1
|
Ma C, Wang J, Li Q, Wu Y, Yu Z, Chao Y, Liu Z, Chen G. Injectable oxidized high-amylose starch hydrogel scaffold for macrophage-mediated glioblastoma therapy. Biomaterials 2025; 318:123128. [PMID: 39884130 DOI: 10.1016/j.biomaterials.2025.123128] [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: 11/07/2024] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Glioblastoma, characterized by rapid proliferation and invasiveness, is largely resistant to current treatment modalities. A major obstacle is the blood-brain barrier (BBB), which restricts the delivery of therapeutic agents as well as the infiltration of effective immune cells into glioblastoma. In this study, we developed an injectable oxidized high-amylose starch hydrogel (OHASM) to serve as a biomaterial scaffold for the delivery of macrophages and macrophage-polarizing drugs, aiming to bypass the BBB and enhance glioblastoma treatment. The in vitro and in vivo experiments confirmed the efficacy of the hydrogel in loading and delivering macrophages and polarizing drugs against glioblastoma. Additionally, the hydrogel's interconnected porous structure was conducive to cellular growth and activity, and its slow release of therapeutics contributed to the extended survival of treated mice in a mouse GL261 glioblastoma tumor model. The immunological mechanisms underlying the therapeutic efficacy were further elucidated, revealing the potential of the hydrogel system to modulate macrophage polarization and induce apoptosis in tumor cells via the poly ADP-ribose polymerase (PARP) pathway. The study underscores the potential of the hydrogel-based macrophage delivery strategy as an effective and safe treatment for glioblastoma, offering a promising avenue for clinical management of this aggressive brain cancer.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Qiaofeng Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Yuzhe Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China.
| |
Collapse
|
2
|
Sun Y, Yan X, Wang D, Zhu J, Su H, Zhu D, Yan D, Tang BZ. Self-Assembly versus Coassembly: An Amphiphilic NIR-II Aggregation-Induced Emission Luminogen for Phototheranostics of Orthotopic Glioblastoma. J Med Chem 2025. [PMID: 40359223 DOI: 10.1021/acs.jmedchem.5c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Glioblastoma (GBM) is the most lethal form of malignant brain tumor, known for its high infiltration, aggressiveness, and poor prognosis. Second near-infrared (NIR-II, 1000-1700 nm) phototheranostic agents bring intriguing opportunities for GBM management owing to their noninvasive nature, controllability, and deeper tissue penetration. Herein, an amphiphilic NIR-II luminogen (PEG-TD) with aggregation-induced emission (AIE) characteristics, along with its hydrophobic counterpart (C6-TD), was meticulously synthesized. Specifically, PEG-TD nanoparticles (NPs), formed through straightforward self-assembly, exhibited superior stability, simplicity, robust reactive oxygen species production efficiency, and excellent photothermal conversion compared to C6-TD NPs, which were fabricated via coassembly with DSPE-mPEG, primarily attributed to the distinct molecular arrangements within the forming aggregates. The inherent advantages of PEG-TD NPs led to significant therapeutic efficacy against GL261 cells under 808 nm laser irradiation. Eventually, NIR-II fluorescence/photothermal duplex imaging-guided combined photodynamic/photothermal therapy was successfully performed in an orthotopic glioblastoma mouse model with minimal adverse effects.
Collapse
Affiliation(s)
- Yan Sun
- China Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry, Northeast Normal University, Changchun 130024, China
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xueke Yan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Zhu
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Huifang Su
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dongxia Zhu
- China Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry, Northeast Normal University, Changchun 130024, China
| | - Dingyuan Yan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ben Zhong Tang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemistry, Institute of Molecular Functional Materials, State Key Laboratory of Neuroscience, Division of Biomedical Engineering and Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, 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
|
Pu J, Yuan K, Tao J, Qin Y, Li Y, Fu J, Li Z, Zhou H, Tang Z, Li L, Gai X, Qin D. Glioblastoma multiforme: an updated overview of temozolomide resistance mechanisms and strategies to overcome resistance. Discov Oncol 2025; 16:731. [PMID: 40353925 PMCID: PMC12069213 DOI: 10.1007/s12672-025-02567-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with high lethality. The typical treatment regimen includes post-surgical radiotherapy and temozolomide (TMZ) chemotherapy, which helps extend survival. Nevertheless, TMZ resistance occurs in approximately 50% of patients. This resistance is primarily associated with the expression of O6-methylguanine-DNA methyltransferase (MGMT), which repairs O6-methylguanine lesions generated by TMZ and is thought to be the major mechanism of drug resistance. Additionally, the mismatch repair and base excision repair pathways play crucial roles in TMZ resistance. Emerging studies also point to drug transport mechanisms, glioma stem cells, and the heterogeneous tumor microenvironment as additional influences on TMZ resistance in gliomas. A better understanding of these mechanisms is vital for developing new treatments to improve TMZ effectiveness, such as DNA repair inhibitors, inhibitors of multidrug transporting proteins, TMZ analogs, and combination therapies targeting multiple pathways. This article discusses the main resistance mechanisms and potential strategies to counteract resistance in GBM patients, aiming to broaden the understanding of these mechanisms for future research and to explore the therapeutic effects of traditional Chinese medicines and their active components in overcoming TMZ resistance.
Collapse
Affiliation(s)
- Jianlin Pu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Tao
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Yongxin Li
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Jing Fu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhong Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Haimei Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhengxiu Tang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Li
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xuesong Gai
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
5
|
Tushe A, Marinelli E, Musca B, Ventura A, Zumerle S, Slukinova O, Zampardi G, Volpin F, Bonaudo C, Della Puppa A, Repellin M, Guerriero G, Lollo G, Mandruzzato S. Drug-loaded nanoparticles induce immunogenic cell death and efficiently target cells from glioblastoma patients. Nanomedicine (Lond) 2025:1-12. [PMID: 40326623 DOI: 10.1080/17435889.2025.2497747] [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: 12/03/2024] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
AIM Glioblastoma multiforme (GBM) is characterized by a highly immunosuppressive tumor microenvironment (TME), posing significant challenges for efficient therapy's outcomes. Nanomedicine combined with immunotherapy holds the potential to modulate the TME and reactivate immune responses. This study proposes a polymeric nanosystem (NPs) encapsulating diaminocyclohexane-platinum II (DACHPt), an oxaliplatin derivative, to induce immunogenic cell death (ICD) in GBM cells. MATERIALS & METHODS An ionic-gelation technique was employed to generate polymeric nanoparticles (NPs) with an approximate size of 200 nm. NPs internalization was analyzed in GBM cell lines, in vitro-derived macrophages, and in leukocytes and tumor cells from GBM patient via flow cytometry and confocal imaging. ICD was assessed by measuring two of its main markers: adenosine triphosphate (ATP) and high-mobility group box 1 (HMGB1). RESULTS NPs were efficiently incorporated by myeloid and tumor cells, but not by lymphocytes. DACHPt-loaded NPs demonstrated enhanced cytotoxicity compared to free drug, with increased ATP and HMGB1 release from GBM cells, confirming ICD induction. CONCLUSIONS Our findings suggest that DACHPt-loaded NPs represent a promising therapeutic strategy capable of targeting both tumor cells and tumor-promoting immune cells while inducing ICD.
Collapse
Affiliation(s)
- Ada Tushe
- Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
| | | | | | | | - Sara Zumerle
- Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
| | - Olga Slukinova
- Department of Surgery, Oncology and Gastroneterology, University of Padua, Padua, Italy
| | | | - Francesco Volpin
- Division of Neurosurgery, University Hospital of Padova, Padova, Italy
| | - Camilla Bonaudo
- Neurosurgery, Department of NEUROFARBA, University of Florence, University Hospital of Careggi, Florence, Italy
| | - Alessandro Della Puppa
- Neurosurgery, Department of NEUROFARBA, University of Florence, University Hospital of Careggi, Florence, Italy
| | - Mathieu Repellin
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, Lyon, France
| | - Giulia Guerriero
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, Lyon, France
| | - Giovanna Lollo
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, Lyon, France
- Institut universitaire de France (IUF), Paris, France
| | - Susanna Mandruzzato
- Veneto Institute of Oncology IOV, IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroneterology, University of Padua, Padua, Italy
| |
Collapse
|
6
|
Zhou JJ, Feng YC, Zhao ML, Guo Q, Zhao XB. Nanotechnology-driven strategies in postoperative cancer treatment: innovations in drug delivery systems. Front Pharmacol 2025; 16:1586948. [PMID: 40371327 PMCID: PMC12075547 DOI: 10.3389/fphar.2025.1586948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
Cancer remains a global health challenge, and this challenge comes with a significant burden. Current treatment modalities, such as surgery, chemotherapy, and radiotherapy, have their limitations. The emergence of nanomedicines presents a new frontier in postoperative cancer treatment, offering potential to inhibit tumor recurrence and manage postoperative complications. This review deeply explores the application and potential of nanomedicines in the treatment of cancer after surgery. In particular, it focuses on local drug delivery systems (LDDS), which consist of in situ injection, implantation, and spraying. LDDS can provide targeted drug delivery and controlled release, which enhancing therapeutic efficacy. At the same time, it minimizes damage to healthy tissues and reduces systemic side effects. The nanostructures of these systems are unique. They facilitate the sustained release of drugs, prolong the effects of treatment, and decrease the frequency of dosing. This is especially beneficial in the postoperative period. Despite their potential, nanomedicines have limitations. These include high production costs, concerns regarding long-term toxicity, and complex regulatory approval processes. This paper aims to analyze several aspects. These include the advantages of nanomedicines, their drug delivery systems, how they combine with multiple treatment methods, and the associated challenges. Future research should focus on certain issues. These issues are stability, tumor specificity, and clinical translation. By addressing these, the delivery methods can be optimized and their therapeutic efficacy enhanced. With the advancements in materials science and biomedical engineering, the future design of LDDS is set to become more intelligent and personalized. It will cater to the diverse needs of clinical treatment and offer hope for better outcomes in cancer patients after surgery.
Collapse
Affiliation(s)
- Jun-Jie Zhou
- The Stomatological Hospital, Anyang Sixth People’s Hospital, Anyang, China
| | | | | | | | | |
Collapse
|
7
|
Saha S, Zhang Y, Gibert MK, Dube C, Hanif F, Mulcahy E, Bednarek S, Marcinkiewicz P, Wang X, Kwak G, Hudson K, Sun Y, Dinda M, Saha T, Guessous F, Cruickshanks N, Colon RR, Dell'Olio LG, Anbu R, Kefas B, Kumar P, Klibanov AL, Schiff D, Suk JS, Hanes J, Mata J, Hafner M, Abounader R. Discovery and therapeutic exploitation of Master Regulatory miRNAs in Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646663. [PMID: 40236125 PMCID: PMC11996502 DOI: 10.1101/2025.04.01.646663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Glioblastoma is a fatal primary malignant brain tumor. Despite therapies involving surgical resection, chemotherapy, and radiation therapy, the average survival for glioblastoma patients remains at approximately 15 months. MicroRNAs (miRNAs) are short noncoding RNA molecules that regulate the expression of the majority of human genes. Numerous genes are concurrently deregulated in glioblastoma. Consequently, molecular monotherapies have failed to achieve improvements in clinical outcomes. Several lines of evidence suggest that simultaneous targeting of several deregulated molecules is required to achieve better therapies. However, the simultaneous targeting of several deregulated oncogenic drivers is severely limited by the fact that the drugs needed to target many deregulated molecules do not currently exist, and because combining several drugs in a clinical setting leads to an exponential increase in toxicity. We hypothesized that we can develop and use miRNA to simultaneously inhibit multiple deregulated genes for more efficacious glioblastoma therapies. The goal of this study was therefore to identify master regulatory microRNAs (miRNAs) and use them to simultaneously target multiple deregulated molecules for GBM therapy. We defined master regulatory miRNAs as those that target several deregulated genes in glioblastoma. To find master regulatory miRNAs, we first used PAR-CLIP screenings to identify all targets of all miRNAs in glioblastoma cells. We then analyzed TCGA tumor data to determine which of these targets are deregulated in human tumors. We developed and used an algorithm to rank these targets for significance in glioblastoma malignancy based on their magnitude of deregulation, frequency of deregulation, and correlation with patient survival. We then ranked the miRNAs for their capacity of targeting multiple glioblastoma-deregulated genes and therefore the potential to exhibit strong anti-tumor effects when delivered as therapy. Using this strategy, we selected two tumor suppressor master regulatory miRNAs, miR-340, miR-382 and an oncogenic master regulatory miRNA, miR-17. We validated the target genes of the miRNAs and showed that they form part of important glioblastoma regulatory pathways. We then showed that the miRNAs (miR-340 and miR-582) or the miR-17 inhibitor have strong inhibitory effects on glioblastoma cell growth, survival, invasion, stemness and in vivo tumor growth. Ultimately, we developed and successfully tested a new therapeutic approach to delivery miR-340 using MRI guided focused ultrasound and microbubbles (FUS-MB) and special brain penetrating nanoparticles (BPN). This approach resulted in a substantial reduction in tumor volume and prolongation of the survival of glioblastoma-bearing mice and can be translated into clinical trials. We therefore developed and successfully tested a novel strategy to discover and deliver miRNAs for glioblastoma and cancer therapy.
Collapse
|
8
|
Li Z, Du L, Du B, Ullah Z, Zhang Y, Tu Y, Zhou Y, Guo B. Inorganic and hybrid nanomaterials for NIR-II fluorescence imaging-guided therapy of Glioblastoma and perspectives. Theranostics 2025; 15:5616-5665. [PMID: 40365286 PMCID: PMC12068291 DOI: 10.7150/thno.112204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 05/15/2025] Open
Abstract
Glioblastoma (GBM) is the most invasive and lethal brain tumor, with limited therapeutic options due to its highly infiltrative nature, resistance to conventional therapies, and blood-brain barriers. Recent advancements in near-infrared II (NIR-II) fluorescence imaging have facilitated greater tissue penetration, improved resolution, and real-time visualization of GBM, providing a promising approach for precise diagnosis and treatment. The inorganic and hybrid NIR-II fluorescent materials have developed rapidly for NIR-II fluorescence imaging-guided diagnosis and therapy of many diseases, including GBM. Herein, we offer a timely update to explore the contribution of inorganic/hybrid NIR-II fluorescent nanomaterials, such as quantum dots, rare-earth-doped nanoparticles, carbon-based nanomaterials, and metal nanoclusters in imaging-guided treatment for GBM. These nanomaterials provide high photostability, strong fluorescence intensity, and tunable optical properties, allowing for multimodal imaging and enhanced therapeutic efficacy. Additionally, their integration with modern therapeutic strategies, such as photothermal therapy, chemodynamic therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, has shown significant potential in overcoming the limitations of traditional treatments. Looking forward, future advancements including safe body clearance, long-term biocompatibility, efficient BBB penetration, and extended emission wavelengths beyond 1500 nm could enhance the theranostic outcomes. The integration of dual imaging with immunotherapy and AI-driven strategies will further enhance precision and accelerate the clinical translation of smart theranostic platforms for GBM treatment.
Collapse
Affiliation(s)
- Zhigang Li
- Department of Medical Imaging, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen 518110, China
| | - Lixin Du
- Department of Medical Imaging, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen 518110, China
| | - Binghua Du
- Department of Medical Imaging, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen 518110, China
| | - Zia Ullah
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yinghe Zhang
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yanyang Tu
- Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou City, Guangdong Province, China
| | - Ying Zhou
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
9
|
Zhao B, Wu J, Zhang T, Han M, Zhang C, Rong X, Zhang R, Chen X, Peng F, Jin J, Liu S, Dong X, Zhao S. A spatial transcriptomics study of MES-like and mono/macro cells in gliomas. Sci Rep 2025; 15:12730. [PMID: 40222970 PMCID: PMC11994772 DOI: 10.1038/s41598-025-95277-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Gliomas, including both glioblastoma multiforme (GBM) and lower-grade glioma (LGG), present a substantial challenge in neuro-oncology because of genetic heterogeneity and unsatisfactory prognosis. This study aimed to conduct a comprehensive multi-omics analysis of gliomas using various bioinformatics approaches to identify potential therapeutic targets and prognostic markers. A comprehensive analysis was conducted on 1327 sequencing data samples alongside their relevant clinical information sourced from The Cancer Genome Atlas (TCGA) pertaining to glioblastoma (GBM), low-grade glioma (LGG), the Chinese Glioma Genome Atlas (CCGA) and University of California Santa Cruz Xena (UCSC Xena) datasets. These tools were employed for gene expression profiling, survival analysis, and cell communication mapping. Spatial transcriptomics revealed the localization of mesenchymal (MES)-like malignant tumors, and drug sensitivity analysis was performed to evaluate responses to quinpirole and meropenem. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) framework was utilized to gauge the responsiveness to immunotherapy. The MES-like malignant and monocyte/macrophage (mono/macro) cell subsets showed high hallmark scores, playing key roles in the tumor microenvironment. MES-like malignant marker gene scores correlated with overall survival across datasets, whereas mono/macro marker gene scores were significant in the TCGA-LGG and CCGA datasets. Key interactions between these cell types were found, especially with CD14-ITGB2, LGALS1-CD69, and APOE-TREM2. The mono/macro cell subset demonstrated better immune therapy responsiveness, as indicated by lower TIDE scores. Spatial transcriptomics revealed that MES-like malignant tumors are predominantly localized in four distinct regions, with the marker genes CHI3L1 and ADM confirming these locations. Drug sensitivity analysis revealed differential responses of the MES-like malignant cell subset to quinpirole and meropenem. Our results offer fresh perspectives on the differential roles of MES-like malignant and monocyte/macrophage cell subsets in tumor progression and immune modulation, providing novel insights into glioma biology.
Collapse
Affiliation(s)
- Boyan Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Jianing Wu
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Tiehui Zhang
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong, China
| | - Mingyang Han
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Cheng Zhang
- University of Toronto Scarborough 1265 Military Trail, Scarborough, ON, M1C 1A4, Canada
| | - Xuan Rong
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, 150001, Heilongjiang, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Jin
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Shiya Liu
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Xingli Dong
- Central Laboratory, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
| | - Shiguang Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China.
- Department of Neurosurgery, Shenzhen University General Hospital, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
10
|
Wang F, Zhou H, Tian Y, Wang X, Huang Y, Tu Y, Li L, Zhen H. ELK4 induced upregulation of HOMER3 promotes the proliferation and metastasis in glioma via Wnt/β-catenin/EMT signaling pathway. Biol Direct 2025; 20:48. [PMID: 40205485 PMCID: PMC11980352 DOI: 10.1186/s13062-025-00643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Glioma is an aggressive brain tumor characterized by its high invasiveness, which complicates prognosis and contributes to patient resistance against various treatment options. The HOMER family, consisting of HOMER1, HOMER2, and HOMER3, has been implicated in various cancers, yet their specific roles in glioma remain inadequately understood. This study conducted a comprehensive pan-cancer analysis to evaluate the expression profiles of HOMER family members across different tumor types, utilizing data from public databases such as TCGA and GTEx. Our findings indicate significant dysregulation of HOMER1, HOMER2, and HOMER3 in multiple cancers, with HOMER3 emerging as a potential prognostic biomarker, particularly for lower-grade glioma. Elevated expression levels of HOMER3 were associated with shorter overall survival and disease-specific survival in LGG patients, supported by Cox regression analysis that confirmed HOMER3 as an independent prognostic factor. Furthermore, HOMER3 expression correlated positively with advanced clinical stages and key tumor markers. To elucidate the mechanisms behind HOMER3 dysregulation, we identified ELK4 as a transcription factor that binds to the HOMER3 promoter, promoting its expression in glioma cells. Functional assays demonstrated that silencing HOMER3 significantly reduced glioma cell proliferation and metastatic potential in vitro and in vivo, highlighting its oncogenic role. Additionally, HOMER3 was found to influence the Wnt/β-catenin/EMT signaling pathway, with knockdown resulting in altered expression of critical EMT markers. Collectively, our results indicated that HOMER3 plays a crucial role in glioma progression and metastasis, underscoring its potential as a therapeutic target and prognostic biomarker in glioma management.
Collapse
Affiliation(s)
- Furong Wang
- Department of Pathology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
- Department of Pathology, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Yu Tian
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Xiaoling Wang
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Youcai Huang
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Yanyang Tu
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China.
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China.
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China.
| | - Liwen Li
- Department of Bioscience, College of Life Sciences, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
11
|
Piyadasa H, Oberlton B, Ribi M, Ranek JS, Averbukh I, Leow K, Amouzgar M, Liu CC, Greenwald NF, McCaffrey EF, Kumar R, Ferrian S, Tsai AG, Filiz F, Fullaway CC, Bosse M, Varra SR, Kong A, Sowers C, Gephart MH, Nuñez-Perez P, Yang E, Travers M, Schachter MJ, Liang S, Santi MR, Bucktrout S, Gherardini PF, Connolly J, Cole K, Barish ME, Brown CE, Oldridge DA, Drake RR, Phillips JJ, Okada H, Prins R, Bendall SC, Angelo M. Multi-omic landscape of human gliomas from diagnosis to treatment and recurrence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642624. [PMID: 40161803 PMCID: PMC11952471 DOI: 10.1101/2025.03.12.642624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Gliomas are among the most lethal cancers, with limited treatment options. To uncover hallmarks of therapeutic escape and tumor microenvironment (TME) evolution, we applied spatial proteomics, transcriptomics, and glycomics to 670 lesions from 310 adult and pediatric patients. Single-cell analysis shows high B7H3+ tumor cell prevalence in glioblastoma (GBM) and pleomorphic xanthoastrocytoma (PXA), while most gliomas, including pediatric cases, express targetable tumor antigens in less than 50% of tumor cells, potentially explaining trial failures. Longitudinal samples of isocitrate dehydrogenase (IDH)-mutant gliomas reveal recurrence driven by tumor-immune spatial reorganization, shifting from T-cell and vasculature-associated myeloid cell-enriched niches to microglia and CD206+ macrophage-dominated tumors. Multi-omic integration identified N-glycosylation as the best classifier of grade, while the immune transcriptome best predicted GBM survival. Provided as a community resource, this study opens new avenues for glioma targeting, classification, outcome prediction, and a baseline of TME composition across all stages.
Collapse
Affiliation(s)
- Hadeesha Piyadasa
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Oberlton
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Mikaela Ribi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Jolene S. Ranek
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Inna Averbukh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ke Leow
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Meelad Amouzgar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Candace C. Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah F. Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin F. McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rashmi Kumar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Selena Ferrian
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Albert G. Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ferda Filiz
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Alex Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Cameron Sowers
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Pablo Nuñez-Perez
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - EnJun Yang
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Mike Travers
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Samantha Liang
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Maria R. Santi
- Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | | | - Pier Federico Gherardini
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - John Connolly
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Kristina Cole
- Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Michael E. Barish
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Christine E. Brown
- Departments of Hematology & Hematopoietic Cell Transplantation and Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Derek A. Oldridge
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, Children’s Hospital of Philadelphia, PA, USA
| | - Richard R. Drake
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Joanna J. Phillips
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Hideho Okada
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Robert Prins
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Neurosurgery, UCLA, Los Angeles, CA, USA
| | - Sean C. Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
12
|
Wang L, Xu P, Li X, Zhang Q. Comprehensive bioinformatics analysis identified HMGB3 as a promising immunotherapy target for glioblastoma multiforme. Discov Oncol 2025; 16:478. [PMID: 40192954 PMCID: PMC11977083 DOI: 10.1007/s12672-025-02235-6] [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: 12/16/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM) presents significant therapeutic challenges due to its heterogeneous tumorigenicity, drug resistance, and immunosuppression. Although several molecular markers have been developed, there still lack of sensitive molecular for accurately detection. Studying the mechanisms underlying the development of GBM and finding relevant prognostic biomarkers remains crucial. METHODS Single-cell RNA sequencing, bulk RNA-seq, and cancer immune cycle activities of GBM were used to assess the expression of different molecular related to GBM. Bioinformatics analyses were carried to evaluate the functional of the high mobility group protein B3 (HMGB3) in GBM. RESULTS HMGB3 was highly expressed in GBM tissues and influenced the interpatient and intratumoral transcriptomic heterogeneity as well as immunosuppression in GBM. HMGB3 also contributes to a no inflamed tumor microenvironment (TME) and has an inhibitory effect on tumor-associated immune cell infiltration. Besides, HMGB3 participated GBM chemotherapeutic sensitivity and negative correlation with 140 medicines. CONCLUSION HMGB3 as a heterogeneous and immunosuppressive molecule in the GBM TME, making it a potential target for precision therapy for GBM.
Collapse
Affiliation(s)
- Libin Wang
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
- Medical Research Center, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Peizhi Xu
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
- Department of Neurosurgery, The 6th Affiliated Hospital of Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Xinglong Li
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
- Medical Research Center, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| | - Qinghua Zhang
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| |
Collapse
|
13
|
Liu T, Sun T, Chen X, Wu J, Sun X, Liu X, Yan H, Fu Q, Fan Z, Wang X, Cheng P, Cheng W, Wu A. Targeting ARPC1B Overcomes Immune Checkpoint Inhibitor Resistance in Glioblastoma by Reversing Protumorigenic Macrophage Polarization. Cancer Res 2025; 85:1236-1252. [PMID: 39841088 DOI: 10.1158/0008-5472.can-24-2286] [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: 07/05/2024] [Revised: 11/07/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
Immunotherapy has elicited significant improvements in outcomes for patients with several tumor types. However, the immunosuppressive microenvironment in glioblastoma (GBM) restricts the therapeutic efficacy of immune checkpoint blockade (ICB). In this study, we investigated the components of the immune microenvironment that contribute to ICB failure in GBM to elucidate the underlying causes of immunotherapeutic resistance. Macrophages were identified as a main contributor to ICB resistance. Expression of actin-related protein 2/3 complex subunit 1B (ARPC1B), a regulatory subunit of the Arp2/3 complex, was elevated in GBM and correlated with macrophage enrichment and prognosis. ARPC1B in tumor cells increased STAT1 expression and subsequent IL10 production, which induced a protumorigenic macrophage state. Mechanistically, ARPC1B inhibited the ubiquitination and degradation of STAT1 by preventing the E3 ubiquitin ligase NEDD4L from binding to STAT1 and by supporting the interaction between STAT1 and the deubiquitinase USP7. Inhibiting ARPC1B reshaped the immunosuppressive microenvironment and increased the efficacy of ICB in GBM models. This study highlights the important role of ARPC1B in macrophage-mediated immunosuppression and proposes a combination treatment regimen for GBM immunotherapy. Significance: ARPC1B induces macrophage-mediated immunosuppression by activating a STAT1/IL10 axis and can be targeted to improve the efficacy of immune checkpoint blockade in glioblastoma.
Collapse
Affiliation(s)
- Tianqi Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Sun
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianqi Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoqian Sun
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xing Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Haixu Yan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiang Fu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zirong Fan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiangyu Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
He H, Yan M, Ye K, Shi R, Tong L, Zhang S, Zhu Y, Zhan R. Predicting Prognosis and Immunotherapy Response in Glioblastoma (GBM) With a 5-Gene CAF-Risk Signature. Cancer Rep (Hoboken) 2025; 8:e70158. [PMID: 40226936 PMCID: PMC11995297 DOI: 10.1002/cnr2.70158] [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: 08/09/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAF) represent significant constituents within the extracellular matrix (ECM) across a range of cancers. Nevertheless, there exists a scarcity of direct proof concerning the function of CAF in glioblastoma (GBM). AIMS This study endeavors to probe the participation of CAF in GBM by developing and validating a CAF-risk signature and exploring its correlation with immune infiltration and immunotherapy responsiveness. METHODS AND RESULTS To fulfill these objectives, mRNA expression profiles of GBM samples and their corresponding clinical data were retrieved from two databases. First, stromal CAF-associated genes were identified by weighted gene co-expression network analysis (WGCNA). This method constructs co-expression networks and pinpoints gene modules with similar expression patterns to detect relevant genes. Subsequently, a CAF-risk signature was established via univariate and LASSO Cox regression analyses. Thereafter, gene set enrichment analysis (GSEA) and single-sample gene set enrichment analysis (ssGSEA) were carried out to investigate the underlying molecular mechanisms. The immune status was evaluated with several R packages, and the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm was utilized to assess the response to immunotherapy. Validation was performed using single-cell RNA sequencing (scRNA) datasets, the Cancer Cell Line Encyclopedia (CCLE), and the Human Protein Atlas (HPA). Eventually, a 5-gene (ITGA5, MMP14, FN1, COL5A1, and COL6A1) prognostic CAF model was constructed. Notably, immune infiltration analysis demonstrated a significant correlation between Treg, Macrophage, and CAF risk scores. Moreover, TIDE analysis suggested a decreased responsiveness to immunotherapy in high CAF-risk patients. In addition, GSEA showed significant enrichment of the transforming growth factor alpha (TGF-α), inflammatory response, and epithelial-mesenchymal transition (EMT) pathways in this subgroup. Finally, the validation through scRNA, CCLE, and HPA datasets confirmed these findings. CONCLUSION The 5-gene CAF-risk signature exhibited accurate prognostic predictions and efficiently evaluated clinical immunotherapy responses among GBM patients. These results offer robust evidence regarding the implication of CAF in GBM and underscore the potential clinical value of personalized anti-CAF therapies in conjunction with immunotherapy.
Collapse
Affiliation(s)
- Haifeng He
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Min Yan
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Ke Ye
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Rui Shi
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Luqing Tong
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Shengxiang Zhang
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yu Zhu
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Renya Zhan
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
15
|
Lee J, Kang Y, Lee H, Saravanakumar G, Park SA, Ahn S, Kim WJ. Amplifying glioblastoma immunotherapy: T cell shielding through Nitric oxide/reactive oxygen species scavenging nanoparticles Potentiates anti-PD-1. Biomaterials 2025; 315:122904. [PMID: 39490061 DOI: 10.1016/j.biomaterials.2024.122904] [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: 09/02/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
Despite the success of immune checkpoint blockade (ICB) therapy in various cancers, its efficacy faces challenges in glioblastoma (GBM) due to the immunosuppressive cold-tumor microenvironment. The scarcity of tumor-infiltrating T cells and the suppression of T cell activity significantly limit therapeutic outcomes in GBM. Nitric oxide (NO) and reactive oxygen species (ROS) from tumor-associated myeloid cells (TAMCs) are key contributors to T cell suppression, reducing ICB therapy effectiveness. In this study, we developed NO-ROS scavenging micelles that effectively scavenge both NO and ROS, protecting T cells from their exhausting effects. This leads to a significant increase in T cell infiltration and activation. Moreover, when combined with αPD-1, the survival rate increases to 40 % up to 120 days, enhancing therapeutic efficacy compared to αPD-1 alone. This approach not only protects T cells from the inhibitory effects of NO and ROS but also has the potential to reshape the tumor microenvironment, overcoming T cell suppression in cold tumors.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Yeoul Kang
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hyori Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | | | - Soon A Park
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea; OmniaMed Co., Ltd, Pohang, 37666, Republic of Korea.
| |
Collapse
|
16
|
Mao M, Lei Y, Ma X, Xie HY. Challenges and Emerging Strategies of Immunotherapy for Glioblastoma. Chembiochem 2025; 26:e202400848. [PMID: 39945240 DOI: 10.1002/cbic.202400848] [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: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
Glioblastoma (GBM) is recognized as the most lethal primary malignant tumor of the central nervous system. Although traditional treatments can somewhat prolong patient survival, the overall prognosis remains grim. Immunotherapy has become an effective method for GBM treatment. Oncolytic virus, checkpoint inhibitors, CAR T cells and tumor vaccines have all been applied in this field. Moreover, the combining of immunotherapy with traditional radiotherapy, chemotherapy, or gene therapy can further improve the treatment outcome. This review systematically summarizes the features of GBM, the recent progress of immunotherapy in overcoming GBM.
Collapse
Affiliation(s)
- Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yao Lei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Hai-Yan Xie
- Chemical Biology Center, Peking University, Beijing, 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
17
|
Vollmuth P, Karschnia P, Sahm F, Park YW, Ahn SS, Jain R. A Radiologist's Guide to IDH-Wildtype Glioblastoma for Efficient Communication With Clinicians: Part II-Essential Information on Post-Treatment Imaging. Korean J Radiol 2025; 26:368-389. [PMID: 40015559 PMCID: PMC11955384 DOI: 10.3348/kjr.2024.0983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/30/2024] [Indexed: 03/01/2025] Open
Abstract
Owing to recent advancements in various postoperative treatment modalities, such as radiation, chemotherapy, antiangiogenic treatment, and immunotherapy, the radiological and clinical assessment of patients with isocitrate dehydrogenase-wildtype glioblastoma using post-treatment imaging has become increasingly challenging. This review highlights the challenges in differentiating treatment-related changes such as pseudoprogression, radiation necrosis, and pseudoresponse from true tumor progression and aims to serve as a guideline for efficient communication with clinicians for optimal management of patients with post-treatment imaging.
Collapse
Affiliation(s)
- Philipp Vollmuth
- Division for Computational Radiology & Clinical AI (CCIBonn.ai), Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany
- Medical Faculty Bonn, University of Bonn, Bonn, Germany
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
- Department of Neurosurgery, Friedrich-Alexander-University University, Erlangen-Nuremberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Rajan Jain
- Department of Radiology, New York University Grossman School of Medicine, New York, USA
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, USA
| |
Collapse
|
18
|
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
|
19
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
20
|
Rivera D, Bouras A, Mattioli M, Anastasiadou M, Pacentra AC, Pelcher O, Koziel C, Schupper AJ, Chanenchuk T, Carlton H, Ivkov R, Hadjipanayis CG. Magnetic hyperthermia therapy enhances the chemoradiosensitivity of glioblastoma. Sci Rep 2025; 15:10532. [PMID: 40148452 PMCID: PMC11950323 DOI: 10.1038/s41598-025-95544-3] [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: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer and is resistant to standard-of-care chemoradiation therapy (CRT). Magnetic hyperthermia therapy (MHT) exposes magnetic iron oxide nanoparticles (MIONPs) to an alternating magnetic field (AMF) to generate local hyperthermia. This study evaluated MHT-mediated enhancement of CRT in preclinical GBM models. Cell viability and apoptosis were assessed in GBM cell lines after water bath heating with radiation and/or temozolomide. Heating efficiency of MIONPs after intracranial delivery was measured in healthy mice. MHT with CRT was performed in syngeneic and patient-derived xenograft (PDX) GBM tumors. Tissue sections were analyzed for γ-H2AX, HSP90, CD4 + T cells, and microglial cells. Tumor burden and survival were assessed. Hyperthermia with radiation and temozolomide significantly reduced cell viability and increased apoptosis. Hyperthermia predominantly exhibited additive to synergistic interactions with both treatment modalities and reduced doses needed for tumor cell growth inhibition. In vivo, MHT with CRT decreased tumor burden and increased survival in PDX and syngeneic models. Immunohistochemistry showed increased γ-H2AX, HSP90, microglial activation, and CD4 + T cells after MHT in combination with CRT. Overall, adjuvant hyperthermia enhances CRT efficacy in GBM cells, with MHT improving survival outcomes in rodents. Sufficient intracranial heating and MIONP retention for repeated treatments was achieved, supporting further clinical translation.
Collapse
Affiliation(s)
- Daniel Rivera
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Milena Mattioli
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Maria Anastasiadou
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Chiara Pacentra
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Olivia Pelcher
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Corrine Koziel
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Alexander J Schupper
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Tori Chanenchuk
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Hayden Carlton
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Constantinos G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA.
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
21
|
Wang Z, Li P, Zeng X, Guo J, Zhang C, Fan Z, Wang Z, Zhu P, Chen Z. CAR-T therapy dilemma and innovative design strategies for next generation. Cell Death Dis 2025; 16:211. [PMID: 40148310 PMCID: PMC11950394 DOI: 10.1038/s41419-025-07454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has shown remarkable curative effects on hematological tumors, driving the exponential growth in CAR-T-related research. Although CD19-targeting CAR-T-cell therapy has displayed remarkable promise in clinical trials, many obstacles are arising that limit its therapeutic efficacy in tumor immunotherapy. The "dilemma" of CAR-T cell-based tumor therapy includes lethal cytotoxicity, restricted trafficking, limited tumor infiltration, an immunosuppressive microenvironment, immune resistance and limited potency. The solution to CAR-T-cell therapy's dilemma requires interdisciplinary strategies, including synthetic biology-based ON/OFF switch, bioinstructive scaffolds, nanomaterials, oncolytic viruses, CRISPR screening, intestinal microbiota and its metabolites. In this review, we will introduce and summarize these interdisciplinary-based innovative technologies for the next generation CAR-T-cell design and delivery to overcome the key barriers of current CAR-T cells.
Collapse
Affiliation(s)
- Zhiwei Wang
- The First Affiliated Hospital of Henan University, 475004, Kaifeng, China
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China
| | - Peixian Li
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China
| | - Xiaoyu Zeng
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China
| | - Jing Guo
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China
| | - Cheng Zhang
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China
| | - Zusen Fan
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Zhiwei Wang
- The First Affiliated Hospital of Henan University, 475004, Kaifeng, China.
| | - Pingping Zhu
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China
| | - Zhenzhen Chen
- School of Life Sciences, Zhengzhou University, 100 Kexue Road, Zhengzhou, 450001, China.
| |
Collapse
|
22
|
Tang J, Karbhari N, Campian JL. Therapeutic Targets in Glioblastoma: Molecular Pathways, Emerging Strategies, and Future Directions. Cells 2025; 14:494. [PMID: 40214448 PMCID: PMC11988183 DOI: 10.3390/cells14070494] [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: 02/14/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by rapid growth, invasive infiltration into surrounding brain tissue, and resistance to conventional therapies. Despite advancements in surgery, radiotherapy, and chemotherapy, median survival remains approximately 15 months, underscoring the urgent need for innovative treatments. Key considerations informing treatment development include oncogenic genetic and epigenetic alterations that may dually serve as therapeutic targets and facilitate treatment resistance. Various immunotherapeutic strategies have been explored and continue to be refined for their anti-tumor potential. Technical aspects of drug delivery and blood-brain barrier (BBB) penetration have been addressed through novel vehicles and techniques including the incorporation of nanotechnology. Molecular profiling has emerged as an important tool to individualize treatment where applicable, and to identify patient populations with the most drug sensitivity. The goal of this review is to describe the spectrum of potential GBM therapeutic targets, and to provide an overview of key trial outcomes. Altogether, the progress of clinical and preclinical work must be critically evaluated in order to develop therapies for GBM with the strongest therapeutic efficacy.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Nishika Karbhari
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| |
Collapse
|
23
|
Wen J, Wu D, Le Y, Yin Z, Chen M, Shen Y, Wu X, Liu K, Luo K, Shu Z, Shu Q, Ouyang D. Engineered nanovesicles targeting SERPINE1 overcome temozolomide resistance in glioblastoma. Cell Signal 2025; 132:111763. [PMID: 40139622 DOI: 10.1016/j.cellsig.2025.111763] [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: 12/06/2024] [Revised: 01/15/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor with limited treatment options due to its resistance to temozolomide (TMZ). This study explores a novel therapeutic approach using engineered cell membrane nanovesicles loaded with SERPINE1 inhibitors to combat TMZ resistance. High-throughput sequencing identified pivotal genes associated with resistance, while the nanovesicles demonstrated excellent stability and the ability to cross the blood-brain barrier. Functional assays revealed significant suppression of GBM cell viability, migration, and invasion, accompanied by reduced expression of SERPINE1 and VEGF, suggesting inhibition of angiogenesis and tumor progression. These findings highlight the potential of SERPINE1-targeted nanovesicles as an innovative and effective strategy for overcoming TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jianping Wen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China.
| | - Dongxu Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yi Le
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zonghua Yin
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Minglong Chen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yulong Shen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Xia Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kebo Liu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kun Luo
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zhicheng Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Qingxia Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Dongsheng Ouyang
- The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
24
|
Takahashi M, Mukhamejanova D, Jasewicz H, Acharya N, Moon JJ, Hara T. Opportunities to Modulate Tumor Ecosystem Toward Successful Glioblastoma Immunotherapy. Cancer Sci 2025. [PMID: 40123277 DOI: 10.1111/cas.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Over the past decade, the failure of multiple clinical trials has confirmed the need for a systematic and comprehensive understanding of glioblastoma (GBM). Current immunotherapies aiming to harness the immune system to achieve anti-tumor effects remain largely ineffective, highlighting the complexities of the GBM microenvironment. However, our recent understanding of immune niches within the central nervous system provides both opportunities and challenges in translating these insights into successful immunotherapy implementation. We discuss these strategies, including targeting multiple antigens within the heterogeneous GBM microenvironment, identifying new druggable targets to abrogate immunosuppression, and understanding niche-specific immune cell functionality to modulate tumor-immune-stroma interactions.
Collapse
Affiliation(s)
- Mariko Takahashi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Darina Mukhamejanova
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurosurgery, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biology, Nazarbayev University, Astana, Kazakhstan
| | - Himani Jasewicz
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Nandini Acharya
- Pelotonia Institute for Immuno-Oncology, OSUCCC-James, The Ohio State University, Columbus, Ohio, USA
- Department of Neurology, the Neuroscience Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Toshiro Hara
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurosurgery, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
25
|
Meehan B, Adnani L, Zhu X, Tawil N, Garnier D, Nakano I, Huang S, Rak J. Curative timed NK cell-based immunochemotherapy aborts brain tumour recurrence driven by mesenchymal glioma stem cells. Acta Neuropathol Commun 2025; 13:64. [PMID: 40119461 PMCID: PMC11927124 DOI: 10.1186/s40478-025-01984-3] [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: 08/24/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
High grade gliomas (HGG) are incurable brain cancers, where inevitable disease recurrence is driven by tumour-initiating glioma stem cells (GSCs). GSCs survive and expand in the brain after surgery, radiation and temozolomide (TMZ) chemotherapy, amidst weak immune and natural killer (NK) cell surveillance. The present study was designed to understand how to enhance the contribution of innate immunity to post TMZ disease control. Strikingly, molecular subtypes of HGG impacted the repertoire of NK cell sensitivity markers across human HGG transcriptomes, and in a panel of GSCs with either proneural (PN-GSC) or mesenchymal (MES-GSC) phenotypes. Indeed, only MES-GSCs (but not PN-GSCs) were enriched for NK cell ligands and sensitive to NK-mediated cytotoxicity in vitro. While NK cells alone had no effect on HGG progression in vivo, the post-chemotherapy (TMZ) recurrence of MES-GSC-driven xenografts was aborted by timed intracranial injection of live or irradiated NK (NK92MI) cells, resulting in long term survival of animals. This curative effect declined when NK cell administration was delayed relative to TMZ exposure pointing to limits of the immune control over resurging residual tumour stem cell populations that survived chemotherapy. Overall, these results suggest that chemotherapy-dependent tumour depopulation may create a unique window of opportunity for NK-mediated intervention with curative effects restricted to a subset of HGGs driven by mesenchymal brain tumour initiating cells.
Collapse
Affiliation(s)
- Brian Meehan
- Research Institute of the McGill University Health Centre, 1001 Decarie Boul, Montreal, QC, H4A 3J1, Canada
| | - Lata Adnani
- Research Institute of the McGill University Health Centre, 1001 Decarie Boul, Montreal, QC, H4A 3J1, Canada
| | - Xianbing Zhu
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Nadim Tawil
- Research Institute of the McGill University Health Centre, 1001 Decarie Boul, Montreal, QC, H4A 3J1, Canada
| | | | - Ichiro Nakano
- Department of Neurosurgery, Hokuto Social Medical Corporation, Hokuto Hospital, Kisen-7-5 Inadacho, Obihiro, 080-0833, Hokkaido, Japan
| | - Sidong Huang
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Janusz Rak
- Research Institute of the McGill University Health Centre, 1001 Decarie Boul, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
26
|
He J, Li H, Zhang B, Liang G, Zhang L, Zhao W, Zhao W, Zhang YJ, Wang ZX, Li JF. Convolutional neural network-assisted Raman spectroscopy for high-precision diagnosis of glioblastoma. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125615. [PMID: 39721487 DOI: 10.1016/j.saa.2024.125615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Glioblastoma multiforme (GBM) is the most lethal intracranial tumor with a median survival of approximately 15 months. Due to its highly invasive properties, it is particularly difficult to accurately identify the tumor margins intraoperatively. The current gold standard for diagnosing GBM during surgery is pathology, but it is time-consuming. Under these circumstances, we developed a method combining Raman spectroscopy (RS) with convolutional neural networks (CNN) to distinguish GBM. Analysis of the spectra of normal brain samples (478 spectra) and GBM samples (462 spectra) from 29 in situ intracranial tumor-bearing mice showed that this method identified GBM tissue with 96.8 % accuracy. Subsequently, spectral analysis of 23 normal human brain tissues (223 spectra) versus 21 tissues from patients with pathologically diagnosed GBM (267 spectra) revealed that the accuracy of this method was 93.9 %. Most importantly, for the difference peaks in the spectra of GBM and normal brain tissue, the common difference peaks in the mouse and human spectra were at 750 cm-1, 1440 cm-1, and 1586 cm-1, which emphasized the differences in cytochrome C and lipids between GBM samples and normal brain samples in both mice and human. The preliminary results showed that CNN-assisted RS is simple to operate and can rapidly and accurately identify whether it is GBM tissue or normal brain tissue.
Collapse
Affiliation(s)
- Jiawei He
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Hongmei Li
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Bingchang Zhang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Gehao Liang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Liang Zhang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Wentao Zhao
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Wenpeng Zhao
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China
| | - Yue-Jiao Zhang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China.
| | - Zhan-Xiang Wang
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China.
| | - Jian-Feng Li
- Department of Neurosurgery and Department of Neuroscience, The First Affiliated Hospital of Xiamen University, School of Medicine, College of Chemistry and Chemical Engineering, College of Energy, Institute of Artificial Intelligence, State Key Laboratory for Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361102, China; Scientific Research Foundation of State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen 361005, China.
| |
Collapse
|
27
|
Ali H, Zhou N, Chen L, van Hijfte L, Karri V, Zhou Y, Habashy K, Arrieta VA, Kim KS, Duffy J, Yeeravalli R, Tiek DM, Song X, Mishra S, Lee-Chang C, Ahmed AU, Heiland DH, Sonabend AM, Dmello C. Targeting CHEK2-YBX1&YBX3 regulatory hub to potentiate immune checkpoint blockade response in gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642289. [PMID: 40161682 PMCID: PMC11952400 DOI: 10.1101/2025.03.09.642289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Although GBM's immunosuppressive environment is well known, the tumor's resistance to CD8+ T cell killing is not fully understood. Our previous study identified Checkpoint Kinase 2 (Chek2) as the key driver of CD8+ T cell resistance in mouse glioma through an in vivo CRISPR screen and demonstrated that Chk2 inhibition, combined with PD-1/PD-L1 blockade, significantly enhanced CD8+ T cell-mediated tumor killing and improved survival in preclinical model. Here, we aimed to elucidate the immunosuppressive function of Chek2. Immunoprecipitation (IP) followed by mass spectrometry (MS) and phosphoproteomics identified an association between Chek2 with the DNA/RNA-binding proteins YBX1 and YBX3 that are implicated in transcriptional repression of pro-inflammatory genes. Single-gene knock-out and overexpression studies of CHEK2, YBX1, and YBX3 in multiple glioma cell lines revealed that these proteins positively regulate each other's expression. RNA sequencing coupled with chromatin immunoprecipitation-sequencing (ChIP-seq) analysis demonstrated common inflammatory genes repressed by CHK2-YBX1&YBX3 hub. Targeting one of the hub proteins, YBX1, with the YBX1 inhibitor SU056 led to degradation of CHK2-YBX1&YBX3 hub. Targeting of this hub by SU056 led to enhanced antigen presentation and antigen specific CD8+ T cell proliferation. Further, combination of SU056 with ICB significantly improved survival in multiple glioma models. Collectively, these findings reveal an immunosuppressive mechanism mediated by the CHK2-YBX1&YBX3 hub proteins. Therefore, CHK2-YBX1&YBX3 hub targeting in combination with immune checkpoint blockade therapies in gliomas is warranted.
Collapse
|
28
|
Rocha JR, Krause RF, Ribeiro CE, Oliveira NDA, Ribeiro de Sousa L, Leandro Santos J, Castro SDM, Valadares MC, Cunha Xavier Pinto M, Pavam MV, Lima EM, Antônio Mendanha S, Bakuzis AF. Near Infrared Biomimetic Hybrid Magnetic Nanocarrier for MRI-Guided Thermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13094-13110. [PMID: 38973727 PMCID: PMC11891835 DOI: 10.1021/acsami.4c03434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/09/2024]
Abstract
Cell-membrane hybrid nanoparticles (NPs) are designed to improve drug delivery, thermal therapy, and immunotherapy for several diseases. Here, we report the development of distinct biomimetic magnetic nanocarriers containing magnetic nanoparticles encapsulated in vesicles and IR780 near-infrared dyes incorporated in the membranes. Distinct cell membranes are investigated, red blood cell (RBC), melanoma (B16F10), and glioblastoma (GL261). Hybrid nanocarriers containing synthetic lipids and a cell membrane are designed. The biomedical applications of several systems are compared. The inorganic nanoparticle consisted of Mn-ferrite nanoparticles with a core diameter of 15 ± 4 nm. TEM images show many multicore nanostructures (∼40 nm), which correlate with the hydrodynamic size. Ultrahigh transverse relaxivity values are reported for the magnetic NPs, 746 mM-1s-1, decreasing respectively to 445 mM-1s-1 and 278 mM-1s-1 for the B16F10 and GL261 hybrid vesicles. The ratio of relaxivities r2/r1 decreased with the higher encapsulation of NPs and increased for the biomimetic liposomes. Therapeutic temperatures are achieved by both, magnetic nanoparticle hyperthermia and photothermal therapy. Photothermal conversion efficiency ∼25-30% are reported. Cell culture revealed lower wrapping times for the biomimetic vesicles. In vivo experiments with distinct routes of nanoparticle administration were investigated. Intratumoral injection proved the nanoparticle-mediated PTT efficiency. MRI and near-infrared images showed that the nanoparticles accumulate in the tumor after intravenous or intraperitoneal administration. Both routes benefit from MRI-guided PTT and demonstrate the multimodal theranostic applications for cancer therapy.
Collapse
Affiliation(s)
| | - Rafael Freire Krause
- Institute
of Physics, Federal University of Goiás, Goianiâ, Goiás 74690-900, Brazil
| | | | | | | | | | | | - Marize Campos Valadares
- ToxIn
− Laboratory of Education and Research in In Vitro Toxicology, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
| | - Mauro Cunha Xavier Pinto
- Department
of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, Goianiâ, Goiás 74690-900, Brazil
| | - Marcilia Viana Pavam
- FarmaTec
− Laboratory of Pharmaceutical Technology, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
- CNanoMed
− Nanomedicine Integrated Research Center, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
| | - Eliana Martins Lima
- FarmaTec
− Laboratory of Pharmaceutical Technology, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
- CNanoMed
− Nanomedicine Integrated Research Center, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
| | - Sebastião Antônio Mendanha
- Institute
of Physics, Federal University of Goiás, Goianiâ, Goiás 74690-900, Brazil
- FarmaTec
− Laboratory of Pharmaceutical Technology, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
- CNanoMed
− Nanomedicine Integrated Research Center, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
| | - Andris Figueiroa Bakuzis
- Institute
of Physics, Federal University of Goiás, Goianiâ, Goiás 74690-900, Brazil
- CNanoMed
− Nanomedicine Integrated Research Center, Federal University of Goiás, Goianiâ, Goiás 74690-631, Brazil
| |
Collapse
|
29
|
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
|
30
|
Jin S, Li T, Liu L, Gao T, Zhang T, Yuan D, Di J, Guo Z, Luo Z, Yuan H, Liu J. V-domain immunoglobulin suppressor of T-cell activation and programmed death receptor 1 dual checkpoint blockade enhances antitumour immunity and survival in glioblastoma. Br J Pharmacol 2025; 182:1306-1323. [PMID: 39626657 DOI: 10.1111/bph.17404] [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: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The current therapy cannot meet the needs of glioblastoma (GBM). V-domain immunoglobulin suppressor of T-cell activation (VISTA) is significantly up-regulated in GBM patients; however, its therapeutic potential in GBM is still unclear. EXPERIMENTAL APPROACH Flow cytometry was used to detect the expression of VISTA and the co-expression pattern of VISTA and programmed death receptor 1 (PD-1) on brain infiltrating lymphocytes of GBM mice. Monoclonal antibody therapy was used to evaluate the therapeutic effect of α-VISTA monotherapy and α-VISTA combined with α-PD-1 on GBM mice. Transcriptome analysis, flow cytometry, and immunofluorescence were used to detect changes of immune microenvironment in mouse brain tumours. Immunofluorescence and TCGA data analysis were used to further validate the combined treatment strategy on patient data. KEY RESULTS Compared with normal mice, the frequency of VISTA expression and co-expression of VISTA and PD-1 on tumour-infiltrating lymphocytes (TILs) in tumour-bearing mice was increased. Anti-VISTA monotherapy significantly up-regulated multiple immune stimulation-related pathways and moderately prolonged mouse survival time. Blocking the immune checkpoint VISTA and PD-1 significantly prolonged the survival time of mice and cured about 80% of the mice; CD8+ T cells played an important role in this process. In addition, we found that the expression of VISTA and PD-1 was significantly up-regulated in GBM patients by immunofluorescence, and patients with high expression of VISTA and PD-1 were associated with poor overall survival. This combination of blocking the immune checkpoint VISTA and PD-1 may achieve clinical transformation in GBM.
Collapse
Affiliation(s)
- Shasha Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tao Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ting Gao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Dingyi Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianwen Di
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhanying Guo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhijie Luo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
31
|
Wang L, Gu M, Zhang X, Kong T, Liao J, Zhang D, Li J. Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413367. [PMID: 39854126 PMCID: PMC11905078 DOI: 10.1002/advs.202413367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/14/2024] [Indexed: 01/26/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
Collapse
Affiliation(s)
- Liyin Wang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Min Gu
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Xiaoli Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | | | - Jun Liao
- Institute of Systems BiomedicineBeijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Dan Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Jingwu Li
- The First Hospital of China Medical UniversityLiaoning110001China
| |
Collapse
|
32
|
Wang R, Zhang Y, Guo Y, Zeng W, Li J, Wu J, Li N, Zhu A, Li J, Di L, Cao P. Plant-derived nanovesicles: Promising therapeutics and drug delivery nanoplatforms for brain disorders. FUNDAMENTAL RESEARCH 2025; 5:830-850. [PMID: 40242551 PMCID: PMC11997602 DOI: 10.1016/j.fmre.2023.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/17/2023] [Accepted: 09/10/2023] [Indexed: 04/18/2025] Open
Abstract
Plant-derived nanovesicles (PDNVs), including plant extracellular vesicles (EVs) and plant exosome-like nanovesicles (ELNs), are natural nano-sized membranous vesicles containing bioactive molecules. PDNVs consist of a bilayer of lipids that can effectively encapsulate hydrophilic and lipophilic drugs, improving drug stability and solubility as well as providing increased bioavailability, reduced systemic toxicity, and enhanced target accumulation. Bioengineering strategies can also be exploited to modify the PDNVs to achieve precise targeting, controlled drug release, and massive production. Meanwhile, they are capable of crossing the blood-brain barrier (BBB) to transport the cargo to the lesion sites without harboring human pathogens, making them excellent therapeutic agents and drug delivery nanoplatform candidates for brain diseases. Herein, this article provides an initial exposition on the fundamental characteristics of PDNVs, including biogenesis, uptake process, isolation, purification, characterization methods, and source. Additionally, it sheds light on the investigation of PDNVs' utilization in brain diseases while also presenting novel perspectives on the obstacles and clinical advancements associated with PDNVs.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingjie Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yumiao Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wei Zeng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jinge Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jie Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Anran Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
33
|
Zheng Y, Shi J. EFNB1 drives glioma progression and shapes the immune microenvironment: a potential prognostic biomarker. Discov Oncol 2025; 16:249. [PMID: 40014231 PMCID: PMC11868007 DOI: 10.1007/s12672-025-01867-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
Gliomas, a heterogeneous group of tumors affecting the brain and spinal cord, present a significant clinical challenge. Ephrin B1 (EFNB1) has been implicated in various malignancies. However, its role in gliomas remained poorly understood. Hence, this study aimed to elucidate the connection between EFNB1 and the progression of glioma. A retrospective RNA-seq analysis was conducted by utilizing the data from glioma patients in the TCGA and CGGA databases. Kaplan-Meier survival analysis and multivariate regression models were employed to evaluate the prognostic significance of EFNB1. RT-PCR was used to quantify EFNB1 expression in glioma tissues and cell lines. Meanwhile, in vitro assays were carried out to assess its functional roles in glioma cells. Our findings demonstrated that EFNB1 expression was significantly elevated in gliomas and other cancers. Moreover, high EFNB1 expression was closely correlated with advanced clinical stages and poor prognosis. Notably, multivariate analysis identified EFNB1 as an independent prognostic factor for overall survival. KEGG pathway analysis suggested that EFNB1 was involved in critical biological processes, including the cell cycle, protein processing in the endoplasmic reticulum, Epstein-Barr virus infection, and Salmonella infection. Furthermore, EFNB1 expression was associated with immune cell infiltration, particularly Th2 cells, macrophages, and plasmacytoid dendritic cells. In glioma cells, EFNB1 expression was markedly increased. Consequently, functional experiments demonstrated that EFNB1 knockdown inhibited glioma cell proliferation, invasion, and migration. These results highlighted EFNB1 as a novel independent prognostic biomarker and suggest its potential role in shaping the immunological microenvironment of gliomas.
Collapse
Affiliation(s)
- Yungui Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, China
| | - Jiasong Shi
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, China.
| |
Collapse
|
34
|
Lv P, Zhang Y, Wu W, Jiang X, Xiang W. Pan-cancer analysis identifies ADAM12 as a prognostic biomarker and indicator of immune infiltration in glioma. Sci Rep 2025; 15:6314. [PMID: 39984619 PMCID: PMC11845722 DOI: 10.1038/s41598-025-90121-0] [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: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
ADAM12, part of the adisintegrin and metalloproteases (ADAMs) family, has been widely reported in recent years to be associated with various malignant tumor behaviors, including migration, invasion, and treatment resistance. However, its role at the pan-cancer level remains insufficiently characterized. In this study, pan-cancer data were utilized to elucidate the expression patterns, prognostic significance, and potential roles of ADAM12 within the tumor immune microenvironment. An in-depth analysis of ADAM12 in gliomas was also conducted. Our findings revealed that ADAM12 expression is markedly overexpressed in glioma tissues, enhances glioma cell malignancy, and is associated with a worse prognosis. These results suggest that ADAM12 may serve as a biomarker for predicting glioma malignancy and patient prognosis, as well as a potential therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Peng Lv
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yanbin Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China.
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
35
|
Li J, Cui Y, Jiang X, Chi X, Li H, Ma X, Tang Y, Huang D, Liu Z. Surface-engineered bio-manufactured gas vesicles for multimodal imaging of glioma. J Nanobiotechnology 2025; 23:116. [PMID: 39966815 PMCID: PMC11834395 DOI: 10.1186/s12951-025-03203-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Integrated imaging techniques offer enhanced medical insights into the central nervous system by combining different modalities. In glioma diagnosis, the challenge often lies in delivering contrast agents effectively across the blood-brain barrier. We present an integrated multimodal imaging biohybrid, GV@pCY5, which enables blood-brain barrier penetrating as well as fluorescence and ultrasound (FL/US) imaging capabilities. This biohybrid is created by decorating a far-red-fluorescent cyanine dye (CY5) onto polyethyleneimine (PEI)-coated gas vesicles (GV). The layer-by-layer assembly improves the stability and performance of GV@pCY5 under ultrasound, thanks to the hydration shell variation induced by PEI. Given to the blood-brain barrier penetrating ability, GV@pCY5 demonstrates increase both in fluorescence and ultrasound imaging performance compared to single-component systems, proving effective for glioma diagnosis in vivo. This study underscores the potential of the FL/US platform for dual ratiometric imaging of various cerebral conditions.
Collapse
Affiliation(s)
- Juanjuan Li
- Faculty of Animal Science and Technology, Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
| | - Yutong Cui
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
- Hainan Forestry Science Research Institute, Haikou, 571199, China
| | - Xiaoli Jiang
- Faculty of Animal Science and Technology, Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
| | - Xue Chi
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
| | - Hong Li
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
| | - Xiang Ma
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
| | - Yanqiong Tang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China
| | - Denggao Huang
- Department of Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, China
| | - Zhu Liu
- Faculty of Animal Science and Technology, Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China.
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, China.
| |
Collapse
|
36
|
Look T, Sankowski R, Bouzereau M, Fazio S, Sun M, Buck A, Binder N, Mastall M, Prisco F, Seehusen F, Frei J, Wyss C, Snijder B, Nombela Arrieta C, Weller M, Pascolo S, Weiss T. CAR T cells, CAR NK cells, and CAR macrophages exhibit distinct traits in glioma models but are similarly enhanced when combined with cytokines. Cell Rep Med 2025; 6:101931. [PMID: 39889712 PMCID: PMC11866521 DOI: 10.1016/j.xcrm.2025.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/17/2024] [Accepted: 01/03/2025] [Indexed: 02/03/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising immunotherapy against cancer. Although there is a growing interest in other cell types, a comparison of CAR immune effector cells in challenging solid tumor contexts is lacking. Here, we compare mouse and human NKG2D-CAR-expressing T cells, natural killer (NK) cells, and macrophages against glioblastoma, the most aggressive primary brain tumor. In vitro we show that T cell cancer killing is CAR dependent, whereas intrinsic cytotoxicity overrules CAR dependence for NK cells, and CAR macrophages reduce glioma cells in co-culture assays. In orthotopic immunocompetent glioma mouse models, systemically administered CAR T cells demonstrate superior accumulation in the tumor, and each immune cell type induces distinct changes in the tumor microenvironment. An otherwise low therapeutic efficacy is significantly enhanced by co-expression of pro-inflammatory cytokines in all CAR immune effector cells, underscoring the necessity for multifaceted cell engineering strategies to overcome the immunosuppressive solid tumor microenvironment.
Collapse
Affiliation(s)
- Thomas Look
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Manon Bouzereau
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Serena Fazio
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Miaomiao Sun
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Alicia Buck
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland; Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Niklas Binder
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Maximilian Mastall
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Julia Frei
- Department of Dermatology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Conrad Wyss
- Department of Dermatology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Cesar Nombela Arrieta
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Steve Pascolo
- Department of Dermatology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
37
|
Wang K, Wan J, Zheng R, Xiao Y, Lv F, Ge H, Yang G, Cheng Y. SPP1 as a Prognostic and Immunotherapeutic Biomarker in Gliomas and Other Cancer Types: A Pan-Cancer Study. J Inflamm Res 2025; 18:2247-2265. [PMID: 39963684 PMCID: PMC11832131 DOI: 10.2147/jir.s505237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Background Gliomas, including glioblastoma (GBM), present significant treatment challenges due to their poor prognosis and complex tumor microenvironment. This study investigates the role of Secreted Phosphoprotein 1 (SPP1) as a prognostic and immunotherapeutic biomarker in gliomas and other cancers through pan-cancer analysis. Methods A comprehensive pan-cancer analysis was conducted using datasets from UCSC TCGA Pan-Cancer, TCGA-GBM, UALCAN, and single-cell sequencing data from GEO and TISCH. The correlation of SPP1 expression with overall survival (OS), progression-free survival (PFS), immune cell infiltration, and immune checkpoint markers was analyzed. Functional validation was performed via SPP1 knockdown in glioma cell lines to evaluate effects on proliferation, invasion, and immune interactions. Results SPP1 was found to be overexpressed in 27 tumor types, with high expression correlating with poor OS, PFS, and increased immune cell infiltration, particularly with CD8+ T cells and macrophages. Single-cell analysis indicated SPP1 enrichment in macrophages interacting with malignant GBM cells. Knockdown of SPP1 significantly inhibited glioma cell proliferation, invasion, and promoted apoptosis. Conclusion The findings suggest that SPP1 is a promising target for immunotherapy, potentially improving outcomes for patients with gliomas and other cancers. Further research is warranted to explore SPP1-targeted therapies and their efficacy in clinical settings.
Collapse
Affiliation(s)
- Kan Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150007, People’s Republic of China
| | - Jinxin Wan
- Department of Neurosurgery, Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai City, Guangdong Province, 519090, People’s Republic of China
| | - Ruipeng Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150007, People’s Republic of China
| | - Yifei Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150007, People’s Republic of China
| | - Fengjun Lv
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150007, People’s Republic of China
| | - Haitao Ge
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150007, People’s Republic of China
| | - Guang Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, People’s Republic of China
| | - Yu Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150007, People’s Republic of China
| |
Collapse
|
38
|
Hui H, Zhou F, Pei S, Zhou W, Shang J, Wang P, Deng Z, Zhou X. Thioredoxin related transmembrane protein 1acts as a prognostic indictor and promotes proliferation and TMZ resistance of lower-grade glioma. Sci Rep 2025; 15:5246. [PMID: 39939413 PMCID: PMC11822014 DOI: 10.1038/s41598-025-89908-y] [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: 12/01/2024] [Accepted: 02/10/2025] [Indexed: 02/14/2025] Open
Abstract
Thioredoxin Related Transmembrane Protein 1 (TMX1) encodes a thiol-disulfide oxidoreductase and regulates cleavage, formation, or isomerization of disulfide bonds among cysteine residues in proteins. Using bioinformatic analysis, we firstly prove TMX1 is significantly upregulated in lower-grade gliomas (LGGs) and associated with poor prognosis of LGG patients. Besides, it's also highly expressed in higher WHO grade, 1p/19q non-co-deleted and isocitrate dehydrogenase (IDH) wildtype LGGs, thus we established a TMX1-based nomogram model, which exhibits a strong and stable predictive ability in the prognosis of LGG patients. Then, we also noticed that TMX1 was significantly associated with the immune cell infiltrations in LGGs, especially in B cell, CD8+ T cell, CD4+T cell, dendritic cell, macrophage and neutrophil. Meanwhile, it was also highly correlated to the expressions of MKI67, PCNA, PROM1 and SOX2 in LGGs, and that higher TMX1 LGGs showed a stronger resistance to temozolomide (TMZ). Finally, in our in vitro and in vivo experiments, we verified that TMX1 is highly expressed in LGG patients clinically, and it not only regulates the proliferative ability of SW1088 and SW1783 cells both in vitro and in vivo, it can also be inhibited to increase the TMZ therapy sensitivity in vivo. These results revealed that TMX1 acts as a strong prognostic biomarker in LGGs, and targeting TMX1 can be an efficient way to increase the TMZ therapy in LGG patients.
Collapse
Affiliation(s)
- Hongyan Hui
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Clinical Pharmacy, Henan Province Key Subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, Weihui, 453100, Henan, People's Republic of China
| | - Fang Zhou
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Clinical Pharmacy, Henan Province Key Subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, Weihui, 453100, Henan, People's Republic of China
| | - Sujuan Pei
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Clinical Pharmacy, Henan Province Key Subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, Weihui, 453100, Henan, People's Republic of China
| | - Wenyan Zhou
- Department of Medical Imaging, The First Affiliated Hospital of Xinxiang Medical University, 453100, Weihui, Henan, People's Republic of China
| | - Jie Shang
- Medical Science, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
| | - Pengwei Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Clinical Pharmacy, Henan Province Key Subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, Weihui, 453100, Henan, People's Republic of China
| | - Zhijian Deng
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Clinical Pharmacy, Henan Province Key Subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, Weihui, 453100, Henan, People's Republic of China
| | - Xiang Zhou
- Brain Hospital, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, People's Republic of China.
| |
Collapse
|
39
|
Gordon KS, Perez CR, Garmilla A, Lam MSY, Aw JJY, Datta A, Lauffenburger DA, Pavesi A, Birnbaum ME. Pooled screening for CAR function identifies novel IL-13Rα2-targeted CARs for treatment of glioblastoma. J Immunother Cancer 2025; 13:e009574. [PMID: 39933837 PMCID: PMC11815465 DOI: 10.1136/jitc-2024-009574] [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: 05/08/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) therapies have demonstrated potent efficacy in treating B-cell malignancies, but have yet to meaningfully translate to solid tumors. Nonetheless, they are of particular interest for the treatment of glioblastoma, which is an aggressive form of brain cancer with few effective therapeutic options, due to their ability to cross the highly selective blood-brain barrier. METHODS Here, we use our pooled screening platform, CARPOOL, to expedite the discovery of CARs with antitumor functions necessary for solid tumor efficacy. We performed selections in primary human T cells expressing a library of 1.3×106 third generation CARs targeting IL-13Rα2, a cancer testis antigen commonly expressed in glioblastoma. Selections were performed for cytotoxicity, proliferation, memory formation, and persistence on repeated antigen challenge. RESULTS Each enriched CAR robustly produced the phenotype for which it was selected, and one enriched CAR triggered potent cytotoxicity and long-term proliferation on in vitro tumor rechallenge. It also showed significantly improved persistence and comparable tumor control in a microphysiological human in vitro model and a xenograft model of human glioblastoma, but also demonstrated increased off-target recognition of IL-13Rα1. CONCLUSION Taken together, this work demonstrates the utility of extending CARPOOL to diseases beyond hematological malignancies and represents the largest exploration of signaling combinations in human primary cells to date.
Collapse
Affiliation(s)
- Khloe S Gordon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Singapore-MIT Alliance for Research and Technology Centre, Singapore
| | - Caleb R Perez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Singapore-MIT Alliance for Research and Technology Centre, Singapore
| | - Andrea Garmilla
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Maxine S Y Lam
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
| | - Joey J Y Aw
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
| | - Anisha Datta
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Michael E Birnbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Singapore-MIT Alliance for Research and Technology Centre, Singapore
| |
Collapse
|
40
|
Robinson SD, Filippopoulou C, Besta S, Samuels M, Betrán AL, Abu Ajamieh M, Vella V, Jones W, Giamas G. Spatial biology - unravelling complexity within the glioblastoma microenvironment. Trends Mol Med 2025:S1471-4914(25)00014-0. [PMID: 39934022 DOI: 10.1016/j.molmed.2025.01.014] [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: 12/05/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025]
Abstract
The advent and refinement of state-of-the-art spatial biology technologies have facilitated analysis that combines the advantages of high-throughput single cell analysis with techniques that preserve tissue architecture. This combination of cellular phenotyping with retained spatial context provides a much greater understanding of cellular interactions within the tumour microenvironment (TME). For glioblastoma, with its significant intra-tumoural heterogeneity, cellular plasticity, and complex TME, appreciating and understanding these spatial patterns may prove key to improving patient outcomes. This review examines the advances in spatial biology techniques, discusses how these methodologies are being applied to study glioblastoma, and explores how spatial information improves understanding of the TME. Ultimately, it is this spatial context that will accelerate the identification of more effective treatments for glioblastoma.
Collapse
Affiliation(s)
- Stephen D Robinson
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK; Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, BN2 5BD, UK.
| | - Chrysa Filippopoulou
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Simoni Besta
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mark Samuels
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Andrea L Betrán
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Maha Abu Ajamieh
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Viviana Vella
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - William Jones
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK; International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
41
|
Liang Z, Zhao S, Liu Y, Cheng C. The promise of mitochondria in the treatment of glioblastoma: a brief review. Discov Oncol 2025; 16:142. [PMID: 39924629 PMCID: PMC11807951 DOI: 10.1007/s12672-025-01891-y] [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: 10/01/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Glioblastoma (GBM) is a prevalent and refractory type of brain tumor. Over the past two decades, there have been minimal advancements in GBM therapy. The current standard treatment involves surgical excision followed by radiation and chemotherapy. Compared to other tumors, GBM is more challenging to treat due to the presence of glioma stem-like cells (GSCs) and the blood-brain barrier, resulting in an extremely low survival rate. Mitochondria play a critical role in tumor respiration, metabolism, and multiple signaling pathways involved in tumor formation, progression, and cell apoptosis. Consequently, mitochondria represent promising targets for developing novel anticancer agents, including those targeting oxidative phosphorylation, reactive oxygen species (ROS), mitochondrial transfer, and mitophagy. This review outlines the mitochondrial-related therapeutic targets in GBM, highlighting the potential of mitochondria as a target for GBM treatment.
Collapse
Affiliation(s)
- Zhuo Liang
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Songyun Zhao
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yuankun Liu
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Chao Cheng
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
42
|
Yu W, Gui S, Peng L, Luo H, Xie J, Xiao J, Yilamu Y, Sun Y, Cai S, Cheng Z, Tao Z. STAT3-controlled CHI3L1/SPP1 positive feedback loop demonstrates the spatial heterogeneity and immune characteristics of glioblastoma. Dev Cell 2025:S1534-5807(25)00034-6. [PMID: 39933531 DOI: 10.1016/j.devcel.2025.01.014] [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: 08/05/2024] [Revised: 10/30/2024] [Accepted: 01/17/2025] [Indexed: 02/13/2025]
Abstract
Proneural-mesenchymal transition (PMT) is a phenotypic alteration and contributes to the malignant progression of glioblastoma (GBM). Macrophages, as a main infiltrating component of the tumor immune microenvironment (TIM), control the biological processes of PMT; however, the mechanisms driving this process remain largely unknown. Here, the overall landscape of tumor and nontumor cells was described by scMulti-omics technology. Then, we demonstrated that chitinase-3-like protein 1 (CHI3L1) played a critical role in maintaining mesenchymal (MES) status and reprogramming macrophage phenotype using C57BL/6 and NSG mice models derived from PN20 cells. Mechanistically, osteopontin (OPN)/ITGB1 maintained the activation of nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways by establishing a positive feedback loop with the CHI3L1-STAT3 axis, resulting in PMT. CHI3L1 enhanced the phosphorylation, nuclear localization, and transcriptional activity of STAT3 via directly binding its coiled-coil domain (CCD). Importantly, we screened and validated that hygromycin B (HB), an inhibitor of the STAT3-CCD domain, disrupted the CHI3L1-STAT3 interaction, thereby reducing the tumor burden in vitro and in vivo.
Collapse
Affiliation(s)
- Wanli Yu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Shikai Gui
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Lunshan Peng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Haitao Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jiabao Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Juexian Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yimuran Yilamu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Yi Sun
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Shihao Cai
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Zujue Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Zhennan Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China; Neurosurgical Institute, Nanjing University, Nanjing 210008, Jiangsu, China.
| |
Collapse
|
43
|
Du W, Na J, Zhong L, Zhang P. Advances in preclinical and clinical studies of oncolytic virus combination therapy. Front Oncol 2025; 15:1545542. [PMID: 39990685 PMCID: PMC11842258 DOI: 10.3389/fonc.2025.1545542] [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: 12/15/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Oncolytic viruses represent a distinct class of viruses that selectively infect and destroy tumor cells while sparing normal cells. Despite their potential, oncolytic viruses encounter several challenges as standalone therapies. Consequently, the combination of oncolytic viruses with other therapeutic modalities has emerged as a prominent research focus. This paper summarizes the tumor-killing mechanisms of oncolytic viruses, explores their integration with radiotherapy, chemotherapy, immune checkpoint inhibitors, CAR-T, and CAR-NK therapies, and provides an overview of related clinical trials. By synthesizing these advancements, this study seeks to offer valuable insights for the clinical translation of oncolytic virus combination therapies.
Collapse
Affiliation(s)
- Wenlong Du
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Pumin Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| |
Collapse
|
44
|
Fidaleo AM, Bach MD, Orbeta S, Abdullaev IF, Martino N, Adam AP, Boulos MA, Dulin NO, Paul AR, Kuo YH, Mongin AA. LRRC8A-containing anion channels promote glioblastoma proliferation via a WNK1/mTORC2-dependent mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.636139. [PMID: 39975357 PMCID: PMC11838495 DOI: 10.1101/2025.02.02.636139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Leucine-rich repeat-containing protein 8A (LRRC8A) is the essential subunit of ubiquitous volume-regulated anion channels (VRACs). LRCC8A is overexpressed in several cancers and promotes negative survival outcomes via a poorly defined mechanism. Here, we explored the role of LRRC8A and VRACs in the progression of glioblastoma (GBM), the most common and deadly primary brain tumor. We found that, as compared to healthy controls, LRRC8A mRNA was strongly upregulated in surgical GBM specimens, patient-derived GBM cell lines, and GBM datasets from The Cancer Genome Atlas (TCGA). Our in-silico analysis indicated that patients belonging to the lowest LRRC8A expression quartile demonstrated a trend for extended life expectancy. In patient-derived GBM cultures, siRNA-driven LRRC8A knockdown reduced cell proliferation and additionally decreased intracellular chloride levels and inhibited activity of mTOR complex 2. The antiproliferative effect of LRRC8A downregulation was recapitulated with a pharmacological inhibitor of VRAC. Our ensuing biochemical and molecular biology analyses established that the LRRC8A-containing VRACs facilitate GBM proliferation via a new mechanism involving non-enzymatic actions of the chloride-sensitive protein kinase WNK1. Accordingly, the chloride-bound WNK1 stimulates mTORC2 and the mTORC2-dependent protein kinases AKT and SGK, which promote proliferation. These findings establish the new mTORC2-centric axis for VRAC dependent regulation of cellular functions and uncover potential targets for GBM intervention.
Collapse
Affiliation(s)
- Antonio M Fidaleo
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Martin D Bach
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Shaina Orbeta
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Iskandar F Abdullaev
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Mateo A Boulos
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Alexandra R Paul
- Department of Neurosurgery, Albany Medical College, Albany, NY, USA
| | - Yu-Hung Kuo
- Neurosurgery, Luminis Health Anne Arundel Medical Center, Annapolis, MD, USA
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
45
|
Asey B, Pantel TF, Mohme M, Zghaibeh Y, Dührsen L, Silverbush D, Schüller U, Drexler R, Ricklefs FL. Peripheral blood-derived immune cell counts as prognostic indicators and their relationship with DNA methylation subclasses in glioblastoma patients. Brain Pathol 2025:e13334. [PMID: 39901324 DOI: 10.1111/bpa.13334] [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: 08/27/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Glioblastomas are known for their immunosuppressive tumor microenvironment, which may explain the failure of most clinical trials in the past decade. Recent studies have emphasized the significance of stratifying glioblastoma patients to predict better therapeutic responses and survival outcomes. This study aims to investigate the prognostic relevance of peripheral immune cell counts sampled prior to surgery, with a special focus on methylation-based subclassification. Peripheral blood was sampled in patients with newly diagnosed (n = 176) and recurrent (n = 41) glioblastoma at the time of surgery and analyzed for neutrophils, monocytes, leukocytes, platelets, neutrophil-lymphocyte ratio, lymphocyte-monocyte ratio, and platelet-lymphocyte ratio. Peripheral immune cell counts were correlated with patients' survival after combined radiochemotherapy. In addition, 850 k genome-wide DNA methylation was assessed on tissue for defining tumor subclasses and performing cell-type deconvolution. In newly diagnosed glioblastoma, patients with higher peripheral neutrophil counts had an unfavorable overall survival (OS) (p = 0.01, median overall-survival (mOS) 17.0 vs. 10.0 months). At the time of first recurrence, a significant decrease of peripheral immune cell counts was observed, and elevated monocyte (p = 0.03), neutrophil (p = 0.04), and platelet (p = 0.01) counts were associated with poorer survival outcomes. DNA methylation subclass-stratified analysis revealed a significant survival influence of neutrophils (p = 0.007) and lymphocytes (p = 0.04) in the mesenchymal (MES) subclass. Integrating deconvolution of matched tumor tissue showed that platelets and monocytes were correlated with a more differentiated, tumor-progressive cell state, and peripheral immune cell counts were most accurately reflected in tissue of the MES subclass. This study illustrates a restricted prognostic significance of peripheral immune cell counts in newly diagnosed glioblastoma and a constrained representation in matched tumor tissue, but it demonstrates a more pertinent situation at the time of recurrence and after DNA methylation-based stratification.
Collapse
Affiliation(s)
- Benedikt Asey
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias F Pantel
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yahya Zghaibeh
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lasse Dührsen
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dana Silverbush
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Research Institute Children's Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Richard Drexler
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Zhu A, Shao S, Hu J, Tu W, Song Z, Liu Y, Liu J, Zhang Q, Li J. Hydrogen sulfide-generating semiconducting polymer nanoparticles for amplified radiodynamic-ferroptosis therapy of orthotopic glioblastoma. MATERIALS HORIZONS 2025; 12:973-986. [PMID: 39552555 DOI: 10.1039/d4mh01356e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
A variety of therapeutic strategies are available to treat glioblastoma (GBM), but the tumor remains one of the deadliest due to its aggressive invasiveness, restrictive blood-brain barrier (BBB), and exceptional resistance to drugs. In this study, we present a hydrogen sulfide (H2S)-generating semiconducting polymer nanoparticle (PFeD@Ang) for amplified radiodynamic-ferroptosis therapy of orthotopic glioblastoma. Our results show that in an acidic tumor microenvironment (TME), H2S donors produce large amounts of H2S, which inhibits mitochondrial respiration and alleviates cellular hypoxia, thus enhancing the radiodynamic effect during X-ray irradiation; meanwhile, Fe3+ is reduced to Fe2+ by tannic acid in an acidic TME, which promotes an iron-dependent cell death process in tumors. H2S facilitates the ferroptosis process by increasing the local H2O2 concentration via inhibiting catalase activity. This kind of amplified radiodynamic-ferroptosis therapeutic strategy could remarkably inhibit glioma progression in an orthotopic GBM mouse model. Our study demonstrates the potential of PFeD@Ang for GBM treatment via targeted delivery and combinational therapeutic actions of RDT and ferroptosis therapy.
Collapse
Affiliation(s)
- Anni Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Shuai Shao
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jinyuan Hu
- Faculty of Arts and Sciences, Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Beijing Normal University, Zhuhai 519087, China
| | - Wenzhi Tu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| | - Zheming Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Yue Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Jiansheng Liu
- Department of Neurology, Shanghai Xuhui District Central Hospital, Zhongshan-Xuhui Hospital Fudan University, Shanghai 200032, China.
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
47
|
Wang Y, Li G, Su J, Liu Y, Zhang X, Zhang G, Wu Z, Li J, Wang X, Zhang Y, Bai M, Yao Y, Wang R, Shao K. Tumor-Associated Macrophages Nano-Reprogrammers Induce "Gear Effect" to Empower Glioblastoma Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406839. [PMID: 39797442 DOI: 10.1002/smll.202406839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/24/2024] [Indexed: 01/13/2025]
Abstract
Glioblastoma (GBM), the most malignant brain tumor with high prevalence, remains highly resistant to the existing immunotherapies due to the significant immunosuppression within tumor microenvironment (TME), predominantly manipulated by M2-phenotypic tumor-associated macrophages (M2-TAMs). Here in this work, an M2-TAMs targeted nano-reprogrammers, MG5-S-IMDQ, is established by decorating the mannose molecule as the targeting moiety as well as the toll-like receptor (TLR) 7/8 agonist, imidazoquinoline (IMDQ) on the dendrimeric nanoscaffold. MG5-S-IMDQ demonstrated an excellent capacity of penetrating the blood-brain barrier (BBB) as well as selectively targeting M2-TAMs in the GBM microenvironment, leading to a phenotype transformation and function restoration of TAMs shown as heightened phagocytic activity toward tumor cells, enhanced cytotoxic effects, and improved tumor antigen cross-presentation capability. In the meantime, by induction of a function-oriented "gear effect", MG5-S-IMDQ treatment extended its impact systemically by enhancing the infiltration of type I conventional dendritic cells (cDC1s) into the tumor sites and bolstering adaptive immune responses. In sum, by precisely working on M2-TAMs as a unique target in tumor situ, the nano-reprogrammers successfully established a robust immune network that worked synergistically to combat tumors. This facile nanoplatform-based immunomodulatory strategy, serving as a powerful and convenient immune monotherapy or as a complementary treatment alongside other therapies like surgery, provided deep insights for advancing translational study in GBM.
Collapse
Affiliation(s)
- Yang Wang
- Cancer Hospital of Dalian University of Technology, Dalian University of Technology, Shenyang, 110042, China
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Guangzhe Li
- State Key Laboratory of Fine Chemicals, Department of Pharmacy, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jianlong Su
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Yiming Liu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Xiaomai Zhang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Guanyi Zhang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Zhihao Wu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jinrong Li
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Xu Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Yuxuan Zhang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Mingrui Bai
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Yuanhang Yao
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Ruimin Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Kun Shao
- Cancer Hospital of Dalian University of Technology, Dalian University of Technology, Shenyang, 110042, China
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
48
|
Thomas MPH, Ajaib S, Tanner G, Bulpitt AJ, Stead LF. GBMPurity: A Machine Learning Tool for Estimating Glioblastoma Tumour Purity from Bulk RNA-seq Data. Neuro Oncol 2025:noaf026. [PMID: 39891579 DOI: 10.1093/neuonc/noaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) presents a significant clinical challenge due to its aggressive nature and extensive heterogeneity. Tumour purity, the proportion of malignant cells within a tumour, is an important covariate for understanding the disease, having direct clinical relevance or obscuring signal of the malignant portion in molecular analyses of bulk samples. However, current methods for estimating tumour purity are non-specific and technically demanding. Therefore, we aimed to build a reliable and accessible purity estimator for GBM. METHODS We developed GBMPurity, a deep-learning model specifically designed to estimate the purity of IDH-wildtype primary GBM from bulk RNA-seq data. The model was trained using simulated pseudobulk tumours of known purity from labelled single-cell data acquired from the GBmap resource. The performance of GBMPurity was evaluated and compared to several existing tools using independent datasets. RESULTS GBMPurity outperformed existing tools, achieving a mean absolute error of 0.15 and a concordance correlation coefficient of 0.88 on validation datasets. We demonstrate the utility of GBMPurity through inference on bulk RNA-seq samples and observe reduced purity of the Proneural molecular subtype relative to the Classical, attributed to the increased presence of healthy brain cells. CONCLUSIONS GBMPurity provides a reliable and accessible tool for estimating tumour purity from bulk RNA-seq data, enhancing the interpretation of bulk RNA-seq data and offering valuable insights into GBM biology. To facilitate the use of this model by the wider research community, GBMPurity is available as a web-based tool at: https://gbmdeconvoluter.leeds.ac.uk/.
Collapse
Affiliation(s)
- Morgan P H Thomas
- School of Computer Science, University of Leeds, UK
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Shoaib Ajaib
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Georgette Tanner
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | - Lucy F Stead
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| |
Collapse
|
49
|
Khilar S, Dembinska-Kenner A, Hall H, Syrmos N, Ligarotti GKI, Plaha P, Apostolopoulos V, Chibbaro S, Barbagallo GMV, Ganau M. Towards a New Dawn for Neuro-Oncology: Nanomedicine at the Service of Drug Delivery for Primary and Secondary Brain Tumours. Brain Sci 2025; 15:136. [PMID: 40002469 PMCID: PMC11852924 DOI: 10.3390/brainsci15020136] [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: 12/12/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background/Objectives: Primary and secondary brain tumours often hold devastating prognoses and low survival rates despite the application of maximal neurosurgical resection, and state-of-the-art radiotherapy and chemotherapy. One limiting factor in their management is that several antineoplastic agents are unable to cross the blood-brain barrier (BBB) to reach the tumour microenvironment. Nanomedicine could hold the potential to become an effective means of drug delivery to overcome previous hurdles towards effective neuro-oncological treatments. (2) Methods: A scoping review following the PRISMA-ScR guidelines and checklist was conducted using key terms input into PubMed to find articles that reflect emerging trends in the utilisation of nanomedicine in drug delivery for primary and secondary brain tumours. (3) Results: The review highlights various strategies by which different nanoparticles can be exploited to bypass the BBB; we provide a synthesis of the literature on the ongoing contributions to therapeutic protocols based on chemotherapy, immunotherapy, focused ultrasound, radiotherapy/radiosurgery, and radio-immunotherapy. (4) Conclusions: The emerging trends summarised in this scoping review indicate encouraging advantageous properties of nanoparticles as potential effective drug delivery mechanisms; however, there are still nanotoxicity issues that largely remain to be addressed before the translation of these innovations from laboratory to clinical practice.
Collapse
Affiliation(s)
- Smita Khilar
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Antonina Dembinska-Kenner
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Helen Hall
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Nikolaos Syrmos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Puneet Plaha
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Vasileios Apostolopoulos
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
| | - Salvatore Chibbaro
- Neurosurgery Unit, Department of Medical and Surgical Sciences and Neurosciences, Siena University, 53100 Siena, Italy
| | | | - Mario Ganau
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, UK; (S.K.); (H.H.)
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
50
|
Cheng W, Duan Z, Chen H, Wang Y, Wang C, Pan Y, Wu J, Wang N, Qu H, Xue X. Macrophage membrane-camouflaged pure-drug nanomedicine for synergistic chemo- and interstitial photodynamic therapy against glioblastoma. Acta Biomater 2025; 193:392-405. [PMID: 39800099 DOI: 10.1016/j.actbio.2025.01.016] [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: 09/23/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Glioblastoma (GBM) persists as a highly fatal malignancy, with current clinical treatments showing minimal progress over years. Interstitial photodynamic therapy (iPDT) holds promise due to its minimally invasive nature and low toxicity but is impeded by poor photosensitizer penetration and inadequate GBM targeting. Here, we developed a biomimetic pure-drug nanomedicine (MM@CT), which co-assembles the photosensitizer chlorin e6 (Ce6) and the first-line chemotherapeutic drug (temozolomide, TMZ) for GBM, then camouflaged with macrophage membranes. This design eliminates the need for traditional excipients, ensuring formulation safety and achieving exceptionally high drug loading with 73.2 %. By leveraging the biomimetic properties of macrophage membranes, MM@CT evades clearance by the mononuclear phagocyte system and can overcome blood circulatory barriers to target intracranial GBM tumors due to its inherent tumor-homing ability. Consequently, this targeted strategy enables precise delivery of TMZ to the tumor site while significantly enhancing Ce6 accumulation within the tumor tissue. Upon intra-tumoral irradiation using an optical fiber, activated Ce6 synergizes with TMZ to exert both cytotoxic effects from chemotherapy and unique advantages from iPDT simultaneously attacking GBM tumors in a dual manner. In subcutaneous and intracranial GBM mouse models, MM@CT exhibits remarkable anti-tumor efficacy with minimal systemic toxicity, emerging as a promising GBM treatment strategy. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) remains a formidable brain cancer, posing significant therapeutic challenges due to the presence of the blood-brain barrier (BBB) and tumor heterogeneity. To overcome these obstacles, we have developed MM@CT, a biomimetic nanomedicine with exceptional drug loading efficiency of 73.2 %. MM@CT incorporates the photosensitizer Ce6 and chemotherapy agent TMZ, encapsulated within nanoparticles and camouflaged with macrophage membranes. This innovative design enables efficient BBB penetration, precise tumor targeting, and synergistic application of chemotherapy and photodynamic therapy. Encouragingly, preclinical evaluations have demonstrated substantial antitumor activity with minimal systemic toxicity, positioning MM@CT as a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanjun Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ning Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|