1
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
2
|
Li J, Wu W, Ye L, Zheng B. Hyperglycemia as driver of glioblastoma progression: Insights from Mendelian randomization and single-cell transcriptomics. Brain Res 2025; 1858:149636. [PMID: 40210146 DOI: 10.1016/j.brainres.2025.149636] [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: 11/06/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Hyperglycemia and diabetes may influence GBM progression by altering tumor metabolism and the tumor microenvironment. However, the causal relationship between blood glucose levels and GBM remains unclear. METHODS Mendelian randomization (MR) analysis was performed using GWAS data from the UK Biobank and FinnGen databases, with fasting blood glucose, plasma glucose, cerebrospinal fluid (CSF) glucose, and diabetes as exposures. Single-cell RNA sequencing of GBM mouse models on high-glucose and control diets was conducted to explore the cellular landscape of the tumor microenvironment under hyperglycemic conditions. Additionally, gene set enrichment analysis (GSEA) was performed on transcriptomic data from brain tissues of diabetic patients to assess the activity of GBM-related pathways. RESULTS MR analysis demonstrated a significant genetic relationship between elevated fasting blood glucose and GBM risk, with an odds ratio (OR) of 40.991 (95 % CI: 2.066-813.447, p = 0.015). Type 2 diabetes (T2D) also showed a potential causal link with GBM, with the Weighted Median and Inverse Variance Weighted methods yielding ORs of 2.740 (95 % CI: 1.033-7.273, p = 0.043) and 2.100 (95 % CI: 1.029-4.287, p = 0.042), respectively. Single-cell transcriptomic analysis of GBM mouse models revealed an increased proportion of GBM tumor stem cells and pro-tumorigenic M2 macrophages in the high-glucose diet (HGD) group. GSEA of diabetic patient brain tissue revealed heightened activity of GBM-related pathways, particularly in astrocytes, endothelial cells, and neurons. CONCLUSION These findings suggest that hyperglycemia may actively contribute to GBM progression by promoting cellular changes within the tumor microenvironment and activating GBM-related pathways in brain tissues.
Collapse
Affiliation(s)
- Jin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wenjing Wu
- The Central Laboratory, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Liguo Ye
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
3
|
Dang Y, Chen Y, Chen J, Yuan G, Pan Y. Machine learning unravels the mysteries of glioma typing and treatment. Biochem Biophys Rep 2025; 42:101969. [PMID: 40129966 PMCID: PMC11930589 DOI: 10.1016/j.bbrep.2025.101969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/25/2025] [Accepted: 02/28/2025] [Indexed: 03/26/2025] Open
Abstract
Gliomas, which are complex primary malignant brain tumors known for their heterogeneous and invasive nature, present substantial challenges for both treatment and prognosis. Recent advancements in whole-genome studies have opened new avenues for investigating glioma mechanisms and therapies. Through single-cell analysis, we identified a specific cluster of cancer cell-related genes within gliomas. By leveraging diverse datasets and employing non-negative matrix factorization (NMF), we developed a glioma subtyping method grounded in this identified gene set. Our exploration delved into the clinical implications and underlying regulatory frameworks of the newly defined subtype classification, revealing its intimate ties to glioma malignancy and prognostic outcomes. Comparative assessments between the identified subtypes revealed differences in clinical features, immune modulation, and the tumor microenvironment (TME). Using tools such as the limma R package, weighted gene co-expression network analysis (WGCNA), machine learning methodologies, survival analyses, and protein-protein interaction (PPI) networks, we identified key driver genes influencing subtype differentiation while quantifying associated outcomes. This study not only sheds light on the biological mechanisms within gliomas but also paves the way for precise molecular targeted therapies within this intricate disease landscape.
Collapse
Affiliation(s)
- Ying Dang
- The Second Medical College of Lanzhou University, Lanzhou, Gansu, 730030, PR China
| | - Youhu Chen
- Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province, 710032, PR China
| | - Jie Chen
- The Northern Medical District, Chinese PLA General Hospital, Beijing, 100089, PR China
| | - Guoqiang Yuan
- The Second Medical College of Lanzhou University, Lanzhou, Gansu, 730030, PR China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, 730030, PR China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University. Lanzhou, Gansu, 730030, PR China
| | - Yawen Pan
- The Second Medical College of Lanzhou University, Lanzhou, Gansu, 730030, PR China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, 730030, PR China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University. Lanzhou, Gansu, 730030, PR China
| |
Collapse
|
4
|
Mahboubi E, Mondanizadeh M, Almasi-Hashiani A, Ghasemi E. Exploring the effects of doxorubicin on survival rates in glioma patients: a comprehensive systematic review. Eur J Med Res 2025; 30:425. [PMID: 40437615 PMCID: PMC12121047 DOI: 10.1186/s40001-025-02674-5] [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: 01/12/2025] [Accepted: 05/09/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Glioma is still one of the most aggressive and common types of brain and central nervous system cancer, with few effective treatment options. Despite progress in therapy, there is a requirement for new methods to enhance patient outcomes. Doxorubicin (DOX), a type of anthracycline that has been proven to be effective in different cancers, encounters difficulties in treating glioma because of the blood-brain barrier. This systematic review is intended to assess the effects of DOX on the survival and quality of life of glioma patients. METHODS We conducted a thorough search and found 1576 records, from which 10 studies met our inclusion criteria. These studies, published between 1973 and 2023, were examined for overall survival (OS), progression-free survival (PFS), median time to progression (mTTP), response rate, and toxicity profiles. RESULTS The studies included in the analysis showed that OS ranged from 6 to 18.5 months, indicating potential improved outcomes with liposomal and PEGylated forms of DOX. PFS rates varied from 15 to 58%, and mTTP ranged from 11 to 39.83 weeks. Response rates varied from 19 to 88%, and while DOX was generally well-tolerated, some hematological and non-hematological toxicities were observed. DOX shows promise in prolonging survival and slowing glioma progression, especially with advanced delivery methods. CONCLUSIONS However, variations in research, small sample sizes, and lack of uniformity highlight the need for further investigation. Overcoming these limitations in future studies is crucial to maximize DOX's effectiveness in glioma treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Elyar Mahboubi
- Department of Biotechnology and Molecular Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mahdieh Mondanizadeh
- Department of Biotechnology and Molecular Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Markazi, Islamic Republic of Iran.
- Traditional and Complementary Medicine Research Center, School of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Elham Ghasemi
- Department of Biotechnology and Molecular Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
5
|
Yan X, Li R, Xu J, Liu H, He M, Jiang X, Ren C, Zhou Q. ARHGDIB as a prognostic biomarker and modulator of the immunosuppressive microenvironment in glioma. Cancer Immunol Immunother 2025; 74:204. [PMID: 40372473 PMCID: PMC12081808 DOI: 10.1007/s00262-025-04063-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Glioma, a prevalent malignant intracranial tumor, exhibits limited therapeutic efficacy due to its immunosuppressive microenvironment, leading to a poor prognosis for patients. ARHGDIB is implicated in the remodeling of the tumor microenvironment and plays a significant role in the pathogenesis of various tumors. However, its regulatory effect within the immune microenvironment of glioma remains unclear. METHODS The mRNA expression pattern of ARHGDIB was analyzed using public databases, and its expression was further validated in our collected cohort through quantitative PCR (qPCR) and immunohistochemistry (IHC). Kaplan-Meier survival analysis and LASSO-Cox regression were employed to ascertain the clinical significance of ARHGDIB in glioma. Subsequently, we systematically evaluated the association between ARHGDIB expression and immune characteristics within the glioma microenvironment, as well as its potential to predict treatment response in glioma. Additionally, in vitro experiments were conducted to elucidate the role of ARHGDIB in remodeling the glioma microenvironment and promoting tumor malignancy progression. RESULTS Combined with bioinformatics analysis of public databases and validation with qPCR and IHC on our cohort, our findings indicate that ARHGDIB is markedly overexpressed in glioma and correlates with poor patient prognosis, thereby serving as a potential biomarker for adverse outcomes in glioma. Functional enrichment and immune infiltration analyses reveal that ARHGDIB is implicated in the recruitment of immunosuppressive cells, such as M2 macrophages and neutrophils, contributing to the alteration of the glioma immunosuppressive microenvironment and hindering the immune response. Further investigations through single-cell sequencing, immunohistochemistry, immunofluorescence, and in vitro experiments demonstrate that ARHGDIB exhibits an expression pattern akin to CD163, with its overexpression inducing M2 macrophage polarization and facilitating glioma cell proliferation and migration. CONCLUSIONS ARHGDIB emerges as a novel marker for tumor-associated macrophages, playing a crucial role in shaping the immunosuppressive microenvironment and representing a promising prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Xuejun Yan
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.
| | - Rongnian Li
- Xiangtan Hospital of Traditional Chinese Medicine, Xiangtan, Hunan, China
| | - Jing Xu
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Hua Liu
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Minmin He
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Caiping Ren
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, China.
| | - Quanwei Zhou
- Department of Neurosurgery, The National Key Clinical Specialty, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Goo J, Lee JS, Park J, Jeon SI, Kim J, Yun WS, Shim N, Moon Y, Song S, Kim J, Cho H, Jeong JY, Lee JS, Han S, Lee HJ, Koh WG, Chang WS, Kim TI, Kim K. Dual Delivery of Light/Prodrug Nanoparticles Using Tumor-Implantable Micro Light-Emitting Diode on an Optofluidic System for Combinational Glioma Treatment. ACS NANO 2025; 19:17393-17409. [PMID: 40296430 DOI: 10.1021/acsnano.4c17421] [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: 04/30/2025]
Abstract
Glioma is a highly lethal tumor with a poor prognosis, in which the presence of the blood-brain barrier (BBB) and skull significantly limits treatment options. To address this, a tumor-implantable optofluidic system (LED-SC), consisting of a microsized LED (microLED) and a microsyringe chip (SC), is proposed to deliver both light and prodrug nanoparticles (PNPs) directly to brain glioma. The LED-SC combines microLED and SC to enable intratumoral administration of light and PNPs for chemophotodynamic therapy. PNPs, self-assembled nanoparticles of verteporfin (VPF)-doxorubicin (DOX) prodrug, are cleaved by the enzyme cathepsin B, releasing active drugs specifically within tumor cells. In vitro studies show that PNPs are taken up by glioma cells and exhibit enhanced cytotoxicity under light irradiation. The PNPs-loaded LED-SC can be implanted into glioma, wherein PNPs are slowly diffused through the tumor, bypassing the BBB, and it also ensures effective light delivery in glioma beneath the skull, boosting chemo-photodynamic therapy. In glioma mouse models, PNP-loaded LED-SC implantation showed a 3.9-fold improvement in PNP delivery efficiency over intravenous administration, leading to better drug distribution and therapeutic results. The PNPs-loaded LED-SC offers a promising and minimally invasive solution for glioma treatment, overcoming the barriers of the BBB and skull while reducing systemic toxicity.
Collapse
Affiliation(s)
- Jagyeong Goo
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jun Seo Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Junwon Park
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Jeongrae Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Wan Su Yun
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Nayeon Shim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Yujeong Moon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Sunejeong Song
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Jinseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Hanhee Cho
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Ju Yeon Jeong
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Ju Seung Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Sangheon Han
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyeon-Ju Lee
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Won Seok Chang
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
- Gradutate Program in Innovative Biomaterials Convergence, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| |
Collapse
|
7
|
Pu J, Yuan K, Tao J, Qin Y, Li Y, Fu J, Li Z, Zhou H, Tang Z, Li L, Gai X, Qin D. Glioblastoma multiforme: an updated overview of temozolomide resistance mechanisms and strategies to overcome resistance. Discov Oncol 2025; 16:731. [PMID: 40353925 PMCID: PMC12069213 DOI: 10.1007/s12672-025-02567-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with high lethality. The typical treatment regimen includes post-surgical radiotherapy and temozolomide (TMZ) chemotherapy, which helps extend survival. Nevertheless, TMZ resistance occurs in approximately 50% of patients. This resistance is primarily associated with the expression of O6-methylguanine-DNA methyltransferase (MGMT), which repairs O6-methylguanine lesions generated by TMZ and is thought to be the major mechanism of drug resistance. Additionally, the mismatch repair and base excision repair pathways play crucial roles in TMZ resistance. Emerging studies also point to drug transport mechanisms, glioma stem cells, and the heterogeneous tumor microenvironment as additional influences on TMZ resistance in gliomas. A better understanding of these mechanisms is vital for developing new treatments to improve TMZ effectiveness, such as DNA repair inhibitors, inhibitors of multidrug transporting proteins, TMZ analogs, and combination therapies targeting multiple pathways. This article discusses the main resistance mechanisms and potential strategies to counteract resistance in GBM patients, aiming to broaden the understanding of these mechanisms for future research and to explore the therapeutic effects of traditional Chinese medicines and their active components in overcoming TMZ resistance.
Collapse
Affiliation(s)
- Jianlin Pu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Tao
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Yongxin Li
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Jing Fu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhong Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Haimei Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhengxiu Tang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Li
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xuesong Gai
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
8
|
Gonzalez-Prada I, Barcelos Ribeiro A, Dion M, Magariños B, Lapoujade C, Rousseau A, Concheiro A, Garcion E, Alvarez-Lorenzo C. Disulfiram-loaded electrospun fibers with antimicrobial and antitumoral properties for glioblastoma treatment. J Control Release 2025; 381:113615. [PMID: 40086760 DOI: 10.1016/j.jconrel.2025.113615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/02/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Glioblastoma (GB) is a malignant brain tumor with low survival rates and a high recurrence ratio due to limited therapeutic arsenal. The repurposed drug disulfiram (DSF), approved for alcoholism treatment, shows promising anticancer and antimicrobial activity, but its poor biopharmaceutical profile hinders its clinical use. This work aimed to develop DSF-loaded silk fibroin (SF) electrospun fibers for controlled release in the postsurgical resection cavity. Incorporating hydroxypropyl-β-cyclodextrin (HPβCD), which formed inclusion complexes with DSF, enhanced drug release rate and antimicrobial activity (>3 logCFUs reduction) against Staphylococcus aureus and Pseudomonas aeruginosa. Addition of CuCl2 enabled in situ formation of Cu(DDC)2 complexes, further boosting antimicrobial and in vitro antitumoral effects of the nanofibers (≤ 500 nm) while maintaining adequate mechanical properties. Selective toxicity of DSF and DSF-loaded fibers against glioblastoma cells, while sparing against astrocytes, highlights the potential of the nanofibers for targeted brain cancer therapy. Increased potency of DSF at low concentrations when combined with SF fibers, HPβCD and copper was remarkable. Thus, DSF delivery and bioavailability can be significantly optimized through electrospun nanofibers, which may also allow for more precise dosing. Combination with radiotherapy was also explored to assess the translational potential of DSF as part of a combination therapy regimen for glioblastoma. In vivo studies in a rat model simulating GB surgery confirmed the safety of selected formulations in healthy brain tissue. However, findings suggest that DSF-loaded fibers alone may be insufficient for complete tumor eradication, indicating the need for combination with existing therapies to target residual tumor cells effectively.
Collapse
Affiliation(s)
- Iago Gonzalez-Prada
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain
| | - Arthur Barcelos Ribeiro
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Marine Dion
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Beatriz Magariños
- Departamento de Microbiología y Parasitología, Facultad de Biología-CIBUS and Aquatic One Health Research Center (iARCUS), Universidade de Santiago de Compostela, Spain
| | - Clémentine Lapoujade
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France
| | - Audrey Rousseau
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France; Department of Pathology, University Hospital of Angers, F-49000 Angers, France
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain
| | - Emmanuel Garcion
- Inserm UMR 1307, CNRS UMR 6075, Université de Nantes, CRCI2NA, Université d'Angers, 49000 Angers, France; PACEM (Plateforme d'Analyse Cellulaire et Moléculaire), Université d'Angers, SFR 4208, F-49000 Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000 Angers, France.
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain.
| |
Collapse
|
9
|
Zhang H, Feng K, Han M, Shi Y, Zhang Y, Wu J, Yang W, Wang X, Di L, Wang R. Homologous magnetic targeted immune vesicles for amplifying immunotherapy via ferroptosis activation augmented photodynamic therapy against glioblastoma. J Control Release 2025; 383:113816. [PMID: 40334815 DOI: 10.1016/j.jconrel.2025.113816] [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: 02/06/2025] [Revised: 04/25/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
In spite of noteworthy breakthroughs in clinical treatments, immune checkpoint blockade (ICB) therapy is often hindered by T lymphocyte dysfunction in the immunosuppressive microenvironment of glioblastoma (GBM). Herein, GBM-derived exosomes (GBM-Exos) co-encapsulate ferroptosis inducer arsenic trioxide (ATO) and NIR photosensitizer IR780, modified with superparamagnetic iron oxide nanoparticle (SPION), to construct homologous magnetic targeted immune vesicles (Sp-Exo/AI) for reinvigorating anti-tumor immunity. SPION modified GBM-Exos display capacities of tumor accumulation and blood-brain barrier penetration. Notably, reactive oxygen species metabolism is disturbed by ferroptosis activation augmented photodynamic therapy (PDT), hence triggering tumor cell lysis and mitochondrial damage to reshape tumor microenvironment (TME) and transform GBM from immune "cold" to "hot". Accordingly, the tumor specific T lymphocytes function and phenotype transformation of macrophages were promoted to stimulate robust innate and adaptive immunities. Significantly, the remarkable ferroptosis activation augmented PDT combining with programmed death-1 antibody actives long-term immune memory and inhibits distal tumor metastasis. Superior anti-tumor effect of Sp-Exo/AI in the recurrence model, breast cancer model and patient-derived model were observed as well. Altogether, the presented homologous magnetic targeted immune vesicles exhibit substantial potential for amplifying immune response in "cold" tumors like GBM through revising immunosuppressive TME.
Collapse
Affiliation(s)
- Hanwen Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Kuanhan Feng
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| | - Yali Shi
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yingjie Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Wu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wanyi Yang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinrui Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Liuqing Di
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruoning Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
10
|
Zhao S, Qu Y, Sun Z, Zhang S, Xia M, Shi Y, Wang J, Wang Y, Zhong Z, Meng F. Glioblastoma Cell Lysate and Adjuvant Nanovaccines via Strategic Vaccination Completely Regress Established Murine Tumors. Adv Healthc Mater 2025; 14:e2500911. [PMID: 40270217 DOI: 10.1002/adhm.202500911] [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: 02/19/2025] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Tumor vaccines have shown great promise for treating various malignancies; however, glioblastoma (GBM), characterized by its immunosuppressive tumor microenvironment, high heterogeneity, and limited accessibility, has achieved only modest clinical benefits. Here, it is reported that GBM cell lysate nanovaccines boosted with TLR9 agonist CpG ODN (GlioVac) via a strategic vaccination regimen achieve complete regression of malignant murine GBM tumors. Subcutaneous administration of GlioVac promotes uptake by cervical lymph nodes and antigen presentation cells, bolstering antigen cross-presentation and infiltration of GBM-specific CD8+ T cells into the tumor. Notably, a regimen involving two subcutaneous and three intravenous vaccinations not only activates systemic anti-GBM immunity but also further enhances the tumor infiltration of cytotoxic T lymphocytes, effectively reshaping the "cold" GBM tumor into a "hot" tumor. This approach led to a state of tumor-free survival in 5 out of 7 mice bearing the established GL261 GBM model with complete protection from tumor rechallenge. In an orthotopic hRas-GBM model induced by a lentiviral plasmid, GlioVac resulted in ≈100% complete tumor regression. These findings suggest that GlioVac provides a personalized therapeutic vaccine strategy for glioblastoma.
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, P. R. 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, P. R. 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, P. R. China
| | - Shuo Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. 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, P. R. China
| | - Yan Shi
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. 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, P. R. China
| | - Yuan Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. 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, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. 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, P. R. China
| |
Collapse
|
11
|
Branco F, Cunha J, Mendes M, Sousa JJ, Vitorino C. 3D Bioprinting Models for Glioblastoma: From Scaffold Design to Therapeutic Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501994. [PMID: 40116532 DOI: 10.1002/adma.202501994] [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: 01/28/2025] [Indexed: 03/23/2025]
Abstract
Conventional in vitro models fail to accurately mimic the tumor in vivo characteristics, being appointed as one of the causes of clinical attrition rate. Recent advances in 3D culture techniques, replicating essential physical and biochemical cues such as cell-cell and cell-extracellular matrix interactions, have led to the development of more realistic tumor models. Bioprinting has emerged to advance the creation of 3D in vitro models, providing enhanced flexibility, scalability, and reproducibility. This is crucial for the development of more effective drug treatments, and glioblastoma (GBM) is no exception. GBM, the most common and deadly brain cancer, remains a major challenge, with a median survival of only 15 months post-diagnosis. This review highlights the key components needed for 3D bioprinted GBM models. It encompasses an analysis of natural and synthetic biomaterials, along with crosslinking methods to improve structural integrity. Also, it critically evaluates current 3D bioprinted GBM models and their integration into GBM-on-a-chip platforms, which hold noteworthy potential for drug screening and personalized therapies. A versatile development framework grounded on Quality-by-Design principles is proposed to guide the design of bioprinting models. Future perspectives, including 4D bioprinting and machine learning approaches, are discussed, along with the current gaps to advance the field further.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - João J Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| |
Collapse
|
12
|
Zhang R, Song Y, Yao J, Qin H, Ye Y, Gao J, Zhang C, Han D, Gao M, Chen H, Chen X, Zhao S, Liu K, Tu Y, Xu Z. RNA-Seq Reveals the Mechanism of Synergistic Hydrogen-Chemotherapy Based on Active Magnesium Micromotors for Inhibiting Glioblastoma Recurrence by Modulating Tumor Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408809. [PMID: 40259480 DOI: 10.1002/smll.202408809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Indexed: 04/23/2025]
Abstract
Postoperative recurrence of glioblastoma (GBM) is a key contributing factor to the unfavorable prognosis of patients. Chemotherapy has been extensively employed as a postoperative treatment for GBM; however, the produced drug resistance significantly undermines the chemotherapeutic efficacy. Herein, a multifunctional system based on magnesium micromotor (Mg-Motor-DOX) is designed and fabricated that can generate hydrogen gas in situ and actively deliver the chemotherapeutic drug doxorubicin (DOX). Utilizing a temperature-sensitive hydrogel, Mg-Motor-DOX is administrated in situ to the residual cavity of the tumor after subtotal GBM resection. The produced H2 by the Mg-water reaction not only propels the motion of motors but also functions as an antioxidant to effectively alleviate the neuroinflammation caused by GBM resection. The H2 bubbles create a pronounced vortex flow in situ, greatly enhancing the DOX penetration and the sensitivity of GBM cells to DOX. Therefore, synergistic hydrogen-chemotherapy significantly inhibits the recurrence of the in situ GBM model. RNA-Seq technology further elucidates the role of the strategy in modulating the tumor immune microenvironment via converting cold tumors into hot tumors, thereby establishing a theoretical foundation for the clinical implementation of synergistic hydrogen-chemotherapy.
Collapse
Affiliation(s)
- Ruotian Zhang
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yanzhen Song
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiawei Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hanfeng Qin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yicheng Ye
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junbin Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Cheng Zhang
- University of Toronto Scarborough, 1265 Military Trail, Scarborough, ON, M1C 1A4, Canada
| | - Dayong Han
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ming Gao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hao Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Kun Liu
- Experimental Education/Administration Center, National Demonstration Center for Experimental Education of Basic Medical Sciences, Key Laboratory of Functional Proteomics of Guangdong Province, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yingfeng Tu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhili Xu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| |
Collapse
|
13
|
Wu G, Chen Y, Chen C, Liu J, Wu Q, Zhang Y, Chen R, Xiao J, Su Y, Shi H, Yu C, Wang M, Ouyang Y, Jiang A, Chen Z, Ye X, Shen C, Reheman A, Li X, Liu M, Shen J. Role and mechanisms of exercise therapy in enhancing drug treatment for glioma: a review. Front Immunol 2025; 16:1576283. [PMID: 40370453 PMCID: PMC12075166 DOI: 10.3389/fimmu.2025.1576283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/02/2025] [Indexed: 05/16/2025] Open
Abstract
Gliomas, particularly glioblastoma (GBM), are among the most aggressive and challenging brain tumors to treat. Although current therapies such as chemotherapy, radiotherapy, and targeted treatments have extended patient survival to some extent, their efficacy remains limited and is often accompanied by severe side effects. In recent years, exercise therapy has gained increasing attention as an adjunctive treatment in clinical and research settings. Exercise not only improves patients' physical function and cognitive abilities but may also enhance the efficacy of conventional drug treatments by modulating the immune system, suppressing inflammatory responses, and improving blood-brain barrier permeability. This review summarizes the potential mechanisms of exercise in glioma treatment, including enhancing immune surveillance through activation of natural killer (NK) cells and T cells, and increasing drug penetration by improving blood-brain barrier function. Additionally, studies suggest that exercise can synergize with chemotherapy and immunotherapy, improving treatment outcomes while reducing drug-related side effects. Although the application of exercise therapy in glioma patients is still in the exploratory phase, existing evidence indicates its significant clinical value as an adjunctive approach, with the potential to become a new standard in glioma treatment in the future.
Collapse
Affiliation(s)
- Guanghui Wu
- Department of Neurosurgery, Ningde Clinical Medical College, Fujian Medical University, Ningde, Fujian, China
- Department of Neurosurgery, Ningde Municipal Hospital, Ningde Normal University, Ningde, Fujian, China
| | - Yisheng Chen
- Department of Neurosurgery, Ningde Clinical Medical College, Fujian Medical University, Ningde, Fujian, China
- Department of Neurosurgery, Ningde Municipal Hospital, Ningde Normal University, Ningde, Fujian, China
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, United States
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
- Department of Neurosurgery and Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Chong Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianling Liu
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Qiaowu Wu
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Yazhen Zhang
- School of Physical Education, Ningde Normal University, Ningde, Fujian, China
| | - Runqiong Chen
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Jianzhong Xiao
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Yusheng Su
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Haojun Shi
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Chunsheng Yu
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Miao Wang
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Yifan Ouyang
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Airong Jiang
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Zhengzhou Chen
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Xiao Ye
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Chengwan Shen
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Aikebaier Reheman
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Xianjun Li
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Ming Liu
- Department of Neurosurgery, Ningde Clinical Medical College, Fujian Medical University, Ningde, Fujian, China
- Department of Neurosurgery, Ningde Municipal Hospital, Ningde Normal University, Ningde, Fujian, China
| | - Jiancheng Shen
- Department of Neurosurgery, Ningde Clinical Medical College, Fujian Medical University, Ningde, Fujian, China
- Department of Neurosurgery, Ningde Municipal Hospital, Ningde Normal University, Ningde, Fujian, China
| |
Collapse
|
14
|
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
|
15
|
Zhao B, Wu J, Zhang T, Han M, Zhang C, Rong X, Zhang R, Chen X, Peng F, Jin J, Liu S, Dong X, Zhao S. A spatial transcriptomics study of MES-like and mono/macro cells in gliomas. Sci Rep 2025; 15:12730. [PMID: 40222970 PMCID: PMC11994772 DOI: 10.1038/s41598-025-95277-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Gliomas, including both glioblastoma multiforme (GBM) and lower-grade glioma (LGG), present a substantial challenge in neuro-oncology because of genetic heterogeneity and unsatisfactory prognosis. This study aimed to conduct a comprehensive multi-omics analysis of gliomas using various bioinformatics approaches to identify potential therapeutic targets and prognostic markers. A comprehensive analysis was conducted on 1327 sequencing data samples alongside their relevant clinical information sourced from The Cancer Genome Atlas (TCGA) pertaining to glioblastoma (GBM), low-grade glioma (LGG), the Chinese Glioma Genome Atlas (CCGA) and University of California Santa Cruz Xena (UCSC Xena) datasets. These tools were employed for gene expression profiling, survival analysis, and cell communication mapping. Spatial transcriptomics revealed the localization of mesenchymal (MES)-like malignant tumors, and drug sensitivity analysis was performed to evaluate responses to quinpirole and meropenem. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) framework was utilized to gauge the responsiveness to immunotherapy. The MES-like malignant and monocyte/macrophage (mono/macro) cell subsets showed high hallmark scores, playing key roles in the tumor microenvironment. MES-like malignant marker gene scores correlated with overall survival across datasets, whereas mono/macro marker gene scores were significant in the TCGA-LGG and CCGA datasets. Key interactions between these cell types were found, especially with CD14-ITGB2, LGALS1-CD69, and APOE-TREM2. The mono/macro cell subset demonstrated better immune therapy responsiveness, as indicated by lower TIDE scores. Spatial transcriptomics revealed that MES-like malignant tumors are predominantly localized in four distinct regions, with the marker genes CHI3L1 and ADM confirming these locations. Drug sensitivity analysis revealed differential responses of the MES-like malignant cell subset to quinpirole and meropenem. Our results offer fresh perspectives on the differential roles of MES-like malignant and monocyte/macrophage cell subsets in tumor progression and immune modulation, providing novel insights into glioma biology.
Collapse
Affiliation(s)
- Boyan Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Jianing Wu
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Tiehui Zhang
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong, China
| | - Mingyang Han
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Cheng Zhang
- University of Toronto Scarborough 1265 Military Trail, Scarborough, ON, M1C 1A4, Canada
| | - Xuan Rong
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, 150001, Heilongjiang, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Jin
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Shiya Liu
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Xingli Dong
- Central Laboratory, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
| | - Shiguang Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China.
- Department of Neurosurgery, Shenzhen University General Hospital, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
16
|
Qiu Q, Deng H, Song P, Liu Y, Zhang M. Lactylation in Glioblastoma: A Novel Epigenetic Modifier Bridging Epigenetic Plasticity and Metabolic Reprogramming. Int J Mol Sci 2025; 26:3368. [PMID: 40244246 PMCID: PMC11989911 DOI: 10.3390/ijms26073368] [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: 01/21/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Glioblastoma, the most common and aggressive primary malignant brain tumor, is characterized by a high rate of recurrence, disability, and lethality. Therefore, there is a pressing need to develop more effective prognostic biomarkers and treatment approaches for glioblastoma. Lactylation, an emerging form of protein post-translational modification, has been closely associated with lactate, a metabolite of glycolysis. Since the initial identification of lactylation sites in core histones in 2019, accumulating evidence has shown the critical role that lactylation plays in glioblastoma development, assessment of poor clinical prognosis, and immunosuppression, which provides a fresh angle for investigating the connection between metabolic reprogramming and epigenetic plasticity in glioblastoma cells. The objective of this paper is to present an overview of the metabolic and epigenetic roles of lactylation in the expanding field of glioblastoma research and explore the practical value of developing novel treatment plans combining targeted therapy and immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Mengxian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.Q.); (H.D.); (P.S.); (Y.L.)
| |
Collapse
|
17
|
Wang H, Wang Q, Chen YH, Chen X, Zheng DK, Xie Z, Feng DM, Liu L, Li J, Liu Y. Phlecarinatones H-N: Abietane-type diterpenoids from Phlegmariurus carinatus with proliferative inhibitory effect on U251 glioblastoma cells. PHYTOCHEMISTRY 2025; 232:114356. [PMID: 39675447 DOI: 10.1016/j.phytochem.2024.114356] [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: 08/03/2024] [Revised: 11/26/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Thirteen abietane-type diterpenoids, including seven previously undescribed compounds and six known analogs, were isolated from the root and aerial parts of Phlegmariurus carinatus. Their structures were elucidated by comprehensive spectroscopic data analysis (UV, IR, NMR, and HRESIMS) and quantum chemical calculations (calculated ECD or 13C NMR). Notably, these compounds exhibited high structural diversity. Compounds 1-8 possessed six distinct fused ring systems. Phlecarinatones I (2) and J (3) were identified as rare 6,7-seco-abietane diterpenoids featuring a five-membered lactone ring B. Compounds (1-12) were evaluated for their anti-proliferative activities in the U251 cell line. In particular, phlecarinatone I (2) exhibited potential inhibitory effects on U251 cell proliferation, with an IC50 value of 13.88 ± 1.82 μM. To elucidate the underlying molecular mechanism, p53 signal pathway was enriched from our RNA-seq data. Further investigations using western blot and immunofluorescence assays confirmed that p53 expression was up-regulated in a concentration-independent manner. Additionally, molecular docking studies suggested 2 likely binds to the MDM2-p53 binding region. Finally, an shRNA-mediated MDM2 knockdown experiment provided definitive evidence that 2 acts as a potent inhibitor of glioma proliferation, operating via the modulation of the MDM2-p53 pathway. These findings suggest that 2 might be a valuable of lead compound with anti-proliferative activity.
Collapse
Affiliation(s)
- Hua Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; Jiangxi Provincal Key Laboratory of Tissue Engineering, 2024SSY06291, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Qiang Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yong-Hong Chen
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Xi Chen
- Xiangya School of Pharmaceutical Sciences, Department of Pharmacy, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dong-Kun Zheng
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zhen Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Du-Min Feng
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Lin Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jing Li
- Xiangya School of Pharmaceutical Sciences, Department of Pharmacy, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Yang Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; Jiangxi Provincal Key Laboratory of Tissue Engineering, 2024SSY06291, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; School of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China.
| |
Collapse
|
18
|
Zhou Q, Ke X, Man J, Jiang J, Ren J, Xue C, Zhang B, Zhang P, Zhao J, Zhou J. Integrated MRI radiomics, tumor microenvironment, and clinical risk factors for improving survival prediction in patients with glioblastomas. Strahlenther Onkol 2025; 201:398-410. [PMID: 39249499 DOI: 10.1007/s00066-024-02283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 07/14/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE To construct a comprehensive model for predicting the prognosis of patients with glioblastoma (GB) using a radiomics method and integrating clinical risk factors, tumor microenvironment (TME), and imaging characteristics. MATERIALS AND METHODS In this retrospective study, we included 148 patients (85 males and 63 females; median age 53 years) with isocitrate dehydrogenase-wildtype GB between January 2016 and April 2022. Patients were randomly divided into the training (n = 104) and test (n = 44) sets. The best feature combination related to GB overall survival (OS) was selected using LASSO Cox regression analyses. Clinical, radiomics, clinical-radiomics, clinical-TME, and clinical-radiomics-TME models were established. The models' concordance index (C-index) was evaluated. The survival curve was drawn using the Kaplan-Meier method, and the prognostic stratification ability of the model was tested. RESULTS LASSO Cox analyses were used to screen the factors related to OS in patients with GB, including MGMT (hazard ratio [HR] = 0.642; 95% CI 0.414-0.997; P = 0.046), TERT (HR = 1.755; 95% CI 1.095-2.813; P = 0.019), peritumoral edema (HR = 1.013; 95% CI 0.999-1.027; P = 0.049), tumor purity (TP; HR = 0.982; 95% CI 0.964-1.000; P = 0.054), CD163 + tumor-associated macrophages (TAMs; HR = 1.049; 95% CI 1.021-1.078; P < 0.001), CD68 + TAMs (HR = 1.055; 95% CI 1.018-1.093; P = 0.004), and the six radiomics features. The clinical-radiomics-TME model had the best survival prediction ability, the C‑index was 0.768 (0.717-0.819). The AUC of 1‑, 2‑, and 3‑year OS prediction in the test set was 0.842, 0.844, and 0.795, respectively. CONCLUSION The clinical-radiomics-TME model is the most effective for predicting the survival of patients with GB. Radiomics features, TP, and TAMs play important roles in the prognostic model.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jiangwei Man
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Surgical, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jian Jiang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Jialiang Ren
- Department of Pharmaceuticals Diagnostics, GE HealthCare, Beijing, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Peng Zhang
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jun Zhao
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, 730030, Lanzhou, Gansu, China.
| |
Collapse
|
19
|
Keshari R, Dewani M, Kaur N, Patel GK, Singh SK, Chandra P, Prasad R, Srivastava R. Lipid Nanocarriers as Precision Delivery Systems for Brain Tumors. Bioconjug Chem 2025; 36:347-366. [PMID: 39937652 DOI: 10.1021/acs.bioconjchem.5c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Brain tumors, particularly glioblastomas, represent the most complicated cancers to treat and manage due to their highly invasive nature and the protective barriers of the brain, including the blood-brain barrier (BBB). The efficacy of currently available treatments, viz., radiotherapy, chemotherapy, and immunotherapy, are frequently limited by major side effects, drug resistance, and restricted drug penetration into the brain. Lipid nanoparticles (LNPs) have emerged as a promising and targeted delivery system for brain tumors. Lipid nanocarriers have gained tremendous attention for brain tumor therapeutics due to multiple drug encapsulation abilities, controlled release, better biocompatibility, and ability to cross the BBB. Herein, a detailed analysis of the design, mechanisms, and therapeutic benefits of LNPs in brain tumor treatment is discussed. Moreover, we also discuss the safety issues and clinical developments of LNPs and their current and future challenges. Further, we also focused on the clinical transformation of LNPs in brain tumor therapy by eliminating side effects and engineering the LNPs to overcome the related biological barriers, which provide personalized, affordable, and low-risk treatment options.
Collapse
Affiliation(s)
- Roshan Keshari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Mahima Dewani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Navneet Kaur
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India-211004
| | - Sumit Kumar Singh
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
20
|
Singh G, Rohit, Kumar P, Aran KR. Targeting EGFR and PI3K/mTOR pathways in glioblastoma: innovative therapeutic approaches. Med Oncol 2025; 42:97. [PMID: 40064710 DOI: 10.1007/s12032-025-02652-1] [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: 11/20/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma (GBM) stands as the most aggressive form of primary brain cancer in adults, characterized by its rapid growth, invasive nature, and a robust propensity to induce angiogenesis, forming new blood vessels to sustain its expansion. GBM arises from astrocytes, star-shaped glial cells, and despite significant progress in understanding its molecular mechanisms, its prognosis remains grim. It is frequently associated with mutations or overexpression of the epidermal growth factor receptor (EGFR), which initiates several downstream signaling pathways. Dysregulation of key signaling pathways, such as EGFR/PTEN/AKT/mTOR, drives tumorigenesis, promotes metastasis and leads to treatment resistance. The modest survival benefits of the conventional treatment of surgical resection followed by radiation and chemotherapy underscore the pressing need for innovative therapeutic approaches. In most the tumor, overexpression of EGFR is found associated with GBM and mutations in its several variants are important for promoting ongoing mitogenic signaling and tumor growth. This receptor inhibits apoptosis and promotes cell survival and proliferation by activating downstream PI3K/AKT/mTOR pathways. This route is typically blocked by PTEN, a crucial tumor suppressor, however, GBM frequently results in abnormalities in this protein. The aim of this review is to explore the molecular foundations of GBM, with a focus on the EGFR and PI3K/mTOR pathways and their impact on tumor behavior. Additionally, this review highlights EGFR and PI3K/AKT/mTOR inhibitors currently in clinical and preclinical trials, addressing treatment resistance, challenges, and future directions.
Collapse
Affiliation(s)
- Gursimran Singh
- Department of Pharmacy Practice, ISF College of Pharmacy (an Autonomous College), Moga, Punjab, 142001, India
| | - Rohit
- Research Scholar, I.K. Gujral Punjab Technical University, Kapurthala, Punjab, 144603, India
| | - Pankaj Kumar
- Department of Pharmacology, Himachal Institute of Pharmaceutical Education and Research (HIPER), Tehsil-Nadaun, Hamirpur, Himachal Pradesh, 177033, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy (an Autonomous College), Moga, Punjab, 142001, India.
| |
Collapse
|
21
|
Liang L, Lv W, Cheng G, Gao M, Sun J, Liu N, Zhang H, Guo B, Liu J, Li Y, Xie S, Wang J, Hei J, Zhang J. Impact of celastrol on mitochondrial dynamics and proliferation in glioblastoma. BMC Cancer 2025; 25:412. [PMID: 40050778 PMCID: PMC11887396 DOI: 10.1186/s12885-025-13733-9] [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: 11/17/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Targeting mitochondrial dynamics offers promising strategies for treating glioblastoma multiforme. Celastrol has demonstrated therapeutic effects on various cancers, but its impact on mitochondrial dynamics in glioblastoma multiforme remains largely unknown. We studied the effects of Celastrol on mitochondrial dynamics, redox homeostasis, and the proliferation. METHODS Mito-Tracker Green staining was conducted on U251, LN229, and U87-MG cells to evaluate the effects of Celastrol on mitochondrial dynamics. The Western blot analysis quantified the expression levels of mitochondrial dynamin, antioxidant enzymes, and cell cycle-related proteins. JC-1 staining was performed to discern mitochondrial membrane potential. Mitochondrial reactive oxygen species were identified using MitoSOX. The proliferative capacity of cells was assessed using Cell Counting Kit-8 analysis, and colony formation assays. Survival analysis was employed to evaluate the therapeutic efficacy of Celastrol in C57BL/6J mice with glioblastoma. RESULTS Our findings suggest that Celastrol (1 and 1.5 µM) promotes mitochondrial fission by downregulating the expression of mitofusin-1. A decrease in mitochondrial membrane potential at 1 and 1.5 µM indicates that Celastrol impaired mitochondrial function. Concurrently, an increase in mitochondrial reactive oxygen species and impaired upregulation of antioxidant enzymes were noted at 1.5 µM, indicating that Celastrol led to an imbalance in mitochondrial redox homeostasis. At both 1 and 1.5 µM, cell proliferation was inhibited, which may be related to the decreased expression levels of Cyclin-dependent kinase 1 and Cyclin B1. Celastrol extended the survival of GBM-afflicted mice. CONCLUSION Celastrol promotes mitochondrial fission in glioblastoma multiforme cells by reducing mitofusin-1 expression, accompanying mitochondrial dysfunction, lower mitochondrial membrane potential, heightened oxidative stress, and decreased Cyclin-dependent kinase 1 and Cyclin B1 levels. This indicates that Celastrol possesses potential for repurposing as an agent targeting mitochondrial dynamics in glioblastoma multiforme, warranting further investigation.
Collapse
Affiliation(s)
- Lei Liang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wenying Lv
- Department of Neurosurgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Gang Cheng
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Mou Gao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Junzhao Sun
- Department of Neurosurgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Ning Liu
- Department of Neurosurgery, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100010, China
| | - Hanbo Zhang
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Baorui Guo
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiayu Liu
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanteng Li
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | | | | | - Junru Hei
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianning Zhang
- Medical School of Chinese PLA, Beijing, 100853, China.
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
22
|
Wang L, Gu M, Zhang X, Kong T, Liao J, Zhang D, Li J. Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413367. [PMID: 39854126 PMCID: PMC11905078 DOI: 10.1002/advs.202413367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/14/2024] [Indexed: 01/26/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
Collapse
Affiliation(s)
- Liyin Wang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Min Gu
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Xiaoli Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | | | - Jun Liao
- Institute of Systems BiomedicineBeijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Dan Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Jingwu Li
- The First Hospital of China Medical UniversityLiaoning110001China
| |
Collapse
|
23
|
Gai S, Yan Q, Li S, Zhong X, Qin Y, Jiang M. Lactoferrin Nanoparticle-Vanadium Complex: A Promising High-Efficiency Agent against Glioblastoma by Triggering Autophagy and Ferroptosis. J Med Chem 2025; 68:4650-4662. [PMID: 39945608 DOI: 10.1021/acs.jmedchem.4c02696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Glioblastoma represents the most aggressive type of brain cancer with minimal clinical advancements in recent decades attributed to the absence of efficient drug delivery strategies. In this study, we synthesized a series of vanadium complexes (V1-V4) and then constructed a lactoferrin (LF)-V4 nanoparticle (NP) delivery system. The nanoplatform crossed the blood-brain barrier by binding to low-density lipoprotein receptor-associated protein-1 and selectively targeted glioblastoma, ultimately inhibiting the growth of in situ glioblastoma tumors. LF-V4 NPs induced autophagic cell death in U87-MG cells by generating reactive oxygen species (ROS) that damaged the mitochondria. Further studies revealed that LF-V4 NPs triggered lipid peroxidation through the accumulation of ROS, the depletion of GSH, and the downregulation of GPX4 and SLC7A11, ultimately leading to ferroptosis in glioblastoma cells.
Collapse
Affiliation(s)
- Shuangshuang Gai
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
- Institute for History and Culture of Science and Technology, Guangxi Minzu University, Nanning 530006, China
| | - Qiwei Yan
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Shan Li
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Xuwei Zhong
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Yiming Qin
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Ming Jiang
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| |
Collapse
|
24
|
Ahmed T, Alam KT. Biomimetic Nanoparticle Based Targeted mRNA Vaccine Delivery as a Novel Therapy for Glioblastoma Multiforme. AAPS PharmSciTech 2025; 26:68. [PMID: 39984771 DOI: 10.1208/s12249-025-03065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
The prognosis for patients with glioblastoma multiforme (GBM), an aggressive and deadly brain tumor, is poor due to the limited therapeutic options available. Biomimetic nanoparticles have emerged as a promising vehicle for targeted mRNA vaccine delivery, thanks to recent advances in nanotechnology. This presents a novel treatment method for GBM. This review explores the potential of using biomimetic nanoparticles to improve the specificity and effectiveness of mRNA vaccine against GBM. These nanoparticles can evade immune detection, cross the blood-brain barrier, & deliver mRNA directly to glioma cells by mimicking natural biological structures. This allows glioma cells to produce tumor-specific antigens that trigger strong immune responses against the tumor. This review discusses biomimetic nanoparticle design strategies, which are critical for optimizing transport and ensuring targeted action. These tactics include surface functionalization and encapsulation techniques. It also highlights the ongoing preclinical research and clinical trials that demonstrate the therapeutic advantages and challenges of this strategy. Biomimetic nanoparticles for mRNA vaccine delivery represent a new frontier in GBM treatment, which could impact the management of this deadly disease and improve patient outcomes by integrating cutting-edge nanotechnology with immunotherapy.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Plot 15, Block B, Bashundhara R/A, Dhaka, 1229, Bangladesh.
| | - Kazi Tasnuva Alam
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Plot 15, Block B, Bashundhara R/A, Dhaka, 1229, Bangladesh
| |
Collapse
|
25
|
Liu Y, Wu H, Liang G. Combined Strategies for Nanodrugs Noninvasively Overcoming the Blood-Brain Barrier and Actively Targeting Glioma Lesions. Biomater Res 2025; 29:0133. [PMID: 39911305 PMCID: PMC11794768 DOI: 10.34133/bmr.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 02/07/2025] Open
Abstract
Drugs for tumor treatment face various challenges, including poor solubility, poor stability, short blood half-life, nontargeting ability, and strong toxic side effects. Fortunately, nanodrug delivery systems provide excellent solution to these problems. However, nanodrugs for glioma treatment also face some key challenges including overcoming the blood-brain barrier (BBB) and, specifically, accumulation in glioma lesions. In this review, we systematically summarize the advantages and disadvantages of combined strategies for nanodrugs noninvasively overcoming BBB and actively targeting glioma lesions to achieve effective glioma therapy. Common noninvasive strategies for nanodrugs overcoming the BBB include bypassing the BBB via the nose-to-brain route, opening the tight junction of the BBB by focused ultrasound with microbubbles, and transendothelial cell transport by intact cell loading, ligand decoration, or cell membrane camouflage of nanodrugs. Actively targeting glioma lesions after overcoming the BBB is another key factor helping nanodrugs accurately treat in situ gliomas. This aim can also be achieved by loading nanodrugs into intact cells and modifying ligand or cell membrane fragments on the surface of nanodrugs. Targeting decorated nanodrugs can guarantee precise glioma killing and avoid side effects on normal brain tissues that contribute to the specific recognition of glioma lesions. Furthermore, the challenges and prospects of nanodrugs in clinical glioma treatment are discussed.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| | - Haigang Wu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan Province 475004, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| |
Collapse
|
26
|
Zhang G, Xu Y, Zhou A, Yu Y, Ning X, Bao H. Bioengineered NanoAid synergistically targets inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NANOSCALE 2025; 17:2753-2768. [PMID: 39831463 DOI: 10.1039/d4nr04557b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Through transcriptomic analysis of patient-derived glioblastoma tissues, we identify an overactivation of inflammatory pathways that contribute to the development of a tumor-promoting microenvironment and therapeutic resistance. To address this critical mechanism, we present NanoAid, a biomimetic nanoplatform designed to target inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NanoAid employs macrophage-membrane-liposome hybrids to optimize the delivery of COX-2 inhibitor parecoxib and paclitaxel. By inheriting macrophage characteristics, NanoAid not only efficiently traverses the blood-brain barrier and precisely accumulates within tumors but also enhances cancer cell uptake, thereby improving overall anticancer efficacy. Notably, the combination of parecoxib and paclitaxel effectively disrupts inflammatory pro-tumor processes while inducing a synergistic effect that inhibits tumor growth, overcomes therapeutic resistance, and minimizes adverse effects. This results in substantial tumor growth inhibition and extends the median survival of tumor-bearing mice. Thus, our study bridges clinical insights with fundamental research, potentially revolutionizing tumor therapy paradigms.
Collapse
Affiliation(s)
- Gui Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Yongle Yu
- Medical College of Guangxi University, Nanning 530004, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Hongguang Bao
- Department of Anaesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 211101, China.
| |
Collapse
|
27
|
Cheng W, Duan Z, Chen H, Wang Y, Wang C, Pan Y, Wu J, Wang N, Qu H, Xue X. Macrophage membrane-camouflaged pure-drug nanomedicine for synergistic chemo- and interstitial photodynamic therapy against glioblastoma. Acta Biomater 2025; 193:392-405. [PMID: 39800099 DOI: 10.1016/j.actbio.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Glioblastoma (GBM) persists as a highly fatal malignancy, with current clinical treatments showing minimal progress over years. Interstitial photodynamic therapy (iPDT) holds promise due to its minimally invasive nature and low toxicity but is impeded by poor photosensitizer penetration and inadequate GBM targeting. Here, we developed a biomimetic pure-drug nanomedicine (MM@CT), which co-assembles the photosensitizer chlorin e6 (Ce6) and the first-line chemotherapeutic drug (temozolomide, TMZ) for GBM, then camouflaged with macrophage membranes. This design eliminates the need for traditional excipients, ensuring formulation safety and achieving exceptionally high drug loading with 73.2 %. By leveraging the biomimetic properties of macrophage membranes, MM@CT evades clearance by the mononuclear phagocyte system and can overcome blood circulatory barriers to target intracranial GBM tumors due to its inherent tumor-homing ability. Consequently, this targeted strategy enables precise delivery of TMZ to the tumor site while significantly enhancing Ce6 accumulation within the tumor tissue. Upon intra-tumoral irradiation using an optical fiber, activated Ce6 synergizes with TMZ to exert both cytotoxic effects from chemotherapy and unique advantages from iPDT simultaneously attacking GBM tumors in a dual manner. In subcutaneous and intracranial GBM mouse models, MM@CT exhibits remarkable anti-tumor efficacy with minimal systemic toxicity, emerging as a promising GBM treatment strategy. STATEMENT OF SIGNIFICANCE: Glioblastoma (GBM) remains a formidable brain cancer, posing significant therapeutic challenges due to the presence of the blood-brain barrier (BBB) and tumor heterogeneity. To overcome these obstacles, we have developed MM@CT, a biomimetic nanomedicine with exceptional drug loading efficiency of 73.2 %. MM@CT incorporates the photosensitizer Ce6 and chemotherapy agent TMZ, encapsulated within nanoparticles and camouflaged with macrophage membranes. This innovative design enables efficient BBB penetration, precise tumor targeting, and synergistic application of chemotherapy and photodynamic therapy. Encouragingly, preclinical evaluations have demonstrated substantial antitumor activity with minimal systemic toxicity, positioning MM@CT as a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Wei Cheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiran Duan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanjun Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ning Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haijing Qu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiangdong Xue
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
28
|
Huang Z, Wang M, Chen Y, Tang H, Tang K, Zhao M, Yang W, Zhou Z, Tian J, Xiang W, Li S, Luo Q, Liu L, Zhao Y, Li T, Zhou J, Chen L. Glioblastoma-derived migrasomes promote migration and invasion by releasing PAK4 and LAMA4. Commun Biol 2025; 8:91. [PMID: 39833606 PMCID: PMC11747271 DOI: 10.1038/s42003-025-07526-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Almost all high-grade gliomas, particularly glioblastoma (GBM), are highly migratory and aggressive. Migrasomes are organelles produced by highly migratory cells capable of mediating intercellular communication. Thus, GBM cells may produce migrasomes during migration. However, it remains unclear whether migrasomes can influence GBM migration and invasion. In this study, we observed the presence and formation of migrasomes in GBM cells. We found that expression levels of key migrasome formation factor, tetraspanin 4 (TSPAN4), correlated positively with pathological grade and poor prognosis of GBM based on the databases and clinical samples analysis. Subsequently, we knocked down TSPAN4 and found that GBM cell migration and invasion were significantly inhibited due to the reduced formation of migrasomes. We further confirmed that migrasomes are enriched in extracellular matrix (ECM)-related proteins such as p21-activating kinase 4 (PAK4) and laminin alpha 4 (LAMA4). Our experimental results suggest that migrasomes promote GBM cells migration by releasing such proteins into the extracellular space. Overall, we identified migrasomes in GBM and the molecular mechanisms by which they regulate them, providing potential targets for treating GBM.
Collapse
Affiliation(s)
- Zhe Huang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Ming Wang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Yitian Chen
- Faculty of Health Sciences, University of Macau, Macau, 999078, PR China
| | - Hua Tang
- Department of Neurosurgery, The People's Hospital of Jianyang City, Chengdu, 641400, PR China
| | - Kuo Tang
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Mingkuan Zhao
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Wei Yang
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Zhengjun Zhou
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Junjie Tian
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Wei Xiang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Shenjie Li
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Qinglian Luo
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Luotong Liu
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Yanru Zhao
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China
| | - Tao Li
- Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| | - Jie Zhou
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China.
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China.
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China.
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China.
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China.
- Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, 646000, PR China.
- Neurological diseases and brain function laboratory, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China.
- Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, PR China.
| |
Collapse
|
29
|
Cui Z, Liu X, E T, Lin H, Wang D, Liu Y, Ruan X, Wang P, Liu L, Xue Y. Effect of SNORD113-3/ADAR2 on glycolipid metabolism in glioblastoma via A-to-I editing of PHKA2. Cell Mol Biol Lett 2025; 30:5. [PMID: 39794701 PMCID: PMC11724473 DOI: 10.1186/s11658-024-00680-9] [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: 05/15/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive brain tumor, characterized by its poor prognosis. Glycolipid metabolism is strongly associated with GBM development and malignant behavior. However, the precise functions of snoRNAs and ADARs in glycolipid metabolism within GBM cells remain elusive. The objective of the present study is to delve into the underlying mechanisms through which snoRNAs and ADARs exert regulatory effects on glycolipid metabolism in GBM cells. METHODS RNA immunoprecipitation and RNA pull-down experiments were conducted to verify the homodimerization of ADAR2 by SNORD113-3, and Sanger sequencing and Western blot experiments were used to detect the A-to-I RNA editing of PHKA2 mRNA by ADAR2. Furthermore, the phosphorylation of EBF1 was measured by in vitro kinase assay. Finally, in vivo studies using nude mice confirmed that SNORD113-3 and ADAR2 overexpression, along with PHKA2 knockdown, could suppress the formation of subcutaneous xenograft tumors and improve the outcome of tumor-bearing nude mice. RESULTS We found that PHKA2 in GBM significantly promoted glycolipid metabolism, while SNORD113-3, ADAR2, and EBF1 significantly inhibited glycolipid metabolism. SNORD113-3 promotes ADAR2 protein expression by promoting ADAR2 homodimer formation. ADAR2 mediates the A-to-I RNA editing of PHKA2 mRNA. Mass spectrometry analysis and in vitro kinase testing revealed that PHKA2 phosphorylates EBF1 on Y256, reducing the stability and expression of EBF1. Furthermore, direct binding of EBF1 to PKM2 and ACLY promoters was observed, suggesting the inhibition of their expression by EBF1. These findings suggest the existence of a SNORD113-3/ADAR2/PHKA2/EBF1 pathway that collectively regulates the metabolism of glycolipid and the growth of GBM cells. Finally, in vivo studies using nude mice confirmed that knockdown of PHKA2, along with overexpression of SNORD113-3 and ADAR2, could obviously suppress GBM subcutaneous xenograft tumor formation and improve the outcome of those tumor-bearing nude mice. CONCLUSIONS Herein, we clarified the underlying mechanism involving the SNORD113-3/ADAR2/PHKA2/EBF1 pathway in the regulation of GBM cell growth and glycolipid metabolism. Our results provide a framework for the development of innovative therapeutic interventions to improve the prognosis of patients with GBM.
Collapse
Affiliation(s)
- Zheng Cui
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China
- Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Tiange E
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Hongda Lin
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Xuelei Ruan
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Ping Wang
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Libo Liu
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Yixue Xue
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China.
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
30
|
Horta M, Soares P, Sarmento B, Leite Pereira C, Lima RT. Nanostructured lipid carriers for enhanced batimastat delivery across the blood-brain barrier: an in vitro study for glioblastoma treatment. Drug Deliv Transl Res 2025:10.1007/s13346-024-01775-8. [PMID: 39760929 DOI: 10.1007/s13346-024-01775-8] [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: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Glioblastoma presents a significant treatment challenge due to the blood-brain barrier (BBB) hindering drug delivery, and the overexpression of matrix metalloproteinases (MMPs), which promotes tumor invasiveness. This study introduces a novel nanostructured lipid carrier (NLC) system designed for the delivery of batimastat, an MMP inhibitor, across the BBB and into the glioblastoma microenvironment. The NLCs were functionalized with epidermal growth factor (EGF) and a transferrin receptor-targeting construct to enhance BBB penetration and entrapment within the tumor microenvironment. NLCs were prepared by ultrasonicator-assisted hot homogenization, followed by surface functionalization with EGF and the construct though carbodiimide chemistry. The construct was successfully conjugated with an efficiency of 81%. Two functionalized NLC formulations, fMbat and fNbat, differing in the surfactant amount, were characterized. fMbat had a size of 302 nm, a polydispersity index (PDI) of 0.298, a ζ-potential (ZP) of -27.1 mV and an 85% functionalization efficiency (%FE), whereas fNbat measured 285 nm, with a PDI of 0.249, a ZP of -28.6 mV and a %FE of 92%. Both formulations achieved a drug loading of 0.42 μg/mg. In vitro assays showed that fNbat was cytotoxic and failed to cross the BBB, while fMbat showed cytocompatibility at concentrations 10 times higher than the drug's IC50. Additionally, fMbat inhibited MMP-2 activity between 11 and 62% across different cell lines and achieved a three-fold increase in BBB penetration upon functionalization. Our results suggest that the fMbat formulation has potential for enhancing GB treatment by overcoming current drug delivery limitations and may be combined with other therapeutic strategies for improved 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
- 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
- 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
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116, Gandra, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
- INEB - Instituto 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
- 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
|
31
|
Jing L, Du J, Dong Y, Li L, Tang Z, Liu X, Zhong Y, Yuan M. Targeted delivery strategy of indocyanine green-mitoxantrone loaded liposomes co-modified with BTP-7 and BR2 for the treatment of glioma. Pharm Dev Technol 2025; 30:90-100. [PMID: 39745268 DOI: 10.1080/10837450.2024.2448619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/07/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025]
Abstract
OBJECTIVE This study aims to develop a dual-ligand-modified targeted drug delivery system by integrating photosensitizers and chemotherapeutic drugs to enhance anti-glioma effects. The system is designed to overcome the blood-brain barrier (BBB) that hinders effective drug delivery, increase drug accumulation in glioma cells, and thereby enhance therapeutic efficacy. METHODS Liposomes were prepared using the film dispersion-ammonium sulfate gradient technique, co-loading the photosensitizer indocyanine green (ICG) and the chemotherapeutic drug mitoxantrone (MTO). The conjugation of BTP-7 and BR2 to the liposome surface was achieved using an organic phase reaction method. The stability, dispersibility, particle size, and potential of the modified liposomes were tested. Their ability to penetrate the BBB and accumulate in glioma was evaluated in BBB models and cellular uptake studies. Additionally, the anti-tumor activity of this combination approach was assessed. RESULTS The resulting liposomes demonstrated significant stability and dispersibility, with an average particle size of 142.3 ± 1.8 nm and a potential of -17.6 mV. BBB model and cellular uptake studies indicated that BTP-7/BR2-ICG/MTO-LP could not only penetrate the BBB but also accumulate in glioma, leading to glioma cell necrosis. The anti-tumor activity evaluation showed that this combination approach exhibited a strong tumor-suppressing effect. CONCLUSION The dual-ligand-modified liposomes developed in this study can penetrate the blood-brain barrier and achieve targeted drug delivery in glioma therapy. The combination of BTP-7 and BR2 not only enhances the carrier's penetration ability but also increases intracellular drug accumulation, thereby improving therapeutic efficacy. This novel therapeutic approach, which combines chemotherapy and photothermal response via dual-ligand-modified liposomes delivered to the tumor site, demonstrates the potential to reduce drug-related side effects and improve treatment outcomes.
Collapse
Affiliation(s)
- Lin Jing
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Jingguo Du
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Yichao Dong
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Lili Li
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Zijun Tang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Yonglong Zhong
- Department of Thoracic Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| |
Collapse
|
32
|
Ambele MA, Maebele LT, Mulaudzi TV, Kungoane T, Damane BP. Advances in nano-delivery of phytochemicals for glioblastoma treatment. DISCOVER NANO 2024; 19:216. [PMID: 39718730 DOI: 10.1186/s11671-024-04172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by cellular and molecular diversity. This diversity presents significant challenges for treatment and leads to poor prognosis. Surgery remains the primary treatment of choice for GBMs, but it often results in tumor recurrence due to complex interactions between GBM cells and the peritumoral brain zone. Phytochemicals have shown promising anticancer activity in in-vitro studies and are being investigated as potential treatments for various cancers, including GBM. However, some phytochemicals have failed to translate their efficacy to pre-clinical studies due to limited penetration into the tumor microenvironment, leading to high toxicity. Thus, combining phytochemicals with nanotechnology has emerged as a promising alternative for treating GBM. This review explores the potential of utilizing specific nanoparticles to deliver known anticancer phytochemicals directly to tumor cells. This method has demonstrated potential in overcoming the challenges of the complex GBM microenvironment, including the tight blood-brain barrier while minimizing damage to healthy brain tissue. Therefore, employing this interdisciplinary approach holds significant promise for developing effective phyto-nanomedicines for GBM and improving patient outcomes.
Collapse
Affiliation(s)
- Melvin Anyasi Ambele
- Department of Oral and Maxillofacial Pathology, Faculty of Health Sciences, School of Dentistry, University of Pretoria, P.O. Box 1266, Pretoria, 0001, South Africa.
- Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, P.O. Box 0084, Gezina, South Africa.
| | - Lorraine Tshegofatso Maebele
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Tsholofelo Kungoane
- Department of Oral and Maxillofacial Pathology, Faculty of Health Sciences, School of Dentistry, University of Pretoria, P.O. Box 1266, Pretoria, 0001, South Africa
| | - Botle Precious Damane
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa.
| |
Collapse
|
33
|
Russo M, Martella N, Gargano D, Fantasma F, Marcovecchio C, Russo V, Oliva MA, Segatto M, Saviano G, Di Bartolomeo S, Arcella A. Lavender Essential Oil and Its Terpenic Components Negatively Affect Tumor Properties in a Cell Model of Glioblastoma. Molecules 2024; 29:6044. [PMID: 39770132 PMCID: PMC11676467 DOI: 10.3390/molecules29246044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive form of brain cancer in adults, characterized by extensive growth, a high recurrence rate, and resistance to treatment. Growing research interest is focusing on the biological roles of natural compounds due to their potential beneficial effects on health. Our research aimed to investigate the effects of lavender essential oil (LEO) on a GBM cell model. Chemical characterization using GC-MS analysis indicated that LEO contains several terpenes, compounds that have been found to exhibit anticancer properties by interfering with key cancer-related pathways in several cancer models. By means of cell biology assays, we demonstrated that LEO impairs cell proliferation and migration, and also reduces oxidative stress in U87 cells. We further observed that Terpinen-4-ol, contained in LEO, was capable of reproducing the effects of the oil on GBM cells. Our results suggest that the terpenic molecules present in LEO could be considered valuable allies alongside conventional therapies against GBM.
Collapse
Affiliation(s)
- Miriam Russo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Noemi Martella
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Deborah Gargano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Francesca Fantasma
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Chiara Marcovecchio
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Veronica Russo
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| | - Maria Antonietta Oliva
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Gabriella Saviano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Sabrina Di Bartolomeo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Antonietta Arcella
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| |
Collapse
|
34
|
Aslan TN. Cationic Micelle-like Nanoparticles as the Carrier of Methotrexate for Glioblastoma Treatment. Molecules 2024; 29:5977. [PMID: 39770065 PMCID: PMC11678594 DOI: 10.3390/molecules29245977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
In the present study, ultra-small, magnetic, oleyl amine-coated Fe3O4 nanoparticles were synthesized and stabilized with a cationic ligand, cetyltrimethylammonium bromide, and an anticancer drug, methotrexate, was incorporated into a micelle-like nanoparticle structure for glioblastoma treatment. Nanoparticles were further characterized for their physicochemical properties using spectroscopic methods. Drug incorporation efficiency, drug loading, and drug release profile of the nanoparticles were investigated. According to the results, max incorporation efficiency% of 89.5 was found for 25 µg/mL of methotrexate-loaded nanoparticles. The cumulative amount of methotrexate released reached 40% at physiological pH and 85% at a pH of 5.0 up to 12 h. The toxicity and anticancer efficacy of the nanoparticles were also studied on U87 cancer and L929 cells. IC50 concentration of nanoparticles reduced cell viability to 49% in U87 and 72% in L929 cells. The cellular uptake of nanoparticles was found to be 1.92-fold higher in U87 than in L929 cells. The total apoptosis% in U87 cells was estimated to be ~10-fold higher than what was observed in the L929 cells. Nanoparticles also inhibited the cell motility and prevented the metastasis of U87 cell lines. Overall, designed nanoparticles are a promising controlled delivery system for methotrexate to the cancer cells to achieve better therapeutic outcomes.
Collapse
Affiliation(s)
- Tuğba Nur Aslan
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Necmettin Erbakan University, Konya 42090, Turkey;
- Science and Technology Research and Application Center (BITAM), Necmettin Erbakan University, Konya 42140, Turkey
| |
Collapse
|
35
|
Mafi A, Hedayati N, Kahkesh S, Khoshayand S, Alimohammadi M, Farahani N, Hushmandi K. The landscape of circRNAs in gliomas temozolomide resistance: Insights into molecular pathways. Noncoding RNA Res 2024; 9:1178-1189. [PMID: 39022676 PMCID: PMC11250881 DOI: 10.1016/j.ncrna.2024.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 07/20/2024] Open
Abstract
As the deadliest type of primary brain tumor, gliomas represent a significant worldwide health concern. Circular RNA (circRNA), a unique non-coding RNA molecule, seems to be one of the most alluring target molecules involved in the pathophysiology of many kinds of cancers. CircRNAs have been identified as prospective targets and biomarkers for the diagnosis and treatment of numerous disorders, particularly malignancies. Recent research has established a clinical link between temozolomide (TMZ) resistance and certain circRNA dysregulations in glioma tumors. CircRNAs may play a therapeutic role in controlling or overcoming TMZ resistance in gliomas and may provide guidance for a novel kind of individualized glioma therapy. To address the biological characteristics of circRNAs and their potential to induce resistance to TMZ, this review has highlighted and summarized the possible roles that circRNAs may play in molecular pathways of drug resistance, including the Ras/Raf/ERK PI3K/Akt signaling pathway and metabolic processes in gliomas.
Collapse
Affiliation(s)
- Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sara Khoshayand
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
36
|
Zhang S, Jiang X, Wei Q, Huang L, Huang Z, Zhang L. RAB32 promotes glioma cell progression by activating the JAK/STAT3 signaling pathway. J Int Med Res 2024; 52:3000605241282384. [PMID: 39628429 PMCID: PMC11615995 DOI: 10.1177/03000605241282384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/22/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the role of RAB32 in glioblastomas and its molecular mechanisms that regulate gliomas. METHODS The expression and prognostic value of RAB32 were evaluated using western blotting and the Gene Expression Profiling Interactive, Chinese Glioma Genome Atlas, and The Cancer Genome Atlas databases. Lentivirus containing sh-RAB32 or OE-RAB32 was used to manipulate RAB32 expression in glioma cells. The effects of RAB32 on cell proliferation, migration, and invasion were determined by western blotting, cell counting kit-8, plate cloning, wound healing, and transwell assays. Gene set enrichment analysis was used to screen for associations between the JAK/STAT3 signaling pathway and RAB32. The role of this pathway was verified using JAK/STAT3 inhibitors. RESULTS RAB32 expression was significantly upregulated in patients with glioma and in glioma cell lines. The expression level was positively correlated with the glioma grade and served as an independent prognostic factor. In vitro experiments revealed that RAB32 knockdown inhibited glioblastoma cell proliferation, migration, and invasion, while the opposite effects were observed with overexpression and could be inhibited by the JAK/STAT3 inhibitor BP-1-102. CONCLUSION RAB32 promotes malignant progression of glioblastoma cells through the JAK/STAT signaling pathway, providing new possibilities for therapeutic targets for glioblastoma.
Collapse
Affiliation(s)
- Sinan Zhang
- Jiamusi University, Jiamusi, Heilongjiang, China
- Department of Laboratory Medicine, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Xudong Jiang
- Department of Laboratory Medicine, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
- Harbin Medical University (Daqing), Daqing, Heilongjiang, China
| | - Qing Wei
- Jiamusi University, Jiamusi, Heilongjiang, China
| | - Liji Huang
- Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou, Guangxi, China
| | - Zhuoyan Huang
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Lina Zhang
- Department of Laboratory Medicine, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| |
Collapse
|
37
|
Kim KS, Habashy K, Gould A, Zhao J, Najem H, Amidei C, Saganty R, Arrieta VA, Dmello C, Chen L, Zhang DY, Castro B, Billingham L, Levey D, Huber O, Marques M, Savitsky DA, Morin BM, Muzzio M, Canney M, Horbinski C, Zhang P, Miska J, Padney S, Zhang B, Rabadan R, Phillips JJ, Butowski N, Heimberger AB, Hu J, Stupp R, Chand D, Lee-Chang C, Sonabend AM. Fc-enhanced anti-CTLA-4, anti-PD-1, doxorubicin, and ultrasound-mediated blood-brain barrier opening: A novel combinatorial immunotherapy regimen for gliomas. Neuro Oncol 2024; 26:2044-2060. [PMID: 39028616 PMCID: PMC11534315 DOI: 10.1093/neuonc/noae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Glioblastoma is a highly aggressive brain cancer that is resistant to conventional immunotherapy strategies. Botensilimab, an Fc-enhanced anti-CTLA-4 antibody (FcE-aCTLA-4), has shown durable activity in "cold" and immunotherapy-refractory cancers. METHODS We evaluated the efficacy and immune microenvironment phenotype of a mouse analogue of FcE-aCTLA-4 in treatment-refractory preclinical models of glioblastoma, both as a monotherapy and in combination with doxorubicin delivered via low-intensity pulsed ultrasound and microbubbles (LIPU/MB). Additionally, we studied 4 glioblastoma patients treated with doxorubicin, anti-PD-1 with concomitant LIPU/MB to investigate the novel effect of doxorubicin modulating FcγR expressions in tumor-associated macrophages/microglia (TAMs). RESULTS FcE-aCTLA-4 demonstrated high-affinity binding to FcγRIV, the mouse ortholog of human FcγRIIIA, which was highly expressed in TAMs in human glioblastoma, most robustly at diagnosis. Notably, FcE-aCTLA-4-mediated selective depletion of intratumoral regulatory T cells (Tregs) via TAM-mediated phagocytosis, while sparing peripheral Tregs. Doxorubicin, a chemotherapeutic drug with immunomodulatory functions, was found to upregulate FcγRIIIA on TAMs in glioblastoma patients who received doxorubicin and anti-PD-1 with concomitant LIPU/MB. In murine models of immunotherapy-resistant gliomas, a combinatorial regimen of FcE-aCTLA-4, anti-PD-1, and doxorubicin with LIPU/MB, achieved a 90% cure rate, that was associated robust infiltration of activated CD8+ T cells and establishment of immunological memory as evidenced by rejection upon tumor rechallenge. CONCLUSIONS Our findings demonstrate that FcE-aCTLA-4 promotes robust immunomodulatory and anti-tumor effects in murine gliomas and is significantly enhanced when combined with anti-PD-1, doxorubicin, and LIPU/MB. We are currently investigating this combinatory strategy in a clinical trial (clinicaltrials.gov NCT05864534).
Collapse
Affiliation(s)
- Kwang-Soo Kim
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Karl Habashy
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andrew Gould
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Junfei Zhao
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, New York, USA
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Hinda Najem
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christina Amidei
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ruth Saganty
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Víctor A Arrieta
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Crismita Dmello
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Li Chen
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel Y Zhang
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brandyn Castro
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leah Billingham
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | - Miguel Muzzio
- Life Science Group, IIT Research Institute (IITRI), Chicago, Illinois, USA
| | | | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peng Zhang
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason Miska
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Surya Padney
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bin Zhang
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raul Rabadan
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, New York, USA
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Joanna J Phillips
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Amy B Heimberger
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jian Hu
- Division of Basic Science Research, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roger Stupp
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dhan Chand
- Agenus Inc., Lexington, Massachusetts, USA
| | - Catalina Lee-Chang
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adam M Sonabend
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
38
|
Chang X, Liu J, Li Y, Li W. Pluronic F127-Complexed PEGylated Poly(glutamic acid)-Cisplatin Nanomedicine for Enhanced Glioblastoma Therapy. Macromol Rapid Commun 2024; 45:e2400662. [PMID: 39264576 DOI: 10.1002/marc.202400662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Glioblastoma is one of the most aggressive and treatment-resistant forms of primary brain cancer, posing significant challenges in effective therapy. This study aimed to enhance the effectiveness of glioblastoma therapy by developing a unique nanomedicine composed of Pluronic F127-complexed PEGylated poly(glutamic acid)-cisplatin (PLG-PEG/PF127-CDDP). PLG-PEG/PF127-CDDP demonstrated an optimal size of 133.97 ± 12.60 nm, facilitating efficient cell uptake by GL261 glioma cells. In vitro studies showed significant cytotoxicity against glioma cells with a half-maximal (50%) inhibitory concentration (IC50) of 12.61 µg mL-1 at 48 h and a 72.53% ± 1.89% reduction in cell invasion. Furthermore, PLG-PEG/PF127-CDDP prolonged the circulation half-life of cisplatin to 9.75 h in vivo, leading to a more than 50% reduction in tumor size on day 16 post-treatment initiation in a murine model of glioma. The treatment significantly elevated lactate levels in GL261 cells, indicating enhanced metabolic disruption. Therefore, PLG-PEG/PF127-CDDP offers a promising approach for glioblastoma therapy due to its effects on improving drug delivery efficiency, therapeutic outcomes, and safety while minimizing systemic side effects. This work underscores the potential of polymer-based nanomedicines in overcoming the challenges of treating brain tumors, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Xiaoyu Chang
- Department of Neurosurgery, the First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Jiaxue Liu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, 5 Jilin Street, Jilin, 132000, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Yunqian Li
- Department of Neurosurgery, the First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Wenliang Li
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, 5 Jilin Street, Jilin, 132000, P. R. China
| |
Collapse
|
39
|
Ismail M, Wang Y, Li Y, Liu J, Zheng M, Zou Y. Stimuli-Responsive Polymeric Nanocarriers Accelerate On-Demand Drug Release to Combat Glioblastoma. Biomacromolecules 2024; 25:6250-6282. [PMID: 39259212 DOI: 10.1021/acs.biomac.4c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with a poor prognosis and limited treatment options. Drug delivery by stimuli-responsive nanocarriers holds great promise for improving the treatment modalities of GBM. At the beginning of the review, we highlighted the stimuli-active polymeric nanocarriers carrying therapies that potentially boost anti-GBM responses by employing endogenous (pH, redox, hypoxia, enzyme) or exogenous stimuli (light, ultrasonic, magnetic, temperature, radiation) as triggers for controlled drug release mainly via hydrophobic/hydrophilic transition, degradability, ionizability, etc. Modifying these nanocarriers with target ligands further enhanced their capacity to traverse the blood-brain barrier (BBB) and preferentially accumulate in glioma cells. These unique features potentially lead to more effective brain cancer treatment with minimal adverse reactions and superior therapeutic outcomes. Finally, the review summarizes the existing difficulties and future prospects in stimuli-responsive nanocarriers for treating GBM. Overall, this review offers theoretical guidelines for developing intelligent and versatile stimuli-responsive nanocarriers to facilitate precise drug delivery and treatment of GBM in clinical settings.
Collapse
Affiliation(s)
- Muhammad Ismail
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan Zou
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
40
|
Cunha Silva L, Branco F, Cunha J, Vitorino C, Gomes C, Carrascal MA, Falcão A, Miguel Neves B, Teresa Cruz M. The potential of exosomes as a new therapeutic strategy for glioblastoma. Eur J Pharm Biopharm 2024; 203:114460. [PMID: 39218361 DOI: 10.1016/j.ejpb.2024.114460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 07/30/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) stands for the most common and aggressive type of brain tumour in adults. It is highly invasive, which explains its short rate of survival. Little is known about its risk factors, and current therapy is still ineffective. Hence, efforts are underway to develop novel and effective treatment approaches against this type of cancer. Exosomes are being explored as a promising strategy for conveying and delivering therapeutic cargo to GBM cells. They can fuse with the GBM cell membrane and, consequently, serve as delivery systems in this context. Due to their nanoscale size, exosomes can cross the blood-brain barrier (BBB), which constitutes a significant hurdle to most chemotherapeutic drugs used against GBM. They can subsequently inhibit oncogenes, activate tumour suppressor genes, induce immune responses, and control cell growth. However, despite representing a promising tool for the treatment of GBM, further research and clinical studies regarding exosome biology, engineering, and clinical applications still need to be completed. Here, we sought to review the application of exosomes in the treatment of GBM through an in-depth analysis of the scientific and clinical studies on the entire process, from the isolation and purification of exosomes to their design and transformation into anti-oncogenic drug delivery systems. Surface modification of exosomes to enhance BBB penetration and GBM-cell targeting is also a topic of discussion.
Collapse
Affiliation(s)
- Leonor Cunha Silva
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Francisco Branco
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3004 535, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, Coimbra 3000-504, Portugal
| | - Mylène A Carrascal
- Tecnimede Group, Sintra 2710-089, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, Coimbra 3000-548, Portugal
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal.
| |
Collapse
|
41
|
Gerlach SL, Metcalf JS, Dunlop RA, Banack SA, Her C, Krishnan VV, Göransson U, Gunasekera S, Slazak B, Cox PA. Kalata B1 Enhances Temozolomide Toxicity to Glioblastoma Cells. Biomedicines 2024; 12:2216. [PMID: 39457529 PMCID: PMC11505038 DOI: 10.3390/biomedicines12102216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 10/28/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive cancer originating in the brain, but unfortunately combination treatments with resection, radiation, and chemotherapy are relatively ineffective. Therefore, novel methods of adjuvant therapy are critically needed. Cyclotides are plant-derived circular peptides that chemosensitize drug-resistant breast cancer to doxorubicin. We analyzed naturally occurring and synthetic cyclotides (Cycloviolacin O3, Cycloviolacin O19, natural Kalata B1, synthetic Kalata B1, and Vitri E) alone and in co-exposure treatments with the drug temozolomide (TMZ) in human glioblastoma cells. The cyclotides were identified by UPLC-PDA and HPLC-UV. The synthetic Kalata B1 sequence was verified with orbitrap LC-MS, and structural confirmation was provided by NMR spectroscopy. The cyclotides displayed dose-dependent cytotoxicity (IC50 values 2.4-21.1 µM) both alone and as chemosensitizers of U-87 MG and T 98 cells to TMZ. In fact, a 16-fold lower concentration of TMZ (100 µM) was needed for significant cytotoxicity in U-87 MG cells co-exposed to synthetic Kalata B (0.5 µM). Similarly, a 15-fold lower concentration of TMZ (75 µM) was required for a significant reduction in cell viability in T 98 cells co-exposed to synthetic Kalata B1 (0.25 µM). Kalata B1 remained stable in human serum stability assays. The data support the assertion that cyclotides may chemosensitize glioblastoma cells to TMZ.
Collapse
Affiliation(s)
- Samantha L. Gerlach
- Department of Biology, Dillard University, New Orleans, LA 70122, USA
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY 83001, USA or (J.S.M.); (R.A.D.); (S.A.B.)
| | - James S. Metcalf
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY 83001, USA or (J.S.M.); (R.A.D.); (S.A.B.)
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA
| | - Rachael A. Dunlop
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY 83001, USA or (J.S.M.); (R.A.D.); (S.A.B.)
| | - Sandra Anne Banack
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY 83001, USA or (J.S.M.); (R.A.D.); (S.A.B.)
| | - Cheenou Her
- Department of Chemistry and Biochemistry, University of California, San Diego, CA 92093, USA;
- Department of Chemistry and Biochemistry, California State University, Fresno, CA 93740, USA;
| | - Viswanathan V. Krishnan
- Department of Chemistry and Biochemistry, California State University, Fresno, CA 93740, USA;
- Department of Medical Pathology and Laboratory Medicine, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Ulf Göransson
- Pharmacognosy, Department of Pharmaceutical Biosciences, Uppsala University, Box 574, 751 23 Uppsala, Sweden; (U.G.); (S.G.); (B.S.)
| | - Sunithi Gunasekera
- Pharmacognosy, Department of Pharmaceutical Biosciences, Uppsala University, Box 574, 751 23 Uppsala, Sweden; (U.G.); (S.G.); (B.S.)
| | - Blazej Slazak
- Pharmacognosy, Department of Pharmaceutical Biosciences, Uppsala University, Box 574, 751 23 Uppsala, Sweden; (U.G.); (S.G.); (B.S.)
- W. Szafer Institute of Botany, Polish Academy of Sciences, 46 Lubicz, 31-512 Cracow, Poland
| | - Paul Alan Cox
- Brain Chemistry Labs, Institute for Ethnomedicine, Jackson, WY 83001, USA or (J.S.M.); (R.A.D.); (S.A.B.)
| |
Collapse
|
42
|
Shin S, Jo H, Agura T, Jeong S, Ahn H, Kim Y, Kang JS. Use of surface-modified porous silicon nanoparticles to deliver temozolomide with enhanced pharmacokinetic and therapeutic efficacy for intracranial glioblastoma in mice. J Mater Chem B 2024; 12:9335-9344. [PMID: 39171683 DOI: 10.1039/d4tb00631c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Glioblastoma (GBM) is one of the most common and fatal primary brain tumors, with a 5-year survival rate of 7.2%. The standard treatment for GBM involves surgical resection followed by chemoradiotherapy, and temozolomide (TMZ) is currently the only approved chemotherapeutic agent for the treatment of GBM. However, hydrolytic instability and insufficient drug accumulation are major challenges that limit the effectiveness of TMZ chemotherapy. To overcome these limitations, we have developed a drug delivery platform utilizing porous silicon nanoparticles (pSiNPs) to improve the stability and blood-brain barrier penetration of TMZ. The pSiNPs are synthesized via electrochemical etching and functionalized with octadecane. The octadecyl-modified pSiNP (pSiNP-C18) demonstrates the superiority of loading efficiency, in vivo stability, and brain accumulation of TMZ. Treatment of intracranial tumor-bearing mice with TMZ-loaded pSiNP-C18 results in a decreased tumor burden and a corresponding increase in survival compared with equivalent free-drug dosing. Furthermore, the mice treated with TMZ-loaded nanoparticles do not exhibit in vivo toxicity, thus underscoring the preclinical potential of the pSiNP-based platform for the delivery of therapeutic agents to gliomas.
Collapse
Affiliation(s)
- Seulgi Shin
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
- Department of Research and Development, N therapeutics Co., Ltd, Seoul 08813, Republic of Korea
| | - Hyejung Jo
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Tomoyo Agura
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Seoyoun Jeong
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Hyovin Ahn
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Yejin Kim
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Jae Seung Kang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
43
|
Meena D, Jha S. Autophagy in glioblastoma: A mechanistic perspective. Int J Cancer 2024; 155:605-617. [PMID: 38716809 DOI: 10.1002/ijc.34991] [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: 01/29/2024] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 06/20/2024]
Abstract
Glioblastoma (GBM) is one of the most lethal malignancies in humans. Even after surgical resection and aggressive radio- or chemotherapies, patients with GBM can survive for less than 14 months. Extreme inter-tumor and intra-tumor heterogeneity of GBM poses a challenge for resolving recalcitrant GBM pathophysiology. GBM tumor microenvironment (TME) exhibits diverse heterogeneity in cellular composition and processes contributing to tumor progression and therapeutic resistance. Autophagy is such a cellular process; that demonstrates a cell-specific and TME context-dependent role in GBM progression, leading to either the promotion or suppression of GBM progression. Autophagy can regulate GBM cell function directly via regulation of survival, migration, and invasion, or indirectly by affecting GBM TME composition such as immune cell population, tumor metabolism, and glioma stem cells. This review comprehensively investigates the role of autophagy in GBM pathophysiology.
Collapse
Affiliation(s)
- Durgesh Meena
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sushmita Jha
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
44
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
45
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
46
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
47
|
Sharapova G, Sabirova S, Gomzikova M, Brichkina A, Barlev NA, Kalacheva NV, Rizvanov A, Markov N, Simon HU. Mitochondrial Protein Density, Biomass, and Bioenergetics as Predictors for the Efficacy of Glioma Treatments. Int J Mol Sci 2024; 25:7038. [PMID: 39000148 PMCID: PMC11241254 DOI: 10.3390/ijms25137038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The metabolism of glioma cells exhibits significant heterogeneity and is partially responsible for treatment outcomes. Given this variability, we hypothesized that the effectiveness of treatments targeting various metabolic pathways depends on the bioenergetic profiles and mitochondrial status of glioma cells. To this end, we analyzed mitochondrial biomass, mitochondrial protein density, oxidative phosphorylation (OXPHOS), and glycolysis in a panel of eight glioma cell lines. Our findings revealed considerable variability: mitochondrial biomass varied by up to 3.2-fold, the density of mitochondrial proteins by up to 2.1-fold, and OXPHOS levels by up to 7.3-fold across the cell lines. Subsequently, we stratified glioma cell lines based on their mitochondrial status, OXPHOS, and bioenergetic fitness. Following this stratification, we utilized 16 compounds targeting key bioenergetic, mitochondrial, and related pathways to analyze the associations between induced changes in cell numbers, proliferation, and apoptosis with respect to their steady-state mitochondrial and bioenergetic metrics. Remarkably, a significant fraction of the treatments showed strong correlations with mitochondrial biomass and the density of mitochondrial proteins, suggesting that mitochondrial status may reflect glioma cell sensitivity to specific treatments. Overall, our results indicate that mitochondrial status and bioenergetics are linked to the efficacy of treatments targeting metabolic pathways in glioma.
Collapse
Affiliation(s)
- Gulnaz Sharapova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (G.S.); (S.S.); (M.G.); (A.B.); (N.A.B.)
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (N.V.K.); (A.R.)
| | - Sirina Sabirova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (G.S.); (S.S.); (M.G.); (A.B.); (N.A.B.)
- Laboratory of Intercellular Communication, Kazan Federal University, 420111 Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (G.S.); (S.S.); (M.G.); (A.B.); (N.A.B.)
- Laboratory of Intercellular Communication, Kazan Federal University, 420111 Kazan, Russia
| | - Anna Brichkina
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (G.S.); (S.S.); (M.G.); (A.B.); (N.A.B.)
- Institute of Systems Immunology, Center for Tumor Biology and Immunology, Philipps University of Marburg, 35043 Marburg, Germany
| | - Nick A Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (G.S.); (S.S.); (M.G.); (A.B.); (N.A.B.)
- Gene Expression Program, Institute of Cytology RAS, 194064 Saint-Petersburg, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Natalia V Kalacheva
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (N.V.K.); (A.R.)
| | - Albert Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (N.V.K.); (A.R.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
- I.K. Akhunbaev Kyrgyz State Medical Academy, Bishkek 720020, Kyrgyzstan
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Hans-Uwe Simon
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (G.S.); (S.S.); (M.G.); (A.B.); (N.A.B.)
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| |
Collapse
|
48
|
Lai G, Wu H, Yang K, Hu K, Zhou Y, Chen X, Fu F, Li J, Xie G, Wang HF, Lv Z, Wu X. Progress of nanoparticle drug delivery system for the treatment of glioma. Front Bioeng Biotechnol 2024; 12:1403511. [PMID: 38919382 PMCID: PMC11196769 DOI: 10.3389/fbioe.2024.1403511] [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: 03/19/2024] [Accepted: 05/17/2024] [Indexed: 06/27/2024] Open
Abstract
Gliomas are typical malignant brain tumours affecting a wide population worldwide. Operation, as the common treatment for gliomas, is always accompanied by postoperative drug chemotherapy, but cannot cure patients. The main challenges are chemotherapeutic drugs have low blood-brain barrier passage rate and a lot of serious adverse effects, meanwhile, they have difficulty targeting glioma issues. Nowadays, the emergence of nanoparticles (NPs) drug delivery systems (NDDS) has provided a new promising approach for the treatment of gliomas owing to their excellent biodegradability, high stability, good biocompatibility, low toxicity, and minimal adverse effects. Herein, we reviewed the types and delivery mechanisms of NPs currently used in gliomas, including passive and active brain targeting drug delivery. In particular, we primarily focused on various hopeful types of NPs (such as liposome, chitosan, ferritin, graphene oxide, silica nanoparticle, nanogel, neutrophil, and adeno-associated virus), and discussed their advantages, disadvantages, and progress in preclinical trials. Moreover, we outlined the clinical trials of NPs applied in gliomas. According to this review, we provide an outlook of the prospects of NDDS for treating gliomas and summarise some methods that can enhance the targeting specificity and safety of NPs, like surface modification and conjugating ligands and peptides. Although there are still some limitations of these NPs, NDDS will offer the potential for curing glioma patients.
Collapse
Affiliation(s)
- Guogang Lai
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hao Wu
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaixia Yang
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaikai Hu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yan Zhou
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiao Chen
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Fan Fu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiayi Li
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guomin Xie
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hai-Feng Wang
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zhongyue Lv
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiping Wu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
49
|
Luo C, He J, Yang Y, Wu K, Fu X, Cheng J, Ming Y, Liu W, Peng Y. SRSF9 promotes cell proliferation and migration of glioblastoma through enhancing CDK1 expression. J Cancer Res Clin Oncol 2024; 150:292. [PMID: 38842611 PMCID: PMC11156731 DOI: 10.1007/s00432-024-05797-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive and prevalent brain tumor that poses significant challenges in treatment. SRSF9, an RNA-binding protein, is essential for cellular processes and implicated in cancer progression. Yet, its function and mechanism in GBM need clarification. METHODS Bioinformatics analysis was performed to explore differential expression of SRSF9 in GBM and its prognostic relevance to glioma patients. SRSF9 and CDK1 expression in GBM cell lines and patients' tissues were quantified by RT-qPCR, Western blot or immunofluorescence assay. The role of SRSF9 in GBM cell proliferation and migration was assessed by MTT, Transwell and colony formation assays. Additionally, transcriptional regulation of CDK1 by SRSF9 was investigated using ChIP-PCR and dual-luciferase assays. RESULTS The elevated SRSF9 expression correlates to GBM stages and poor survival of glioma patients. Through gain-of-function and loss-of-function strategies, SRSF9 was demonstrated to promote proliferation and migration of GBM cells. Bioinformatics analysis showed that SRSF9 has an impact on cell growth pathways including cell cycle checkpoints and E2F targets. Mechanistically, SRSF9 appears to bind to the promoter of CDK1 gene and increase its transcription level, thus promoting GBM cell proliferation. CONCLUSIONS These findings uncover the cellular function of SRSF9 in GBM and highlight its therapeutic potential for GBM.
Collapse
Affiliation(s)
- Chunyuan Luo
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Juan He
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yang Yang
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Wu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Fu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Cheng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Ming
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenrong Liu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
50
|
Niu H, Cao H, Liu X, Chen Y, Cheng Z, Long J, Li F, Sun C, Zuo P. The Significance of the Redox Gene in the Prognosis and Therapeutic Response of Glioma. Am J Clin Oncol 2024; 47:259-270. [PMID: 38318849 DOI: 10.1097/coc.0000000000001086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
OBJECTIVES Glioblastoma (GBM) is a fatal adult central nervous system tumor. Due to its high heterogeneity, the survival rate and prognosis of patients are poor. Thousands of people die of this disease every year all over the world. At present, the treatment of GBM is mainly through surgical resection and the combination of later drugs, radiotherapy, and chemotherapy. An abnormal redox system is involved in the malignant progression and treatment tolerance of glioma, which is the main reason for poor survival and prognosis. The construction of a GBM redox-related prognostic model may be helpful in improving the redox immunotherapy and prognosis of GBM. METHODS Based on glioma transcriptome data and clinical data from The Cancer Genome Atlas, databases, a risk model of redox genes was constructed by univariate and multivariate Cox analysis. The good prediction performance of the model was verified by the internal validation set of The Cancer Genome Atlas, and the external data of Chinese Glioma Genome Atlas. RESULTS The results confirmed that the higher the risk score, the worse the survival of patients. Age and isocitrate dehydrogenase status were significantly correlated with risk scores. The analysis of immune infiltration and immunotherapy found that there were significant differences in the immune score, matrix score, and ESTIMATE score between high and low-risk groups. reverse transcription polymerase chain reaction and immunohistochemical staining of glioma samples confirmed the expression of the hub gene. CONCLUSION Our study suggests that the 5 oxidative-related genes nitricoxidesynthase3 , NCF2 , VASN , FKBP1B , and TXNDC2 are hub genes, which may provide a reliable prognostic tool for glioma clinical treatment.
Collapse
Affiliation(s)
| | | | - Xin Liu
- Department of Molecular Diagnosis Center, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming
| | | | | | - Jinyong Long
- Department of Surgery, Jinping County People's Hospital, Jinping
| | - Fuhua Li
- Department of Surgery, Jinping County People's Hospital, Jinping
| | - Chaoyan Sun
- Department of Emergency, Zhoukou Central Hospital, Zhoukou, China
| | | |
Collapse
|