1
|
Tataranu LG, Turliuc S, Kamel A, Rizea RE, Dricu A, Staicu GA, Baloi SC, Rodriguez SMB, Manole AIM. Glioblastoma Tumor Microenvironment: An Important Modulator for Tumoral Progression and Therapy Resistance. Curr Issues Mol Biol 2024; 46:9881-9894. [PMID: 39329940 PMCID: PMC11430601 DOI: 10.3390/cimb46090588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The race to find an effective treatment for glioblastoma (GBM) remains a critical topic, because of its high aggressivity and impact on survival and the quality of life. Currently, due to GBM's high heterogeneity, the conventional treatment success rate and response to therapy are relatively low, with a median survival rate of less than 20 months. A new point of view can be provided by the comprehension of the tumor microenvironment (TME) in pursuance of the development of new therapeutic strategies to aim for a longer survival rate with an improved quality of life and longer disease-free interval (DFI). The main components of the GBM TME are represented by the extracellular matrix (ECM), glioma cells and glioma stem cells (GSCs), immune cells (microglia, macrophages, neutrophils, lymphocytes), neuronal cells, all of them having dynamic interactions and being able to influence the tumoral growth, progression, and drug resistance thus being a potential therapeutic target. This paper will review the latest research on the GBM TME and the potential therapeutic targets to form an up-to-date strategy.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Neurosurgical Department, University of Medicine and Pharmacy "Carol Davila", 020022 Bucharest, Romania
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy "G. T. Popa", 700115 Iasi, Romania
| | - Amira Kamel
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Radu Eugen Rizea
- Neurosurgical Department, University of Medicine and Pharmacy "Carol Davila", 020022 Bucharest, Romania
- Neurosurgical Department, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | | | - Stefania Carina Baloi
- Biochemistry Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | | | | |
Collapse
|
2
|
Li C, Zhou W, Wang P, Ji P, Wang Y, Guo S, Zhai Y, Xu M, Wang L, Feng F, Liu J. Prognostic Factors for Patients with Primary Gliosarcoma: A Single-Center Retrospective Study. World Neurosurg 2024:S1878-8750(24)01497-9. [PMID: 39216722 DOI: 10.1016/j.wneu.2024.08.128] [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: 06/17/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Primary gliosarcoma is a rare form of malignant central nervous system tumor, with limited understanding regarding its prognostic determinants and effective therapeutic interventions. METHODS The medical records of patients diagnosed with gliosarcoma at Tangdu Hospital between March 2011 and June 2023 were retrospectively analyzed in this study. Patients with a prior history of glioma or those who received preoperative chemoradiotherapy were excluded. Survival analyses were conducted using Kaplan-Meier and Cox regression analysis. RESULTS A total of 77 patients were included in the final analysis, with a median age of 57 years (range: 13-83). The predominant symptom leading to diagnosis was headache, and the temporal lobe was the most frequently affected site. Univariate analysis revealed that age ≤65 years, complete resection, Ki67 ≤ 25%, postoperative Karnofsky Performance Status ≥ 70, adherence to the Stupp protocol, and additional active therapy upon relapse were associated with enhanced survival. Furthermore, multivariate analysis identified complete resection, age ≤65 years, Stupp protocol treatment, and active therapy following relapse were independent predictors of overall survival. Notably, 1 patient experienced subcutaneous metastasis during treatment. CONCLUSIONS The present study's findings suggest that optimal management of primary gliosarcoma entails maximal safe resection, combined with adjuvant radiotherapy and chemotherapy with temozolomide, followed by salvage therapy in case of recurrence. However, the risk of metastases should be carefully monitored during the treatment course.
Collapse
Affiliation(s)
- Chen Li
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China
| | - Wenqian Zhou
- The Fourth Student Brigade of Basic Medical College, Air Force Medical University, Xian, China
| | - Peng Wang
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peigang Ji
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China
| | - Yuan Wang
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China
| | - Shaochun Guo
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China
| | - Yulong Zhai
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China
| | - Meng Xu
- Evidence-Based Social Sciences Research Centre, School of Public Health, Lanzhou University, Lanzhou, China
| | - Liang Wang
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China
| | - Fuqiang Feng
- Department of Neurosurgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jinghui Liu
- Department of Neurosurgery, The Second Affiliated Hospita of Air Force Medical University, Xian, China.
| |
Collapse
|
3
|
Zhang Y, Ma H, Li L, Sun C, Yu C, Wang L, Xu D, Song X, Yu R. Dual-Targeted Novel Temozolomide Nanocapsules Encapsulating siPKM2 Inhibit Aerobic Glycolysis to Sensitize Glioblastoma to Chemotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400502. [PMID: 38651254 DOI: 10.1002/adma.202400502] [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: 01/10/2024] [Revised: 03/07/2024] [Indexed: 04/25/2024]
Abstract
Chemotherapy of glioblastoma (GBM) has not yielded success due to inefficient blood-brain barrier (BBB) penetration and poor glioma tissue accumulation. Aerobic glycolysis, as the main mode of energy supply for GBM, safeguards the rapid growth of GBM while affecting the efficacy of radiotherapy and chemotherapy. Therefore, to effectively inhibit aerobic glycolysis, increase drug delivery efficiency and sensitivity, a novel temozolomide (TMZ) nanocapsule (ApoE-MT/siPKM2 NC) is successfully designed and prepared for the combined delivery of pyruvate kinase M2 siRNA (siPKM2) and TMZ. This drug delivery platform uses siPKM2 as the inner core and methacrylate-TMZ (MT) as the shell component to achieve inhibition of glioma energy metabolism while enhancing the killing effect of TMZ. By modifying apolipoprotein E (ApoE), dual targeting of the BBB and GBM is achieved in a "two birds with one stone" style. The glutathione (GSH) responsive crosslinker containing disulfide bonds ensures "directional blasting" cleavage of the nanocapsules to release MT and siPKM2 in the high GSH environment of glioma cells. In addition, in vivo experiments verify that ApoE-MT/siPKM2 NC has good targeting ability and prolongs the survival of tumor-bearing nude mice. In summary, this drug delivery system provides a new strategy for metabolic therapy sensitization chemotherapy.
Collapse
Affiliation(s)
- Yongkang Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Hongwei Ma
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Linsen Li
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Chen Sun
- Department of Nephrology, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Changshui Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Lansheng Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Duo Xu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Xu Song
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| |
Collapse
|
4
|
Cao X, Li J, Ren J, Peng J, Zhong R, He J, Xu T, Yu Z, Jin H, Hao S, Liu R, Xu B. Minimally-invasive implantable device enhances brain cancer suppression. EMBO Mol Med 2024; 16:1704-1716. [PMID: 38902433 PMCID: PMC11250787 DOI: 10.1038/s44321-024-00091-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Current brain tumor treatments are limited by the skull and BBB, leading to poor prognosis and short survival for glioma patients. We introduce a novel minimally-invasive brain tumor suppression (MIBTS) device combining personalized intracranial electric field therapy with in-situ chemotherapeutic coating. The core of our MIBTS technique is a wireless-ultrasound-powered, chip-sized, lightweight device with all functional circuits encapsulated in a small but efficient "Swiss-roll" structure, guaranteeing enhanced energy conversion while requiring tiny implantation windows ( ~ 3 × 5 mm), which favors broad consumers acceptance and easy-to-use of the device. Compared with existing technologies, competitive advantages in terms of tumor suppressive efficacy and therapeutic resolution were noticed, with maximum ~80% higher suppression effect than first-line chemotherapy and 50-70% higher than the most advanced tumor treating field technology. In addition, patient-personalized therapy strategies could be tuned from the MIBTS without increasing size or adding circuits on the integrated chip, ensuring the optimal therapeutic effect and avoid tumor resistance. These groundbreaking achievements of MIBTS offer new hope for controlling tumor recurrence and extending patient survival.
Collapse
Affiliation(s)
- Xiaona Cao
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jie Li
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jinliang Ren
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jiajin Peng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Ruyue Zhong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jiahao He
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Ting Xu
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Zhenhua Yu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Huawei Jin
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Siqi Hao
- School of Naval Architecture & Ocean Engineering, Guangzhou Maritime University, 101 Hongshan 3rd Road, Huangpu District, Guangzhou, Guangdong, 510725, P.R. China
| | - Ruiwei Liu
- School of Naval Architecture & Ocean Engineering, Guangzhou Maritime University, 101 Hongshan 3rd Road, Huangpu District, Guangzhou, Guangdong, 510725, P.R. China
| | - Bingzhe Xu
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China.
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China.
| |
Collapse
|
5
|
Xiao T, Zheng H, Zu K, Yue Y, Wang Y. Tumor-treating fields in cancer therapy: advances of cellular and molecular mechanisms. Clin Transl Oncol 2024:10.1007/s12094-024-03551-z. [PMID: 38884919 DOI: 10.1007/s12094-024-03551-z] [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: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Tumor-Treating Fields (TTFields) use intermediate-frequency and low-intensity electric fields to inhibit tumor cells. However, their mechanisms are still not well understood. This article reviews their key antitumor mechanisms at the cellular and molecular levels, including inhibition of proliferation, induction of death, disturbance of migration, and activation of the immune system. The multifaceted biological effects in combination with other cancer treatments are also summarized. The deep insight into their mechanism will help develop more potential antitumor treatments.
Collapse
Affiliation(s)
- Tong Xiao
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hao Zheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Kaiyang Zu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Youjia Yue
- School of Biomedical Engineeringg, Capital Medical University, Beijing, 100069, China
| | - Ying Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
6
|
Chen J, Liu Y, Lan J, Liu H, Tang Q, Li Z, Qiu X, Hu W, Xie J, Feng Y, Qin L, Zhang X, Liu J, Chen L. Identification and validation of COL6A1 as a novel target for tumor electric field therapy in glioblastoma. CNS Neurosci Ther 2024; 30:e14802. [PMID: 38887185 PMCID: PMC11183175 DOI: 10.1111/cns.14802] [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: 01/25/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most aggressive primary brain malignancy. Novel therapeutic modalities like tumor electric field therapy (TEFT) have shown promise, but underlying mechanisms remain unclear. The extracellular matrix (ECM) is implicated in GBM progression, warranting investigation into TEFT-ECM interplay. METHODS T98G cells were treated with TEFT (200 kHz, 2.2 V/m) for 72 h. Collagen type VI alpha 1 (COL6A1) was identified as hub gene via comprehensive bioinformatic analysis based on RNA sequencing (RNA-seq) and public glioma datasets. TEFT intervention models were established using T98G and Ln229 cell lines. Pre-TEFT and post-TEFT GBM tissues were collected for further validation. Focal adhesion pathway activity was assessed by western blot. Functional partners of COL6A1 were identified and validated by co-localization and survival analysis. RESULTS TEFT altered ECM-related gene expression in T98G cells, including the hub gene COL6A1. COL6A1 was upregulated in GBM and associated with poor prognosis. Muti-database GBM single-cell analysis revealed high-COL6A1 expression predominantly in malignant cell subpopulations. Differential expression and functional enrichment analyses suggested COL6A1 might be involved in ECM organization and focal adhesion. Western blot (WB), immunofluorescence (IF), and co-immunoprecipitation (Co-IP) experiments revealed that TEFT significantly inhibited expression of COL6A1, hindering its interaction with ITGA5, consequently suppressing the FAK/Paxillin/AKT pathway activity. These results suggested that TEFT might exert its antitumor effects by downregulating COL6A1 and thereby inhibiting the activity of the focal adhesion pathway. CONCLUSION TEFT could remodel the ECM of GBM cells by downregulating COL6A1 expression and inhibiting focal adhesion pathway. COL6A1 could interact with ITGA5 and activate the focal adhesion pathway, suggesting that it might be a potential therapeutic target mediating the antitumor effects of TEFT.
Collapse
Affiliation(s)
- Junyi Chen
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| | - Yuyang Liu
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jinxin Lan
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
| | - Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalHainanChina
| | - Qingyun Tang
- Department of Gastroenterology920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Ze Li
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| | - Xiaoguang Qiu
- Beijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Wentao Hu
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
| | - Jiaxin Xie
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Yaping Feng
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Lilin Qin
- Zhejiang Cancer HospitalZhejiangHangzhouChina
| | - Xin Zhang
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jialin Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| | - Ling Chen
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| |
Collapse
|
7
|
Lan J, Liu Y, Chen J, Liu H, Feng Y, Liu J, Chen L. Advanced tumor electric fields therapy: A review of innovative research and development and prospect of application in glioblastoma. CNS Neurosci Ther 2024; 30:e14720. [PMID: 38715344 PMCID: PMC11077002 DOI: 10.1111/cns.14720] [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: 11/09/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive malignant tumor with a high mortality rate and is the most prevalent primary intracranial tumor that remains incurable. The current standard treatment, which involves surgery along with concurrent radiotherapy and chemotherapy, only yields a survival time of 14-16 months. However, the introduction of tumor electric fields therapy (TEFT) has provided a glimmer of hope for patients with newly diagnosed and recurrent GBM, as it has been shown to extend the median survival time to 20 months. The combination of TEFT and other advanced therapies is a promising trend in the field of GBM, facilitated by advancements in medical technology. AIMS In this review, we provide a concise overview of the mechanism and efficacy of TEFT. In addition, we mainly discussed the innovation of TEFT and our proposed blueprint for TEFT implementation. CONCLUSION Tumor electric fields therapy is an effective and highly promising treatment modality for GBM. The full therapeutic potential of TEFT can be exploited by combined with other innovative technologies and treatments.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| | - Yuyang Liu
- Medical School of Chinese PLABeijingChina
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Junyi Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalHainanChina
| | - Yaping Feng
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jialin Liu
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Ling Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| |
Collapse
|
8
|
Shen Z, Dong T, Yong H, Deng C, Chen C, Chen X, Chen M, Chu S, Zheng J, Li Z, Bai J. FBXO22 promotes glioblastoma malignant progression by mediating VHL ubiquitination and degradation. Cell Death Discov 2024; 10:151. [PMID: 38519492 PMCID: PMC10959977 DOI: 10.1038/s41420-024-01919-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor. Despite comprehensive treatment with traditional surgery, radiotherapy, and chemotherapy, the median survival rate is <14.6% and the 5-year survival rate is only 5%. FBXO22, a substrate receptor of the SCF ubiquitin ligases, has been reported to play a promoting role in melanoma, liver cancer, cervical cancer, and other cancers. However, the function of FBXO22 in GBM has not been reported. In the present study, we demonstrate that FBXO22 is highly expressed in glioma and is positively correlated with worse pathological features and shorter survival of GBM patients. We revealed that FBXO22 promotes GBM cell proliferation, angiogenesis, migration, and tumorigenesis in vitro and in vivo. In terms of mechanism, we reveal that FBXO22 decreases VHL expression by directly mediating VHL ubiquitination degradation, which ultimately increases HIF-1α and VEGFA expression. In addition, our data confirm that there are positive correlations among FBXO22, HIF-1α, and VEGFA expression, and there is a negative correlation between FBXO22 and VHL protein expression in glioma patients. Our study strongly indicates that FBXO22 is a promising diagnostic marker and therapeutic target for glioma patients.
Collapse
Affiliation(s)
- Zhigang Shen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tao Dong
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongmei Yong
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huaian, Jiangsu, China
| | - Chuyin Deng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Changxiu Chen
- Department of Pediatrics, the Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xintian Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Miaolei Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Zhongwei Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Laboratory of Tumor Epigenetics, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
9
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
10
|
Li ZQ, Zhang GS, Liu RQ, Shuai SY, Hu PY, Zheng Q, Xiao SH. Anti-Glioma Effects of Ligustilide or n-Butylphthalide on Their Own and the Synergistic Effects with Temozolomide via PI3K/Akt Signaling Pathway. Onco Targets Ther 2023; 16:983-994. [PMID: 38021448 PMCID: PMC10676728 DOI: 10.2147/ott.s432901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background Ligustilide (LIG) and n-butylphthalide (NBP) have neuroprotective effects in cerebral ischemia; however, their roles in gliomas are not well-known.This study aimed to explore the anti-glioma effects of LIG and NBP individually and the synergistic effects of temozolomide (TMZ) via the PI3K/Akt Signaling Pathway. Materials and Methods Cytotoxicity of LIG and NBP alone and in combination with TMZ in U251 cells was determined using the CCk-8. The effect of compounds alone or in combination on cell migration was detected using the wound healing assay, and the invasion was evaluated by transwell assays, respectively. Cell apoptosis was quantified by flow cytometry and the changed expressions of proteins were detected by Western blotting. Results The results showed that LIG and NBP significantly inhibited the growth of U251 cells at concentrations of 4-10 µg/mL and 1.5-6 µg/mL in a dose-dependent manner (p<0.05, p<0.01). The combination of 20 µg/mL TMZ with LIG in the concentration range of 4-10 µg/mL or with NBP of 0.5-6 µg/mlachieved synergistic effect towardsU251 cells. LIG and NBP, alone or in combination with TMZ, markedly inhibited cell invasion (p< 0.001) and enhanced apoptosis (p< 0.05). The combination of TMZ with LIG or NBP markedly inhibited cell migration (p< 0.001). Western blot analysis showed that LIG, NBP, and TMZ, alone and in combination, significantly decreased the expression of Bcl-2, p-PI3K, and p-Akt, and increased the expression of Bax. Conclusion Both LIG and NBP exert anti-glioma effects on their own through the PI3K/Akt pathway and enhance TMZ-mediated anti-glioma efficiency via the same pathway.
Collapse
Affiliation(s)
- Zi-Qi Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Guo-Song Zhang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Ri-Qun Liu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Shu-Yuan Shuai
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Peng-Yi Hu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Shu-Hua Xiao
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| |
Collapse
|
11
|
Galicka A, Szoka Ł, Radziejewska I, Marcinkiewicz C. Effect of Dimeric Disintegrins Isolated from Vipera lebetina obtusa Venom on Glioblastoma Cellular Responses. Cancers (Basel) 2023; 15:4805. [PMID: 37835499 PMCID: PMC10572073 DOI: 10.3390/cancers15194805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Integrins play a fundamental role in the migration and invasiveness of glioblastoma (GBM) cells, making them suitable targets for innovative cancer therapy. The aim of this study was to evaluate the effect of the RGD homodimeric disintegrin VLO4, isolated from Vipera lebetina obtusa venom, on the adhesion, spreading, migration, and survival of LBC3, LN18, and LN229 cell lines. This disintegrin, as a potent antagonist for α5β1 integrin, showed pro-adhesive properties for these cell lines, the highest for LN229 and the lowest for LBC3. Glioblastoma cells displayed significant differences in the spreading on the immobilized VLO4 and the natural α5β1 integrin ligand, fibronectin. Solubilized VLO4 showed different cytotoxicity and pro-apoptotic properties among tested cell lines, with the highest against LN18 and none against LN229. Moreover, VLO4 revealed an inhibitory effect on the migration of LBC3 and LN18 cell lines, in contrast to LN229 cells, which were not sensitive to this disintegrin. However, LN229 migration was impaired by VLO5, a disintegrin antagonistic to integrin α9β1, used in combination with VLO4. A possible mechanism of action of VLO4 may be related to the downregulation of α5β1 integrin subunit expression, as revealed by Western blot. VLO4 also inhibited cell proliferation and induced caspase-dependent apoptosis in LBC3 and LN18 cell lines. These results indicate that targeting α5β1 integrin by related VLO4 compounds may be useful in the development of integrin-targeted therapy for glioblastoma.
Collapse
Affiliation(s)
- Anna Galicka
- Department of Medical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland;
| | - Łukasz Szoka
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland;
| | - Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland;
| | - Cezary Marcinkiewicz
- Department of Bioengineering, Temple University CoE, Philadelphia, PA 19406, USA
| |
Collapse
|
12
|
Sun Z, Sun X, Yuan Y, Li H, Li X, Yao Z. FCGR2B as a prognostic and immune microenvironmental marker for gliomas based on transcriptomic analysis. Medicine (Baltimore) 2023; 102:e35084. [PMID: 37713871 PMCID: PMC10508392 DOI: 10.1097/md.0000000000035084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/15/2023] [Indexed: 09/17/2023] Open
Abstract
To explore the expression and prognosis of Fc fragment of IgG low affinity IIb receptor (FCGR2B) in glioma and its relationship with immune microenvironment, so as to provide potential molecular targets for the treatment of glioma. We analyzed the gene expression of FCGR2B using the Cancer Genome Atlas database, Chinese Glioma Genome Atlas, Gene Expression Omnibus database and other glioma related databases. Moreover, we generated survival receiver operating characteristic curve, carried out univariate and multivariate Cox analysis and nomograph construction, and analyzed the relationship between FCGR2B and prognosis. According to the median of FCGR2B gene expression value, the differential expression analysis was carried out by high and low grouping method, and the gene ontology, Kyoto encyclopedia of genes and genomes, and gene set enrichment analysis enrichment analysis were carried out to explore the possible mechanism. Then, the correlation between immune score of glioma and prognosis, World Health Organization grade and FCGR2B expression was analyzed. Finally, the correlation between FCGR2B expression and the proportion of tumor infiltrating immune cells, immune checkpoints, tumor mutation load and immune function was analyzed. The expression of FCGR2B in gliomas was higher than that in normal tissues and was associated with poor prognosis. Independent prognostic analysis showed that FCGR2B was an independent prognostic factor for glioma. The analysis of gene ontology and gene set enrichment analysis showed that FCGR2B was closely related to immune-related functions. The analysis of immune scores and prognosis, World Health Organization grade and FCGR2B expression in gliomas indicated that patients with high immune scores had significantly poorer overall survival and higher tumor pathological grade. In addition, immune scores were significantly positively correlated with the expression of FCGR2B. The analysis of tumor infiltrating immune cells suggested that the expression level of FCGR2B affected the immune activity of TME. In addition, the expression of FCGR2B was positively correlated with almost all immune checkpoint molecules including CD28, CD44, TNFSF14, PDCD1LG2, LAIR1, and CD48 and was significantly positively correlated with tumor mutation load. All immunobiological functions of the high expression group of FCGR2B were significantly inhibited. FCGR2B may play an important role in the occurrence, development and invasion of tumor by influencing the tumor microenvironment of immunosuppression. FCGR2B may be an important target for the treatment of glioma.
Collapse
Affiliation(s)
- Zhimin Sun
- Department of Neurosurgery and Radiology, The Third Hospital of Shijiazhuang City, Shijiazhuang, China
| | - Xiaoli Sun
- Department of Neurosurgery and Radiology, The Third Hospital of Shijiazhuang City, Shijiazhuang, China
| | - Yaqin Yuan
- Department of Neurosurgery and Radiology, The Third Hospital of Shijiazhuang City, Shijiazhuang, China
| | - Hongsheng Li
- Department of Neurology, The People Hospital of Xingtai City, Xingtai, China
| | - Xiaona Li
- Department of Pediatrics, The People Hospital of Linxi County, Xingtai, China
| | - Zhigang Yao
- Department of Neurosurgery and Radiology, The Third Hospital of Shijiazhuang City, Shijiazhuang, China
| |
Collapse
|
13
|
Zeng L, Zheng W, Liu X, Zhou Y, Jin X, Xiao Y, Bai Y, Pan Y, Zhang J, Shao C. SDC1-TGM2-FLOT1-BHMT complex determines radiosensitivity of glioblastoma by influencing the fusion of autophagosomes with lysosomes. Theranostics 2023; 13:3725-3743. [PMID: 37441590 PMCID: PMC10334832 DOI: 10.7150/thno.81999] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Rationale: Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Radiotherapy has long been an important treatment for GBM. Despite recent advances in tumor radiotherapy, the prognosis of GBM remains poor due to radioresistance. Autophagy has been reported as a basic factor to prolong the survival of tumor under radiation stress, but the molecular mechanism of how autophagy contributes to GBM radioresistance was still lacking. Methods: We established radioresistant GBM cells and identified their protein profiles by Tandem mass tag (TMT) quantitative proteomic analysis, then chose the radioresistant genes based on the TMT analysis of GBM cells and differentially expressed genes (DEGs) analysis of GEO database. Colony formation, flow cytometry, qPCR, western blotting, mRFP-GFP-LC3, transmission electron microscopy, immunofluorescence, and co-IP assays were conducted to investigate the regulation mechanisms among these new-found molecules. Results: Syndecan 1 (SDC1) and Transglutaminase 2 (TGM2) were both overexpressed in the radioresistant GBM cells and tissues, contributing to the dismal prognosis of radiotherapy. Mechanically, after irradiation, SDC1 carried TGM2 from cell membrane into cytoplasm, and transported to lysosomes by binding to flotillin 1 (FLOT1), then TGM2 recognized the betaine homocysteine methyltransferase (BHMT) on autophagosomes to coordinate the encounter between autophagosomes and lysosomes. Conclusions: The SDC1-TGM2-FLOT1-BHMT copolymer, a novel member of the protein complexes involved in the fusion of lysosomes and autophagosomes, maintained the autophagic flux in the irradiated tumor cells and ultimately enhanced radioresistance of GBM, which provides new insights of the molecular mechanism and therapeutic targets of radioresistant GBM.
Collapse
Affiliation(s)
- Liang Zeng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wang Zheng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuchuan Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoya Jin
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuqi Xiao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Zhou Y, Xing X, Zhou J, Jiang H, Cen P, Jin C, Zhong Y, Zhou R, Wang J, Tian M, Zhang H. Therapeutic potential of tumor treating fields for malignant brain tumors. Cancer Rep (Hoboken) 2023; 6:e1813. [PMID: 36987739 PMCID: PMC10172187 DOI: 10.1002/cnr2.1813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/02/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Malignant brain tumors are among the most threatening diseases of the central nervous system, and despite increasingly updated treatments, the prognosis has not been improved. Tumor treating fields (TTFields) are an emerging approach in cancer treatment using intermediate-frequency and low-intensity electric field and can lead to the development of novel therapeutic options. RECENT FINDINGS A series of biological processes induced by TTFields to exert anti-cancer effects have been identified. Recent studies have shown that TTFields can alter the bioelectrical state of macromolecules and organelles involved in cancer biology. Massive alterations in cancer cell proteomics and transcriptomics caused by TTFields were related to cell biological processes as well as multiple organelle structures and activities. This review addresses the mechanisms of TTFields and recent advances in the application of TTFields therapy in malignant brain tumors, especially in glioblastoma (GBM). CONCLUSIONS As a novel therapeutic strategy, TTFields have shown promising results in many clinical trials, especially in GBM, and continue to evolve. A growing number of patients with malignant brain tumors are being enrolled in ongoing clinical studies demonstrating that TTFields-based combination therapies can improve treatment outcomes.
Collapse
Affiliation(s)
- Youyou Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaoqing Xing
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinyun Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Han Jiang
- Faculty of Science and Technology, Department of Electrical and Computer Engineering, Biomedical Imaging Laboratory (BIG), University of Macau, Taipa, Macau SAR, China
| | - Peili Cen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Bettenfeld R, Claudel J, Kourtiche D, Nadi M, Schlauder C. Design and Modeling of a Device Combining Single-Cell Exposure to a Uniform Electrical Field and Simultaneous Characterization via Bioimpedance Spectroscopy. SENSORS (BASEL, SWITZERLAND) 2023; 23:3460. [PMID: 37050519 PMCID: PMC10098563 DOI: 10.3390/s23073460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 06/19/2023]
Abstract
Previous studies have demonstrated the electropermeabilization of cell membranes exposed to an electric field with moderate intensity (<2 V/cm) and a frequency of <100 MHz. Bioimpedance spectroscopy (BIS) is an electrical characterization technique that can be useful in studying this phenomenon because it is already used for electroporation. In this paper, we report a device designed to perform BIS on single cells and expose them to an electric field simultaneously. It also allows cells to be monitored by visualization through a transparent exposure electrode. This device is based on a lab-on-a-chip (LOC) with a microfluidic cell-trapping system and microelectrodes for BIS characterization. We present numerical simulations that support the design of the LOC. We also describe the fabrication of the LOC and the first electrical characterization of its measurement bandwidth. This first test, performed on reference medium with a conductivity in the same order than human cells, confirms that the measurement capabilities of our device are suitable for electrical cells characterization.
Collapse
|
16
|
Abed T, Ganser K, Eckert F, Stransky N, Huber SM. Ion channels as molecular targets of glioblastoma electrotherapy. Front Cell Neurosci 2023; 17:1133984. [PMID: 37006466 PMCID: PMC10064067 DOI: 10.3389/fncel.2023.1133984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/10/2023] [Indexed: 03/19/2023] Open
Abstract
Therapies with weak, non-ionizing electromagnetic fields comprise FDA-approved treatments such as Tumor Treating Fields (TTFields) that are used for adjuvant therapy of glioblastoma. In vitro data and animal models suggest a variety of biological TTFields effects. In particular, effects ranging from direct tumoricidal, radio- or chemotherapy-sensitizing, metastatic spread-inhibiting, up to immunostimulation have been described. Diverse underlying molecular mechanisms, such as dielectrophoresis of cellular compounds during cytokinesis, disturbing the formation of the spindle apparatus during mitosis, and perforating the plasma membrane have been proposed. Little attention, however, has been paid to molecular structures that are predestinated to percept electromagnetic fields-the voltage sensors of voltage-gated ion channels. The present review article briefly summarizes the mode of action of voltage sensing by ion channels. Moreover, it introduces into the perception of ultra-weak electric fields by specific organs of fishes with voltage-gated ion channels as key functional units therein. Finally, this article provides an overview of the published data on modulation of ion channel function by diverse external electromagnetic field protocols. Combined, these data strongly point to a function of voltage-gated ion channels as transducers between electricity and biology and, hence, to voltage-gated ion channels as primary targets of electrotherapy.
Collapse
Affiliation(s)
- Tayeb Abed
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Medical University Vienna, Vienna, Austria
| | - Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
17
|
Xu S, Luo C, Chen D, Tang L, Cheng Q, Chen L, Liu Z. circMMD reduction following tumor treating fields inhibits glioblastoma progression through FUBP1/FIR/DVL1 and miR-15b-5p/FZD6 signaling. J Exp Clin Cancer Res 2023; 42:64. [PMID: 36932454 PMCID: PMC10021944 DOI: 10.1186/s13046-023-02642-z] [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: 01/02/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Tumor treating fields (TTF) is the latest treatment for GBM. Circular RNA (circRNA) has been demonstrated to play critical roles in tumorigenesis. However, the molecular mechanism of TTF remained largely unknown and the role of circRNA in TTF was not reported. The aim of this study was to elucidate the role and mechanism of circMMD in TTF treatment of GBM. METHODS Divergent primer was designed to verify the existence of circMMD in GBM cells. The prognostic role of circMMD was explored in glioma specimens. The knockdown and overexpressed plasmids were used to evaluate the effect of circMMD on GBM cell proliferation and TTF efficacy. RNA pull-down and RNA immunoprecipitation were performed to identify binding proteins of circMMD. Subcutaneous and intracranial tumor models were established to validate findings in vivo. RESULTS The expression of circMMD was elevated in GBM and its high expression indicated poor prognoses. TTF intervention could reduce circMMD synthesis, which suppressed GBM proliferation and increased TTF-mediated apoptosis. The reduction of circMMD promoted the interaction between FUBP1 and FIR, which decreased DVL1 transcription. Meanwhile, decreased circMMD would promote the activity of miR-15b-5p to degrade FZD6. Finally, the diminished expression of DVL1 and FZD6 expression suppressed the activation of Wnt/β-catenin pathway. CONCLUSIONS Our study revealed a novel mechanism of TTF that TTF-mediated reduction of circMMD could inhibit Wnt/β-catenin pathway to suppress GBM proliferation.
Collapse
Affiliation(s)
- Shengchao Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chengke Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, 410008, China
| | - Lu Tang
- Department of Anesthesiology, Xiangya Hospital, Central South University, ChangshaHunan, 410008, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ling Chen
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Medical School of Chinese PLA, Institute of Neurosurgery of Chinese PLA, Beijing, 100853, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
18
|
Shams S, Patel CB. Anti-cancer mechanisms of action of therapeutic alternating electric fields (tumor treating fields [TTFields]). J Mol Cell Biol 2022; 14:mjac047. [PMID: 35973687 PMCID: PMC9912101 DOI: 10.1093/jmcb/mjac047] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/11/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Despite improved survival outcomes across many cancer types, the prognosis remains grim for certain solid organ cancers including glioblastoma and pancreatic cancer. Invariably in these cancers, the control achieved by time-limited interventions such as traditional surgical resection, radiation therapy, and chemotherapy is short-lived. A new form of anti-cancer therapy called therapeutic alternating electric fields (AEFs) or tumor treating fields (TTFields) has been shown, either by itself or in combination with chemotherapy, to have anti-cancer effects that translate to improved survival outcomes in patients. Although the pre-clinical and clinical data are promising, the mechanisms of TTFields are not fully elucidated. Many investigations are underway to better understand how and why TTFields is able to selectively kill cancer cells and impede their proliferation. The purpose of this review is to summarize and discuss the reported mechanisms of action of TTFields from pre-clinical studies (both in vitro and in vivo). An improved understanding of how TTFields works will guide strategies focused on the timing and combination of TTFields with other therapies, to further improve survival outcomes in patients with solid organ cancers.
Collapse
Affiliation(s)
- Shadi Shams
- Rowan University School of Osteopathic Medicine, Stratford, NJ 08028, USA
| | - Chirag B Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
19
|
Tanzhu G, Chen L, Xiao G, Shi W, Peng H, Chen D, Zhou R. The schemes, mechanisms and molecular pathway changes of Tumor Treating Fields (TTFields) alone or in combination with radiotherapy and chemotherapy. Cell Death Discov 2022; 8:416. [PMID: 36220835 PMCID: PMC9553876 DOI: 10.1038/s41420-022-01206-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor Treating Fields (TTFields) is a physical therapy that uses moderate frequency (100-300 kHz) and low-intensity (1-3 V/cm) alternating electric fields to inhibit tumors. Currently, the Food and Drug Administration approves TTFields for treating recurrent or newly diagnosed glioblastoma (GBM) and malignant pleural mesothelioma (MPM). The classical mechanism of TTFields is mitotic inhibition by hindering the formation of tubulin and spindle. In addition, TTFields inhibits cell proliferation, invasion, migration and induces cell death, such as apoptosis, autophagy, pyroptosis, and cell cycle arrest. Meanwhile, it regulates immune function and changes the permeability of the nuclear membrane, cell membrane, and blood-brain barrier. Based on the current researches on TTFields in various tumors, this review comprehensively summarizes the in-vitro effects, changes in pathways and molecules corresponding to relevant parameters of TTFields (frequency, intensity, and duration). In addition, radiotherapy and chemotherapy are common tumor treatments. Thus, we also pay attention to the sequence and dose when TTFields combined with radiotherapy or chemotherapy. TTFields has inhibitory effects in a variety of tumors. The study of TTFields mechanism is conducive to subsequent research. How to combine common tumor therapy such as radiotherapy and chemotherapy to obtain the maximum benefit is also a problem that's worthy of our attention.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Liu Chen
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Wen Shi
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Haiqin Peng
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P.R. China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, 410008, Changsha, China.
| |
Collapse
|
20
|
Chen B, Zhou X, Yang L, Zhou H, Meng M, Wu H, Liu Z, Zhang L, Li C. Glioma stem cell signature predicts the prognosis and the response to tumor treating fields treatment. CNS Neurosci Ther 2022; 28:2148-2162. [PMID: 36070228 PMCID: PMC9627385 DOI: 10.1111/cns.13956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Glioma stem cells (GSCs) play an important role in glioma recurrence and chemo-radiotherapy (CRT) resistance. Currently, there is a lack of efficient treatment approaches targeting GSCs. This study aimed to explore the potential personalized treatment of patients with GSC-enriched gliomas. METHODS Single-cell RNA sequencing (scRNA-seq) was used to identify the GSC-related genes. Then, machine learning methods were applied for clustering and validation. The least absolute shrinkage and selection operator (LASSO) and COX regression were used to construct the risk scores. Survival analysis was performed. Additionally, the incidence of chemo-radiotherapy resistance, immunotherapy status, and tumor treating field (TTF) therapy response were evaluated in high- and low-risk scores groups. RESULTS Two GSC clusters exhibited significantly different stemness indices, immune microenvironments, and genomic alterations. Based on GSC clusters, 11-gene GSC risk scores were constructed, which exhibited a high predictive value for prognosis. In terms of therapy, patients with high GSC risk scores had a higher risk of resistance to chemotherapy. TTF therapy can comprehensively inhibit the malignant biological characteristics of the high GSC-risk-score gliomas. CONCLUSION Our study constructed a GSC signature consisting of 11 GSC-specific genes and identified its prognostic value in gliomas. TTF is a promising therapeutic approach for patients with GSC-enriched glioma.
Collapse
Affiliation(s)
- Bo Chen
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoxi Zhou
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Liting Yang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina,Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaChina,Clinical Diagnosis and Therapy Center for Glioma, Xiangya HospitalCentral South UniversityChangshaChina
| | - Hongshu Zhou
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Ming Meng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Hao Wu
- Department of Neurosurgery, The Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina,Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaChina,Clinical Diagnosis and Therapy Center for Glioma, Xiangya HospitalCentral South UniversityChangshaChina
| | - Chuntao Li
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaChina,Hypothalamic‐Pituitary Research Center, Xiangya HospitalCentral South UniversityChangshaChina,Clinical Diagnosis and Therapy Center for Glioma, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
21
|
Wang Z, Chen G. Insights about circadian clock in glioma: From molecular pathways to therapeutic drugs. CNS Neurosci Ther 2022; 28:1930-1941. [PMID: 36066207 PMCID: PMC9627379 DOI: 10.1111/cns.13966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 02/06/2023] Open
Abstract
Glioma is characterized as the most aggressive brain tumor that occurred in the central nervous system. The circadian rhythm is an essential cyclic change system generated by the endogenous circadian clock. Current studies found that the circadian clock affects glioma pathophysiology. It is still controversial whether the circadian rhythm disruption is a cause or an effect of tumorigenesis. This review discussed the association between cell cycle and circadian clock and provided a prominent molecular theoretical basis for tumor therapy. We illustrated the external factors affecting the circadian clock including thermodynamics, hypoxia, post-translation, and microRNA, while the internal characteristics concerning the circadian clock in glioma involve stemness, metabolism, radiotherapy sensitivity, and chemotherapy sensitivity. We also summarized the molecular pathways and the therapeutic drugs involved in the glioma circadian rhythm. There are still many questions in this field waiting for further investigation. The results of glioma chronotherapy in sensitizing radiation therapy and chemotherapy have shown great therapeutic potential in improving clinical outcomes. These findings will help us further understand the characteristics of glioma pathophysiology.
Collapse
Affiliation(s)
- Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina,Institute of Stroke ResearchSoochow UniversitySuzhouChina
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina,Institute of Stroke ResearchSoochow UniversitySuzhouChina
| |
Collapse
|
22
|
Xu S, Luo C, Chen D, Tang L, Chen L, Liu Z. Whole transcriptome and proteome analyses identify potential targets and mechanisms underlying tumor treating fields against glioblastoma. Cell Death Dis 2022; 13:721. [PMID: 35982032 PMCID: PMC9388668 DOI: 10.1038/s41419-022-05127-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 01/21/2023]
Abstract
Glioblastoma (GBM) is one of the most malignant types of brain cancer. Tumor treating fields (TTFields) is the up-to-date treatment for GBM. However, its molecular mechanism requires additional investigation. Herein, a novel TTFields system was developed (CL-301A) and its efficiency in suppressing GBM cell proliferation and inducing cell apoptosis was demonstrated. Through the whole proteomic and transcriptomic analyses, a multitude of differentially expressed proteins (1243), mRNAs (4191), miRtNAs (47), lncRNAs (4286), and circRNAs (13,903) were identified. Bioinformatic analysis indicated that TTFields mainly affected nuclear proteins and interrupt cell mitosis-related events. Moreover, the inhibition of autophagy could significantly enhance the anti-GBM activity of TTFields. And CDK2-AS1 might be a target of TTFields to mediate cell cycle arrest via regulating CDK2 mRNA stability. This study provided valuable resources for understanding the mechanism of TTFields, which might further assist the investigation of TTFields in GBM treatment.
Collapse
Affiliation(s)
- Shengchao Xu
- grid.216417.70000 0001 0379 7164Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chengke Luo
- grid.216417.70000 0001 0379 7164Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Lu Tang
- grid.216417.70000 0001 0379 7164Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Ling Chen
- grid.488137.10000 0001 2267 2324Department of Neurosurgery, Chinese People’s Liberation Army of China (PLA) General Hospital, Medical School of Chinese PLA, Institute of Neurosurgery of Chinese PLA, 100853 Beijing, China
| | - Zhixiong Liu
- grid.216417.70000 0001 0379 7164Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Liu Y, Shi Y, Wu M, Liu J, Wu H, Xu C, Chen L. Hypoxia-induced polypoid giant cancer cells in glioma promote the transformation of tumor-associated macrophages to a tumor-supportive phenotype. CNS Neurosci Ther 2022; 28:1326-1338. [PMID: 35762580 PMCID: PMC9344088 DOI: 10.1111/cns.13892] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/28/2022] Open
Abstract
Aims Polypoid giant cancer cells (PGCCs) represent a unique subgroup of stem‐like cells, acting as a critical factor in promoting the recurrence of various solid tumors. The effect of PGCCs on the tumor malignancy of glioma and its immune microenvironment remains unclear. Methods Bioinformatic analysis was performed to investigate the relationship between M2 tumor‐associated macrophages (TAMs) infiltration and survival of glioblastoma (GBM) patients. The spatial location of M2 TAMs in GBM was also investigated using the Ivy Glioblastoma Atlas Project (Ivy GAP) database. PGCCs were quantified in glioma of different grades. CoCl2 was used to induce PGCCs in cultures of A172 cells. PGCCs, and their progeny cells in cultures were further evaluated for morphological features, tumorsphere formation, and TAMs activation. Results The magnitude of M2 TAMs infiltration is significantly correlated with poor survival in GBM patients. M2 TAMs were enriched in the perinecrotic zone (PNZ) of GBM and positively correlated with hypoxic levels. Increased PGCCs were detected in glioma specimens of higher grades. CoCl2 induced hypoxia and the transformation of A172 cultures into PGCCs, producing the progeny cells, PGCCs‐Dau, through asymmetric division. PGCCs and PGCCs‐Dau possessed tumor stem cell‐like features, while PGCCs‐Dau enhanced the polarization of TAMs into an M2 phenotype with relevance to immunosuppression and malignancy in GBM. Conclusions PGCCs promote malignancy and immune‐suppressive microenvironment in GBM. PGCCs or their progeny cells may be a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Yuyang Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ying Shi
- School of Medicine, University of Electronic science and Technology of China, Chengdu, China.,Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Mengwan Wu
- School of Medicine, University of Electronic science and Technology of China, Chengdu, China.,Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Jialin Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Hong Wu
- Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Chuan Xu
- Integrative Cancer Center& Cancer Clinical Research Center, Sichuan Cancer Hospital, Chengdu, China
| | - Ling Chen
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
24
|
Liu Y, Yao R, Shi Y, Liu Y, Liu H, Liu J, Guan Y, Yao Y, Chen L. Identification of CD101 in Glioma: A Novel Prognostic Indicator Expressed on M2 Macrophages. Front Immunol 2022; 13:845223. [PMID: 35350788 PMCID: PMC8957828 DOI: 10.3389/fimmu.2022.845223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/07/2022] [Indexed: 12/21/2022] Open
Abstract
Glioma represents the most common primary intracranial malignancy worldwide, with low overall survival rates and limited therapeutic options. The protein CD101, mainly expressed on several immune cells, has been demonstrated to exert potent effects on blunting T cell immune responses across infectious and autoimmunity diseases. Nevertheless, the prognostic value of CD101 expression and its role in the immune microenvironment of various malignancies currently remains elusive. Herein, by adopting bioinformatics methodology, we comprehensively illustrated the potential function and predictive value of CD101 in stratifying clinical prognosis among patients with glioma, for which a high CD101 level predicted an unfavorable clinical outcome in glioma patients. Results from enrichment analyses manifested that CD101 predominantly expressed on the tumor-associated macrophages and was significantly associated with the immune regulatory processes, as evidenced by its positive correlation with immune-related genes and the putative infiltration of immune cells. Evidence provided by in-situ multicolor immunofluorescence staining further validated our findings at the protein level. Taken together, CD101 may serve as a novel biomarker in predicting clinical prognosis and immune status for glioma patients.
Collapse
Affiliation(s)
- Yuyang Liu
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China.,Senior Department of Neurosurgery, the First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Renqi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ying Shi
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuxiao Liu
- Senior Department of Neurosurgery, the First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongyu Liu
- Senior Department of Neurosurgery, the First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jialin Liu
- Senior Department of Neurosurgery, the First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yunqian Guan
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ling Chen
- Senior Department of Neurosurgery, the First Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
25
|
Hong P, Kudulaiti N, Wu S, Nie J, Zhuang D. Tumor treating fields: a comprehensive overview of the underlying molecular mechanism. Expert Rev Mol Diagn 2021; 22:19-28. [PMID: 34883030 DOI: 10.1080/14737159.2022.2017283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION As a novel treatment modality, tumor treating fields (TTFields) exert low-intensity, medium-frequency electric fields on tumor cells. TTFields' effectiveness and safety have been demonstrated clinically and in the real world for treating glioblastoma, the most common and aggressive primary central nervous system tumor. TTFields therapy has also been approved for the management of malignant mesothelioma, and clinical trials are ongoing for NSCLC, gastric cancer, pancreatic cancer, and other solid tumors. AREAS COVERED This article comprehensively reviews the currently described evidence of TTFields' mechanism of action. TTFields' most evident therapeutic effect is to induce cell death by disrupting mitosis. Moreover, evidence suggests at additional mechanistic complexity, such as delayed DNA repair and heightened DNA replication stress, reversible increase in cell membrane and blood-brain barrier permeability, induction of immune response, and so on. EXPERT OPINION TTFields therapy has been arising as the fourth anti-tumor treatment besides surgery, radiotherapy, and antineoplastic agents in recent years. However, the precise molecular mechanisms underlying the effects of TTFields are not fully understood and some concepts remain controversial. An in-depth understanding of TTFields' effects on tumor cell and tumor microenvironment would be crucial for informing research aimed at further optimizing TTFields' efficacy and developing new combination therapies for clinical applications.
Collapse
Affiliation(s)
- Pengjie Hong
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Nijiati Kudulaiti
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Shuai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Jingtao Nie
- Zai Lab Trading (Shanghai) Co., Ltd., Shanghai, China
| | - Dongxiao Zhuang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| |
Collapse
|
26
|
Wu H, Yang L, Liu H, Zhou D, Chen D, Zheng X, Yang H, Li C, Chang J, Wu A, Wang Z, Ren N, Lv S, Liu Y, Jia M, Lu J, Liu H, Sun G, Liu Z, Liu J, Chen L. Exploring the efficacy of tumor electric field therapy against glioblastoma: An in vivo and in vitro study. CNS Neurosci Ther 2021; 27:1587-1604. [PMID: 34710276 PMCID: PMC8611775 DOI: 10.1111/cns.13750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
AIMS Tumor electric fields therapy (TTFields) is emerging as a novel anti-cancer physiotherapy. Despite recent breakthroughs of TTFields in glioma treatment, the average survival time for glioblastoma patients with TTFields is <2 years, even when used in conjugation with traditional anti-cancer therapies. To optimize TTFields-afforded efficacy against glioblastoma, we investigated the cancer cell-killing effects of various TTFields paradigms using in vitro and in vivo models of glioblastoma. METHODS For in vitro studies, the U251 glioma cell line or primary cell cultures prepared from 20 glioblastoma patients were treated with the tumor electric field treatment (TEFT) system. Cell number, volume, and proliferation were measured after TEFT at different frequencies (100, 150, 180, 200, or 220 kHz), durations (24, 48, or 72 h), field strengths (1.0, 1.5, or 2.2V/cm), and output modes (fixed or random sequence output). A transwell system was used to evaluate the influence of TEFT on the invasiveness of primary glioblastoma cells. For in vivo studies, the therapeutic effect and safety profiles of random sequence electric field therapy in glioblastoma-transplanted rats were assessed by calculating tumor size and survival time and evaluating peripheral immunobiological and blood parameters, respectively. RESULTS In the in vitro settings, TEFT was robustly effective in suppressing cell proliferation of both the U251 glioma cell line and primary glioblastoma cell cultures. The anti-proliferation effects of TEFT were frequency- and "dose" (field strength and duration)-dependent, and contingent on the field sequence output mode, with the random sequence mode (TEFT-R) being more effective than the fixed sequence mode (TEFT-F). Genetic tests were performed in 11 of 20 primary glioblastoma cultures, and 6 different genetic traits were identified them. However, TEFT exhibited comparable anti-proliferation effects in all primary cultures regardless of their genetic traits. TEFT also inhibited the invasiveness of primary glioblastoma cells in transwell experiments. In the in vivo rat model of glioblastoma brain transplantation, treatment with TEFT-F or TEFT-R at frequency of 200 kHz and field strength of 2.2V/cm for 14 days significantly reduced tumor volume by 42.63% (TEFT-F vs. control, p = 0.0002) and 63.60% (TEFT-R vs. control, p < 0.0001), and prolonged animal survival time by 30.15% (TEFT-F vs. control, p = 0.0415) and 69.85% (TEFT-R vs. control, p = 0.0064), respectively. The tumor-bearing rats appeared to be well tolerable to TEFT therapies, showing only moderate increases in blood levels of creatine and red blood cells. Adverse skin reactions were common for TEFT-treated rats; however, skin reactions were curable by local treatment. CONCLUSION Tumor electric field treatment at optimal frequency, strength, and output mode markedly inhibits the cell viability, proliferation, and invasiveness of primary glioblastoma cells in vitro independent of different genetic traits of the cells. Moreover, a random sequence electric field output confers considerable anti-cancer effects against glioblastoma in vivo. Thus, TTFields are a promising physiotherapy for glioblastoma and warrants further investigation.
Collapse
Affiliation(s)
- Hao Wu
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lin Yang
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hanjie Liu
- Beijing Neurosurgical Institute; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dan Zhou
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Xiaoque Zheng
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui Yang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chong Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jiusheng Chang
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Zhifei Wang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Nianjun Ren
- Department of Neurosurgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuyang Liu
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Muyuan Jia
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jian Lu
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Hongyu Liu
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guochen Sun
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jialin Liu
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|