulloblastoma spheroids and that activity is independent of target cell MHC class I expression. Importantly, nonspecific T cell activation against the ONS 2303 medulloblastoma cell line can be reduced by tuning the valency of the αCD3 targeted monomer in the oligomerized CSAN. Intraperitoneal injections of αB7-H3-αCD3 bispecific CSANs were found to effectively cross the blood-tumor barrier into the brain and elicit significant antitumor T cell activity intracranially as well as systemically in an orthotopic medulloblastoma model. Moreover, following treatment with αB7-H3-αCD3 CSANs, intratumoral T cells were found to primarily have a central memory phenotype that displayed significant levels of characteristic activation markers. Collectively, these results demonstrate the ability of our multivalent, bispecific CSANs to direct potent antitumor T cell responses and indicate its potential utility as an alternative or complementary therapy for immune cell targeting of B7-H3+ brain tumors.
Collapse
Affiliation(s)
- Ellie A. Mews
- Department of Medicinal Chemistry, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6 St SE, Minneapolis, MN 55455 United States
| | - Pauline Beckmann
- Department of Pediatrics, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Malcolm Moos Tower, 515 Delaware St SE, Minneapolis, MN 55455 United States
| | - Mahathi Patchava
- Department of Pediatrics, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Malcolm Moos Tower, 515 Delaware St SE, Minneapolis, MN 55455 United States
| | - Yiao Wang
- Department of Medicinal Chemistry, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6 St SE, Minneapolis, MN 55455 United States
| | - David A. Largaespada
- Department of Pediatrics, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Malcolm Moos Tower, 515 Delaware St SE, Minneapolis, MN 55455 United States
| | - Carston R. Wagner
- Department of Medicinal Chemistry, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6 St SE, Minneapolis, MN 55455 United States
- Corresponding Author: Carston R Wagner: Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455 United States;
| |
Collapse
|