1
|
Gennari AG, Doniselli FM, Coley J, Grisoli M, Quaia E, Souchon R, Prada F, DiMeco F. Intraoperative Comparison Between Strain Elastography and Preoperative Magnetic Resonance Imaging Features in High-Grade Gliomas Using Fusion Imaging: A Pilot Study. World Neurosurg 2024:S1878-8750(24)01561-4. [PMID: 39265936 DOI: 10.1016/j.wneu.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
OBJECTIVE To compare the elastographic patterns of high-grade gliomas (HGGs) solid portions and those of adjacent healthy brain parenchyma, on intraoperative ultrasound, with magnetic resonance image (MRI) characteristics. METHODS Clinical records and images of HGGs patients, operated between June and December 2018, were retrospectively reviewed. Fusion images were used to compare preoperative gadolinium-enhanced T1-weighted MRI/fluid-attenuated inversion recovery images (Gd-T1 MRI/FLAIR) to intraoperative strain elastography (SE). FLAIR/Gd-T1 MRI images were used to define enhancement patterns (absent/whole lesion/peripheral) and lesions' characteristics (primary and secondary pattern, further subdivided in solid/necrotic/cystic/infiltrating). HGGs SE patterns were categorized as homogeneous/inhomogeneous, while lesions' primary and secondary patterns as stiff/intermediate/elastic. The SE motive of neighboring healthy brain parenchyma was defined similarly. RESULTS Eighteen patients (M:F, 11:7; mean age: 53 years) harboring 14 glioblastomas (77.8%) and 4 anaplastic astrocytomas (22.2%) were compared. Glioblastomas typically enhanced peripherally and had a primary necrotic pattern (78.6% and 64.3%, respectively), while anaplastic astrocytomas did not enhance and were solid (75% both) at T1-Gd MRI and FLAIR images. At SE anaplastic astrocytomas had a homogeneous stiff primary pattern, whereas the majority of glioblastomas primary patterns were heterogeneous (85.7%) and intermediate (78.6%). CONCLUSIONS Three major SE patterns defined HGGs and adjacent healthy brain parenchyma. SE patterns varied according to HGG histotypes and Gd-T1 MRI/FLAIR characteristics.
Collapse
Affiliation(s)
- Antonio Giulio Gennari
- Department of Neuropediatrics, University Children's Hospital, Zurich, Switzerland; Center for MR Research, University Children's Hospital, Zurich, Switzerland
| | - Fabio Martino Doniselli
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy
| | - Justin Coley
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Marina Grisoli
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy
| | - Emilio Quaia
- Department of Radiology, University of Padova, Padova, Italy
| | | | - Francesco Prada
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, USA; Acoustic Neuroimaging and Therapy Lab, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy; Focused Ultrasound Foundation, Charlottesville, Virginia, USA; Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy.
| | - Francesco DiMeco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy; Department of Neurological Surgery, Johns Hopkins Medical School, Baltimore, Maryland, USA; Department of Oncology and Hemato-oncology, Università degli studi di Milano, Milano, Italy
| |
Collapse
|
2
|
Papacocea SI, Vrinceanu D, Dumitru M, Manole F, Serboiu C, Papacocea MT. Molecular Profile as an Outcome Predictor in Glioblastoma along with MRI Features and Surgical Resection: A Scoping Review. Int J Mol Sci 2024; 25:9714. [PMID: 39273661 PMCID: PMC11395592 DOI: 10.3390/ijms25179714] [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: 07/25/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive malignant tumors of the brain. We queried PubMed for articles about molecular predictor markers in GBM. This scoping review aims to analyze the most important outcome predictors in patients with GBM and to compare these factors in terms of absolute months of survival benefit and percentages. Performing a gross total resection for patients with GBM undergoing optimal chemo- and radiotherapy provides a significant benefit in overall survival compared to those patients who received a subtotal or partial resection. However, compared to IDH-Wildtype GBMs, patients with IDH-Mutant 1/2 GBMs have an increased survival. MGMT promoter methylation status is another strong outcome predictor for patients with GBM. In the reviewed literature, patients with methylated MGMT promoter lived approximately 50% to 90% longer than those with an unmethylated MGMT gene promoter. Moreover, KPS is an important predictor of survival and quality of life, demonstrating that we should refrain from aggressive surgery in important brain areas. As new therapies (such as TTFs) emerge, we are optimistic that the overall median survival will increase, even for IDH-Wildtype GBMs. In conclusion, molecular profiles are stronger outcome predictors than the extent of neurosurgical resection for GBM.
Collapse
Affiliation(s)
- Serban Iancu Papacocea
- Neurosurgery Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniela Vrinceanu
- ENT Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihai Dumitru
- ENT Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Felicia Manole
- ENT Department, Faculty of Medicine, University of Oradea, 410073 Oradea, Romania
| | - Crenguta Serboiu
- Cellular Biology and Histology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Marius Toma Papacocea
- Neurosurgery Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
3
|
Al-khatib SM, Al-Bzour AN, Almajali MN, Jarrad TA, AL-Eitan LN, Abdo N. Analysis of IDH and EGFR as biomarkers in glioblastoma multiforme: A case-control study. Heliyon 2024; 10:e35323. [PMID: 39165999 PMCID: PMC11333891 DOI: 10.1016/j.heliyon.2024.e35323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024] Open
Abstract
Background Glioblastoma multiforme (GBM) is a very aggressive primary central nervous system (CNS) tumor with limited therapeutic options and poor prognosis. This study aimed to analyze the association between single nucleotide polymorphisms (SNPs), including IDH1 rs121913500C > T, IDH2 rs11540478G > A, and EGFR rs1468727C > T, and their association on the risk and overall survival of GBM patients in Jordan. Methods Using a case-control study design involving 63 GBM patients and 226 healthy controls was conducted at King Abdullah University Hospital in Jordan. DNA extraction was performed using formalin-fixed and paraffin-embedded tissue for GBM samples and blood samples for controls. SNPs analysis was performed using the Sequenom iPLEX assay sequencing technique. Survival outcomes were assessed using Cox models and hazard ratios (HR), and single-cell RNA (scRNA) analysis was performed from GSE70630. Results The study showed a significant association between genotype frequency in GBM cases and controls for specific SNPs, including IDH1 rs121913500C > T, and EGFR rs1468727C > T. The G/G genotype of rs11540478 (IDH2) was associated with better prognostic outcomes in GBM patients. The scRNA analysis demonstrated the differential expression of IDH1, IDH2, and EGFR in GBM, with enrichment in central carbon metabolism in cancer. Conclusion Our findings suggest that SNPs, particularly in IDH1 and IDH2 genes and EGFR, may serve as diagnostic and prognostic biomarkers for GBM. While the study underscores the clinical relevance of these genetic variants, further investigations with larger and more diverse populations are essential to validate and extend these associations.
Collapse
Affiliation(s)
- Sohaib M. Al-khatib
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ayah N. Al-Bzour
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad N. Almajali
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Tariq A. Jarrad
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Laith N. AL-Eitan
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Nour Abdo
- Department of Public Health, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
4
|
Yan T, Jiang Q, Ni G, Ma H, Meng Y, Kang G, Xu M, Peng F, Li H, Chen X, Wang M. WZ-3146 acts as a novel small molecule inhibitor of KIF4A to inhibit glioma progression by inducing apoptosis. Cancer Cell Int 2024; 24:221. [PMID: 38937742 PMCID: PMC11209999 DOI: 10.1186/s12935-024-03409-y] [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: 03/12/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Glioma is considered the most common primary malignant tumor of the central nervous system. Although traditional treatments have not achieved satisfactory outcomes, recently, targeted therapies for glioma have shown promising efficacy. However, due to the single-target nature of targeted therapy, traditional targeted therapies are ineffective; thus, novel therapeutic targets are urgently needed. METHODS The gene expression data for glioma patients were derived from the GEO (GSE4290, GSE50161), TCGA and CGGA databases. Next, the upregulated genes obtained from the above databases were cross-analyzed, finally, 10 overlapping genes (BIRC5, FOXM1, EZH2, CDK1, KIF11, KIF4A, NDC80, PBK, RRM2, and TOP2A) were ultimately screened and only KIF4A expression has the strongest correlation with clinical characteristics in glioma patients. Futher, the TCGA and CGGA database were utilized to explore the correlation of KIF4A expression with glioma prognosis. Then, qRT-PCR and Western blot was used to detect the KIF4A mRNA and protein expression level in glioma cells, respectively. And WZ-3146, the small molecule inhibitor targeting KIF4A, were screened by Cmap analysis. Subsequently, the effect of KIF4A knockdown or WZ-3146 treatment on glioma was measured by the MTT, EdU, Colony formation assay and Transwell assay. Ultimately, GSEA enrichment analysis was performed to find that the apoptotic pathway could be regulated by KIF4A in glioma, in addition, the effect of WZ-3146 on glioma apoptosis was detected by flow cytometry and Western blot. RESULTS In the present study, we confirmed that KIF4A is abnormally overexpressed in glioma. In addition, KIF4A overexpression is a key indicator of glioma prognosis; moreover, suppressing KIF4A expression can inhibit glioma progression. We also discovered that WZ-3146, a small molecule inhibitor of KIF4A, can induce apoptosis in glioma cells and exhibit antiglioma effects. CONCLUSION In conclusion, these observations demonstrated that targeting KIF4A can inhibit glioma progression. With further research, WZ-3146, a small molecule inhibitor of KIF4A, could be combined with other molecular targeted drugs to cooperatively inhibit glioma progression.
Collapse
Affiliation(s)
- Tao Yan
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Qing Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, Heilongjiang Province, 150001, China
| | - Guangpu Ni
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Haofeng Ma
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Yun Meng
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Guiqiong Kang
- Central Laboratory, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Meifang Xu
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
- Department of Neurology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Huadong Li
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China.
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China.
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, Heilongjiang Province, 150001, China.
| | - Mingguang Wang
- Department of Neurosurgery, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China.
| |
Collapse
|
5
|
Yun HS, Kramp TR, Palanichamy K, Tofilon PJ, Camphausen K. MGMT inhibition regulates radioresponse in GBM, GSC, and melanoma. Sci Rep 2024; 14:12363. [PMID: 38811596 PMCID: PMC11136993 DOI: 10.1038/s41598-024-61240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
Radiotherapy is the standard treatment for glioblastoma (GBM), but the overall survival rate for radiotherapy treated GBM patients is poor. The use of adjuvant and concomitant temozolomide (TMZ) improves the outcome; however, the effectiveness of this treatment varies according to MGMT levels. Herein, we evaluated whether MGMT expression affected the radioresponse of human GBM, GBM stem-like cells (GSCs), and melanoma. Our results indicated a correlation between MGMT promoter methylation status and MGMT expression. MGMT-producing cell lines ACPK1, GBMJ1, A375, and MM415 displayed enhanced radiosensitivity when MGMT was silenced using siRNA or when inhibited by lomeguatrib, whereas the OSU61, NSC11, WM852, and WM266-4 cell lines, which do not normally produce MGMT, displayed reduced radiosensitivity when MGMT was overexpressed. Mechanistically lomeguatrib prolonged radiation-induced γH2AX retention in MGMT-producing cells without specific cell cycle changes, suggesting that lomeguatrib-induced radiosensitization in these cells is due to radiation-induced DNA double-stranded break (DSB) repair inhibition. The DNA-DSB repair inhibition resulted in cell death via mitotic catastrophe in MGMT-producing cells. Overall, our results demonstrate that MGMT expression regulates radioresponse in GBM, GSC, and melanoma, implying a role for MGMT as a target for radiosensitization.
Collapse
Affiliation(s)
- Hong Shik Yun
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, 9000 Rockville Pike, Building 10, Bethesda, MD, 20892, USA
| | - Tamalee R Kramp
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, 9000 Rockville Pike, Building 10, Bethesda, MD, 20892, USA
| | - Kamalakannan Palanichamy
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Philip J Tofilon
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, 9000 Rockville Pike, Building 10, Bethesda, MD, 20892, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, 9000 Rockville Pike, Building 10, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Zhao H, Meng L, Du P, Liao X, Mo X, Gong M, Chen J, Liao Y. IDH1 mutation produces R-2-hydroxyglutarate (R-2HG) and induces mir-182-5p expression to regulate cell cycle and tumor formation in glioma. Biol Res 2024; 57:30. [PMID: 38760850 PMCID: PMC11100189 DOI: 10.1186/s40659-024-00512-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/02/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2), are present in most gliomas. IDH1 mutation is an important prognostic marker in glioma. However, its regulatory mechanism in glioma remains incompletely understood. RESULTS miR-182-5p expression was increased within IDH1-mutant glioma specimens according to TCGA, CGGA, and online dataset GSE119740, as well as collected clinical samples. (R)-2-hydroxyglutarate ((R)-2HG) treatment up-regulated the expression of miR-182-5p, enhanced glioma cell proliferation, and suppressed apoptosis; miR-182-5p inhibition partially eliminated the oncogenic effects of R-2HG upon glioma cells. By direct binding to Cyclin Dependent Kinase Inhibitor 2 C (CDKN2C) 3'UTR, miR-182-5p inhibited CDKN2C expression. Regarding cellular functions, CDKN2C knockdown promoted R-2HG-treated glioma cell viability, suppressed apoptosis, and relieved cell cycle arrest. Furthermore, CDKN2C knockdown partially attenuated the effects of miR-182-5p inhibition on cell phenotypes. Moreover, CDKN2C knockdown exerted opposite effects on cell cycle check point and apoptosis markers to those of miR-182-5p inhibition; also, CDKN2C knockdown partially attenuated the functions of miR-182-5p inhibition in cell cycle check point and apoptosis markers. The engineered CS-NPs (antagomir-182-5p) effectively encapsulated and delivered antagomir-182-5p, enhancing anti-tumor efficacy in vivo, indicating the therapeutic potential of CS-NPs(antagomir-182-5p) in targeting the miR-182-5p/CDKN2C axis against R-2HG-driven oncogenesis in mice models. CONCLUSIONS These insights highlight the potential of CS-NPs(antagomir-182-5p) to target the miR-182-5p/CDKN2C axis, offering a promising therapeutic avenue against R-2HG's oncogenic influence to glioma.
Collapse
Affiliation(s)
- Haiting Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
- Department of Neurology, Xiangya Hospital, The Central South University (CSU), Changsha, 410008, P.R. China
| | - Li Meng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
- Department of Radiology, Xiangya Hospital, Central South University (CSU), Changsha, 410008, P.R. China
| | - Peng Du
- Department of Neurosurgery, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, 830063, PR China
| | - Xinbin Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, 410008, P.R. China
| | - Xin Mo
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, 410008, P.R. China
| | - Mengqi Gong
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, 410008, P.R. China
| | - Jiaxin Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
- Department of Neurology, Xiangya Hospital, The Central South University (CSU), Changsha, 410008, P.R. China
| | - Yiwei Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China.
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, 410008, P.R. China.
| |
Collapse
|
7
|
Lv W, Lin S, Zuo Z, Huang Z, Wang Y. Involvement of microglia-expressed MS4A6A in the onset of glioblastoma. Eur J Neurosci 2024; 59:2836-2849. [PMID: 38488530 DOI: 10.1111/ejn.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 05/22/2024]
Abstract
Glioblastoma multiforme (GBM) represents the deadliest form of brain tumour, characterized by its low survival rate and grim prognosis. Cytokines released from glioma-associated microglia/macrophages are involved in establishing the tumour microenvironment, thereby crucially promoting GBM progression. MS4A6A polymorphism was confirmed to be associated with neurodegenerative and polymorphism disease pathobiology, but whether it participates in the regulation of GBM and the underlying mechanisms is still not elucidated. Here, we found that MS4A6A was significantly upregulated in GBM patient samples. The results from the single-cell RNA-sequencing (scRNA-seq) database and immunostaining demonstrated the specific expression of MS4A6A in microglial cells. In vitro, microglial overexpression of MS4A6A stimulated the proliferation and migration of glioblastoma cells. Moreover, high MS4A6A mRNA expression was related to poor prognosis in GBM patients. Our study highlights the potential of MS4A6A as a promising biomarker for GBM, which may provide novel strategies for its prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Wenhao Lv
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shengyan Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhenxing Zuo
- Department of Neurosurgery, Tenth people's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Liu H, Bao X, Zeng Z, Liu W, Li M. Analysis of cuproptosis-related genes in prognosis and immune infiltration in grade 4 diffuse gliomas. Heliyon 2024; 10:e29212. [PMID: 38633656 PMCID: PMC11021980 DOI: 10.1016/j.heliyon.2024.e29212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Background Grade 4 diffuse gliomas are highly malignant tumours with poor prognosis. Cuproptosis is a novel form of cell death. Cuproptosis genes are associated with various tumours and affect the prognosis of patients with these tumours. However, the relationship between cuproptosis and grade 4 diffuse gliomas remains unclear. Methods Differentially expressed genes associated with cuproptosis in grade 4 diffuse gliomas were identified. Second, the prognostic model was established by univariate and multivariate COX regression analyses, and the genes (p < 0.05) were selected for subsequent analysis. The endpoint of the study was death. Single-gene analysis was performed in accordance with the expression levels of SLC31A1. Third, based on the expression levels of SLC31A1, gene function enrichment, drug sensitivity, and immune cell infiltration analyses were performed. Finally, the expression and biological functions of SLC31A1 in grade 4 diffuse gliomas were identified using immunohistochemical staining, qRT-PCR, and related biological experiments. Results We identified six coproptosis genes in the grade 4 diffuse gliomas dataset (SLC31A1, PDHA1, GLS, FDX1, LIPT1, and ATP7B). The six key cuproptosis genes of grade 4 diffuse gliomas were analysed using univariate COX analysis. Basic patient data, including age, race, year of diagnosis, sex, and treatment, were included in the univariate COX analysis. Then, multivariate COX analysis was performed for the factors with p < 0.2 in the univariate COX analysis. Age, year of diagnosis, and SLC31A1, PDHA1, and FDX1 levels were found to be independent prognostic factors. A nomogram was constructed using these 5 factors. Through experiments, we found that SLC31A1 had a higher expression level in cancer tissue than that near cancer among the three genes, SLC31A1, PDHA1, and FDX1; therefore, we focused on SLC31A1. According on the expression level of SLC31A1, we performed gene function enrichment, drug sensitivity, and immune cell infiltration analyses. Navitoclax was the most sensitive drug. Differential gene function enrichment was observed for metalloendopeptidase activity. SLC31A1 is expressed in dendritic cells, macrophages, neutrophils, and CD8+T cells. SLC31A1 is highly expressed in grade 4 diffuse gliomas, whereas SLC31A1 knockdown significantly reduces cell proliferation and mobility. Conclusions Age, year of diagnosis, and SLC31A1, PDHA1, and FDX1 expression were independent prognostic factors. A nomogram was constructed based on age, year of diagnosis, and SLC31A1, PDHA1, and FDX1 levels. Through analysis and experimental verification, SLC31A1 was found to affect the prognosis and progression of patients with grade 4 diffuse gliomas and was associated with immune cell infiltration.
Collapse
Affiliation(s)
- Hui Liu
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin Bao
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhirui Zeng
- School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Wei Liu
- Department of Oncology, Changle County People's Hospital, Weifang, China
| | - Meifang Li
- Department of Oncology, Changle County People's Hospital, Weifang, China
| |
Collapse
|
9
|
Rios SA, Oyervides S, Uribe D, Reyes AM, Fanniel V, Vazquez J, Keniry M. Emerging Therapies for Glioblastoma. Cancers (Basel) 2024; 16:1485. [PMID: 38672566 PMCID: PMC11048459 DOI: 10.3390/cancers16081485] [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: 02/16/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma is most commonly a primary brain tumor and the utmost malignant one, with a survival rate of approximately 12-18 months. Glioblastoma is highly heterogeneous, demonstrating that different types of cells from the same tumor can manifest distinct gene expression patterns and biological behaviors. Conventional therapies such as temozolomide, radiation, and surgery have limitations. As of now, there is no cure for glioblastoma. Alternative treatment methods to eradicate glioblastoma are discussed in this review, including targeted therapies to PI3K, NFKβ, JAK-STAT, CK2, WNT, NOTCH, Hedgehog, and TGFβ pathways. The highly novel application of oncolytic viruses and nanomaterials in combating glioblastoma are also discussed. Despite scores of clinical trials for glioblastoma, the prognosis remains poor. Progress in breaching the blood-brain barrier with nanomaterials and novel avenues for targeted and combination treatments hold promise for the future development of efficacious glioblastoma therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Megan Keniry
- School of Integrative Biological and Chemical Sciences, College of Sciences, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; (S.A.R.); (D.U.); (A.M.R.)
| |
Collapse
|
10
|
Manzanares-Guzmán A, Lugo-Fabres PH, Camacho-Villegas TA. vNARs as Neutralizing Intracellular Therapeutic Agents: Glioblastoma as a Target. Antibodies (Basel) 2024; 13:25. [PMID: 38534215 DOI: 10.3390/antib13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Glioblastoma is the most prevalent and fatal form of primary brain tumors. New targeted therapeutic strategies for this type of tumor are imperative given the dire prognosis for glioblastoma patients and the poor results of current multimodal therapy. Previously reported drawbacks of antibody-based therapeutics include the inability to translocate across the blood-brain barrier and reach intracellular targets due to their molecular weight. These disadvantages translate into poor target neutralization and cancer maintenance. Unlike conventional antibodies, vNARs can permeate tissues and recognize conformational or cryptic epitopes due to their stability, CDR3 amino acid sequence, and smaller molecular weight. Thus, vNARs represent a potential antibody format to use as intrabodies or soluble immunocarriers. This review comprehensively summarizes key intracellular pathways in glioblastoma cells that induce proliferation, progression, and cancer survival to determine a new potential targeted glioblastoma therapy based on previously reported vNARs. The results seek to support the next application of vNARs as single-domain antibody drug-conjugated therapies, which could overcome the disadvantages of conventional monoclonal antibodies and provide an innovative approach for glioblastoma treatment.
Collapse
Affiliation(s)
- Alejandro Manzanares-Guzmán
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico
| | - Pavel H Lugo-Fabres
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico
| | - Tanya A Camacho-Villegas
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico
| |
Collapse
|
11
|
Gondarenko E, Mazur D, Masliakova M, Ryabukha Y, Kasheverov I, Utkin Y, Tsetlin V, Shahparonov M, Kudryavtsev D, Antipova N. Subtype-Selective Peptide and Protein Neurotoxic Inhibitors of Nicotinic Acetylcholine Receptors Enhance Proliferation of Patient-Derived Glioblastoma Cell Lines. Toxins (Basel) 2024; 16:80. [PMID: 38393158 PMCID: PMC10891657 DOI: 10.3390/toxins16020080] [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: 12/05/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain cancer, with a poor prognosis. GBM cells, which develop in the environment of neural tissue, often exploit neurotransmitters and their receptors to promote their own growth and invasion. Nicotinic acetylcholine receptors (nAChRs), which play a crucial role in central nervous system signal transmission, are widely represented in the brain, and GBM cells express several subtypes of nAChRs that are suggested to transmit signals from neurons, promoting tumor invasion and growth. Analysis of published GBM transcriptomes revealed spatial heterogeneity in nAChR subtype expression, and functional nAChRs of α1*, α7, and α9 subtypes are demonstrated in our work on several patient-derived GBM microsphere cultures and on the U87MG GBM cell line using subtype-selective neurotoxins and fluorescent calcium mobilization assay. The U87MG cell line shows reactions to nicotinic agonists similar to those of GBM patient-derived culture. Selective α1*, α7, and α9 nAChR neurotoxins stimulated cell growth in the presence of nicotinic agonists. Several cultivating conditions with varying growth factor content have been proposed and tested. The use of selective neurotoxins confirmed that cell cultures obtained from patients are representative GBM models, but the use of media containing fetal bovine serum can lead to alterations in nAChR expression and functioning.
Collapse
Affiliation(s)
- Elena Gondarenko
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Diana Mazur
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Marina Masliakova
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Yana Ryabukha
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Igor Kasheverov
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Yuri Utkin
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Victor Tsetlin
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Mikhail Shahparonov
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Denis Kudryavtsev
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Nadine Antipova
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| |
Collapse
|
12
|
Shah S. Novel Therapies in Glioblastoma Treatment: Review of Glioblastoma; Current Treatment Options; and Novel Oncolytic Viral Therapies. Med Sci (Basel) 2023; 12:1. [PMID: 38249077 PMCID: PMC10801585 DOI: 10.3390/medsci12010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
One of the most prevalent primary malignant brain tumors is glioblastoma (GB). About 6 incidents per 100,000 people are reported annually. Most frequently, these tumors are linked to a poor prognosis and poor quality of life. There has been little advancement in the treatment of GB. In recent years, some innovative medicines have been tested for the treatment of newly diagnosed cases of GB and recurrent cases of GB. Surgery, radiotherapy, and alkylating chemotherapy are all common treatments for GB. A few of the potential alternatives include immunotherapy, tumor-treating fields (TTFs), and medications that target specific cellular receptors. To provide new multimodal therapies that focus on the molecular pathways implicated in tumor initiation and progression in GB, novel medications, delivery technologies, and immunotherapy approaches are being researched. Of these, oncolytic viruses (OVs) are among the most recent. Coupling OVs with certain modern treatment approaches may have significant benefits for GB patients. Here, we discuss several OVs and how they work in conjunction with other therapies, as well as virotherapy for GB. The study was based on the PRISMA guidelines. Systematic retrieval of information was performed on PubMed. A total of 307 articles were found in a search on oncolytic viral therapies for glioblastoma. Out of these 83 articles were meta-analyses, randomized controlled trials, reviews, and systematic reviews. A total of 42 articles were from the years 2018 to 2023. Appropriate studies were isolated, and important information from each of them was understood and entered into a database from which the information was used in this article. One of the most prevalent malignant brain tumors is still GB. Significant promise and opportunity exist for oncolytic viruses in the treatment of GB and in boosting immune response. Making the most of OVs in the treatment of GB requires careful consideration and evaluation of a number of its application factors.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
13
|
Mellor NG, Chung SA, Graham ES, Day BW, Unsworth CP. Eliciting calcium transients with UV nanosecond laser stimulation in adult patient-derived glioblastoma brain cancer cells in vitro. J Neural Eng 2023; 20:066026. [PMID: 37988746 DOI: 10.1088/1741-2552/ad0e7d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/21/2023] [Indexed: 11/23/2023]
Abstract
Objective.Glioblastoma (GBM) is the most common and lethal type of high-grade adult brain cancer. The World Health Organization have classed GBM as an incurable disease because standard treatments have yielded little improvement with life-expectancy being 6-15 months after diagnosis. Different approaches are now crucial to discover new knowledge about GBM communication/function in order to establish alternative therapies for such an aggressive adult brain cancer. Calcium (Ca2+) is a fundamental cell molecular messenger employed in GBM being involved in a wide dynamic range of cellular processes. Understanding how the movement of Ca2+behaves and modulates activity in GBM at the single-cell level is relatively unexplored but holds the potential to yield opportunities for new therapeutic strategies and approaches for cancer treatment.Approach.In this article we establish a spatially and temporally precise method for stimulating Ca2+transients in three patient-derived GBM cell-lines (FPW1, RN1, and RKI1) such that Ca2+communication can be studied from single-cell to larger network scales. We demonstrate that this is possible by administering a single optimized ultra-violet (UV) nanosecond laser pulse to trigger GBM Ca2+transients.Main results.We determine that 1.58µJµm-2is the optimal UV nanosecond laser pulse energy density necessary to elicit a single Ca2+transient in the GBM cell-lines whilst maintaining viability, functionality, the ability to be stimulated many times in an experiment, and to trigger further Ca2+communication in a larger network of GBM cells.Significance.Using adult patient-derived mesenchymal GBM brain cancer cell-lines, the most aggressive form of GBM cancer, this work is the first of its kind as it provides a new effective modality of which to stimulate GBM cells at the single-cell level in an accurate, repeatable, and reliable manner; and is a first step toward Ca2+communication in GBM brain cancer cells and their networks being more effectively studied.
Collapse
Affiliation(s)
- Nicholas G Mellor
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Sylvia A Chung
- Adult Cancer Program, Lowy Cancer Research Centre, The University of New South Wales, Sydney, Australia
| | - E Scott Graham
- Department of Molecular Medicine and Pathology & The Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Bryan W Day
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Charles P Unsworth
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Olatunji G, Aderinto N, Adefusi T, Kokori E, Akinmoju O, Yusuf I, Olusakin T, Muzammil MA. Efficacy of tumour-treating fields therapy in recurrent glioblastoma: A narrative review of current evidence. Medicine (Baltimore) 2023; 102:e36421. [PMID: 38050252 PMCID: PMC10695547 DOI: 10.1097/md.0000000000036421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023] Open
Abstract
Recurrent Glioblastoma presents a formidable challenge in oncology due to its aggressive nature and limited treatment options. Tumour-Treating Fields (TTFields) Therapy, a novel therapeutic modality, has emerged as a promising approach to address this clinical conundrum. This review synthesizes the current evidence surrounding the efficacy of TTFields Therapy in the context of recurrent Glioblastoma. Diverse academic databases were explored to identify relevant studies published within the last decade. Strategic keyword selection facilitated the inclusion of studies focusing on TTFields Therapy's efficacy, treatment outcomes, and patient-specific factors. The review reveals a growing body of evidence suggesting the potential clinical benefits of TTFields Therapy for patients with recurrent Glioblastoma. Studies consistently demonstrate its positive impact on overall survival (OS) and progression-free survival (PFS). The therapy's safety profile remains favorable, with mild to moderate skin reactions being the most commonly reported adverse events. Our analysis highlights the importance of patient selection criteria, with emerging biomarkers such as PTEN mutation status influencing therapy response. Additionally, investigations into combining TTFields Therapy with other treatments, including surgical interventions and novel approaches, offer promising avenues for enhancing therapeutic outcomes. The synthesis of diverse studies underscores the potential of TTFields Therapy as a valuable addition to the armamentarium against recurrent Glioblastoma. The narrative review comprehensively explains the therapy's mechanisms, clinical benefits, adverse events, and future directions. The insights gathered herein serve as a foundation for clinicians and researchers striving to optimize treatment strategies for patients facing the challenging landscape of recurrent Glioblastoma.
Collapse
Affiliation(s)
- Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Ismaila Yusuf
- Department of Medicine and Surgery, Obafemi Awolowo University, Ife, Nigeria
| | - Tobi Olusakin
- College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
15
|
Hu M, Liao X, Tao Y, Chen Y. Advances in oncolytic herpes simplex virus and adenovirus therapy for recurrent glioma. Front Immunol 2023; 14:1285113. [PMID: 38022620 PMCID: PMC10652401 DOI: 10.3389/fimmu.2023.1285113] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Recurrent glioma treatment is challenging due to molecular heterogeneity and treatment resistance commonly observed in these tumors. Researchers are actively pursuing new therapeutic strategies. Oncolytic viruses have emerged as a promising option. Oncolytic viruses selectively replicate within tumor cells, destroying them and stimulating the immune system for an enhanced anticancer response. Among Oncolytic viruses investigated for recurrent gliomas, oncolytic herpes simplex virus and oncolytic adenovirus show notable potential. Genetic modifications play a crucial role in optimizing their therapeutic efficacy. Different generations of replicative conditioned oncolytic human adenovirus and oncolytic HSV have been developed, incorporating specific modifications to enhance tumor selectivity, replication efficiency, and immune activation. This review article summarizes these genetic modifications, offering insights into the underlying mechanisms of Oncolytic viruses' therapy. It also aims to identify strategies for further enhancing the therapeutic benefits of Oncolytic viruses. However, it is important to acknowledge that additional research and clinical trials are necessary to establish the safety, efficacy, and optimal utilization of Oncolytic viruses in treating recurrent glioblastoma.
Collapse
Affiliation(s)
- Mingming Hu
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - XuLiang Liao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Tao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaohui Chen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Tincu (Iurciuc) CE, Andrițoiu CV, Popa M, Ochiuz L. Recent Advancements and Strategies for Overcoming the Blood-Brain Barrier Using Albumin-Based Drug Delivery Systems to Treat Brain Cancer, with a Focus on Glioblastoma. Polymers (Basel) 2023; 15:3969. [PMID: 37836018 PMCID: PMC10575401 DOI: 10.3390/polym15193969] [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: 08/14/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignant tumor, and the most prevalent primary malignant tumor affecting the brain and central nervous system. Recent research indicates that the genetic profile of GBM makes it resistant to drugs and radiation. However, the main obstacle in treating GBM is transporting drugs through the blood-brain barrier (BBB). Albumin is a versatile biomaterial for the synthesis of nanoparticles. The efficiency of albumin-based delivery systems is determined by their ability to improve tumor targeting and accumulation. In this review, we will discuss the prevalence of human glioblastoma and the currently adopted treatment, as well as the structure and some essential functions of the BBB, to transport drugs through this barrier. We will also mention some aspects related to the blood-tumor brain barrier (BTBB) that lead to poor treatment efficacy. The properties and structure of serum albumin were highlighted, such as its role in targeting brain tumors, as well as the progress made until now regarding the techniques for obtaining albumin nanoparticles and their functionalization, in order to overcome the BBB and treat cancer, especially human glioblastoma. The albumin drug delivery nanosystems mentioned in this paper have improved properties and can overcome the BBB to target brain tumors.
Collapse
Affiliation(s)
- Camelia-Elena Tincu (Iurciuc)
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| | - Călin Vasile Andrițoiu
- Apitherapy Medical Center, Balanesti, Nr. 336-337, 217036 Gorj, Romania;
- Specialization of Nutrition and Dietetics, Faculty of Pharmacy, Vasile Goldis Western University of Arad, Liviu Rebreanu Street, 86, 310045 Arad, Romania
| | - Marcel Popa
- Department of Natural and Synthetic Polymers, “Cristofor Simionescu” Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 73, Prof. Dimitrie Mangeron Street, 700050 Iasi, Romania;
- Faculty of Dental Medicine, “Apollonia” University of Iasi, 11, Pacurari Street, 700511 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| | - Lăcrămioara Ochiuz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16, University Street, 700115 Iasi, Romania;
| |
Collapse
|
17
|
Liu H, Zeng Z, Sun P. Prognosis and immunoinfiltration analysis of angiogene-related genes in grade 4 diffuse gliomas. Aging (Albany NY) 2023; 15:9842-9857. [PMID: 37737709 PMCID: PMC10564429 DOI: 10.18632/aging.205054] [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: 05/18/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023]
Abstract
Although angiogenesis critically influences the progression of solid tumors, its contribution to highly malignant, grade 4 diffuse gliomas remains unclear. After analyzing 506 angiogenesis-related genes differentially expressed in grade 4 diffuse gliomas via LASSO and univariate and multivariate COX regression analyses, we constructed a nomogram based on COL22A1, IGFBP2, and MPO that accurately predicted patient survival. The nomogram's performance was validated in an external patient cohort, and a risk score based on the formula COL22A1*0.148+IGFBP2*0.234+MPO*0.145 was used to distinguish high-risk from low-risk patients. Based on differentially expressed genes among risk groups, functional enrichment and drug sensitivity analyses were conducted, and the association between COL22A1, IGFBP2, and MPO expression and infiltrating immune cells and immune checkpoint genes was investigated. We next focused on COL22A1, and verified its overexpression in both glioma cell lines and clinical samples. A pro-oncogenic role for COL22A1, evidenced by impaired proliferation, migration, and invasion capacities, was evidenced upon shRNA-mediated COL22A1 silencing in glioma U87 and LN18 cells. In summary, we present a novel nomogram based on the angiogenesis-related genes COL22A1, IGFBP2, and MPO that allows survival prediction in patients with grade 4 diffuse gliomas. Furthermore, our cellular assays support a pro-oncogenic role for COL22A1 in these tumors.
Collapse
Affiliation(s)
- Hui Liu
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhirui Zeng
- Department of Neurosurgery, Guizhou Medical University, Guiyang, China
| | - Peng Sun
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
18
|
Jain P, Vashist S, Panjiyar BK. Navigating the Immune Challenge in Glioblastoma: Exploring Immunotherapeutic Avenues for Overcoming Immune Suppression. Cureus 2023; 15:e46089. [PMID: 37900496 PMCID: PMC10611557 DOI: 10.7759/cureus.46089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor known for its short survival time, typically 14-18 months from diagnosis to fatality. Managing GBM poses significant challenges due to factors like the formidable blood-brain barrier, the immunosuppressive conditions within GBM, and the intricacies of surgical procedures. Currently, the typical treatment for GBM combines surgical procedures, radiation therapy, and chemotherapy using temozolomide. Unfortunately, this conventional approach has not proven effective in substantially extending the lives of GBM patients. Consequently, researchers are exploring alternative methods for GBM management. One promising avenue receiving attention in recent years is immunotherapy. This approach has successfully treated cancer types like non-small cell lung cancer and blood-related malignancies. Various immunotherapeutic strategies are currently under investigation for GBM treatment, including checkpoint inhibitors, vaccines, chimeric antigen receptor (CAR) T-cell therapy, and oncolytic viruses. A comprehensive review of 26 high-quality studies conducted over the past decade, involving thorough searches of databases such as PubMed and Google Scholar, has been conducted. The findings from this review suggest that while immunotherapeutic strategies show promise, they face significant limitations and challenges in practical application for GBM treatment. The study emphasizes the importance of combining diverse approaches, customizing treatments for individual patients, and ongoing research efforts to improve GBM patients' outlook.
Collapse
Affiliation(s)
- Prateek Jain
- Internal Medicine, Maulana Azad Medical College, Delhi, IND
| | | | - Binay K Panjiyar
- Medicine, Harvard Medical School, Boston, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
19
|
Barkhoudarian G, Badruddoja M, Blondin N, Chowdhary S, Cobbs C, Duic JP, Flores JP, Fonkem E, McClay E, Nabors LB, Salacz M, Taylor L, Vaillant B, Gill J, Kesari S. An expanded safety/feasibility study of the EMulate Therapeutics Voyager™ System in patients with recurrent glioblastoma. CNS Oncol 2023; 12:CNS102. [PMID: 37462385 PMCID: PMC10410686 DOI: 10.2217/cns-2022-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Aim: The EMulate Therapeutics Voyager™ is a simple, wearable, home-use device that uses an alternating electromagnetic field to alter biologic signaling within cells. Objective: To assess the safety/feasibility of the Voyager in the treatment of recurrent glioblastoma (rGBM). Methods: In this study, patients with rGBM were treated with Voyager as monotherapy or in combination with standard chemotherapy at the Investigator's discretion. Safety was assessed by incidence of adverse events associated with the Voyager. Patients were followed until death. Results: A total of 75 patients were enrolled and treated for at least one day with the Voyager (safety population). Device-related adverse events were uncommon and generally did not result in interruption or withdrawal from treatment. There were no serious adverse events associated with Voyager. A total of 60 patients were treated for at least one month (clinical utility population). The median progression-free survival (PFS) was 17 weeks (4.3 months) in the Voyager only group (n = 24) and 21 weeks (5.3 months) in the Voyager + concurrent therapy group (n = 36). The median overall survival (OS) was 7 months in the Voyager only group and 9 months in the Voyager + concurrent therapy group. In patients treated with Voyager + concurrent therapy, the median OS for patients enrolled with their 1st or 2nd recurrence (n = 26) was 10 months, while in patients enrolled with their 3rd or 4th recurrence (n = 10) OS was 7 months. Conclusion: The data support the safety and feasibility of the Voyager for the treatment of rGBM. Further prospective study of the device is warranted. Trial Registration Number: NCT02296580 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Garni Barkhoudarian
- Saint John's Cancer Institute & Pacific Neuroscience Institute at Providence St. John's Health Center, Santa Monica, CA 90404, USA
| | | | - Nicholas Blondin
- Associated Neurologists of Southern Connecticut, Fairfield, CT 06824, USA
| | | | - Charles Cobbs
- Swedish Medical Center, Ben and Catherine Ivy Center For Advanced Brain Tumor Treatment, Seattle, WA 98122, USA
| | | | | | | | - Edward McClay
- cCARE (California Cancer Associates for Research & Excellence), Encinitas, CA 92024, USA
| | - Louis Burt Nabors
- University of Alabama, Division of Neuro-Oncology, Birmingham, AL 35294-3410, USA
| | - Michael Salacz
- University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lynn Taylor
- University of Washington, Seattle, WA 98195, USA
| | | | - Jaya Gill
- Saint John's Cancer Institute & Pacific Neuroscience Institute at Providence St. John's Health Center, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- Saint John's Cancer Institute & Pacific Neuroscience Institute at Providence St. John's Health Center, Santa Monica, CA 90404, USA
| |
Collapse
|
20
|
Baddam SR, Kalagara S, Kuna K, Enaganti S. Recent advancements and theranostics strategies in glioblastoma therapy. Biomed Mater 2023; 18:052007. [PMID: 37582381 DOI: 10.1088/1748-605x/acf0ab] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal malignant brain tumor, and it is challenging to cure with surgery and treatment. The prevention of permanent brain damage and tumor invasion, which is the ultimate cause of recurrence, are major obstacles in GBM treatment. Besides, emerging treatment modalities and newer genetic findings are helping to understand and manage GBM in patients. Accordingly, researchers are focusing on advanced nanomaterials-based strategies for tackling the various problems associated with GBM. In this context, researchers explored novel strategies with various alternative treatment approaches such as early detection techniques and theranostics approaches. In this review, we have emphasized the recent advancement of GBM cellular models and their roles in designing GBM therapeutics. We have added a special emphasis on the novel genetic and drug target findings as well as strategies for early detection. Besides, we have discussed various theranostic approaches such as hyperthermia therapy, phototherapy and image-guided therapy. Approaches utilized for targeted drug delivery to the GBM were also discussed. This article also describes the recentin vivo, in vitroandex vivoadvances using innovative theranostic approaches.
Collapse
Affiliation(s)
- Sudhakar Reddy Baddam
- University of Massachusetts Chan Medical School, RNA Therapeutics Institute,Worcester,MA 01655, United States of America
| | - Sudhakar Kalagara
- Department of Chemistry and Biochemistry,University of the Texas at El Paso, 500 W University Ave,El Paso,TX 79968, United States of America
| | - Krishna Kuna
- Department of Chemistry,University College of Science, Saifabad, Osmania University, Hyderabad,Telangana,India
| | - Sreenivas Enaganti
- Department of Bioinformatics, Averinbiotech Laboratories,208, 2nd Floor, Windsor Plaza, Nallakunta, Hyderabad, Telangana,India
| |
Collapse
|
21
|
Okamoto R, Toya K, Ogino Y, Sato A. Downregulation of long noncoding RNA TP73-AS1 expression confers resistance to temozolomide in human glioblastoma cells. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 43:86-98. [PMID: 37452786 DOI: 10.1080/15257770.2023.2234960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Glioblastoma multiforme (GBM), the most aggressive primary malignant brain tumor, is resistant to conventional radiotherapies and chemotherapies, including temozolomide (TMZ). Overcoming GBM resistance to the chemotherapeutic agent TMZ poses an important therapeutic problem. This study established an association between the long noncoding RNA TP73-AS1 and TMZ sensitivity regulation in human GBM cells (U87MG). Transcriptomic analysis revealed that TP73-AS1 expression was reduced in TMZ-resistant U87MGRT100 cells compared to that in parental U87MG cells. Additionally, TP73-AS1 knockdown in parental U87MG cells decreased their sensitivity to TMZ. Overall, these findings suggest that TP73-AS1 functions as a regulator of TMZ sensitivity in GBM cells.
Collapse
Affiliation(s)
- Ryo Okamoto
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Koki Toya
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yoko Ogino
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Akira Sato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
22
|
Ghanem M, Ghaith AK, Zamanian C, Bon-Nieves A, Bhandarkar A, Bydon M, Quiñones-Hinojosa A. Deep Learning Approaches for Glioblastoma Prognosis in Resource-Limited Settings: A Study Using Basic Patient Demographic, Clinical, and Surgical Inputs. World Neurosurg 2023; 175:e1089-e1109. [PMID: 37088416 DOI: 10.1016/j.wneu.2023.04.072] [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: 01/26/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common brain tumor in the United States, with an annual incidence rate of 3.21 per 100,000. It is the most aggressive type of diffuse glioma and has a median survival of months after treatment. This study aims to assess the accuracy of different novel deep learning models trained on a set of simple clinical, demographic, and surgical variables to assist in clinical practice, even in areas with constrained health care infrastructure. METHODS Our study included 37,095 patients with GBM from the SEER (Surveillance Epidemiology and End Results) database. All predictors were based on demographic, clinicopathologic, and treatment information of the cases. Our outcomes of interest were months of survival and vital status. Concordance index (C-index) and integrated Brier scores (IBS) were used to evaluate the performance of the models. RESULTS The patient characteristics and the statistical analyses were consistent with the epidemiologic literature. The models C-index and IBS ranged from 0.6743 to 0.6918 and from 0.0934 to 0.1034, respectively. Probabilistic matrix factorization (0.6918), multitask logistic regression (0.6916), and logistic hazard (0.6916) had the highest C-index scores. The models with the lowest IBS were the probabilistic matrix factorization (0.0934), multitask logistic regression (0.0935), and logistic hazard (0.0936). These models had an accuracy (1-IBS) of 90.66%; 90.65%, and 90.64%, respectively. The deep learning algorithms were deployed on an interactive Web-based tool for practical use available via https://glioblastoma-survanalysis.herokuapp.com/. CONCLUSIONS Novel deep learning algorithms can better predict GBM prognosis than do baseline methods and can lead to more personalized patient care regardless of extensive electronic health record availability.
Collapse
Affiliation(s)
- Marc Ghanem
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Abdul Karim Ghaith
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Cameron Zamanian
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Antonio Bon-Nieves
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Archis Bhandarkar
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mohamad Bydon
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA.
| | | |
Collapse
|
23
|
Huang HS, Chiang IT, Lawal B, Weng YS, Jeng LB, Kuo YC, Liu YC, Hsu FT. A Novel Isotope-labeled Small Molecule Probe CC12 for Anti-glioma via Suppressing LYN-mediated Progression and Activating Apoptosis Pathways. Int J Biol Sci 2023; 19:3209-3225. [PMID: 37416766 PMCID: PMC10321274 DOI: 10.7150/ijbs.82266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/16/2023] [Indexed: 07/08/2023] Open
Abstract
Background: Glioblastoma multiforme (GBM) is the most lethal malignancy in brain, which is surrounded by the blood-brain barrier (BBB), which limits the efficacy of standard treatments. Developing an effective drug that can penetrate the blood-brain barrier (BBB) remains a critical challenge in the fight against GBM. CC12 (NSC749232) is an anthraquinone tetraheterocyclic homolog with a lipophilic structure that may facilitate penetration of the brain area. Methods: We used temozolomide sensitive and resistance GBM cells and animal model to identify the CC12 delivery, anti-tumor potential and its underlying mechanism. Results: Importantly, toxicity triggered by CC12 was not associated with the methyl guanine-DNA methyl transferase (MGMT) methylation status which revealed a greater application potential compared to temozolomide. Alexa F488 cadaverine-labelled CC12 successfully infiltrated into the GBM sphere; in addition, 68Ga-labeled CC12 was also found in the orthotopic GBM area. After passing BBB, CC12 initiated both caspase-dependent intrinsic/extrinsic apoptosis pathways and apoptosis-inducing factor, EndoG-related caspase-independent apoptosis signaling in GBM. RNA sequence analysis from The Cancer Genome Atlas indicated that LYN was overexpressed in GBM is associated with poorer overall survival. We proved that targeting of LYN by CC12 may diminish GBM progression and suppress it downstream factors such as signal transduction and activator of extracellular signal-regulated kinases (ERK)/transcription 3 (STAT3)/nuclear factor (NF)-κB. CC12 was also found to participate in suppressing GBM metastasis and dysregulation of the epithelial-mesenchymal transition (EMT) through inactivation of the LYN axis. Conclusion: CC12, a newly developed BBB-penetrating drug, was found to possess an anti-GBM capacity via initiating an apoptotic mechanism and disrupting LYN/ERK/STAT3/NF-κB-regulated GBM progression.
Collapse
Affiliation(s)
- Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; and Academia Sinica, Taipei 115, Taiwan, R.O.C
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan, R.O.C
| | - I-Tsang Chiang
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua 500, Taiwan, R.O.C
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Lukang, Changhua 505, Taiwan, R.O.C
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan, R.O.C
- Medical administrative center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; and Academia Sinica, Taipei 115, Taiwan, R.O.C
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan, R.O.C
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Yueh-Shan Weng
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan, R.O.C
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
- Cell Therapy Center, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, R.O.C
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
- Master Program in Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110, Taiwan, R.O.C
| | - Yu-Chang Liu
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua 500, Taiwan, R.O.C
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Lukang, Changhua 505, Taiwan, R.O.C
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan, R.O.C
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan, R.O.C
| |
Collapse
|
24
|
Hasan H, Afzal M, Castresana JS, Shahi MH. A Comprehensive Review of miRNAs and Their Epigenetic Effects in Glioblastoma. Cells 2023; 12:1578. [PMID: 37371047 DOI: 10.3390/cells12121578] [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: 04/18/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma is the most aggressive form of brain tumor originating from glial cells with a maximum life expectancy of 14.6 months. Despite the establishment of multiple promising therapies, the clinical outcome of glioblastoma patients is abysmal. Drug resistance has been identified as a major factor contributing to the failure of current multimodal therapy. Epigenetic modification, especially DNA methylation has been identified as a major regulatory mechanism behind glioblastoma progression. In addition, miRNAs, a class of non-coding RNA, have been found to play a role in the regulation as well as in the diagnosis of glioblastoma. The relationship between epigenetics, drug resistance, and glioblastoma progression has been clearly demonstrated. MGMT hypermethylation, leading to a lack of MGMT expression, is associated with a cytotoxic effect of TMZ in GBM, while resistance to TMZ frequently appears in MGMT non-methylated GBM. In this review, we will elaborate on known miRNAs linked to glioblastoma; their distinctive oncogenic or tumor suppressor roles; and how epigenetic modification of miRNAs, particularly via methylation, leads to their upregulation or downregulation in glioblastoma. Moreover, we will try to identify those miRNAs that might be potential regulators of MGMT expression and their role as predictors of tumor response to temozolomide treatment. Although we do not impact clinical data and survival, we open possible experimental approaches to treat GBM, although they should be further validated with clinically oriented studies.
Collapse
Affiliation(s)
- Hera Hasan
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Afzal
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India
| | - Javier S Castresana
- Department of Biochemistry and Genetics, University of Navarra School of Sciences, 31008 Pamplona, Spain
| | - Mehdi H Shahi
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
25
|
Dejonckheere CS, Thelen A, Simon B, Greschus S, Köksal MA, Schmeel LC, Wilhelm-Buchstab T, Leitzen C. Impact of Postoperative Changes in Brain Anatomy on Target Volume Delineation for High-Grade Glioma. Cancers (Basel) 2023; 15:2840. [PMID: 37345177 DOI: 10.3390/cancers15102840] [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: 04/20/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
High-grade glioma has a poor prognosis, and radiation therapy plays a crucial role in its management. Every step of treatment planning should thus be optimised to maximise survival chances and minimise radiation-induced toxicity. Here, we compare structures needed for target volume delineation between an immediate postoperative magnetic resonance imaging (MRI) and a radiation treatment planning MRI to establish the need for the latter. Twenty-eight patients were included, with a median interval between MRIs (range) of 19.5 (8-50) days. There was a mean change in resection cavity position (range) of 3.04 ± 3.90 (0-22.1) mm, with greater positional changes in skull-distant (>25 mm) resection cavity borders when compared to skull-near (≤25 mm) counterparts (p < 0.001). The mean differences in resection cavity and surrounding oedema and FLAIR hyperintensity volumes were -32.0 ± 29.6% and -38.0 ± 25.0%, respectively, whereas the mean difference in midline shift (range) was -2.64 ± 2.73 (0-11) mm. These data indicate marked short-term volumetric changes and support the role of an MRI to aid in target volume delineation as close to radiation treatment start as possible. Planning adapted to the actual anatomy at the time of radiation limits the risk of geographic miss and might thus improve outcomes in patients undergoing adjuvant radiation for high-grade glioma.
Collapse
Affiliation(s)
| | - Anja Thelen
- Faculty of Medicine, University Bonn, 53127 Bonn, Germany
| | - Birgit Simon
- Department of Radiology, University Hospital Bonn, 53127 Bonn, Germany
| | | | - Mümtaz Ali Köksal
- Department of Radiation Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | | | | | - Christina Leitzen
- Department of Radiation Oncology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
26
|
Iturrioz-Rodríguez N, Sampron N, Matheu A. Current advances in temozolomide encapsulation for the enhancement of glioblastoma treatment. Theranostics 2023; 13:2734-2756. [PMID: 37284445 PMCID: PMC10240814 DOI: 10.7150/thno.82005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/20/2023] [Indexed: 06/08/2023] Open
Abstract
Glioblastoma is the most common and lethal brain tumor in adults. The incorporation of temozolomide (TMZ) into the standard treatment has increased the overall survival rate of glioblastoma patients. Since then, significant advances have been made in understanding the benefits and limitations of TMZ. Among the latter, the unspecific toxicity of TMZ, poor solubility, and hydrolyzation are intrinsic characteristics, whereas the presence of the blood-brain barrier and some tumor properties, such as molecular and cellular heterogeneity and therapy resistance, have limited the therapeutic effects of TMZ in treating glioblastoma. Several reports have revealed that different strategies for TMZ encapsulation in nanocarriers overcome those limitations and have shown that they increase TMZ stability, half-life, biodistribution, and efficacy, offering the promise for future nanomedicine therapies in handling glioblastoma. In this review, we analyze the different nanomaterials used for the encapsulation of TMZ to improve its stability, blood half-life and efficacy, paying special attention to polymer- and lipid-based nanosystems. To improve TMZ drug resistance, present in up to 50% of patients, we detail TMZ combined therapeutic with i) other chemotherapies, ii) inhibitors, iii) nucleic acids, iv) photosensitizers and other nanomaterials for photodynamic therapy, photothermal therapy, and magnetic hyperthermia, v) immunotherapy, and vi) other less explored molecules. Moreover, we describe targeting strategies, such as passive targeting, active targeting to BBB endothelial cells, glioma cells, and glioma cancer stem cells, and local delivery, where TMZ has demonstrated an improved outcome. To finish our study, we include possible future research directions that could help decrease the time needed to move from bench to bedside.
Collapse
Affiliation(s)
| | - Nicolas Sampron
- Cellular Oncology group, Biodonostia Health Research Institute, San Sebastian, Spain
| | - Ander Matheu
- Cellular Oncology group, Biodonostia Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento (CIBERfes), Carlos III Institute, Madrid, Spain
| |
Collapse
|
27
|
Lau A, Le N, Nguyen C, Kandpal RP. Signals transduced by Eph receptors and ephrin ligands converge on MAP kinase and AKT pathways in human cancers. Cell Signal 2023; 104:110579. [PMID: 36572189 DOI: 10.1016/j.cellsig.2022.110579] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Eph receptors, the largest known family of receptor tyrosine kinases, and ephrin ligands have been implicated in a variety of human cancers. The novel bidirectional signaling events initiated by binding of Eph receptors to their cognate ephrin ligands modulate many cellular processes such as proliferation, metastasis, angiogenesis, invasion, and apoptosis. The relationships between the abundance of a unique subset of Eph receptors and ephrin ligands with associated cellular processes indicate a key role of these molecules in tumorigenesis. The combinatorial expression of these molecules converges on MAP kinase and/or AKT/mTOR signaling pathways. The intracellular target proteins of the initial signal may, however, vary in some cancers. Furthermore, we have also described the commonality of up- and down-regulation of individual receptors and ligands in various cancers. The current state of research in Eph receptors illustrates MAP kinase and mTOR pathways as plausible targets for therapeutic interventions in various cancers.
Collapse
Affiliation(s)
- Andreas Lau
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Nghia Le
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Claudia Nguyen
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Raj P Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
28
|
Loras A, Gonzalez-Bonet LG, Gutierrez-Arroyo JL, Martinez-Cadenas C, Marques-Torrejon MA. Neural Stem Cells as Potential Glioblastoma Cells of Origin. Life (Basel) 2023; 13:life13040905. [PMID: 37109434 PMCID: PMC10145968 DOI: 10.3390/life13040905] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor in adults and it remains incurable. These tumors are very heterogeneous, resistant to cytotoxic therapies, and they show high rates of invasiveness. Therefore, patients face poor prognosis, and the survival rates remain very low. Previous research states that GBM contains a cell population with stem cell characteristics called glioma stem cells (GSCs). These cells are able to self-renew and regenerate the tumor and, therefore, they are partly responsible for the observed resistance to therapies and tumor recurrence. Recent data indicate that neural stem cells (NSCs) in the subventricular zone (SVZ) are the cells of origin of GBM, that is, the cell type acquiring the initial tumorigenic mutation. The involvement of SVZ-NSCs is also associated with GBM progression and recurrence. Identifying the cellular origin of GBM is important for the development of early detection techniques and the discovery of early disease markers. In this review, we analyze the SVZ-NSC population as a potential GBM cell of origin, and its potential role for GBM therapies.
Collapse
Affiliation(s)
- Alba Loras
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon de la Plana, Spain
| | - Julia L. Gutierrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon de la Plana, Spain
| | | | - Maria Angeles Marques-Torrejon
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
- Correspondence: ; Tel.: +34-964-387-478
| |
Collapse
|
29
|
Fresnais M, Jung I, Klein UB, Miller AK, Turcan S, Haefeli WE, Burhenne J, Longuespée R. Important Requirements for Desorption/Ionization Mass Spectrometric Measurements of Temozolomide-Induced 2'-Deoxyguanosine Methylations in DNA. Cancers (Basel) 2023; 15:cancers15030716. [PMID: 36765673 PMCID: PMC9913758 DOI: 10.3390/cancers15030716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
In clinical pharmacology, drug quantification is mainly performed from the circulation for pharmacokinetic purposes. Finely monitoring the chemical effect of drugs at their chemical sites of action for pharmacodynamics would have a major impact in several contexts of personalized medicine. Monitoring appropriate drug exposure is particularly challenging for alkylating drugs such as temozolomide (TMZ) because there is no flow equilibrium that would allow reliable conclusions to be drawn about the alkylation of the target site from plasma concentrations. During the treatment of glioblastoma, it appears, therefore, promising to directly monitor the alkylating effect of TMZ rather than plasma exposure, ideally at the site of action. Mass spectrometry (MS) is a method of choice for the quantification of methylated guanines and, more specifically, of O6-methylguanines as a marker of TMZ exposure at the site of action. Depending on the chosen strategy to analyze modified purines and 2'-deoxynucleosides, the analysis of methylated guanines and 2'-deoxyguanosines is prone to important artefacts due to the overlap between masses of (i) guanines from DNA and RNA, and (ii) different methylated species of guanines. Therefore, the specific analysis of O6-methyl-2'deoxyguanosine, which is the product of the TMZ effect, is highly challenging. In this work, we report observations from matrix-assisted laser desorption/ionization (MALDI), and desorption electrospray ionization (DESI) MS analyses. These allow for the construction of a decision tree to initiate studies using desorption/ionization MS for the analysis of 2'-deoxyguanosine methylations induced by TMZ.
Collapse
Affiliation(s)
- Margaux Fresnais
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Ina Jung
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Uli B. Klein
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Aubry K. Miller
- Cancer Drug Development, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sevin Turcan
- Neurology Clinic and National Center for Tumor Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Rémi Longuespée
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- Correspondence: author:
| |
Collapse
|
30
|
Cystic Glioblastoma: A Mimicker of Infection? A Case Report and Literature Review. Case Rep Oncol Med 2023; 2023:7348188. [PMID: 36704640 PMCID: PMC9873443 DOI: 10.1155/2023/7348188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequent malignant and aggressive type of glioma. Most cases of GBM present as a single solitary solid tumor; however, there are rare instances in which it may present as a cystic lesion. Here, we report an even rarer case of GBM presenting as bilateral multicystic lesions, mimicking infectious etiology. Our case highlights the importance of identifying clinical features of cystic GBM to ensure early diagnosis and treatment. A literature review was conducted in PubMed, looking at the common characteristics and treatment options for cystic GBM.
Collapse
|
31
|
Safaei R, Mojtahedi H, Hanaei S, Razavi A, Esmaeili M, Sadr M, Rezaei A, Edalatfar M, Kashani HK, Sadeghi-Naini M, Darbeheshti F, Gharehdaghi J, Forouzesh M, Ebrahimi A, Rezaei N. MGMT Gene rs1625649 Polymorphism in Iranian Patients with Brain Glioblastoma: A Case Control Study. Avicenna J Med Biotechnol 2023; 15:48-52. [PMID: 36789113 PMCID: PMC9895979 DOI: 10.18502/ajmb.v15i1.11424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/01/2022] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor with poor prognosis and high potential of dispersion to other brain tissues in adult. Effective and modern choices of treatment including chemotherapy with alkylating agents marginally extend survival of GBM. However, alkylating agents can lead to highly harmful mismatch during DNA replication causing apoptosis and cell death. Accordingly, O6-Methylguanine-DNA methyltransferase (MGMT) removes alkyl adducts, thereby causing resistance to alkylating drugs. Single-Nucleotide Polymorphisms (SNPs) in MGMT promoter region may play a role in the regulation of MGMT expression and prediction of glioma development risk. In order to evaluate the clinical significance of rs1625649 SNP in the MGMT promoter region of glioblastoma, genomic DNA from a series of 54 patients with GBM and 50 healthy individuals in Iranian population were collected for tetra ARMS PCR amplification. None of the "A" or "C" alleles were associated with tumor occurrence, the "AA" genotype was more frequent in healthy subjects, and the "AC" genotype was 4.6 times more common in patients with GBM. The longest survival time was observed in the "CC" genotype; however, this difference was not statistically significant. On the other hand, homozygous rs1625649 (AA genotype) was significantly associated with a better survival than the cases with heterozygous rs1625649 (CA genotype) or wild type rs1625649 (CC genotype), predicting better response to temozolomide-based chemotherapy.
Collapse
Affiliation(s)
- Reyhaneh Safaei
- Department of Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hanieh Mojtahedi
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sara Hanaei
- Department of Neurosurgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadehsadat Razavi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzie Esmaeili
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Sadr
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezou Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Edalatfar
- Department of Neurosurgery, lmam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Khayat Kashani
- Department of Neurosurgery, lmam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Sadeghi-Naini
- Department of Neurosurgery, Lorestan University of Medical Sciences, KhorramAbad, Iran
| | - Farzaneh Darbeheshti
- Department of Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jaber Gharehdaghi
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Mehdi Forouzesh
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Abdolali Ebrahimi
- Department of Pathology, lmam Hossein Hospital, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Godugu K, Hay BA, Glinsky GV, Mousa SA. Discovery of novel thyrointegrin αvβ3 antagonist fb-PMT (NP751) in the management of human glioblastoma multiforme. Neurooncol Adv 2023; 5:vdac180. [PMID: 36879662 PMCID: PMC9985163 DOI: 10.1093/noajnl/vdac180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Thyrointegrin αvβ3 receptors are unique molecular cancer therapeutic targets because of their overexpression on cancer and rapidly dividing blood vessel cells compared and quiescent on normal cells. A macromolecule, TriAzole Tetraiodothyroacetic acid (TAT) conjugated to polyethylene glycol with a lipophilic 4-fluorobenyl group (fb-PMT and NP751), interacts with high affinity (0.21 nM) and specificity with the thyrointegrin αvβ3 receptors on the cell surface without nuclear translocation in contrast to the non-polymer conjugated TAT. Methods The following in vitro assays were carried out to evaluate NP751 including binding affinity to different integrins, transthyretin (TTR)-binding affinity, glioblastoma multiforme (GBM) cell adhesion, proliferation assays, nuclear translocations, chorioallantoic membrane model of angiogenesis, and microarray for molecular mechanisms. Additionally, in vivo studies were carried out to evaluate the anticancer efficacy of NP751, its biodistribution, and brain GBM tumor versus plasma levels kinetics. Results NP751 demonstrated a broad spectrum of antiangiogenesis and anticancer efficacy in experimental models of angiogenesis and xenografts of human GBM cells. Tumor growth and cancer cells' viability were markedly decreased (by > 90%; P < .001) in fb-PMT-treated U87-luc or 3 different primary human GBM xenograft-bearing mice based on tumor in vivo imaging system (IVIS) imaging and histopathological examination, without relapse upon treatment discontinuation. Additionally, it effectively transports across the blood-brain barrier via its high-affinity binding to plasma TTR with high retention in brain tumors. NP751-induced effects on gene expression support the model of molecular interference at multiple key pathways essential for GBM tumor progression and vascularization. Conclusions fb-PMT is a potent thyrointegrin αvβ3 antagonist with potential impact on GBM tumor progression.
Collapse
Affiliation(s)
- Kavitha Godugu
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer & NanoPharmaceuticals LLC, Rensselaer, New York, USA
| | - Bruce A Hay
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer & NanoPharmaceuticals LLC, Rensselaer, New York, USA
| | - Gennadi V Glinsky
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer & NanoPharmaceuticals LLC, Rensselaer, New York, USA
| |
Collapse
|
33
|
Sun X, Klingbeil O, Lu B, Wu C, Ballon C, Ouyang M, Wu XS, Jin Y, Hwangbo Y, Huang YH, Somerville TDD, Chang K, Park J, Chung T, Lyons SK, Shi J, Vogel H, Schulder M, Vakoc CR, Mills AA. BRD8 maintains glioblastoma by epigenetic reprogramming of the p53 network. Nature 2023; 613:195-202. [PMID: 36544023 PMCID: PMC10189659 DOI: 10.1038/s41586-022-05551-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/10/2022] [Indexed: 12/24/2022]
Abstract
Inhibition of the tumour suppressive function of p53 (encoded by TP53) is paramount for cancer development in humans. However, p53 remains unmutated in the majority of cases of glioblastoma (GBM)-the most common and deadly adult brain malignancy1,2. Thus, how p53-mediated tumour suppression is countered in TP53 wild-type (TP53WT) GBM is unknown. Here we describe a GBM-specific epigenetic mechanism in which the chromatin regulator bromodomain-containing protein 8 (BRD8) maintains H2AZ occupancy at p53 target loci through the EP400 histone acetyltransferase complex. This mechanism causes a repressive chromatin state that prevents transactivation by p53 and sustains proliferation. Notably, targeting the bromodomain of BRD8 displaces H2AZ, enhances chromatin accessibility and engages p53 transactivation. This in turn enforces cell cycle arrest and tumour suppression in TP53WT GBM. In line with these findings, BRD8 is highly expressed with H2AZ in proliferating single cells of patient-derived GBM, and is inversely correlated with CDKN1A, a canonical p53 target that encodes p21 (refs. 3,4). This work identifies BRD8 as a selective epigenetic vulnerability for a malignancy for which treatment has not improved for decades. Moreover, targeting the bromodomain of BRD8 may be a promising therapeutic strategy for patients with TP53WT GBM.
Collapse
Affiliation(s)
- Xueqin Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Olaf Klingbeil
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Bin Lu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Caizhi Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Carlos Ballon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Meng Ouyang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Xiaoli S Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Genetics Program, Stony Brook University, Stony Brook, NY, USA
| | - Ying Jin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Yon Hwangbo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Yu-Han Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jung Park
- Department of Neurosurgery, Zucker School of Medicine at Hofstra Northwell, Lake Success, NY, USA
| | - Taemoon Chung
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Scott K Lyons
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Junwei Shi
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Michael Schulder
- Department of Neurosurgery, Zucker School of Medicine at Hofstra Northwell, Lake Success, NY, USA
| | | | - Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
34
|
Alswailem R, Alqahtani FY, Aleanizy FS, Alrfaei BM, Badran M, Alqahtani QH, Abdelhady HG, Alsarra I. MicroRNA-219 loaded chitosan nanoparticles for treatment of glioblastoma. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:198-207. [PMID: 35762105 DOI: 10.1080/21691401.2022.2092123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recent evidence has implicated microRNA-219 (miR-219) in regulation of gene contributed in glioblastoma (GBM) pathogenesis. This study aimed to prepare miR-219 in chitosan (CS) nanoparticles (NPs), characterize and investigate their efficacy on human GBM cell line (U87 MG). NPs were prepared using ionic gelation method. The influence of process parameters on physicochemical characteristics of NPs was investigated. Apoptotic effect of miR-219 was examined on U87 MG cells. Formulated NPs showed particle size of 109 ± 2.18 nm, with poly dispersity index equal to 0.2 ± 0.05, and zeta potential of +20.5 ± 0.7 mV. Entrapment efficiency of miR-219 in loaded NP has reached 95%. The in vitro release study demonstrated sustained release pattern of miR-219 from CS-NPs. Gel retardation assay has confirmed the integrity of miR-219 after production process. The fabricated NPs reduced the survival of U87 MG cells to 78% after 24 h of post-transfection, and into 67.5% after 48 h. However, fibroblasts were not affected by the NPs, revealing their specificity for GBM cells. Given the tumour suppressing function of miR-219, and advantage of CS-NPs for gene delivery to the central nervous system, the presented NPs have a great potential for treatment of GBM.
Collapse
Affiliation(s)
- Rawan Alswailem
- Drug sector, Saudi Food and Drug Authority, Riyadh, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fulwah Yahya Alqahtani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fadilah Sfouq Aleanizy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bahauddeen M Alrfaei
- Department of Cellular Therapy and Cancer Research, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health, Riyadh, Saudi Arabia
| | - Mohammad Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Qamraa Hamad Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Ibrahim Alsarra
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Dewdney B, Ursich L, Fletcher EV, Johns TG. Anoctamins and Calcium Signalling: An Obstacle to EGFR Targeted Therapy in Glioblastoma? Cancers (Basel) 2022; 14:cancers14235932. [PMID: 36497413 PMCID: PMC9740065 DOI: 10.3390/cancers14235932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Glioblastoma is the most common form of high-grade glioma in adults and has a poor survival rate with very limited treatment options. There have been no significant advancements in glioblastoma treatment in over 30 years. Epidermal growth factor receptor is upregulated in most glioblastoma tumours and, therefore, has been a drug target in recent targeted therapy clinical trials. However, while many inhibitors and antibodies for epidermal growth factor receptor have demonstrated promising anti-tumour effects in preclinical models, they have failed to improve outcomes for glioblastoma patients in clinical trials. This is likely due to the highly plastic nature of glioblastoma tumours, which results in therapeutic resistance. Ion channels are instrumental in the development of many cancers and may regulate cellular plasticity in glioblastoma. This review will explore the potential involvement of a class of calcium-activated chloride channels called anoctamins in brain cancer. We will also discuss the integrated role of calcium channels and anoctamins in regulating calcium-mediated signalling pathways, such as epidermal growth factor signalling, to promote brain cancer cell growth and migration.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6319-1023
| | - Lauren Ursich
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| | - Terrance G. Johns
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
36
|
Aggarwal P, Luo W, Pehlivan KC, Hoang H, Rajappa P, Cripe TP, Cassady KA, Lee DA, Cairo MS. Pediatric versus adult high grade glioma: Immunotherapeutic and genomic considerations. Front Immunol 2022; 13:1038096. [PMID: 36483545 PMCID: PMC9722734 DOI: 10.3389/fimmu.2022.1038096] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022] Open
Abstract
High grade gliomas are identified as malignant central nervous tumors that spread rapidly and have a universally poor prognosis. Historically high grade gliomas in the pediatric population have been treated similarly to adult high grade gliomas. For the first time, the most recent classification of central nervous system tumors by World Health Organization has divided adult from pediatric type diffuse high grade gliomas, underscoring the biologic differences between these tumors in different age groups. The objective of our review is to compare high grade gliomas in the adult versus pediatric patient populations, highlighting similarities and differences in epidemiology, etiology, pathogenesis and therapeutic approaches. High grade gliomas in adults versus children have varying clinical presentations, molecular biology background, and response to chemotherapy, as well as unique molecular targets. However, increasing evidence show that they both respond to recently developed immunotherapies. This review summarizes the distinctions and commonalities between the two in disease pathogenesis and response to therapeutic interventions with a focus on immunotherapy.
Collapse
Affiliation(s)
- Payal Aggarwal
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | | | - Hai Hoang
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Prajwal Rajappa
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Dean A. Lee
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
37
|
Semreen AM, Alsoud LO, El-Huneidi W, Ahmed M, Bustanji Y, Abu-Gharbieh E, El-Awady R, Ramadan WS, Alqudah MA, Shara M, Abuhelwa AY, Soares NC, Semreen MH, Alzoubi KH. Metabolomics Analysis Revealed Significant Metabolic Changes in Brain Cancer Cells Treated with Paclitaxel and/or Etoposide. Int J Mol Sci 2022; 23:13940. [PMID: 36430415 PMCID: PMC9693830 DOI: 10.3390/ijms232213940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer of the central nervous system (CNS) is ranked as the 19th most prevalent form of the disease in 2020. This study aims to identify candidate biomarkers and metabolic pathways affected by paclitaxel and etoposide, which serve as potential treatments for glioblastoma, and are linked to the pathogenesis of glioblastoma. We utilized an untargeted metabolomics approach using the highly sensitive ultra-high-performance liquid chromatography-electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESI-QTOF-MS) for identification. In this study, 92 and 94 metabolites in U87 and U373 cell lines were profiled, respectively. The produced metabolites were then analyzed utilizing t-tests, volcano plots, and enrichment analysis modules. Our analysis revealed distinct metabolites to be significantly dysregulated (nutriacholic acid, L-phenylalanine, L-arginine, guanosine, ADP, hypoxanthine, and guanine), and to a lesser extent, mevalonic acid in paclitaxel and/or etoposide treated cells. Furthermore, both urea and citric acid cycles, and metabolism of polyamines and amino acids (aspartate, arginine, and proline) were significantly enriched. These findings can be used to create a map that can be utilized to assess the antitumor effect of paclitaxel and/or etoposide within the studied cancer cells.
Collapse
Affiliation(s)
- Ahlam M. Semreen
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Leen Oyoun Alsoud
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Yasser Bustanji
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Eman Abu-Gharbieh
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Raafat El-Awady
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad A.Y. Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohd Shara
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmad Y. Abuhelwa
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Karem H. Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
38
|
Satvat N, Korczynski O, Müller-Eschner M, Othman AE, Schöffling V, Keric N, Ringel F, Sommer C, Brockmann MA, Reder S. A Rapid Late Enhancement MRI Protocol Improves Differentiation between Brain Tumor Recurrence and Treatment-Related Contrast Enhancement of Brain Parenchyma. Cancers (Basel) 2022; 14:cancers14225523. [PMID: 36428617 PMCID: PMC9688406 DOI: 10.3390/cancers14225523] [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: 09/22/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Differentiation between tumor recurrence and treatment-related contrast enhancement in MRI can be difficult. Late enhancement MRI up to 75 min after contrast agent application has been shown to improve differentiation between tumor recurrence and treatment-related changes. We investigated the diagnostic performance of late enhancement using a rapid MRI protocol optimized for clinical workflow. METHODS Twenty-three patients with 28 lesions suspected for glioma recurrence underwent MRI including T1-MPRAGE-series acquired 2 and 20 min after contrast agent administration. Early contrast series were subtracted from late contrast series using motion correction. Contrast enhancing lesions were retrospectively and independently evaluated by two readers blinded to the patients' later clinical course and histology with or without the use of late enhancement series. Sensitivity, specificity, NPV, and PPV were calculated for both readers by comparing results of MRI with histological samples. RESULTS Using standard MR sequences, sensitivity, specificity, PPV, and NPV were 0.84, 0, 0.875, and 0 (reader 1) and 0.92, 0, 0.885, and 0 (reader 2), respectively. Early late enhancement increased sensitivity, specificity, PPV, and NPV to 1 for each value and for both readers. Inter-reader reliability increased from 0.632 (standard MRI sequences) to 1.0 (with early late enhancement). CONCLUSION The described rapid late enhancement MRI protocol improves MRI-based discrimination between tumor tissue and treatment-related changes of the brain parenchyma.
Collapse
Affiliation(s)
- Neda Satvat
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Oliver Korczynski
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Matthias Müller-Eschner
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Ahmed E. Othman
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Vanessa Schöffling
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Naureen Keric
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Clemens Sommer
- Department of Neuropathology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | - Marc A. Brockmann
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-6131-17-7139
| | - Sebastian Reder
- Department of Neuroradiology, University Medical Centre, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| |
Collapse
|
39
|
Foo CY, Munir N, Kumaria A, Akhtar Q, Bullock CJ, Narayanan A, Fu RZ. Medical Device Advances in the Treatment of Glioblastoma. Cancers (Basel) 2022; 14:5341. [PMID: 36358762 PMCID: PMC9656148 DOI: 10.3390/cancers14215341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 07/30/2023] Open
Abstract
Despite decades of research and the growing emergence of new treatment modalities, Glioblastoma (GBM) frustratingly remains an incurable brain cancer with largely stagnant 5-year survival outcomes of around 5%. Historically, a significant challenge has been the effective delivery of anti-cancer treatment. This review aims to summarize key innovations in the field of medical devices, developed either to improve the delivery of existing treatments, for example that of chemo-radiotherapy, or provide novel treatments using devices, such as sonodynamic therapy, thermotherapy and electric field therapy. It will highlight current as well as emerging device technologies, non-invasive versus invasive approaches, and by doing so provide a detailed summary of evidence from clinical studies and trials undertaken to date. Potential limitations and current challenges are discussed whilst also highlighting the exciting potential of this developing field. It is hoped that this review will serve as a useful primer for clinicians, scientists, and engineers in the field, united by a shared goal to translate medical device innovations to help improve treatment outcomes for patients with this devastating disease.
Collapse
Affiliation(s)
- Cher Ying Foo
- Imperial College School of Medicine, Imperial College London, Fulham Palace Rd., London W6 8RF, UK
| | - Nimrah Munir
- QV Bioelectronics Ltd., 1F70 Mereside, Alderley Park, Nether Alderley, Cheshire SK10 4TG, UK
| | - Ashwin Kumaria
- Department of Neurosurgery, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham NG7 2UH, UK
| | - Qasim Akhtar
- QV Bioelectronics Ltd., 1F70 Mereside, Alderley Park, Nether Alderley, Cheshire SK10 4TG, UK
| | - Christopher J. Bullock
- QV Bioelectronics Ltd., 1F70 Mereside, Alderley Park, Nether Alderley, Cheshire SK10 4TG, UK
| | - Ashwin Narayanan
- QV Bioelectronics Ltd., 1F70 Mereside, Alderley Park, Nether Alderley, Cheshire SK10 4TG, UK
| | - Richard Z. Fu
- QV Bioelectronics Ltd., 1F70 Mereside, Alderley Park, Nether Alderley, Cheshire SK10 4TG, UK
- School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Michael, Smith Building, Dover St., Manchester M13 9PT, UK
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford Royal, Stott Lane, Salford M6 8HD, UK
| |
Collapse
|
40
|
Sumera, Anwer F, Waseem M, Fatima A, Malik N, Ali A, Zahid S. Molecular Docking and Molecular Dynamics Studies Reveal Secretory Proteins as Novel Targets of Temozolomide in Glioblastoma Multiforme. Molecules 2022; 27:7198. [PMID: 36364024 PMCID: PMC9653723 DOI: 10.3390/molecules27217198] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 10/13/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a tumor of glial origin and is the most malignant, aggressive and prevalent type, with the highest mortality rate in adult brain cancer. Surgical resection of the tumor followed by Temozolomide (TMZ) therapy is currently available, but the development of resistance to TMZ is a common limiting factor in effective treatment. The present study investigated the potential interactions of TMZ with several secretory proteins involved in various molecular and cellular processes in GBM. Automated docking studies were performed using AutoDock 4.2, which showed an encouraging binding affinity of TMZ towards all targeted proteins, with the strongest interaction and binding affinity with GDF1 and SLIT1, followed by NPTX1, CREG2 and SERPINI, among the selected proteins. Molecular dynamics (MD) simulations of protein-ligand complexes were performed via CABS-flex V2.0 and the iMOD server to evaluate the root-mean-square fluctuations (RMSFs) and measure protein stability, respectively. The results showed that docked models were more flexible and stable with TMZ, suggesting that it may be able to target putative proteins implicated in gliomagenesis that may impact radioresistance. However, additional in vitro and in vivo investigations can ascertain the potential of the selected proteins to serve as novel targets for TMZ for GBM treatment.
Collapse
Affiliation(s)
- Sumera
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Farha Anwer
- Integrative Biology Laboratory, Department of Industrial Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Maaz Waseem
- Integrative Biology Laboratory, Department of Industrial Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Areeba Fatima
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Nishat Malik
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Amjad Ali
- Integrative Biology Laboratory, Department of Industrial Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Saadia Zahid
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| |
Collapse
|
41
|
McCarthy L, Verma G, Hangel G, Neal A, Moffat BA, Stockmann JP, Andronesi OC, Balchandani P, Hadjipanayis CG. Application of 7T MRS to High-Grade Gliomas. AJNR Am J Neuroradiol 2022; 43:1378-1395. [PMID: 35618424 PMCID: PMC9575545 DOI: 10.3174/ajnr.a7502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/11/2022] [Indexed: 01/26/2023]
Abstract
MRS, including single-voxel spectroscopy and MR spectroscopic imaging, captures metabolites in high-grade gliomas. Emerging evidence indicates that 7T MRS may be more sensitive to aberrant metabolic activity than lower-field strength MRS. However, the literature on the use of 7T MRS to visualize high-grade gliomas has not been summarized. We aimed to identify metabolic information provided by 7T MRS, optimal spectroscopic sequences, and areas for improvement in and new applications for 7T MRS. Literature was found on PubMed using "high-grade glioma," "malignant glioma," "glioblastoma," "anaplastic astrocytoma," "7T," "MR spectroscopy," and "MR spectroscopic imaging." 7T MRS offers higher SNR, modestly improved spatial resolution, and better resolution of overlapping resonances. 7T MRS also yields reduced Cramér-Rao lower bound values. These features help to quantify D-2-hydroxyglutarate in isocitrate dehydrogenase 1 and 2 gliomas and to isolate variable glutamate, increased glutamine, and increased glycine with higher sensitivity and specificity. 7T MRS may better characterize tumor infiltration and treatment effect in high-grade gliomas, though further study is necessary. 7T MRS will benefit from increased sample size; reductions in field inhomogeneity, specific absorption rate, and acquisition time; and advanced editing techniques. These findings suggest that 7T MRS may advance understanding of high-grade glioma metabolism, with reduced Cramér-Rao lower bound values and better measurement of smaller metabolite signals. Nevertheless, 7T is not widely used clinically, and technical improvements are necessary. 7T MRS isolates metabolites that may be valuable therapeutic targets in high-grade gliomas, potentially resulting in wider ranging neuro-oncologic applications.
Collapse
Affiliation(s)
- L McCarthy
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| | - G Verma
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - G Hangel
- Department of Neurosurgery (G.H.)
- High-field MR Center (G.H.), Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - A Neal
- Department of Medicine (A.N.), Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
- Department of Neurology (A.N.), Royal Melbourne Hospital, Melbourne, Australia
| | - B A Moffat
- The Melbourne Brain Centre Imaging Unit (B.A.M.), Department of Radiology, The University of Melbourne, Melbourne, Australia
| | - J P Stockmann
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - O C Andronesi
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - P Balchandani
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - C G Hadjipanayis
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| |
Collapse
|
42
|
Shu T, Ding L, Fang Z, Yu S, Chen L, Moser MAJ, Zhang W, Qin Z, Zhang B. Lethal Electric Field Thresholds for Cerebral Cells With Irreversible Electroporation and H-FIRE Protocols: An In Vitro Three-Dimensional Cell Model Study. J Biomech Eng 2022; 144:1140297. [PMID: 35445240 DOI: 10.1115/1.4054381] [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: 01/24/2022] [Indexed: 11/08/2022]
Abstract
The lethal electric field (LEF) thresholds for three typical cerebral cells, including a malignant glioblastoma (GBM) cell line and two cell lines from the healthy blood-brain barrier (BBB), treated by irreversible electroporation (IRE) or high-frequency irreversible electroporation (H-FIRE) protocols were investigated in an in vitro three-dimensional (3D) cell model. A conventional IRE protocol (90 pulses, 1 Hz, and 100-μs pulse duration) and three novel H-FIRE protocols (1-3-1, 0.5-1-0.5, and 1-1-1) were used to treat the cerebral cells in both 3D single-cell and two-cell models. The electrical conductivity of the 3D cell model under different electric field strengths were characterized with the method of electrochemical impedance spectroscopy (EIS). Based on EIS, a numerical electrothermal model of electroporation was built for the determination of the LEF threshold with different protocols and temperature monitoring. Cell viability was assessed by fluorescence staining 6 h after the treatment. The results showed no thermal lethal effect on cells when these protocols were used. The LEF threshold for GBM cells was significantly lower than that of the healthy BBB cells. These results suggest the possibility of selective ablation of human cerebral GBM by IRE and H-FIRE treatments with no injury or reversible injury to healthy cells, and the potential use of IRE or H-FIRE for transient disruption of the BBB to allow chemotherapy to reach the tumor.
Collapse
Affiliation(s)
- Ting Shu
- Intelligent Energy-Based Tumor Ablation Laboratory, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Lujia Ding
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Zheng Fang
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Shuangquan Yu
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Michael A J Moser
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Wenjun Zhang
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Bing Zhang
- Intelligent Energy-Based Tumor Ablation Laboratory, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| |
Collapse
|
43
|
Chiesa S, Mangraviti A, Martini M, Cenci T, Mazzarella C, Gaudino S, Bracci S, Martino A, Della Pepa GM, Offi M, Gessi M, Russo R, Martucci M, Beghella Bartoli F, Larocca LM, Lauretti L, Olivi A, Pallini R, Balducci M, D'Alessandris QG. Clinical and NGS predictors of response to regorafenib in recurrent glioblastoma. Sci Rep 2022; 12:16265. [PMID: 36171338 PMCID: PMC9519741 DOI: 10.1038/s41598-022-20417-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 09/13/2022] [Indexed: 11/09/2022] Open
Abstract
Predictive factors for response to regorafenib in recurrent glioblastoma, IDH-wildtype, are scarcely recognized. The objective of this study was to identify molecular predictive factors for response to regorafenib using a clinically available platform. We analyzed a prospective cohort of 30 patients harboring recurrent glioblastoma, IDH-wildtype, and treated with regorafenib. Next-generation sequencing (NGS) analysis was performed on DNA extracted from paraffin-embedded tissues using a clinically available platform. Moreover, MGMT methylation and EGFRvIII expression analyses were performed. Six-month progression-free survival (PFS) was 30% and median overall survival (OS) was 7.5 months, in line with literature data. NGS analysis revealed a mutation in the EGFR pathway in 18% of cases and a mutation in the mitogen-activated protein-kinase (MAPK) pathway in 18% of cases. In the remaining cases, no mutations were detected. Patients carrying MAPK pathway mutation had a poor response to regorafenib treatment, with a significantly shorter PFS and a nonsignificantly shorter OS compared to EGFR-mutated patients (for PFS, 2.5 vs 4.5 months, p = 0.0061; for OS, 7 vs 9 months, p = 0.1076). Multivariate analysis confirmed that MAPK pathway mutations independently predicted a shorter PFS after regorafenib treatment (p = 0.0188). The negative prognostic role of MAPK pathway alteration was reinforced when we combined EGFR-mutated with EGFRvIII-positive cases. Recurrent glioblastoma tumors with an alteration in MAPK pathway could belong to the mesenchymal subtype and respond poorly to regorafenib treatment, while EGFR-altered cases have a better response to regorafenib. We thus provide a molecular selection criterion easy to implement in the clinical practice.
Collapse
Affiliation(s)
- Silvia Chiesa
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Antonella Mangraviti
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Maurizio Martini
- Depatrment of Pathology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Tonia Cenci
- Depatrment of Pathology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Ciro Mazzarella
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Simona Gaudino
- Department of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Serena Bracci
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Antonella Martino
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Giuseppe M Della Pepa
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Martina Offi
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Marco Gessi
- Depatrment of Pathology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Rosellina Russo
- Department of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Matia Martucci
- Department of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Francesco Beghella Bartoli
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Luigi M Larocca
- Depatrment of Pathology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Liverana Lauretti
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Alessandro Olivi
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Roberto Pallini
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy.
| | - Mario Balducci
- Department of Radiation Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| | - Quintino Giorgio D'Alessandris
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168, Rome, Italy
| |
Collapse
|
44
|
Surgical Treatment of Glioblastoma: State-of-the-Art and Future Trends. J Clin Med 2022; 11:jcm11185354. [PMID: 36143001 PMCID: PMC9505564 DOI: 10.3390/jcm11185354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/17/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022] Open
Abstract
Glioblastoma (GBM) is a highly aggressive disease and is associated with poor prognosis despite treatment advances in recent years. Surgical resection of tumor remains the main therapeutic option when approaching these patients, especially when combined with adjuvant radiochemotherapy. In the present study, we conducted a comprehensive literature review on the state-of-the-art and future trends of the surgical treatment of GBM, emphasizing topics that have been the object of recent study.
Collapse
|
45
|
Substrate viscosity impairs temozolomide-mediated inhibition of glioblastoma cells' growth. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166513. [PMID: 35932892 DOI: 10.1016/j.bbadis.2022.166513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/16/2022] [Accepted: 07/29/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND The mechanical state of the extracellular environment of the brain cells considerably affects their phenotype during the development of central nervous system (CNS) pathologies, and when the cells respond to drugs. The reports on the evaluation of the viscoelastic properties of different brain tumors have shown that both tissue stiffness and viscosity can be altered during cancer development. Although a compelling number of reports established the role of substrate stiffness on the proliferation, motility, and drug sensitivity of brain cancer cells, there is a lack of parallel data in terms of alterations in substrate viscosity. METHODS Based on viscoelasticity measurements of rat brain samples using strain rheometry, polyacrylamide (PAA) hydrogels mimicking elastic and viscous parameters of the tissues were prepared. Optical microscopy and flow cytometry were employed to assess the differences in glioblastoma cells morphology, proliferation, and cytotoxicity of anticancer drug temozolomide (TZM) due to increased substrate viscosity. RESULTS Our results indicate that changes in substrate viscosity affect the proliferation of untreated glioma cells to a lesser extent, but have a significant impact on the apoptosis-associated depolarization of mitochondria and level of DNA fragmentation. This suggests that viscosity sensing and stiffness sensing machinery can activate different signaling pathways in glioma cells. CONCLUSION Collected data indicate that viscosity should be considered an important parameter in in vitro polymer-based cell culture systems used for drug screening.
Collapse
|
46
|
Xie S, Hong Z, Li Y, Wang J, Wang J, Li S, Liu Y. RNF216 Alleviates Radiation-Induced Apoptosis and DNA Damage Through Regulating Ubiquitination-Mediated Degradation of p53 in Glioblastoma. Mol Neurobiol 2022; 59:4703-4717. [PMID: 35594003 DOI: 10.1007/s12035-022-02868-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
Glioblastoma (GBM) is the most common and lethal subtype of glioma, characterized by uncontrolled cancer cell proliferation, extensive infiltration, and therapeutic resistance. Ring finger protein 216 (RNF216) is a RING-type E3 ubiquitin ligase aberrantly expressed in multiple human cancers. Tumor protein 53 (p53) is a transcription factor that acts as a tumor suppressor. This study aimed to compare the RNF216 expression in GBM tissues and normal peritumoral tissues and to examine the effects of RNF216 overexpression/knockdown on tumorigenesis, radioresistance, and the p53 pathway in GBM. The results showed that RNF216 was overexpressed in GBM tissues and cell lines, and high RNF216 expression was related to a poor prognosis. RNF216 overexpression promoted GBM cell growth and inhibited apoptosis, while RNF216 knockdown impaired GBM cell growth and enhanced cell death. RNF216 was also highly expressed in recurrent GBM tissues compared with paired primary tumors. The upregulation of RNF216 not only facilitated GBM cell growth but also protected cells against X-ray irradiation-induced apoptosis and DNA damage, while RNF216 knockdown exerted opposite effects. Moreover, the implantation of GBM cells with RNF216 silencing suppressed tumorigenesis and increased radiosensitivity of mice bearing GBM xenografts. Further analysis revealed that RNF216 overexpression reduced the stability of p53 protein via ubiquitination and negatively regulated the p53 pathway, while RNF216 knockdown preserved the p53 protein. In conclusion, RNF216 effectively attenuated radiation-induced apoptosis and DNA damage in GBM via inducing ubiquitination-mediated degradation of p53. These findings suggest the potential therapeutic use of RNF216 inhibition for tumorigenesis and therapeutic resistance in GBM.
Collapse
Affiliation(s)
- Songwang Xie
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Zhen Hong
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yan Li
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Junyong Wang
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Jian Wang
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Shaoquan Li
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yongchang Liu
- Department of Neurovascular Intervention, Cangzhou Central Hospital, Cangzhou, Hebei, China.
| |
Collapse
|
47
|
Mechanical Properties of the Extracellular Environment of Human Brain Cells Drive the Effectiveness of Drugs in Fighting Central Nervous System Cancers. Brain Sci 2022; 12:brainsci12070927. [PMID: 35884733 PMCID: PMC9313046 DOI: 10.3390/brainsci12070927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
The evaluation of nanomechanical properties of tissues in health and disease is of increasing interest to scientists. It has been confirmed that these properties, determined in part by the composition of the extracellular matrix, significantly affect tissue physiology and the biological behavior of cells, mainly in terms of their adhesion, mobility, or ability to mutate. Importantly, pathophysiological changes that determine disease development within the tissue usually result in significant changes in tissue mechanics that might potentially affect the drug efficacy, which is important from the perspective of development of new therapeutics, since most of the currently used in vitro experimental models for drug testing do not account for these properties. Here, we provide a summary of the current understanding of how the mechanical properties of brain tissue change in pathological conditions, and how the activity of the therapeutic agents is linked to this mechanical state.
Collapse
|
48
|
Werner M, Lyu C, Stadlbauer B, Schrader I, Buchner A, Stepp H, Sroka R, Pohla H. The role of shikonin in improving 5-aminolevulinic acid-based photodynamic therapy and chemotherapy on glioblastoma stem cells. Photodiagnosis Photodyn Ther 2022; 39:102987. [PMID: 35760350 DOI: 10.1016/j.pdpdt.2022.102987] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022]
Abstract
Glioblastoma multiforme remains a malignant neoplasia with a median survival of less than two years and without satisfactory therapeutic options. The so-called glioblastoma stem cells escape the established radio- and chemotherapies and lead to tumor recurrence in most cases. The alkaloid Shikonin with its various anti stem cell properties and the interstitial photodynamic therapy with 5-aminolevulinic acid seem to be promising new options in the therapy of glioblastoma. In this study, in vitro investigations were performed to observe the influence of Shikonin on viability, proliferation, induction of apoptosis and the capability of forming tumor spheres in U-87 MG and the primary glioblastoma cell line GB14. The combined effect with the chemotherapeutic temozolomide and photodynamic treatment on the mRNA expression of glioma specific stem cell markers and further examined intracellular protoporphyrin IX accumulation under Shikonin treatment was analyzed. Shikonin effectively inhibited the capability of forming tumor spheres and enhanced temozolomide effectiveness in the reduction of proliferation and in the induction of apoptosis. Additionally, Shikonin increased the mRNA expression of the tumor suppressing Neurofibromatosis type 1 (NF1) gene and showed modulating effects on intracellular protoporphyrin IX.
Collapse
Affiliation(s)
- Maxim Werner
- Laboratory of Tumor Immunology, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany.
| | - Chen Lyu
- Laboratory of Tumor Immunology, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany
| | - Birgit Stadlbauer
- Laboratory of Tumor Immunology, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany; Department of Urology, LMU Klinikum, University Munich, 82151 Planegg, Germany
| | - Isabel Schrader
- Laboratory of Tumor Immunology, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany
| | - Alexander Buchner
- Laboratory of Tumor Immunology, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany; Department of Urology, LMU Klinikum, University Munich, 82151 Planegg, Germany
| | - Herbert Stepp
- Department of Urology, LMU Klinikum, University Munich, 82151 Planegg, Germany; Laser-Forschungslabor, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany
| | - Ronald Sroka
- Department of Urology, LMU Klinikum, University Munich, 82151 Planegg, Germany; Laser-Forschungslabor, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany
| | - Heike Pohla
- Laboratory of Tumor Immunology, LIFE Center, LMU Klinikum, University Munich, 82151 Planegg, Germany; Department of Urology, LMU Klinikum, University Munich, 82151 Planegg, Germany
| |
Collapse
|
49
|
Catanzaro D, Milani G, Bozza A, Bernardi M, Chieregato K, Menarin M, Merlo A, Celli P, Belli R, Peroni D, Pozzato A, Pozzato G, Raneri FA, Volpin L, Ruggeri M, Astori G. Selective cell cycle arrest in glioblastoma cell lines by quantum molecular resonance alone or in combination with temozolomide. Br J Cancer 2022; 127:824-835. [PMID: 35715634 PMCID: PMC9427848 DOI: 10.1038/s41416-022-01865-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background Glioblastoma is the most aggressive form of brain cancer, characterised by high proliferation rates and cell invasiveness. Despite advances in surgery and radio-chemotherapy, patients continue to have poor prognoses, with a survival rate of 14–15 months. Thus, new therapeutic strategies are needed. Non-ionising electromagnetic fields represent an emerging option given the potential advantages of safety, low toxicity and the possibility to be combined with other therapies. Methods Here, the anticancer activity of quantum molecular resonance (QMR) was investigated. For this purpose, three glioblastoma cell lines were tested, and the QMR effect was evaluated on cancer cell proliferation rate and aggressiveness. To clarify the QMR mechanism of action, the proteomic asset after stimulation was delineated. Mesenchymal stromal cells and astrocytes were used as healthy controls. Results QMR affected cancer cell proliferation, inducing a significant arrest of cell cycle progression and reducing cancer tumorigenicity. These parameters were not altered in healthy control cells. Proteomic analysis suggested that QMR acts not only on DNA replication but also on the machinery involved in the mitotic spindle assembly and chromosome segregation. Moreover, in a combined therapy assessment, QMR significantly enhanced temozolomide efficacy. Conclusions QMR technology appears to be a promising tool for glioblastoma treatment.
Collapse
Affiliation(s)
- Daniela Catanzaro
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy.,CORIS, Consorzio per la Ricerca Sanitaria, Via N. Giustiniani, 2, 35128, Padova, Italy
| | - Gloria Milani
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy.,CORIS, Consorzio per la Ricerca Sanitaria, Via N. Giustiniani, 2, 35128, Padova, Italy
| | - Angela Bozza
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy.,CORIS, Consorzio per la Ricerca Sanitaria, Via N. Giustiniani, 2, 35128, Padova, Italy
| | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy.,CORIS, Consorzio per la Ricerca Sanitaria, Via N. Giustiniani, 2, 35128, Padova, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy.,CORIS, Consorzio per la Ricerca Sanitaria, Via N. Giustiniani, 2, 35128, Padova, Italy
| | - Martina Menarin
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Anna Merlo
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Paola Celli
- Genetic Unit, Vicenza Hospital, Vicenza, Italy
| | - Romina Belli
- Mass Spectrometry and Proteomics Facility, Department of Cellular, Computational and Integrative Biology, CIBIO University of Trento, Trento, Italy
| | - Daniele Peroni
- Mass Spectrometry and Proteomics Facility, Department of Cellular, Computational and Integrative Biology, CIBIO University of Trento, Trento, Italy
| | | | | | | | - Lorenzo Volpin
- Department of Neurosurgery, Vicenza Hospital, Vicenza, Italy
| | | | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy.
| |
Collapse
|
50
|
Mehidine H, Devaux B, Varlet P, Abi Haidar D. Comparative Study Between a Customized Bimodal Endoscope and a Benchtop Microscope for Quantitative Tissue Diagnosis. Front Oncol 2022; 12:881331. [PMID: 35686105 PMCID: PMC9171499 DOI: 10.3389/fonc.2022.881331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/31/2022] [Indexed: 12/24/2022] Open
Abstract
Nowadays, surgical removal remains the standard method to treat brain tumors. During surgery, the neurosurgeon may encounter difficulties to delimitate tumor boundaries and the infiltrating areas as they have a similar visual appearance to adjacent healthy zones. These infiltrating residuals increase the tumor recurrence risk, which decreases the patient’s post-operation survival time. To help neurosurgeons improve the surgical act by accurately delimitating healthy from cancerous areas, our team is developing an intraoperative multimodal imaging tool. It consists of a two-photon fluorescence fibered endomicroscope that is intended to provide a fast, real-time, and reliable diagnosis information. In parallel to the instrumental development, a large optical database is currently under construction in order to characterize healthy and tumor brain tissues with their specific optical signature using multimodal analysis of the endogenous fluorescence. Our previous works show that this multimodal analysis could provide a reliable discrimination response between different tissue types based on several optical indicators. Here, our goal is to show that the two-photon fibered endomicroscope is able to provide, based on the same approved indicators in the tissue database, the same reliable response that could be used intraoperatively. We compared the spectrally resolved and time-resolved fluorescence signal, generated by our two-photon bimodal endoscope from 46 fresh brain tissue samples, with a similar signal provided by a standard reference benchtop multiphoton microscope that has been validated for tissue diagnosis. The higher excitation efficiency and collection ability of an endogenous fluorescence signal were shown for the endoscope setup. Similar molecular ratios and fluorescence lifetime distributions were extracted from the two compared setups. Spectral discrimination ability of the bimodal endoscope was validated. As a preliminary step before tackling multimodality, the ability of the developed bimodal fibered endoscope to excite and to collect efficiently as well as to provide a fast exploitable high-quality signal that is reliable to discriminate different types of human brain tissues was validated.
Collapse
Affiliation(s)
| | - Bertrand Devaux
- Université Paris Cité - Faculté de Médecine Paris Descartes, Paris, France.,Service de Neurochirurgie, Hôpital Lariboisière, Paris, France.,Department of Neurosurgery, GHU Paris Psychiatrie et Neuroscience, Paris, France
| | - Pascale Varlet
- Université Paris Cité - Faculté de Médecine Paris Descartes, Paris, France.,Department of Neuropathology, GHU Paris-Psychiatrie et Neurosciences, Sainte-Anne Hospital, Paris, France.,IMA BRAIN, INSERM UMR S1266, Centre de Psychiatrie et de Neurosciences, Paris, France
| | - Darine Abi Haidar
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, Orsay, France.,Université Paris Cité, IJCLab, Orsay, France
| |
Collapse
|