1
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
2
|
Asioli S, Gatto L, Vardy U, Agostinelli C, Di Nunno V, Righi S, Tosoni A, Ambrosi F, Bartolini S, Giannini C, Franceschi E. Immunophenotypic Profile of Adult Glioblastoma IDH-Wildtype Microenvironment: A Cohort Study. Cancers (Basel) 2024; 16:3859. [PMID: 39594814 PMCID: PMC11592556 DOI: 10.3390/cancers16223859] [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: 07/21/2024] [Revised: 10/15/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Glioblastoma IDH-wildtype (GBM IDH-wt) is the most aggressive brain tumor in adults and is characterized by an immunosuppressive microenvironment. Different factors shaping its tumor microenvironment (TME) regulate tumor progression and treatment response. The aim of this study was to characterize the main immunosuppressive elements of the GBM IDH-wt TME. Methods: Immunohistochemistry for CD3, CD4, CD8, CD163, programmed death ligand 1 (PD-L1) and programmed death 1 (PD1) was performed on surgical tumor specimens from patients diagnosed with GBM IDH-wt, according to the CNS WHO 2021 criteria. The impact of categorical variables on time-dependent outcomes such as overall survival (OS) and progression-free survival (PFS) has been estimated through the Kaplan-Meier method. Results: We included 30 patients (19 males and 11 females), median age of 59.8 years (range 40.2-69.1 years). All patients underwent surgery followed by temozolomide concurrent with and adjuvant to radiotherapy. MGMT was methylated in 14 patients (47%) and unmethylated in 16 patients (53%). The overall absolute percentages of CD4+ lymphocytes, both intratumoral and perivascular, were significantly more represented than CD8+ lymphocytes in the TME (p = 0.02). A low density of CD4+ lymphocytes (≤10%) was found to be a favorable prognostic factor for GBM outcome (p = 0.02). Patients with MGMT methylated and unmethylated tumors exhibited a distinct TME composition, with a significant higher number of perivascular CD8+ lymphocytes (p = 0.002), intratumoral CD8+ lymphocytes (p = 0.0024) and perivascular CD4+ lymphocytes (p = 0.014) in MGMT unmethylated tumors. PD-L1 expression in tumor cell surface was observed in four tumors (13.3%), and PD1 expression in infiltrating T lymphocytes was observed in nine (30%) tumors, with predominantly perivascular distribution. Conclusions: MGMT methylated and unmethylated tumors exhibit different immune profiles, likely reflecting the different biology of these tumors. The expression of PD-L1 in GBM IDH-wt patients is confined to a small subpopulation. While we found a significant association between low CD4+ lymphocyte density (≤10%) and survival, given the small numbers of our cohort, the prognostic value of CD4+ lymphocyte density will need to be validated in large-scale studies.
Collapse
Affiliation(s)
- Sofia Asioli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40127 Bologna, Italy; (S.A.); (C.G.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Lidia Gatto
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Uri Vardy
- School of Medicine and Surgery, University of Bologna, 40138 Bologna, Italy;
| | - Claudio Agostinelli
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Vincenzo Di Nunno
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Simona Righi
- Pathology Unit, Maggiore Hospital-AUSL Bologna, 40133 Bologna, Italy; (S.R.); (F.A.)
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Francesca Ambrosi
- Pathology Unit, Maggiore Hospital-AUSL Bologna, 40133 Bologna, Italy; (S.R.); (F.A.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Caterina Giannini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40127 Bologna, Italy; (S.A.); (C.G.)
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| |
Collapse
|
3
|
Ferrarese R, Joseph K, Andrieux G, Haase IV, Zanon F, Kling E, Izzo A, Corrales E, Schwabenland M, Prinz M, Ravi VM, Boerries M, Heiland DH, Carro MS. ZBTB18 regulates cytokine expression and affects microglia/macrophage recruitment and commitment in glioblastoma. Commun Biol 2024; 7:1472. [PMID: 39516530 PMCID: PMC11549471 DOI: 10.1038/s42003-024-07144-y] [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: 12/06/2023] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Glioma associated macrophages/microglia (GAMs) play an important role in glioblastoma (GBM) progression, due to their massive recruitment to the tumor site and polarization to a tumor promoting phenotype. GAMs secrete a variety of cytokines, which facilitate tumor cell growth and invasion, and prevent other immune cells from mounting an immune response against the tumor. Here, we demonstrate that zinc finger and BTB containing domain 18 (ZBTB18), a transcriptional repressor with tumor suppressive function in glioblastoma, impairs the production of key cytokines, which function as chemoattractant for GAMs. Consistently, we observe a reduced migration of GAMs when ZBTB18 is expressed by glioblastoma cells, both in cell culture and in vivo experiments. Moreover, RNA sequencing analysis shows that the presence of ZBTB18 in glioblastoma cells alters the commitment of conditioned microglia, suggesting the loss of the immune-suppressive phenotype and the acquisition of pro-inflammatory features. Thus, therapeutic approaches to increase ZBTB18 expression in GBM cells could represent an effective adjuvant to immune therapy in GBM.
Collapse
Affiliation(s)
- Roberto Ferrarese
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
- Laboratory of General Pathology and Immunology, University of Insubria, Varese, Italy
| | - Kevin Joseph
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ira Verena Haase
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Francesca Zanon
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Eva Kling
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Annalisa Izzo
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Eyleen Corrales
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Marius Schwabenland
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for NeuroModulation (NeuroModul), University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for NeuroModulation (NeuroModul), University of Freiburg, Freiburg, Germany
| | - Vidhya Madapusi Ravi
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center, University of Freiburg, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center, University of Freiburg, Freiburg, Germany
| | - Maria Stella Carro
- Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg, Germany.
- Laboratory of General Pathology and Immunology, University of Insubria, Varese, Italy.
| |
Collapse
|
4
|
Frederico SC, Raphael I, Nisnboym M, Huq S, Schlegel BT, Sneiderman CT, Jackson SA, Jain A, Olin MR, Rood BR, Pollack IF, Hwang EI, Rajasundaram D, Kohanbash G. Transcriptomic observations of intra and extracellular immunotherapy targets for pediatric brain tumors. Expert Rev Clin Immunol 2024; 20:1411-1420. [PMID: 39114885 DOI: 10.1080/1744666x.2024.2390023] [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: 05/14/2024] [Accepted: 08/04/2024] [Indexed: 08/20/2024]
Abstract
OBJECTIVES Despite surgical resection, chemoradiation, and targeted therapy, brain tumors remain a leading cause of cancer-related death in children. Immunotherapy has shown some promise and is actively being investigated for treating childhood brain tumors. However, a critical step in advancing immunotherapy for these patients is to uncover targets that can be effectively translated into therapeutic interventions. METHODS In this study, our team performed a transcriptomic analysis across pediatric brain tumor types to identify potential targets for immunotherapy. Additionally, we assessed components that may impact patient response to immunotherapy, including the expression of genes essential for antigen processing and presentation, inhibitory ligands and receptors, interferon signature, and overall predicted T cell infiltration. RESULTS We observed distinct expression patterns across tumor types. These included elevated expression of antigen genes and antigen processing machinery in some tumor types while other tumors had elevated inhibitory checkpoint receptors, known to be associated with response to checkpoint inhibitor immunotherapy. CONCLUSION These findings suggest that pediatric brain tumors exhibit distinct potential for specific immunotherapies. We believe our findings can guide investigators in their assessment of appropriate immunotherapy classes and targets in pediatric brain tumors.
Collapse
Affiliation(s)
- Stephen C Frederico
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michal Nisnboym
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Sakibul Huq
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brent T Schlegel
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chaim T Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sydney A Jackson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anya Jain
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R Olin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Brian R Rood
- Division of Oncology, Children's National Medical Center, Washington, DC, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugene I Hwang
- Division of Oncology, Children's National Medical Center, Washington, DC, USA
| | | | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Pike SC, Wiencke JK, Zhang Z, Molinaro AM, Hansen HM, Koestler DC, Christensen BC, Kelsey KT, Salas LA. Glioma immune microenvironment composition calculator (GIMiCC): a method of estimating the proportions of eighteen cell types from DNA methylation microarray data. Acta Neuropathol Commun 2024; 12:170. [PMID: 39468647 PMCID: PMC11514818 DOI: 10.1186/s40478-024-01874-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
A scalable platform for cell typing in the glioma microenvironment can improve tumor subtyping and immune landscape detection as successful immunotherapy strategies continue to be sought and evaluated. DNA methylation (DNAm) biomarkers for molecular classification of tumor subtypes have been developed for clinical use. However, tools that predict the cellular landscape of the tumor are not well-defined or readily available. We developed the Glioma Immune Microenvironment Composition Calculator (GIMiCC), an approach for deconvolution of cell types in gliomas using DNAm data. Using data from 17 isolated cell types, we describe the derivation of the deconvolution libraries in the biological context of selected genomic regions and validate deconvolution results using independent datasets. We utilize GIMiCC to illustrate that DNAm-based estimates of immune composition are clinically relevant and scalable for potential clinical implementation. In addition, we utilize GIMiCC to identify composition-independent DNAm alterations that are associated with high immune infiltration. Our future work aims to optimize GIMiCC and advance the clinical evaluation of glioma.
Collapse
Affiliation(s)
- Steven C Pike
- Integrative Neuroscience at Dartmouth, Guarini School of Graduate and Advanced Studies at Dartmouth College, Hanover, NH, USA
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Ze Zhang
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, Medical Center, University of Kansas, Kansas City, KS, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Karl T Kelsey
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Lucas A Salas
- Integrative Neuroscience at Dartmouth, Guarini School of Graduate and Advanced Studies at Dartmouth College, Hanover, NH, USA.
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA.
| |
Collapse
|
6
|
Gallus M, Young JS, Cook Quackenbush S, Khasraw M, de Groot J, Okada H. Chimeric antigen receptor T-cell therapy in patients with malignant glioma-From neuroimmunology to clinical trial design considerations. Neuro Oncol 2024:noae203. [PMID: 39450490 DOI: 10.1093/neuonc/noae203] [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] [Indexed: 10/26/2024] Open
Abstract
Clinical trials evaluating chimeric antigen receptor (CAR) T-cell therapy in patients with malignant gliomas have shown some early promise in pediatric and adult patients. However, the long-term benefits and safety for patients remain to be established. The ultimate success of CAR T-cell therapy for malignant glioma will require the integration of an in-depth understanding of the immunology of the central nervous system (CNS) parenchyma with strategies to overcome the paucity and heterogeneous expression of glioma-specific antigens. We also need to address the cold (immunosuppressive) microenvironment, exhaustion of the CAR T-cells, as well as local and systemic immunosuppression. Here, we discuss the basics and scientific considerations for CAR T-cell therapies and highlight recent clinical trials. To help identify optimal CAR T-cell administration routes, we summarize our current understanding of CNS immunology and T-cell homing to the CNS. We also discuss challenges and opportunities related to clinical trial design and patient safety/monitoring. Finally, we provide our perspective on future prospects in CAR T-cell therapy for malignant gliomas by discussing combinations and novel engineering strategies to overcome immuno-regulatory mechanisms. We hope this review will serve as a basis for advancing the field in a multiple discipline-based and collaborative manner.
Collapse
Affiliation(s)
- Marco Gallus
- Department of Neurological Surgery, Unversity of California San Fracisco, San Francisco, California, USA
| | - Jacob S Young
- Department of Neurological Surgery, Unversity of California San Fracisco, San Francisco, California, USA
| | | | - Mustafa Khasraw
- The Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - John de Groot
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
- Department of Neurological Surgery, Unversity of California San Fracisco, San Francisco, California, USA
| | - Hideho Okada
- The Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
- Department of Neurological Surgery, Unversity of California San Fracisco, San Francisco, California, USA
| |
Collapse
|
7
|
Zhao L, Shireman J, Probelsky S, Rigg B, Wang X, Huff WX, Kwon JH, Dey M. CCL21 Induces Plasmacytoid Dendritic Cell Migration and Activation in a Mouse Model of Glioblastoma. Cancers (Basel) 2024; 16:3459. [PMID: 39456552 PMCID: PMC11506458 DOI: 10.3390/cancers16203459] [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: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that are traditionally divided into two distinct subsets: myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). pDCs are known for their ability to secrete large amounts of cytokine type I interferons (IFN- α). In our previous work, we have demonstrated that pDC infiltration promotes glioblastoma (GBM) tumor immunosuppression through decreased IFN-α secretion via TLR-9 signaling and increased suppressive function of regulatory T cells (Tregs) via increased IL-10 secretion, resulting in poor overall outcomes in mouse models of GBM. Further dissecting the overall mechanism of pDC-mediated GBM immunosuppression, in this study, we identified CCL21 as highly upregulated by multiple GBM cell lines, which recruit pDCs to tumor sites via CCL21-CCR7 signaling. Furthermore, pDCs are activated by CCL21 in the GBM microenvironment through intracellular signaling of β-arrestin and CIITA. Finally, we found that CCL21-treated pDCs directly suppress CD8+ T cell proliferation without affecting regulatory T cells (Tregs) differentiation, which is considered the canonical pathway of immunotolerant regulation. Taken together, our results show that pDCs play a multifaced role in GBM immunosuppression, and CCL21 could be a novel therapeutic target in GBM to overcome pDC-mediated immunosuppression.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Jack Shireman
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Samantha Probelsky
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Bailey Rigg
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Xiaohu Wang
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Wei X. Huff
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Jae H. Kwon
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Mahua Dey
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| |
Collapse
|
8
|
Wang H, Yao L, Chen J, Li Y, Su Z, Liu Y, Li W, Xiong Y, Gao H, Zhang X, Zhou Y. The dual role of POSTN in maintaining glioblastoma stem cells and the immunosuppressive phenotype of microglia in glioblastoma. J Exp Clin Cancer Res 2024; 43:252. [PMID: 39227950 PMCID: PMC11373117 DOI: 10.1186/s13046-024-03175-9] [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: 06/14/2024] [Accepted: 08/24/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an immunosuppressive, universally lethal cancer driven by glioblastoma stem cells (GSCs). The interplay between GSCs and immunosuppressive microglia plays crucial roles in promoting the malignant growth of GBM; however, the molecular mechanisms underlying this crosstalk are unclear. This study aimed to investigate the role of POSTN in maintaining GSCs and the immunosuppressive phenotype of microglia. METHODS The expression of POSTN in GBM was identified via immunohistochemistry, quantitative real-time PCR, and immunoblotting. Tumorsphere formation assay, Cell Counting Kit-8 assay and immunofluorescence were used to determine the key role of POSTN in GSC maintenance. ChIP-seq and ChIP-PCR were conducted to confirm the binding sequences of β-catenin in the promoter region of FOSL1. Transwell migration assays, developmental and functional analyses of CD4+ T cells, CFSE staining and analysis, enzyme-linked immunosorbent assays and apoptosis detection tests were used to determine the key role of POSTN in maintaining the immunosuppressive phenotype of microglia and thereby promoting the immunosuppressive tumor microenvironment. Furthermore, the effects of POSTN on GSC maintenance and the immunosuppressive phenotype of microglia were investigated in a patient-derived xenograft model and orthotopic glioma mouse model, respectively. RESULTS Our findings revealed that POSTN secreted from GSCs promotes GSC self-renewal and tumor growth via activation of the αVβ3/PI3K/AKT/β-catenin/FOSL1 pathway. In addition to its intrinsic effects on GSCs, POSTN can recruit microglia and upregulate CD70 expression in microglia through the αVβ3/PI3K/AKT/NFκB pathway, which in turn promotes Treg development and functionality and supports the formation of an immunosuppressive tumor microenvironment. In both in vitro models and orthotopic mouse models of GBM, POSTN depletion disrupted GSC maintenance, decreased the recruitment of immunosuppressive microglia and suppressed GBM growth. CONCLUSION Our findings reveal that POSTN plays critical roles in maintaining GSCs and the immunosuppressive phenotype of microglia and provide a new therapeutic target for treating GBM.
Collapse
Affiliation(s)
- Hao Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Lin Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jinming Chen
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Yanyan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zuopeng Su
- Department of Neurosurgery, Minhang Hospital of Fudan University, Shanghai, 201199, China
| | - Yongsheng Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Wen Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yun Xiong
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Heyang Gao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xiao Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Youxin Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
9
|
Li L, Zhang T, Xiao M, Lu Y, Gao L. Brain macrophage senescence in glioma. Semin Cancer Biol 2024; 104-105:46-60. [PMID: 39098625 DOI: 10.1016/j.semcancer.2024.07.005] [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: 06/07/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Gliomas are a diverse group of primary central nervous system neoplasms with no curative therapies available. Brain macrophages comprise microglia in the brain parenchyma, border-associated macrophages in the meningeal-choroid plexus-perivascular space and monocyte-derived macrophages infiltrating the brain. With the great improvement of our recognition of brain macrophages, diverse macrophage populations have been found in the context of glioma, which exhibit functional and phenotypic heterogeneity. We have long thought that brain macrophage senescence is detrimental, manifested by specialized forms of persistent cell cycle arrest and chronic low-grade inflammation. Persistent senescence of macrophages may result in immune dysfunction, potentially contributing to glioma initiation and development. Given the crucial roles played by brain macrophages in glioma, we unravel how brain macrophages undergo reprogramming and their contribution to glioma. We outline general molecular alterations and specific biomarkers in senescent brain macrophages, as well as functional changes (such as metabolism, autophagy, phagocytosis, antigen presentation, and infiltration and recruitment). In addition, recent advances in genetic regulation and mechanisms linked to senescent brain macrophages are discussed. In particular, this review emphasizes the contribution of senescent brain macrophages to glioma, which may drive translational efforts to utilize brain macrophages as a prognostic marker or/and treatment target in glioma. An in-depth comprehending of how brain macrophage senescence functionally influences the tumor microenvironment will be key to our development of innovative therapeutics for glioma.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Tianhe Zhang
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China
| | - Meiling Xiao
- Department of Rehabilitation, The Central Hospital of Shenyang Medical College, Shenyang, Liaoning 110024, China
| | - Yu Lu
- Rehabilitation Medicine Department, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China.
| | - Lin Gao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
10
|
Li C, Niu C, Chen L, Yu B, Luo F, Qie J, Yang H, Qian J, Chu Y. Classical Monocytes Shuttling for Precise Delivery of Nanotherapeutics to Glioblastoma. Adv Healthc Mater 2024:e2400925. [PMID: 39212635 DOI: 10.1002/adhm.202400925] [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: 03/11/2024] [Revised: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor for which current therapies have limited efficacy. Immunosuppression and difficulties in accessing tumors with therapeutic agents are major obstacles for GBM treatments. Classical monocytes (CMs) possess the strongest infiltration among myeloid cells recruited into tumors during tumorigenesis. In this study, CMs are utilized to deliver the small-molecule CUDC-907 encapsulated in nanoparticles (907-NPs@CMs) for GBM therapy. Hitchhiking on CMs enables more 907-NPs to successfully penetrate the blood-brain barrier (BBB) and reach the interior of tumors. Results demonstrate that 907-NPs@CMs significantly improve the survival rates by suppressing tumor growth and reversing the immunosuppression of tumor microenvironment (TME). Furthermore, the high delivery efficiency of CMs reduces the amount of CUDC-907 required for treatments, reducing the physiological toxicity and off-target effects caused by high doses. 907-NPs@CMs is a safe and versatile therapeutic system that provides a platform for targeted drug delivery to tumors and the ability to treat GBM through a combination of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Congwen Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Congyi Niu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lin Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingbo Qie
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hui Yang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiawen Qian
- Shanghai Fifth People's Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Science, Fudan University, Shanghai, 200030, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, 200032, China
| |
Collapse
|
11
|
Han Y, Hu J, Pan J, Song X, Zhou Y, Zhang J, Yang Y, Shi X, Sun M, Yang J. LPS exposure alleviates multiple tissues damage by facilitating macrophage efferocytosis. Int Immunopharmacol 2024; 135:112283. [PMID: 38772299 DOI: 10.1016/j.intimp.2024.112283] [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: 04/02/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
Toll-like receptors (TLRs) play a crucial role in mediating immune responses by recognizing pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), as well as facilitating apoptotic cell (ACs) clearance (efferocytosis), thus contributing significantly to maintaining homeostasis and promoting tissue resolution. In this study, we investigate the impact of TLR agonists on macrophage efferocytosis. Our findings demonstrate that pretreatment with the TLR agonist lipopolysaccharide (LPS) significantly enhances macrophage phagocytic ability, thereby promoting efferocytosis both in vitro and in vivo. Moreover, LPS pretreatment confers tissue protection against damage by augmenting macrophage efferocytic capacity in murine models. Further examination reveals that LPS modulates efferocytosis by upregulating the expression of Tim4.These results underscore the pivotal role of TLR agonists in regulating the efferocytosis process and suggest potential therapeutic avenues for addressing inflammatory diseases. Overall, our study highlights the intricate interplay between LPS pretreatment and efferocytosis in maintaining tissue homeostasis and resolving inflammation.
Collapse
Affiliation(s)
- Yuwen Han
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Jiukun Hu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Jinlin Pan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Xueyan Song
- Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Yuanshuai Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Jun Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China
| | - Yue Yang
- Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China; Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, China
| | - Xiaohua Shi
- Department of Gastroenterology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 1 Lijiang Road, Suzhou 215153, China
| | - Minxuan Sun
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Jiangsu Key Lab of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Keling Road No. 88, Suzhou 215163, China.
| | - Jiao Yang
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, China.
| |
Collapse
|
12
|
Tian Z, Jia W, Wang Z, Mao H, Zhang J, Shi Q, Li X, Song S, Zhang J, Zhu Y, Yang B, Huang C, Huang J. Clinical significance of immune-related antigen CD58 in gliomas and analysis of its potential core related gene clusters. Heliyon 2024; 10:e29275. [PMID: 38699747 PMCID: PMC11063413 DOI: 10.1016/j.heliyon.2024.e29275] [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: 03/23/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Background The clinical significance of immune-related antigen CD58 in gliomas remains uncertain. The aim of this study was to examine the clinical importance and possible core related genes of CD58 in gliomas. Methods Pan-cancer analysis was to observe the association between CD58 and different tumors, glioma RNA sequencing data and clinical sample analyses were used to observe the relationship between CD58 and glioma, shRNA interference models were to observe the impact of CD58 on glioma cell function, and four glioma datasets and two online analysis platforms were used to explore the core related genes affecting the correlation between CD58 and glioma. Results High CD58 expression was associated with worse prognosis in various tumors and higher malignancy in glioma. Down regulation of CD58 expression was linked to decreased proliferation, increased apoptosis, and reduced metastasis in glioma cells. The pathways involved in CD58-related effects were enriched for immune cell adhesion and immune factor activation, and the core genes were CASP1, CCL2, IL18, MYD88, PTPRC, and TLR2. The signature of CD58 and its core-related genes showed superior predictive power for glioma prognosis. Conclusion High CD58 expression is correlated with more malignant glioma types, and also an independent risk factor for mortality in glioma. CD58 and its core-related genes may serve as novel biomarkers for diagnosing and treating glioma.
Collapse
Affiliation(s)
- Zhi Tian
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Wei Jia
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Zhao Wang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Hui Mao
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jingjing Zhang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Qiongya Shi
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Xing Li
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Shaoyu Song
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jiao Zhang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Yingjie Zhu
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Bo Yang
- Department of Pathology, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Chunhai Huang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008 PR China
| |
Collapse
|
13
|
Scuderi SA, Ardizzone A, Salako AE, Pantò G, De Luca F, Esposito E, Capra AP. Pentraxin 3: A Main Driver of Inflammation and Immune System Dysfunction in the Tumor Microenvironment of Glioblastoma. Cancers (Basel) 2024; 16:1637. [PMID: 38730589 PMCID: PMC11083335 DOI: 10.3390/cancers16091637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Brain tumors are a heterogeneous group of brain neoplasms that are highly prevalent in individuals of all ages worldwide. Within this pathological framework, the most prevalent and aggressive type of primary brain tumor is glioblastoma (GB), a subtype of glioma that falls within the IV-grade astrocytoma group. The death rate for patients with GB remains high, occurring within a few months after diagnosis, even with the gold-standard therapies now available, such as surgery, radiation, or a pharmaceutical approach with Temozolomide. For this reason, it is crucial to continue looking for cutting-edge therapeutic options to raise patients' survival chances. Pentraxin 3 (PTX3) is a multifunctional protein that has a variety of regulatory roles in inflammatory processes related to extracellular matrix (ECM). An increase in PTX3 blood levels is considered a trustworthy factor associated with the beginning of inflammation. Moreover, scientific evidence suggested that PTX3 is a sensitive and earlier inflammation-related marker compared to the short pentraxin C-reactive protein (CRP). In several tumoral subtypes, via regulating complement-dependent and macrophage-associated tumor-promoting inflammation, it has been demonstrated that PTX3 may function as a promoter of cancer metastasis, invasion, and stemness. Our review aims to deeply evaluate the function of PTX3 in the pathological context of GB, considering its pivotal biological activities and its possible role as a molecular target for future therapies.
Collapse
Affiliation(s)
- Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Ayomide Eniola Salako
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
- University of Florence, 50121 Florence, Italy
| | - Giuseppe Pantò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| |
Collapse
|
14
|
Wang H, Zhang X, Zhang Y, Shi T, Zhang Y, Song X, Liu B, Wang Y, Wei J. Targeting PCSK9 to upregulate MHC-II on the surface of tumor cells in tumor immunotherapy. BMC Cancer 2024; 24:445. [PMID: 38600469 PMCID: PMC11007992 DOI: 10.1186/s12885-024-12148-2] [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: 10/12/2023] [Accepted: 03/20/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9), the last member of the proprotein convertase family, functions as a classic regulator of low-density lipoprotein (LDL) by interacting with low-density lipoprotein receptor (LDLR). Recent studies have shown that PCSK9 can affect the occurrence and development of tumors and can be used as a novel therapeutic target. However, a comprehensive pan-cancer analysis of PCSK9 has yet to be conducted. METHODS The potential oncogenic effects of PCSK9 in 33 types of tumors were explored based on the datasets of The Cancer Genome Atlas (TCGA) dataset. In addition, the immune regulatory role of PCSK9 inhibition was evaluated via in vitro cell coculture and the tumor-bearing mouse model. Finally, the antitumor efficacy of targeted PCSK9 combined with OVA-II vaccines was verified. RESULTS Our results indicated that PCSK9 was highly expressed in most tumor types and was significantly correlated with late disease stage and poor prognosis. Additionally, PCSK9 may regulate the tumor immune matrix score, immune cell infiltration, immune checkpoint expression, and major histocompatibility complex expression. Notably, we first found that dendritic cell (DC) infiltration and major histocompatibility complex-II (MHC-II) expression could be upregulated by PCSK9 inhibition and improve CD8+ T cell activation in the tumor immune microenvironment, thereby achieving potent tumor control. Combining PCSK9 inhibitors could enhance the efficacies of OVA-II tumor vaccine monotherapy. CONCLUSIONS Conclusively, our pan-cancer analysis provided a more comprehensive understanding of the oncogenic and immunoregulatory roles of PCSK9 and demonstrated that targeting PCSK9 could increase the efficacy of long peptide vaccines by upregulating DC infiltration and MHC-II expression on the surface of tumor cells. This study reveals the critical oncogenic and immunoregulatory roles of PCSK9 in various tumors and shows the promise of PCSK9 as a potent immunotherapy target.
Collapse
Affiliation(s)
- Hanbing Wang
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Xin Zhang
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Yipeng Zhang
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Tao Shi
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Yue Zhang
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Xueru Song
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Baorui Liu
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China
| | - Yue Wang
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China.
| | - Jia Wei
- Department of Oncology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, No. 321, Zhongshan Road, 210008, Nanjing, China.
| |
Collapse
|
15
|
Yalcin F, Haneke H, Efe IE, Kuhrt LD, Motta E, Nickl B, Flüh C, Synowitz M, Dzaye O, Bader M, Kettenmann H. Tumor associated microglia/macrophages utilize GPNMB to promote tumor growth and alter immune cell infiltration in glioma. Acta Neuropathol Commun 2024; 12:50. [PMID: 38566120 PMCID: PMC10985997 DOI: 10.1186/s40478-024-01754-7] [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: 03/14/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Tumor-associated microglia and blood-derived macrophages (TAMs) play a central role in modulating the immune suppressive microenvironment in glioma. Here, we show that GPNMB is predominantly expressed by TAMs in human glioblastoma multiforme and the murine RCAS-PDGFb high grade glioma model. Loss of GPNMB in the in vivo tumor microenvironment results in significantly smaller tumor volumes and generates a pro-inflammatory innate and adaptive immune cell microenvironment. The impact of host-derived GPNMB on tumor growth was confirmed in two distinct murine glioma cell lines in organotypic brain slices from GPNMB-KO and control mice. Using published data bases of human glioma, the elevated levels in TAMs could be confirmed and the GPNMB expression correlated with a poorer survival.
Collapse
Affiliation(s)
- Fatih Yalcin
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute of Pathology, Christian-Albrecht University of Kiel, Kiel, Germany
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Hannah Haneke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ibrahim E Efe
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Leonard D Kuhrt
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Edyta Motta
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Bernadette Nickl
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Charlotte Flüh
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Omar Dzaye
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Helmut Kettenmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
16
|
Huang JH, Chen Y, Kang YB, Yao ZJ, Song JH. The potential crosstalk genes and molecular mechanisms between glioblastoma and periodontitis. Sci Rep 2024; 14:5970. [PMID: 38472293 DOI: 10.1038/s41598-024-56577-2] [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: 07/14/2023] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
Despite clinical and epidemiological evidence suggestive of a link between glioblastoma (GBM) and periodontitis (PD), the shared mechanisms of gene regulation remain elusive. In this study, we identify differentially expressed genes (DEGs) that overlap between the GEO datasets GSE4290 [GBM] and GSE10334 [PD]. Functional enrichment analysis was conducted, and key modules were identified using protein-protein interaction (PPI) network and weighted gene co-expression network analysis (WGCNA). The expression levels of CXCR4, LY96, and C3 were found to be significantly elevated in both the test dataset and external validation dataset, making them key crosstalk genes. Additionally, immune cell landscape analysis revealed elevated expression levels of multiple immune cells in GBM and PD compared to controls, with the key crosstalk genes negatively associated with Macrophages M2. FLI1 was identified as a potential key transcription factor (TF) regulating the three key crosstalk genes, with increased expression in the full dataset. These findings contribute to our understanding of the immune and inflammatory aspects of the comorbidity mechanism between GBM and PD.
Collapse
Affiliation(s)
- Jian-Huang Huang
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian, China.
| | - Yao Chen
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian, China
| | - Yuan-Bao Kang
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian, China
| | - Zheng-Jian Yao
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian, China
| | - Jian-Hua Song
- Department of Neurosurgery, Affiliated Hospital of Putian University, Putian, Fujian, China
| |
Collapse
|
17
|
Yu C, Hsieh K, Cherry DR, Nehlsen AD, Resende Salgado L, Lazarev S, Sindhu KK. Immune Escape in Glioblastoma: Mechanisms of Action and Implications for Immune Checkpoint Inhibitors and CAR T-Cell Therapy. BIOLOGY 2023; 12:1528. [PMID: 38132354 PMCID: PMC10741174 DOI: 10.3390/biology12121528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Glioblastoma, the most common primary brain cancer in adults, is characterized by a poor prognosis and resistance to standard treatments. The advent of immunotherapy has revolutionized the treatment of several cancers in recent years but has failed to demonstrate benefit in patients with glioblastoma. Understanding the mechanisms by which glioblastoma exerts tumor-mediated immune suppression in both the tumor microenvironment and the systemic immune landscape is a critical step towards developing effective immunotherapeutic strategies. In this review, we discuss the current understanding of immune escape mechanisms in glioblastoma that compromise the efficacy of immunotherapies, with an emphasis on immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy. In parallel, we review data from preclinical studies that have identified additional therapeutic targets that may enhance overall treatment efficacy in glioblastoma when administered alongside existing immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kunal K. Sindhu
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.Y.); (D.R.C.); (A.D.N.); (L.R.S.); (S.L.)
| |
Collapse
|
18
|
Xu M, Cheng Y, Meng R, Yang P, Chen J, Qiao Z, Wu J, Qian K, Li Y, Wang P, Zhou L, Wang T, Sheng D, Zhang Q. Enhancement of Microglia Functions by Developed Nano-Immuno-Synergist to Ameliorate Immunodeficiency for Malignant Glioma Treatment. Adv Healthc Mater 2023; 12:e2301861. [PMID: 37573475 DOI: 10.1002/adhm.202301861] [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: 06/12/2023] [Revised: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Resident microglia are key factors in mediating immunity against brain tumors, but the microglia in malignant glioma are functionally impaired. Little immunotherapy is explored to restore microglial function against glioma. Herein, oleanolic acid (OA) (microglia "restorer") and D PPA-1 peptide (immune checkpoint blockade) are integrated on a nano-immuno-synergist (D PAM@OA) to work coordinately. The self-assembled OA core is coated with macrophage membrane for efficient blood-brain barrier penetration and microglia targeting, on which D PPA-1 peptide is attached via acid-sensitive bonds for specific release in tumor microenvironment. With the enhanced accumulation of the dual drugs in their respective action sites, D PAM@OA effectively promotes the recruitment and activation of effector T cells by inhibiting aberrant activation of Signal transducer and activator of transcription (STAT-3) pathway in microglia, and assists activated effector T cells in killing tumor cells by blocking elevated immune checkpoint proteins in malignant glioma. Eventually, as adjuvant therapy, the rationally designed nano-immuno-synergist hinders malignant glioma progression and recurrence with or without temozolomide. The work demonstrates the feasibility of a nano-formulation for microglia-based immunotherapy, which may provide a new direction for the treatment of brain tumors.
Collapse
Affiliation(s)
- Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jian Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhen Qiao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
19
|
Silvin A, Qian J, Ginhoux F. Brain macrophage development, diversity and dysregulation in health and disease. Cell Mol Immunol 2023; 20:1277-1289. [PMID: 37365324 PMCID: PMC10616292 DOI: 10.1038/s41423-023-01053-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Brain macrophages include microglia in the parenchyma, border-associated macrophages in the meningeal-choroid plexus-perivascular space, and monocyte-derived macrophages that infiltrate the brain under various disease conditions. The vast heterogeneity of these cells has been elucidated over the last decade using revolutionary multiomics technologies. As such, we can now start to define these various macrophage populations according to their ontogeny and their diverse functional programs during brain development, homeostasis and disease pathogenesis. In this review, we first outline the critical roles played by brain macrophages during development and healthy aging. We then discuss how brain macrophages might undergo reprogramming and contribute to neurodegenerative disorders, autoimmune diseases, and glioma. Finally, we speculate about the most recent and ongoing discoveries that are prompting translational attempts to leverage brain macrophages as prognostic markers or therapeutic targets for diseases that affect the brain.
Collapse
Affiliation(s)
- Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Jiawen Qian
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, 94800, France.
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, 138648, Republic of Singapore.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, 169856, Singapore.
| |
Collapse
|
20
|
Sim J, Park J, Moon JS, Lim J. Dysregulation of inflammasome activation in glioma. Cell Commun Signal 2023; 21:239. [PMID: 37723542 PMCID: PMC10506313 DOI: 10.1186/s12964-023-01255-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/01/2023] [Indexed: 09/20/2023] Open
Abstract
Gliomas are the most common brain tumors characterized by complicated heterogeneity. The genetic, molecular, and histological pathology of gliomas is characterized by high neuro-inflammation. The inflammatory microenvironment in the central nervous system (CNS) has been closely linked with inflammasomes that control the inflammatory response and coordinate innate host defenses. Dysregulation of the inflammasome causes an abnormal inflammatory response, leading to carcinogenesis in glioma. Because of the clinical importance of the various physiological properties of the inflammasome in glioma, the inflammasome has been suggested as a promising treatment target for glioma management. Here, we summarize the current knowledge on the contribution of the inflammasomes in glioma and therapeutic insights. Video Abstract.
Collapse
Affiliation(s)
- JeongMin Sim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea
| | - JeongMan Park
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Jaejoon Lim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, 11160, Republic of Korea.
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University College of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam, 13496, Republic of Korea.
| |
Collapse
|
21
|
Tao JC, Yu D, Shao W, Zhou DR, Wang Y, Hou SQ, Deng K, Lin N. Interactions between microglia and glioma in tumor microenvironment. Front Oncol 2023; 13:1236268. [PMID: 37700840 PMCID: PMC10493873 DOI: 10.3389/fonc.2023.1236268] [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: 06/07/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023] Open
Abstract
Gliomas, the most prevalent primary tumors in the central nervous system, are marked by their immunosuppressive properties and consequent poor patient prognosis. Current evidence emphasizes the pivotal role of the tumor microenvironment in the progression of gliomas, largely attributed to tumor-associated macrophages (brain-resident microglia and bone marrow-derived macrophages) that create a tumor microenvironment conducive to the growth and invasion of tumor cells. Yet, distinguishing between these two cell subgroups remains a challenge. Thus, our review starts by analyzing the heterogeneity between these two cell subsets, then places emphasis on elucidating the complex interactions between microglia and glioma cells. Finally, we conclude with a summary of current attempts at immunotherapy that target microglia. However, given that independent research on microglia is still in its initial stages and has many shortcomings at the present time, we express our related concerns and hope that further research will be carried out to address these issues in the future.
Collapse
Affiliation(s)
- Jin-Cheng Tao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dong Yu
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Wei Shao
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Dong-Rui Zhou
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Yu Wang
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Shi-Qiang Hou
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Ke Deng
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Lin
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui, China
| |
Collapse
|
22
|
Castillo SP, Galvez-Cancino F, Liu J, Pollard SM, Quezada SA, Yuan Y. The tumour ecology of quiescence: Niches across scales of complexity. Semin Cancer Biol 2023; 92:139-149. [PMID: 37037400 DOI: 10.1016/j.semcancer.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/06/2023] [Accepted: 04/08/2023] [Indexed: 04/12/2023]
Abstract
Quiescence is a state of cell cycle arrest, allowing cancer cells to evade anti-proliferative cancer therapies. Quiescent cancer stem cells are thought to be responsible for treatment resistance in glioblastoma, an aggressive brain cancer with poor patient outcomes. However, the regulation of quiescence in glioblastoma cells involves a myriad of intrinsic and extrinsic mechanisms that are not fully understood. In this review, we synthesise the literature on quiescence regulatory mechanisms in the context of glioblastoma and propose an ecological perspective to stemness-like phenotypes anchored to the contemporary concepts of niche theory. From this perspective, the cell cycle regulation is multiscale and multidimensional, where the niche dimensions extend to extrinsic variables in the tumour microenvironment that shape cell fate. Within this conceptual framework and powered by ecological niche modelling, the discovery of microenvironmental variables related to hypoxia and mechanosignalling that modulate proliferative plasticity and intratumor immune activity may open new avenues for therapeutic targeting of emerging biological vulnerabilities in glioblastoma.
Collapse
Affiliation(s)
- Simon P Castillo
- Centre for Evolution and Cancer & Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Felipe Galvez-Cancino
- Immune Regulation and Tumor Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Jiali Liu
- Immune Regulation and Tumor Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine and Cancer Research UK Scotland Centre, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sergio A Quezada
- Immune Regulation and Tumor Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer & Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK.
| |
Collapse
|
23
|
Roddy AC, McInerney CE, Flannery T, Healy EG, Stewart JP, Spence VJ, Walsh J, Salto-Tellez M, McArt DG, Prise KM. Transcriptional Profiling of a Patient-Matched Cohort of Glioblastoma (IDH-Wildtype) for Therapeutic Target and Repurposing Drug Identification. Biomedicines 2023; 11:biomedicines11041219. [PMID: 37189838 DOI: 10.3390/biomedicines11041219] [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: 03/30/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive adult brain tumor. Despite multi-modal therapies, GBM recurs, and patients have poor survival (~14 months). Resistance to therapy may originate from a subpopulation of tumor cells identified as glioma-stem cells (GSC), and new treatments are urgently needed to target these. The biology underpinning GBM recurrence was investigated using whole transcriptome profiling of patient-matched initial and recurrent GBM (recGBM). Differential expression analysis identified 147 significant probes. In total, 24 genes were validated using expression data from four public cohorts and the literature. Functional analyses revealed that transcriptional changes to recGBM were dominated by angiogenesis and immune-related processes. The role of MHC class II proteins in antigen presentation and the differentiation, proliferation, and infiltration of immune cells was enriched. These results suggest recGBM would benefit from immunotherapies. The altered gene signature was further analyzed in a connectivity mapping analysis with QUADrATiC software to identify FDA-approved repurposing drugs. Top-ranking target compounds that may be effective against GSC and GBM recurrence were rosiglitazone, nizatidine, pantoprazole, and tolmetin. Our translational bioinformatics pipeline provides an approach to identify target compounds for repurposing that may add clinical benefit in addition to standard therapies against resistant cancers such as GBM.
Collapse
Affiliation(s)
- Aideen C Roddy
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Caitríona E McInerney
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Tom Flannery
- Department of Neurosurgery, Royal Victoria Hospital, Belfast Health and Social Care Trust, Belfast BT12 6BA, UK
| | - Estelle G Healy
- Regional Service for Neuropathology, Institute of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust, Belfast BT12 6BA, UK
| | - James P Stewart
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Veronica J Spence
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Jamie Walsh
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Manuel Salto-Tellez
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
- Integrated Pathology Unit, Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Darragh G McArt
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Kevin M Prise
- Patrick G. Johnson Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, UK
| |
Collapse
|
24
|
Arrieta VA, Dmello C, McGrail DJ, Brat DJ, Lee-Chang C, Heimberger AB, Chand D, Stupp R, Sonabend AM. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Invest 2023; 133:e163447. [PMID: 36647828 PMCID: PMC9843050 DOI: 10.1172/jci163447] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Immune checkpoint blockade (ICB) has revolutionized modern cancer therapy, arousing great interest in the neuro-oncology community. While several reports show that subsets of patients with glioma exhibit durable responses to immunotherapy, the efficacy of this treatment has not been observed for unselected patient populations, preventing its broad clinical implementation for gliomas and glioblastoma (GBM). To exploit the maximum therapeutic potential of ICB for patients with glioma, understanding the different aspects of glioma-related tumor immune responses is of critical importance. In this Review, we discuss contributing factors that distinguish subsets of patients with glioma who may benefit from ICB. Specifically, we discuss (a) the complex interaction between the tumor immune microenvironment and glioma cells as a potential influence on immunotherapy responses; (b) promising biomarkers for responses to immune checkpoint inhibitors; and (c) the potential contributions of peripheral immune cells to therapeutic responses.
Collapse
Affiliation(s)
- Víctor A. Arrieta
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Program of Combined Studies in Medicine (PECEM), Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel J. McGrail
- Center for Immunotherapy and Precision Immuno-Oncology and
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Daniel J. Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dhan Chand
- Agenus Bio, Lexington, Massachusetts, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
25
|
Takacs GP, Kreiger CJ, Luo D, Tian G, Garcia JS, Deleyrolle LP, Mitchell DA, Harrison JK. Glioma-derived CCL2 and CCL7 mediate migration of immune suppressive CCR2 +/CX3CR1 + M-MDSCs into the tumor microenvironment in a redundant manner. Front Immunol 2023; 13:993444. [PMID: 36685592 PMCID: PMC9854274 DOI: 10.3389/fimmu.2022.993444] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized in part by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive, hematopoietic cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a potent subset of myeloid cells, expressing monocytic (M)-MDSC markers, distinguished by dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate into the TME. This study evaluated the T cell suppressive function and migratory properties of CCR2+/CX3CR1+ MDSCs. Bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Recombinant and glioma-derived CCL2 and CCL7 induce the migration of CCR2+/CX3CR1+ MDSCs with similar efficacy. KR158B-CCL2 and -CCL7 knockdown murine gliomas contain equivalent percentages of CCR2+/CX3CR1+ MDSCs compared to KR158B gliomas. Combined neutralization of CCL2 and CCL7 completely blocks CCR2-expressing cell migration to KR158B cell conditioned media. CCR2+/CX3CR1+ cells are also reduced within KR158B gliomas upon combination targeting of CCL2 and CCL7. High levels of CCL2 and CCL7 are also associated with negative prognostic outcomes in GBM patients. These data provide a more comprehensive understanding of the function of CCR2+/CX3CR1+ MDSCs and the role of CCL2 and CCL7 in the recruitment of these immune suppressive cells and further support the significance of targeting this chemokine axis in GBM.
Collapse
Affiliation(s)
- Gregory P. Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christian J. Kreiger
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Defang Luo
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Guimei Tian
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Julia S. Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Loic P. Deleyrolle
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Duane A. Mitchell
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
26
|
Microglia and Brain Macrophages as Drivers of Glioma Progression. Int J Mol Sci 2022; 23:ijms232415612. [PMID: 36555253 PMCID: PMC9779147 DOI: 10.3390/ijms232415612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Evidence is accumulating that the tumour microenvironment (TME) has a key role in the progression of gliomas. Non-neoplastic cells in addition to the tumour cells are therefore finding increasing attention. Microglia and other glioma-associated macrophages are at the centre of this interest especially in the context of therapeutic considerations. New ideas have emerged regarding the role of microglia and, more recently, blood-derived brain macrophages in glioblastoma (GBM) progression. We are now beginning to understand the mechanisms that allow malignant glioma cells to weaken microglia and brain macrophage defence mechanisms. Surface molecules and cytokines have a prominent role in microglia/macrophage-glioma cell interactions, and we discuss them in detail. The involvement of exosomes and microRNAs forms another focus of this review. In addition, certain microglia and glioma cell pathways deserve special attention. These "synergistic" (we suggest calling them "Janus") pathways are active in both glioma cells and microglia/macrophages where they act in concert supporting malignant glioma progression. Examples include CCN4 (WISP1)/Integrin α6β1/Akt and CHI3L1/PI3K/Akt/mTOR. They represent attractive therapeutic targets.
Collapse
|
27
|
Bottlenecks and opportunities in immunotherapy for glioma: a narrative review. JOURNAL OF BIO-X RESEARCH 2022. [DOI: 10.1097/jbr.0000000000000135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Helbig D, Klein S. Immune checkpoint inhibitors for unresectable or metastatic pleomorphic dermal sarcomas. Front Oncol 2022; 12:975342. [PMID: 36465341 PMCID: PMC9712951 DOI: 10.3389/fonc.2022.975342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/03/2022] [Indexed: 04/04/2024] Open
Abstract
Pleomorphic dermal sarcomas (PDS) are rare neoplasms of the skin that occur in UV-exposed sites in the elderly, but represent the most common cutaneous sarcomas. Although the majority of PDS can be surgically removed, local recurrences occur in up to 28%, usually occurring within the first two years after primary excision. Metastases are diagnosed in up to 20% of cases, mainly observed in the skin, lymph nodes and lungs, preferentially affecting patients with underlying hemato-oncologic diseases. Similar to other UV-induced tumors, PDS are inflammatory and immunogenic tumors (with a high number of CD4+/CD8+ tumor-infiltrating lymphocytes (TILs) and checkpoint molecule expression such as PD-L1, LAG-3, TIGIT) with a very high mutational burden. The most common genetic alterations include UV-induced TP53 loss of function mutations, followed by alterations in the CDKN2A/B gene. Rarely, targetable genetic alterations can be detected. Compelling experimental data and clinical reports about PD-1/PD-L1-blocking antibodies in patients with PDS suggest its use as first line treatment in unresectable or metastatic tumor stages. However, individual ("off-line") patient management should be discussed in an interdisciplinary tumor board based on molecular genetic testing, mutational burden, PD-L1 expression, and evidence of tumor-infiltrating lymphocytes in addition to comorbities of the individual patient.
Collapse
Affiliation(s)
- Doris Helbig
- Department of Dermatology, University Hospital Cologne, Cologne, Germany
| | - Sebastian Klein
- Department of Hematology and Stem Cell Transplantation, University Duisburg-Essen, University Hospital Essen, Essen, Germany
| |
Collapse
|
29
|
Wang H, Feng Y, Zhang Y, Wang T, Xu H, Zhi Y, Feng Y, Tian L, Yuan K. Siglec10-An immunosuppressor and negative predictor of survival prognosis in gliomas. Front Genet 2022; 13:873655. [PMID: 36468012 PMCID: PMC9709431 DOI: 10.3389/fgene.2022.873655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 10/28/2022] [Indexed: 07/24/2023] Open
Abstract
Glioma is a type of tumor occurring in the central nervous system. In recent decades, specific gene mutations and molecular aberrations have been used to conduct the glioma classification and clinical decisions. Siglec10 is a member of the sialic acid-binding immunoglobulin superfamily. In this study, we investigated the expression and functions of siglec10 in gliomas. We analyzed the siglec10 expression in glioma patients with immunohistochemical (IHC) staining and evaluated the survival prognosis. The high siglec10 expression had a shorter survival prognosis than the low siglec10 expression in patients, especially in malignant gliomas. Bioinformatic datasets, including TCGA and CGGA, validated the IHC results and discovered the expression of siglec10 was higher in the malignant subtype than a benign subtype of gliomas. So, siglec10 is associated with the poor prognosis of gliomas. Furthermore, the related mechanisms of siglec10 in gliomas were investigated by functional enrichment analysis, including GSEA, GO, and KEGG analysis. Siglec10 was correlated with inflammatory mediators, inflammatory cells, and inflammatory pathways in gliomas. Siglec10 might take part in the immune response in the tumor microenvironment to induce glioma's progression and metastasis. This study showed siglec10 was a biomarker in glioma, and it might be the potential target of glioma immunotherapy in the future.
Collapse
Affiliation(s)
- Hesong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yanyan Feng
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yuxiang Zhang
- Department of Neurosurgery, Sanbo Brain Hospital Capital Medical University, Beijing, China
| | - Ting Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Heng Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuxing Zhi
- Qi-Huang Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyin Feng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Lichun Tian
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Yuan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Pang L, Khan F, Heimberger AB, Chen P. Mechanism and therapeutic potential of tumor-immune symbiosis in glioblastoma. Trends Cancer 2022; 8:839-854. [PMID: 35624002 PMCID: PMC9492629 DOI: 10.1016/j.trecan.2022.04.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal form of brain tumor in human adults. Myeloid-lineage cells, including macrophages, microglia, myeloid-derived suppressor cells (MDSCs), and neutrophils, are the most frequent types of cell in the GBM tumor microenvironment (TME) that contribute to tumor progression. Emerging experimental evidence indicates that symbiotic interactions between cancer cells and myeloid cells are critical for tumor growth and immunotherapy resistance in GBM. In this review, we discuss the molecular mechanisms whereby cancer cells shape a myeloid cell-mediated immunosuppressive TME and, reciprocally, how such myeloid cells affect tumor progression and immunotherapy efficiency in GBM. Moreover, we highlight tumor-T cell symbiosis and summarize immunotherapeutic strategies intercepting this co-dependency in GBM.
Collapse
Affiliation(s)
- Lizhi Pang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Fatima Khan
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amy B Heimberger
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
31
|
Microglia-T cell conversations in brain cancer progression. Trends Mol Med 2022; 28:951-963. [PMID: 36075812 DOI: 10.1016/j.molmed.2022.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 12/26/2022]
Abstract
The highly immunosuppressive and heterogeneous milieu of brain malignancies contributes to their dismal prognosis. Regardless of their cellular origin, brain tumors grow in an environment with various specialized organ-resident cells. Although homeostatic microglia contribute to a healthy brain, conversations between disease-associated microglia and T cells compromise their individual and collective capacity to curb malignant growth. We review the mechanisms of T cell-microglia interactions and discuss how their collaboration fosters heterogeneity and immunosuppression in brain cancers. Because of the importance of microglia and T cells in the brain tumor microenvironment, it is crucial to understand their interactions to derive innovative therapeutics.
Collapse
|
32
|
The Yin and Yang of toll-like receptors in endothelial dysfunction. Int Immunopharmacol 2022; 108:108768. [DOI: 10.1016/j.intimp.2022.108768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
|
33
|
Man SM, Jenkins BJ. Context-dependent functions of pattern recognition receptors in cancer. Nat Rev Cancer 2022; 22:397-413. [PMID: 35355007 DOI: 10.1038/s41568-022-00462-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 02/07/2023]
Abstract
The immune system plays a critical role in shaping all facets of cancer, from the early initiation stage through to metastatic disease and resistance to therapy. Our understanding of the importance of the adaptive arm of the immune system in antitumour immunity has led to the implementation of immunotherapy with immune checkpoint inhibitors in numerous cancers, albeit with differing efficacy. By contrast, the clinical utility of innate immunity in cancer has not been exploited, despite dysregulated innate immunity being a feature of at least one-third of all cancers associated with tumour-promoting chronic inflammation. The past two decades have seen innate immune pattern recognition receptors (PRRs) emerge as critical regulators of the immune response to microbial infection and host tissue damage. More recently, it has become apparent that in many cancer types, PRRs play a central role in modulating a vast array of tumour-inhibiting and tumour-promoting cellular responses both in immune cells within the tumour microenvironment and directly in cancer cells. Herein, we provide a comprehensive overview of the fast-evolving field of PRRs in cancer, and discuss the potential to target PRRs for drug development and biomarker discovery in a wide range of oncology settings.
Collapse
Affiliation(s)
- Si Ming Man
- Division of Immunity, Inflammation and Infection, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
34
|
Lin YJ, Wu CYJ, Wu JY, Lim M. The Role of Myeloid Cells in GBM Immunosuppression. Front Immunol 2022; 13:887781. [PMID: 35711434 PMCID: PMC9192945 DOI: 10.3389/fimmu.2022.887781] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Gliomas are intrinsic brain tumors that originate from glial cells. Glioblastoma (GBM) is the most aggressive glioma type and resistant to immunotherapy, mainly due to its unique immune environment. Dimensional data analysis reveals that the intra-tumoral heterogeneity of immune cell populations in the glioma microenvironment is largely made up of cells of myeloid lineage. Conventional therapies of combined surgery, chemotherapy and radiotherapy have achieved limited improvements in the prognosis of glioma patients, as myeloid cells are prominent mediators of immune and therapeutic responses—like immunotherapy resistance—in glioma. Myeloid cells are frequently seen in the tumor microenvironment (TME), and they are polarized to promote tumorigenesis and immunosuppression. Reprogramming myeloid cells has emerged as revolutionary, new types of immunotherapies for glioma treatment. Here we detail the current advances in classifying epigenetic, metabolic, and phenotypic characteristics and functions of different populations of myeloid cells in glioma TME, including myeloid-derived suppressor cells (MDSCs), glioma-associated microglia/macrophages (GAMs), glioma-associated neutrophils (GANs), and glioma-associated dendritic cells (GADCs), as well as the mechanisms underlying promotion of tumorigenesis. The final goal of this review will be to provide new insights into novel therapeutic approaches for specific targeting of myeloid cells to improve the efficacy of current treatments in glioma patients.
Collapse
Affiliation(s)
- Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Medical Foundation, Linkou Medical Center, Taoyuan, Taiwan.,Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Neurosurgery, Chang Gung Medical Foundation, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Janet Yuling Wu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
35
|
Xia H, Deng L, Meng S, Liu X, Zheng C. Single-Cell Transcriptome Profiling Signatures and Alterations of Microglia Associated With Glioblastoma Associate Microglia Contribution to Tumor Formation. Pathol Oncol Res 2022; 28:1610067. [PMID: 35693633 PMCID: PMC9176381 DOI: 10.3389/pore.2022.1610067] [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: 09/08/2021] [Accepted: 05/05/2022] [Indexed: 11/13/2022]
Abstract
Glioblastoma (GBM), which occasionally occurs in pediatric patients, is the most common tumor of the central nervous system in adults. Clinically, GBM is classified as low-grade to high-grade (from 1 to 4) and is characterized by late discovery, limited effective treatment methods, and poor efficacy. With the development of immunotherapy technology, effective GBM treatment strategies are of great significance. The main immune cells found in the GBM tumor microenvironment are macrophages and microglia (MG). Both these monocytes play important roles in the occurrence and development of GBM. Macrophages are recruited during tumorigenesis, whereas MG is present in the brain during embryonic development. Interestingly, the accumulation of these monocytes is inversely proportional to the survival of adult GBM patients but not the pediatric GBM patients. This study used single-cell RNA-seq data to reveal the heterogeneity of MG in tumor lesions and to explore the role of different MG subtypes in the occurrence and development of GBM. The results may help find new targets for immunotherapy of GBM.
Collapse
Affiliation(s)
- Hailong Xia
- Department of Neurosurgery, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Lei Deng
- Department of Neurosurgery, Bishan District People's Hospital, Chongqing, China
| | - Shu Meng
- Internal Medicine, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Xipeng Liu
- Department of Neurosurgery, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Chao Zheng
- Department of Neurosurgery, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| |
Collapse
|
36
|
Microbial-Derived Toll-like Receptor Agonism in Cancer Treatment and Progression. Cancers (Basel) 2022; 14:cancers14122923. [PMID: 35740589 PMCID: PMC9221178 DOI: 10.3390/cancers14122923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Toll like receptors (TLRs) are a group of transmembrane receptors belonging to the class of pattern recognition receptors (PRR), which are involved in recognition of pathogen associated molecular patterns (PAMPs), inducing immune response. During the past decade, a number of preclinical and clinical breakthroughs in the field of TLR agonists has immerged in cancer research and some of these agents have performed exceptionally well in clinical trials. Based on evidence from scientific studies, we draw attention to several microbial based TLR agonists and discuss their relevance in various cancer and explore various microbial based TLR agonists for developing effective immunotherapeutic strategies against cancer. Abstract Toll-like receptors (TLRs) are typical transmembrane proteins, which are essential pattern recognition receptors in mediating the effects of innate immunity. TLRs recognize structurally conserved molecules derived from microbes and damage-associated molecular pattern molecules that play an important role in inflammation. Since the first discovery of the Toll receptor by the team of J. Hoffmann in 1996, in Drosophila melanogaster, numerous TLRs have been identified across a wide range of invertebrate and vertebrate species. TLR stimulation leads to NF-κB activation and the subsequent production of pro-inflammatory cytokines and chemokines, growth factors and anti-apoptotic proteins. The expression of TLRs has also been observed in many tumors, and their stimulation results in tumor progression or regression, depending on the TLR and tumor type. The anti-tumoral effects can result from the activation of anti-tumoral immune responses and/or the direct induction of tumor cell death. The pro-tumoral effects may be due to inducing tumor cell survival and proliferation or by acting on suppressive or inflammatory immune cells in the tumor microenvironment. The aim of this review is to draw attention to the effects of TLR stimulation in cancer, the activation of various TLRs by microbes in different types of tumors, and, finally, the role of TLRs in anti-cancer immunity and tumor rejection.
Collapse
|
37
|
Menna G, Mattogno PP, Donzelli CM, Lisi L, Olivi A, Della Pepa GM. Glioma-Associated Microglia Characterization in the Glioblastoma Microenvironment through a 'Seed-and Soil' Approach: A Systematic Review. Brain Sci 2022; 12:718. [PMID: 35741603 PMCID: PMC9220868 DOI: 10.3390/brainsci12060718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background and aim: Ever since the discovery of tumor-associated immune cells, there has been growing interest in the understanding of the mechanisms underlying the crosstalk between these cells and tumor cells. A "seed and soil" approach has been recently introduced to describe the glioblastoma (GBM) landscape: tumor microenvironments act as fertile "soil" and interact with the "seed" (glial and stem cells compartment). In the following article, we provide a systematic review of the current evidence pertaining to the characterization of glioma-associated macrophages and microglia (GAMs) and microglia and macrophage cells in the glioma tumor microenvironment (TME). Methods: An online literature search was launched on PubMed Medline and Scopus using the following research string: "((Glioma associated macrophages OR GAM OR Microglia) AND (glioblastoma tumor microenvironment OR TME))". The last search for articles pertinent to the topic was conducted in February 2022. Results: The search of the literature yielded a total of 349 results. A total of 235 studies were found to be relevant to our research question and were assessed for eligibility. Upon a full-text review, 58 articles were included in the review. The reviewed papers were further divided into three categories based on their focus: (1) Microglia maintenance of immunological homeostasis and protection against autoimmunity; (2) Microglia crosstalk with dedifferentiated and stem-like glioblastoma cells; (3) Microglia migratory behavior and its activation pattern. Conclusions: Aggressive growth, inevitable recurrence, and scarce response to immunotherapies are driving the necessity to focus on the GBM TME from a different perspective to possibly disentangle its role as a fertile 'soil' for tumor progression and identify within it feasible therapeutic targets. Against this background, our systematic review confirmed microglia to play a paramount role in promoting GBM progression and relapse after treatments. The correct and extensive understanding of microglia-glioma crosstalk could help in understanding the physiopathology of this complex disease, possibly opening scenarios for improvement of treatments.
Collapse
Affiliation(s)
- Grazia Menna
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Pier Paolo Mattogno
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Carlo Maria Donzelli
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Lucia Lisi
- Institute of Pharmacology, Catholic University of Rome, 00168 Rome, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Giuseppe Maria Della Pepa
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| |
Collapse
|
38
|
Eisemann T, Wechsler-Reya RJ. Coming in from the cold: overcoming the hostile immune microenvironment of medulloblastoma. Genes Dev 2022; 36:514-532. [PMID: 35680424 PMCID: PMC9186392 DOI: 10.1101/gad.349538.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Medulloblastoma is an aggressive brain tumor that occurs predominantly in children. Despite intensive therapy, many patients die of the disease, and novel therapies are desperately needed. Although immunotherapy has shown promise in many cancers, the low mutational burden, limited infiltration of immune effector cells, and immune-suppressive microenvironment of medulloblastoma have led to the assumption that it is unlikely to respond to immunotherapy. However, emerging evidence is challenging this view. Here we review recent preclinical and clinical studies that have identified mechanisms of immune evasion in medulloblastoma, and highlight possible therapeutic interventions that may give new hope to medulloblastoma patients and their families.
Collapse
Affiliation(s)
- Tanja Eisemann
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA.,Department of Pediatrics, University of California at San Diego, La Jolla, California 92161, USA
| |
Collapse
|
39
|
Xiao K, Zhao S, Yuan J, Pan Y, Song Y, Tang L. Construction of Molecular Subtypes and Related Prognostic and Immune Response Models Based on M2 Macrophages in Glioblastoma. Int J Gen Med 2022; 15:913-926. [PMID: 35115817 PMCID: PMC8801375 DOI: 10.2147/ijgm.s343152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To identify the molecular subtypes of glioblastoma multiforme (GBM) related to M2 macrophage-based prognostic genes, then to preliminarily explore their biological functions and construct immunotherapy response gene models. MATERIAL AND METHODS We used R language to analyze GBM microarray data, and other tools, including xCell and CIBERSORTx, to identify subtypes of GBM that related to M2 macrophages. The process started with the exploration of biological functions of the two subtypes by pathway analyses and GSEA, and continued with a combined procedure of constructing an M2 macrophage-related prognostic gene model and exploring the immune treatment response for GBM. RESULTS A high abundance of M2 macrophages in GBM was associated with poor prognosis. According to M2 macrophage-related prognostic genes, GBM was divided into two subtypes (cluster A and cluster B). The differential gene enrichment analysis of the two clusters showed that cluster A was less enriched in M2 macrophages and had immunopotential. The M2score, which was constructed based on M2 macrophage-related prognostic genes, was not only related to the survival and prognosis of patients with GBM, but also predictive of the effectiveness of immunotherapy in these patients. This result has been effectively verified in an external data set. CONCLUSION GBM was successfully divided into two subtypes according to M2-macrophage-related prognostic genes. In GBM, a high M2score may indicate better clinical outcome and enhancement of the immunotherapy response.
Collapse
Affiliation(s)
- Kai Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Shushan Zhao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Jian Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yimin Pan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Ya Song
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
40
|
Pediatric glioblastoma: mechanisms of immune evasion and potential therapeutic opportunities. Cancer Immunol Immunother 2022; 71:1813-1822. [PMID: 35020009 DOI: 10.1007/s00262-021-03131-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
Pediatric glioblastoma is relatively rare compared with its adult counterpart but is associated with a similarly grim prognosis. Available data indicate that pediatric glioblastomas are molecularly distinct from adult tumors, and relatively little is known about the pediatric glioblastoma tumor microenvironment (TME). Cancer immunotherapy has emerged as a new pillar of cancer treatment and is revolutionizing the care of patients with many advanced solid tumors, including melanoma, non-small cell lung cancer, head and neck cancer, and renal cell carcinoma. Unfortunately, attempts to treat adult glioblastoma with current immunotherapies have had limited success to date. Nevertheless, the immune milieu in pediatric glioblastoma is distinct from that found in adult tumors, and evidence suggests that pediatric tumors are less immunosuppressive. As a result, immunotherapies should be specifically evaluated in the pediatric context. The purpose of this review is to explore known and emerging mechanisms of immune evasion in pediatric glioblastoma and highlight potential opportunities for implementing immunotherapy in the treatment of these devastating pediatric brain tumors.
Collapse
|
41
|
ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent glioblastoma. NATURE CANCER 2021; 2:1372-1386. [PMID: 35121903 PMCID: PMC8818262 DOI: 10.1038/s43018-021-00260-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022]
Abstract
Only a subset of recurrent glioblastoma (rGBM) responds to anti-PD-1 immunotherapy. Previously, we reported enrichment of BRAF/PTPN11 mutations in 30% of rGBM that responded to PD-1 blockade. Given that BRAF and PTPN11 promote MAPK/ERK signaling, we investigated whether activation of this pathway is associated with response to PD-1 inhibitors in rGBM, including patients that do not harbor BRAF/PTPN11 mutations. Here we show that immunohistochemistry for ERK1/2 phosphorylation (p-ERK), a marker of MAPK/ERK pathway activation, is predictive of overall survival following adjuvant PD-1 blockade in two independent rGBM patient cohorts. Single-cell RNA-sequencing and multiplex immunofluorescence analyses revealed that p-ERK was mainly localized in tumor cells and that high-p-ERK GBMs contained tumor-infiltrating myeloid cells and microglia with elevated expression of MHC class II and associated genes. These findings indicate that ERK1/2 activation in rGBM is predictive of response to PD-1 blockade and is associated with a distinct myeloid cell phenotype.
Collapse
|
42
|
Himes BT, Geiger PA, Ayasoufi K, Bhargav AG, Brown DA, Parney IF. Immunosuppression in Glioblastoma: Current Understanding and Therapeutic Implications. Front Oncol 2021; 11:770561. [PMID: 34778089 PMCID: PMC8581618 DOI: 10.3389/fonc.2021.770561] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults an carries and carries a terrible prognosis. The current regiment of surgical resection, radiation, and chemotherapy has remained largely unchanged in recent years as new therapeutic approaches have struggled to demonstrate benefit. One of the most challenging hurdles to overcome in developing novel treatments is the profound immune suppression found in many GBM patients. This limits the utility of all manner of immunotherapeutic agents, which have revolutionized the treatment of a number of cancers in recent years, but have failed to show similar benefit in GBM therapy. Understanding the mechanisms of tumor-mediated immune suppression in GBM is critical to the development of effective novel therapies, and reversal of this effect may prove key to effective immunotherapy for GBM. In this review, we discuss the current understanding of tumor-mediated immune suppression in GBM in both the local tumor microenvironment and systemically. We also discuss the effects of current GBM therapy on the immune system. We specifically explore some of the downstream effectors of tumor-driven immune suppression, particularly myeloid-derived suppressor cells (MDSCs) and other immunosuppressive monocytes, and the manner by which GBM induces their formation, with particular attention to the role of GBM-derived extracellular vesicles (EVs). Lastly, we briefly review the current state of immunotherapy for GBM and discuss additional hurdles to overcome identification and implementation of effective therapeutic strategies.
Collapse
Affiliation(s)
- Benjamin T Himes
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Philipp A Geiger
- Department of Neurosurgery, University Hospital Innsbruck, Tirol, Austria
| | | | - Adip G Bhargav
- Department of Neurosurgery, University of Kansas, Kansas City, KS, United States
| | - Desmond A Brown
- Surgical Neurology Branch, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
43
|
Xun Y, Yang H, Kaminska B, You H. Toll-like receptors and toll-like receptor-targeted immunotherapy against glioma. J Hematol Oncol 2021; 14:176. [PMID: 34715891 PMCID: PMC8555307 DOI: 10.1186/s13045-021-01191-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023] Open
Abstract
Glioma represents a fast proliferating and highly invasive brain tumor which is resistant to current therapies and invariably recurs. Despite some advancements in anti-glioma therapies, patients’ prognosis remains poor. Toll-like receptors (TLRs) act as the first line of defense in the immune system being the detectors of those associated with bacteria, viruses, and danger signals. In the glioma microenvironment, TLRs are expressed on both immune and tumor cells, playing dual roles eliciting antitumoral (innate and adaptive immunity) and protumoral (cell proliferation, migration, invasion, and glioma stem cell maintenance) responses. Up to date, several TLR-targeting therapies have been developed aiming at glioma bulk and stem cells, infiltrating immune cells, the immune checkpoint axis, among others. While some TLR agonists exhibited survival benefit in clinical trials, it attracts more attention when they are involved in combinatorial treatment with radiation, chemotherapy, immune vaccination, and immune checkpoint inhibition in glioma treatment. TLR agonists can be used as immune modulators to enhance the efficacy of other treatment, to avoid dose accumulation, and what brings more interests is that they can potentiate immune checkpoint delayed resistance to PD-1/PD-L1 blockade by upregulating PD-1/PD-L1 overexpression, thus unleash powerful antitumor responses when combined with immune checkpoint inhibitors. Herein, we focus on recent developments and clinical trials exploring TLR-based treatment to provide a picture of the relationship between TLR and glioma and their implications for immunotherapy.
Collapse
Affiliation(s)
- Yang Xun
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong Province, China
| | - Hua Yang
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong Province, China
| | - Bozena Kaminska
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou, 510095, China.,Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Hua You
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou, 510095, China.
| |
Collapse
|
44
|
Tanaka S, Ohgidani M, Hata N, Inamine S, Sagata N, Shirouzu N, Mukae N, Suzuki SO, Hamasaki H, Hatae R, Sangatsuda Y, Fujioka Y, Takigawa K, Funakoshi Y, Iwaki T, Hosoi M, Iihara K, Mizoguchi M, Kato TA. CD206 Expression in Induced Microglia-Like Cells From Peripheral Blood as a Surrogate Biomarker for the Specific Immune Microenvironment of Neurosurgical Diseases Including Glioma. Front Immunol 2021; 12:670131. [PMID: 34267749 PMCID: PMC8276757 DOI: 10.3389/fimmu.2021.670131] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/04/2021] [Indexed: 01/14/2023] Open
Abstract
Targeting the unique glioma immune microenvironment is a promising approach in developing breakthrough immunotherapy treatments. However, recent advances in immunotherapy, including the development of immune checkpoint inhibitors, have not improved the outcomes of patients with glioma. A way of monitoring biological activity of immune cells in neural tissues affected by glioma should be developed to address this lack of sensitivity to immunotherapy. Thus, in this study, we sought to examine the feasibility of non-invasive monitoring of glioma-associated microglia/macrophages (GAM) by utilizing our previously developed induced microglia-like (iMG) cells. Primary microglia (pMG) were isolated from surgically obtained brain tissues of 22 patients with neurological diseases. iMG cells were produced from monocytes extracted from the patients’ peripheral blood. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed a significant correlation of the expression levels of representative markers for M1 and M2 microglia phenotypes between pMG and the corresponding iMG cells in each patient (Spearman’s correlation coefficient = 0.5225, P <0.0001). Synchronous upregulation of CD206 expression levels was observed in most patients with glioma (6/9, 66.7%) and almost all patients with glioblastoma (4/5, 80%). Therefore, iMG cells can be used as a minimally invasive tool for monitoring the disease-related immunological state of GAM in various brain diseases, including glioma. CD206 upregulation detected in iMG cells can be used as a surrogate biomarker of glioma.
Collapse
Affiliation(s)
- Shunya Tanaka
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuhiro Hata
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shogo Inamine
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriaki Sagata
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noritoshi Shirouzu
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobutaka Mukae
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi O Suzuki
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideomi Hamasaki
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryusuke Hatae
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuhei Sangatsuda
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yutaka Fujioka
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Takigawa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Funakoshi
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Iwaki
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masako Hosoi
- Department of Psychosomatic Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Koji Iihara
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Mizoguchi
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
45
|
Park JH, de Lomana ALG, Marzese DM, Juarez T, Feroze A, Hothi P, Cobbs C, Patel AP, Kesari S, Huang S, Baliga NS. A Systems Approach to Brain Tumor Treatment. Cancers (Basel) 2021; 13:3152. [PMID: 34202449 PMCID: PMC8269017 DOI: 10.3390/cancers13133152] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Brain tumors are among the most lethal tumors. Glioblastoma, the most frequent primary brain tumor in adults, has a median survival time of approximately 15 months after diagnosis or a five-year survival rate of 10%; the recurrence rate is nearly 90%. Unfortunately, this prognosis has not improved for several decades. The lack of progress in the treatment of brain tumors has been attributed to their high rate of primary therapy resistance. Challenges such as pronounced inter-patient variability, intratumoral heterogeneity, and drug delivery across the blood-brain barrier hinder progress. A comprehensive, multiscale understanding of the disease, from the molecular to the whole tumor level, is needed to address the intratumor heterogeneity resulting from the coexistence of a diversity of neoplastic and non-neoplastic cell types in the tumor tissue. By contrast, inter-patient variability must be addressed by subtyping brain tumors to stratify patients and identify the best-matched drug(s) and therapies for a particular patient or cohort of patients. Accomplishing these diverse tasks will require a new framework, one involving a systems perspective in assessing the immense complexity of brain tumors. This would in turn entail a shift in how clinical medicine interfaces with the rapidly advancing high-throughput (HTP) technologies that have enabled the omics-scale profiling of molecular features of brain tumors from the single-cell to the tissue level. However, several gaps must be closed before such a framework can fulfill the promise of precision and personalized medicine for brain tumors. Ultimately, the goal is to integrate seamlessly multiscale systems analyses of patient tumors and clinical medicine. Accomplishing this goal would facilitate the rational design of therapeutic strategies matched to the characteristics of patients and their tumors. Here, we discuss some of the technologies, methodologies, and computational tools that will facilitate the realization of this vision to practice.
Collapse
Affiliation(s)
- James H. Park
- Institute for Systems Biology, Seattle, WA 98109, USA; (J.H.P.); (S.H.)
| | | | - Diego M. Marzese
- Balearic Islands Health Research Institute (IdISBa), 07010 Palma, Spain;
| | - Tiffany Juarez
- St. John’s Cancer Institute, Santa Monica, CA 90401, USA; (T.J.); (S.K.)
| | - Abdullah Feroze
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA; (A.F.); (A.P.P.)
| | - Parvinder Hothi
- Swedish Neuroscience Institute, Seattle, WA 98122, USA; (P.H.); (C.C.)
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Seattle, WA 98122, USA
| | - Charles Cobbs
- Swedish Neuroscience Institute, Seattle, WA 98122, USA; (P.H.); (C.C.)
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Seattle, WA 98122, USA
| | - Anoop P. Patel
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA; (A.F.); (A.P.P.)
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Brotman-Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
| | - Santosh Kesari
- St. John’s Cancer Institute, Santa Monica, CA 90401, USA; (T.J.); (S.K.)
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA 98109, USA; (J.H.P.); (S.H.)
| | - Nitin S. Baliga
- Institute for Systems Biology, Seattle, WA 98109, USA; (J.H.P.); (S.H.)
- Departments of Microbiology, Biology, and Molecular Engineering Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
46
|
Zhang W, Liu Z, Liu B, Jiang M, Yan S, Han X, Shen H, Na M, Wang Y, Ren Z, Liu B, Jiang Z, Gao Y, Lin Z. GNG5 is a novel oncogene associated with cell migration, proliferation, and poor prognosis in glioma. Cancer Cell Int 2021; 21:297. [PMID: 34098960 PMCID: PMC8186147 DOI: 10.1186/s12935-021-01935-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 04/13/2021] [Indexed: 12/16/2022] Open
Abstract
Background Although many biomarkers have been reported for detecting glioma, the prognosis for the disease remains poor, and therefore, new biomarkers need to be identified. GNG5, which is part of the G-protein family, has been associated with different malignant tumors, though the role of GNG5 in glioma has not been studied. Therefore, we aimed to identify the relationship between GNG5 and glioma prognosis and identify a new biomarker for the diagnosis and treatment of gliomas. Methods We used data on more than a thousand gliomas from multiple databases and clinical data to determine the expression of GNG5 in glioma. Based on clinical data and CGGA database, we identified the correlation between GNG5 and multiple molecular and clinical features and prognosis using various analytical methods. Co-expression analysis and GSEA were performed to detect GNG5-related genes in glioma and possible signaling pathways involved. ESTIMATE, ssGSEA, and TIMER were used to detect the relationship between GNG5 and the immune microenvironment. Functional experiments were performed to explore the function of GNG5 in glioma cells. Results GNG5 is highly expressed in gliomas, and its expression level is positively correlated with pathological grade, histological type, age, and tumor recurrence and negatively correlated with isocitrate dehydrogenase mutation, 1p/19 co-deletion, and chemotherapy. Moreover, GNG5 as an independent risk factor was negatively correlated with the overall survival time. GSEA revealed the potential signaling pathways involved in GNG5 function in gliomas, including cell adhesion molecules signaling pathway. The ssGSEA, ESTIMATE, and TIMER based analysis indicated a correlation between GNG5 expression and various immune cells in glioma. In vivo and in vitro experiments showed that GNG5 could participate in glioma cell proliferation and migration. Conclusions Based on the large data platform and the use of different databases to corroborate results obtained using various datasets, as well as in vitro and in vivo experiments, our study reveals for the first time that GNG5, as an oncogene, is overexpressed in gliomas and can inhibit the proliferation and migration of glioma cells and lead to poor prognosis of patients. Thus, GNG5 is a potential novel biomarker for the clinical diagnosis and treatment of gliomas. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01935-7.
Collapse
Affiliation(s)
- Wang Zhang
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China.,Department of Orthopaedics, Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Zhendong Liu
- Department of Orthopaedics, Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Binchao Liu
- Department of Neurosurgery of Xing, Tai People's Hospital, Xing Tai, China
| | - Miaomiao Jiang
- Department of the Pathology, The First Affiliate Hospital of Harbin Medical University, Harbin, China
| | - Shi Yan
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Xian Han
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Hong Shen
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Meng Na
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Yanbiao Wang
- Department of Orthopaedics, Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Zhishuai Ren
- Department of Orthopaedics, Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Binfeng Liu
- Department of Orthopaedics, Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Zhenfeng Jiang
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China
| | - Yanzheng Gao
- Department of Orthopaedics, Department of Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No. 7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| | - Zhiguo Lin
- Department of Neurosurgery, The First Affiliate Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
47
|
Fan D, Yue Q, Chen J, Wang C, Yu R, Jin Z, Yin S, Wang Q, Chen L, Liao X, Peng C, Zhang J, Cao Z, Mao Y, Huang R, Chen L, Li C. Reprogramming the immunosuppressive microenvironment of IDH1 wild-type glioblastoma by blocking Wnt signaling between microglia and cancer cells. Oncoimmunology 2021; 10:1932061. [PMID: 34123575 PMCID: PMC8183516 DOI: 10.1080/2162402x.2021.1932061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The vast majority (>90%) of glioblastoma (GBM) patients belong to the isocitrate dehydrogenase 1 wild type (IDH1WT) group which exhibits a poor prognosis with a median survival of less than 15 months. This study demonstrated numerous immunosuppressive genes as well as β-catenin gene, pivotal for Wnt/β-catenin signaling, were upregulated in 206 IDH1WT glioma patients using the Chinese Glioma Genome Atlas (CGGA) database. The increase in microglia with an immunosuppressive phenotype and the overexpression of β-catenin protein were further verified in IDH1WT GBM patients and IDH1WT GL261 glioma allografts. Subsequently, we found that IDH1WT GL261 cell-derived conditioned medium activated Wnt/β-catenin signaling in primary microglia and triggered their transition to an immunosuppressive phenotype. Blocking Wnt/β-catenin signaling not only attenuated microglial polarization to the immunosuppressive subtype but also reactivated immune responses in IDH1WT GBM allografts by simultaneously enhancing cytotoxic CD8+ T cell infiltration and downregulating regulatory T cells. Positron emission tomography imaging demonstrated enhanced proinflammatory activities in IDH1WT GBM allografts after the blockade of Wnt signaling. Finally, gavage administration of a Wnt signaling inhibitor significantly restrained tumor proliferation and improved the survival of model mice bearing IDH1WT GBM allografts. Depletion of CD8+ T cells remarkably abrogated the therapeutic efficacy induced by the Wnt signaling inhibitor. Overall, the present work indicates that the crosstalk between IDH1WT glioma cells and immunosuppressive microglia is important in maintaining the immunosuppressive glioma microenvironment. Blocking Wnt/β-catenin signaling is a promising complement for IDH1WT GBM treatment by improving the hostile immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Dandan Fan
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Qi Yue
- Department of Neurosurgery, Huashan Hospital and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Cong Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Ruilin Yu
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Ziyi Jin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Shujie Yin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Qinyue Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Luo Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Xueling Liao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Chengyuan Peng
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jianpin Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhonglian Cao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruimin Huang
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Tianqiao and Chrissy Chen Institute for Translational Research, Shanghai, China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Cai X, Yuan F, Zhu J, Yang J, Tang C, Cong Z, Ma C. Glioma-Associated Stromal Cells Stimulate Glioma Malignancy by Regulating the Tumor Immune Microenvironment. Front Oncol 2021; 11:672928. [PMID: 33996602 PMCID: PMC8117153 DOI: 10.3389/fonc.2021.672928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022] Open
Abstract
Background The glioma-associated stromal cell (GASC) is a recently identified type of cell in the glioma microenvironment and may be a prognostic marker for glioma. However, the potential mechanisms of GASCs in the glioma microenvironment remain largely unknown. In this work, we aimed to explore the mechanisms of GASCs in gliomas, particularly in high-grade gliomas (HGG). Methods We used glioma datasets from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). We utilized the Single-sample Gene Set Enrichment Analysis (ssGSEA) algorithm to discriminate between patients with high or low GASC composition. The xCELL and CIBERSORT algorithms were used to analyze the composition of stromal cells and immune cells. Risk score and a nomogram model were constructed for prognostic prediction of glioma. Results We observed for the first time that the levels of M2 macrophages and immune checkpoints (PD-1, PD-L1, PD-L2, TIM3, Galectin-9, CTLA-4, CD80, CD86, CD155, and CIITA) were significantly higher in the high GASC group and showed positive correlation with the GASC score in all glioma population and the HGG population. Copy number variations of DR3 and CIITA were higher in the high-GASC group. THY1, one of the GASC markers, exhibited lower methylation in the high GASC group. The constructed risk score was an independent predictor of glioma prognostics. Finally, a credible nomogram based on the risk score was established. Conclusions GASCs stimulate glioma malignancy through the M2 macrophage, and are associated with the level of immune checkpoints in the glioma microenvironment. The methylation of THY1 could be used as prognostic indicator and treatment target for glioma. However, further studies are required to verify these findings.
Collapse
Affiliation(s)
- Xiangming Cai
- School of Medicine, Southeast University, Nanjing, China
| | - Feng Yuan
- School of Medicine, Nanjing University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Junhao Zhu
- School of Medicine, Nanjing Medical University, Nanjing, China
| | - Jin Yang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chao Tang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Zixiang Cong
- School of Medicine, Nanjing University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chiyuan Ma
- School of Medicine, Southeast University, Nanjing, China.,School of Medicine, Nanjing University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China.,School of Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Akintola OO, Reardon DA. The Current Landscape of Immune Checkpoint Blockade in Glioblastoma. Neurosurg Clin N Am 2021; 32:235-248. [PMID: 33781505 DOI: 10.1016/j.nec.2020.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The glioblastoma tumor microenvironment is highly immunosuppressed. This immunosuppressive state is engineered by inhibitory molecules secreted by tumor cells that limit activation of immune effector cells, drive T-cell exhaustion, and enhance the immunosuppressive action of tumor-associated myeloid cells. Immunotherapeutic approaches have sought to combat glioblastoma microenvironment immunosuppression with agents such as immune checkpoint inhibitors. Although immune checkpoint blockade in glioblastoma has yielded disappointing results thus far, there is significant interest in the combination of immune checkpoint blockade with other approaches to enhance response.
Collapse
Affiliation(s)
- Oluwatosin O Akintola
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Massachusetts General Hospital Cancer Center, 450 Brookline Avenue, Boston, MA 02215-5450, USA.
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| |
Collapse
|
50
|
Caponegro MD, Oh K, Madeira MM, Radin D, Sterge N, Tayyab M, Moffitt RA, Tsirka SE. A distinct microglial subset at the tumor-stroma interface of glioma. Glia 2021; 69:1767-1781. [PMID: 33704822 DOI: 10.1002/glia.23991] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/09/2021] [Accepted: 03/02/2021] [Indexed: 02/01/2023]
Abstract
The characterization of the tumor microenvironment (TME) in high grade gliomas (HGG) has generated significant interest in an effort to understand how neoplastic lesions in the central nervous system (CNS) are supported and to devise novel therapeutic targets. The TME of the CNS contains unique and specialized cells, including the resident myeloid cells, microglia. Myeloid involvement in HGG, such as glioblastoma, is associated with poor outcomes. Glioma-associated microglia and infiltrating monocytes/macrophages (GAM) accumulate within the neoplastic lesion where they facilitate tumor growth and drive immunosuppression. However, it has been difficult to differentiate whether microglia and macrophages have similar or distinct roles in pathology, and if the spatial organization of these cells informs outcomes. Here, we characterize the tumor-stroma border and identify peritumoral GAM (PGAM) as a unique subpopulation of GAM. Using data mining and analyses of samples derived from both murine and human sources we show that PGAM exhibit a pro-inflammatory and chemotactic phenotype that is associated with peripheral monocyte recruitment, and decreased overall survival. PGAM act as a unique subset of GAM at the tumor-stroma interface. We define a novel gene signature to identify these cells and suggest that PGAM constitute a cellular target of the TME.
Collapse
Affiliation(s)
- Michael D Caponegro
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Ki Oh
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Biomedical Informatics, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Miguel M Madeira
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Daniel Radin
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Nicholas Sterge
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Maryam Tayyab
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Richard A Moffitt
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Biomedical Informatics, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Pathology, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Stony Brook Cancer Center, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| | - Stella E Tsirka
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA.,Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|