1
|
Yoo K, Yun HH, Jung SY, Im CN, Riew TR, Lee MY, Lee JH. KRIBB11 Exerts Anticancer Effects on A172 Glioblastoma Cells via the Cdh1/SKP2/p27 and HSF1/p53/p21 Pathways. Cancer Genomics Proteomics 2025; 22:467-477. [PMID: 40280718 PMCID: PMC12041876 DOI: 10.21873/cgp.20514] [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: 01/28/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/AIM KRIBB11, a heat shock factor1 (HSF1) inhibitor, sensitizes cancer cells to several anticancer drugs. We have previously demonstrated that KRIBB11 alone induced the apoptosis of A172 glioblastoma cells. However, the molecular basis of its anticancer activity remains unclear. Hence, we aimed to examine the alterations in cell cycle regulators and the relevance of HSF1 activity following KRIBB11 treatment in A172 cells. MATERIALS AND METHODS The expression levels of p21, p27, and p53 were determined using western blotting or real-time PCR. Alterations in p27 levels were induced using small interfering RNA and retroviral transfection. SKP2 degradation was analyzed through a cycloheximide chase assay. RESULTS p21 and p27 exhibited opposite expression profiles in A172 cells following KRIBB11 treatment, with p21 accumulating and p27 decreasing, respectively. Further experiments revealed that p21 induction could be attributed to HSF1-dependent p53 accumulation, which is responsible for cell cycle arrest and apoptosis. In contrast, p27 reduction was not reproduced by HSF1 silencing; however, further suppression of p27 accelerated poly (ADP-ribose) polymerase cleavage by KRIBB11 treatment, which was partially reversed by p27 overexpression. Thus, the reduction in p27 levels by KRIBB11 appeared favorable for apoptosis, suggesting that p27 functions as an oncogene in A172 cells. Subsequently, we demonstrated that the decrease in p27 levels following KRIBB11 exposure was mediated by the accumulation of the SKP2 protein, accompanied by a reduction in Cdh1 ubiquitin ligase. CONCLUSION KRIBB11 induces apoptotic cell death in A172 cells through two axes: HSF1-dependent p53/p21 accumulation and Cdh1/SKP2-dependent reduction of p27.
Collapse
Affiliation(s)
- Kyunghyun Yoo
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hye Hyeon Yun
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soon-Young Jung
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang-Nim Im
- The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tae-Ryong Riew
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mun-Yong Lee
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea;
- The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
2
|
Veviorskiy A, Mkrtchyan GV, Osipov AN, Izumchenko E, Ozerov IV, Aliper A, Zhavoronkov A, Scheibye-Knudsen M. Variability in radiotherapy outcomes across cancer types: a comparative study of glioblastoma multiforme and low-grade gliomas. Aging (Albany NY) 2025; 17:550-562. [PMID: 40015963 PMCID: PMC11892922 DOI: 10.18632/aging.206212] [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: 10/16/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025]
Abstract
Radiotherapy is a crucial treatment option for various cancers. However, the results of radiotherapy can vary widely across different cancer types and even among patients with the same type of cancer. This variability presents a major challenge in optimizing treatment strategies and improving patient survival. Here, we collected radiotherapy phenotype and expression data from 32 TCGA cancer datasets and performed overall survival analysis for 32 cancer types. Additionally, we conducted a signaling pathway enrichment analysis to identify key pathways involved in radiotherapy resistance and sensitivity. Our findings show that radiotherapy improves survival outcomes in certain cancer types, such as glioblasoma multiforme (GBM), while worsening outcomes in others, such as low-grade glioma (LGG). Next, we focused on exploring the differences in radiotherapy outcomes between GBM and LGG, focusing on the molecular mechanisms contributing to these variations. We identify differential regulation of pathways related to programmed cell death, DNA repair, telomere maintenance, chromosome condensation, antiviral responses, and interferon signaling between GBM and LGG patients perhaps explaining radiotherapy efficacy. A genetic analysis confirmed the importance of immune response and radiotherapy outcome for LGG patients. These insights underscore the importance of personalized treatment approaches and the need for further research to improve radiotherapy outcomes in cancer patients.
Collapse
Affiliation(s)
| | - Garik V. Mkrtchyan
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Denmark
| | | | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Ivan V. Ozerov
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Alex Aliper
- Insilico Medicine AI Limited, Abu Dhabi, UAE
| | - Alex Zhavoronkov
- Insilico Medicine AI Limited, Abu Dhabi, UAE
- Insilico Medicine Hong Kong Limited, Hong Kong Science and Technology Park, Hong Kong
- Insilico Medicine Canada Inc., René-Lévesque Blvd W, Montreal, Quebec H3B 4W8, Canada
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Denmark
| |
Collapse
|
3
|
Baskaran AB, Kozel OA, Venkatesh O, Wainwright DA, Sonabend AM, Heimberger AB, Lukas RV. Immune Checkpoint Inhibitors in Glioblastoma IDHwt Treatment: A Systematic Review. Cancers (Basel) 2024; 16:4148. [PMID: 39766048 PMCID: PMC11674442 DOI: 10.3390/cancers16244148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE A glioblastoma (GBM) is a primary brain tumor with significant unmet therapeutic needs. Immune checkpoint inhibitors (ICIs) have marked therapeutic benefits in many different cancers but have yet to show benefit for most GBM patients in phase III trials. METHODS A systematic review querying ClinicalTrials.gov for prospective clinical trials investigating ICI in GBM between 1950 and July 2024 was performed. Search terms comprised 11 distinct ICIs. Data abstracted include clinical trial NCT numbers with study titles and status, enrollment information, interventions, and more. Clinical trial identifying information, interventions, and outcomes were extracted. RESULTS One hundred and seventeen clinical trials were identified; four were phase 3. Most involved PD-1 or CTLA-4 blockade as monotherapy or in combination with standard-of-care. The large, randomized trials included CHECKMATE 143, CHECKMATE 498, CHECKMATE 548, and NRG BN007. These showed no improvement in median overall survival or progression-free survival in unselected patients. Biomarker-directed analyses suggest that a subset of GBM patients may benefit. CONCLUSIONS ICI for the treatment of GBM has not demonstrated clear evidence of efficacy thus far. This review serves as a quick reference of ICI trial results in GBM. Biomarker-driven patient selection and/or novel approaches to overcome resistance mechanisms remain areas of viable inquiry.
Collapse
Affiliation(s)
| | - Olivia A. Kozel
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Omkar Venkatesh
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA;
| | - Derek A. Wainwright
- Departments of Cancer Biology & Neurological Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA;
| | - Adam M. Sonabend
- Department of Neurosurgery, Northwestern University, Chicago, IL 60208, USA; (A.M.S.); (A.B.H.)
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL 60611, USA;
| | - Amy B. Heimberger
- Department of Neurosurgery, Northwestern University, Chicago, IL 60208, USA; (A.M.S.); (A.B.H.)
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL 60611, USA;
| | - Rimas Vincas Lukas
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL 60611, USA;
- Department of Neurology, Northwestern University, Chicago, IL 60208, USA
| |
Collapse
|
4
|
Hameedat F, Mendes BB, Conniot J, Di Filippo LD, Chorilli M, Schroeder A, Conde J, Sousa F. Engineering nanomaterials for glioblastoma nanovaccination. NATURE REVIEWS MATERIALS 2024; 9:628-642. [DOI: 10.1038/s41578-024-00684-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 01/03/2025]
|
5
|
Zhou L, Xiang H, Liu S, Chen H, Yang Y, Zhang J, Cai W. Folic Acid Functionalized AQ4N/Gd@PDA Nanoplatform with Real-Time Monitoring of Hypoxia Relief and Enhanced Synergistic Chemo/Photothermal Therapy in Glioma. Int J Nanomedicine 2024; 19:3367-3386. [PMID: 38617794 PMCID: PMC11012807 DOI: 10.2147/ijn.s451921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Purpose Hypoxia is often associated with glioma chemoresistance, and alleviating hypoxia is also crucial for improving treatment efficacy. However, although there are already some methods that can improve efficacy by alleviating hypoxia, real-time monitoring that can truly achieve hypoxia relief and efficacy feedback still needs to be explored. Methods AQ4N/Gd@PDA-FA nanoparticles (AGPF NPs) were synthesized using a one-pot method and were characterized. The effects of AGPF NPs on cell viability, cellular uptake, and apoptosis were investigated using the U87 cell line. Moreover, the effectiveness of AGPF NPs in alleviating hypoxia was explored in tumor-bearing mice through photoacoustic imaging. In addition, the diagnosis and treatment effect of AGPF NPs were evaluated by magnetic resonance imaging (MRI) and bioluminescent imaging (BLI) on orthotopic glioma mice respectively. Results In vitro experiments showed that AGPF NPs had good dispersion, stability, and controlled release. AGPF NPs were internalized by cells through endocytosis, and could significantly reduce the survival rate of U87 cells and increase apoptosis under irradiation. In addition, we monitored blood oxygen saturation at the tumor site in real-time through photoacoustic imaging (PAI), and the results showed that synergistic mild-photothermal therapy/chemotherapy effectively alleviated tumor hypoxia. Finally, in vivo anti-tumor experiments have shown that synergistic therapy can effectively alleviate hypoxia and inhibit the growth of orthotopic gliomas. Conclusion This work not only provides an effective means for real-time monitoring of the dynamic feedback between tumor hypoxia relief and therapeutic efficacy, but also offers a potential approach for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Longjiang Zhou
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Haitao Xiang
- Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, People’s Republic of China
| | - Susu Liu
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi’an, 710126, People’s Republic of China
| | - Honglin Chen
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Yuanwei Yang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Jianyong Zhang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Wei Cai
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| |
Collapse
|
6
|
Shields LB, O'Dell P, Daniels MW, Sevak PR, Highfield HA, Sinicrope KD, Sun DA, Spalding AC. Impact of Reirradiation Utilizing Fractionated Stereotactic Radiotherapy for Recurrent Glioblastoma. Cureus 2024; 16:e53001. [PMID: 38406061 PMCID: PMC10894660 DOI: 10.7759/cureus.53001] [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] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Patients with recurrent glioblastoma (GBM) have limited treatment options. This study determined whether patients with recurrent GBM treated with initial radiation/temozolomide (TMZ) and reirradiation using fractionated stereotactic radiotherapy (FSRT) had improved outcomes. MATERIALS AND METHODS We identified 95 patients with recurrent GBM, 50 of whom underwent FSRT at recurrence and 45 who had systemic treatment only (control). The median total FSRT dose at the time of GBM recurrence was 30 Gy in five fractions of the gadolinium-enhanced tumor only. RESULTS With a median follow-up of 18 months, the progression-free survival (PFS) and overall survival (OS) following initial GBM diagnosis were longer in the reirradiation group compared to the control group (13.5 vs. 7.5 months [p=0.001] and 24.6 vs. 12.6 months [p<0.001], respectively). For patients who underwent reirradiation, the median time interval between the end of the initial radiation and reirradiation was 15.2 months. The median OS after GBM recurrence was longer in the reirradiation group versus the control group (9.9 vs. 3.5 months [p<0.001]), with a one-year OS survival rate of 22%. The hazard ratio for death of patients in the reirradiation group was 0.31 [0.19-0.50]. The reirradiation group had a higher percentage of patients who received bevacizumab (BEV, 62.0% vs. 28.9%, p=0.002) and a lower percentage of patients whose TMZ was discontinued due to toxicity (8.0% vs. 28.9%, p=0.017) compared to the control group. CONCLUSIONS Reirradiation utilizing FSRT was associated with improved PFS and OS after GBM recurrence compared to the control group who did not receive additional irradiation.
Collapse
Affiliation(s)
- Lisa B Shields
- Norton Neuroscience Institute, Norton Healthcare, Louisville, USA
| | - Patrick O'Dell
- Norton Cancer Institute, Norton Healthcare, Louisville, USA
| | - Michael W Daniels
- Bioinformatics and Biostatistics, University of Louisville, Louisville, USA
| | - Parag R Sevak
- Norton Cancer Institute, Norton Healthcare, Louisville, USA
| | - Hilary A Highfield
- Clinical Pathology Accreditation (CPA) Laboratory, Norton Healthcare, Louisville, USA
| | | | - David A Sun
- Norton Neuroscience Institute, Norton Healthcare, Louisville, USA
| | | |
Collapse
|
7
|
Xu L, Duan H, Zou Y, Wang J, Liu H, Wang W, Zhu X, Chen J, Zhu C, Yin Z, Zhao X, Wang Q. Xihuang Pill-destabilized CD133/EGFR/Akt/mTOR cascade reduces stemness enrichment of glioblastoma via the down-regulation of SOX2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154764. [PMID: 36963368 DOI: 10.1016/j.phymed.2023.154764] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/20/2023] [Accepted: 03/12/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Our previous study found that XHP could induce GBM cells to undergo apoptosis. A lot of evidence suggests that glioma stem-like cells (GSCs) are key factors that contribute to disease progression and poor prognosis of glioblastoma multiforme (GBM). Traditional Chinese medicine has been applied in clinical practice as a complementary and alternative therapy for glioma. PURPOSE To evaluate the effect and the potential molecular mechanism of Xihuang pill (XHP) on GSCs. METHODS UPLC-QTOF-MS analysis was used for constituent analysis of XHP. Using network pharmacology and bioinformatics methods, a molecular network targeting GSCs by the active ingredients in XHP was constructed. Cell viability, self-renewal ability, apoptosis, and GSC markers were detected by CCK-8 assay, tumor sphere formation assay and flow cytometry, respectively. The interrelationship between GSC markers (CD133 and SOX2) and key proteins of the EGFR/Akt/mTOR signaling pathway was evaluated using GEPIA and verified by western blot. A GBM cell line stably overexpressing Akt was constructed using lentivirus to evaluate the role of Akt signaling in the regulation of glioma stemness. The effect of XHP on glioma growth was analyzed by a subcutaneously transplanted glioma cell model in nude mice, hematoxylin-eosin staining was used to examine pathological changes, TUNEL staining was used to detect apoptosis in tumor tissues, and the expression of GSC markers in tumor tissues was identified by western blot and immunofluorescence. RESULTS Bioinformatics analysis showed that 55 matched targets were related to XHP targets and glioma stem cell targets. In addition to causing apoptosis, XHP could diminish the number of GBM 3D spheroids, the proportion of CD133-positive cells and the expression level of GSC markers (CD133 and SOX2) in vitro. Furthermore, XHP could attenuate the expression of CD133, EGFR, p-Akt, p-mTOR and SOX2 in GBM spheres. Overexpression of Akt significantly increased the expression level of SOX2, which was prohibited in the presence of XHP. XHP reduced GSC markers including CD133 and SOX2, and impeded the development of glioma growth in xenograft mouse models in vivo. CONCLUSION We demonstrate for the first time that XHP down-regulates stemness, restrains self-renewal and induces apoptosis in GSCs and impedes glioma growth by down-regulating SOX2 through destabilizing the CD133/EGFR/Akt/mTOR cascade.
Collapse
Affiliation(s)
- Lanyang Xu
- Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China; Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hao Duan
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yuheng Zou
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jing Wang
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Huaxi Liu
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wanyu Wang
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiao Zhu
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiali Chen
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chuanwu Zhu
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhixin Yin
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaoshan Zhao
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Qirui Wang
- Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, China; Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
8
|
Targeted nano-delivery of chemotherapy via intranasal route suppresses in vivo glioblastoma growth and prolongs survival in the intracranial mouse model. Drug Deliv Transl Res 2023; 13:608-626. [PMID: 36245060 DOI: 10.1007/s13346-022-01220-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2022] [Indexed: 12/30/2022]
Abstract
Nanotechnology-based drug delivery platforms have shown great potential in overcoming the limitations of conventional therapy for glioblastoma (GBM). However, permeation across the blood-brain barrier (BBB), physiological complexity of the brain, and glioma targeting strategies cannot entirely meet the challenging requirements of distinctive therapeutic delivery stages. The objective of this research is to fabricate lipid nanoparticles (LNPs) for the co-delivery of paclitaxel (PTX) and miltefosine (HePc) a proapoptotic agent decorated with transferrin (Tf-PTX-LNPs) and investigate its anti-glioma activity both in vitro and in vivo orthotopic NOD/SCID GBM mouse model. The present study demonstrates the anti-glioma effect of the dual drug combination of PTX and proapoptotic HePc lipid-based transferrin receptor (TfR) targeted alternative delivery (direct nose to brain transportation) of the nanoparticulate system (Tf-PTX-LNPs, 364 ± 5 nm, -43 ± 9 mV) to overcome the O6-methylguanine-DNA methyltransferase induce drug-resistant for improving the effectiveness of GBM therapy. The resulting nasally targeted LNPs present good biocompatibility, stability, high BBB transcytosis through selective TfR-mediated uptake by tumor cells, and effective tumor penetration in the brain of GBM induced mice. We observed markedly enhanced anti-proliferative efficacy of the targeted LNPs in U87MG cells compared to free drug. Nasal targeted LNPs had shown significantly improved brain concentration (Cmax fivefold and AUC0-24 4.9 fold) with early tmax (0.5 h) than the free drug. In vivo intracranial GBM-bearing targeted LNPs treated mice exhibited significantly prolonged survival with improved anti-tumor efficacy accompanied by reduced toxicity compared to systemic Taxol® and nasal free drug. These findings indicate that the nasal delivery of targeted synergistic nanocarrier holds great promise as a non-invasive adjuvant chemotherapy therapy of GBM.
Collapse
|
9
|
Liang J, Sun J, Liu A, Chen L, Ma X, Liu X, Zhang C. Saikosaponin D improves chemosensitivity of glioblastoma by reducing the its stemness maintenance. Biochem Biophys Rep 2022; 32:101342. [PMID: 36186734 PMCID: PMC9516410 DOI: 10.1016/j.bbrep.2022.101342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Chemotherapy is one of the important adjuvant methods for the treatment of glioblastoma (GBM), and chemotherapy resistance is a clinical problem that neurooncologists need to solve urgently. It is reported that Saikosaponin D (SSD), an active component of Bupleurum chinense, had various of antitumor activities and could also enhance the chemosensitivity of liver cancer and other tumors. However, it is not clear whether it has an effect on the chemosensitivity of glioma and its specific mechanism. Methods The CCK8 assay, Wound healing assay and Matrigel invasion assay were used to detect the effect of SSD on the phenotype of GBM cells. We detected the effect of SSD on the chemosensitivity of GSM by Flow cytometry, LDH content and MTT assay. Then, we used cell plate cloning, semi-quantitative PCR and western blotting experiments to detect the effect of SSD on the stem potential of GBM cells. Finally, the effect of SSD on the chemosensitivity of GBM and its potential mechanism were verified by nude mouse experiments in vivo. Results firstly, we found that SSD could partially inhibit the malignant phenotype of LN-229 cells, including inhibiting migration, invasion and apoptosis, and increasing the apoptosis rate and lactate dehydrogenase (LDH) release of LN-229 cells under the treatment of temozolomide (TMZ), that is to say, increasing the chemotherapy effect of TMZ on the cells. In addition, we unexpectedly found that SSD could partially inhibit the colony forming ability of LN-229 cells, which directly related to the stemness maintenance potential of cancer stem cells. Subsequently, our results showed that SSD could inhibit the gene and protein expression of stemness factors (OCT4, SOX2, c-Myc and Klf4) in LN-229 cells. Finally, we verified that SSD could improve the chemotherapy effect of TMZ by inhibiting the stem potential of glioblastoma in vivo nude mice. Conclusion this research can provide a certain theoretical basis for the application of SSD in the chemotherapy resistance of GBM and its mechanism of action, and provide a new hope for the clinical treatment of glioblastoma. SSD could inhibit the malignant phenotype of LN-229 cells, increase the chemotherapy effect of TMZ on the cells. SSD could inhibit the colony forming ability of LN-229 cells, and also inhibit their gene and protein expression of stemness factors. We verified that SSD could improve the chemotherapy effect of TMZ by inhibiting the stem potential of glioblastoma.
Collapse
|
10
|
Dhiwar PS, Matada GSP, Raghavendra NM, Ghara A, Singh E, Abbas N, Andhale GS, Shenoy GP, Sasmal P. Current updates on EGFR and HER2 tyrosine kinase inhibitors for the breast cancer. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02934-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2025]
|
11
|
Rodriguez SMB, Staicu GA, Sevastre AS, Baloi C, Ciubotaru V, Dricu A, Tataranu LG. Glioblastoma Stem Cells-Useful Tools in the Battle against Cancer. Int J Mol Sci 2022; 23:ijms23094602. [PMID: 35562993 PMCID: PMC9100635 DOI: 10.3390/ijms23094602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma stem cells (GSCs) are cells with a self-renewal ability and capacity to initiate tumors upon serial transplantation that have been linked to tumor cell heterogeneity. Most standard treatments fail to completely eradicate GSCs, causing the recurrence of the disease. GSCs could represent one reason for the low efficacy of cancer therapy and for the short relapse time. Nonetheless, experimental data suggest that the presence of therapy-resistant GSCs could explain tumor recurrence. Therefore, to effectively target GSCs, a comprehensive understanding of their biology and the survival and developing mechanisms during treatment is mandatory. This review provides an overview of the molecular features, microenvironment, detection, and targeting strategies of GSCs, an essential information required for an efficient therapy. Despite the outstanding results in oncology, researchers are still developing novel strategies, of which one could be targeting the GSCs present in the hypoxic regions and invasive edge of the glioblastoma.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Clinical Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.M.B.R.); (V.C.); (L.G.T.)
| | - Georgiana-Adeline Staicu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy, 200349 Craiova, Romania; (G.-A.S.); (C.B.)
| | - Ani-Simona Sevastre
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Carina Baloi
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy, 200349 Craiova, Romania; (G.-A.S.); (C.B.)
| | - Vasile Ciubotaru
- Neurosurgical Department, Clinical Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.M.B.R.); (V.C.); (L.G.T.)
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy, 200349 Craiova, Romania; (G.-A.S.); (C.B.)
- Correspondence:
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Clinical Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.M.B.R.); (V.C.); (L.G.T.)
- Department 6—Clinical Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
12
|
Hsu JBK, Lee TY, Cheng SJ, Lee GA, Chen YC, Le NQK, Huang SW, Kuo DP, Li YT, Chang TH, Chen CY. Identification of Differentially Expressed Genes in Different Glioblastoma Regions and Their Association with Cancer Stem Cell Development and Temozolomide Response. J Pers Med 2021; 11:jpm11111047. [PMID: 34834399 PMCID: PMC8625522 DOI: 10.3390/jpm11111047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
The molecular heterogeneity of gene expression profiles of glioblastoma multiforme (GBM) are the most important prognostic factors for tumor recurrence and drug resistance. Thus, the aim of this study was to identify potential target genes related to temozolomide (TMZ) resistance and GBM recurrence. The genomic data of patients with GBM from The Cancer Genome Atlas (TCGA; 154 primary and 13 recurrent tumors) and a local cohort (29 primary and 4 recurrent tumors), samples from different tumor regions from a local cohort (29 tumor and 25 peritumoral regions), and Gene Expression Omnibus data (GSE84465, single-cell RNA sequencing; 3589 cells) were included in this study. Critical gene signatures were identified based an analysis of differentially expressed genes (DEGs). DEGs were further used to evaluate gene enrichment levels among primary and recurrent GBMs and different tumor regions through gene set enrichment analysis. Protein-protein interactions (PPIs) were incorporated into gene regulatory networks to identify the affected metabolic pathways. The enrichment levels of 135 genes were identified in the peritumoral regions as being risk signatures for tumor recurrence. Fourteen genes (DVL1, PRKACB, ARRB1, APC, MAPK9, CAMK2A, PRKCB, CACNA1A, ERBB4, RASGRF1, NF1, RPS6KA2, MAPK8IP2, and PPM1A) derived from the PPI network of 135 genes were upregulated and involved in the regulation of cancer stem cell (CSC) development and relevant signaling pathways (Notch, Hedgehog, Wnt, and MAPK). The single-cell data analysis results indicated that 14 key genes were mainly expressed in oligodendrocyte progenitor cells, which could produce a CSC niche in the peritumoral region. The enrichment levels of 336 genes were identified as biomarkers for evaluating TMZ resistance in the solid tumor region. Eleven genes (ARID5A, CDC42EP3, CDKN1A, FLT3, JUNB, MAP2K3, MYBPC2, RGS14, RNASEK, TBC1D30, and TXNDC11) derived from the PPI network of 336 genes were upregulated and may be associated with a high risk of TMZ resistance; these genes were identified in both the TCGA and local cohorts. Furthermore, the expression patterns of ARID5A, CDKN1A, and MAP2K3 were identical to the gene signatures of TMZ-resistant cell lines. The identified enrichment levels of the two gene sets expressed in tumor and peritumoral regions are potentially helpful for evaluating TMZ resistance in GBM. Moreover, these key genes could be used as biomarkers, potentially providing new molecular strategies for GBM treatment.
Collapse
Affiliation(s)
- Justin Bo-Kai Hsu
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (J.B.-K.H.); (G.A.L.); (S.-W.H.)
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
| | - Tzong-Yi Lee
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, China;
- School of Life and Health Science, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Sho-Jen Cheng
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (J.B.-K.H.); (G.A.L.); (S.-W.H.)
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yung-Chieh Chen
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Nguyen Quoc Khanh Le
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Professional Master’s Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Shiu-Wen Huang
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (J.B.-K.H.); (G.A.L.); (S.-W.H.)
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Duen-Pang Kuo
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yi-Tien Li
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 110, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (T.-H.C.); (C.-Y.C.); Tel.: +886-2-2737-2181 (C.-Y.C.)
| | - Cheng-Yu Chen
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; (S.-J.C.); (Y.-C.C.); (N.Q.K.L.); (D.-P.K.); (Y.-T.L.)
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei 110, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (T.-H.C.); (C.-Y.C.); Tel.: +886-2-2737-2181 (C.-Y.C.)
| |
Collapse
|
13
|
Frosina G. Radiotherapy of High-Grade Gliomas: First Half of 2021 Update with Special Reference to Radiosensitization Studies. Int J Mol Sci 2021; 22:8942. [PMID: 34445646 PMCID: PMC8396323 DOI: 10.3390/ijms22168942] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023] Open
Abstract
Albeit the effort to develop targeted therapies for patients with high-grade gliomas (WHO grades III and IV) is evidenced by hundreds of current clinical trials, radiation remains one of the few effective therapeutic options for them. This review article analyzes the updates on the topic "radiotherapy of high-grade gliomas" during the period 1 January 2021-30 June 2021. The high number of articles retrieved in PubMed using the search terms ("gliom* and radio*") and manually selected for relevance indicates the feverish research currently ongoing on the subject. During the last semester, significant advances were provided in both the preclinical and clinical settings concerning the diagnosis and prognosis of high-grade gliomas, their radioresistance, and the inevitable side effects of their treatment with radiation. The novel information concerning tumor radiosensitization was of special interest in terms of therapeutic perspective and was discussed in detail.
Collapse
Affiliation(s)
- Guido Frosina
- Mutagenesis & Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
14
|
Wagner PM, Prucca CG, Caputto BL, Guido ME. Adjusting the Molecular Clock: The Importance of Circadian Rhythms in the Development of Glioblastomas and Its Intervention as a Therapeutic Strategy. Int J Mol Sci 2021; 22:8289. [PMID: 34361055 PMCID: PMC8348990 DOI: 10.3390/ijms22158289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Gliomas are solid tumors of the central nervous system (CNS) that originated from different glial cells. The World Health Organization (WHO) classifies these tumors into four groups (I-IV) with increasing malignancy. Glioblastoma (GBM) is the most common and aggressive type of brain tumor classified as grade IV. GBMs are resistant to conventional therapies with poor prognosis after diagnosis even when the Stupp protocol that combines surgery and radiochemotherapy is applied. Nowadays, few novel therapeutic strategies have been used to improve GBM treatment, looking for higher efficiency and lower side effects, but with relatively modest results. The circadian timing system temporally organizes the physiology and behavior of most organisms and daily regulates several cellular processes in organs, tissues, and even in individual cells, including tumor cells. The potentiality of the function of the circadian clock on cancer cells modulation as a new target for novel treatments with a chronobiological basis offers a different challenge that needs to be considered in further detail. The present review will discuss state of the art regarding GBM biology, the role of the circadian clock in tumor progression, and new chrono-chemotherapeutic strategies applied for GBM treatment.
Collapse
Affiliation(s)
- Paula M. Wagner
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (P.M.W.); (C.G.P.); (B.L.C.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - César G. Prucca
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (P.M.W.); (C.G.P.); (B.L.C.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Beatriz L. Caputto
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (P.M.W.); (C.G.P.); (B.L.C.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Mario E. Guido
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (P.M.W.); (C.G.P.); (B.L.C.)
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| |
Collapse
|
15
|
Adjusting the Molecular Clock: The Importance of Circadian Rhythms in the Development of Glioblastomas and Its Intervention as a Therapeutic Strategy. Int J Mol Sci 2021; 22:8289. [PMID: 34361055 PMCID: PMC8348990 DOI: 10.3390/ijms22158289;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Gliomas are solid tumors of the central nervous system (CNS) that originated from different glial cells. The World Health Organization (WHO) classifies these tumors into four groups (I-IV) with increasing malignancy. Glioblastoma (GBM) is the most common and aggressive type of brain tumor classified as grade IV. GBMs are resistant to conventional therapies with poor prognosis after diagnosis even when the Stupp protocol that combines surgery and radiochemotherapy is applied. Nowadays, few novel therapeutic strategies have been used to improve GBM treatment, looking for higher efficiency and lower side effects, but with relatively modest results. The circadian timing system temporally organizes the physiology and behavior of most organisms and daily regulates several cellular processes in organs, tissues, and even in individual cells, including tumor cells. The potentiality of the function of the circadian clock on cancer cells modulation as a new target for novel treatments with a chronobiological basis offers a different challenge that needs to be considered in further detail. The present review will discuss state of the art regarding GBM biology, the role of the circadian clock in tumor progression, and new chrono-chemotherapeutic strategies applied for GBM treatment.
Collapse
|
16
|
Tang X, Zuo C, Fang P, Liu G, Qiu Y, Huang Y, Tang R. Targeting Glioblastoma Stem Cells: A Review on Biomarkers, Signal Pathways and Targeted Therapy. Front Oncol 2021; 11:701291. [PMID: 34307170 PMCID: PMC8297686 DOI: 10.3389/fonc.2021.701291] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) remains the most lethal and common primary brain tumor, even after treatment with multiple therapies, such as surgical resection, chemotherapy, and radiation. Although great advances in medical development and improvements in therapeutic methods of GBM have led to a certain extension of the median survival time of patients, prognosis remains poor. The primary cause of its dismal outcomes is the high rate of tumor recurrence, which is closely related to its resistance to standard therapies. During the last decade, glioblastoma stem cells (GSCs) have been successfully isolated from GBM, and it has been demonstrated that these cells are likely to play an indispensable role in the formation, maintenance, and recurrence of GBM tumors, indicating that GSCs are a crucial target for treatment. Herein, we summarize the current knowledge regarding GSCs, their related signaling pathways, resistance mechanisms, crosstalk linking mechanisms, and microenvironment or niche. Subsequently, we present a framework of targeted therapy for GSCs based on direct strategies, including blockade of the pathways necessary to overcome resistance or prevent their function, promotion of GSC differentiation, virotherapy, and indirect strategies, including targeting the perivascular, hypoxic, and immune niches of the GSCs. In summary, targeting GSCs provides a tremendous opportunity for revolutionary approaches to improve the prognosis and therapy of GBM, despite a variety of challenges.
Collapse
Affiliation(s)
- Xuejia Tang
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China.,Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chenghai Zuo
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pengchao Fang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guojing Liu
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yongyi Qiu
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Huang
- Department of Neurosurgery, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Rongrui Tang
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|