1
|
Kisby T, Borst GR, Coope DJ, Kostarelos K. Targeting the glioblastoma resection margin with locoregional nanotechnologies. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01020-2. [PMID: 40369318 DOI: 10.1038/s41571-025-01020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
Surgical resection is the first stage of treatment for patients diagnosed with resectable glioblastoma and is followed by a combination of adjuvant radiotherapy and systemic single-agent chemotherapy, which is typically commenced 4-6 weeks after surgery. This delay creates an interval during which residual tumour cells residing in the resection margin can undergo uninhibited proliferation and further invasion, even immediately after surgery, thus limiting the effectiveness of adjuvant therapies. Recognition of the postsurgical resection margin and peri-marginal zones as important anatomical clinical targets and the need to rethink current strategies can galvanize opportunities for local, intraoperative approaches, while also generating a new landscape of innovative treatment modalities. In this Perspective, we discuss opportunities and challenges for developing locoregional therapeutic strategies to target the glioblastoma resection margin as well as emerging opportunities offered by nanotechnology in this clinically transformative setting. We also discuss how persistent barriers to clinical translation can be overcome to offer a potential path forward towards broader acceptability of such advanced technologies.
Collapse
Affiliation(s)
- Thomas Kisby
- Centre for Nanotechnology in Medicine, Faculty of Biology & Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Gerben R Borst
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health & Manchester Cancer Research Centre, Manchester Academic Health Science Centre (MAHSC), University of Manchester, Manchester, UK
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - David J Coope
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Salford Royal, Salford, UK
| | - Kostas Kostarelos
- Centre for Nanotechnology in Medicine, Faculty of Biology & Medicine and Health, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK.
- Nanomedicine Lab, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Barcelona, Spain.
- Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
2
|
Zhang K, Zhang Y, Xiang P, Wang Y, Li Y, Jiang S, Zhang Y, Chen M, Su W, Li X, Li S. Advances in T Cell-Based Cancer Immunotherapy: From Fundamental Mechanisms to Clinical Prospects. Mol Pharm 2025. [PMID: 40359327 DOI: 10.1021/acs.molpharmaceut.4c01502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
T cells and their T cell receptors (TCRs) play crucial roles in the adaptive immune system's response against pathogens and tumors. However, immunosenescence, characterized by declining T cell function and quantity with age, significantly impairs antitumor immunity. Recent years have witnessed remarkable progress in T cell-based cancer treatments, driven by a deeper understanding of T cell biology and innovative screening technologies. This review comprehensively examines T cell maturation mechanisms, T cell-mediated antitumor responses, and the implications of thymic involution on T cell diversity and cancer prognosis. We discuss recent advances in adoptive T cell therapies, including tumor-infiltrating lymphocyte (TIL) therapy, engineered T cell receptor (TCR-T) therapy, and chimeric antigen receptor T cell (CAR-T) therapy. Notably, we highlight emerging DNA-encoded library technologies in mammalian cells for high-throughput screening of TCR-antigen interactions, which are revolutionizing the discovery of novel tumor antigens and optimization of TCR affinity. The review also explores strategies to overcome challenges in the solid tumor microenvironment and emerging approaches to enhance the efficacy of T cell therapy. As our understanding of T cell biology deepens and screening technologies advances, T cell-based immunotherapies show increasing promise for delivering durable clinical benefits to a broader patient population.
Collapse
Affiliation(s)
- Kaili Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Pan Xiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Wang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yifan Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuze Jiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yuxuan Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Min Chen
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaoling Li
- Cell Biotechnology Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin 300308, China
- National Clinical Research Center for Cancer, Tianjin 300060, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300090, China
| | - Shuai Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
3
|
Galvez-Cancino F, Navarrete M, Beattie G, Puccio S, Conde-Gallastegi E, Foster K, Morris Y, Sahwangarrom T, Karagianni D, Liu J, Lee AJX, Garyfallos DA, Simpson AP, Mastrokalos GT, Nannini F, Costoya C, Anantharam V, Cianciotti BC, Bradley L, Garcia-Diaz C, Clements M, Shroff A, Vahid Dastjerdi F, Rota EM, Sheraz S, Bentham R, Uddin I, Walczak H, Lladser A, Reading JL, Chester KA, Pule MA, Brennan PM, Marguerat S, Parrinello S, Peggs KS, McGranahan N, Lugli E, Litchfield K, Pollard SM, Quezada SA. Regulatory T cell depletion promotes myeloid cell activation and glioblastoma response to anti-PD1 and tumor-targeting antibodies. Immunity 2025; 58:1236-1253.e8. [PMID: 40280128 DOI: 10.1016/j.immuni.2025.03.021] [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: 06/26/2023] [Revised: 10/28/2024] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Glioblastoma is invariably lethal and responds poorly to immune checkpoint blockade. Here, we examined the impact of regulatory T (Treg) cell depletion on glioblastoma progression and immunotherapy responsiveness. In human glioblastoma, elevated Treg cell signatures correlated with poorer survival outcomes, with these cells expressing high levels of CD25. In Nf1-/-Pten-/-EGFRvIII+ glioblastoma-bearing mice, a single dose of non-interleukin-2 (IL-2) blocking (NIB) anti-CD25 (anti-CD25NIB) antibody depleted Treg cells and promoted CD8+ T cell clonal expansion and partial tumor control, further enhanced by programmed cell death-1 (PD1)-blockade. Treg cell depletion induced interferon-γ (IFN-γ)-dependent tumor microenvironment remodeling, increasing Fcγ receptor (FcγR) expression on intratumoral myeloid cells and enhancing phagocytosis. Combination of anti-CD25NIB with anti-EGFRvIII tumor-targeting antibodies resulted in complete tumor control. Anti-human CD25NIB treatment of glioblastoma patient-derived tumor fragments effectively depleted Treg cells and activated CD8+ T cells. These findings underscore the therapeutic relevance of Treg targeting in glioblastoma and unveil potent combination strategies for anti-CD25NIB based on innate cell activation.
Collapse
Affiliation(s)
- Felipe Galvez-Cancino
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK; Immune Regulation Laboratory, Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Mariela Navarrete
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Gordon Beattie
- CRUK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK; Bioinformatics Hub, UCL Cancer Institute, University College London, London, UK
| | - Simone Puccio
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Enrique Conde-Gallastegi
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Kane Foster
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Yasmin Morris
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Teerapon Sahwangarrom
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Despoina Karagianni
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Jiali Liu
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alvin J X Lee
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Dimitrios A Garyfallos
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alexander P Simpson
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Gerasimos-Theodoros Mastrokalos
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Francesco Nannini
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Cristobal Costoya
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Varshaa Anantharam
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | | | - Leanne Bradley
- Centre for Regenerative Medicine and Institute for Regeneration and Repair, & Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Claudia Garcia-Diaz
- Neurogenesis and Brain Cancer Group, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Melanie Clements
- Neurogenesis and Brain Cancer Group, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Aditya Shroff
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, London WC1E 6DD, UK
| | | | - Enrique Miranda Rota
- Recombinant Antibody Therapeutics Group, UCL Cancer Institute, London WC1E 6DD, UK
| | - Shahida Sheraz
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Robert Bentham
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Imran Uddin
- CRUK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, London WC1E 6DD, UK; Institute of Biochemistry I & CECAD Cluster of Excellence, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Alvaro Lladser
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - James L Reading
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kerry A Chester
- Recombinant Antibody Therapeutics Group, UCL Cancer Institute, London WC1E 6DD, UK
| | - Martin A Pule
- Research Department of Haematology, Cancer Institute, University College London, Paul O'Gorman Building, London WC1E 6DD, UK
| | - Paul M Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Samuel Marguerat
- Bioinformatics Hub, UCL Cancer Institute, University College London, London, UK
| | - Simona Parrinello
- Neurogenesis and Brain Cancer Group, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Karl S Peggs
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Nicholas McGranahan
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Enrico Lugli
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Kevin Litchfield
- The Tumour Immunogenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, London WC1E 6DD, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine and Institute for Regeneration and Repair, & Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sergio A Quezada
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK.
| |
Collapse
|
4
|
Ellenbogen Y, Zadeh G. A new paradigm for immunotherapy in glioblastoma. Nat Med 2025; 31:1404-1405. [PMID: 40164727 DOI: 10.1038/s41591-025-03607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Affiliation(s)
- Yosef Ellenbogen
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Gelareh Zadeh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada.
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Long GV, Shklovskaya E, Satgunaseelan L, Mao Y, da Silva IP, Perry KA, Diefenbach RJ, Gide TN, Shivalingam B, Buckland ME, Gonzalez M, Caixeiro N, Vergara IA, Bai X, Rawson RV, Hsiao E, Palendira U, Phan TG, Menzies AM, Carlino MS, Quek C, Grimmond SM, Vissers JHA, Yeo D, Rasko JEJ, Khasraw M, Neyns B, Reardon DA, Ashley DM, Wheeler H, Back M, Scolyer RA, Drummond J, Wilmott JS, Rizos H. Neoadjuvant triplet immune checkpoint blockade in newly diagnosed glioblastoma. Nat Med 2025; 31:1557-1566. [PMID: 40016450 DOI: 10.1038/s41591-025-03512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/14/2025] [Indexed: 03/01/2025]
Abstract
Glioblastoma (GBM) is an aggressive primary adult brain tumor that rapidly recurs after standard-of-care treatments, including surgery, chemotherapy and radiotherapy. While immune checkpoint inhibitor therapies have transformed outcomes in many tumor types, particularly when used neoadjuvantly or as a first-line treatment, including in melanoma brain metastases, they have shown limited efficacy in patients with resected or recurrent GBM. The lack of efficacy has been attributed to the scarcity of tumor-infiltrating lymphocytes (TILs), an immunosuppressive tumor microenvironment and low tumor mutation burden typical of GBM tumors, plus exclusion of large molecules from the brain parenchyma. We hypothesized that upfront neoadjuvant combination immunotherapy, administered with disease in situ, could induce a stronger immune response than treatment given after resection or after recurrence. Here, we present a case of newly diagnosed IDH-wild-type, MGMT promoter unmethylated GBM, treated with a single dose of neoadjuvant triplet immunotherapy (anti-programmed cell death protein 1 plus anti-cytotoxic T-lymphocyte protein 4 plus anti-lymphocyte-activation gene 3) followed by maximal safe resection 12 days later. The anti-programmed cell death protein 1 drug was bound to TILs in the resected GBM and there was marked TIL infiltration and activation compared with the baseline biopsy. After 17 months, there is no definitive sign of recurrence. If used first line, before safe maximal resection, checkpoint inhibitors are capable of immune activation in GBM and may induce a response. A clinical trial of first-line neoadjuvant combination checkpoint inhibitor therapy in newly diagnosed GBM is planned (GIANT; trial registration no. NCT06816927 ).
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.
- Mater Hospital, North Sydney, New South Wales, Australia.
- Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| | - Elena Shklovskaya
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| | - Laveniya Satgunaseelan
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Yizhe Mao
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Inês Pires da Silva
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Kristen A Perry
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Russell J Diefenbach
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| | - Tuba N Gide
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Brindha Shivalingam
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Michael E Buckland
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Maria Gonzalez
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| | - Nicole Caixeiro
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Ismael A Vergara
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Xinyu Bai
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Robert V Rawson
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - Edward Hsiao
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Umaimainthan Palendira
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Tri Giang Phan
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, North Sydney, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Blacktown Hospital, Blacktown, New South Wales, Australia
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Camelia Quek
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sean M Grimmond
- Collaborative Centre for Genomic Cancer Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph H A Vissers
- Collaborative Centre for Genomic Cancer Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Dannel Yeo
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Centenary Institute, Camperdown, New South Wales, Australia
| | - John E J Rasko
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Centenary Institute, Camperdown, New South Wales, Australia
| | | | - Bart Neyns
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Helen Wheeler
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Michael Back
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - James Drummond
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
- North Shore Radiology & Nuclear Medicine, St Leonards, New South Wales, Australia
- Brain Imaging Laboratory, The Brain Cancer Group, St Leonards, New South Wales, Australia
| | - James S Wilmott
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Helen Rizos
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| |
Collapse
|
6
|
Ellert-Miklaszewska A, Pilanc P, Poleszak K, Roura AJ, Cyranowski S, Ghosh M, Baluszek S, Pasierbinska M, Gielniewski B, Swatler J, Hovorova Y, Wojnicki K, Kaminska B. 7aaRGD - a novel SPP1/integrin signaling-blocking peptide reverses immunosuppression and improves anti-PD-1 immunotherapy outcomes in experimental gliomas. J Exp Clin Cancer Res 2025; 44:132. [PMID: 40281508 PMCID: PMC12032770 DOI: 10.1186/s13046-025-03393-9] [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: 09/19/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) present clinical benefits in many cancer patients but invariably fail in glioblastoma (GBM), the most common and deadly primary brain tumor. The lack of ICIs efficacy in GBM is attributed to the accumulation of tumor-reprogrammed glioma-associated myeloid cells (GAMs) that create a "cold" immunosuppressive tumor microenvironment (TME), impeding the infiltration and activation of effector T cells. GBM-derived αvβ3/αvβ5-integrin ligands, including SPP1, were shown to mediate the emergence of GAMs. We hypothesized that a combination strategy aiming to block the reprogramming of GAMs using a synthetic 7aaRGD peptide that targets SPP1/integrin signaling might overcome resistance to ICIs and reinvigorate anti-tumor immunity. METHODS Matrigel invasion assay was used to test the efficacy of 7aaRGD in glioma-microglia co-cultures. We determined the impact of 7aaRGD, administered as a monotherapy or combined with PD-1 blockade, on tumor growth, GAMs accumulation and phenotypes, arginase-1 levels and neovasculature in experimental gliomas. The effects of treatments on the tumor immune landscape were dissected using multiparameter flow cytometry, immunocytochemistry, cytokine profiling and RNA-seq analysis of sorted GAMs followed by CITE-seq based data deconvolution. RESULTS 7aaRGD efficiently blocked microglia-dependent invasion of human and mouse glioma cells in vitro. Intratumorally delivered 7aaRGD alone did not reduce tumor growth in orthotopic gliomas but prevented the emergence of immunosuppressive GAMs and led to normalization of peritumoral blood vessels. Combining 7aaRGD with anti-PD-1 antibody resulted in reduced tumor growth, with an increase in the number of proliferating, interferon-ɣ producing CD8+T cells and depletion of regulatory T cells. Transcriptomic profiles of myeloid cells were altered by the combined treatment, reflecting the restored "hot" inflammatory TME and boosted immunotherapy responses. Intratumoral administration of 7aaRGD similarly modified the phenotypes of GAMs in human U87-MG gliomas in immunocompromised mice. Exploration of transcriptomic datasets revealed that high expression of integrin receptor coding genes in pre-treatment biopsies was associated with a poorer response to immune check-point blockade in patients with several types of cancers. CONCLUSIONS We demonstrate that combining the blockade of SPP1/integrin signaling with ICIs modifies innate immunity and reinvigorates adaptive antitumor responses, which paves the way to improve immunotherapy outcomes in GBM.
Collapse
Affiliation(s)
| | - Paulina Pilanc
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Katarzyna Poleszak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Adria-Jaume Roura
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Salwador Cyranowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Mitrajit Ghosh
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Szymon Baluszek
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Maria Pasierbinska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bartłomiej Gielniewski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Yuliana Hovorova
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
7
|
Rezazadeh‐Gavgani E, Majidazar R, Lotfinejad P, Kazemi T, Shamekh A. Immune Checkpoint Molecules: A Review on Pathways and Immunotherapy Implications. Immun Inflamm Dis 2025; 13:e70196. [PMID: 40243372 PMCID: PMC12004596 DOI: 10.1002/iid3.70196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Today, treating cancer patients with monoclonal antibodies (mAbs), by targeting immune checkpoints, is one of the most outstanding immunotherapeutic methods. Immune checkpoints are special molecules having regulatory role in immune system responses. Once these molecules are presented on cancer cells, these cells will be capable of evading the immune system through their own specific pathways. This Evasion can be prevented by counterbalancing immune system responses with immune checkpoints related antibodies. AIMS The current study aimed to highlight immunotherapy and its methods, describe the immune checkpoints pathways, outline the immune checkpoint inhibitors (ICIs), and recent advances in this field, and sketch an outlook on the best treatment options for the most prevalent cancers. MATERIALS & METHODS This research implemented a narrative review method. A comprehensive literature review on the history, molecular and cellular biology, and the clinical aspects of immune checkpoint molecules was performed to illustrate the pathways involved in various cancers. Also, currently-available and future potential immunotherapies targeting these pathways were extracted from the searched studies. RESULTS The immune checkpoint family consists of many molecules, including CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and TIGIT. Attempts to modify these molecules in cancer treatment led to the development of therapeutic monoclonal antibodies. Most of these antibodies have entered clinical studies and some of them have been approved by the Food and Drug Administration (FDA) to be used in cancer patients' treatment plans. DISCUSSION With these novel treatments and the combination therapies they offer, there is also hope for better treatment outcomes for the previously untreatable metastatic cancers. In spite of the beneficial aspects of immune checkpoint therapy, similar to other treatments, they may cause side effects in some patients. Therefore, more studies are needed to reduce the probable side effects and uncover their underlying mechanism. CONCLUSION Based on the data shown in this review, there is still a lack of knowledge about the complete properties of ICIs and the possible combination therapies that we may be able to implement to achieve a better treatment response in cancer patients.
Collapse
Affiliation(s)
| | - Reza Majidazar
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Parisa Lotfinejad
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Tohid Kazemi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Ali Shamekh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
8
|
Sandhbor P, John G, Bhat S, Goda JS. Immune response recalibration using immune therapy and biomimetic nano-therapy against high-grade gliomas and brain metastases. Asian J Pharm Sci 2025; 20:101021. [PMID: 40224727 PMCID: PMC11987628 DOI: 10.1016/j.ajps.2025.101021] [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: 04/30/2024] [Revised: 08/07/2024] [Accepted: 10/03/2024] [Indexed: 04/15/2025] Open
Abstract
Although with aggressive standards of care like surgical resection, chemotherapy, and radiation, high-grade gliomas (HGGs) and brain metastases (BM) treatment has remained challenging for more than two decades. However, technological advances in this field and immunotherapeutic strategies have revolutionized the treatment of HGGs and BM. Immunotherapies like immune checkpoint inhibitors, CAR-T targeting, oncolytic virus-based therapy, bispecific antibody treatment, and vaccination approaches, etc., are emerging as promising avenues offering new hope in refining patient's survival benefits. However, selective trafficking across the blood-brain barrier (BBB), immunosuppressive tumor microenvironment (TME), metabolic alteration, and tumor heterogeneity limit the therapeutic efficacy of immunotherapy for HGGs and BM. Furthermore, to address this concern, the NanoBioTechnology-based bioinspired delivery system has been gaining tremendous attention in recent years. With technological advances such as Trojan horse targeting and infusing/camouflaging nanoparticles surface with biological molecules/cells like immunocytes, erythrocytes, platelets, glioma cell lysate and/or integrating these strategies to get hybrid membrane for homotypic recognition. These biomimetic nanotherapy offers advantages over conventional nanoparticles, focusing on greater target specificity, increased circulation stability, higher active loading capacity, BBB permeability (inherent inflammatory chemotaxis of neutrophils), decreased immunogenicity, efficient metabolism-based combinatorial effects, and prevention of tumor recurrence by induction of immunological memory, etc. provide new age of improved immunotherapies outcomes against HGGs and BM. In this review, we emphasize on neuro-immunotherapy and the versatility of these biomimetic nano-delivery strategies for precise targeting of hard-to-treat and most lethal HGGs and BM. Moreover, the challenges impeding the clinical translatability of these approaches were addressed to unmet medical needs of brain cancers.
Collapse
Affiliation(s)
- Puja Sandhbor
- Institute for NanoBioTechnology, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore 21218, USA
| | - Geofrey John
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Kharghar 410210, India
- Homi Bhabha National Institute, Anushakti Nagar 400094, India
| | - Sakshi Bhat
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Kharghar 410210, India
- Homi Bhabha National Institute, Anushakti Nagar 400094, India
| | - Jayant S. Goda
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer, Tata Memorial Centre, Kharghar 410210, India
- Homi Bhabha National Institute, Anushakti Nagar 400094, India
| |
Collapse
|
9
|
Lin Q, Wei Y, Xu G, Wang L, Ling F, Chen X, Cheng Y, Zhou Y. Integrative multi-omic profiling of the neoantigen landscape of glioblastoma for the development of therapeutic vaccines reveals vast heterogeneity in immunogenic signatures. Front Oncol 2025; 15:1507632. [PMID: 40190555 PMCID: PMC11968714 DOI: 10.3389/fonc.2025.1507632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/13/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Glioblastoma (GBM) is the most common primary brain malignancy. Few neoantigens have been tested in trials as the cancer vaccine against GBM. Methods To better understand the neoantigen landscape and its associated tumor microenvironment (TME) for the optimized vaccine design of our initiated GBM trial, we apply the integrative multi-omics approach to comprehensively profile the mutation, HLA typing, TCR/BCR repertoire, immune cell components on the tumor tissue and peripheral blood mononuclear cell (PMBC) specimen of 24 GBM patients. Results On average, 148 mutated genes and 200 mutated sites per patient were identified, with no predominant mutated sites and genes in this cohort. Diversified HLA genotypes and expression rate across A, B, and C alleles, with A30:01&A11:01, B13:02, and C06:02, as the most frequent genotypes at respective alleles. Clustered CDR3 of TCR/BCR existed in tumor tissue with decreased richness compared with PMBC. NK and Th1 cells were revealed as the predominant immune cells within the tumor microenvironment (TME). Neoantigens were feasible predicted and designed for each patient, with an average number of 107. Very few neoantigens were shared by more than two patients and no dominant neoantigen could be identified. A minimum of 11-peptide bulk was required to cover this 24-patient cohort, guaranteeing each patient could have at least one neoantigen. Discussion In summary, our data reveals a heterogeneous landscape of the neoantigen and its associated immune TME of GBM, based on which a peptide bulk is feasibly developed to cover these patients as a cohort.
Collapse
Affiliation(s)
- Qingtang Lin
- Department of Neurosurgery, Brain Tumor and Skull-Base Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yukui Wei
- Department of Neurosurgery, Brain Tumor and Skull-Base Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Geng Xu
- Department of Neurosurgery, Brain Tumor and Skull-Base Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Leiming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Feng Ling
- Department of Neurosurgery, Brain Tumor and Skull-Base Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaojie Chen
- Base&Byte Biotechnology Co., Ltd, Beijing, China
| | - Ye Cheng
- Department of Neurosurgery, Brain Tumor and Skull-Base Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yiming Zhou
- Base&Byte Biotechnology Co., Ltd, Beijing, China
| |
Collapse
|
10
|
Radin DP. AMPA Receptor Modulation in the Treatment of High-Grade Glioma: Translating Good Science into Better Outcomes. Pharmaceuticals (Basel) 2025; 18:384. [PMID: 40143160 PMCID: PMC11945080 DOI: 10.3390/ph18030384] [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: 01/15/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma (GB) treatment, despite consisting of surgical resection paired with radiation, temozolomide chemotherapy and tumor-treating fields, yields a median survival of 15-20 months. One of the more recently appreciated hallmarks of GB aggressiveness is the co-opting of neurotransmitter signaling mechanisms that normally sustain excitatory synaptic communication in the CNS. AMPA-glutamate receptor (AMPAR) signaling governs the majority of excitatory synaptic activity in the mammalian brain. AMPAR activation in glioma cells activates cellular pathways that enhance proliferation and invasion and confer resistance to approved GB therapeutics. In addition, this review places a specific emphasis on discussing the redefined GB cytoarchitecture that consists of neuron-to-glioma cell synapses, whose oncogenic activity is driven by AMPAR activation on glioma cells, and the discovery of tumor microtubes, which propagate calcium signals throughout the tumor network in order to enhance resistance to complete surgical resection and radiotherapy. These new discoveries notwithstanding, some evidence suggests that AMPAR activation can produce excitotoxicity in tumor cells. This disparity warrants a closer examination at how AMPAR modulation can be leveraged to produce more durable outcomes in the treatment of GB and tumors in peripheral organs that express AMPAR.
Collapse
Affiliation(s)
- Daniel P Radin
- Stony Brook Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| |
Collapse
|
11
|
Wang X, Li T, Slebos RJC, Chaudhary R, Guevara-Patino JA, Bonomi M, Saba NF, Chung CH. Clinical significance of peripheral T-cell repertoire in head and neck squamous cell carcinoma treated with cetuximab and nivolumab. Cancer Immunol Immunother 2025; 74:142. [PMID: 40056190 PMCID: PMC11890688 DOI: 10.1007/s00262-025-03993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
Immunotherapy holds significant promise for treating head and neck squamous cell carcinoma (HNSCC), yet responses are limited to a subset of patients. This research investigates whether analyzing the peripheral T-cell receptor (TCR) repertoire could help identify patients who are more likely to benefit from a combination treatment of cetuximab and nivolumab. We report here updated correlative analysis using all samples profiled with deep immunoSEQ assay to study the peripheral TCR repertoires in peripheral blood mononuclear cells from patients enrolled in a phase I/II trial (NCT03370276). TCR repertoires were analyzed in 67 patients. Of these, 64 had available baseline data. Overall, our findings confirm that a more polyclonal peripheral TCR repertoire is associated with improved response to concurrent cetuximab and nivolumab in HNSCC. While the baseline productive Simpson clonality did not reach statistically significant differences in response groups, significant differences were observed within the HPV-negative subgroup and among patients who had received previous ICI therapy. Additionally, the TCR diversity at baseline and early follow-up was associated with overall survival. TRBV/TRBJ gene usage analysis also identified specific gene pairs associated with patient outcomes. Furthermore, our analysis indicates that the TCR clonality patterns are modulated by prior treatment exposures and tumor HPV status, suggesting a cohort expansion within these subgroups for further validation. Together, this study demonstrates the feasibility of leveraging the peripheral T-cell repertoire profiling and clonality dynamics as predictive biomarkers for immunotherapy efficacy in HNSCC.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
- Immuno-Oncology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| | - Tingyi Li
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Robbert J C Slebos
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Ritu Chaudhary
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Jose A Guevara-Patino
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Marcelo Bonomi
- Department of Internal Medicine, The Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| |
Collapse
|
12
|
Tsuboi N, Rivera-Caraballo KA, Sahu U, Pacholczyk R, Douglass E, Johnson TS, Wang Q, Kolhe R, Hedrick CC, Munn DH, Hong B. Blocking Feedback Immunosuppression of Antigen Presentation in Brain Tumor During Oncolytic Virotherapy with oHSV-mshPKR. Mol Cancer Ther 2025; 24:444-452. [PMID: 39711419 PMCID: PMC11879753 DOI: 10.1158/1535-7163.mct-24-0629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Glioblastoma (GBM) is the most frequent malignant brain tumor. We recently discovered that oncolytic herpes simplex virus engineered to disable tumor-intrinsic protein kinase R (PKR) signaling (oHSV-shPKR) could increase oHSV oncolysis and antitumor immune response. However, in this study, we show that disabling tumor-intrinsic PKR signaling can also induce the activation of the indoleamine 2,3-dioxygenase (IDO) signaling pathway. Both GBM tumor progression and oHSV intratumoral therapy increased infiltration of IDO+CD11c+ dendritic cells (DC) into the tumor. The coculture of oHSV-infected human GBM neurospheres with monocyte-derived DCs (MoDC) dramatically increased IDO signaling activation in MoDCs through type-I IFN signaling. Addition of IDO inhibitor (indoximod) in the coculture significantly increased MoDC activation and reduced the consumption of tryptophan. Combining indoximod and oHSV significantly inhibited tumor growth and induced antigen-specific CD8+ T-cell activation. These results suggest that inhibition of the IDO pathway could significantly block feedback immunosuppression during oncolytic virotherapy of GBM.
Collapse
Affiliation(s)
- Nobushige Tsuboi
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | | | - Upasana Sahu
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Rafal Pacholczyk
- Department of Biochemistry and Molecular Biology, Georgia Cancer Center at Augusta University, 1410 Laney Walker Blvd, Augusta, GA, USA
| | - Eugene Douglass
- Department of Pharmaceutical & Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Theodore S. Johnson
- Department of Pediatrics, Pediatric Immunotherapy Program, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Qin Wang
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Ravindra Kolhe
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Catherine C. Hedrick
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - David H. Munn
- Department of Pediatrics, Pediatric Immunotherapy Program, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Bangxing Hong
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| |
Collapse
|
13
|
Lopez de Rodas M, Villalba-Esparza M, Sanmamed MF, Chen L, Rimm DL, Schalper KA. Biological and clinical significance of tumour-infiltrating lymphocytes in the era of immunotherapy: a multidimensional approach. Nat Rev Clin Oncol 2025; 22:163-181. [PMID: 39820025 DOI: 10.1038/s41571-024-00984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have improved clinical outcomes across several solid tumour types. Prominent efforts have focused on understanding the anticancer mechanisms of these agents, identifying biomarkers of response and uncovering resistance mechanisms to develop new immunotherapeutic approaches. This research has underscored the crucial roles of the tumour microenvironment and, particularly, tumour-infiltrating lymphocytes (TILs) in immune-mediated tumour elimination. Numerous studies have evaluated the prognostic and predictive value of TILs and the mechanisms that govern T cell dysfunction, fuelled by technical developments in single-cell transcriptomics, proteomics, high-dimensional spatial platforms and advanced computational models. However, questions remain regarding the definition of TILs, optimal strategies to study them, specific roles of different TIL subpopulations and their clinical implications in different treatment contexts. Additionally, most studies have focused on the abundance of major TIL subpopulations but have not developed standardized quantification strategies or analysed other crucial aspects such as their functional profile, spatial distribution and/or arrangement, tumour antigen-reactivity, clonal diversity and heterogeneity. In this Review, we discuss a conceptual framework for the systematic study of TILs and summarize the evidence regarding their biological properties and biomarker potential for ICI therapy. We also highlight opportunities, challenges and strategies to support future developments in this field.
Collapse
Affiliation(s)
- Miguel Lopez de Rodas
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba-Esparza
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada and Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
14
|
Jang HJ, Park JW. Microenvironmental Drivers of Glioma Progression. Int J Mol Sci 2025; 26:2108. [PMID: 40076738 PMCID: PMC11900340 DOI: 10.3390/ijms26052108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Gliomas, particularly glioblastoma (GBM), are among the most challenging brain tumors due to their complex and dynamic tumor microenvironment (TME). The TME plays a pivotal role in tumor progression, immune evasion, and resistance to therapy through intricate interactions among glioma cells, immune components, neurons, astrocytes, the extracellular matrix, and the blood-brain barrier. Targeting the TME has demonstrated potential, with immunotherapies such as checkpoint inhibitors and neoadjuvant therapies enhancing immune responses. Nonetheless, overcoming the immunosuppressive landscape and metabolic adaptations continues to pose significant challenges. This review explores the diverse cellular and molecular mechanisms that shape the glioma TME. A deeper understanding of these mechanisms holds promise for providing novel therapeutic opportunities to improve glioma treatment outcomes.
Collapse
Affiliation(s)
- Hyun Ji Jang
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
| | - Jong-Whi Park
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
15
|
Vallieri N, Datsi A. Immune Cell Interplay in the Fight Against GBM. Cancers (Basel) 2025; 17:817. [PMID: 40075663 PMCID: PMC11899300 DOI: 10.3390/cancers17050817] [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: 01/26/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Despite multimodal therapies, the treatment of glioblastoma remains challenging. In addition to the very complex mechanisms of cancer cells, including specialized phenotypes that enable them to proliferate, invade tissues, and evade immunosurveillance, they exhibit a pronounced resistance to chemo- and radiotherapy. More advanced tumors create a hypoxic environment that supports their proliferation and survival, while robust angiogenesis ensures a constant supply of nutrients. In GBM, these structures are very pronounced and contribute to the creation and maintenance of a highly immunosuppressive microenvironment that promotes tumor growth and immune escape. In addition, the high accumulation of immunosuppressive tumor-infiltrating leukocytes and other cells, the pronounced expression of immune checkpoint molecules, and the low mutational burden, i.e., the low number of neoantigens, are hallmarks of GBM and contribute to the challenge of therapeutic approaches. Here, we review a number of mechanisms that GBM exploits to support tumor growth and potential treatments. These include new chemotherapeutics, tumor treating fields, and small molecules, including compounds targeting angiogenesis or blockers of tyrosine kinases that inhibit tumor cell proliferation and survival. In addition, we focus on immunotherapies such as immune checkpoint blockade or cell therapies, in particular vaccination with dendritic cells and CAR-T cells, which can either kill GBM cells directly or bypass immunosuppression by modulating the tumor microenvironment or boosting the patient's own immune response.
Collapse
Affiliation(s)
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, 40225 Duesseldorf, Germany;
| |
Collapse
|
16
|
Mo Y, Fan D, Wang W, Wang S, Yan Y, Zhao Z. Identification of inflammation-related genes signature to establish a prognostic model in MGMT unmethylated glioblastoma patients. Discov Oncol 2025; 16:154. [PMID: 39932605 DOI: 10.1007/s12672-025-01894-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Patients with unmethylated O6-methylguanine-DNA methyltransferase promoter (uMGMT) glioblastoma (GBM) have a poor prognosis. Inflammatory response can affect the prognosis, for it may have a significant impact on the tumor microenvironment (TME). This study aims to identify a prognostic signature of inflammation-related genes, which can predict the prognosis of uMGMT GBM patients. METHODS We examined the gene expression, somatic mutations, and overall survival of 159 GBM patients with uMGMT using the TCGA and CGGA databases. We identified molecular subtypes of uMGMT GBM patients based on the expression of inflammation-related genes. Furthermore, we determined principal component analysis (PCA), gene ontology (GO) analysis, pathway analysis and immune infiltration analysis between high and low-inflammation subtypes. We also examined the spatial and longitudinal heterogeneity of these two subtypes. The LASSO-Cox analyses were used to develop an inflammation-related prognostic model. RESULTS Our findings indicate that patients with uMGMT GBM can be divided into high-inflammation and low-inflammation subtypes. Patients with high levels of inflammation are more likely to develop an immunosuppressive microenvironment, which stimulates the production of immunosuppressive cytokines, immune checkpoints, and immunosuppressive cells. Nine inflammation-related genes (EREG, BDKRB1, DCBLD2, CD14, AHR, CLEC5A, LTA, SLC4A4, and LY6E) were found to have excellent predictive potential for patient survival in the prognostic model. CONCLUSIONS In conclusion, we created a new prognostic model including 9 inflammation-related genes. This model has produced meaningful results in evaluating patient prognosis, which may help with future therapeutic strategies for patients with uMGMT GBM.
Collapse
Affiliation(s)
- Yunzhao Mo
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Dandan Fan
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Wei Wang
- Department of Pathology, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Shenchuan Wang
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China
| | - Yingyu Yan
- Quality Management Department, The 921st Hospital of the Joint Logistics Support Force, Changsha, 410008, China
| | - Zhenyu Zhao
- Department of Neurosurgery, General Hospital of Southern Theater Command, Guangzhou, 510010, China.
| |
Collapse
|
17
|
Lunavat TR, Nieland L, van de Looij SM, de Reus AJEM, Couturier CP, Farran CAE, Miller TE, Lill JK, Schübel M, Xiao T, Ianni ED, Woods EC, Sun Y, Rufino-Ramos D, van Solinge TS, Mahjoum S, Grandell E, Li M, Mangena V, Dunn GP, Jenkins RW, Mempel TR, Breakefield XO, Breyne K. Intratumoral gene delivery of 4-1BBL boosts IL-12-triggered anti-glioblastoma immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636330. [PMID: 39975249 PMCID: PMC11838556 DOI: 10.1101/2025.02.03.636330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The standard of care in high-grade gliomas has remained unchanged in the past 20 years. Efforts to replicate effective immunotherapies in non-cranial tumors have led to only modest therapeutical improvements in glioblastoma (GB). Here, we demonstrate that intratumoral administration of recombinant interleukin-12 (rIL-12) promotes local cytotoxic CD8 POS T cell accumulation and conversion into an effector-like state, resulting in a dose-dependent survival benefit in preclinical GB mouse models. This tumor-reactive CD8 T cell response is further supported by intratumoral rIL-12-sensing dendritic cells (DCs) and is accompanied by the co-stimulatory receptor 4-1BB expression on both cell types. Given that DCs and CD8 POS T cells are functionally suppressed in the tumor microenvironments of de novo and recurrent glioma patients, we tested whether anti-tumor response at the rIL-12-inflamed tumor site could be enhanced with 4-1BBL, the ligand of 4-1BB. 4-1BBL was delivered using an adeno-associated virus (AAV) vector targeting GFAP-expressing cells and resulted in prolonged survival of rIL-12 treated GB-bearing mice. This study establishes that tumor antigen-specific CD8 T cell activity can be directed using an AAV-vector-mediated gene therapy approach, effectively enhancing anti-GB immunity.
Collapse
|
18
|
Yan T, Liao Q, Chen Z, Xu Y, Zhu W, Hu P, Zhang S, Wu Y, Shu L, Liu J, Luo M, Shu H, Sheng Y, Wang L, Xu C, Lei C, Wang H, Ye Q, Yang L, Zhu X. β-Ketoenamine covalent organic framework nanoplatform combined with immune checkpoint blockade via photodynamic immunotherapy inhibit glioblastoma progression. Bioact Mater 2025; 44:531-543. [PMID: 39584065 PMCID: PMC11583667 DOI: 10.1016/j.bioactmat.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/02/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
The synergistic approach of combining photodynamic immunotherapy with endogenous clearance of PD-L1 immune checkpoint blockade therapy holds promise for enhancing survival outcomes in glioblastoma (GBM) patients. The observed upregulation of O-GlcNAc glycolysis in tumors may contribute to the stabilization of endogenous PD-L1 protein, facilitating tumor immune evasion. This study presents a pH-adapted excited state intramolecular proton transfer (ESIPT)-isomerized β-ketoamide-based covalent organic framework (COF) nanoplatform (denoted as OT@COF-RVG). Temozolomide (TMZ) and OSMI-4 (O-GlcNAc transferase inhibitor) were integrated into COF cavities, then modified on the surface with polyethylene glycol and the rabies virus peptide RVG-29, showing potential for sensitizing TMZ chemotherapy and initiating photodynamic therapy (PDT). By inhibiting O-GlcNAc and promoting lysosomal degradation of PD-L1, OT@COF-RVG enhanced the effectiveness of immune checkpoint blockade (ICB) therapy. Additionally, treatment with OT@COF-RVG led to a notable elevation in reactive oxygen species (ROS) levels, thereby re-establishing an immunostimulatory state, inducing immunogenic cell death (ICD). In summary, our research unveiled a correlation between O-GlcNAc in GBM and the evasion of immune responses by tumors, while showcasing the potential of OT@COF-RVG in reshaping the immunosuppressive microenvironment of GBM and offering a more effective approach to immunotherapy in clinical settings.
Collapse
Affiliation(s)
- Tengfeng Yan
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, Jiangxi Province, 330000, China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi Province, 330000, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Qiuye Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Zhihao Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Wenping Zhu
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi Province, 330000, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Ping Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Si Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Yanze Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Lei Shu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Junzhe Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Min Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Hongxin Shu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Yilei Sheng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Li Wang
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, 330022, China
| | - Chun Xu
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Chang Lei
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Hongming Wang
- College of Chemistry and Chemical Engineering and Jiangxi Provincial Key Laboratory of Functional Crystalline Materials Chemistry Nanchang University, Nanchang, 330031, China
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan University, 430060, Wuhan, China
| | - Li Yang
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi Province, 330000, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330000, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang, Jiangxi Province, 330000, China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi Province, 330000, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, 330000, China
| |
Collapse
|
19
|
Juarez TM, Gill JM, Minev BR, Sharma A, Kesari S. Neoadjuvant clinical trials in adults with newly diagnosed high-grade glioma: A systematic review. Crit Rev Oncol Hematol 2025; 206:104596. [PMID: 39675399 DOI: 10.1016/j.critrevonc.2024.104596] [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: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND High-grade gliomas are devastating cancers that remain incurable with standard surgical resection and radiochemotherapy. Although beneficial against neoplasms, radiation lowers lymphocyte counts, weakens immune activation, and recruits suppressive myeloid cells impairing immune responses. Tumor environments treated with radiation experience long-term immunosuppression, reducing immunotherapy effectiveness and contributing to recurrence. Investigating pre-radiation treatments in newly diagnosed patients could identify active agents, assess immunotherapy impact, and enable multiomic analyses without radiation-induced confounding factors. This literature review was conducted to describe the feasibility, safety, and outcomes of postsurgical, pre-radiation clinical trials for adults with newly diagnosed high-grade glioma. METHODS A systematic review was performed of the English-language literature reporting results of clinical trials for adults with newly diagnosed high-grade glioma administered postsurgical treatment prior to radiation therapy. A search was conducted in PubMed and references cited in research and review articles were also considered. RESULTS From 1991 to 2024, 52 clinical trials were identified: 3 phase I, 38 phase II, 4 phase III, and 7 of unknown phase. Nine trials were randomized, 24 were multicenter trials, 21 investigated temozolomide-containing regimens, and 12 focused on inoperable tumors, involving a total of 2737 patients. CONCLUSION Pre-radiation neoadjuvant studies are feasible and may identify active drugs. This is particularly relevant in the era of personalized medicine with brain-penetrant drugs, targeted therapy, and immuno-oncology advancements. Investigating pre-radiation treatments in newly diagnosed high-grade glioma is a viable approach to rapidly identify active and inactive regimens while the immune system and tumor microenvironment remain intact.
Collapse
Affiliation(s)
| | | | - Boris R Minev
- Calidi Biotherapeutics, San Diego, CA, USA; Department of Radiation Medicine and Applied Sciences, University of California, San Diego, CA 92093, USA
| | - Akanksha Sharma
- Pacific Neuroscience Institute, Santa Monica, CA, USA; Department of Translational Neuroscience, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute, Santa Monica, CA, USA; Department of Translational Neuroscience, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA.
| |
Collapse
|
20
|
Badani A, Ozair A, Khasraw M, Woodworth GF, Tiwari P, Ahluwalia MS, Mansouri A. Immune checkpoint inhibitors for glioblastoma: emerging science, clinical advances, and future directions. J Neurooncol 2025; 171:531-547. [PMID: 39570554 DOI: 10.1007/s11060-024-04881-2] [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: 09/21/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Glioblastoma (GBM), the most common and aggressive primary central nervous system (CNS) tumor in adults, continues to have a dismal prognosis. Across hundreds of clinical trials, few novel approaches have translated to clinical practice while survival has improved by only a few months over the past three decades. Randomized controlled trials of immune checkpoint inhibitors (ICIs), which have seen impressive success for advanced or metastatic extracranial solid tumors, have so far failed to demonstrate a clinical benefit for patients with GBM. This has been secondary to GBM heterogeneity, the unique immunosuppressive CNS microenvironment, immune-evasive strategies by cancer cells, and the rapid evolution of tumor on therapy. This review aims to summarize findings from major clinical trials of ICIs for GBM, review historic failures, and describe currently promising avenues of investigation. We explore the biological mechanisms driving ICI responses, focusing on the role of the tumor microenvironment, immune evasion, and molecular biomarkers. Beyond conventional monotherapy approaches targeting PD-1, PD-L1, CTLA-4, we describe emerging approaches for GBM, such as dual-agent ICIs, and combination of ICIs with oncolytic virotherapy, antigenic peptide vaccines, chimeric antigenic receptor (CAR) T-cell therapy, along with nanoparticle-based delivery systems to enhance ICI efficacy. We highlight potential strategies for improving patient selection and treatment personalization, along with real-time, longitudinal monitoring of therapeutic responses through advanced imaging and liquid biopsy techniques. Integrated radiomics, tissue, and plasma-based analyses, may potentially uncover immunotherapeutic response signatures, enabling early, adaptive therapeutic adjustments. By specifically targeting current therapeutic challenges, outcomes for GBM patients may potentially be improved.
Collapse
Affiliation(s)
- Aarav Badani
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neuroscience, University of California, Berkeley, CA, USA
| | - Ahmad Ozair
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mustafa Khasraw
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, NC, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Brain Tumor Center, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- University of Maryland - Medicine Institute for Neuroscience Discovery (UM-MIND), Baltimore, MD, USA
| | - Pallavi Tiwari
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- William S. Middleton Memorial Veterans Affairs (VA) Healthcare, Madison, WI, USA
| | - Manmeet S Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
21
|
Sabahi M, Fathi Jouzdani A, Sadeghian Z, Dabbagh Ohadi MA, Sultan H, Salehipour A, Maniakhina L, Rezaei N, Adada B, Mansouri A, Borghei-Razavi H. CAR-engineered NK cells versus CAR T cells in treatment of glioblastoma; strength and flaws. J Neurooncol 2025; 171:495-530. [PMID: 39538038 DOI: 10.1007/s11060-024-04876-z] [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: 09/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary brain tumor that carries a grim prognosis. Because of the dearth of treatment options available for treatment of GBM, Chimeric Antigen Receptor (CAR)-engineered T cell and Natural Killer (NK) therapy could provide alternative strategies to address the challenges in GBM treatment. In these approaches, CAR T and NK cells are engineered for cancer-specific immunotherapy by recognizing surface antigens independently of major histocompatibility complex (MHC) molecules. However, the efficacy of CAR T cells is hindered by GBM's downregulation of its targeted antigens. CAR NK cells face similar challenges, but, in contrast, they offer advantages as off-the-shelf allogeneic products, devoid of graft-versus-host disease (GVHD) risk as well as anti-cancer activity beyond CAR specificity, potentially reducing the risk of relapse or resistance. Despite CAR T cell therapies being extensively studied in clinical settings, the use of CAR-modified NK cells in GBM treatment remains largely in the preclinical stage. This review aims to discuss recent advancements in NK cell and CAR T cell therapies for GBM, including methods for introducing CARs into both NK cells and T cells, addressing manufacturing challenges, and providing evidence supporting the efficacy of these approaches from preclinical and early-phase clinical studies. The comprehensive evaluation of CAR-engineered NK cells and CAR T cells seeks to identify the optimal therapeutic approach for GBM, contributing to the development of effective immunotherapies for this devastating disease.
Collapse
Affiliation(s)
- Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Ali Fathi Jouzdani
- Neurosurgery Research Group (NRG), Hamadan University of Medical Sciences, Hamadan, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohre Sadeghian
- Department of Pathology & Laboratory Medicine, Cleveland Clinic Florida, Weston, FL, USA
| | | | - Hadi Sultan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Arash Salehipour
- Neurosurgery Research Group (NRG), Hamadan University of Medical Sciences, Hamadan, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Lana Maniakhina
- Department of Neurosurgery, Geisinger and Geisinger Commonwealth School of Medicine, Wilkes-Barre, PA, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Badih Adada
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | - Hamid Borghei-Razavi
- Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, FL, USA
| |
Collapse
|
22
|
Eckert T, Zobaer MS, Boulos J, Alexander-Bryant A, Baker TG, Rivers C, Das A, Vandergrift WA, Martinez J, Zukas A, Lindhorst SM, Patel S, Strickland B, Rowland NC. Immune Resistance in Glioblastoma: Understanding the Barriers to ICI and CAR-T Cell Therapy. Cancers (Basel) 2025; 17:462. [PMID: 39941829 PMCID: PMC11816167 DOI: 10.3390/cancers17030462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary malignant brain tumor, with fewer than 5% of patients surviving five years after diagnosis. The introduction of immune checkpoint inhibitors (ICIs), followed by chimeric antigen receptor (CAR) T-cell therapy, marked major advancements in oncology. Despite demonstrating efficacy in other blood and solid cancers, these therapies have yielded limited success in clinical trials for both newly diagnosed and recurrent GBM. A deeper understanding of GBM's resistance to immunotherapy is essential for enhancing treatment responses and translating results seen in other cancer models. OBJECTIVES In this review, we examine clinical trial outcomes involving ICIs and CAR-T for GBM patients and explore the evasive mechanisms of GBM and the tumor microenvironment. FINDINGS AND DISCUSSION Multiple clinical trials investigating ICIs in GBM have shown poor outcomes, with no significant improvement in progression-free survival (PFS) or overall survival (OS). Results from smaller case studies with CAR-T therapy have warranted further investigation. However, no large-scale trials or robust studies have yet established these immunotherapeutic approaches as definitive treatment strategies. Future research should shift focus from addressing the scarcity of functional T cells to exploiting the abundant myeloid-derived cells within the tumor microenvironment. CONCLUSIONS Translating these therapies into effective treatments for glioblastoma in humans remains a significant challenge. The highly immunosuppressive nature of GBM and its tumor microenvironment continue to hinder the success of these innovative immunotherapeutic approaches. Targeting the myeloid-derived compartment may lead to more robust and sustained immune responses.
Collapse
Affiliation(s)
- Thomas Eckert
- School of Medicine, University of South Carolina, Columbia, SC 29209, USA
- MUSC Institute for Neuroscience Discovery (MIND), Medical University of South Carolina, Charleston, SC 29425, USA; (M.S.Z.); (T.G.B.); (N.C.R.)
| | - MS Zobaer
- MUSC Institute for Neuroscience Discovery (MIND), Medical University of South Carolina, Charleston, SC 29425, USA; (M.S.Z.); (T.G.B.); (N.C.R.)
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
| | - Jessie Boulos
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; (J.B.); (A.A.-B.)
| | | | - Tiffany G. Baker
- MUSC Institute for Neuroscience Discovery (MIND), Medical University of South Carolina, Charleston, SC 29425, USA; (M.S.Z.); (T.G.B.); (N.C.R.)
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charlotte Rivers
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Arabinda Das
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
| | - William A. Vandergrift
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
| | - Jaime Martinez
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
| | - Alicia Zukas
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Scott M. Lindhorst
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sunil Patel
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
| | - Ben Strickland
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nathan C. Rowland
- MUSC Institute for Neuroscience Discovery (MIND), Medical University of South Carolina, Charleston, SC 29425, USA; (M.S.Z.); (T.G.B.); (N.C.R.)
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (A.D.); (W.A.V.); (J.M.); (A.Z.); (S.M.L.); (S.P.); (B.S.)
| |
Collapse
|
23
|
Su H, Peng Y, Wu Y, Zeng X. Overcoming immune evasion with innovative multi-target approaches for glioblastoma. Front Immunol 2025; 16:1541467. [PMID: 39911397 PMCID: PMC11794508 DOI: 10.3389/fimmu.2025.1541467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Glioblastoma (GBM) cells leverage complex endogenous and environmental regulatory mechanisms to drive proliferation, invasion, and metastasis. Tumor immune evasion, facilitated by a multifactorial network, poses a significant challenge to effective therapy, as evidenced by the limited clinical benefits of monotherapies, highlighting the adaptive nature of immune evasion. This review explores glioblastoma's immune evasion mechanisms, the role of ICIs in the tumor microenvironment, and recent clinical advancements, offering theoretical insights and directions for monotherapy and combination therapy in glioblastoma management.
Collapse
Affiliation(s)
- Hai Su
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Peng
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yilong Wu
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoli Zeng
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi “Flagship” Oncology Department of Synergy for Chinese and Western Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
24
|
Gong G, Jiang L, Zhou J, Su Y. Advancements in targeted and immunotherapy strategies for glioma: toward precision treatment. Front Immunol 2025; 15:1537013. [PMID: 39877359 PMCID: PMC11772277 DOI: 10.3389/fimmu.2024.1537013] [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: 11/29/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
In recent years, significant breakthroughs have been made in cancer therapy, particularly with the development of molecular targeted therapies and immunotherapies, owing to advances in tumor molecular biology and molecular immunology. High-grade gliomas (HGGs), characterized by their high malignancy, remain challenging to treat despite standard treatment regimens, including surgery, radiotherapy, chemotherapy, and tumor treating fields (TTF). These therapies provide limited efficacy, highlighting the need for novel treatment strategies. Molecular targeted therapies and immunotherapy have emerged as promising avenues for improving treatment outcomes in high-grade gliomas. This review explores the current status and recent advancements in targeted and immunotherapeutic approaches for high-grade gliomas.
Collapse
Affiliation(s)
- Guangyuan Gong
- Department of Intensive Care Medicine, Jiangsu Provincial People’s Hospital Chongqing Hospital (Qijiang District People’s Hospital), Chongqing, China
| | - Lang Jiang
- Department of Intensive Care Medicine, Jiangsu Provincial People’s Hospital Chongqing Hospital (Qijiang District People’s Hospital), Chongqing, China
| | - Jing Zhou
- Department of Thoracic Surgery, Jiangsu Provincial People’s Hospital Chongqing Hospital (Qijiang District People’s Hospital), Chongqing, China
| | - Yuanchao Su
- Department of Emergency Medicine, Jiangsu Provincial People’s Hospital Chongqing Hospital (Qijiang District People’s Hospital), Chongqing, China
| |
Collapse
|
25
|
Yuzhakova DV, Sachkova DA, Izosimova AV, Yashin KS, Yusubalieva GM, Baklaushev VP, Mozherov AM, Shcheslavskiy VI, Shirmanova MV. Fluorescence Lifetime Imaging of NAD(P)H in Patients' Lymphocytes: Evaluation of Efficacy of Immunotherapy. Cells 2025; 14:97. [PMID: 39851525 PMCID: PMC11764258 DOI: 10.3390/cells14020097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The wide variability in clinical responses to anti-tumor immunotherapy drives the search for personalized strategies. One of the promising approaches is drug screening using patient-derived models composed of tumor and immune cells. In this regard, the selection of an appropriate in vitro model and the choice of cellular response assay are critical for reliable predictions. Fluorescence lifetime imaging microscopy (FLIM) is a powerful, non-destructive tool that enables direct monitoring of cellular metabolism on a label-free basis with a potential to resolve metabolic rearrangements in immune cells associated with their reactivity. OBJECTIVE The aim of the study was to develop a patient-derived glioma explant model enriched by autologous peripheral lymphocytes and explore FLIM of the redox-cofactor NAD(P)H in living lymphocytes to measure the responses of the model to immune checkpoint inhibitors. METHODS The light microscopy, FLIM of NAD(P)H and flow cytometry were used. RESULTS The results demonstrate that the responsive models displayed a significant increase in the free NAD(P)H fraction α1 after treatment, associated with a shift towards glycolysis due to lymphocyte activation. The non-responsive models exhibited no alterations or a decrease in the NAD(P)H α1 after treatment. The FLIM data correlated well with the standard assays of immunotherapy drug response in vitro, including morphological changes, the T-cells activation marker CD69, and the tumor cell proliferation index Ki67. CONCLUSIONS The proposed platform that includes tumor explants co-cultured with lymphocytes and the NAD(P)H FLIM assay represents a promising solution for the patient-specific immunotherapeutic drug screening.
Collapse
Affiliation(s)
- Diana V. Yuzhakova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Daria A. Sachkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Anna V. Izosimova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Konstantin S. Yashin
- Department of Neurosurgery, Privolzsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia;
| | - Gaukhar M. Yusubalieva
- Federal Research and Clinical Center, Federal Medical and Biological Agency, 28 Orekhovy Blvd., 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
- Laboratory of Molecular Mechanisms of Regeneration and Aging, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Research and Clinical Center, Federal Medical and Biological Agency, 28 Orekhovy Blvd., 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
- Laboratory of Molecular Mechanisms of Regeneration and Aging, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., 119991 Moscow, Russia
| | - Artem M. Mozherov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Vladislav I. Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
- R&D Department, Becker&Hickl GmbH, 7-9 Nunsdorfer Ring, 12277 Berlin, Germany
| | - Marina V. Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| |
Collapse
|
26
|
de Melo SM, Elias Nunes da Silva ME, Torloni MR, Riera R, De Cicco K, Latorraca CO, Pinto ACPN. Anti-PD-1 and anti-PD-L1 antibodies for glioma. Cochrane Database Syst Rev 2025; 1:CD012532. [PMID: 39777725 PMCID: PMC11707826 DOI: 10.1002/14651858.cd012532.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and aggressive adult glioma (16-month median survival). Its immunosuppressive microenvironment limits the efficacy of immune checkpoint inhibitors (ICIs). OBJECTIVES To assess the effects of the ICIs antibodies anti-programmed cell death 1 (anti-PD-1) and anti-programmed cell death ligand 1 (anti-PD-L1) in treating adults with diffuse glioma. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, and clinical trials registers on 8 March 2024. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating adults with diffuse glioma treated with anti-PD-1/PD-L1 compared to placebo or other therapies used alone or with other ICIs. Primary outcomes were overall survival (OS), progression-free survival (PFS), and serious adverse events (SAE). Secondary outcomes were overall response rate (ORR), quality of life (QoL), and less serious AEs. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methods. MAIN RESULTS We included seven RCTs evaluating anti-PD-1 treatment in recurrent (N = 4) and newly diagnosed (N = 3) grade 4 glioma participants. The analysis encompassed 1953 participants; sample sizes ranged from 35 to 716. Meta-analyses were not possible due to heterogeneity and the small number of studies. Most trials had high risk of bias. Nivolumab versus bevacizumab in people with recurrent GBM (1 trial, 369 participants) Nivolumab probably does not increase OS (hazard ratio (HR) 1.04, 95% confidence interval (CI) 0.83 to 1.30; 1.3% more, 95% CI 6.30 fewer to 7.80 more; 369 participants; moderate-certainty evidence) or PFS (HR 1.97, 95% CI 1.57 to 2.48; 16.40% more, 95% CI 12.40 more to 19.00 more; 369 participants; moderate-certainty evidence). The evidence for SAE is very uncertain (risk ratio (RR) 1.20, 95% CI 0.74 to 1.92; 347 participants). Nivolumab probably does not increase ORR (RR 0.34, 95% CI 0.18 to 0.63; 309 participants; moderate-certainty evidence), but may not increase less serious AEs (RR 1.03, 95% CI 0.96 to 1.10; 347 participants; low-certainty evidence). Nivolumab plus bevacizumab 10 mg/kg versus nivolumab plus bevacizumab 3 mg/kg in people with recurrent GBM (1 trial, 90 participants) Nivolumab plus bevacizumab 10 mg/kg may not increase OS (HR 1.39, 95% CI 0.86 to 2.25; 9.90% more, 95% CI 5.20 fewer to 18.80 more; 90 participants; low-certainty evidence). The evidence for PFS (HR 1.23, 95% CI 0.78 to 1.93; 5.80% more, 95% CI 8.20 fewer to 14.20 more; 90 participants) and SAE (RR 1.19, 95% CI 0.79 to 1.79; 90 participants) is very uncertain. Nivolumab may not increase less serious AEs (RR 1.02, 95% CI 0.96 to 1.09; low-certainty evidence; 90 participants). Pembrolizumab plus bevacizumab versus pembrolizumab in people with recurrent GBM (1 trial, 80 participants) The evidence for OS (HR 1.03, 95% CI 0.65 to 1.63; 0.30% more, 95% CI 7.60 fewer to 2.90 more; 80 participants), PFS (HR 0.97, 95% CI 0.61 to 1.54: 0.40% fewer, 95% CI 9.20 fewer to 2.80 more; 80 participants), SAE (RR 1.32, 95% CI 0.75 to 2.42; 80 participants), and ORR (RR 12.76, 95% CI 0.77 to 210.27; 80 participants) is very uncertain. Pembrolizumab plus bevacizumab may not increase less serious AEs (RR 1.04, 95% CI 0.96 to 1.13; 80 participants; low-certainty evidence). Neoadjuvant (before surgical resection) and adjuvant (after surgical resection) pembrolizumab versus adjuvant-only pembrolizumab in people with recurrent GBM (1 trial, 35 participants) The evidence for OS (HR 0.39, 95% CI 0.17 to 0.92; 25.20% fewer, 95% CI 37.10 fewer to 2.10 fewer; 35 participants), PFS (HR 0.43, 95% CI 0.20 to 0.91; 30.10% fewer, 95% CI 52.20 fewer to 3.60 fewer; 35 participants), and SAE (RR 1.00, 95% CI 0.31 to 3.28; 32 participants) is very uncertain. Nivolumab plus radiotherapy versus temozolomide plus radiotherapy in people with newly diagnosed unmethylated GBM (1 trial, 560 participants) Nivolumab plus radiotherapy probably does not increase OS (HR 1.31, 95% CI 1.09 to 1.58 months; 8.30% more, 95% CI 2.80 more to 12.90 more; 560 participants) and PFS (HR 1.38, 95% CI 1.15 to 1.65 months; 7.50% more, 95% CI 3.60 more to 10.30 more; 560 participants; moderate-certainty evidence). The evidence for SAE is very uncertain (RR 0.87, 95% CI 0.65 to 1.18; 553 participants). It may not increase ORR (RR 1.08, 95% CI 0.43 to 2.69; 560 participants; low-certainty evidence) and probably does not increase less serious AEs (RR 1.00, 95% CI 0.96 to 1.04; 560 participants; moderate-certainty evidence). The evidence for time to deterioration of QoL is very uncertain (HR 0.76, 95% CI 0.59 to 0.99; 560 participants). Nivolumab plus temozolomide plus radiotherapy versus placebo plus temozolomide plus radiotherapy in people with newly diagnosed methylated GBM (1 trial, 716 participants) Nivolumab plus temozolomide plus radiotherapy probably does not increase OS (HR 1.10, 95% CI 0.92 to 1.32; 3.50 more, 95% CI 3.80 fewer to 9.60 more; 716 participants) and PFS (HR 1.10, 95% CI 0.92 to 1.32; 3.00 more, 95% CI 3.50 fewer to 7.90 more; 716 participants), and probably increases SAE (RR 2.91, 95% CI 2.05 to 4.12; 709 participants; moderate-certainty evidence). It does not increase less serious AEs (RR 1.02, 95% CI 1.00 to 1.04; 709 participants; high-certainty evidence). Adjuvant nivolumab plus temozolomide versus temozolomide in older people with GBM (1 trial, 103 participants) Nivolumab plus temozolomide probably does not increase OS (HR 0.85, 95% CI 0.54 to 1.33; 3.10 fewer, 95% CI 15.80 fewer to 3.60 more; 103 participants; moderate-certainty evidence) and PFS (HR 0.77, 95% CI 0.49 to 1.19; 5.40 fewer, 95% CI 19.10 fewer to 2.40 more; 103 participants; moderate-certainty evidence). The evidence for SAE is very uncertain (RR 1.58, 95% CI 0.88 to 2.81; 103 participants). The evidence for QoL is very uncertain (results only reported graphically; 103 participants). AUTHORS' CONCLUSIONS In recurrent GBM, nivolumab alone probably has no benefit. Anti-PD1 plus bevacizumab may also be ineffective based on low- to very low-certainty evidence. Neoadjuvant plus adjuvant pembrolizumab may improve OS and PFS, but this was based on only one small trial and very low-certainty evidence. In newly diagnosed GBM, nivolumab plus radiotherapy in unmethylated and plus radiotherapy plus temozolomide in methylated GBM probably has no benefit. In older participants, adjuvant nivolumab probably offers no benefit.
Collapse
Affiliation(s)
- Suely M de Melo
- Departamento de Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de Sao Paulo (UNIFESP), São Paulo, Brazil
- Neuro Oncologia, Hospital do Coração de São Paulo, São Paulo, Brazil
- Saúde Baseada em Evidências, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Rachel Riera
- Núcleo de Ensino e Pesquisa em Saúde Baseada em Evidências e Avaliação Tecnológica em Saúde (NEP-Sbeats) , Universidade Federal de São Paulo, São Paulo, Brazil
- Cochrane Afilliate Rio de Janeiro, Petrópolis, Brazil
- Center of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | | | - Ana Carolina Pereira Nunes Pinto
- Saúde Baseada em Evidências, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Cochrane Brazil, São Paulo, Brazil
- Iberoamerican Cochrane Centre - Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Departamento de Ciências Biológicas e da Saúde, Universidade Federal do Amapá, Macapá, Brazil
| |
Collapse
|
27
|
Sun AK, Fan S, Choi SW. Exploring Multiplex Immunohistochemistry (mIHC) Techniques and Histopathology Image Analysis: Current Practice and Potential for Clinical Incorporation. Cancer Med 2025; 14:e70523. [PMID: 39764760 PMCID: PMC11705464 DOI: 10.1002/cam4.70523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/10/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND By simultaneously staining multiple immunomarkers on a single tissue section, multiplexed immunohistochemistry (mIHC) enhances the amount of information that can be observed in a single tissue section and thus can be a powerful tool to visualise cellular interactions directly in the tumour microenvironment. Performing mIHC remains technically and practically challenging, and this technique has many limitations if not properly validated. However, with proper validation, heterogeneity between histopathological images can be avoided. AIMS This review aimed to summarize the currently used methods and to propose a standardised method for effective mIHC. MATERIALS AND METHODS An extensive literature review was conducted to identify different methods currently in use for mIHC. RESULTS Guidelines for antibody selection, panel design, antibody validation and analytical strategies are given. The advantages and disadvantages of each method are discussed. CONCLUSION This review summarizes widely used pathology imaging software and discusses the potential for automation of pathology image analysis so that mIHC technology can be a truly powerful tool for research as well as clinical use.
Collapse
Affiliation(s)
- Aria Kaiyuan Sun
- Department of Anaesthesiology, School of Clinical Medicine, Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalGuangzhouChina
| | - Siu Wai Choi
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Faculty of MedicineThe University of Hong KongHong KongHong Kong
| |
Collapse
|
28
|
Abdullaeva S, Chubarev V, Valeeva A, Preferanskaya N, Neganova M, Smolyarchuk E, Liu J, Sukocheva O, Samsonov M, Alyautdin R. Analysis of Clinical Success and Molecular Mechanisms of Action of Novel Anti-glioblastoma Drugs: A Review. Curr Med Chem 2025; 32:1082-1102. [PMID: 38299393 DOI: 10.2174/0109298673281283240101053940] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Gliomas and glioblastomas (GBM) are common primary malignant brain tumors, which are highly malignant and have a poor prognosis. The presence of cancer stem cells with unrestricted proliferative capacity and ability to generate glial neoplastic cells, the diffuse nature of GBM, and other specific factors of GBM contribute to poor results of drug therapy in patients with GBM. Despite the worldwide efforts to improve the treatment, many novel anti-GBM drugs are active just in vitro, in silico, and in preclinical trials, and they sometimes demonstrate poor or no activity in clinical trials. In this paper, we have casually selected and analyzed the most promising evidence-based results related to glioblastoma treatment at FDA and Clinical Trials.gov databases. It was observed that the most prospective trend in the development of anti-GBM drugs is combination therapy vs. monotherapy. Our analysis of clinical trials has allowed us to predict that the most promising combination therapy that has shown the best results in patient's surveillance should include drugs that block different growth-promoting signals in glioblastoma cells and that are activated by the V600E BRAF mutation. One drug should inhibit signals from the BRAF protein, whereas the second drug in combination should inhibit signals from the MEK protein. METHODS The content of this review is based on information obtained from PubMed, ClinicalTrials.- gov, and the U.S. Food and Drug Administration (https://www.fda.gov/). In ClinicalTrials.gov, we retrieved studies published from January 1, 2015. In the data search, "Glioblastoma" was used as the keyword. A study was deleted if it studied remedies for concomitant tumor diseases, as well as if it did not include descriptions of treatment methods and/or if GBM was not mentioned. The analysis of the effectiveness of treatment was carried out according to the increasing overall survival in GBM patients, compared to the gold standard for this cancer. RESULTS GBM patients treated with novel immunotherapy agents and drugs acting on epigenetic factors and receptor tyrosine kinase inhibitors have shown encouraging potential for future development in clinic. However, combinations of drugs have led to more significant improvements in the results and an increase in life expectancy of patients. For example, the combination of nivolumab and ipilimumab showed a 72% increase in life expectancy compared to using nivolumab alone (9.8 vs. 16.85). CONCLUSION Combining anti-GBM drugs appears to be a key direction for increasing treatment effectiveness and overall survival. Radiotherapy of GBM can increase the effect of combination drug therapy.
Collapse
Affiliation(s)
- Sabina Abdullaeva
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Vladimir Chubarev
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Anna Valeeva
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Nina Preferanskaya
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij pr. 1, Chernogolovka, 142432, Russian Federation
- Laboratory of Redox-activity Molecular System, FRC Kazan Scientific Center, Arbuzov Institute of Organic and Physical Chemistry, Russian Academy of Sciences, Akad. Arbuzov st. 8, Kazan, 420088, Russia
| | - Elena Smolyarchuk
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Mikhail Samsonov
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
- R-Pharm JSC, Leninsky pr., 111- 1, Moscow, 119421, Russian Federation
| | - Renad Alyautdin
- Department of Pharmacology, Sechenov University, Trubetskaya Street, 8-2, Moscow, 119991, Russian Federation
- Scientific Centre for Expert Evaluation of Medicinal Products, 8/2., Petrovsky Boulevard, Moscow, 127051, Russian Federation
| |
Collapse
|
29
|
Kesari S, Wojcinski A, Pabla S, Seager RJ, Gill JM, Carrillo JA, Wagle N, Park DJ, Nguyen M, Truong J, Takasumi Y, Chaiken L, Chang SC, Barkhoudarian G, Kelly DF, Juarez TM. Pre-radiation Nivolumab plus ipilimumab in patients with newly diagnosed high-grade gliomas. Oncoimmunology 2024; 13:2432728. [PMID: 39572979 PMCID: PMC11587836 DOI: 10.1080/2162402x.2024.2432728] [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: 06/17/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
The limited success of immune checkpoint inhibitors (ICIs) in the adjuvant setting for glioblastoma highlights the need to explore administering ICIs prior to immunosuppressive radiation. To address the feasibility and safety of this approach, we conducted a phase I study in patients with newly diagnosed Grade 3 and Grade 4 gliomas. Patients received nivolumab 300 mg every 2 weeks and ipilimumab 1 mg/kg every 6 weeks until disease progression or unacceptable toxicity. Fifteen patients were treated, with four patients on dexamethasone at treatment initiation and five tumors having MGMT promoter methylated. Treatment began a median of 38 days post-surgery. The most common treatment-related adverse events (AEs) were rash, pruritus, fatigue, nausea, and anorexia. Grade 3 AEs were lipase increased (n = 2), anorexia (n = 1), pruritus (n = 1), and rash (n = 3), and one Grade 4 cerebral edema occurred. Median progression-free survival (mPFS) was 1.3 months and median overall survival (mOS) was 19.3 months (95% CI, 12.9-NA). Three patients deferred conventional radiochemotherapy for over seven months while ten eventually received it. Progressing tumors tended to exhibit higher LAG-3 levels at baseline compared to shrinking tumors. Analysis of paired pre-treatment and post-progression tissue (n = 5) showed trends of up-regulated TGF-β, ERBB2, ERBB3, and ERBB4 signaling pathways, downregulated PPAR signaling, decreased B cell proportions, and increased monocytes proportions in tumors post-treatment. We show nivolumab plus ipilimumab can be safely administered prior to standard radiotherapy for newly diagnosed gliomas and is operationally feasible. Clinicaltrials.gov NCT03425292 registered February 7, 2018.
Collapse
Affiliation(s)
- Santosh Kesari
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | | | | | | | - Jaya M. Gill
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Jose A. Carrillo
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Naveed Wagle
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - David J. Park
- Providence St. Jude Medical Center, Department of Hematology and Oncology, Fullerton, CA, USA
| | - Minhdan Nguyen
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Judy Truong
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Yuki Takasumi
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Department of Pathology, Santa Monica, CA, USA
| | - Lisa Chaiken
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Department of Radiology, Santa Monica, CA, USA
| | - Shu-Ching Chang
- Providence St. Vincent Medical Center, Clinical Research Program Services, Portland, OR, USA
| | | | - Daniel F. Kelly
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Tiffany M. Juarez
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
- Saint John’s Cancer Institute, Translational Neurosciences, Santa Monica, CA, USA
- CureScience Institute, San Diego, CA, USA
| |
Collapse
|
30
|
Liu Y, Zhou F, Ali H, Lathia JD, Chen P. Immunotherapy for glioblastoma: current state, challenges, and future perspectives. Cell Mol Immunol 2024; 21:1354-1375. [PMID: 39406966 PMCID: PMC11607068 DOI: 10.1038/s41423-024-01226-x] [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: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive and lethal type of brain tumor in human adults. The standard of care offers minimal clinical benefit, and most GBM patients experience tumor recurrence after treatment. In recent years, significant advancements have been made in the development of novel immunotherapies or other therapeutic strategies that can overcome immunotherapy resistance in many advanced cancers. However, the benefit of immune-based treatments in GBM is limited because of the unique brain immune profiles, GBM cell heterogeneity, and immunosuppressive tumor microenvironment. In this review, we present a detailed overview of current immunotherapeutic strategies and discuss the challenges and potential molecular mechanisms underlying immunotherapy resistance in GBM. Furthermore, we provide an in-depth discussion regarding the strategies that can overcome immunotherapy resistance in GBM, which will likely require combination therapies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Fei Zhou
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Heba Ali
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
- Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA
| | - Peiwen Chen
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.
| |
Collapse
|
31
|
Wu J, Wang N. Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review). Mol Med Rep 2024; 30:221. [PMID: 39364736 PMCID: PMC11462401 DOI: 10.3892/mmr.2024.13344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/11/2023] [Indexed: 10/05/2024] Open
Abstract
Glioblastoma (GBM) is the most common central nervous system malignancy in adults. GBM may be classified as grade IV diffuse astrocytoma according to the 2021 World Health Organization revised classification of central nervous system tumors, which means it is the most aggressive, invasive, undifferentiated type of tumor. Immune checkpoint blockade (ICB), particularly anti‑programmed cell death protein‑1 (PD‑1)/PD‑1 ligand‑1 immunotherapy, has been confirmed to be successful across several tumor types. However, in GBM, this treatment is still uncommon and the efficacy is unpredictable, and <10% of patients show long‑term responses. Recently, numerous studies have been conducted to explore what factors may indicate or affect the ICB response rate in GBM, including molecular alterations, immune expression signatures and immune infiltration. The present review aimed to summarize the current progress to improve the understanding of immunotherapy for GBM.
Collapse
Affiliation(s)
- Jianheng Wu
- Department of Neurosurgery, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Nannan Wang
- Department of Gastroenterology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| |
Collapse
|
32
|
Liu J, Peng J, Jiang J, Liu Y. Clinical immunotherapy in glioma: current concepts, challenges, and future perspectives. Front Immunol 2024; 15:1476436. [PMID: 39555054 PMCID: PMC11564147 DOI: 10.3389/fimmu.2024.1476436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Glioma is one of the common tumors in the central nervous system, and its treatment methods (surgery, radiotherapy, and chemotherapy) lack specificity and have a poor prognosis. With the development of immunology, cell biology, and genomics, tumor immunotherapy has ushered in a new era of tumor therapy, achieving significant results in other invasive cancers such as advanced melanoma and advanced non-small cell lung cancer. Currently, the clinical trials of immunotherapy in glioma are also progressing rapidly. Here, this review summarizes promising immunotherapy methods in recent years, reviews the current status of clinical trials, and discusses the challenges and prospects of glioma immunotherapy.
Collapse
Affiliation(s)
- Jun Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Neurosurgery, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Jingjian Peng
- Department of Neurosurgery, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Jian Jiang
- Department of Neurosurgery, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Zhou QQ, Guo J, Wang Z, Li J, Chen M, Xu Q, Zhu L, Xu Q, Wang Q, Pan H, Pan J, Zhu Y, Song M, Liu X, Wang J, Zhang Z, Zhang L, Wang Y, Cai H, Chen X, Lu G. Rapid visualization of PD-L1 expression level in glioblastoma immune microenvironment via machine learning cascade-based Raman histopathology. J Adv Res 2024; 65:257-271. [PMID: 38072311 PMCID: PMC11519053 DOI: 10.1016/j.jare.2023.12.002] [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: 08/04/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 02/13/2024] Open
Abstract
INTRODUCTION Combination immunotherapy holds promise for improving survival in responsive glioblastoma (GBM) patients. Programmed death-ligand 1 (PD-L1) expression in immune microenvironment (IME) is the most important predictive biomarker for immunotherapy. Due to the heterogeneous distribution of PD-L1, post-operative histopathology fails to accurately capture its expression in residual tumors, making intra-operative diagnosis crucial for GBM treatment strategies. However, the current methods for evaluating the expression of PD-L1 are still time-consuming. OBJECTIVE To overcome the PD-L1 heterogeneity and enable rapid, accurate, and label-free imaging of PD-L1 expression level in GBM IME at the tissue level. METHODS We proposed a novel intra-operative diagnostic method, Machine Learning Cascade (MLC)-based Raman histopathology, which uses a coordinate localization system (CLS), hierarchical clustering analysis (HCA), support vector machine (SVM), and similarity analysis (SA). This method enables visualization of PD-L1 expression in glioma cells, CD8+ T cells, macrophages, and normal cells in addition to the tumor/normal boundary. The study quantified PD-L1 expression levels using the tumor proportion, combined positive, and cellular composition scores (TPS, CPS, and CCS, respectively) based on Raman data. Furthermore, the association between Raman spectral features and biomolecules was examined biochemically. RESULTS The entire process from signal collection to visualization could be completed within 30 min. In an orthotopic glioma mouse model, the MLC-based Raman histopathology demonstrated a high average accuracy (0.990) for identifying different cells and exhibited strong concordance with multiplex immunofluorescence (84.31 %) and traditional pathologists' scoring (R2 ≥ 0.9). Moreover, the peak intensities at 837 and 874 cm-1 showed a positive linear correlation with PD-L1 expression level. CONCLUSIONS This study introduced a new and extendable diagnostic method to achieve rapid and accurate visualization of PD-L1 expression in GBM IMB at the tissular level, leading to great potential in GBM intraoperative diagnosis for guiding surgery and post-operative immunotherapy.
Collapse
Affiliation(s)
- Qing-Qing Zhou
- Department of Radiology, Jinling Hospital, Affiliated Nanjing Medical University, Nanjing, China; Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Jingxing Guo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China.
| | - Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China; Nanjing Nuoyuan Medical Devices Co. Ltd, Nanjing, China
| | - Jianrui Li
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meng Chen
- Nanjing Nuoyuan Medical Devices Co. Ltd, Nanjing, China
| | - Qiang Xu
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lijun Zhu
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qing Xu
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiang Wang
- Department of Neurosurgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hao Pan
- Department of Neurosurgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jing Pan
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yong Zhu
- School of Science, China Pharmaceutical University, Nanjing, China
| | - Ming Song
- Department of Mathmatical Sciences, The University of Texas at Dallas, Richardson, USA
| | - Xiaoxue Liu
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiandong Wang
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhiqiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China; Nanjing Nuoyuan Medical Devices Co. Ltd, Nanjing, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Guangming Lu
- Department of Radiology, Jinling Hospital, Affiliated Nanjing Medical University, Nanjing, China; Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| |
Collapse
|
34
|
Chen J, Cui J, Jiao B, Zheng Z, Yu H, Wang H, Zhang G, Lai S, Gan Z, Yu Q. Biomimetic Nanosensitizer Potentiates Efficient Glioblastoma Gene-Radiotherapy through Synergistic Hypoxia Mitigation and PLK1 Silencing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58241-58261. [PMID: 39287499 DOI: 10.1021/acsami.4c11566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Postoperative radiotherapy currently stands as the cornerstone of glioblastoma (GBM) treatment. Nevertheless, low-dose radiotherapy has been proven ineffective for GBM, due to hypoxia in the GBM microenvironment, which renders the resistance to radiation-induced cell death. Moreover, the overexpression of the PLK1 gene in glioma cells enhances GBM proliferation, invasion, metastasis, and resistance to radiation. This study introduced a hybrid membrane-camouflaged biomimetic lipid nanosensitizer (CNL@miPA), which efficiently encapsulated gold nanoclusters (PA) and miR-593-5p by a chimeric membrane derived from lipids, cancer cells, and natural killer cells. CNL@miPA exhibited exceptional blood-brain barrier and tumor tissue penetration, effectively ameliorating hypoxia and synergizing with radiotherapy. By enabling prolonged miRNA circulation in the bloodstream and achieving high enrichment at the tumor site, CNL@miPA significantly suppressed tumor growth in combination treatment, thereby significantly extending the survival period of treated mice. Overall, the developed biomimetic nanosensitizer represented an efficient and multifunctional targeted delivery system, offering a novel strategy for gene-radiotherapy of GBM.
Collapse
Affiliation(s)
- Jiawei Chen
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajunzi Cui
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Binbin Jiao
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Ziyan Zheng
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Haiwang Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hanbing Wang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Guan Zhang
- Department of Urology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shicong Lai
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
- The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
35
|
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
|
36
|
Rodriguez SMB, Tataranu LG, Kamel A, Turliuc S, Rizea RE, Dricu A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int J Mol Sci 2024; 25:10765. [PMID: 39409094 PMCID: PMC11477435 DOI: 10.3390/ijms251910765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma is known to be one of the most aggressive and fatal human cancers, with a poor prognosis and resistance to standard treatments. In the last few years, many solid tumor treatments have been revolutionized with the help of immunotherapy. However, this type of treatment has failed to improve the results in glioblastoma patients. Effective immunotherapeutic strategies may be developed after understanding how glioblastoma achieves tumor-mediated immune suppression in both local and systemic landscapes. Biomarkers may help identify patients most likely to benefit from this type of treatment. In this review, we discuss the use of immunotherapy in glioblastoma, with an emphasis on immune checkpoint inhibitors and the factors that influence clinical response. A Pubmed data search was performed for all existing information regarding immune checkpoint inhibitors used for the treatment of glioblastoma. All data evaluating the ongoing clinical trials involving the use of ICIs either as monotherapy or in combination with other drugs was compiled and analyzed.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Amira Kamel
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania;
| |
Collapse
|
37
|
Ghosh S, Rothlin CV. Feeding the wrath with myelin. Trends Immunol 2024; 45:729-731. [PMID: 39341708 PMCID: PMC11471388 DOI: 10.1016/j.it.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
Kloosterman and colleagues studied molecular and cellular changes during radiation therapy and disease recurrence across molecular subtypes of glioblastoma. They uncovered a distinct immune-cancer cell metabolic crosstalk during proneural/oligodendrocyte progenitor cell-like to mesenchymal-like transition, wherein macrophages feed on cholesterol-rich myelin debris to provide lipids to mesenchymal tumor cells, thereby fueling glioblastoma growth.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Carla V Rothlin
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
38
|
Dobersalske C, Rauschenbach L, Hua Y, Berliner C, Steinbach A, Grüneboom A, Kokkaliaris KD, Heiland DH, Berger P, Langer S, Tan CL, Stenzel M, Landolsi S, Weber F, Darkwah Oppong M, Werner RA, Gull H, Schröder T, Linsenmann T, Buck AK, Gunzer M, Stuschke M, Keyvani K, Forsting M, Glas M, Kipnis J, Steindler DA, Reinhardt HC, Green EW, Platten M, Tasdogan A, Herrmann K, Rambow F, Cima I, Sure U, Scheffler B. Cranioencephalic functional lymphoid units in glioblastoma. Nat Med 2024; 30:2947-2956. [PMID: 39085419 PMCID: PMC11485206 DOI: 10.1038/s41591-024-03152-x] [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: 06/26/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
The ecosystem of brain tumors is considered immunosuppressed, but our current knowledge may be incomplete. Here we analyzed clinical cell and tissue specimens derived from patients presenting with glioblastoma or nonmalignant intracranial disease to report that the cranial bone (CB) marrow, in juxtaposition to treatment-naive glioblastoma tumors, harbors active lymphoid populations at the time of initial diagnosis. Clinical and anatomical imaging, single-cell molecular and immune cell profiling and quantification of tumor reactivity identified CD8+ T cell clonotypes in the CB that were also found in the tumor. These were characterized by acute and durable antitumor response rooted in the entire T cell developmental spectrum. In contrast to distal bone marrow, the CB niche proximal to the tumor showed increased frequencies of tumor-reactive CD8+ effector types expressing the lymphoid egress marker S1PR1. In line with this, cranial enhancement of CXCR4 radiolabel may serve as a surrogate marker indicating focal association with improved progression-free survival. The data of this study advocate preservation and further exploitation of these cranioencephalic units for the clinical care of glioblastoma.
Collapse
Affiliation(s)
- Celia Dobersalske
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
| | - Laurèl Rauschenbach
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
- Center for Translational Neuroscience and Behavioral Science (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Yichao Hua
- Department of Applied Computational Cancer Research, IKIM, University Hospital Essen, Essen, Germany
| | - Christoph Berliner
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Anita Steinbach
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Konstantinos D Kokkaliaris
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- DKTK, German Cancer Consortium, partner site Frankfurt/Mainz, Quantitative Spatial Cancer Biology Laboratory, University Hospital Frankfurt, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Dieter H Heiland
- DKTK, German Cancer Consortium, partner site Freiburg, Translational Neurosurgery, Microenvironment and Immunology Research Laboratory, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University Clinic Erlangen, Erlangen, Germany
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pia Berger
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
| | - Sarah Langer
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
| | - Chin L Tan
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- DKTK, German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
| | - Martin Stenzel
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Somaya Landolsi
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- DKTK, German Cancer Consortium, partner site Frankfurt/Mainz, Quantitative Spatial Cancer Biology Laboratory, University Hospital Frankfurt, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Flora Weber
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Marvin Darkwah Oppong
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
- Center for Translational Neuroscience and Behavioral Science (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- University Hospital Frankfurt, Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
- The Russell H. Morgan Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hanah Gull
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
- Center for Translational Neuroscience and Behavioral Science (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Thomas Schröder
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Thomas Linsenmann
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Matthias Gunzer
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Radiation Oncology, University Hospital Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University Hospital Essen, Essen, Germany
| | - Michael Forsting
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Martin Glas
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Center for Translational Neuroscience and Behavioral Science (C-TNBS), University of Duisburg-Essen, Essen, Germany
- Department of Neurology, Division of Neurooncology, University Hospital Essen, Essen, Germany
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - Dennis A Steindler
- Steindler Consulting, Boston, MA, USA
- The Eshelman Institute for Innovation, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hans Christian Reinhardt
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
- Center of Medical Biotechnology (ZMB), University Duisburg-Essen, Essen, Germany
| | - Edward W Green
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- DKTK, German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
| | - Michael Platten
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- DKTK, German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz, Germany
- German Cancer Research Center-Hector Cancer Institute at the Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alpaslan Tasdogan
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Center of Medical Biotechnology (ZMB), University Duisburg-Essen, Essen, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Florian Rambow
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Applied Computational Cancer Research, IKIM, University Hospital Essen, Essen, Germany
- Center of Medical Biotechnology (ZMB), University Duisburg-Essen, Essen, Germany
| | - Igor Cima
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany
| | - Ulrich Sure
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
- Center for Translational Neuroscience and Behavioral Science (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Björn Scheffler
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, a partnership between DKFZ and University Hospital Essen, University Duisburg-Essen, Essen, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- DKFZ Division Translational Neurooncology at the WTZ, University Medicine Essen, Essen, Germany.
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany.
- Center of Medical Biotechnology (ZMB), University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
39
|
Hotchkiss KM, Karschnia P, Schreck KC, Geurts M, Cloughesy TF, Huse J, Duke ES, Lathia J, Ashley DM, Nduom EK, Long G, Singh K, Chalmers A, Ahluwalia MS, Heimberger A, Bagley S, Todo T, Verhaak R, Kelly PD, Hervey-Jumper S, de Groot J, Patel A, Fecci P, Parney I, Wykes V, Watts C, Burns TC, Sanai N, Preusser M, Tonn JC, Drummond KJ, Platten M, Das S, Tanner K, Vogelbaum MA, Weller M, Whittle JR, Berger MS, Khasraw M. A brave new framework for glioma drug development. Lancet Oncol 2024; 25:e512-e519. [PMID: 39362262 PMCID: PMC11983286 DOI: 10.1016/s1470-2045(24)00190-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 10/05/2024]
Abstract
Patients with brain tumours are motivated to participate in clinical trials involving repeat tissue sampling. Normalising the use of neoadjuvant and staged surgical trials necessitates collaboration among patients, regulatory agencies, and researchers. Initial and repetitive tissue sampling plays a crucial role in enhancing our understanding of resistance mechanisms and vulnerabilities in brain tumour therapy. Standardising biopsy techniques and ensuring technical uniformity across institutions are vital for effective interinstitutional collaboration. Although liquid biopsy technologies hold promise, they are not yet ready to replace tissue analysis. Clear communication about the risks and benefits of biopsies is essential, particularly regarding potential postoperative deficits. Changes in mindset and neurosurgical culture are imperative to achieve much needed breakthroughs in the development of new, effective therapies for brain tumours.
Collapse
Affiliation(s)
- Kelly M Hotchkiss
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Philipp Karschnia
- Department of Neurosurgery, Friedrich-Alexander University, Erlangen, Germany; Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Karisa C Schreck
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marjolein Geurts
- Department of Neurology, Erasmus MC Cancer Center, Rotterdam, Netherlands; Department of Medical Oncology, Erasmus MC Cancer Center, Rotterdam, Netherlands
| | | | - Jason Huse
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth S Duke
- Division of Oncology 2, Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Justin Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Edjah K Nduom
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Georgina Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Mater and Royal North Shore Hospitals, Sydney, NSW, Australia
| | - Kirit Singh
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | | | | | - Amy Heimberger
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | - Stephen Bagley
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tomoki Todo
- Department of Surgical Neuro-Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Roel Verhaak
- Department of Surgical Neuro-Oncology, Yale University, New Haven, CT, USA
| | - Patrick D Kelly
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shawn Hervey-Jumper
- Department of Neurological Surgery, UCSF Brain Tumor Center, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - John de Groot
- Department of Neurological Surgery, UCSF Brain Tumor Center, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Anoop Patel
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Peter Fecci
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Ian Parney
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Victoria Wykes
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK; Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Colin Watts
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK; Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Terry C Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Nader Sanai
- Barrow Neurological Institute, Phoenix, AZ, USA
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Joerg Christian Tonn
- Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Katharine J Drummond
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Surgery, University of Melbourne, Parkville, VIC, Australia
| | - Michael Platten
- Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; Clinical Cooperation Unit Brain Tumor Immunology (D170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sunit Das
- Division of Neurosurgery, University of Toronto, Toronto, ON, Canada
| | - Kirk Tanner
- National Brain Tumor Society, Newton, MA, USA
| | | | - Michael Weller
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA; Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - James R Whittle
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Personalised Oncology Division, Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Mitchel S Berger
- Department of Neurological Surgery, UCSF Brain Tumor Center, University of California San Francisco, San Francisco, CA, USA; Weill Institute for Neuroscience, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
40
|
Xing Y, Yasinjan F, Geng H, He M, Yang M, Gao Y, Zhang J, Zhang L, Guo B. A scientometric analysis of immunotherapies for gliomas: Focus on GBM. Asian J Surg 2024; 47:4271-4280. [PMID: 38448290 DOI: 10.1016/j.asjsur.2024.02.138] [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: 09/20/2023] [Revised: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024] Open
Abstract
Gliomas are the most prevalent primary malignant brain tumors worldwide, with glioblastoma (GBM) being the most common and aggressive type. The standard therapy for GBM has remained unchanged for nearly two decades, with no significant improvement in survival outcomes. Despite several barriers such as the tumor microenvironment (TME) and blood-brain barrier, immunotherapies bring new hope for the treatment of GBM. To better understand the development and progress of immunotherapies in GBM, we made this scientometric analysis of this field. A total of 3753 documents were obtained from the Web of Science Core Collection, with publication years ranging from 1999 to 2022. The Web of Science platform, CiteSpace, and VOS viewer were used to conduct the scientometric analysis. The results of scientometric analysis showed that this field has recently become a popular topic of interest. The United States had the most publications among 89 countries or regions. Keyword analysis indicated significant areas in the field of immunotherapies for GBM, especially TME, immune checkpoint blockades (ICBs), chimeric antigen receptor T (CAR-T) cells, vaccines, and oncolytic viruses (OVs). Overall, we hope that this scientometric analysis can provide insights for researchers and promote the development of this field.
Collapse
Affiliation(s)
- Yang Xing
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Feroza Yasinjan
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Huayue Geng
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Minghua He
- College of Computer Science and Technology, Jilin University, ChangChun, China
| | - Mei Yang
- Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yufei Gao
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Jinnan Zhang
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, China.
| | - Ling Zhang
- Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China.
| |
Collapse
|
41
|
Inocencio JF, Mitrasinovic S, Asad M, Parney IF, Zang X, Himes BT. Immune checkpoint pathways in glioblastoma: a diverse and evolving landscape. Front Immunol 2024; 15:1424396. [PMID: 39346924 PMCID: PMC11427296 DOI: 10.3389/fimmu.2024.1424396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Immune checkpoint (IC) inhibition in glioblastoma (GBM) has not shown promising results in the last decade compared to other solid tumors. Several factors contributing to the lack of immunotherapy response include the profound immunosuppressive nature of GBM, highly redundant signaling pathways underlying immune checkpoints, and the negative immunogenic impact of current standard of care on the tumor microenvironment. In this review, we will discuss various ICs in the context of GBM, their interplay with the tumor immune microenvironment, relevant pre-clinical and clinical studies, and the impact of current treatment modalities on GBM IC blockade therapy. Understanding the molecular mechanisms that drive ICs, and how they contribute to an immunosuppressive tumor microenvironment is critical in advancing IC inhibition therapy in GBM. Furthermore, revisiting current treatment modalities and their impact on the immune landscape is instrumental in designing future combinatorial therapies that may overcome treatment resistance.
Collapse
Affiliation(s)
- Julio F Inocencio
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Stefan Mitrasinovic
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mohammad Asad
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Benjamin T Himes
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
42
|
Skadborg SK, Maarup S, Draghi A, Borch A, Hendriksen S, Mundt F, Pedersen V, Mann M, Christensen IJ, Skjøth-Ramussen J, Yde CW, Kristensen BW, Poulsen HS, Hasselbalch B, Svane IM, Lassen U, Hadrup SR. Nivolumab Reaches Brain Lesions in Patients with Recurrent Glioblastoma and Induces T-cell Activity and Upregulation of Checkpoint Pathways. Cancer Immunol Res 2024; 12:1202-1220. [PMID: 38885356 PMCID: PMC11369628 DOI: 10.1158/2326-6066.cir-23-0959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/10/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor with poor prognosis. Although immunotherapy is being explored as a potential treatment option for patients with GBM, it is unclear whether systemic immunotherapy can reach and modify the tumor microenvironment in the brain. We evaluated immune characteristics in patients receiving the anti-PD-1 immune checkpoint inhibitor nivolumab 1 week prior to surgery, compared with control patients receiving salvage resection without prior nivolumab treatment. We observed saturating levels of nivolumab bound to intratumorally and tissue-resident T cells in the brain, implicating saturating levels of nivolumab reaching brain tumors. Following nivolumab treatment, significant changes in T-cell activation and proliferation were observed in the tumor-resident T-cell population, and peripheral T cells upregulated chemokine receptors related to brain homing. A strong nivolumab-driven upregulation in compensatory checkpoint inhibition molecules, i.e., TIGIT, LAG-3, TIM-3, and CTLA-4, was observed, potentially counteracting the treatment effect. Finally, tumor-reactive tumor-infiltrating lymphocytes (TIL) were found in a subset of nivolumab-treated patients with prolonged survival, and neoantigen-reactive T cells were identified in both TILs and blood. This indicates a systemic response toward GBM in a subset of patients, which was further boosted by nivolumab, with T-cell responses toward tumor-derived neoantigens. Our study demonstrates that nivolumab does reach the GBM tumor lesion and enhances antitumor T-cell responses both intratumorally and systemically. However, various anti-inflammatory mechanisms mitigate the clinical efficacy of the anti-PD-1 treatment.
Collapse
Affiliation(s)
- Signe K. Skadborg
- Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Simone Maarup
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- National Center for Cancer Immune Therapy, CCIT-DK, Copenhagen University Hospital, Herlev, Denmark.
| | - Arianna Draghi
- National Center for Cancer Immune Therapy, CCIT-DK, Copenhagen University Hospital, Herlev, Denmark.
| | - Annie Borch
- Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Sille Hendriksen
- Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Filip Mundt
- Novo Nordisk Foundation Center for Protein Research, CPR, University of Copenhagen, Copenhagen, Denmark.
| | - Vilde Pedersen
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Pathology, The Bartholin Institute, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, CPR, University of Copenhagen, Copenhagen, Denmark.
- Research Department Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | - Ib J. Christensen
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Jane Skjøth-Ramussen
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Neurosurgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Christina W. Yde
- Center for Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Bjarne W. Kristensen
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Pathology, The Bartholin Institute, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| | - Hans S. Poulsen
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Benedikte Hasselbalch
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Inge M. Svane
- National Center for Cancer Immune Therapy, CCIT-DK, Copenhagen University Hospital, Herlev, Denmark.
| | - Ulrik Lassen
- Department of Oncology, DCCC Brain Tumor Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Sine R. Hadrup
- Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
43
|
Chen J, Liang RS, Zhuang BB, Chen HD, Liu S, Zhang GL, Shi SS. Cordycepin inhibits glioma growth by downregulating PD-L1 expression via the NOD-like receptor/NFKB1/STAT1 axis. Chem Biol Interact 2024; 400:111178. [PMID: 39084503 DOI: 10.1016/j.cbi.2024.111178] [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: 05/23/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024]
Abstract
Glioma is a serious primary malignant tumor of the human central nervous system with a poor prognosis and a high recurrence rate; however, inhibition of immune checkpoints can greatly improve the survival rate of patients. The purpose of this study was to investigate the regulation of PD-L1 by cordycepin and the mechanism of its anti-tumor action. The results of previous studies indicate that cordycepin has good anti-proliferative and anti-migratory activities and can induce apoptosis in U251 and T98G cells in vitro. Here, transcriptome sequencing showed that cordycepin may exert anti-tumor effects through the NOD-like receptor signaling pathway. Further intervention with BMS-1, a small molecule inhibitor of PD-L1, was used to explore whether inhibition of PD-L1 affected the regulation of the NOD-like receptor signaling pathway by cordycepin. Mechanistically, on the one hand, cordycepin regulated the expression of NFKB1 and STAT1 through the NOD-like receptor signaling pathway, thereby inhibiting the expression of PD-L1. In addition, inhibition of PD-L1 enhanced the regulation by cordycepin of the NOD-like receptor signaling pathway. On the other hand, cordycepin directly upregulated expression of STAT1 and downregulated that of PD-L1. In vivo studies further showed that cordycepin could downregulate expression of PD-L1 and NFKB1 and upregulate that of STAT1 in glioma xenograft tumor tissues, consistent with the results of in vitro studies. The results suggest that cordycepin may down-regulate the expression of PD-L1 through NOD-like receptor signaling pathway and NFKB signaling pathway, thereby inhibiting the immune escape of glioma, and can be developed as a PD-L1 inhibitor. Our results therefore provide a theoretical foundation for the use of cordycepin in treatment of glioma and enrich our understanding of its pharmacological mechanism.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China.
| | - Ri-Sheng Liang
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Bing-Bo Zhuang
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Hao-Dong Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Shuang Liu
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Guo-Liang Zhang
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29# Xinquan Road, Fuzhou, 350001, Fujian, China
| |
Collapse
|
44
|
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
|
45
|
Wang Y, Zhang Y, Chen Y, Wang S, Liu W, Liu Z, Hu M. [ 18F]AlF-NOTA-PCP2: a novel PET/CT tracer for enhanced PD-L1 heterogeneity imaging and comparative analysis with [ 18F]AlF-NOTA-WL12 in glioblastoma xenografts. Eur J Nucl Med Mol Imaging 2024; 51:3161-3175. [PMID: 38713298 DOI: 10.1007/s00259-024-06743-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE The unsatisfactory efficacy of PD-L1 antibodies in glioblastoma (GBM) is largely due to the temporal and spatial heterogeneity of PD-L1 expression. Molecular imaging can enhance understanding of the tumor immune microenvironment and guide immunotherapy. However, highly sensitive imaging agents capable of effectively visualizing PD-L1 heterogeneity are limited. This study introduces a novel PET tracer, offering improved imaging of PD-L1 heterogeneity in GBM xenografts, with a comparative analysis to [18F]AlF-NOTA-WL12. METHODS [18F]AlF-NOTA-PCP2 was synthesized with high purity and its affinity for PD-L1 was characterized using surface plasmon resonance (SPR) and cell binding assays. Its specificity for PD-L1 was evaluated both in vitro using various cell lines and in vivo with GBM xenograft models in NOD/SCID mice. PET/CT imaging was conducted to evaluate the tracer's biodistribution, pharmacokinetics, and ability to quantify tumoral spatial heterogeneity of PD-L1 expression. A focused comparative analysis between [18F]AlF-NOTA-PCP2 and [18F]AlF-NOTA-WL12 was conducted, examining binding affinity, biodistribution, pharmacokinetics, and imaging effectiveness in GBM xenografts. Additionally, human radiation dosimetry estimates compared the safety profiles of both tracers. RESULTS [18F]AlF-NOTA-PCP2 demonstrated high radiochemical purity (> 95%) and a strong affinity for PD-L1, comparable to [18F]AlF-NOTA-WL12. In vitro and in vivo studies confirmed its specificity for PD-L1, with increased uptake in PD-L1 expressing cells and tumors. Toxicological profiles indicated no significant abnormalities in serum biochemical indicators or major organ tissues. MicroPET/CT imaging showed [18F]AlF-NOTA-PCP2's effectiveness in visualizing PD-L1 expression levels and spatial heterogeneity in GBM xenografts. Comparative studies revealed [18F]AlF-NOTA-PCP2's improved pharmacokinetic properties, including higher tumor-to-blood ratios and lower nonspecific liver uptake, as well as reduced radiation exposure compared to [18F]AlF-NOTA-WL12. CONCLUSION [18F]AlF-NOTA-PCP2 distinguishes itself as an exceptionally sensitive PET/CT tracer, adept at non-invasively and accurately quantifying PD-L1 expression and its spatial heterogeneity in tumors, especially in GBM. Its favorable pharmacokinetic properties, safety profile, and high affinity for PD-L1 highlight its potential for enhancing the precision of cancer immunotherapy and guiding individualized treatment strategies. While promising, its clinical translation, especially in brain imaging, necessitates further validation in clinical trials.
Collapse
Affiliation(s)
- Yong Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yunhao Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shijie Wang
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wei Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Zhiguo Liu
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
46
|
Ius T, Somma T, Pasqualetti F, Berardinelli J, Vitulli F, Caccese M, Cella E, Cenciarelli C, Pozzoli G, Sconocchia G, Zeppieri M, Gerardo C, Caffo M, Lombardi G. Local therapy in glioma: An evolving paradigm from history to horizons (Review). Oncol Lett 2024; 28:440. [PMID: 39081966 PMCID: PMC11287108 DOI: 10.3892/ol.2024.14573] [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: 02/13/2024] [Accepted: 06/14/2024] [Indexed: 08/02/2024] Open
Abstract
Despite the implementation of multimodal treatments after surgery, glioblastoma (GBM) remains an incurable disease, posing a significant challenge in neuro-oncology. In this clinical setting, local therapy (LT), a developing paradigm, has received significant interest over time due to its potential to overcome the drawbacks of conventional therapy options for GBM. The present review aimed to trace the historical development, highlight contemporary advances and provide insights into the future horizons of LT in GBM management. In compliance with the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols criteria, a systematic review of the literature on the role of LT in GBM management was conducted. A total of 2,467 potentially relevant articles were found and, after removal of duplicates, 2,007 studies were screened by title and abstract (Cohen's κ coefficient=0.92). Overall, it emerged that 15, 10 and 6 clinical studies explored the clinical efficiency of intraoperative local treatment modalities, local radiotherapy and local immunotherapy, respectively. GBM recurrences occur within 2 cm of the radiation field in 80% of cases, emphasizing the significant influence of local factors on recurrence. This highlights the urgent requirement for LT strategies. In total, three primary reasons have thus led to the development of numerous LT solutions in recent decades: i) Intratumoral implants allow the blood-brain barrier to be bypassed, resulting in limited systemic toxicity; ii) LT facilitates bridging therapy between surgery and standard treatments; and iii) given the complexity of GBM, targeting multiple components of the tumor microenvironment through ligands specific to various elements could have a synergistic effect in treatments. Considering the spatial and temporal heterogeneity of GBM, the disease prognosis could be significantly improved by a combination of therapeutic strategies in the era of precision medicine.
Collapse
Affiliation(s)
- Tamara Ius
- Unit of Neurosurgery, Head-Neck and Neurosciences Department, University Hospital of Udine, I-33100 Udine, Italy
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | | | - Jacopo Berardinelli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | - Francesca Vitulli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | - Mario Caccese
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
| | - Eugenia Cella
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
- Medical Oncology 2, San Martino Hospital-IRCCS, I-16131 Genoa Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council, I-00133 Roma, Italy
| | - Giacomo Pozzoli
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, I-00168 Rome, Italy
| | - Giuseppe Sconocchia
- Institute of Translational Pharmacology, National Research Council, I-00133 Roma, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, I-33100 Udine, Italy
| | - Caruso Gerardo
- Unit of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University Hospital of Messina, I-98125 Messina, Italy
| | - Maria Caffo
- Unit of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University Hospital of Messina, I-98125 Messina, Italy
| | - Giuseppe Lombardi
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
| |
Collapse
|
47
|
de Dios O, Ramírez-González MA, Gómez-Soria I, Segura-Collar B, Manosalva J, Megías D, De Andrea CE, Fernández-Rubio L, Hernández-Laín A, Sepúlveda-Sánchez JM, Rodriguez-Ruiz ME, Pérez-Núñez Á, Wainwright DA, Gargini R, Sánchez-Gómez P. NKG2C/ KLRC2 tumor cell expression enhances immunotherapeutic efficacy against glioblastoma. J Immunother Cancer 2024; 12:e009210. [PMID: 39214651 PMCID: PMC11367385 DOI: 10.1136/jitc-2024-009210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Activating and inhibitory receptors of natural killer (NK) cells such as NKp, NKG2, or CLEC are highly relevant to cold tumors including glioblastoma (GBM). Here, we aimed to characterize the expression of these receptors in GBM to gain insight into their potential role as modulators of the intratumoral microenvironment. METHODS We performed a transcriptomic analysis of several NK receptors with a focus on the activating receptor encoded by KLRC2, NKG2C, among bulk and single-cell RNA sequencing GBM data sets. We also evaluated the effects of KLRC2-overexpressing GL261 cells in mice treated with or without programmed cell death protein-1 (PD-1) monoclonal antibody (mAb). Finally, we analyzed samples from two clinical trials evaluating PD-1 mAb effects in patients with GBM to determine the potential of NKG2C to serve as a biomarker of response. RESULTS We observed significant expression of several inhibitory NK receptors on GBM-infiltrating NK and T cells, which contrasts with the strong expression of KLRC2 on tumor cells, mainly at the infiltrative margin. Neoplastic KLRC2 expression was associated with a reduction in the number of myeloid-derived suppressor cells and with a higher level of tumor-resident lymphocytes. A stronger antitumor activity after PD-1 mAb treatment was observed in NKG2Chigh-expressing tumors both in mouse models and patients with GBM whereas the expression of inhibitory NK receptors showed an inverse association. CONCLUSIONS This study explored the role of neoplastic NKG2C/KLRC2 expression in shaping the immune profile of GBM and suggests that it is a predictive biomarker for positive responses to immune checkpoint inhibitor treatment in patients with GBM. Future studies could further validate this finding in prospective trials.
Collapse
Affiliation(s)
- Olaya de Dios
- Neurooncology Unit, Chronic Disease Deparment (UFIEC), Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - M Angeles Ramírez-González
- Neurooncology Unit, Chronic Disease Deparment (UFIEC), Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Irene Gómez-Soria
- Neurooncology Unit, Chronic Disease Deparment (UFIEC), Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Berta Segura-Collar
- Neurooncology Unit, Instituto de Investigaciones Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Anatomical Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Juliana Manosalva
- Advanced Microscopy Unit, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Diego Megías
- Advanced Microscopy Unit, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Carlos E De Andrea
- Department of Anatomy, Physiology and Pathology, Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Leticia Fernández-Rubio
- Division of Immunology and Immunotherapy, Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Pamplona, Navarra, Spain
| | - Aurelio Hernández-Laín
- Neurooncology Unit, Instituto de Investigaciones Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Neuropathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Juan M Sepúlveda-Sánchez
- Neurooncology Unit, Instituto de Investigaciones Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- Hospital HM Sanchinarro, Centro Integral Oncologico Clara Campal, Madrid, Spain
| | - Maria E Rodriguez-Ruiz
- Division of Immunology and Immunotherapy, Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Pamplona, Navarra, Spain
- Department of Radiation Oncology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Ángel Pérez-Núñez
- Department of Neurosurgery, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Surgery, Universidad Complutense de Madrid, Facultad de Medicina, Madrid, Spain
| | - Derek A Wainwright
- Department of Neurological Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Ricardo Gargini
- Neurooncology Unit, Instituto de Investigaciones Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Anatomical Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pilar Sánchez-Gómez
- Neurooncology Unit, Chronic Disease Deparment (UFIEC), Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| |
Collapse
|
48
|
Lv K, Du X, Chen C, Yu Y. Research hotspots and trend of glioblastoma immunotherapy: a bibliometric and visual analysis. Front Oncol 2024; 14:1361530. [PMID: 39175478 PMCID: PMC11339877 DOI: 10.3389/fonc.2024.1361530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Background Glioblastoma (GBM) is one of the common malignant tumors of the central nervous system (CNS), characterized by rapid proliferation, heterogeneity, aggressiveness, proneness to recurrence after surgery, and poor prognosis. There is increasing evidence that tumorigenesis is inextricably linked to immune escape, and immunotherapy is undoubtedly an important complement to clinical treatment options for GBM, and will be a focus and hot topic in GBM treatment research. The purpose of this study was to visualize and analyze the scientific results and research trends of immunotherapy for GBM. Methods Publications concerning immunotherapy for GBM were retrieved from the Web of Science Core Collection (WOScc) database. Bibliometric and visual analysis was performed mainly using CiteSpace and R software, and the Online Analysis Platform of Literature Metrology (https://bibliometric.com/app) for countries/regions, authors, journals, references and keywords related to publications in the field. Results Among totally 3491 publications retrieved in this field, 1613 publications were finally obtained according to the screening criteria, including 1007 articles (62.43%) and 606 reviews (37.57%). The number of publications increased year by year, with an average growth rate (AGR) of 17.41%. Such a number was the largest in the USA (717, 44.45%), followed by China (283, 17.55%), and the USA showed the strongest international collaboration. Among the research institutions, Duke Univ (94, 5.83%) was the largest publisher in the field, followed by Harvard Med Sch (70, 4.34%). In addition, the most prolific authors in this field were OHN H SAMPSON (51) and MICHAEL LIM (43), and the degree of collaboration (DC) between authors was 98.26%. Among the co-cited authors, STUPP R (805) was the most cited author, followed by REARDON DA (448). The journal with the most published publications was FRONTIERS IN IMMUNOLOGY (75), and the most cited journal in terms of co-citation was CLIN CANCER RES (1322), followed by CANCER RES (1230). The high-frequency keyword included glioblastoma (672) and immunotherapy (377). Cluster analysis was performed on the basis of keyword co-occurrence analysis, yielding 17 clusters, based on which the current research status and future trends in the field of immunotherapy for GBM were identified. Conclusion Immunotherapy is currently a novel treatment strategy for GBM that has attracted much attention. In the future, it is necessary to strengthen cooperation and exchanges between countries and institutions towards relevant research to promote the development of this field. Immunotherapy is expected to be an important part of the future treatment strategy for GBM, and it has already become a hot spot of current research and will be the key focus of future research.
Collapse
Affiliation(s)
- Keren Lv
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xue Du
- Yaan People’s Hospital, Sichuan University West China Hospital Yaan Hospital, Yaan, Sichuan, China
| | - Chunbao Chen
- Chengdu Pidu District People's Hospital, the 3rd Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yina Yu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Hendriksen JD, Locallo A, Maarup S, Debnath O, Ishaque N, Hasselbach B, Skjøth-Rasmussen J, Yde CW, Poulsen HS, Lassen U, Weischenfeldt J. Immunotherapy drives mesenchymal tumor cell state shift and TME immune response in glioblastoma patients. Neuro Oncol 2024; 26:1453-1466. [PMID: 38695342 PMCID: PMC11300009 DOI: 10.1093/neuonc/noae085] [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] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Glioblastoma is a highly aggressive type of brain tumor for which there is no curative treatment available. Immunotherapies have shown limited responses in unselected patients, and there is an urgent need to identify mechanisms of treatment resistance to design novel therapy strategies. METHODS Here we investigated the phenotypic and transcriptional dynamics at single-cell resolution during nivolumab immune checkpoint treatment of glioblastoma patients. RESULTS We present the integrative paired single-cell RNA-seq analysis of 76 tumor samples from patients in a clinical trial of the PD-1 inhibitor nivolumab and untreated patients. We identify a distinct aggressive phenotypic signature in both tumor cells and the tumor microenvironment in response to nivolumab. Moreover, nivolumab-treatment was associated with an increased transition to mesenchymal stem-like tumor cells, and an increase in TAMs and exhausted and proliferative T cells. We verify and extend our findings in large external glioblastoma dataset (n = 298), develop a latent immune signature and find 18% of primary glioblastoma samples to be latent immune, associated with mesenchymal tumor cell state and TME immune response. Finally, we show that latent immune glioblastoma patients are associated with shorter overall survival following immune checkpoint treatment (P = .0041). CONCLUSIONS We find a resistance mechanism signature in one fifth of glioblastoma patients associated with a tumor-cell transition to a more aggressive mesenchymal-like state, increase in TAMs and proliferative and exhausted T cells in response to immunotherapy. These patients may instead benefit from neuro-oncology therapies targeting mesenchymal tumor cells.
Collapse
Affiliation(s)
- Josephine D Hendriksen
- The Finsen Laboratory, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
| | - Alessio Locallo
- The Finsen Laboratory, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
| | - Simone Maarup
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
- Department of Radiation Biology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Olivia Debnath
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Digital Health Center, Berlin, Germany
| | - Naveed Ishaque
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Digital Health Center, Berlin, Germany
| | - Benedikte Hasselbach
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
- Department of Oncology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Jane Skjøth-Rasmussen
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
- Department of Neurosurgery, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Christina Westmose Yde
- Department of Genomic Medicine, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Hans S Poulsen
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
- Department of Radiation Biology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Lassen
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
- Department of Oncology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Joachim Weischenfeldt
- The Finsen Laboratory, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The DCCC Brain Tumor Center, Danish Comprehensive Cancer Center, Denmark
| |
Collapse
|
50
|
Phung LH, Nejo T, Okada H. Lessons from Post-Immunotherapy Tumor Tissues in Clinical Trials: How Can We Fuel the Tumor Microenvironment in Gliomas? Vaccines (Basel) 2024; 12:862. [PMID: 39203988 PMCID: PMC11359082 DOI: 10.3390/vaccines12080862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Despite recent advancements in cancer immunotherapy, many patients with gliomas and glioblastomas have yet to experience substantial therapeutic benefits. Modulating the tumor microenvironment (TME) of gliomas, which is typically "cold", is crucial for improving treatment outcomes. Clinical tumor specimens obtained post-immunotherapy provide invaluable insights. However, access to such post-immunotherapy samples remains limited, even in clinical trials, as tumor tissues are often collected only at tumor relapse. Recent studies of neoadjuvant immunotherapy provided important insights by incorporating surgical resections of post-treatment tumors. Moreover, pre-surgical immunotherapies are increasingly integrated into clinical trial designs to evaluate treatment efficacy. These investigations reveal critical information, particularly regarding the delivery success of therapeutic agents, the expansion and persistence of immune products, and the cellular and molecular changes induced in the TME. In this review, we assess the findings on post-treatment tumor specimens obtained from recent immunotherapy clinical trials on gliomas, highlight the importance of these samples for understanding therapeutic impacts, and discuss proactive investigation approaches for future clinical trials.
Collapse
Affiliation(s)
- Lan Hoc Phung
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA; (L.H.P.); (T.N.)
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Takahide Nejo
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA; (L.H.P.); (T.N.)
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA; (L.H.P.); (T.N.)
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| |
Collapse
|