1
|
Iorio AL, Lenci E, Marzano C, Bucaletti E, Tirinnanzi B, Casati G, Giunti L, Dallari C, Credi C, Sardi I, Trabocchi A. Oxime Linked Doxorubicin Glycoconjugates Improve the Specific Targeting of Glioblastoma in High-Grade Glioma Therapy. ACS Med Chem Lett 2024; 15:1953-1960. [PMID: 39563793 PMCID: PMC11571026 DOI: 10.1021/acsmedchemlett.4c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
The treatment of glioblastoma (GBM) represents an urgent challenge for public health due to the inability to effectively deliver anticancer agents, such as doxorubicin (DOX), through the blood-brain barrier (BBB). Herein we report the synthesis of two novel DOX glycoconjugates using an oxime linkage that maintained the intercalation capability of the planar anthracycline ring of DOX, as demonstrated by UV-vis and fluorescence experiments in the presence of DNA. The biological effect of DOX glycoconjugates was evaluated in GBM cell lines, showing an enhanced cytotoxic and pro-apoptotic effect of 7 as compared to 4 and to conventional DOX. These data were confirmed in an in vitro coculture BBB model in which DOX glycoconjugate 7 showed high capability to cross a cellular monolayer and exert its cytotoxic effect on GBM cells. The results show that conjugation with glucose may represent a helpful tool to increase chemotherapy effectiveness in poor-responding GBM patients.
Collapse
Affiliation(s)
- Anna Lisa Iorio
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, 50139 Florence, Italy
| | - Elena Lenci
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| | - Chiara Marzano
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Elisabetta Bucaletti
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| | - Bianca Tirinnanzi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Giacomo Casati
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Laura Giunti
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Caterina Dallari
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Florence, Italy
- National Institute of Optics National Research Council, 50019 Sesto Fiorentino, Florence, Italy
| | - Caterina Credi
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Florence, Italy
- National Institute of Optics National Research Council, 50019 Sesto Fiorentino, Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Andrea Trabocchi
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
2
|
Peng G, Feng Y, Wang X, Huang W, Li Y. The mitochondria-related gene risk mode revealed p66Shc as a prognostic mitochondria-related gene of glioblastoma. Sci Rep 2024; 14:11418. [PMID: 38763954 PMCID: PMC11102912 DOI: 10.1038/s41598-024-62083-2] [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: 11/01/2023] [Accepted: 05/13/2024] [Indexed: 05/21/2024] Open
Abstract
Numerous studies have highlighted the pivotal role of mitochondria-related genes (MRGs) in the initiation and progression of glioblastoma (GBM). However, the specific contributions of MRGs coding proteins to GBM pathology remain incompletely elucidated. The identification of prognostic MRGs in GBM holds promise for the development of personalized targeted therapies and the enhancement of patient prognosis. We combined differential expression with univariate Cox regression analysis to screen prognosis-associated MRGs in GBM. Based on the nine MRGs, the hazard ratio model was conducted using a multivariate Cox regression algorithm. SHC-related survival, pathway, and immune analyses in GBM cohorts were obtained from the Biomarker Exploration of the Solid Tumor database. The proliferation and migration of U87 cells were measured by CCK-8 and transwell assay. Apoptosis in U87 cells was evaluated using flow cytometry. Confocal microscopy was employed to measure mitochondrial reactive oxygen species (ROS) levels and morphology. The expression levels of SHC1 and other relevant proteins were examined via western blotting. We screened 15 prognosis-associated MRGs and constructed a 9 MRGs-based model. Validation of the model's risk score confirmed its efficacy in predicting the prognosis of patients with GBM. Furthermore, analysis revealed that SHC1, a constituent MRG of the prognostic model, was upregulated and implicated in the progression, migration, and immune infiltration of GBM. In vitro experiments elucidated that p66Shc, the longest isoform of SHC1, modulates mitochondrial ROS production and morphology, consequently promoting the proliferation and migration of U87 cells. The 9 MRGs-based prognostic model could predict the prognosis of GBM. SHC1 was upregulated and correlated with the prognosis of patients by involvement in immune infiltration. Furthermore, in vitro experiments demonstrated that p66Shc promotes U87 cell proliferation and migration by mediating mitochondrial ROS production. Thus, p66Shc may serve as a promising biomarker and therapeutic target for GBM.
Collapse
Affiliation(s)
- Gang Peng
- Department of Phamacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Yabo Feng
- PET-CT Center, Chenzhou First People's Hospital, Chenzhou, 423000, Hunan, People's Republic of China
| | - Xiangyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Weicheng Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Yang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
3
|
Zhao G, Deng Z, Li X, Wang H, Chen G, Feng M, Zhou Y. Targeting EZH2 regulates the biological characteristics of glioma stem cells via the Notch1 pathway. Exp Brain Res 2023; 241:2409-2418. [PMID: 37644332 DOI: 10.1007/s00221-023-06693-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Glioma is the most common malignant brain tumor, and its behavior is closely related to the presence of glioma stem cells (GSCs). We found that the enhancer of zeste homolog 2 (EZH2) is highly expressed in glioma and that its expression is correlated with the prognosis of glioblastoma multiforme (GBM) in two databases: The Cancer Genome Atlas and the Chinese Glioma Genome Atlas. Additionally, EZH2 is known to regulate the stemness-associated gene expression, proliferation, and invasion ability of GSCs, which may be achieved through the activation of the STAT3 and Notch1 pathways. Furthermore, we demonstrated the effect of the EZH2-specific inhibitor GSK126 on GSCs; these results not only corroborate our hypothesis, but also provide a potential novel treatment approach for glioma.
Collapse
Affiliation(s)
- Guozheng Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Department of Neurosurgery, Suzhou Ninth People's Hospital, Suzhou, 215000, China
| | - Zhitong Deng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Department of Neurosurgery, The First Affiliated Hospital of Huzhou University, Huzhou, 313000, China
| | - Xuetao Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Hao Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Guangliang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Ming Feng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Youxin Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| |
Collapse
|
4
|
Scherm A, Ippen FM, Hau P, Baurecht H, Wick W, Gempt J, Knüttel H, Leitzmann MF, Seliger C. Targeted therapies in patients with newly diagnosed glioblastoma-A systematic meta-analysis of randomized clinical trials. Int J Cancer 2023; 152:2373-2382. [PMID: 36647335 DOI: 10.1002/ijc.34433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023]
Abstract
Glioblastoma (GB) is the most common malignant primary brain tumor in adults. The standard of care for newly diagnosed GB involves surgical resection followed by radiochemotherapy with temozolomide, with or without tumor-treating fields. In recent years, various efforts have been made to identify suitable molecularly targeted treatment options for malignant brain tumors. This meta-analysis provides an overview of recently published randomized controlled trials (RCTs) with and without molecular stratification, analyzing targeted agents in patients with newly diagnosed GB. The Cochrane Library, MEDLINE (Ovid), ClinicalTrials.gov, WHO's International Clinical Trials Registry Platform, and Google Scholar were searched for RCTs on targeted therapies in patients with newly diagnosed glioblastoma. Hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS) were extracted and pooled in a random-effects meta-analysis. Twelve RCTs (n = 3941 patients) involving protein kinase inhibitors, proteasome and histone deacetylase inhibitors, anti-angiogenic approaches and poly (ADP-ribose) polymerase (PARP) inhibitors were included in the meta-analysis. None of the targeted agents achieved a significant benefit with regard to OS (HR = 0.98 [95% confidence interval (CI) 0.86-1.11, P = .7731]). By comparison, targeted therapy showed a benefit for PFS (HR = 0.83 [95% CI 0.74-0.94, P = .0037]), especially for patients with an unmethylated O6-methylguanine-DNA-methyltransferase (MGMT) promoter (0.75 [95% CI 0.56-0.99, P = .0440]). Prolongation of PFS was largely driven by VEGF inhibition with bevacizumab (HR = 0.70 [95% CI 0.61-0.80, P = .0000]). VEGF inhibition with bevacizumab prolonged PFS in patients with newly diagnosed glioblastoma compared to standard care. However, no improvement in OS was observed with any of the targeted agents.
Collapse
Affiliation(s)
- Angelika Scherm
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | | | - Peter Hau
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, Regensburg University Hospital, Regensburg, Germany
| | - Hansjörg Baurecht
- Institute of Epidemiology and Preventive Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Wolfgang Wick
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Research Center (DKFZ) & German Cancer Center (DKTK), Heidelberg, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Helge Knüttel
- University Library, Regensburg University, Regensburg, Germany
| | - Michael F Leitzmann
- Institute of Epidemiology and Preventive Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Corinna Seliger
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
5
|
Tamtaji OR, Razavi ZS, Razzaghi N, Aschner M, Barati E, Mirzaei H. Quercetin and Glioma: Which signaling pathways are involved? Curr Mol Pharmacol 2022; 15:962-968. [DOI: 10.2174/1874467215666220211094136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Gliomas are the most common brain tumors. These tumors commonly exhibit continuous growth without invading surrounding brain tissues. Dominant remedial approaches suffer limited therapy and survival rates. Although some progress has been made in conventional glioma treatments, these breakthroughs have not yet proven sufficient for treating this malignancy. The remedial options are limited given gliomas' aggressive metastasis and drug resistance. Quercetin, a flavonoid, is an anti-oxidative, anti-allergic, antiviral, anti-inflammatory, and anticancer compound. Multiple lines of evidence have shown that Quercetin has anti-tumor effects, documenting this natural compound exerts its pharmacological effects by targeting a variety of cellular and molecular processes, i.e., apoptosis, metastasis, and autophagy. Herein, we summarize various cellular and molecular pathways that are affected by Quercetin in gliomas.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zahra Sadat Razavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Nazanin Razzaghi
- Laboratory Sciences Research Centre, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - Erfaneh Barati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
6
|
Sfifou F, Hakkou EM, Bouaiti ELA, Slaoui M, Errihani H, Al Bouzidi A, Abouqal R, El Ouahabi A, Cherradi N. Correlation of immunohistochemical expression of HIF-1alpha and IDH1 with clinicopathological and therapeutic data of moroccan glioblastoma and survival analysis. Ann Med Surg (Lond) 2021; 69:102731. [PMID: 34466221 PMCID: PMC8384773 DOI: 10.1016/j.amsu.2021.102731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/15/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Glioblastomas are aggressive primary intracranial tumours of the central nervous system causing significant mortality and morbidity worldwide. OBJECTIVE This study aims to evaluate the prognostic value of tissue expression by immunostaining of hypoxia-inducible factor (HIF-1α), isocitrate dehydrogenase 1 (IDH1), and tumour protein p53 in glioblastoma in Moroccan patients. The association of HIF-1α, IDH1, and p53 expression with the clinicopathological data and overall patient survival (OS) was also evaluated. MATERIALS AND METHODS Confirmed glioblastomas were included in this study. Twenty-two tissue samples were obtained by neurosurgical intervention resulting from total resection, and subtotal resection or biopsy. Karnofsky index, histological type of tumour, and the status of IDH1, p53 protein, and HIF-1α expression by immunostaining were reported. RESULTS The majority of the patients were males (64%) with a sex ratio of 1.75. The average age was 54 ± 13. Median follow-up was 10.10 months and median overall survival was 10 months. The expression of HIF-1α was high in 10 samples (45%) and low in 12 (55%). There was a statistically significant difference in OS of 85% at 12 months for the subgroup of patients "HIF-1α negative IDH1 positive" p = 0.038, the unadjusted analysis showed that the group "HIF-1α positive, IDH1 positive" was a poor prognostic factor, the HR was 0.08 (95% CI: 0.009-0.756, p = 0.027). CONCLUSION Patients with negative HIF-1α expression and positive IDH1 expression have a better prognosis, suggesting that these two biomarkers may be useful in the search for new approaches for targeted therapy in glioblastoma.
Collapse
Affiliation(s)
- Fatima Sfifou
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
- Pathological Anatomy Department, Hospital of Specialities in Rabat, Morocco
| | - El Mehdi Hakkou
- Neurosurgery Department, Hospital of Specialities in Rabat, Morocco
| | - EL Arbi Bouaiti
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Rabat Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Meriem Slaoui
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Hassan Errihani
- National Oncology Centre Sidi Mohamed Ben Abdallah in Rabat, Morocco
| | - Abderrahmane Al Bouzidi
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Redouane Abouqal
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Rabat Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | | | - Nadia Cherradi
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy. Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
- Pathological Anatomy Department, Hospital of Specialities in Rabat, Morocco
| |
Collapse
|
7
|
Khalafallah AM, Rakovec M, Bettegowda C, Jackson CM, Gallia GL, Weingart JD, Lim M, Esquenazi Y, Zacharia BE, Goldschmidt E, Ziu M, Ivan ME, Venteicher AS, Nduom EK, Mamelak AN, Chu RM, Yu JS, Sheehan JP, Nahed BV, Carter BS, Berger MS, Sawaya R, Mukherjee D. A Crowdsourced Consensus on Supratotal Resection Versus Gross Total Resection for Anatomically Distinct Primary Glioblastoma. Neurosurgery 2021; 89:712-719. [PMID: 34320218 DOI: 10.1093/neuros/nyab257] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Gross total resection (GTR) of contrast-enhancing tumor is associated with increased survival in primary glioblastoma. Recently, there has been increasing interest in performing supratotal resections (SpTRs) for glioblastoma. OBJECTIVE To address the published results, which have varied in part due to lack of consensus on the definition and appropriate use of SpTR. METHODS A crowdsourcing approach was used to survey 21 neurosurgical oncologists representing 14 health systems nationwide. Participants were presented with 11 definitions of SpTR and asked to rate the appropriateness of each definition. Participants reviewed T1-weighed postcontrast and fluid-attenuated inversion-recovery magnetic resonance imaging for 22 anatomically distinct glioblastomas. Participants were asked to assess the tumor location's eloquence, the perceived equipoise of enrolling patients in a randomized trial comparing gross total to SpTR, and their personal treatment plans. RESULTS Most neurosurgeons surveyed (n = 18, 85.7%) agree that GTR plus resection of some noncontrast enhancement is an appropriate definition for SpTR. Overall, moderate inter-rater agreement existed regarding eloquence, equipoise, and personal treatment plans. The 4 neurosurgeons who had performed >10 SpTRs for glioblastomas in the past year were more likely to recommend it as their treatment plan (P < .005). Cases were divided into 3 anatomically distinct groups based upon perceived eloquence. Anterior temporal and right frontal glioblastomas were considered the best randomization candidates. CONCLUSION We established a consensus definition for SpTR of glioblastoma and identified anatomically distinct locations deemed most amenable to SpTR. These results may be used to plan prospective trials investigating the potential clinical utility of SpTR for glioblastoma.
Collapse
Affiliation(s)
- Adham M Khalafallah
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Maureen Rakovec
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher M Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gary L Gallia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jon D Weingart
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center, Houston, Texas, USA
| | - Brad E Zacharia
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ezequiel Goldschmidt
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Mateo Ziu
- Inova Neuroscience and Spine Institute, University of Virginia Medical School-Inova Campus, Falls Church, Virginia, USA
| | - Michael E Ivan
- Sylvester Comprehensive Cancer Center, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Andrew S Venteicher
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Edjah K Nduom
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Adam N Mamelak
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ray M Chu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - John S Yu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Brian V Nahed
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Raymond Sawaya
- Division of Surgery, Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Debraj Mukherjee
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
8
|
A Z, J SW, A M, E L, I W, W R, J JG. LY294002 and sorafenib as inhibitors of intracellular survival pathways in the elimination of human glioma cells by programmed cell death. Cell Tissue Res 2021; 386:17-28. [PMID: 34236519 PMCID: PMC8526469 DOI: 10.1007/s00441-021-03481-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/02/2021] [Indexed: 11/05/2022]
Abstract
Gliomas are aggressive brain tumors with very high resistance to chemotherapy throughout the overexpression of multiple intracellular survival pathways. Therefore, the aim of the present study was to investigate for the first time the anticancer activity of LY294002, phosphatidylinositol 3-kinase (PI3K) inhibitor and sorafenib, and rapidly accelerated fibrosarcoma kinase (Raf) inhibitor in the elimination of human glioma cells by programmed cell death. MOGGCCM (anaplastic astrocytoma, III) and T98G (glioblastoma multiforme, IV) cell lines incubated with LY294002 and/or sorafenib were used in the experiments. Simultaneous treatment with both drugs was more effective in the elimination of cancer cells on the way of apoptosis with no significant necrotic effect than single application. It was correlated with decreasing the mitochondrial membrane potential and activation of caspase 3 and 9. The expression of Raf and PI3K was also inhibited. Blocking of those kinases expression by specific siRNA revealed significant apoptosis induction, exceeding the level observed after LY294002 and sorafenib treatment in non-transfected lines but only in MOGGCCM cells. Our results indicated that combination of LY294002 and sorafenib was very efficient in apoptosis induction in glioma cells. Anaplastic astrocytoma cells turned out to be more sensitive for apoptosis induction than glioblastoma multiforme after blocking PI3K and Raf expression with siRNA.
Collapse
Affiliation(s)
- Zając A
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland.
| | - Sumorek-Wiadro J
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| | - Maciejczyk A
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| | - Langner E
- Department of Medical Biology, Institute of Agricultural Medicine, Lublin, Poland
| | - Wertel I
- 1st Department of Gynecology, University School of Medicine, Lublin, Poland
| | - Rzeski W
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland.,Department of Medical Biology, Institute of Agricultural Medicine, Lublin, Poland
| | - Jakubowicz-Gil J
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| |
Collapse
|
9
|
Cui Y, Yang Z, Wang H, Yan Y, Huang Q, Gong Z, Hong F, Zhang X, Li W, Chen J, Xu T. Identification of CDKL3 as a critical regulator in development of glioma through regulating RRM2 and the JNK signaling pathway. Cancer Sci 2021; 112:3150-3162. [PMID: 34097336 PMCID: PMC8353949 DOI: 10.1111/cas.15010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/12/2022] Open
Abstract
Glioma is one of the most commonly diagnosed intracranial malignancies. The molecular mechanism underlying the development of glioma is still largely unknown. In this study, we present the first report concerning the function and mechanism of cyclin‐dependent kinase‐like 3 (CDKL3) in the development and prognosis of glioma. It is shown that CDKL3 was upregulated in glioma tissues and could independently predict poor prognosis of patients. Silencing CDKL3 in glioma cells could inhibit cell proliferation and migration and induce cell apoptosis and cell cycle arrest, whereas the overexpression of CDKL3 promoted cell proliferation. The in vivo experiments also indicated that knockdown of CDKL3 significantly suppressed tumor growth of glioma. Gene expression profiling of CDKL3 knockdown U87 cells identified RRM2 as a potential target of CDKL3, which was proved to have direct interaction with CDKL3. Given similar effects on glioma development with CDKL3, knockdown of RRM2 could rescue the effects of CDKL3 overexpression on glioma cells. Moreover, knockdown of CDKL3 or RRM2 suppressed the activity of JNK signaling, whereas CDKL3 overexpression produced the opposite effect. In conclusion, our results identified CDKL3 as a promotor for glioma, probably through the regulation of RRM2 and activation of the JNK signalling pathway, highlighting the significance of CDKL3 as a promising therapeutic target of glioma.
Collapse
Affiliation(s)
- Yong Cui
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai, China
| | - Zhigang Yang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongxiang Wang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yong Yan
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qilin Huang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhenyu Gong
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fan Hong
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xu Zhang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weiqing Li
- Department of Pathology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Juxiang Chen
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
10
|
Lensoside Aβ as an Adjuvant to the Anti-Glioma Potential of Sorafenib. Cancers (Basel) 2021; 13:cancers13112637. [PMID: 34072003 PMCID: PMC8198162 DOI: 10.3390/cancers13112637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Flavonoids are plant polyphenolic secondary metabolites, commonly consumed in the human diet. Lensoside Aβ is a quercetin glycoside isolated from the aerial parts of lentil (Lens culinaris) organs. The activity of this secondary metabolite, especially in terms of its anticancer potential, has been poorly studied. Currently, there are no published data about the effect of this flavonoid on gliomas, which are so-far incurable, aggressive neoplasms of the central nervous system with a highly infiltrative character. In this study, we found that lensoside Aβ itself exhibits poor anti-glioma properties but exerts a strongly potentiated effect in combination with sorafenib (inhibitor of Raf kinase) on apoptosis induction in cancer cells. Our results have shown that sorafenib with lensoside Aβ seems to be a promising combination that might be useful in glioma therapy. Additionally, the former observation, pointing to the key role of flavonoids as adjuvants in chemotherapy, is confirmed. Abstract Aim: The anti-glioma effect of lensoside Aβ alone and in combination with sorafenib (pro-survival Raf kinase inhibitor) was evaluated for the first time in terms of programmed cell death induction in anaplastic astrocytoma and glioblastoma multiforme cell lines as an experimental model. Apoptosis, autophagy, and necrosis were identified microscopically (fluorescence and scanning microscopes) and confirmed by flow cytometry (mitochondrial membrane potential MMP and cell death). The expression of apoptotic (caspase 3) and autophagic markers (beclin 1) as well as Raf kinase were estimated by immunoblotting. The FTIR method was used to determine the interaction of the studied drugs with lipid and protein groups within cells, while the modes of drug action within the cells were assessed with the FLIM technique. Results: Lensoside Aβ itself does not exhibit anti-glioma activity but significantly enhances the anti-cancer potential of sorafenib, initiating mainly apoptosis of up to 90% of cells. It was correlated with an increased level of active caspase 3, a reduced MMP value, and a lower level of Raf kinase. The interaction with membrane structures led to morphological changes typical of programmed death. Conclusions: Our results indicate that lensoside Aβ plays an important role as an adjuvant in chemotherapy with sorafenib and may be a potential candidate in anti-glioma combination therapy.
Collapse
|
11
|
Effect of hnRNPA2/B1 on the proliferation and apoptosis of glioma U251 cells via the regulation of AKT and STAT3 pathways. Biosci Rep 2021; 40:225046. [PMID: 32463472 PMCID: PMC7350891 DOI: 10.1042/bsr20190318] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Glioma is the most common malignant tumor in the human central nervous system. Although heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2/B1) was previously presumed to be a tumor-promoting gene, the relationship between hnRNPA2/B1 and glioma is unclear. Targeting hnRNPA2/B1 interference in glioma cells can significantly inhibit proliferation and increase apoptosis of human glioma cells in vitro. In a tumor xenograft model, knockdown of hnRNPA2/B1 suppressed tumor growth in glioma cells in vivo. In terms of a mechanism, the knockdown of hnRNPA2/B1 led to inactivation of the AKT and STAT3 signaling pathways, which ultimately reduced the expression of B-cell lymphoma-2 (Bcl-2), CyclinD1 and proliferating cell nuclear antigen (PCNA). Collectively, these data suggest that the inhibition of hnRNPA2/B1 can reduce the growth of gliomas through STAT3 and AKT signaling pathways, and this inhibition is expected to be a therapeutic target for gliomas.
Collapse
|
12
|
Yang C, Zhan H, Zhao Y, Wu Y, Li L, Wang H. MEX3A contributes to development and progression of glioma through regulating cell proliferation and cell migration and targeting CCL2. Cell Death Dis 2021; 12:14. [PMID: 33414423 PMCID: PMC7791131 DOI: 10.1038/s41419-020-03307-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/03/2023]
Abstract
Glioma is one of the most commonly diagnosed intracranial malignant tumors with extremely high morbidity and mortality, whose treatment was seriously limited because of the unclear molecular mechanism. In this study, in order to identify a novel therapeutic target for glioma treatment, we explored the functions and mechanism of MEX3A in regulating glioma. The immunohistochemical staining of MEX3A in glioma and normal tissues revealed the upregulation of MEX3A and further indicated the relationship between high MEX3A expression and higher malignancy as well as poorer prognosis of glioma. In vitro loss-of-function and gain-of-function experiments comprehensively demonstrated that MEX3A may promote glioma development through regulating cell proliferation, cell apoptosis, cell cycle, and cell migration. In vivo experiments also suggested the inhibition of glioma growth by MEX3A knockdown. Moreover, our mechanistic study identifies CCL2 as a potential downstream target of MEX3A, which possesses similar regulatory effects on glioma development with MEX3A and could attenuate the promotion of glioma induced by MEX3A overexpression. Overall, MEX3A was identified as a potential tumor promoter in glioma development and therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Chao Yang
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Haoqiang Zhan
- Department of Neurosurgery, The Six Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yiqing Zhao
- Department of Neurosurgery, TongJi hospital of TongJi Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
| | - Yasong Wu
- Department of Neurosurgery, TongJi hospital of TongJi Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130012, China
| | - Heping Wang
- Department of Neurosurgery, TongJi hospital of TongJi Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China.
| |
Collapse
|
13
|
Wang H, Tao Z, Feng M, Li X, Deng Z, Zhao G, Yin H, Pan T, Chen G, Feng Z, Li Y, Zhou Y. Dual PLK1 and STAT3 inhibition promotes glioblastoma cells apoptosis through MYC. Biochem Biophys Res Commun 2020; 533:368-375. [PMID: 32962858 DOI: 10.1016/j.bbrc.2020.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 09/05/2020] [Indexed: 12/27/2022]
Abstract
Glioblastoma (GBM) is the deadliest primary brain tumor that is highly resistant to current treatments. Polo-like kinase 1 (PLK1) and signal transducer and activator of transcription 3 (STAT3) are highly expressed in gliomas, especially GBM. Previous studies have shown reciprocal activation between PLK1 and STAT3 and that they regulate the same pools of MYC downstream. We have demonstrated that PLK1 and STAT3 levels are elevated in gliomas compared with those in normal brain tissues, and high expression of both PLK1 and STAT3 is associated with poor prognosis in TCGA. Moreover, there was direct or indirect reciprocal regulation between PLK1 and STAT3. Furthermore, we found that PLK1 and STAT3 can regulate the same pools of MYC downstream. Compared to monotherapy, combined treatment of glioma cells with PLK1 and STAT3 inhibitors, BI2536 and Stattic, respectively, showed lower expression of MYC, synergistic induction of cell invasion and apoptosis in vitro, and tumor inhibition in xenografts. PLK1 and STAT3 were able to directly regulate the expression of MYC and induce apoptosis of glioma cells through the regulation of MYC. These findings may help develop a therapeutic strategy for dual inhibition of PLK1 and STAT3 against the tumorigenesis of glioma.
Collapse
Affiliation(s)
- Hao Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhennan Tao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ming Feng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuetao Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhitong Deng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guozheng Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haoran Yin
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tingzheng Pan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guangliang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zibin Feng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanyan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Youxin Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
Fathi Kazerooni A, Akbari H, Shukla G, Badve C, Rudie JD, Sako C, Rathore S, Bakas S, Pati S, Singh A, Bergman M, Ha SM, Kontos D, Nasrallah M, Bagley SJ, Lustig RA, O'Rourke DM, Sloan AE, Barnholtz-Sloan JS, Mohan S, Bilello M, Davatzikos C. Cancer Imaging Phenomics via CaPTk: Multi-Institutional Prediction of Progression-Free Survival and Pattern of Recurrence in Glioblastoma. JCO Clin Cancer Inform 2020; 4:234-244. [PMID: 32191542 PMCID: PMC7113126 DOI: 10.1200/cci.19.00121] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
PURPOSE To construct a multi-institutional radiomic model that supports upfront prediction of progression-free survival (PFS) and recurrence pattern (RP) in patients diagnosed with glioblastoma multiforme (GBM) at the time of initial diagnosis. PATIENTS AND METHODS We retrospectively identified data for patients with newly diagnosed GBM from two institutions (institution 1, n = 65; institution 2, n = 15) who underwent gross total resection followed by standard adjuvant chemoradiation therapy, with pathologically confirmed recurrence, sufficient follow-up magnetic resonance imaging (MRI) scans to reliably determine PFS, and available presurgical multiparametric MRI (MP-MRI). The advanced software suite Cancer Imaging Phenomics Toolkit (CaPTk) was leveraged to analyze standard clinical brain MP-MRI scans. A rich set of imaging features was extracted from the MP-MRI scans acquired before the initial resection and was integrated into two distinct imaging signatures for predicting mean shorter or longer PFS and near or distant RP. The predictive signatures for PFS and RP were evaluated on the basis of different classification schemes: single-institutional analysis, multi-institutional analysis with random partitioning of the data into discovery and replication cohorts, and multi-institutional assessment with data from institution 1 as the discovery cohort and data from institution 2 as the replication cohort. RESULTS These predictors achieved cross-validated classification performance (ie, area under the receiver operating characteristic curve) of 0.88 (single-institution analysis) and 0.82 to 0.83 (multi-institution analysis) for prediction of PFS and 0.88 (single-institution analysis) and 0.56 to 0.71 (multi-institution analysis) for prediction of RP. CONCLUSION Imaging signatures of presurgical MP-MRI scans reveal relatively high predictability of time and location of GBM recurrence, subject to the patients receiving standard first-line chemoradiation therapy. Through its graphical user interface, CaPTk offers easy accessibility to advanced computational algorithms for deriving imaging signatures predictive of clinical outcome and could similarly be used for a variety of radiomic and radiogenomic analyses.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hamed Akbari
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gaurav Shukla
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiation Oncology, Christiana Care Helen F. Graham Cancer Center and Research Institute, Newark, DE.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Chaitra Badve
- Department of Radiology, University Hospitals-Seidman Cancer Center, Cleveland, OH.,Case Comprehensive Cancer Center, Cleveland, OH
| | - Jeffrey D Rudie
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA
| | - Chiharu Sako
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Saima Rathore
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sarthak Pati
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ashish Singh
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mark Bergman
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sung Min Ha
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Despina Kontos
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - MacLean Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephen J Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Robert A Lustig
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Donald M O'Rourke
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Andrew E Sloan
- Case Western Reserve University School of Medicine, Cleveland, OH.,Case Comprehensive Cancer Center, Cleveland, OH.,Department of Neurologic Surgery, University Hospitals-Seidman Cancer Center, Cleveland, OH
| | - Jill S Barnholtz-Sloan
- Case Western Reserve University School of Medicine, Cleveland, OH.,Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michel Bilello
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
15
|
Chavda V, Patel V, Yadav D, Shah J, Patel S, Jin JO. Therapeutics and Research Related to Glioblastoma: Advancements and Future Targets. Curr Drug Metab 2020; 21:186-198. [DOI: 10.2174/1389200221666200408083950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 11/28/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022]
Abstract
Glioblastoma, the most common primary brain tumor, has been recognized as one of the most lethal and
fatal human tumors. It has a dismal prognosis, and survival after diagnosis is less than 15 months. Surgery and radiotherapy
are the only available treatment options at present. However, numerous approaches have been made to upgrade
in vivo and in vitro models with the primary goal of assessing abnormal molecular pathways that would be
suitable targets for novel therapeutic approaches. Novel drugs, delivery systems, and immunotherapy strategies to
establish new multimodal therapies that target the molecular pathways involved in tumor initiation and progression in
glioblastoma are being studied. The goal of this review was to describe the pathophysiology, neurodegeneration
mechanisms, signaling pathways, and future therapeutic targets associated with glioblastomas. The key features have
been detailed to provide an up-to-date summary of the advancement required in current diagnosis and therapeutics
for glioblastoma. The role of nanoparticulate system graphene quantum dots as suitable therapy for glioblastoma has
also been discussed.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| | - Jigar Shah
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Snehal Patel
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| |
Collapse
|
16
|
Sugier D, Sugier P, Jakubowicz-Gil J, Winiarczyk K, Kowalski R. Essential Oil from Arnica Montana L. Achenes: Chemical Characteristics and Anticancer Activity. Molecules 2019; 24:molecules24224158. [PMID: 31744121 PMCID: PMC6891426 DOI: 10.3390/molecules24224158] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/03/2019] [Accepted: 11/14/2019] [Indexed: 12/24/2022] Open
Abstract
Mountain arnica Arnica montana L. is a source of several metabolite classes with diverse biological activities. The chemical composition of essential oil and its major volatile components in arnica may vary depending on the geographical region, environmental factors, and plant organ. The objective of this study was to characterize the chemical composition of essential oil derived from A. montana achenes and to investigate its effect on induction of apoptosis and autophagy in human anaplastic astrocytoma MOGGCCM and glioblastoma multiforme T98G cell lines. The chemical composition of essential oil extracted from the achenes was examined with the use of Gas Chromatography–Mass Spectrometry GC-MS. Only 16 components of the essential oil obtained from the achenes of 3-year-old plants and 18 components in the essential oil obtained from the achenes of 4-year-old plants constituted ca. 94.14% and 96.38% of the total EO content, respectively. The main components in the EO from the arnica achenes were 2,5-dimethoxy-p-cymene (39.54 and 44.65%), cumene (13.24 and 10.71%), thymol methyl ether (8.66 and 8.63%), 2,6-diisopropylanisole (8.55 and 8.41%), decanal (7.31 and 6.28%), and 1,2,2,3-tetramethylcyclopent-3-enol (4.33 and 2.94%) in the 3- and 4-year-old plants, respectively. The essential oils were found to exert an anticancer effect by induction of cell death in anaplastic astrocytoma and glioblastoma multiforme cells. The induction of apoptosis at a level of 25.7–32.7% facilitates the use of this secondary metabolite in further studies focused on the development of glioma therapy in the future. Probably, this component plays a key role in the anticancer activity against the MOGGCCM and T98G cell lines. The present study is the first report on the composition and anticancer activities of essential oil from A. montana achenes, and further studies are required to explore its potential for future medicinal purposes.
Collapse
Affiliation(s)
- Danuta Sugier
- Department of Industrial and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - Piotr Sugier
- Department of Botany, Mycology and Ecology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 19 Akademicka Street, 20-033 Lublin, Poland
- Correspondence: ; Tel.: +48-81-537-59-46
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 19 Akademicka Street, 20-033 Lublin, Poland;
| | - Krystyna Winiarczyk
- Department of Cell Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, 19 Akademicka Street, 20-033 Lublin, Poland;
| | - Radosław Kowalski
- Department of Analysis and Evaluation of Food Quality, University of Life Sciences in Lublin, 8 Skromna Street, 20-704 Lublin, Poland;
| |
Collapse
|
17
|
Fathi Kazerooni A, Bakas S, Saligheh Rad H, Davatzikos C. Imaging signatures of glioblastoma molecular characteristics: A radiogenomics review. J Magn Reson Imaging 2019; 52:54-69. [PMID: 31456318 DOI: 10.1002/jmri.26907] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, the advent and development of genomic assessment methods and computational approaches have raised the hopes for identifying therapeutic targets that may aid in the treatment of glioblastoma. However, the targeted therapies have barely been successful in their effort to cure glioblastoma patients, leaving them with a grim prognosis. Glioblastoma exhibits high heterogeneity, both spatially and temporally. The existence of different genetic subpopulations in glioblastoma allows this tumor to adapt itself to environmental forces. Therefore, patients with glioblastoma respond poorly to the prescribed therapies, as treatments are directed towards the whole tumor and not to the specific genetic subregions. Genomic alterations within the tumor develop distinct radiographic phenotypes. In this regard, MRI plays a key role in characterizing molecular signatures of glioblastoma, based on regional variations and phenotypic presentation of the tumor. Radiogenomics has emerged as a (relatively) new field of research to explore the connections between genetic alterations and imaging features. Radiogenomics offers numerous advantages, including noninvasive and global assessment of the tumor and its response to therapies. In this review, we summarize the potential role of radiogenomic techniques to stratify patients according to their specific tumor characteristics with the goal of designing patient-specific therapies. Level of Evidence: 5 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2020;52:54-69.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hamidreza Saligheh Rad
- Quantitative MR Imaging and Spectroscopy Group (QMISG), Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Lee YK, Lee KW, Kim M, Lee Y, Yoo J, Hwangbo C, Park KH, Kim KD. Chelidonine Induces Caspase-Dependent and Caspase-Independent Cell Death through G 2/M Arrest in the T98G Human Glioblastoma Cell Line. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:6318179. [PMID: 31239863 PMCID: PMC6556348 DOI: 10.1155/2019/6318179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/07/2019] [Indexed: 11/18/2022]
Abstract
Chelidonium majus L. (family Papaveraceae), commonly known as greater celandine or tetterwort, has been reported to have antibacterial and anticancer effects and chelidonine is known as a functional metabolite extracted from C.
Collapse
Affiliation(s)
- Yeon-Kyeong Lee
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Ki Won Lee
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Minju Kim
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Yerin Lee
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Jiyun Yoo
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
- Division of Life Science, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Cheol Hwangbo
- Division of Life Science, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Ki Hun Park
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Kwang Dong Kim
- Division of Applied Life Science (BK21Plus), Gyeongsang National University, Jinju 660-701, Republic of Korea
- Division of Life Science, Gyeongsang National University, Jinju 660-701, Republic of Korea
- PMBBRC, Gyeongsang National University, Jinju 660-701, Republic of Korea
| |
Collapse
|
19
|
Kurihara R, Horibe T, Shimizu E, Torisawa A, Gaowa A, Kohno M, Kawakami K. A novel interleukin‐13 receptor alpha 2‐targeted hybrid peptide for effective glioblastoma therapy. Chem Biol Drug Des 2019; 94:1402-1413. [DOI: 10.1111/cbdd.13517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/20/2019] [Accepted: 03/03/2019] [Indexed: 01/22/2023]
Affiliation(s)
- Ryohsuke Kurihara
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| | - Tomohisa Horibe
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| | - Eiko Shimizu
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| | - Aya Torisawa
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| | - Arong Gaowa
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| | - Masayuki Kohno
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| | - Koji Kawakami
- Department of Pharmacoepidemiology Graduate School of Medicine and Public Health Kyoto University Kyoto Japan
| |
Collapse
|
20
|
Combined Treatment with Doxorubicin and Rapamycin Is Effective against In Vitro and In Vivo Models of Human Glioblastoma. J Clin Med 2019; 8:jcm8030331. [PMID: 30857276 PMCID: PMC6462908 DOI: 10.3390/jcm8030331] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 11/17/2022] Open
Abstract
Despite numerous clinical trials, glioblastoma (GBM) remains a tumor that is difficult to treat. The aim of this study was to investigate the potential of a new pharmacological approach, combining doxorubicin (Dox) and rapamycin (Rapa), in in vitro and in vivo GBM models. Cytotoxic and anti-proliferative effects of Rapa plus Dox treatments were analyzed in GBM cell lines. The in vivo effectiveness of these treatments was investigated in an orthotopic xenograft mice model of GBM. In vitro results demonstrated that prolonged exposure to Rapa sensitize GBM cells to Dox treatments. In vivo results demonstrated that Rapa (5 mg/kg) plus Dox (5 mg/kg) determined the major tumor growth inhibition (-97.29% vs. control) but results in greater toxicity. The combination Rapa plus Dox (2.5 mg/kg) showed a tumor inhibition like Rapa plus Dox (5 mg/kg) with a toxicity comparable to Rapa alone. Thus, this study demonstrated the efficacy of this pharmacological approach, providing the rationale for a clinical application of this combinational therapy in "poor-responder" GBM patients.
Collapse
|
21
|
Cha J, Kim P. Time series assessment of the effects of hypoxic stress on glioma tumorsphere development within engineered microscale niches. Biomaterials 2018; 194:171-182. [PMID: 30605825 DOI: 10.1016/j.biomaterials.2018.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023]
Abstract
A rapidly outgrowing tumor mass is liable to suffer from a shortage of oxygen and nutrients due to the diffusion limit. These features evidently prevail in glioblastoma, resulting in extensive hypoxic regions throughout the tumor mass. While there may be a strong link between hypoxic regions and glioblastoma malignancy, the effects of hypoxia stress on each tumorigenic step and their interrelation during the progression remain unexplored due to the limited information from current assay platforms. Here, we suggest a tumor microenvironment array platform (TMAP) to describe a time series assessment of glioblastoma tumorsphere (TS) within a microscale niche. TMAP enables to observe the overall tumorigenic process of glioblastoma TSs over the cultivation time, simultaneously quantifying the features with the biophysical parameters. Through the time series assessment, we observed the induction of hypoxic stress within the mature TSs, which rendered intratumoral phenotypic changes to become more malignant and modulated their microenvironmental niches to enhance angiogenic proliferation, immune recruitment, and even drug response. Based on the finding that the tumorigenic parameters were highly correlated only in mature TSs, we conclude that the effects of hypoxic stress systematically affect the process that drives a glioblastoma to higher malignancy.
Collapse
Affiliation(s)
- Junghwa Cha
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea; KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea; KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
22
|
Da Ros M, Iorio AL, De Gregorio V, Fantappiè O, Laffi G, de Martino M, Pisano C, Genitori L, Sardi I. Aldoxorubicin and Temozolomide combination in a xenograft mice model of human glioblastoma. Oncotarget 2018; 9:34935-34944. [PMID: 30405885 PMCID: PMC6201851 DOI: 10.18632/oncotarget.26183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/15/2018] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is still an incurable disease. The front-line Temozolomide (TMZ)-based therapy suffers from poor efficacy, underlining the need of new therapies. Preclinically, Aldoxorubicin (Aldox), a novel prodrug of Doxorubicin (Dox), has been successfully tested against GBM, encouraging the study of its association with other agents. For the first time, we evaluated the effectiveness of Aldox combined to TMZ in preclinical models of GBM. Our in vitro results demonstrated that the anti–glioma effect of Aldox was more marked than TMZ and their combination increased the killing effect of the anthracycline in TMZ-resistant GBM cells. Moreover, unlike Dox, Aldox was able to accumulate in P-glycoprotein (P-gp)-overexpressed cells due to a negative regulation of the P-gp function. We also compared efficacy and safety of weekly administrations of Aldox (16 mg/kg), with or without TMZ (0.9 mg/kg, daily injections), in the U87 xenograft mouse model. Aldox therapy induced a moderate tumor volume inhibition (TVI) and an increased survival rate (+12.5% vs vehicle). On the other hand, when combined to TMZ, Aldox caused a significant TVI (P=0.0175 vs vehicle) and delayed the mortality during the experimental period, although TVI and endpoint survival percentage (+37.5% vs vehicle) were not significantly different from TMZ alone. Our preliminary data showed that Aldox exerts anti–glioma effects in vitro and in vivo. It also enhances its antitumor activity when combined with TMZ, resulting in a superior efficacy compared to the single agents, without adverse side effects.
Collapse
Affiliation(s)
- Martina Da Ros
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Anna Lisa Iorio
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Veronica De Gregorio
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Ornella Fantappiè
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giacomo Laffi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maurizio de Martino
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | | | - Lorenzo Genitori
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Department of Pediatric Oncology, Meyer Children's Hospital, Florence, Italy
| |
Collapse
|
23
|
Xu J, Fang J, Cheng Z, Fan L, Hu W, Zhou F, Shen H. Overexpression of the Kininogen-1 inhibits proliferation and induces apoptosis of glioma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:180. [PMID: 30068373 PMCID: PMC6090912 DOI: 10.1186/s13046-018-0833-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022]
Abstract
Background Glioma is the most common primary central nervous system tumor derived from glial cells. Kininogen-1 (KNG1) can exert antiangiogenic properties and inhibit proliferation of endothelial cells. The effect of KNG1 on the glioma is rarely reported, so our purpose in to explore its mechanism in glioma cells. Methods The differentially expressed genes (DEGs) were identified based on The Cancer Genome Atlas (TCGA) database. The KNG1-vector was transfected into the two glioma cells. The viability, apoptosis and cell cycle of glioma cells and microvessel density (MVD) were detected by cell counting kit-8 assay, flow cytometry and immunohistochemistry, respectively. The expression were measured by quantitative real-time PCR and Western blot, respectively. A tumor mouse model was established to determine apoptosis rate of brain tissue by terminal deoxynucleotidyl transfer-mediated dUTP nick end labeling (TUNEL) analysis. Results KNG1 was identified as the core gene and lowly expressed in the glioma cells. Overexpression of KNG1 inhibited cell viability and angiogenesis of glioma cells. Overexpression of KNG1 promoted the apoptosis and G1 phase cell cycle arrest of glioma cells. Moreover, the expressions of VEGF, cyclinD1, ki67, caspase-3/9 and XIAP were regulated by overexpression of KNG1. In addition, overexpression of KNG1 inhibited the activity of PI3K/Akt. Furthermore, overexpression of KNG1 decreased the tumor growth and promoted the apoptosis of decreased by overexpression of KNG1 in vivo. . Conclusions Overexpression of KNG1 suppresses glioma progression by inhibiting the proliferation and promoting apoptosis of glioma cells, providing a therapeutic strategy for the malignant glioma.
Collapse
Affiliation(s)
- Jinfang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Jun Fang
- Department of Radiotherapy, Zhejiang Cancer Hospital, No.1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang Province, 310022, China
| | - Zhonghao Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Longlong Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Weiwei Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| | - Feng Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China.
| | - Hong Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, Zhejiang Province, 310009, China
| |
Collapse
|
24
|
Kumar M, Sharma G, Kumar R, Singh B, Katare OP, Raza K. Lysine-Based C60-Fullerene Nanoconjugates for Monomethyl Fumarate Delivery: A Novel Nanomedicine for Brain Cancer Cells. ACS Biomater Sci Eng 2018; 4:2134-2142. [DOI: 10.1021/acsbiomaterials.7b01031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Manish Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, NH-8, District Ajmer, Rajasthan 305 817, India
| | - Gajanand Sharma
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Sector 14, Chandigarh 160 014, India
| | - Rajendra Kumar
- UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles and Nanocomposites, Panjab University, Sector 14, Chandigarh 160 014, India
| | - Bhupinder Singh
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Sector 14, Chandigarh 160 014, India
- UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles and Nanocomposites, Panjab University, Sector 14, Chandigarh 160 014, India
| | - Om Prakash Katare
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Sector 14, Chandigarh 160 014, India
| | - Kaisar Raza
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, NH-8, District Ajmer, Rajasthan 305 817, India
| |
Collapse
|
25
|
Gaetani P, Hulleman E, Levi D, Quarto M, Scorsetti M, Helin K, Simonelli M, Colombo P, Baena RRY. Expression of the Transcription Factor HEY1 in Glioblastoma: A Preliminary Clinical Study. TUMORI JOURNAL 2018; 96:97-102. [DOI: 10.1177/030089161009600116] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aims and background The hairy/enhancer of split (E(spl))-related family of transcription factors (HES and HEY) are established targets of the notch signaling pathway, which has been implicated in different developmental processes, tumor formation and the self-renewal of neural stem cells. We determined the expression of HEY1 in human malignant gliomas to investigate whether its expression might be related to prognosis. Methods The expression of HEY1 was studied by in situ hybridization on 62 cases of glioblastoma. Patients were treated with surgery followed by chemotherapy and radiotherapy. We considered as end points of the study the overall survival time and progression-free interval. Correlations between HEY1 expression and tumor grade/patient overall survival and free interval before recurrence were analyzed using univariate analysis. Results Based on the in situ hybridization results, HEY1 expression rate was reported as negative staining in 13 cases (20.6%), as weak staining in 11 cases (17.3%), as moderate staining in 21 cases (33.3%), and as strong staining in 17 cases. We considered in the analysis the cumulative expression of HEY1 at in situ hybridization (Hey Index) as negative in 13 cases and positive in 49 cases (77.78%). The overall survival (P = 0.002) and the free-interval (P = 0.012) were significantly longer in patients who were negative for HEY1 expression. Conclusions Our data suggest that expression of HEY1 might be used as a marker to distinguish glioblastoma patients with a relatively good prognosis from those at high-risk, and that, in the future, HEY1 might represent a therapeutic target.
Collapse
Affiliation(s)
- Paolo Gaetani
- Department of Neurosurgery, IRCCS Istituto Clinico Humanitas, Rozzano (MI), Italy
| | | | - Daniel Levi
- Department of Neurosurgery, IRCCS Istituto Clinico Humanitas, Rozzano (MI), Italy
| | | | - Marta Scorsetti
- Department of Radiotherapy, IRCCS Istituto Clinico Humanitas, Rozzano (MI), Italy
| | - Kristian Helin
- Biotech Research and Innovation Centre and Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Matteo Simonelli
- Department of Oncology, IRCCS Istituto Clinico Humanitas, Rozzano (MI), Italy
| | | | | |
Collapse
|
26
|
Fann LY, Chen Y, Chu DC, Weng SJ, Chu HC, Wu ATH, Lee JF, Ali AAA, Chen TC, Huang HS, Ma KH. Identification and preclinical evaluation of the small molecule, NSC745887, for treating glioblastomas via suppressing DcR3-associated signaling pathways. Oncotarget 2017; 9:11922-11937. [PMID: 29552282 PMCID: PMC5844718 DOI: 10.18632/oncotarget.23714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/11/2017] [Indexed: 11/25/2022] Open
Abstract
The small-molecule naphtha [2,3-f]quinoxaline-7,12-dione (NSC745887) can effectively inhibit the proliferation of various cancers by trapping DNA-topoisomerase cleavage. The aim of this study was to elucidate cellular responses of NSC745887 in human glioblastoma multiforme (GBM, U118MG and U87MG cells) and investigate the underlying molecular mechanisms. NSC745887 reduced the cell survival rate and increased the sub-G1 population in dose- and time-dependent manners in GBM cells. Moreover, NSC745887 increased expression of γH2AX and caused DNA fragmentation leading to DNA damage. Furthermore, Annexin V/propidium iodide and Br-dTP staining showed the apoptotic effect of NSC745887 in GBM cells. DNA repair proteins of ataxia-telangiectasia mutated (ATM), ATM and Rad3-related, and decoy receptor 3 also decreased with NSC745887 treatment. In addition, NSC745887 caused apoptosis by the caspase-8/9-caspase-3-poly(ADP-ribose) polymerase cascade. An in vivo study indicated that NSC745887 suppressed the [18F]-FDG-specific uptake value in brain tumors. Histological staining also indicated a decrease in Ki-67 and increases in γH2AX and cleaved caspase-3 in the brain tumor area. These data provide preclinical evidence for NSC745887 as a potential new small molecule drug for managing glioblastomas.
Collapse
Affiliation(s)
- Li-Yun Fann
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Nursing and Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan, ROC.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ying Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Da-Chen Chu
- Department of Nursing and Department of Neurosurgery, Taipei City Hospital, Taipei, Taiwan, ROC
| | - Shao-Ju Weng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Heng-Cheng Chu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Alexander T H Wu
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Jiann-Fong Lee
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ahmed Atef Ahmed Ali
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsung-Chih Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsu-Shan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC.,Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Kuo-Hsing Ma
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
27
|
Qu S, Wang T, Huang J. Presence of the minor allele of microRNA205 rs3842530 polymorphism increases 18FDG uptake in patients with breast cancer via targeting VEGF. Mol Med Rep 2017; 17:636-642. [PMID: 29115451 DOI: 10.3892/mmr.2017.7914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 12/12/2016] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are regarded as key regulators of gene expression involved in the pathogenesis of various diseases. Numerous single nucleotide polymorphisms (SNPs) in miRNA genes have been found to be associated with human diseases by affecting the processing process of miRNAs. In the present study, patients with breast cancer underwent a PET scan, and the maximum standard uptake value (SUVmax)/partial volume‑corrected standard uptake value (SUVpvc) were determined in each individual. The samples were collected and genotyped for rs3842530. Statistical analysis was performed to evaluate the difference between the genotype groups. The results demonstrated that miR‑205 downregulated the expression of vascular endothelial growth factor (VEGF) by binding to its 3'untranslated region. The introduction of exogenous miRNA, which mimicked miR‑205, decreased the protein and mRNA expression levels of VEGF and, consistently, the suppression of endogenous miR‑205 resulted in an increase in the expression levels of VEGF. Furthermore, it was found that the expression of mature miR‑205 was markedly reduced by the presence of rs3842530. 18F‑fluorodeoxyglucose (18FDG) metabolism, including SUVmax and SUVpvc, are important parameters of PET, and dysregulation of the expression of VEGF has been reported to be associated with an altered 18FDG metabolism. In the present study, it was found that the presence of minor allele rs3842530 was correlated with increased SUVmax and SUVpvc, which may have been mediated by release of the physiologically inhibited expression of VEGF. Therefore, VEGF was a direct target of miR‑205, and the presence of rs3842530 compromised the expression of miR‑205, suggesting it is a promising biomarker for the metabolism of 18FDG.
Collapse
Affiliation(s)
- Shiying Qu
- Department of Nuclear Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Tie Wang
- Department of Nuclear Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Jingwei Huang
- Department of Nuclear Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
28
|
Romano-Feinholz S, Salazar-Ramiro A, Muñoz-Sandoval E, Magaña-Maldonado R, Hernández Pedro N, Rangel López E, González Aguilar A, Sánchez García A, Sotelo J, Pérez de la Cruz V, Pineda B. Cytotoxicity induced by carbon nanotubes in experimental malignant glioma. Int J Nanomedicine 2017; 12:6005-6026. [PMID: 28860763 PMCID: PMC5573058 DOI: 10.2147/ijn.s139004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Despite multiple advances in the diagnosis of brain tumors, there is no effective treatment for glioblastoma. Multiwalled carbon nanotubes (MWCNTs), which were previously used as a diagnostic and drug delivery tool, have now been explored as a possible therapy against neoplasms. However, although the toxicity profile of nanotubes is dependent on the physicochemical characteristics of specific particles, there are no studies exploring how the effectivity of the carbon nanotubes (CNTs) is affected by different methods of production. In this study, we characterize the structure and biocompatibility of four different types of MWCNTs in rat astrocytes and in RG2 glioma cells as well as the induction of cell lysis and possible additive effect of the combination of MWCNTs with temozolomide. We used undoped MWCNTs (labeled simply as MWCNTs) and nitrogen-doped MWCNTs (labeled as N-MWCNTs). The average diameter of both pristine MWCNTs and pristine N-MWCNTs was ~22 and ~35 nm, respectively. In vitro and in vivo results suggested that these CNTs can be used as adjuvant therapy along with the standard treatment to increase the survival of rats implanted with malignant glioma.
Collapse
Affiliation(s)
| | - Alelí Salazar-Ramiro
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery (NINN), Mexico City
| | | | - Roxana Magaña-Maldonado
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery (NINN), Mexico City
| | | | | | | | | | - Julio Sotelo
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery (NINN), Mexico City
| | | | - Benjamín Pineda
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery (NINN), Mexico City
| |
Collapse
|
29
|
Song G, Luo T, Dong L, Liu Q. Extracellular diffusion quantified by magnetic resonance imaging during rat C6 glioma cell progression. ACTA ACUST UNITED AC 2017; 50:e5403. [PMID: 28678913 PMCID: PMC5496150 DOI: 10.1590/1414-431x20175403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 05/02/2017] [Indexed: 12/31/2022]
Abstract
Solution reflux and edema hamper the convection-enhanced delivery of the standard treatment for glioma. Therefore, a real-time magnetic resonance imaging (MRI) method was developed to monitor the dosing process, but a quantitative analysis of local diffusion and clearance parameters has not been assessed. The objective of this study was to compare diffusion into the extracellular space (ECS) at different stages of rat C6 gliomas, and analyze the effects of the extracellular matrix (ECM) on the diffusion process. At 10 and 20 days, after successful glioma modeling, gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA) was introduced into the ECS of rat C6 gliomas. Diffusion parameters and half-life of the reagent were then detected using MRI, and quantified according to the mathematical model of diffusion. The main ECM components [chondroitin sulfate proteoglycans (CSPGs), collagen IV, and tenascin C] were detected by immunohistochemical and immunoblot analyses. In 20-day gliomas, Gd-DTPA diffused more slowly and derived higher tortuosity, with lower clearance rate and longer half-life compared to 10-day gliomas. The increased glioma ECM was associated with different diffusion and clearance parameters in 20-day rat gliomas compared to 10-day gliomas. ECS parameters were altered with C6 glioma progression from increased ECM content. Our study might help better understand the glioma microenvironment and provide benefits for interstitial drug delivery to treat brain gliomas.
Collapse
Affiliation(s)
- G Song
- Department of Radiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - T Luo
- Department of Radiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - L Dong
- Department of Radiology, The Secondary Affiliated Hospital, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Q Liu
- Department of Radiology, The Secondary Affiliated Hospital, Baotou Medical College, Baotou, Inner Mongolia, China
| |
Collapse
|
30
|
Wang W, Cho HY, Rosenstein-Sisson R, Marín Ramos NI, Price R, Hurth K, Schönthal AH, Hofman FM, Chen TC. Intratumoral delivery of bortezomib: impact on survival in an intracranial glioma tumor model. J Neurosurg 2017; 128:695-700. [PMID: 28409734 DOI: 10.3171/2016.11.jns161212] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Glioblastoma (GBM) is the most prevalent and the most aggressive of primary brain tumors. There is currently no effective treatment for this tumor. The proteasome inhibitor bortezomib is effective for a variety of tumors, but not for GBM. The authors' goal was to demonstrate that bortezomib can be effective in the orthotopic GBM murine model if the appropriate method of drug delivery is used. In this study the Alzet mini-osmotic pump was used to bring the drug directly to the tumor in the brain, circumventing the blood-brain barrier; thus making bortezomib an effective treatment for GBM. METHODS The 2 human glioma cell lines, U87 and U251, were labeled with luciferase and used in the subcutaneous and intracranial in vivo tumor models. Glioma cells were implanted subcutaneously into the right flank, or intracranially into the frontal cortex of athymic nude mice. Mice bearing intracranial glioma tumors were implanted with an Alzet mini-osmotic pump containing different doses of bortezomib. The Alzet pumps were introduced directly into the tumor bed in the brain. Survival was documented for mice with intracranial tumors. RESULTS Glioma cells were sensitive to bortezomib at nanomolar quantities in vitro. In the subcutaneous in vivo xenograft tumor model, bortezomib given intravenously was effective in reducing tumor progression. However, in the intracranial glioma model, bortezomib given systemically did not affect survival. By sharp contrast, animals treated with bortezomib intracranially at the tumor site exhibited significantly increased survival. CONCLUSIONS Bypassing the blood-brain barrier by using the osmotic pump resulted in an increase in the efficacy of bortezomib for the treatment of intracranial tumors. Thus, the intratumoral administration of bortezomib into the cranial cavity is an effective approach for glioma therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Axel H Schönthal
- 3Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | | |
Collapse
|
31
|
Pereira MSL, Klamt F, Thomé CC, Worm PV, de Oliveira DL. Metabotropic glutamate receptors as a new therapeutic target for malignant gliomas. Oncotarget 2017; 8:22279-22298. [PMID: 28212543 PMCID: PMC5400663 DOI: 10.18632/oncotarget.15299] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022] Open
Abstract
Metabotropic glutamate receptors (mGluR) are predominantly involved in maintenance of cellular homeostasis of central nervous system. However, evidences have suggested other roles of mGluR in human tumors. Aberrant mGluR signaling has been shown to participate in transformation and maintenance of various cancer types, including malignant brain tumors. This review intends to summarize recent findings regarding the involvement of mGluR-mediated intracellular signaling pathways in progression, aggressiveness, and recurrence of malignant gliomas, mainly glioblastomas (GBM), highlighting the potential therapeutic applications of mGluR ligands. In addition to the growing number of studies reporting mGluR gene or protein expression in glioma samples (resections, lineages, and primary cultures), pharmacological blockade in vitro of mGluR1 and mGluR3 by selective ligands has been shown to be anti-proliferative and anti-migratory, decreasing activation of MAPK and PI3K pathways. In addition, mGluR3 antagonists promoted astroglial differentiation of GBM cells and also enabled cytotoxic action of temozolomide (TMZ). mGluR3-dependent TMZ toxicity was supported by increasing levels of MGMT transcripts through an intracellular signaling pathway that sequentially involves PI3K and NF-κB. Further, continuous pharmacological blockade of mGluR1 and mGluR3 have been shown to reduced growth of GBM tumor in two independent in vivo xenograft models. In parallel, low levels of mGluR3 mRNA in GBM resections may be a predictor for long survival rate of patients. Since several Phase I, II and III clinical trials are being performed using group I and II mGluR modulators, there is a strong scientifically-based rationale for testing mGluR antagonists as an adjuvant therapy for malignant brain tumors.
Collapse
Affiliation(s)
- Mery Stefani Leivas Pereira
- Department of Biochemistry, Laboratory of Cellular Neurochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre RS, Brazil
| | - Fábio Klamt
- Department of Biochemistry, Laboratory of Cellular Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre RS, Brazil
| | - Chairini Cássia Thomé
- Department of Biochemistry, Laboratory of Cellular Neurochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre RS, Brazil
| | - Paulo Valdeci Worm
- Department of Neurosurgery, Cristo Redentor Hospital – GHC – Porto Alegre RS, Brazil
- Department of Neurosurgery, São José Hospital, Complexo Hospitalar Santa Casa, Porto Alegre RS, Brazil
| | - Diogo Losch de Oliveira
- Department of Biochemistry, Laboratory of Cellular Neurochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre RS, Brazil
| |
Collapse
|
32
|
Jakubowicz-Gil J, Bądziul D, Langner E, Wertel I, Zając A, Rzeski W. Temozolomide and sorafenib as programmed cell death inducers of human glioma cells. Pharmacol Rep 2017; 69:779-787. [PMID: 28587939 DOI: 10.1016/j.pharep.2017.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 01/27/2017] [Accepted: 03/10/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Gliomas are aggressive brain tumors with very high resistance to chemotherapy. Therefore, the aim of the present study was to investigate the effectiveness of sorafenib and Temozolomide in elimination of human glioma cells through apoptosis and autophagy. METHODS MOGGCCM (anaplastic astrocytoma) and T98G (glioblastoma multiforme) cell lines incubated with sorafenib and/or Temozolomide were used in the experiments. Cell morphology (ER stress, apoptosis, autophagy, and necrosis) was analyzed microscopically while apoptosis and mitochondrial membrane potential were assessed with flow cytometry. Beclin1, LC3, p62, Hsp27, and Hsp72 levels were analyzed by immunoblotting. The activity of caspase 3, 8, and 9 was evaluated fluorometrically. Expression of Hsps was blocked by transfection with specific siRNA. RESULTS In MOGGCCM cells, Temozolomide most frequently induced autophagy, which was accompanied by decreased p62 and increased beclin1 and LC3II levels. Sorafenib initiated mainly apoptosis. Additional incubation with Temozolomide, synergistically potentiated the pro-apoptotic properties of sorafenib, but it was mediated in a caspase-independent way. In T98G cells, the effect of the analyzed drugs on programmed cell death induction was different from that in MOGGCCM cells. Sorafenib induced autophagy, while Temozolomide initiated mainly apoptosis. After simultaneous drug application, apoptosis dominated, suggesting synergistic action of both drugs. Inhibition of Hsp27 and Hsp72 expression increased the sensitivity of both cell lines to ER stress and, to a lesser extent, to induction of apoptosis, but not autophagy. CONCLUSIONS Sorafenib and Temozolomide applied in combination are potent apoptosis inducers in T98G and MOGGCCM cells. ER stress precedes the elimination. Blocking of Hsp expression has a greater impact on ER stress rather than apoptosis induction.
Collapse
Affiliation(s)
- Joanna Jakubowicz-Gil
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Lublin, Poland.
| | - Dorota Bądziul
- Department of Human Physiology, University of Rzeszow, Rzeszów, Poland.
| | - Ewa Langner
- Department of Medical Biology, Institute of Agricultural Medicine, Lublin, Poland.
| | - Iwona Wertel
- 1st Department of Gynecology, University School of Medicine, Lublin, Poland.
| | - Adrian Zając
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Lublin, Poland.
| | - Wojciech Rzeski
- Department of Medical Biology, Institute of Agricultural Medicine, Lublin, Poland; Department of Immunology and Virology, Maria Curie-Sklodowska University, Lublin, Poland.
| |
Collapse
|
33
|
Diagnostic and Therapeutic Biomarkers in Glioblastoma: Current Status and Future Perspectives. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8013575. [PMID: 28316990 PMCID: PMC5337853 DOI: 10.1155/2017/8013575] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/13/2016] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is a primary neuroepithelial tumor of the central nervous system, characterized by an extremely aggressive clinical phenotype. Patients with GBM have a poor prognosis and only 3–5% of them survive for more than 5 years. The current GBM treatment standards include maximal resection followed by radiotherapy with concomitant and adjuvant therapies. Despite these aggressive therapeutic regimens, the majority of patients suffer recurrence due to molecular heterogeneity of GBM. Consequently, a number of potential diagnostic, prognostic, and predictive biomarkers have been investigated. Some of them, such as IDH mutations, 1p19q deletion, MGMT promoter methylation, and EGFRvIII amplification are frequently tested in routine clinical practice. With the development of sequencing technology, detailed characterization of GBM molecular signatures has facilitated a more personalized therapeutic approach and contributed to the development of a new generation of anti-GBM therapies such as molecular inhibitors targeting growth factor receptors, vaccines, antibody-based drug conjugates, and more recently inhibitors blocking the immune checkpoints. In this article, we review the exciting progress towards elucidating the potential of current and novel GBM biomarkers and discuss their implications for clinical practice.
Collapse
|
34
|
Bayat N, Ebrahimi-Barough S, Norouzi-Javidan A, Saberi H, Tajerian R, Ardakan MMM, Shirian S, Ai A, Ai J. Apoptotic effect of atorvastatin in glioblastoma spheroids tumor cultured in fibrin gel. Biomed Pharmacother 2016; 84:1959-1966. [DOI: 10.1016/j.biopha.2016.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 10/29/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022] Open
|
35
|
Ramachandran C, Portalatin GM, Prado AM, Quirin KW, Escalon E, Melnick SJ. In Vivo Antitumor Effect of Supercritical CO 2 Extract of Mango Ginger ( Curcuma amada Roxb) in U-87MG Human Glioblastoma Nude Mice Xenografts. J Evid Based Complementary Altern Med 2016; 22:260-267. [PMID: 27436761 DOI: 10.1177/2156587216659390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one the most aggressive and lethal human neoplasms with poor prognosis and very limited positive treatment options. The antitumor effect of supercritical CO2 extract of mango ginger ( Curcuma amada Roxb) (CA) with and without irinotecan (IR) was analyzed in U-87MG human glioblastoma multiforme (GBM) cells in vitro and in nude mice xenografts. CA is highly cytotoxic to GBM cells and is synergistic with IR as indicated by the combination index values of <1 in the CompuSyn analysis. CA inhibits tumor growth rate in GBM xenografts, the inhibition rate being higher than in IR treated group. GBM xenograft mice treated with IR + CA combination showed almost complete inhibition of tumor growth rate. Gene expression analysis of xenograft tumors indicated that IR + CA treatment significantly downregulated anti-apoptotic (Bcl-2 and mutant p53), inflammation-associated (COX-2) and cell division-associated (CCNB2) genes and upregulated pro-apoptotic genes (p21 and caspase-3). These results confirmed the therapeutic efficiency of IR + CA combination against GBM and the need for further clinical investigations.
Collapse
Affiliation(s)
- Cheppail Ramachandran
- 1 Nicklaus Children's Hospital, Miami, FL, USA.,2 Dharma Biomedical LLC, Miami, FL, USA
| | | | | | | | | | - Steven J Melnick
- 1 Nicklaus Children's Hospital, Miami, FL, USA.,2 Dharma Biomedical LLC, Miami, FL, USA
| |
Collapse
|
36
|
Correa BR, de Araujo PR, Qiao M, Burns SC, Chen C, Schlegel R, Agarwal S, Galante PAF, Penalva LOF. Functional genomics analyses of RNA-binding proteins reveal the splicing regulator SNRPB as an oncogenic candidate in glioblastoma. Genome Biol 2016; 17:125. [PMID: 27287018 PMCID: PMC4901439 DOI: 10.1186/s13059-016-0990-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/24/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and aggressive type of brain tumor. Currently, GBM has an extremely poor outcome and there is no effective treatment. In this context, genomic and transcriptomic analyses have become important tools to identify new avenues for therapies. RNA-binding proteins (RBPs) are master regulators of co- and post-transcriptional events; however, their role in GBM remains poorly understood. To further our knowledge of novel regulatory pathways that could contribute to gliomagenesis, we have conducted a systematic study of RBPs in GBM. RESULTS By measuring expression levels of 1542 human RBPs in GBM samples and glioma stem cell samples, we identified 58 consistently upregulated RBPs. Survival analysis revealed that increased expression of 21 RBPs was also associated with a poor prognosis. To assess the functional impact of those RBPs, we modulated their expression in GBM cell lines and performed viability, proliferation, and apoptosis assays. Combined results revealed a prominent oncogenic candidate, SNRPB, which encodes core spliceosome machinery components. To reveal the impact of SNRPB on splicing and gene expression, we performed its knockdown in a GBM cell line followed by RNA sequencing. We found that the affected genes were involved in RNA processing, DNA repair, and chromatin remodeling. Additionally, genes and pathways already associated with gliomagenesis, as well as a set of general cancer genes, also presented with splicing and expression alterations. CONCLUSIONS Our study provides new insights into how RBPs, and specifically SNRPB, regulate gene expression and directly impact GBM development.
Collapse
Affiliation(s)
- Bruna R Correa
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil
- Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | | | - Mei Qiao
- Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Suzanne C Burns
- Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Chen Chen
- Georgetown University Medical Center, Washington, DC, USA
| | | | - Seema Agarwal
- Georgetown University Medical Center, Washington, DC, USA
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil.
| | - Luiz O F Penalva
- Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA.
- Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX, USA.
| |
Collapse
|
37
|
Endovascular therapies for malignant gliomas: Challenges and the future. J Clin Neurosci 2016; 26:26-32. [DOI: 10.1016/j.jocn.2015.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/25/2015] [Indexed: 12/17/2022]
|
38
|
Jia A, Pannullo SC, Minkowitz S, Taube S, Chang J, Parashar B, Christos P, Wernicke AG. Innovative Hypofractionated Stereotactic Regimen Achieves Excellent Local Control with No Radiation Necrosis: Promising Results in the Management of Patients with Small Recurrent Inoperable GBM. Cureus 2016; 8:e536. [PMID: 27096136 PMCID: PMC4835149 DOI: 10.7759/cureus.536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Management of recurrent glioblastoma multiforme (GBM) remains a challenge. Several institutions reported that a single fraction of ≥ 20 Gy for small tumor burden results in excellent local control; however, this is at the expense of a high incidence of radiation necrosis (RN). Therefore, we developed a hypofractionation pattern of 33 Gy/3 fractions, which is a radiobiological equivalent of 20 Gy, with the aim to lower the incidence of RN. We reviewed records of 21 patients with recurrent GBM treated with hypofractionated stereotactic radiation therapy (HFSRT) to their 22 respective lesions. Sixty Gy fractioned external beam radiotherapy was performed as first-line treatment. Median time from primary irradiation to HFSRT was 9.6 months (range: 3.1 – 68.1 months). In HFSRT, a median dose of 33 Gy in 11 Gy fractions was delivered to the 80% isodose line that encompassed the target volume. The median tumor volume was 1.07 cm3 (range: 0.11 – 16.64 cm3). The median follow-up time after HFSRT was 9.3 months (range: 1.7 – 33.6 months). Twenty-one of 23 lesions treated (91.3%) achieved local control while 2/23 (8.7%) progressed. Median time to progression outside of the treated site was 5.2 months (range: 2.2 – 9.6 months). Progression was treated with salvage chemotherapy. Five of 21 patients (23.8%) were alive at the end of this follow-up; two patients remain disease-free. The remaining 16/21 patients (76.2%) died of disease. Treatment was well tolerated by all patients with no acute CTC/RTOG > Grade 2. There was 0% incidence of RN. A prospective trial will be underway to validate these promising results.
Collapse
Affiliation(s)
- Angela Jia
- Stich Radiation Oncology, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Susan C Pannullo
- Neurological Surgery, NewYork-Presbyterian/Weill Cornell Medical Center
| | | | - Shoshana Taube
- Stich Radiation Oncology, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Jenghwa Chang
- Stich Radiation Oncology, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Bhupesh Parashar
- Stich Radiation Oncology, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Paul Christos
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, NewYork-Presbyterian/Weill Cornell Medical Center
| | | |
Collapse
|
39
|
Abstract
Imaging is integral to the management of patients with brain tumors. Conventional structural imaging provides exquisite anatomic detail but remains limited in the evaluation of molecular characteristics of intracranial neoplasms. Quantitative and physiologic biomarkers derived from advanced imaging techniques have been increasingly utilized as problem-solving tools to identify glioma grade and assess response to therapy. This chapter provides a comprehensive overview of the imaging strategies used in the clinical assessment of patients with gliomas and describes how novel imaging biomarkers have the potential to improve patient management.
Collapse
Affiliation(s)
- Whitney B Pope
- Radiological Sciences, Ronald Reagan Medical Center, Los Angeles, CA, USA.
| | - Ibrahim Djoukhadar
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Alan Jackson
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| |
Collapse
|
40
|
Wang H, Xu T, Jiang Y, Xu H, Yan Y, Fu D, Chen J. The challenges and the promise of molecular targeted therapy in malignant gliomas. Neoplasia 2015; 17:239-55. [PMID: 25810009 PMCID: PMC4372648 DOI: 10.1016/j.neo.2015.02.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/06/2015] [Indexed: 11/18/2022] Open
Abstract
Malignant gliomas are the most common malignant primary brain tumors and one of the most challenging forms of cancers to treat. Despite advances in conventional treatment, the outcome for patients remains almost universally fatal. This poor prognosis is due to therapeutic resistance and tumor recurrence after surgical removal. However, over the past decade, molecular targeted therapy has held the promise of transforming the care of malignant glioma patients. Significant progress in understanding the molecular pathology of gliomagenesis and maintenance of the malignant phenotypes will open opportunities to rationally develop new molecular targeted therapy options. Recently, therapeutic strategies have focused on targeting pro-growth signaling mediated by receptor tyrosine kinase/RAS/phosphatidylinositol 3-kinase pathway, proangiogenic pathways, and several other vital intracellular signaling networks, such as proteasome and histone deacetylase. However, several factors such as cross-talk between the altered pathways, intratumoral molecular heterogeneity, and therapeutic resistance of glioma stem cells (GSCs) have limited the activity of single agents. Efforts are ongoing to study in depth the complex molecular biology of glioma, develop novel regimens targeting GSCs, and identify biomarkers to stratify patients with the individualized molecular targeted therapy. Here, we review the molecular alterations relevant to the pathology of malignant glioma, review current advances in clinical targeted trials, and discuss the challenges, controversies, and future directions of molecular targeted therapy.
Collapse
Affiliation(s)
- Hongxiang Wang
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ying Jiang
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hanchong Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yong Yan
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Da Fu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
41
|
Premkumar DR, Jane EP, Pollack IF. Cucurbitacin-I inhibits Aurora kinase A, Aurora kinase B and survivin, induces defects in cell cycle progression and promotes ABT-737-induced cell death in a caspase-independent manner in malignant human glioma cells. Cancer Biol Ther 2015; 16:233-43. [PMID: 25482928 DOI: 10.4161/15384047.2014.987548] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Because STAT signaling is commonly activated in malignant gliomas as a result of constitutive EGFR activation, strategies for inhibiting the EGFR/JAK/STAT cascade are of significant interest. We, therefore, treated a panel of established glioma cell lines, including EGFR overexpressors, and primary cultures derived from patients diagnosed with glioblastoma with the JAK/STAT inhibitor cucurbitacin-I. Treatment with cucurbitacin-I depleted p-STAT3, p-STAT5, p-JAK1 and p-JAK2 levels, inhibited cell proliferation, and induced G2/M accumulation, DNA endoreduplication, and multipolar mitotic spindles. Longer exposure to cucurbitacin-I significantly reduced the number of viable cells and this decrease in viability was associated with cell death, as confirmed by an increase in the subG1 fraction. Our data also demonstrated that cucurbitacin-I strikingly downregulated Aurora kinase A, Aurora kinase B and survivin. We then searched for agents that exhibited a synergistic effect on cell death in combination with cucurbitacin-I. We found that cotreatment with cucurbitacin-I significantly increased Bcl(-)2/Bcl(-)xL family member antagonist ABT-737-induced cell death regardless of EGFR/PTEN/p53 status of malignant human glioma cell lines. Although >50% of the cucurbitacin-I plus ABT-737 treated cells were annexin V and propidium iodide positive, PARP cleavage or caspase activation was not observed. Pretreatment of z-VAD-fmk, a pan caspase inhibitor did not inhibit cell death, suggesting a caspase-independent mechanism of cell death. Genetic inhibition of Aurora kinase A or Aurora kinase B or survivin by RNA interference also sensitized glioma cells to ABT-737, suggesting a link between STAT activation and Aurora kinases in malignant gliomas.
Collapse
Key Words
- Aurora kinases
- BSA, bovine serum albumin
- DMSO, dimethyl sulfoxide
- EGFR, epidermal growth factor receptor
- FITC, fluorescein isothiocyanate
- Glioma
- MTS, 3-[4, 5-dimethylthiazol- 2yl]-5-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]-2H, tetrazolium
- NF-кB, nuclear factor кB
- PAGE, polyacrylamide gel electrophoresis
- PBS, phosphate-buffered saline
- PDGFR, platelet derived growth factor receptor
- PI, propidium iodide
- PI3K, Phosphatidylinositol 3-Kinase
- TBS, Tris-buffered saline
- TRAIL, tumor necrosis factor–related apoptosis inducing ligand
- caspase-independent cell death
- cell cycle arrest
Collapse
Affiliation(s)
- Daniel R Premkumar
- a Department of Neurosurgery ; University of Pittsburgh School of Medicine ; Pittsburgh , PA USA
| | | | | |
Collapse
|
42
|
Abstract
Epilepsy develops in more than 70-90% of oligodendroglial tumors and represents a favorable indicator for long-term survival if present as the first clinical sign. Presence of IDH1 mutation is frequently associated with seizures in oligodendrogliomas, next to alterations of glutamate and GABA metabolism in the origin of glioma-associated epilepsy. Treatment by surgery or radiotherapy results in seizure freedom in about two-thirds of patients, and chemotherapy to a seizure reduction in about 50%. Symptomatic anticonvulsive therapy with levetiracetam and valproic acid as monotherapy are both evidence-based drugs for the partial epilepsies, and their effective use in brain tumors is supported by a large amount of additional data. Pharmacoresistance against anticonvulsants is more prevalent among oligodendrogliomas, occurring in about 40% despite polytherapy with two anticonvulsants or more. Toxic signs of anticonvulsants in brain tumors involve cognition, bone marrow and skin. Previous neurosurgery, radiation therapy or chemotherapy add to the risks of cognitive dysfunction.
Collapse
Affiliation(s)
- Melissa Kerkhof
- Department of Neurology, Medical Center The Hague, The Netherlands
| | - Christa Benit
- Department of Neurology, Medical Center The Hague, The Netherlands
| | | | - Charles J Vecht
- Service Neurologie Mazarin, GH Pitié-Salpêtrière, Paris, France
| |
Collapse
|
43
|
Preusser M, Lim M, Hafler DA, Reardon DA, Sampson JH. Prospects of immune checkpoint modulators in the treatment of glioblastoma. Nat Rev Neurol 2015; 11:504-14. [PMID: 26260659 DOI: 10.1038/nrneurol.2015.139] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Glioblastoma is the most common primary brain tumour in adults. Prognosis is poor: even with the current gold-standard first-line treatment—maximal safe resection and combination of radiotherapy with temozolomide chemotherapy—the median overall survival time is only approximately 15-17 months, because the tumour recurs in virtually all patients, and no commonly accepted standard treatment for recurrent disease exists. Several targeted agents have failed to improve patient outcomes in glioblastoma. Immunotherapy with immune checkpoint inhibitors such as ipilimumab, nivolumab, and pembrolizumab has provided relevant clinical improvements in other advanced tumours for which conventional therapies have had limited success, making immunotherapy an appealing strategy in glioblastoma. This Review summarizes current knowledge on immune checkpoint modulators and evaluates their potential role in glioblastoma on the basis of preclinical studies and emerging clinical data. Furthermore, we discuss challenges that need to be considered in the clinical development of drugs that target immune checkpoint pathways in glioblastoma, such as specific properties of the immune system in the CNS, issues with radiological response assessment, and potential interactions with established and emerging treatment strategies.
Collapse
Affiliation(s)
- Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Centre CNS Tumours Unit, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Michael Lim
- Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, Yale New Haven Hospital, 15 York Street, PO Box 208018, New Haven, CT 06520, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 2134, Boston, MA 02215, USA
| | - John H Sampson
- Division of Neurosurgery, 220 Sands Building, Research Drive, Duke University School of Medicine, Durham, NC 27705, USA
| |
Collapse
|
44
|
Zhang SD, Leung KL, McCrudden CM, Kwok HF. The Prognostic Significance of Combining VEGFA, FLT1 and KDR mRNA Expressions in Brain Tumors. J Cancer 2015; 6:812-8. [PMID: 26284131 PMCID: PMC4532977 DOI: 10.7150/jca.11975] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/12/2015] [Indexed: 02/07/2023] Open
Abstract
Tumor cells require angiogenesis to deliver nutrients and oxygen to support their fast growth and metabolism. The vascular endothelial growth factor (VEGF) pathway plays an important role in promoting angiogenesis, including tumor-induced angiogenesis. Recent clinical trials have demonstrated the benefit of targeting VEGF in the treatment of glioblastoma. However, the prognostic significance of the expression of VEGFA and its receptors VEGFR1 (FLT1) and VEGFR2 (KDR) are still largely elusive. In the present study, we aimed to investigate the prognostic significance of these three factors, alone or in combination, in glioma patients. Gene mRNA expression was extracted from three independent brain tumor cohorts totaling 242 patients and the association between gene expression and survival was tested. We found that when VEGFA, FLT1 and KDR expressions were considered alone, only VEGFA demonstrated a significant association with patient survival. Patients with high expression of both VEGFA and either receptor had significantly worse survival than patients expressing both factors at a low level. Importantly, we found that those patients whose tumors overexpressed all three genes had a significantly shorter survival compared to those patients with a low level expression of these genes. Our results suggest that a high level expression of VEGFA and its receptors, both FLT1 and KDR, may be required for brain tumor progression, and that these three factors should be considered together as a prognostic indicator for brain tumor patients.
Collapse
Affiliation(s)
- Shu-Dong Zhang
- 2. Center for Cancer Research & Cell Biology and School of Pharmacy, Queen's University Belfast, BT9 7BL, Belfast, United Kingdom
| | - Ka Lai Leung
- 1. Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau SAR
- 2. Center for Cancer Research & Cell Biology and School of Pharmacy, Queen's University Belfast, BT9 7BL, Belfast, United Kingdom
| | - Cian M. McCrudden
- 2. Center for Cancer Research & Cell Biology and School of Pharmacy, Queen's University Belfast, BT9 7BL, Belfast, United Kingdom
| | - Hang Fai Kwok
- 1. Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau SAR
- 2. Center for Cancer Research & Cell Biology and School of Pharmacy, Queen's University Belfast, BT9 7BL, Belfast, United Kingdom
- 3. Cancer Research UK Cambridge Institute, University of Cambridge, CB2 0RE, Cambridge, United Kingdom
| |
Collapse
|
45
|
Supramolecular nanoscale assemblies for cancer diagnosis and therapy. J Control Release 2015; 213:152-167. [PMID: 26160308 DOI: 10.1016/j.jconrel.2015.06.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/26/2015] [Accepted: 06/27/2015] [Indexed: 02/08/2023]
Abstract
Nanocarriers based on polymers, metals and lipids have been extensively developed for cancer therapy and diagnosis due to their ability to enhance drug accumulation in cancer cells and decrease undesired drug toxicity in healthy tissues. Overcoming multidrug resistance by designing proper drug nanocarriers will improve outcome of existing oncologic treatments such as chemotherapy and radiotherapy. In this article the relation between physicochemical properties and capacity of a nanosystem to deliver therapeutic agents into pathological sites is discussed. Most promising examples of drug delivery systems are reviewed, and, in particular, the design of a carbohydrate based matrix with entrapped gold nanoparticles is highlighted.
Collapse
|
46
|
NVP-BKM120 potentiates apoptosis in tumor necrosis factor-related apoptosis-inducing ligand-resistant glioma cell lines via upregulation of Noxa and death receptor 5. Int J Oncol 2015; 47:506-16. [PMID: 26044191 PMCID: PMC4501659 DOI: 10.3892/ijo.2015.3035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/15/2015] [Indexed: 01/14/2023] Open
Abstract
We previously observed that glioma cells are differentially sensitive to TRAIL-induced toxicity. Based on our observation that TRAIL-resistant glioma cell lines typically exhibited high levels of Akt activation, we hypothesized that inhibition of Akt signaling using the PI3 kinase inhibitor NVP-BKM120 could promote TRAIL-induced apoptosis in gliomas. We assessed this combination in established and primary cultured glioma cells. Combination treatment led to significant cellular death when compared to either drug alone, but had no effect in normal human astrocytes, and demonstrated activation of the caspase cascade. This enhanced apoptosis appears dependent upon the loss of mitochondrial membrane potential and the release of Smac/DIABLO, AIF and cytochrome c into the cytosol. The upregulation of Noxa and sequestration of Mcl-1 by Noxa were important factors for cell death. Knockdown of Noxa abrogated apoptosis and suggested dependency on Noxa in combination-induced apoptosis. BKM120 upregulated cell surface expression of death receptor 5 (DR5), but did not increase levels of the other major TRAIL receptor, death receptor 4 (DR4). This study demonstrates that antagonizing apoptosis-resistance pathways, such as the PI3/Akt pathway, in combination with death receptor activation, may induce cell death in TRAIL-resistant glioma.
Collapse
|
47
|
Systematic review and meta-analysis of phase I/II targeted therapy combined with radiotherapy in patients with glioblastoma multiforme: quality of report, toxicity, and survival. J Neurooncol 2015; 123:307-14. [PMID: 25975195 DOI: 10.1007/s11060-015-1802-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
To perform a systematic review and meta-analysis of severe adverse events (SAE) reported in early trials combining molecularly targeted therapies (MTT) with radiotherapy (RT), and to compare them to standard therapy. A summary data meta-analysis was performed and compared to the historical standard. Inclusion criteria were phase I and/or II trials published between 2000 and 2011, with glioblastoma multiforme patients treated with RT and MTT. Pooled incidence rates (IR) of SAE were estimated as well as the pooled median progression-free survival (PFS) and overall survival (OS). Nineteen prospective trials (9 phase I, 1 phase I/II and 9 phase II) out of 29 initially selected were included (n = 755 patients). The exact number of patients who had experienced SAE was mentioned in 37 % of the trials, concerning only 17 % of the patients. Information such as the period during which adverse events were monitored, the planned treatment duration, and late toxicity were not reported in the trials. The pooled IR of overall SAE was 131.2 (95 % CI 88.8-193.7) per 1000 person-months compared to 74.7 (63.6-87.8) for standard therapy (p < 0.01). Significant differences were observed for gastrointestinal events (p = 0.05) and treatment-related deaths (p = 0.02), in favour of standard therapy. No significant difference was observed in PFS and OS. Reporting a summary of toxicity data in early clinical trials should be stringently standardized. The use of MTT with RT compared to standard therapy increased SAE while yielded comparable survival in glioblastoma multiforme patients.
Collapse
|
48
|
Aithal MGS, Rajeswari N. Validation of housekeeping genes for gene expression analysis in glioblastoma using quantitative real-time polymerase chain reaction. Brain Tumor Res Treat 2015; 3:24-9. [PMID: 25977903 PMCID: PMC4426273 DOI: 10.14791/btrt.2015.3.1.24] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/23/2015] [Accepted: 04/02/2015] [Indexed: 11/21/2022] Open
Abstract
Background Quantitative real-time polymerase chain reaction (qPCR) is the most reliable tool for gene expression studies. Selection of housekeeping genes (HKGs) that are having most stable expression is critical to carry out accurate gene expression profiling. There is no 'universal' HKG having stable expression in all kinds of tissues under all experimental conditions. Methods The present study aims to identify most appropriate HKGs for gene expression analysis in glioblastoma (GBM) samples. Based on literature survey, six most commonly used HKGs that are invariant in GBM were chosen. We performed qPCR using RNA from formalin fixed paraffin embedded GBM samples and normal brain samples to investigate the expression pattern of HPRT, GAPDH, TBP, B2M, B2M, RPL13A, and RN18S1 with different abundance. A simple Δcycle threshold approach was employed to calculate the fold change. Results Our study shows that the expression of RPL13A and TBP were found to be most stable across all the samples and are thus suitable for gene expression analysis in human GBM. Except for TBP, none of the other conventionally used HKGs in GBM studies e.g., HPRT and GAPDH were found to be suitable as they showed variation in RNA expression. Conclusion Validation of HKGs is therefore immensely specific for a particular experimental setup and is crucial in assessing any new setup.
Collapse
Affiliation(s)
- Madhuri G S Aithal
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, India
| | - Narayanappa Rajeswari
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, India
| |
Collapse
|
49
|
Yuan G, Yan S, Xue H, Zhang P, Sun J, Li G. JSI-124 suppresses invasion and angiogenesis of glioblastoma cells in vitro. PLoS One 2015; 10:e0118894. [PMID: 25789853 PMCID: PMC4366361 DOI: 10.1371/journal.pone.0118894] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/13/2015] [Indexed: 01/13/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the utmost malignant tumors. Excessive angiogenesis and invasiveness are the major reasons for their uncontrolled growth and resistance toward conventional strategies resulting in poor prognosis. In this study, we found that low-dose JSI-124 reduced invasiveness and tumorigenicity of GBM cells. JSI-124 effectively inhibited VEGF expression in GBM cells. In a coculture study, JSI-124 completely prevented U87MG cell–mediated capillary formation of HUVECs and the migration of HUVECs when cultured alone or cocultured with U87MG cells. Furthermore, JSI-124 inhibited VEGF-induced cell proliferation, motility, invasion and the formation of capillary-like structures in HUVECs in a dose-dependent manner. JSI-124 suppressed VEGF-induced p-VEGFR2 activity through STAT3 signaling cascade in HUVECs. Immunohistochemistry analysis showed that the expression of CD34, Ki67, p-STAT3 and p-VEGFR2 protein in xenografts was remarkably decreased. Taken together, our findings provide the first evidence that JSI-124 effectively inhibits tumor angiogenesis and invasion, which might be a viable drug in anti-angiogenesis and anti-invasion therapies.
Collapse
Affiliation(s)
- Guang Yuan
- Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, P.R. China
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, P.R. China
- Department of Neurosurgery, Central Hospital of Zibo City, 54 Gongqingtuan Xi Road, Zibo, 255036, P.R. China
| | - Shaofeng Yan
- Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, P.R. China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, P.R. China
| | - Ping Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, P.R. China
| | - Jintang Sun
- Institute of Basic Medical Sciences and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, 44 Wenhua Xi Road, Jinan, 250012, P.R. China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, P.R. China
- Brain Science Research Institute, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, P.R. China
- * E-mail:
| |
Collapse
|
50
|
Cha J, Koh I, Choi Y, Lee J, Choi C, Kim P. Tapered microtract array platform for antimigratory drug screening of human glioblastoma multiforme. Adv Healthc Mater 2015; 4:405-11. [PMID: 25230171 DOI: 10.1002/adhm.201400384] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/13/2014] [Indexed: 01/24/2023]
Abstract
Understanding the effects of topographic characteristics on tumor cell migration is important for the development of new anti-migratory therapies. However, simplified in vitro culture systems often lead to inaccurate results regarding the efficacy of drugs. Histopathologically, glioblastoma multiform (GBM) cells migrate along the orientation of thin, elongated anatomical structures, such as white-matter tracts. Here, a tapered microtract array platform which mimics the anatomical features of brain tissue is introduced. This platform enables optimization of design for platform fabrication depending on topographic effects. By monitoring the migration of GBM cells on a simple tapered microtract, a saltatory migration resembling the migratory phenotype of human GBM cells in vivo is observed. The platform effectively induces the native characteristics and behavior of cells by topographic cues, allowing to observe the critical point for crawling to saltatory transition. Furthermore, this platform can be applied to efficiently screen anti-cancer drug by inhibiting associated signaling pathways on GBM cells. In conclusion, the microtract array platform reported here may provide a better understanding of the effects of topographic characteristics on cell migration, and may also be useful to determine the efficacy of antimigratory drugs for glioblastoma cells with cellular and molecular research and high-throughput screening.
Collapse
Affiliation(s)
- Junghwa Cha
- Department of Bio and Brain Engineering; KAIST; Daejeon 305-701 Korea
| | - Ilkyoo Koh
- Department of Bio and Brain Engineering; KAIST; Daejeon 305-701 Korea
| | - Yemuk Choi
- Department of Bio and Brain Engineering; KAIST; Daejeon 305-701 Korea
| | - Jungwhoi Lee
- Department of Bio and Brain Engineering; KAIST; Daejeon 305-701 Korea
| | - Chulhee Choi
- Department of Bio and Brain Engineering; KAIST; Daejeon 305-701 Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering; KAIST; Daejeon 305-701 Korea
| |
Collapse
|