1
|
Nussinov R, Yavuz BR, Jang H. Tumors and their microenvironments: Learning from pediatric brain pathologies. Biochim Biophys Acta Rev Cancer 2025; 1880:189328. [PMID: 40254040 PMCID: PMC12124968 DOI: 10.1016/j.bbcan.2025.189328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Early clues to tumors and their microenvironments come from embryonic development. Here we review the literature and consider whether the embryonic brain and its pathologies can serve as a better model. Among embryonic organs, the brain is the most heterogenous and complex, with multiple lineages leading to wide spectrum of cell states and types. Its dysregulation promotes neurodevelopmental brain pathologies and pediatric tumors. Embryonic brain pathologies point to the crucial importance of spatial heterogeneity over time, akin to the tumor microenvironment. Tumors dedifferentiate through genetic mutations and epigenetic modulations; embryonic brains differentiate through epigenetic modulations. Our innovative review proposes learning developmental brain pathologies to target tumor evolution-and vice versa. We describe ways through which tumor pharmacology can learn from embryonic brains and their pathologies, and how learning tumor, and its microenvironment, can benefit targeting neurodevelopmental pathologies. Examples include pediatric low-grade versus high-grade brain tumors as in rhabdomyosarcomas and gliomas.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
2
|
Chen P, Liu Y, Huang H, Li M, Xie H, Roy S, Gu J, Jin J, Deng K, Du L, Guo B. Genetically Engineered IL12/CSF1R-Macrophage Membrane-Liposome Hybrid Nanovesicles for NIR-II Fluorescence Imaging-Guided and Membrane-Targeted Mild Photothermal-Immunotherapy of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500131. [PMID: 40279543 DOI: 10.1002/advs.202500131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/19/2025] [Indexed: 04/27/2025]
Abstract
It is a big challenge for precision therapy of glioblastoma, mainly due to the existence of blood-brain barrier (BBB), tumor immunosuppressive microenvironment (TIM), and lack of efficient treatment paradigms. Herein, a theranostic nanoplatform for the second near-infrared window (NIR-II) fluorescence imaging-guided membrane-targeted mild photothermal-immunotherapy of glioblastoma using genetically engineered CSF1R/IL12-macrophage membrane (MM)-liposome hybrid nanovesicles, is reported. By mimicking lipophilic membrane probe (Dil) with octadecyl chains, a NIR-II emissive photothermal dye (IRC18), which realizes labeling of nanovesicle lipid bilayers for biodistribution tracing, glioblastoma diagnosis, and molecular imaging of tumoral microenvironment, is synthesized. Importantly, MM and c-RGD-decorated liposome together offer BBB crossing, tumor targeting, and long-term circulation; while, the genetically overexpressed CSF1R and IL12 on MM surface contribute to effective modulation of M2-to-M1 macrophage repolarization and local promotion of T cell cytotoxicity in glioblastoma microenvironment, respectively. Notably, through membrane fusion, IRC18 dyes translocate from nanovesicle lipid bilayers to glioblastoma membranes, which achieve membrane-targeted mild photothermal therapy to ablate primary tumor and induce immunogenic cell death to promote antigen presentation. More importantly, the combined blockade of the CSF1-CSF1R axis and IL-12 enrichment not only reprograms the tumor microenvironment through macrophage M1 repolarization but also activates cytotoxic T cells, ultimately achieving complete glioblastoma eradication. This research provides an efficient theranostic paradigm for glioblastoma treatment.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Traumatic Orthopedics, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yue Liu
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Menglong Li
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Hui Xie
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jian Jin
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Kai Deng
- Department of Traumatic Orthopedics, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Lixin Du
- Department of Medical Imaging, Shenzhen Longhua District Key Laboratory of Neuroimaging, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, 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
|
3
|
Zhao S, Sun Z, Xia M, Guo B, Qu Y, Wang J, Zhong Z, Meng F. Polymersome-enabled brain codelivery of STAT3 siRNA and CpG oligonucleotide boosts chemo-immunotherapy of malignant glioma. J Control Release 2025; 383:113764. [PMID: 40274070 DOI: 10.1016/j.jconrel.2025.113764] [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: 11/23/2024] [Revised: 03/22/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Malignant glioma represents one of the most aggressive primary tumors of the central nervous system. The immunotherapy of glioma is restrained by low immunogenicity, an immunosuppressive environment, and challenges in delivering therapeutics and immune-modulating agents. Here, we demonstrate that the systemic brain codelivery of STAT3 siRNA and CpG oligonucleotide using ApoE peptide-functionalized nano-polymersomes (tNano-S&C) significantly boosts the efficacy of chemo-immunotherapy for malignant glioma when combined with temozolomide (TMZ). The administration of STAT3 siRNA via tNano-S&C effectively knocked down STAT3 expression in glioma cells, resulting in increased sensitivity to TMZ treatment and enhancing immunogenic cell death. Furthermore, tNano-S&C was efficiently taken up by dendritic cells (DCs), inducing DC maturation and proinflammatory cytokine secretion. Interestingly, intravenous injections of tNano-S&C in orthotopic murine glioma LCPN models revealed elevated accumulation at the tumor site, in cervical lymph nodes (CLNs) and the spleen, and within antigen-presenting cells (APCs). This delivery system effectively enhanced the outcomes of chemo-immunotherapy with TMZ, leading to a marked extension of median survival time and complete regression in 25% mice. tNano-S&C treatment reduced M2 phenotype glioma associated macrophages and regulatory T cells, while increasing the recruitment of cytotoxic T lymphocytes. These findings suggest that this polymersome-enabled brain codelivery of STAT3 siRNA and immunoadjuvants provides an appealing strategy to effectively reshape the tumor immune microenvironment and boost the efficacy of chemo-immunotherapy of malignant glioma.
Collapse
Affiliation(s)
- Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Zhiwei Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Mingyu Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Yanyi Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Jingyi Wang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
4
|
Beylerli O, Gareev I, Musaev E, Roumiantsev S, Chekhonin V, Ahmad A, Chao Y, Yang G. New approaches to targeted drug therapy of intracranial tumors. Cell Death Discov 2025; 11:111. [PMID: 40113789 PMCID: PMC11926108 DOI: 10.1038/s41420-025-02358-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Intracranial tumors encompass a heterogeneous group of neoplasms, including gliomas, meningiomas, pituitary adenomas, schwannomas, craniopharyngiomas, ependymomas, medulloblastomas, and primary central nervous system lymphomas. These tumors present significant challenges due to their diverse molecular characteristics, critical locations, and the unique obstacles posed by the blood-brain barrier (BBB) and blood-tumor barrier (BTB), which limit the efficacy of systemic therapies. Recent advances in molecular biology and genomics have enabled the identification of specific molecular pathways and targets, paving the way for innovative precision therapies. This review examines the current state of targeted therapies for intracranial tumors, including receptor tyrosine kinase (RTK) inhibitors, PI3K/AKT/mTOR inhibitors, RAF/MEK/ERK pathway inhibitors, IDH mutation inhibitors, immune checkpoint inhibitors, and CAR-T cell therapies. Emphasis is placed on the role of the BBB and BTB in modulating drug delivery and therapeutic outcomes. Strategies to overcome these barriers, such as focused ultrasound, nanoparticle-based delivery systems, and convection-enhanced delivery, are also explored. Furthermore, the manuscript reviews clinical trial data, highlighting successes and limitations across different tumor types. It delves into emerging therapeutic approaches, including combination of regimens and personalized treatments based on molecular profiling. By synthesizing the latest research, this article aims to provide a comprehensive understanding of the advancements and ongoing challenges in the targeted treatment of intracranial tumors. The findings underscore the necessity for innovative delivery systems and more extensive clinical trials to optimize therapeutic strategies. This review aspires to inform future research and clinical practices, aiming to improve patient outcomes and quality of life in the management of these complex and life-threatening conditions.
Collapse
Affiliation(s)
- Ozal Beylerli
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, Russian Federation.
| | - Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, Russian Federation
| | - Elmar Musaev
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Sergey Roumiantsev
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Endocrinology Research Center, Moscow, Russian Federation
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Yuan Chao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Province Neuroscience Institute, Harbin, China.
| |
Collapse
|
5
|
Norollahi SE, Morovat S, Keymoradzadeh A, Hamzei A, Modaeinama M, Soleimanmanesh N, Soleimanmanesh Y, Najafizadeh A, Bakhshalipour E, Alijani B, Samadani AA. Transforming agents: The power of structural modifications in glioblastoma multiforme therapy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:41-56. [PMID: 39701498 DOI: 10.1016/j.pbiomolbio.2024.12.001] [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: 09/20/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024]
Abstract
Glioblastoma (GBM) is a very deadly type of brain tumor with a poor prognosis and a short survival rate. Recent advancements in understanding GBM's molecular and genetic characteristics have led to the development of various therapeutic and diagnostic strategies. Key elements such as microRNAs, lncRNAs, exosomes, angiogenesis, and chromatin modifications are highlighted, alongside significant epigenetic alterations that impact therapy and diagnosis. Despite these advancements, molecular classifications have not improved patient outcomes due to intratumoral diversity complicating targeted therapies. In this article, it is tried to emphasize the potential of investigating the epigenetic landscape of GBM, particularly identifying patients with diffuse hypermethylation at gene promoters associated with better outcomes. Integrating epigenetic and genetic data has enhanced the identification of glioma subtypes with high diagnostic precision. The reversibility of epigenetic changes offers promising therapeutic prospects, as recent insights into the "epigenetic orchestra" suggest new avenues for innovative treatment modalities for this challenging cancer. In this review article, we focus on the roles of translational elements and their alterations in the context of GBM diagnosis and therapy.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran; Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran
| | - Saman Morovat
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Arman Keymoradzadeh
- Department of Neurosurgery, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Hamzei
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Morteza Modaeinama
- Department of Neurosurgery, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | - Ali Najafizadeh
- School of Paramedicine Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Elahe Bakhshalipour
- School of Paramedicine Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Babak Alijani
- Department of Neurosurgery, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
6
|
Ren B, Liang J, Liu Y, Zhang Y, Ma X, Lei P, Gao J, Ma W. Proguanil inhibits proliferation and migration in glioblastoma development through targeting CSF1R receptor. Cell Signal 2025; 127:111550. [PMID: 39662608 DOI: 10.1016/j.cellsig.2024.111550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/14/2024] [Accepted: 12/01/2024] [Indexed: 12/13/2024]
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant tumor of the central nervous system, characterized by high morbidity and invasive potential, necessitating urgent development of novel therapeutic strategies. Studies have shown that colony stimulating factor-1 receptor (CSF1R) is abnormally expressed in a variety of solid tumors, which is closely related to the development of tumor cells. In this study, the CSF1R/cell membrane Chromatographic model was successfully constructed, and was used to screen active compounds targeting CSF1R from more than 60 compounds. Among these, Proguanil exhibited the strongest affinity with retention time of 69 min, and a KD value of (6.73 ± 0.05) × 10-7 M. Proguanil effectively inhibited the growth of U87MG cells in vitro and in vivo by inducing G0/G1 phase cell cycle arrest and suppressing U87MG cells migration. More importantly, we found that Proguanil's inhibitory effect on U87MG cell growth and migration was positively correlated with CSF1R expression, and this effect diminished following CSF1R knockdown and Proguanil demonstrated synergistic effects with CSF1R-targeting positive drugs (BLZ945 and GW2580). Furthermore, Proguanil was found to inhibit CSF1R phosphorylation along with downstream signaling pathways such as PTEN/AKT/mTOR and Ras/MEK1/2/ERK1/2, thereby regulating cell cycle-related molecules (p21, CDK4, and CyclinD1) and cell migration-related molecule MMP3. Meanwhile, Proguanil targeted CSF1R to inhibit M2-type polarization of tumor-associated macrophages (TAMs) and their proliferation, thus altering the tumor microenvironment while indirectly suppressing the proliferation and migration of U87MG cells. Taken together, these findings suggest that Proguanil may serve as a promising CSF1R antagonist for GBM treatment.
Collapse
Affiliation(s)
- Bingxi Ren
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Jinna Liang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong university, Xi'an, 710061, China
| | - Yanhong Liu
- Department of Pharmacy, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, 710018, China
| | - Yuxiu Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Xiaoyu Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Panpan Lei
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Jiapan Gao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Weina Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| |
Collapse
|
7
|
Liu S, Li H, Xi S, Zhang Y, Sun T. Advancing CNS Therapeutics: Enhancing Neurological Disorders with Nanoparticle-Based Gene and Enzyme Replacement Therapies. Int J Nanomedicine 2025; 20:1443-1490. [PMID: 39925682 PMCID: PMC11806685 DOI: 10.2147/ijn.s457393] [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: 12/31/2023] [Accepted: 12/12/2024] [Indexed: 02/11/2025] Open
Abstract
Given the complexity of the central nervous system (CNS) and the diversity of neurological conditions, the increasing prevalence of neurological disorders poses a significant challenge to modern medicine. These disorders, ranging from neurodegenerative diseases to psychiatric conditions, not only impact individuals but also place a substantial burden on healthcare systems and society. A major obstacle in treating these conditions is the blood-brain barrier (BBB), which restricts the passage of therapeutic agents to the brain. Nanotechnology, particularly the use of nanoparticles (NPs), offers a promising solution to this challenge. NPs possess unique properties such as small size, large surface area, and modifiable surface characteristics, enabling them to cross the BBB and deliver drugs directly to the affected brain regions. This review focuses on the application of NPs in gene therapy and enzyme replacement therapy (ERT) for neurological disorders. Gene therapy involves altering or manipulating gene expression and can be enhanced by NPs designed to carry various genetic materials. Similarly, NPs can improve the efficacy of ERT for lysosomal storage disorders (LSDs) by facilitating enzyme delivery to the brain, overcoming issues like immunogenicity and instability. Taken together, this review explores the potential of NPs in revolutionizing treatment options for neurological disorders, highlighting their advantages and the future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Haisong Li
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Shiwen Xi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- International Center of Future Science, Jilin University, Changchun, People’s Republic of China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
8
|
Kelly CL, Wydrzynska M, Phelan MM, Osharovich S, Delikatny EJ, Sée V, Poptani H. Hypoxia Dependent Inhibition of Glioblastoma Cell Proliferation, Invasion, and Metabolism by the Choline-Kinase Inhibitor JAS239. Metabolites 2025; 15:76. [PMID: 39997701 PMCID: PMC11857610 DOI: 10.3390/metabo15020076] [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: 12/18/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Elevated choline kinase alpha (ChoK) levels are observed in most solid tumors, including glioblastomas (GBM), and ChoK inhibitors have demonstrated limited efficacy in GBM models. Given that hypoxia is associated with resistance to GBM therapy, we hypothesized that tumor hypoxia could be responsible for the limited response. Therefore, we evaluated the effects of hypoxia on the function of JAS239, a potent ChoK inhibitor in four GBM cell lines. Methods: Rodent (F98 and 9L) and human (U-87 MG and U-251 MG) GBM cell lines were subjected to 72 h of hypoxic conditioning and treated with JAS239 for 24 h. NMR metabolomic measurements and analyses were performed to evaluate the signaling pathways involved. In addition, cell proliferation, cell cycle progression, and cell invasion parameters were measured in 2D cell monolayers as well as in 3D cell spheroids, with or without JAS239 treatment, in normoxic or hypoxic cells to assess the effect of hypoxia on JAS239 function. Results: Hypoxia and JAS239 treatment led to significant changes in the cellular metabolic pathways, specifically the phospholipid and glycolytic pathways, associated with a reduction in cell proliferation via induced cell cycle arrest. Interestingly, JAS239 also impaired GBM invasion. However, effects from JAS239 were variable depending on the cell line, reflecting the inherent heterogeneity of GBMs. Conclusions: Our findings indicate that JAS239 and hypoxia can deregulate cellular metabolism, inhibit cell proliferation, and alter cell invasion. These results may be useful for designing new therapeutic strategies based on ChoK inhibition, which can act on multiple pro-tumorigenic features.
Collapse
Affiliation(s)
- Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK;
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool L69 7BE, UK;
| | - Martyna Wydrzynska
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool L69 7BE, UK;
| | - Marie M. Phelan
- High Field NMR Facility, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool L69 7ZX, UK;
| | - Sofya Osharovich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (E.J.D.)
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (E.J.D.)
| | - Violaine Sée
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool L69 7BE, UK;
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK;
| |
Collapse
|
9
|
Horta M, Soares P, Leite Pereira C, Lima RT. Emerging Approaches in Glioblastoma Treatment: Modulating the Extracellular Matrix Through Nanotechnology. Pharmaceutics 2025; 17:142. [PMID: 40006509 PMCID: PMC11859630 DOI: 10.3390/pharmaceutics17020142] [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: 12/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Glioblastoma's (GB) complex tumor microenvironment (TME) promotes its progression and resistance to therapy. A critical component of TME is the extracellular matrix (ECM), which plays a pivotal role in promoting the tumor's invasive behavior and aggressiveness. Nanotechnology holds significant promise for GB treatment, with the potential to address challenges posed by both the blood-brain barrier and the GB ECM. By enabling targeted delivery of therapeutic and diagnostic agents, nanotechnology offers the prospect of improving treatment efficacy and diagnostic accuracy at the tumor site. This review provides a comprehensive exploration of GB, including its epidemiology, classification, and current treatment strategies, alongside the intricacies of its TME. It highlights nanotechnology-based strategies, focusing on nanoparticle formulations such as liposomes, polymeric nanoparticles, and gold nanoparticles, which have shown promise in GB therapy. Furthermore, it explores how different emerging nanotechnology strategies modulate the ECM to overcome the challenges posed by its high density, which restricts drug distribution within GB tumors. By emphasizing the intersection of nanotechnology and GB ECM, this review underscores an innovative approach to advancing GB treatment. It addresses the limitations of current therapies, identifies new research avenues, and emphasizes the potential of nanotechnology to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Horta
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paula Soares
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Catarina Leite Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Raquel T. Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
10
|
Yalamandala BN, Huynh TMH, Lien HW, Pan WC, Iao HM, Moorthy T, Chang YH, Hu SH. Advancing brain immunotherapy through functional nanomaterials. Drug Deliv Transl Res 2025:10.1007/s13346-024-01778-5. [PMID: 39789307 DOI: 10.1007/s13346-024-01778-5] [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] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
Glioblastoma (GBM), a highly aggressive brain tumor, poses significant treatment challenges due to its highly immunosuppressive microenvironment and the brain immune privilege. Immunotherapy activating the immune system and T lymphocyte infiltration holds great promise against GBM. However, the brain's low immunogenicity and the difficulty of crossing the blood-brain barrier (BBB) hinder therapeutic efficacy. Recent advancements in immune-actuated particles for targeted drug delivery have shown the potential to overcome these obstacles. These particles interact with the BBB by rapidly and reversibly disrupting its structure, thereby significantly enhancing targeting and penetrating delivery. The BBB targeting also minimizes potential long-term damage. At GBM, the particles demonstrated effective chemotherapy, chemodynamic therapy, photothermal therapy (PTT), photodynamic therapy (PDT), radiotherapy, or magnetotherapy, facilitating tumor disruption and promoting antigen release. Additionally, components of the delivery system retained autologous tumor-associated antigens and presented them to dendritic cells (DCs), ensuring prolonged immune activation. This review explores the immunosuppressive mechanisms of GBM, existing therapeutic strategies, and the role of nanomaterials in enhancing immunotherapy. We also discuss innovative particle-based approaches designed to traverse the BBB by mimicking innate immune functions to improve treatment outcomes for brain tumors.
Collapse
Affiliation(s)
- Bhanu Nirosha Yalamandala
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Thi My Hue Huynh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Hui-Wen Lien
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Wan-Chi Pan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Hoi Man Iao
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Thrinayan Moorthy
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Yun-Hsuan Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan.
| |
Collapse
|
11
|
Modestov A, Zolotovskaia M, Suntsova M, Zakharova G, Seryakov A, Jovcevska I, Mlakar J, Poddubskaya E, Moisseev A, Vykhodtsev G, Roumiantsev S, Sorokin M, Tkachev V, Simonov A, Buzdin A. Bioinformatic and clinical experimental assay uncovers resistance and susceptibility mechanisms of human glioblastomas to temozolomide and identifies new combined and individual survival biomarkers outperforming MGMT promoter methylation. Ther Adv Med Oncol 2024; 16:17588359241292269. [PMID: 39525666 PMCID: PMC11544758 DOI: 10.1177/17588359241292269] [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: 07/01/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive and lethal central nervous system (CNS) tumor. The treatment strategy is mainly surgery and/or radiation therapy, both combined with adjuvant temozolomide (TMZ) chemotherapy. Historically, methylation of MGMT gene promoter is used as the major biomarker predicting individual tumor response to TMZ. Objectives This research aimed to analyze genes and molecular pathways of DNA repair as biomarkers for sensitivity to TMZ treatment in GBM using updated The Cancer Genome Atlas (TCGA) data and validate the results on experimental datasets. Methods Survival analysis of GBM patients under TMZ therapy and hazard ratio (HR) calculation were used to assess all putative biomarkers on World Health Organization CNS5 reclassified TCGA project collection of molecular profiles and experimental multicenter GBM patient cohort. Pathway activation levels were calculated for 38 DNA repair pathways. TMZ sensitivity pathway was reconstructed using a human interactome model built using pairwise interactions extracted from 51,672 human molecular pathways. Results We found that expression/activation levels of seven and six emerging gene/pathway biomarkers served as high-quality positive (HR < 0.61) and negative (HR > 1.63), respectively, patient survival biomarkers performing better than MGMT methylation. Positive survival biomarkers were enriched in the processes of ATM-dependent checkpoint activation and cell cycle arrest whereas negative-in excision DNA repair. We also built and characterized gene pathways which were informative for GBM patient survival following TMZ administration (HR 0.18-0.44, p < 0.0009; area under the curve 0.68-0.9). Conclusion In this study, a comprehensive analysis of the expression of 361 DNA repair genes and activation levels of 38 DNA repair pathways revealed 13 potential survival biomarkers with increased prognostic potential compared to MGMT methylation. We algorithmically reconstructed the TMZ sensitivity pathway with strong predictive capacity in GBM.
Collapse
Affiliation(s)
| | - Marianna Zolotovskaia
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Endocrinology Research Center, Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia
| | - Maria Suntsova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Endocrinology Research Center, Moscow, Russia
| | - Galina Zakharova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Ivana Jovcevska
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Mlakar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Aleksey Moisseev
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Endocrinology Research Center, Moscow, Russia
| | | | | | | | | | | | - Anton Buzdin
- I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Endocrinology Research Center, Dmitriya Ulyanova Str. 11, Moscow 117036, Russia
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow, Russia
- Oncobox LLC, Moscow 119991, Russia
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| |
Collapse
|
12
|
Mu Y, Zhang Z, Zhou H, Jin M, Ma L, Liu B, Ma C, Hu X, Zhang Y, Wang DA. A biomimetic targeted nanosystem delivering synergistic inhibitors for glioblastoma immune microenvironment reprogramming and treatment. Mater Today Bio 2024; 28:101222. [PMID: 39296357 PMCID: PMC11407969 DOI: 10.1016/j.mtbio.2024.101222] [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: 06/03/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/21/2024] Open
Abstract
Efficient drug delivery across the blood-brain barrier is imperative for treating glioblastoma (GBM). This study utilized the GBM cell membrane to construct a biomimetic targeted nanosystem (GMNPs@AMD/RAPA) that hierarchically releases the CXCR4 antagonist AMD3100 and the mTOR pathway inhibitor rapamycin (RAPA) for reprogramming the tumor immune microenvironment and suppressing the progression of GBM. By initially inhibiting the CXCL12/CXCR4 axis, the tumor microenvironment (TME) was reprogrammed to enhance the infiltration of cytotoxic T lymphocytes (CTLs) into the TME while suppressing tumor cell survival, proliferation, and angiogenesis. Subsequently, through further cellular uptake and degradation of the nanoparticles, the mTOR pathway inhibitor RAPA was released, further suppressing the tumor progression. This study successfully combined chemotherapy and immunotherapy, achieving effective synergistic therapeutic effects, and suppressing the progression of GBM.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Min Jin
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Cheng Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Xu Hu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Engineering of China, Chengdu, Sichuan, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
13
|
Tydings CW, Singh B, Smith AW, Ledwitch KV, Brown BP, Lovly CM, Walker AS, Meiler J. Analysis of EGFR binding hotspots for design of new EGFR inhibitory biologics. Protein Sci 2024; 33:e5141. [PMID: 39275996 PMCID: PMC11400634 DOI: 10.1002/pro.5141] [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: 02/29/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/16/2024]
Abstract
The epidermal growth factor (EGF) receptor (EGFR) is activated by the binding of one of seven EGF-like ligands to its ectodomain. Ligand binding results in EGFR dimerization and stabilization of the active receptor conformation subsequently leading to activation of downstream signaling. Aberrant activation of EGFR contributes to cancer progression through EGFR overexpression/amplification, modulation of its positive and negative regulators, and/or activating mutations within EGFR. EGFR targeted therapeutic antibodies prevent dimerization and interaction with endogenous ligands by binding the ectodomain of EGFR. However, these antibodies have had limited success in the clinic, partially due to EGFR ectodomain resistance mutations, and are only applicable to a subset of patients with EGFR-driven cancers. These limitations suggest that alternative EGFR targeted biologics need to be explored for EGFR-driven cancer therapy. To this end, we analyze the EGFR interfaces of known inhibitory biologics with determined structures in the context of endogenous ligands, using the Rosetta macromolecular modeling software to highlight the most important interactions on a per-residue basis. We use this analysis to identify the structural determinants of EGFR targeted biologics. We suggest that commonly observed binding motifs serve as the basis for rational design of new EGFR targeted biologics, such as peptides, antibodies, and nanobodies.
Collapse
Affiliation(s)
- Claiborne W. Tydings
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Bhuminder Singh
- Department of Medicine – Division of Gastroenterology, Hepatology, and NutritionVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Adam W. Smith
- Department of Chemistry and BiochemistryTexas Tech UniversityLubbockTexasUSA
| | - Kaitlyn V. Ledwitch
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Benjamin P. Brown
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Christine M. Lovly
- Department of Medicine – Division of Hematology and OncologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Allison S. Walker
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Department of Biological SciencesVanderbilt UniversityNashvilleTennesseeUSA
| | - Jens Meiler
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
- Institute for Drug DiscoveryLeipzig University Medical SchoolLeipzigSACGermany
| |
Collapse
|
14
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
15
|
Saqib M, Zahoor A, Rahib A, Shamim A, Mumtaz H. Clinical and translational advances in primary brain tumor therapy with a focus on glioblastoma-A comprehensive review of the literature. World Neurosurg X 2024; 24:100399. [PMID: 39386927 PMCID: PMC11462364 DOI: 10.1016/j.wnsx.2024.100399] [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: 06/08/2023] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
This comprehensive review paper examines the most updated state of research on glioblastoma, an aggressive brain tumor with limited treatment options. By analyzing 76 recent studies, from translational and basic sciences, to clinical trials, we highlight various aspects of glioblastoma and shed light on potential therapeutic strategies. The interplay between tumor cells, neural progenitor cells, and the tumor microenvironment is explored. Targeting the PI3K-Akt-mTOR pathway through extracellular-vesicle (EV)-mediated signaling emerges as a potential therapeutic strategy. Personalized modeling approaches utilizing patient-specific MRI data offer promise for optimizing treatment strategies. The response of glioblastoma stem cells (GSCs) to different treatment modalities is examined, emphasizing the need to inhibit the transformation of proneural (PN) GSCs into resistant mesenchymal (MES) GSCs. Metabolic therapy and combination therapies show potential in reversing treatment resistance and inhibiting both PN and MES GSCs. Immunotherapy, targeted approaches, and molecular dynamics in gliomas are discussed, providing insights into early-stage diagnosis and treatment. Additionally, the potential use of Zika virus as an oncolytic agent is explored. Analysis of phase 0 to 3 clinical trials reveal promising outcomes for various experimental treatments, highlighting the importance of combination therapies, predictive signatures, and patient selection strategies. Specific compounds demonstrate potential therapeutic benefits and tolerability. Phase 3 trials indicate the efficacy of DCVax-L in improving survival rates and depatux-m in prolonging progression-free survival. These findings emphasize the importance of personalized treatment approaches and continued exploration of targeted therapies, immunotherapies, and tumor biology understanding in shaping the future of glioblastoma treatment.
Collapse
Affiliation(s)
| | | | - Ahmed Rahib
- Nowshera Medical College, Nowshera, Pakistan
| | - Amna Shamim
- King Edward Medical University, Lahore, Pakistan
| | | |
Collapse
|
16
|
Jiang Q, Yang X, Deng T, Yan J, Guo F, Mo L, An S, Huang Q. Comprehensive machine learning-based integration develops a novel prognostic model for glioblastoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200838. [PMID: 39072291 PMCID: PMC11278295 DOI: 10.1016/j.omton.2024.200838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/09/2024] [Accepted: 06/14/2024] [Indexed: 07/30/2024]
Abstract
In this study, we developed a new prognostic model for glioblastoma (GBM) based on an integrated machine learning algorithm. We used univariate Cox regression analysis to identify prognostic genes by combining six GBM cohorts. Based on the prognostic genes, 10 machine learning algorithms were integrated into 117 algorithm combinations, and the artificial intelligence prognostic signature (AIPS) with the greatest average C-index was chosen. The AIPS was compared with 10 previously published models by univariate Cox analysis and the C-index. We compared the differences in prognosis, tumor immune microenvironment (TIME), and immunotherapy sensitivity between the high and low AIPS score groups. The AIPS based on the random survival forest algorithm with the highest average C-index (0.868) was selected. Compared with the previous 10 prognostic models, our AIPS has the highest C-index. The AIPS was closely linked to the clinical features of GBM. We discovered that patients in the low score group had improved prognoses, a more active TIME, and were more sensitive to immunotherapy. Finally, we verified the expression of several key genes by western blotting and immunohistochemistry. We identified an ideal prognostic signature for GBM, which might provide new insights into stratified treatment approaches for GBM patients.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xiawei Yang
- Transplant Medical Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Teng Deng
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Sanqi An
- Biosafety Level-3 Laboratory, Life Sciences Institute & Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Qianrong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| |
Collapse
|
17
|
Lashkarboloki M, Jahanbakhshi A, Mowla SJ, Bjeije H, Soltani BM. Oncogenic roles of long non-coding RNAs in essential glioblastoma signaling pathways. J Neurogenet 2024; 38:62-78. [PMID: 39169886 DOI: 10.1080/01677063.2024.2390403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and diffuse type of glioma with the lowest survival rate in patients. The recent failure of multiple treatments suggests that targeting several targets at once may be a different strategy to overcome GBM carcinogenesis. Normal function of oncogenes and tumor suppressor genes need for the preservation of regular cellular processes, so any defects in these genes' activity, operate the corresponding signaling pathways, which initiate carcinogenic processes. Long non-coding RNAs (lncRNAs) that can be found in the cytoplasm or nucleus of the cells, control the transcription and translation of genes. LncRNAs perform a variety of functions, including epigenetic alteration, protein modification and stability, transcriptional regulation, and competition for miRNA that regulate mRNA translation through sponging miRNAs. Identification of various oncogenic lncRNAs and their multiple roles in brain cancers making them potential candidates for use as glioma diagnostic, prognostic, and therapeutic targets in the future. This study highlighted multiple oncogenic lncRNAs and classified them into different signaling pathways based on the regulated target genes in glioblastoma.
Collapse
Affiliation(s)
- Mina Lashkarboloki
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amin Jahanbakhshi
- Skull Base Research Center, Rasool Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Javad Mowla
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Bjeije
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bahram M Soltani
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
18
|
Zelisko N, Lesyk R, Stoika R. Structure, unique biological properties, and mechanisms of action of transforming growth factor β. Bioorg Chem 2024; 150:107611. [PMID: 38964148 DOI: 10.1016/j.bioorg.2024.107611] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Transforming growth factor β (TGF-β) is a ubiquitous molecule that is extremely conserved structurally and plays a systemic role in human organism. TGF-β is a homodimeric molecule consisting of two subunits joined through a disulphide bond. In mammals, three genes code for TGF-β1, TGF-β2, and TGF-β3 isoforms of this cytokine with a dominating expression of TGF-β1. Virtually, all normal cells contain TGF-β and its specific receptors. Considering the exceptional role of fine balance played by the TGF-β in anumber of physiological and pathological processes in human body, this cytokine may be proposed for use in medicine as an immunosuppressant in transplantology, wound healing and bone repair. TGFb itself is an important target in oncology. Strategies for blocking members of TGF-β signaling pathway as therapeutic targets have been considered. In this review, signalling mechanisms of TGF-β1 action are addressed, and their role in physiology and pathology with main focus on carcinogenesis are described.
Collapse
Affiliation(s)
- Nataliya Zelisko
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine
| |
Collapse
|
19
|
Xu S, Ma Y, Jiang X, Wang Q, Ma W. CD39 transforming cancer therapy by modulating tumor microenvironment. Cancer Lett 2024; 597:217072. [PMID: 38885807 DOI: 10.1016/j.canlet.2024.217072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
CD39 is a pivotal enzyme in cancer, regulating immune response and tumor progression via extracellular ATP and adenosine in the tumor microenvironment (TME). Beyond its established immunoregulatory function, CD39 influences cancer cell angiogenesis and metabolism, opening new frontiers for therapeutic interventions. Current research faces gaps in understanding CD39's full impact across cancer types, with ongoing debates about its potential beyond modulating immune evasion. This review distills CD39's multifaceted roles, examining its dual actions and implications for cancer prognosis and treatment. We analyze the latest therapeutic strategies, highlighting the need for an integrated approach that combines molecular insights with TME dynamics to innovate cancer care. This synthesis underscores CD39's integral role, charting a course for precision oncology that seeks to unravel controversies and harness CD39's therapeutic promise for improved cancer outcomes.
Collapse
Affiliation(s)
- Suling Xu
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang, 315020, China.
| | - Yuhan Ma
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang, 315020, China.
| | - Xinyu Jiang
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang, 315020, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Thiruvengadam M. Radioresistance in brain tumors: Strategies for improved radiotherapy outcomes. BRAIN & SPINE 2024; 4:102912. [PMID: 39247725 PMCID: PMC11377130 DOI: 10.1016/j.bas.2024.102912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024]
Affiliation(s)
- Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Science, Konkuk University, Seoul, 05029, Republic of Korea
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, 602 105, Tamil Nadu, India
| |
Collapse
|
21
|
Hamza MN, Koziel S, Pietrenko-Dabrowska A. Design and experimental validation of a metamaterial-based sensor for microwave imaging in breast, lung, and brain cancer detection. Sci Rep 2024; 14:16177. [PMID: 39003304 PMCID: PMC11246499 DOI: 10.1038/s41598-024-67103-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
This study proposes an innovative geometry of a microstrip sensor for high-resolution microwave imaging (MWI). The main intended application of the sensor is early detection of breast, lung, and brain cancer. The proposed design consists of a microstrip patch antenna fed by a coplanar waveguide with a metamaterial (MTM) layer-based lens implemented on the back side, and an artificial magnetic conductor (AMC) realized on as a separate layer. The analysis of the AMC's permeability and permittivity demonstrate that the structure exhibits negative epsilon (ENG) qualities near the antenna resonance point. In addition, reflectivity, transmittance, and absorption are also studied. The sensor prototype has been manufactures using the FR4 laminate. Excellent electrical and field characteristics of the structure are confirmed through experimental validation. At the resonance frequency of 4.56 GHz, the realized gain reaches 8.5 dBi, with 3.8 dBi gain enhancement contributed by the AMC. The suitability of the presented sensor for detecting brain tumors, lung cancer, and breast cancer has been corroborated through extensive simulation-based experiments performed using the MWI system model, which employs four copies of the proposed sensor, as well as the breast, lung, and brain phantoms. As demonstrated, the directional radiation pattern and enhanced gain of the sensor enable precise tumor size discrimination. The proposed sensor offers competitive performance in comparison the state-of-the-art sensors described in the recent literature, especially with respect to as gain, pattern directivity, and impedance matching, all being critical for MWI.
Collapse
Affiliation(s)
- Musa N Hamza
- Department of Physics, College of Science, University of Raparin, Sulaymaniyah, 46012, Iraq.
| | - Slawomir Koziel
- Engineering Optimization & Modeling Center, Reykjavik University, 102, Reykjavik, Iceland
- Faculty of Electronics, Telecommunications and Informatics, Gdansk University of Technology, 80-233, Gdansk, Poland
| | - Anna Pietrenko-Dabrowska
- Faculty of Electronics, Telecommunications and Informatics, Gdansk University of Technology, 80-233, Gdansk, Poland
| |
Collapse
|
22
|
Zhu H, Wang YF, Wang ZG, Pang DW, Liu SL. Regulation of Protein Conformation Enables Cell-Selective Targeting of Virus-Mimicking Nanoparticles for siRNA Therapy of Glioblastoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401640. [PMID: 38710154 DOI: 10.1002/adma.202401640] [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: 01/31/2024] [Revised: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Orthotopic glioblastoma (GBM) has an aggressive growth pattern and complex pathogenesis, becoming one of the most common and deadly tumors of the central nervous system (CNS). The emergence of RNA therapies offers promise for the treatment of GBM. However, the efficient and precise delivery of RNA drugs to specific tumor cells in the brain with high cellular heterogeneity remains ongoing. Here, a strategy is proposed to regulate protein conformation through lipid nanoenvironments to custom-design virus-mimicking nanoparticles (VMNs) with excellent selective cell targeting capabilities, leading to efficient and precise delivery of small interfering RNA for effective treatment of GBM. The optimized VMNs not only retain the ability to cross the blood-brain barrier and release the RNA by lysosomal escape like natural viruses but also ensure precise enrichment in the GBM area. This study lays the conceptual foundation for the custom design of VMNs with superior cell-selective targeting capabilities and opens up the possibility of RNA therapies for the efficient treatment of GBM and CNS tumors.
Collapse
Affiliation(s)
- Han Zhu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, P. R. China
| | - Yi-Fan Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, P. R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, P. R. China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
23
|
Fan Q, Kuang L, Wang B, Yin Y, Dong Z, Tian N, Wang J, Yin T, Wang Y. Multiple Synergistic Effects of the Microglia Membrane-Bionic Nanoplatform on Mediate Tumor Microenvironment Remodeling to Amplify Glioblastoma Immunotherapy. ACS NANO 2024; 18:14469-14486. [PMID: 38770948 DOI: 10.1021/acsnano.4c01253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Glioblastoma (GBM) is a lethal brain tumor with high levels of malignancy. Most chemotherapy agents show serious systemic cytotoxicity and restricted delivery effectiveness due to the impediments of the blood-brain barrier (BBB). Immunotherapy has developed great potential for aggressive tumor treatments. Disappointingly, its efficacy against GBM is hindered by the immunosuppressive tumor microenvironment (TME) and BBB. Herein, a multiple synergistic immunotherapeutic strategy against GBM was developed based on the nanomaterial-biology interaction. We have demonstrated that this BM@MnP-BSA-aPD-1 can transverse the BBB and target the TME, resulting in amplified synergetic effects of metalloimmunotherapy and photothermal immunotherapy (PTT). The journey of this nanoformulation within the TME contributed to the activation of the stimulator of the interferon gene pathway, the initiation of the immunogenic cell death effect, and the inhibition of the programmed cell death-1/programmed cell death ligand 1 (PD-1/PD-L1) signaling axis. This nanomedicine revitalizes the immunosuppressive TME and evokes the cascade effect of antitumor immunity. Therefore, the combination of BM@MnP-BSA-aPD-1 and PTT without chemotherapeutics presents favorable benefits in anti-GBM immunotherapy and exhibits immense potential for clinical translational applications.
Collapse
Affiliation(s)
- Qin Fan
- School of Medicine, Chongqing University, Chongqing 400044, China
| | - Lei Kuang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Bingyi Wang
- School of Medicine, Chongqing University, Chongqing 400044, China
| | - Ying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zhufeng Dong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Nixin Tian
- School of Medicine, Chongqing University, Chongqing 400044, China
| | - Jiaojiao Wang
- School of Medicine, Chongqing University, Chongqing 400044, China
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yazhou Wang
- School of Medicine, Chongqing University, Chongqing 400044, China
| |
Collapse
|
24
|
Mokhtarpour K, Akbarzadehmoallemkolaei M, Rezaei N. A viral attack on brain tumors: the potential of oncolytic virus therapy. J Neurovirol 2024; 30:229-250. [PMID: 38806994 DOI: 10.1007/s13365-024-01209-8] [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: 12/01/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/30/2024]
Abstract
Managing malignant brain tumors remains a significant therapeutic hurdle that necessitates further research to comprehend their treatment potential fully. Oncolytic viruses (OVs) offer many opportunities for predicting and combating tumors through several mechanisms, with both preclinical and clinical studies demonstrating potential. OV therapy has emerged as a potent and effective method with a dual mechanism. Developing innovative and effective strategies for virus transduction, coupled with immune checkpoint inhibitors or chemotherapy drugs, strengthens this new technique. Furthermore, the discovery and creation of new OVs that can seamlessly integrate gene therapy strategies, such as cytotoxic, anti-angiogenic, and immunostimulatory, are promising advancements. This review presents an overview of the latest advancements in OVs transduction for brain cancer, focusing on the safety and effectiveness of G207, G47Δ, M032, rQNestin34.5v.2, C134, DNX-2401, Ad-TD-nsIL12, NSC-CRAd-S-p7, TG6002, and PVSRIPO. These are evaluated in both preclinical and clinical models of various brain tumors.
Collapse
Affiliation(s)
- Kasra Mokhtarpour
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
| | - Milad Akbarzadehmoallemkolaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, 1419733151, Iran
| | - Nima Rezaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, 1419733151, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417653761, Iran.
| |
Collapse
|
25
|
Al-Otaibi S, Rehman A, Raza A, Alyami J, Saba T. CVG-Net: novel transfer learning based deep features for diagnosis of brain tumors using MRI scans. PeerJ Comput Sci 2024; 10:e2008. [PMID: 38855235 PMCID: PMC11157570 DOI: 10.7717/peerj-cs.2008] [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: 11/07/2023] [Accepted: 04/01/2024] [Indexed: 06/11/2024]
Abstract
Brain tumors present a significant medical challenge, demanding accurate and timely diagnosis for effective treatment planning. These tumors disrupt normal brain functions in various ways, giving rise to a broad spectrum of physical, cognitive, and emotional challenges. The daily increase in mortality rates attributed to brain tumors underscores the urgency of this issue. In recent years, advanced medical imaging techniques, particularly magnetic resonance imaging (MRI), have emerged as indispensable tools for diagnosing brain tumors. Brain MRI scans provide high-resolution, non-invasive visualization of brain structures, facilitating the precise detection of abnormalities such as tumors. This study aims to propose an effective neural network approach for the timely diagnosis of brain tumors. Our experiments utilized a multi-class MRI image dataset comprising 21,672 images related to glioma tumors, meningioma tumors, and pituitary tumors. We introduced a novel neural network-based feature engineering approach, combining 2D convolutional neural network (2DCNN) and VGG16. The resulting 2DCNN-VGG16 network (CVG-Net) extracted spatial features from MRI images using 2DCNN and VGG16 without human intervention. The newly created hybrid feature set is then input into machine learning models to diagnose brain tumors. We have balanced the multi-class MRI image features data using the Synthetic Minority Over-sampling Technique (SMOTE) approach. Extensive research experiments demonstrate that utilizing the proposed CVG-Net, the k-neighbors classifier outperformed state-of-the-art studies with a k-fold accuracy performance score of 0.96. We also applied hyperparameter tuning to enhance performance for multi-class brain tumor diagnosis. Our novel proposed approach has the potential to revolutionize early brain tumor diagnosis, providing medical professionals with a cost-effective and timely diagnostic mechanism.
Collapse
Affiliation(s)
- Shaha Al-Otaibi
- Department of Information Systems, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amjad Rehman
- Artificial Intelligence & Data Analytics Lab CCIS, Prince Sultan University, Riyadh, Saudi Arabia
| | - Ali Raza
- Institute of Computer Science, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, Pakistan
| | - Jaber Alyami
- Department of Diagnostic Radiology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tanzila Saba
- Artificial Intelligence & Data Analytics Lab CCIS, Prince Sultan University, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Mastronuzzi A, Quaglietta L, Schiavello E, Carai A. Editorial: 2021 WHO classification of pediatric brain tumors: a final wedding between morphology and molecular biology? Front Mol Neurosci 2024; 17:1423298. [PMID: 38779356 PMCID: PMC11109396 DOI: 10.3389/fnmol.2024.1423298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Angela Mastronuzzi
- Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, Instituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Lucia Quaglietta
- Pediatric Neuro-Oncology, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Elisabetta Schiavello
- Pediatric Oncology Unit, Fondazione Instituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Carai
- Neurosciences, Neurosurgery Unit, Bambino Gesù Children's Hospital, Instituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
27
|
Ahirwar K, Kumar A, Srivastava N, Saraf SA, Shukla R. Harnessing the potential of nanoengineered siRNAs carriers for target responsive glioma therapy: Recent progress and future opportunities. Int J Biol Macromol 2024; 266:131048. [PMID: 38522697 DOI: 10.1016/j.ijbiomac.2024.131048] [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: 11/07/2023] [Revised: 01/19/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
Past scientific testimonials in the field of glioma research, the deadliest tumor among all brain cancer types with the life span of 10-15 months after diagnosis is considered as glioblastoma multiforme (GBM). Even though the availability of treatment options such as chemotherapy, radiotherapy, and surgery, are unable to completely cure GBM due to tumor microenvironment complexity, intrinsic cellular signalling, and genetic mutations which are involved in chemoresistance. The blood-brain barrier is accountable for restricting drugs entry at the tumor location and related biological challenges like endocytic degradation, short systemic circulation, and insufficient cellular penetration lead to tumor aggression and progression. The above stated challenges can be better mitigated by small interfering RNAs (siRNA) by knockdown genes responsible for tumor progression and resistance. However, siRNA encounters with challenges like inefficient cellular transfection, short circulation time, endogenous degradation, and off-target effects. The novel functionalized nanocarrier approach in conjunction with biological and chemical modification offers an intriguing potential to address challenges associated with the naked siRNA and efficiently silence STAT3, coffilin-1, EGFR, VEGF, SMO, MGMT, HAO-1, GPX-4, TfR, LDLR and galectin-1 genes in GBM tumor. This review highlights the nanoengineered siRNA carriers, their recent advancements, future perspectives, and strategies to overcome the systemic siRNA delivery challenges for glioma treatment.
Collapse
Affiliation(s)
- Kailash Ahirwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P. 226002, India
| | - Ankit Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P. 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P. 226002, India
| | - Shubhini A Saraf
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P. 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P. 226002, India.
| |
Collapse
|
28
|
Tomoszková S, Škarda J, Lipina R. Potential Diagnostic and Clinical Significance of Selected Genetic Alterations in Glioblastoma. Int J Mol Sci 2024; 25:4438. [PMID: 38674026 PMCID: PMC11050250 DOI: 10.3390/ijms25084438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma is currently considered the most common and, unfortunately, also the most aggressive primary brain tumor, with the highest morbidity and mortality rates. The average survival of patients diagnosed with glioblastoma is 14 months, and only 2% of patients survive 3 years after surgery. Based on our clinical experience and knowledge from extensive clinical studies, survival is mainly related to the molecular biological properties of glioblastoma, which are of interest to the general medical community. Our study examined a total of 71 retrospective studies published from 2016 through 2022 and available on PubMed that deal with mutations of selected genes in the pathophysiology of GBM. In conclusion, we can find other mutations within a given gene group that have different effects on the prognosis and quality of survival of a patient with glioblastoma. These mutations, together with the associated mutations of other genes, as well as intratumoral heterogeneity itself, offer enormous potential for further clinical research and possible application in therapeutic practice.
Collapse
Affiliation(s)
- Silvia Tomoszková
- Neurosurgery Clinic, University Hospital Ostrava, 17. listopadu 1790/5, 708 00 Ostrava, Czech Republic;
- Medical Faculty, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic;
| | - Jozef Škarda
- Medical Faculty, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic;
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, 17. listopadu 1790/5, 708 00 Ostrava, Czech Republic
| | - Radim Lipina
- Neurosurgery Clinic, University Hospital Ostrava, 17. listopadu 1790/5, 708 00 Ostrava, Czech Republic;
- Medical Faculty, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic;
| |
Collapse
|
29
|
Buccarelli M, Castellani G, Fiorentino V, Pizzimenti C, Beninati S, Ricci-Vitiani L, Scattoni ML, Mischiati C, Facchiano F, Tabolacci C. Biological Implications and Functional Significance of Transglutaminase Type 2 in Nervous System Tumors. Cells 2024; 13:667. [PMID: 38667282 PMCID: PMC11048792 DOI: 10.3390/cells13080667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Transglutaminase type 2 (TG2) is the most ubiquitously expressed member of the transglutaminase family. TG2 catalyzes the transamidation reaction leading to several protein post-translational modifications and it is also implicated in signal transduction thanks to its GTP binding/hydrolyzing activity. In the nervous system, TG2 regulates multiple physiological processes, such as development, neuronal cell death and differentiation, and synaptic plasticity. Given its different enzymatic activities, aberrant expression or activity of TG2 can contribute to tumorigenesis, including in peripheral and central nervous system tumors. Indeed, TG2 dysregulation has been reported in meningiomas, medulloblastomas, neuroblastomas, glioblastomas, and other adult-type diffuse gliomas. The aim of this review is to provide an overview of the biological and functional relevance of TG2 in the pathogenesis of nervous system tumors, highlighting its involvement in survival, tumor inflammation, differentiation, and in the resistance to standard therapies.
Collapse
Affiliation(s)
- Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Claudio Tabolacci
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
30
|
Sojka C, Sloan SA. Gliomas: a reflection of temporal gliogenic principles. Commun Biol 2024; 7:156. [PMID: 38321118 PMCID: PMC10847444 DOI: 10.1038/s42003-024-05833-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024] Open
Abstract
The hijacking of early developmental programs is a canonical feature of gliomas where neoplastic cells resemble neurodevelopmental lineages and possess mechanisms of stem cell resilience. Given these parallels, uncovering how and when in developmental time gliomagenesis intersects with normal trajectories can greatly inform our understanding of tumor biology. Here, we review how elapsing time impacts the developmental principles of astrocyte (AS) and oligodendrocyte (OL) lineages, and how these same temporal programs are replicated, distorted, or circumvented in pathological settings such as gliomas. Additionally, we discuss how normal gliogenic processes can inform our understanding of the temporal progression of gliomagenesis, including when in developmental time gliomas originate, thrive, and can be pushed towards upon therapeutic coercion.
Collapse
Affiliation(s)
- Caitlin Sojka
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
31
|
Sun J, Wu S, Zhao W, Xue S, Zhang L, Ren J. MAPK-activated protein kinase 2 is associated with poor prognosis of glioma patients and immune inhibition in glioma. Front Oncol 2024; 14:1307992. [PMID: 38322416 PMCID: PMC10844562 DOI: 10.3389/fonc.2024.1307992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction An effective therapeutic method to noticeably improve the prognosis of glioma patients has not been developed thus far. MAPK-activated protein kinase 2 (MAPKAPK2) is a serine/threonine kinase, which is involved in tumorigenesis, tumor growth, metastasis, and the inflammatory process. The clinical significance and molecular function of MAPKAPK2 in glioma remain unclear. Methods MAPKAPK2 expression in human glioma tissues was detected by immunohistochemistry and analyzed from the transcriptome sequencing data in TCGA and CGGA. Prognostic nomogram was constructed to predict the survival risk of individual patients. GO and KEGG enrichment analyses were performed to analyze the function and pathways MAPKAPK2 involved. Single-cell RNA sequencing data was used to analyze the cell types in which MAPKAPK2 was enriched. Flow cytometry was used for cell cycle and apoptosis detection. The ability of cell proliferation and migration was analyzed by CCK8 and cell migration assay, respectively. Correlation analyses were performed to analyze the relationship of MAPKAPK2 with immune infiltration, immune regulators, chemokine, and chemokine receptors. Results MAPKAPK2 was not only aberrantly upregulated in glioma tissues but also correlated with poor clinical characteristics. Moreover, MAPKAPK2 was prevalent in isocitrate dehydrogenase (IDH) wild-type and 1p/19q non-codeletion glioma cohorts and predicted poor prognosis of glioma patients. MAPKAPK2 may be involved in cell proliferation, cell migration, DNA damage repair, and immune regulation in glioma. MAPKAPK2 was enriched in microglia/macrophages and malignant tumor cells. Further investigation into cellular function revealed that inhibiting MAPKAPK2 suppressed the proliferation and migration of glioblastoma multiforme (GBM) cells in vitro. The inhibition of MAPKAPK2 significantly induced the G1 cell cycle arrest and cell apoptosis of GBM cells. Consistent with the enriched function of MAPKAPK2 in immune regulation, MAPKAPK2 was correlated with immune cell infiltration in glioma tissues. Mechanistically, a series of immune regulators, immunomodulatory chemokine, and chemokine receptors were positively correlated with MAPKAPK2 expression. Discussion Our findings provide evidence of the clinical relevance of MAPKAPK2 in prognosis evaluation of glioma patients and highlight the underlying significance of MAPKAPK2 in glioma therapy.
Collapse
Affiliation(s)
- Jinmin Sun
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sicheng Wu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenyu Zhao
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Senrui Xue
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Zhang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ren
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
32
|
Louise Kelly C, Wydrzynska M, Phelan MM, Osharovich S, Delikatny EJ, Sée V, Poptani H. Inhibition of glioblastoma cell proliferation and invasion by the choline-kinase inhibitor JAS239 varies with cell type and hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576078. [PMID: 38293093 PMCID: PMC10827177 DOI: 10.1101/2024.01.17.576078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background Elevated choline kinase alpha (ChoK) is observed in most solid tumours including glioblastomas (GBM), yet until recently, inhibitors of ChoK have demonstrated limited efficacy in GBM models. Given that hypoxia is associated with GBM therapy resistance, we hypothesised that tumour hypoxia could be responsible for such limitations. We therefore evaluated in GBM cells, the effect of hypoxia on the function of JAS239, a potent ChoK inhibitor. Methods Rodent (F98 and 9L) and human (U-87 MG and U-251 MG) GBM cell lines were subjected to 72 hours of hypoxia conditioning and treated with JAS239 for 24 hours. NMR metabolomic measurements and analyses were performed to evaluate the signalling pathways involved. In addition, cell proliferation, cell cycle progression and cell invasion were measured in cell monolayers and 3D spheroids, with or without JAS239 treatment in normoxic or hypoxic cells to assess how hypoxia affects JAS239 function. Results Hypoxia and JAS239 treatment led to significant changes in the cellular metabolic pathways, specifically the phospholipid and glycolytic pathways associated with a reduction in cell proliferation via induced cell cycle arrest. Interestingly, JAS239 also impaired GBM invasion. However, JAS239 effects were variable depending on the cell line, reflecting the inherent heterogeneity observed in GBMs. Conclusion Our findings indicate that JAS239 and hypoxia can deregulate cellular metabolism, inhibit proliferation and alter cell invasion. These results may be useful for the design of new therapeutic strategies based on ChoK inhibition that can act on multiple pro-tumorigenic features.
Collapse
Affiliation(s)
- Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Martyna Wydrzynska
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- High field NMR facility, Department of Biochemistry & Systems Biology, University of Liverpool, UK
| | - Sofya Osharovich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Violaine Sée
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
33
|
Di Salle G, Tumminello L, Laino ME, Shalaby S, Aghakhanyan G, Fanni SC, Febi M, Shortrede JE, Miccoli M, Faggioni L, Cosottini M, Neri E. Accuracy of Radiomics in Predicting IDH Mutation Status in Diffuse Gliomas: A Bivariate Meta-Analysis. Radiol Artif Intell 2024; 6:e220257. [PMID: 38231039 PMCID: PMC10831518 DOI: 10.1148/ryai.220257] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 01/18/2024]
Abstract
Purpose To perform a systematic review and meta-analysis assessing the predictive accuracy of radiomics in the noninvasive determination of isocitrate dehydrogenase (IDH) status in grade 4 and lower-grade diffuse gliomas. Materials and Methods A systematic search was performed in the PubMed, Scopus, Embase, Web of Science, and Cochrane Library databases for relevant articles published between January 1, 2010, and July 7, 2021. Pooled sensitivity and specificity across studies were estimated. Risk of bias was evaluated using Quality Assessment of Diagnostic Accuracy Studies-2, and methods were evaluated using the radiomics quality score (RQS). Additional subgroup analyses were performed according to tumor grade, RQS, and number of sequences used (PROSPERO ID: CRD42021268958). Results Twenty-six studies that included 3280 patients were included for analysis. The pooled sensitivity and specificity of radiomics for the detection of IDH mutation were 79% (95% CI: 76, 83) and 80% (95% CI: 76, 83), respectively. Low RQS scores were found overall for the included works. Subgroup analyses showed lower false-positive rates in very low RQS studies (RQS < 6) (meta-regression, z = -1.9; P = .02) compared with adequate RQS studies. No substantial differences were found in pooled sensitivity and specificity for the pure grade 4 gliomas group compared with the all-grade gliomas group (81% and 86% vs 79% and 79%, respectively) and for studies using single versus multiple sequences (80% and 77% vs 79% and 82%, respectively). Conclusion The pooled data showed that radiomics achieved good accuracy performance in distinguishing IDH mutation status in patients with grade 4 and lower-grade diffuse gliomas. The overall methodologic quality (RQS) was low and introduced potential bias. Keywords: Neuro-Oncology, Radiomics, Integration, Application Domain, Glioblastoma, IDH Mutation, Radiomics Quality Scoring Supplemental material is available for this article. Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
- Gianfranco Di Salle
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Lorenzo Tumminello
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Maria Elena Laino
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Sherif Shalaby
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Gayane Aghakhanyan
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Salvatore Claudio Fanni
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Maria Febi
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Jorge Eduardo Shortrede
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Mario Miccoli
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Lorenzo Faggioni
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Mirco Cosottini
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Emanuele Neri
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| |
Collapse
|
34
|
Li X, Peng Z, An K, Xue M, Wang Z, Xia J, Qi Z, Shu X. Temsirolimus is a promising immunomodulatory agent for enhanced transplantation outcomes. Transpl Immunol 2023; 81:101952. [PMID: 37918580 DOI: 10.1016/j.trim.2023.101952] [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: 08/02/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Identifying effective immunosuppressive strategies is critical for addressing immunological rejection following organ transplantation. This study explores the potential immunosuppressive effects and mechanisms of temsirolimus, a rapamycin derivative, in organ transplantation. METHODS A mouse cardiac allograft model was established using a cervical cannula technique with BALB/c donors and C57BL/6 recipients. Mice were administered temsirolimus intragastrically and graft survival was evaluated. Histological staining was used to assess pathological changes. The BrdU assay was used to measure splenic T cell proliferation. Flow cytometry was used to quantify regulatory T cells (Tregs), CD4+ T cells, and CD8+ T cells. ELISA and qPCR assays were used to determine Foxp3, IL-4, IFN-γ, and TGF-β expression. RESULTS Temsirolimus displayed potent immunosuppressive effects at 20 mg/kg/day, significantly inhibiting T cell proliferation (84.6%, P < 0.0001) and prolonging graft survival (median 49 days vs. 8.5 days in controls, P < 0.0001). However, median survival decreased to 34.5 days upon withdrawal. Temsirolimus also reduced splenic CD4+ and CD8+ T cells (2.85% and 2.92%, P < 0.001) and antibody levels (IgM, IgG1, IgG2) by 11.85-29.09% (P < 0.0001) and increased Tregs, Foxp3, IL-4 (P < 0.01), and TGF-β (P < 0.05), while decreasing IFN-γ (P < 0.001). CONCLUSIONS Temsirolimus exhibited potent immunosuppressive effects, emerging as a strong candidate to mitigate organ transplant rejection.
Collapse
Affiliation(s)
- Xianguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zuojie Peng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke An
- Department of Physiology, Xuzhou Medical University, Xuzhou 221009, China
| | - Mengjiao Xue
- Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenzhen Wang
- Department of Pharmacy, Zhoukou Central Hospital, Zhoukou 466000, China
| | - Junjie Xia
- Organ Transplantation Institute, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361100, China.
| | - Zhongquan Qi
- Medical College of Guangxi University, Guangxi University, Nanning 530004, China.
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
35
|
Braidotti S, Curci D, Zampieri D, Covino C, Zanon D, Maximova N, Sala R. Iron Bioavailability in the Extracellular Environment Is More Relevant Than the Intracellular One in Viability and Gene Expression: A Lesson from Oligodendroglioma Cells. Biomedicines 2023; 11:2940. [PMID: 38001941 PMCID: PMC10668974 DOI: 10.3390/biomedicines11112940] [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: 09/13/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Oligodendroglioma (OG) is a brain tumor that contributes to <1% of brain tumor diagnoses in the pediatric population. Unfortunately, pediatric OG remains without definitive molecular characteristics to aid in diagnosis, and little is known about the tumor microenvironment. Tumor cells' metabolism and proliferation rate are generally higher than those of healthy cells, so their iron demand is also significantly higher. This consideration underlines the great importance of iron for tumor development and progression. In this context, this study aims to evaluate the effect of iron in a cellular in vitro model of human oligodendroglioma brain tumor. Cell morphology, the effect of siderotic medium on cell growth, iron uptake, and the expression of iron-metabolism-related genes were evaluated via optic microscopy, ICP-MS, confocal microscopy, and real-time PCR, respectively. This study underlines the great importance of iron for tumor development and progression and also the possibility of reducing the available iron concentration to determine an antiproliferative effect on OG. Therefore, every attempt can be promising to defeat OG for which there are currently no long-term curative therapies.
Collapse
Affiliation(s)
- Stefania Braidotti
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Debora Curci
- Advanced Translational Diagnostic Laboratory, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Daniele Zampieri
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Cesare Covino
- Advanced Light and Electron Microscopy Imaging Centre (ALEMBIC), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Davide Zanon
- Pharmacy and Clinical Pharmacology Department, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Natalia Maximova
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Roberto Sala
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy;
| |
Collapse
|
36
|
Giordano F, D'Amico M, Montalto FI, Malivindi R, Chimento A, Conforti FL, Pezzi V, Panno ML, Andò S, De Amicis F. Cdk4 Regulates Glioblastoma Cell Invasion and Stemness and Is Target of a Notch Inhibitor Plus Resveratrol Combined Treatment. Int J Mol Sci 2023; 24:10094. [PMID: 37373242 DOI: 10.3390/ijms241210094] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive types of cancer characterized by poor patient outcomes. To date, it is believed that the major cause of its recurrence and chemoresistance is represented by the enrichment of GBM stem cells (GSCs) sustained by the abnormal activation of a number of signaling pathways. In this study, we found that in GBM cells, treatment with low toxicity doses of the γ-secretase inhibitor RO4929097 (GSI), blocking the Notch pathway activity, in combination with resveratrol (RSV) was able to reverse the basal mesenchymal phenotype to an epithelial-like phenotype, affecting invasion and stemness interplay. The mechanism was dependent on cyclin D1 and cyclin-dependent kinase (CDK4), leading to a reduction of paxillin (Pxn) phosphorylation. Consequently, we discovered the reduced interaction of Pxn with vinculin (Vcl), which, during cell migration, transmits the intracellular forces to the extracellular matrix. The exogenous expression of a constitutively active Cdk4 mutant prevented the RSV + GSI inhibitory effects in GBM cell motility/invasion and augmented the expression of stemness-specific markers, as well as the neurosphere sizes/forming abilities in untreated cells. In conclusion, we propose that Cdk4 is an important regulator of GBM stem-like phenotypes and invasive capacity, highlighting how the combined treatment of Notch inhibitors and RSV could be prospectively implemented in the novel therapeutic strategies to target Cdk4 for these aggressive brain tumors.
Collapse
Affiliation(s)
- Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Maria D'Amico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Francesca Ida Montalto
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
37
|
Doghish AS, Elballal MS, Elazazy O, Elesawy AE, Shahin RK, Midan HM, Sallam AAM, Elbadry AM, Mohamed AK, Ishak NW, Hassan KA, Ayoub AM, Shalaby RE, Elrebehy MA. miRNAs as potential game-changers in bone diseases: Future medicinal and clinical uses. Pathol Res Pract 2023; 245:154440. [PMID: 37031531 DOI: 10.1016/j.prp.2023.154440] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023]
Abstract
MicroRNAs (miRNAs), short, highly conserved non-coding RNA, influence gene expression by sequential mechanisms such as mRNA breakdown or translational repression. Many biological processes depend on these regulating substances, thus changes in their expression have an impact on the maintenance of cellular homeostasis and result in the emergence of a variety of diseases. Relevant studies have shown in recent years that miRNAs are involved in many stages of bone development and growth. Additionally, abnormal production of miRNA in bone tissues has been closely associated with the development of numerous bone disorders, such as osteonecrosis, bone cancer, and bone metastases. Many pathological processes, including bone loss, metastasis, the proliferation of osteosarcoma cells, and differentiation of osteoblasts and osteoclasts, are under the control of miRNAs. By bringing together the most up-to-date information on the clinical relevance of miRNAs in such diseases, this study hopes to further the study of the biological features of miRNAs in bone disorders and explore their potential as a therapeutic target.
Collapse
|
38
|
Zhou X, Jin G, Zhang J, Liu F. Recruitment mechanisms and therapeutic implications of tumor-associated macrophages in the glioma microenvironment. Front Immunol 2023; 14:1067641. [PMID: 37153567 PMCID: PMC10157099 DOI: 10.3389/fimmu.2023.1067641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
As one of the main components of the glioma immune microenvironment, glioma-associated macrophages (GAMs) have increasingly drawn research interest. Primarily comprised of resident microglias and peripherally derived mononuclear macrophages, GAMs are influential in a variety of activities such as tumor cell resistance to chemotherapy and radiotherapy as well as facilitation of glioma pathogenesis. In addition to in-depth research of GAM polarization, study of mechanisms relevant in tumor microenvironment recruitment has gradually increased. Suppression of GAMs at their source is likely to produce superior therapeutic outcomes. Here, we summarize the origin and recruitment mechanism of GAMs, as well as the therapeutic implications of GAM inhibition, to facilitate future glioma-related research and formulation of more effective treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Fusheng Liu
- *Correspondence: Junwen Zhang, ; Fusheng Liu,
| |
Collapse
|