1
|
Thanh HD, Lee S, Nguyen TT, Huu TN, Ahn EJ, Cho SH, Kim MS, Moon KS, Jung C. Temozolomide promotes matrix metalloproteinase 9 expression through p38 MAPK and JNK pathways in glioblastoma cells. Sci Rep 2024; 14:14341. [PMID: 38906916 PMCID: PMC11192740 DOI: 10.1038/s41598-024-65398-2] [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: 12/29/2023] [Accepted: 06/19/2024] [Indexed: 06/23/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and deadly brain cancer. Temozolomide (TMZ) is the standard chemotherapeutic agent for GBM, but the majority of patients experience recurrence and invasion of tumor cells. We investigated whether TMZ treatment of GBM cells regulates matrix metalloproteinases (MMPs), which have the main function to promote tumor cell invasion. TMZ effectively killed GL261, U343, and U87MG cells at a concentration of 500 µM, and surviving cells upregulated MMP9 expression and its activity but not those of MMP2. TMZ also elevated levels of MMP9 mRNA and MMP9 promoter activity. Subcutaneous graft tumors survived from TMZ treatment also exhibited increased expression of MMP9 and enhanced gelatinolytic activity. TMZ-mediated MMP9 upregulation was specifically mediated through the phosphorylation of p38 and JNK. This then stimulates AP-1 activity through the upregulation of c-Fos and c-Jun. Inhibition of the p38, JNK, or both pathways counteracted the TMZ-induced upregulation of MMP9 and AP-1. This study proposes a potential adverse effect of TMZ treatment for GBM: upregulation of MMP9 expression potentially associated with increased invasion and poor prognosis. This study also provides valuable insights into the molecular mechanisms by which TMZ treatment leads to increased MMP9 expression in GBM cells.
Collapse
Affiliation(s)
- Hien Duong Thanh
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Sueun Lee
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-Si, 58245, Jeollanam-Do, Korea
| | - Thuy Thi Nguyen
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Thang Nguyen Huu
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Eun-Jung Ahn
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun, 58128, Jeollanam-Do, Korea
| | - Sang-Hee Cho
- Department of Hemato-Oncology, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Min Soo Kim
- Department of Statistics, College of Natural Sciences, Chonnam National University, Gwangju, 61186, Korea
| | - Kyung-Sub Moon
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun, 58128, Jeollanam-Do, Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea.
| |
Collapse
|
2
|
Krauze AV, Zhao Y, Li MC, Shih J, Jiang W, Tasci E, Cooley Zgela T, Sproull M, Mackey M, Shankavaram U, Tofilon P, Camphausen K. Revisiting Concurrent Radiation Therapy, Temozolomide, and the Histone Deacetylase Inhibitor Valproic Acid for Patients with Glioblastoma-Proteomic Alteration and Comparison Analysis with the Standard-of-Care Chemoirradiation. Biomolecules 2023; 13:1499. [PMID: 37892181 PMCID: PMC10604983 DOI: 10.3390/biom13101499] [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: 08/24/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common brain tumor with an overall survival (OS) of less than 30% at two years. Valproic acid (VPA) demonstrated survival benefits documented in retrospective and prospective trials, when used in combination with chemo-radiotherapy (CRT). PURPOSE The primary goal of this study was to examine if the differential alteration in proteomic expression pre vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA as compared to standard-of-care CRT. The second goal was to explore the associations between the proteomic alterations in response to VPA/RT/TMZ correlated to patient outcomes. The third goal was to use the proteomic profile to determine the mechanism of action of VPA in this setting. MATERIALS AND METHODS Serum obtained pre- and post-CRT was analyzed using an aptamer-based SOMAScan® proteomic assay. Twenty-nine patients received CRT plus VPA, and 53 patients received CRT alone. Clinical data were obtained via a database and chart review. Tests for differences in protein expression changes between radiation therapy (RT) with or without VPA were conducted for individual proteins using two-sided t-tests, considering p-values of <0.05 as significant. Adjustment for age, sex, and other clinical covariates and hierarchical clustering of significant differentially expressed proteins was carried out, and Gene Set Enrichment analyses were performed using the Hallmark gene sets. Univariate Cox proportional hazards models were used to test the individual protein expression changes for an association with survival. The lasso Cox regression method and 10-fold cross-validation were employed to test the combinations of expression changes of proteins that could predict survival. Predictiveness curves were plotted for significant proteins for VPA response (p-value < 0.005) to show the survival probability vs. the protein expression percentiles. RESULTS A total of 124 proteins were identified pre- vs. post-CRT that were differentially expressed between the cohorts who received CRT plus VPA and those who received CRT alone. Clinical factors did not confound the results, and distinct proteomic clustering in the VPA-treated population was identified. Time-dependent ROC curves for OS and PFS for landmark times of 20 months and 6 months, respectively, revealed AUC of 0.531, 0.756, 0.774 for OS and 0.535, 0.723, 0.806 for PFS for protein expression, clinical factors, and the combination of protein expression and clinical factors, respectively, indicating that the proteome can provide additional survival risk discrimination to that already provided by the standard clinical factors with a greater impact on PFS. Several proteins of interest were identified. Alterations in GALNT14 (increased) and CCL17 (decreased) (p = 0.003 and 0.003, respectively, FDR 0.198 for both) were associated with an improvement in both OS and PFS. The pre-CRT protein expression revealed 480 proteins predictive for OS and 212 for PFS (p < 0.05), of which 112 overlapped between OS and PFS. However, FDR-adjusted p values were high, with OS (the smallest p value of 0.586) and PFS (the smallest p value of 0.998). The protein PLCD3 had the lowest p-value (p = 0.002 and 0.0004 for OS and PFS, respectively), and its elevation prior to CRT predicted superior OS and PFS with VPA administration. Cancer hallmark genesets associated with proteomic alteration observed with the administration of VPA aligned with known signal transduction pathways of this agent in malignancy and non-malignancy settings, and GBM signaling, and included epithelial-mesenchymal transition, hedgehog signaling, Il6/JAK/STAT3, coagulation, NOTCH, apical junction, xenobiotic metabolism, and complement signaling. CONCLUSIONS Differential alteration in proteomic expression pre- vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA. Using pre- vs. post-data, prognostic proteins emerged in the analysis. Using pre-CRT data, potentially predictive proteins were identified. The protein signals and hallmark gene sets associated with the alteration in the proteome identified between patients who received VPA and those who did not, align with known biological mechanisms of action of VPA and may allow for the identification of novel biomarkers associated with outcomes that can help advance the study of VPA in future prospective trials.
Collapse
Affiliation(s)
- Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Yingdong Zhao
- Computational and Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, USA; (Y.Z.); (M.-C.L.); (J.S.)
| | - Ming-Chung Li
- Computational and Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, USA; (Y.Z.); (M.-C.L.); (J.S.)
| | - Joanna Shih
- Computational and Systems Biology Branch, Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, USA; (Y.Z.); (M.-C.L.); (J.S.)
| | - Will Jiang
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Erdal Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Theresa Cooley Zgela
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Mary Sproull
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Megan Mackey
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Philip Tofilon
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA (T.C.Z.); (U.S.); (P.T.)
| |
Collapse
|
3
|
Antitumor Potential of Antiepileptic Drugs in Human Glioblastoma: Pharmacological Targets and Clinical Benefits. Biomedicines 2023; 11:biomedicines11020582. [PMID: 36831117 PMCID: PMC9953000 DOI: 10.3390/biomedicines11020582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma (GBM) is characterized by fast-growing cells, genetic and phenotypic heterogeneity, and radio-chemo-therapy resistance, contributing to its dismal prognosis. Various medical comorbidities are associated with the natural history of GBM. The most disabling and greatly affecting patients' quality of life are neurodegeneration, cognitive impairment, and GBM-related epilepsy (GRE). Hallmarks of GBM include molecular intrinsic mediators and pathways, but emerging evidence supports the key role of non-malignant cells within the tumor microenvironment in GBM aggressive behavior. In this context, hyper-excitability of neurons, mediated by glutamatergic and GABAergic imbalance, contributing to GBM growth strengthens the cancer-nervous system crosstalk. Pathogenic mechanisms, clinical features, and pharmacological management of GRE with antiepileptic drugs (AEDs) and their interactions are poorly explored, yet it is a potentially promising field of research in cancer neuroscience. The present review summarizes emerging cooperative mechanisms in oncogenesis and epileptogenesis, focusing on the neuron-to-glioma interface. The main effects and efficacy of selected AEDs used in the management of GRE are discussed in this paper, as well as their potential beneficial activity as antitumor treatment. Overall, although still many unclear processes overlapping in GBM growth and seizure onset need to be elucidated, this review focuses on the intriguing targeting of GBM-neuron mutual interactions to improve the outcome of the so challenging to treat GBM.
Collapse
|
4
|
Tyagunova EE, Zakharov AS, Glukhov AI, Dobrokhotova VZ, Shlapakov TI, Kozlov VV, Korotkova NV, Tyagunova TE. Features of epileptiform activity in patients with diagnosed glioblastoma: from genetic and biochemical mechanisms to clinical aspects. HEAD AND NECK TUMORS (HNT) 2022. [DOI: 10.17650/2222-1468-2022-12-3-102-113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction. glioblastomas multiforme (grade Iv gliomas) are common and the most aggressive primary tumors of the brain with very unfavorable prognosis. In all previously published papers on epileptiform activity in glioblastomas, not enough information on encephalogram results is presented.Aim. To study the features of epileptiform activity in patients with glioblastomas and development of a plan for further study of these patients.Materials and methods. An analysis of articles from Elsevier, Embase, Scopus, The Cochrane Library, global Health, Russian Science Citation Index (RSCI) databases, Scholar, google, web of Science, pubmed search engines and scientific electronic library CyberLeninka was performed. materials were selected considering journal indexing system and citations, scientific novelty of the studies, statistical significance of the results. publications repeating data from previous articles or describing animal experiments were excluded from analysis.Results. During the study, data on mechanisms of epileptiform activity pathogenesis, predisposing factors (tumor location in the temporal, frontal or parietal lobes, IDH-1 and / or IDH-2 gene mutations), treatment options in patients with glioblastomas were systemized. Additionally, and original plan of data accumulation for clinical studied taking into account limitations of the previous studies was developed to increase quality of results interpretation.Conclusion. Epileptiform symptoms in glioblastomas negatively affect patients’ quality of life and lifespan. Currently, researchers actively search for an effective method of treatment of epileptic seizures in patients with glioblastomas. The most effective is combination of temozolomide with valproate and levetiracetam due to good control of seizure frequency, low toxicity, and pharmacological synergy between the drugs.
Collapse
Affiliation(s)
- E. E. Tyagunova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | - A. S. Zakharov
- Pavlov Ryazan State Medical University, Ministry of Health of Russia
| | - A. I. Glukhov
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia; M.V. Lomonosov Moscow State University
| | - V. Z. Dobrokhotova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia; N. N. Blokhin National Research Institute of Oncology, Ministry of Health of Russia
| | - T. I. Shlapakov
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | - V. V. Kozlov
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | - N. V. Korotkova
- Pavlov Ryazan State Medical University, Ministry of Health of Russia
| | | |
Collapse
|
5
|
NMDA receptor signaling induces the chemoresistance of temozolomide via upregulation of MGMT expression in glioblastoma cells. J Neurooncol 2022; 160:375-388. [DOI: 10.1007/s11060-022-04154-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022]
|
6
|
Zhu Y, Chen Z, Kim SN, Gan C, Ryl T, Lesjak MS, Rodemerk J, Zhong RD, Wrede K, Dammann P, Sure U. Characterization of Temozolomide Resistance Using a Novel Acquired Resistance Model in Glioblastoma Cell Lines. Cancers (Basel) 2022; 14:cancers14092211. [PMID: 35565340 PMCID: PMC9101568 DOI: 10.3390/cancers14092211] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Temozolomide (TMZ) is the first-line drug for chemotherapy of GBM, the most aggressive and incurable brain tumor. Acquired chemoresistance is a hallmark that causes the poor prognosis of GBM. Therefore, understanding the underlying mechanisms by using a proper model becomes emergent. Previous models usually take weeks/months and are often not fully representative of characteristics of TMZ resistance. We established an acute acquired TMZ resistance model using GBM cell lines with different genomic backgrounds. In response to TMZ, the resistant cells showed less susceptibility and sustained regrowth, high clonogenicity, reduced DNA damage accompanied by attenuated MMR, shortened G2/M arrest, uncontrolled DNA replication, and evasion of apoptosis. Moreover, these TMZ resistant cells presented stem cell properties that are critical for chemoresistance. Thus, our model recapitulates all key features of TMZ resistance and is believed to be a promising model to study the underlying mechanisms and define therapeutics for GBM in the future. Abstract Temozolomide (TMZ) is the first line of standard therapy in glioblastoma (GBM). However, relapse occurs due to TMZ resistance. We attempted to establish an acquired TMZ resistance model that recapitulates the TMZ resistance phenotype and the relevant gene signature. Two GBM cell lines received two cycles of TMZ (150 µM) treatment for 72 h each. Regrown cells (RG2) were defined as TMZ resistant cells. MTT assay revealed significantly less susceptibility and sustained growth of RG2 compared with parental cells after TMZ challenge. TMZ-induced DNA damage significantly decreased in 53BP1-foci reporter transduced-RG2 cells compared with parental cells, associated with downregulation of MSH2 and MSH6. Flow cytometry revealed reduced G2/M arrest, increased EdU incorporation and suppressed apoptosis in RG2 cells after TMZ treatment. Colony formation and neurosphere assay demonstrated enhanced clonogenicity and neurosphere formation capacity in RG2 cells, accompanied by upregulation of stem markers. Collectively, we established an acute TMZ resistance model that recapitulated key features of TMZ resistance involving impaired mismatch repair, redistribution of cell cycle phases, increased DNA replication, reduced apoptosis and enhanced self-renewal. Therefore, this model may serve as a promising research tool for studying mechanisms of TMZ resistance and for defining therapeutic approaches to GBM in the future.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
- Correspondence: ; Tel.: +0049-201-723-1231
| | - Zhen Chen
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
| | - Su Na Kim
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
| | - Chao Gan
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
| | - Tatsiana Ryl
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
| | - Michaela Silvia Lesjak
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
| | - Jan Rodemerk
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
| | - Rong De Zhong
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
| | - Karsten Wrede
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
| | - Philipp Dammann
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
| | - Ulrich Sure
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (Z.C.); (S.N.K.); (C.G.); (T.R.); (M.S.L.); (J.R.); (R.D.Z.); (K.W.); (P.D.); (U.S.)
- Center for Translational Neuro- & Behavioral Sciences (C-TNBS), University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
7
|
Cheng M, Wang Q, Chen L, Zhao D, Tang J, Xu J, He Z. LncRNA UCA1/miR-182-5p/MGMT axis modulates glioma cell sensitivity to TMZ through MGMT-related DNA damage pathways. Hum Pathol 2022; 123:59-73. [DOI: 10.1016/j.humpath.2022.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/30/2022]
|
8
|
Mukesh RK, Kalam AA, Nag J, Jaikumar VS, Kunnakkadan U, Kumar NA, Suma SM, Rajavelu A, Johnson JB. Chandipura virus induces cell death in cancer cell lines of human origin and promotes tumor regression in vivo. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:254-265. [PMID: 34761105 PMCID: PMC8554107 DOI: 10.1016/j.omto.2021.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 11/25/2022]
Abstract
Chandipura virus (CHPV) is an emerging human pathogen of great clinical significance. In this study, we have investigated the susceptibility pattern of both normal and cancer cell lines of human origin to wild-type (wt) CHPV in order to explore the possibility of developing CHPV as an oncolytic vector (OV). Marked cytopathic effect along with enhanced virus output was observed in cancer cell lines (HeLa, A549, U-138, PC-3, and HepG2) in comparison to normal human adult dermal fibroblast (HADF) cells. At an MOI of 0.1, cancer cell lines were differentially susceptible to CHPV, with cells like HeLa and U-138 having pronounced cell death, while the PC-3 were comparatively resistant. All cell lines used in the study except U-138 restricted CHPV infection to varying degrees with IFN-β pre-treatment and supplementation of interferon (IFN) could neither activate the IFN signaling pathway in U-138 cells. Finally, U-138 tumor xenografts established in non-obese diabetic severe combined immunodeficiency (NOD/SCID) mice showed significant delay in tumor growth in the CHPV-challenged animals. Thus, targeted cytopathic effect in cancer cells at a very low dose with restricted replication in normal cells offers a rationale to exploit CHPV as an oncolytic vector in the future.
Collapse
Affiliation(s)
- Reshma Koolaparambil Mukesh
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Azeem Abdul Kalam
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Joydeep Nag
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Vishnu Sunil Jaikumar
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Umerali Kunnakkadan
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram 695581, Kerala, India
| | - Nisha Asok Kumar
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | | | - Arumugam Rajavelu
- Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - John Bernet Johnson
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| |
Collapse
|
9
|
Hwang K, Kim J, Kang SG, Jung TY, Kim JH, Kim SH, Kang SH, Hong YK, Kim TM, Kim YJ, Choi BS, Chang JH, Kim CY. Levetiracetam as a sensitizer of concurrent chemoradiotherapy in newly diagnosed glioblastoma: An open-label phase 2 study. Cancer Med 2021; 11:371-379. [PMID: 34845868 PMCID: PMC8729048 DOI: 10.1002/cam4.4454] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND An open-label single-arm phase 2 study was conducted to evaluate the role of levetiracetam as a sensitizer of concurrent chemoradiotherapy (CCRT) for patients with newly diagnosed glioblastoma. This study aimed to determine the survival benefit of levetiracetam in conjunction with the standard treatment for glioblastoma. METHODS Major eligibility requirements included histologically proven glioblastoma in the supratentorial region, patients 18 years or older, and Eastern Cooperative Oncology Group (ECOG) performance status of 0-2. Levetiracetam was given at 1,000-2,000 mg daily in two divided doses during CCRT and adjuvant chemotherapy thereafter. The primary and the secondary endpoints were 6-month progression-free survival (6mo-PFS) and 24-month overall survival (24mo-OS), respectively. Outcomes of the study group were compared to those of an external control group. RESULTS Between July 2016 and January 2019, 76 patients were enrolled, and 73 patients were included in the final analysis. The primary and secondary outcomes were improved in the study population compared to the external control (6mo-PFS, 84.9% vs. 72.3%, p = 0.038; 24mo-OS, 58.0% vs. 39.9%, p = 0.018), but the differences were less prominent in a propensity score-matched analysis (6mo-PFS, 88.0% vs. 76.9%, p = 0.071; 24mo-OS, 57.1% vs. 38.8%, p = 0.054). In exploratory subgroup analyses, some results suggested that patients with ages under 65 years or unmethylated MGMT promoter might have a greater survival benefit from the use of levetiracetam. CONCLUSIONS The use of levetiracetam during CCRT in patients with newly diagnosed glioblastoma may result in improved outcomes, but further investigations are warranted.
Collapse
Affiliation(s)
- Kihwan Hwang
- Department of Neurosurgery, Internal Medicine, Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Junhyung Kim
- Department of Neurosurgery, Internal Medicine, Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Young Jung
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Chonnam National University College of Medicine, Hwasun, Republic of Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Se-Hyuk Kim
- Department of Neurosurgery, Ajou University Hospital, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Shin-Hyuk Kang
- Department of Neurosurgery, Korea University Medical Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong-Kil Hong
- Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Jung Kim
- Department of Neurosurgery, Internal Medicine, Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Byung Se Choi
- Department of Neurosurgery, Internal Medicine, Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chae-Yong Kim
- Department of Neurosurgery, Internal Medicine, Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| |
Collapse
|
10
|
Synergistic Anticancer Effect of Glycolysis and Histone Deacetylases Inhibitors in a Glioblastoma Model. Biomedicines 2021; 9:biomedicines9121749. [PMID: 34944565 PMCID: PMC8698815 DOI: 10.3390/biomedicines9121749] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/16/2021] [Accepted: 11/22/2021] [Indexed: 11/23/2022] Open
Abstract
Over the last decade, we have seen tremendous progress in research on 2-deoxy-D-glucose (2-DG) and its analogs. Clinical trials of 2-DG have demonstrated the challenges of using 2-DG as a monotherapy, due to its poor drug-like characteristics, leading researchers to focus on improving its bioavailability to tissue and organs. Novel 2-DG analogs such as WP1122 and others have revived the old concept of glycolysis inhibition as an effective anticancer strategy. Combined with other potent cytotoxic agents, inhibitors of glycolysis could synergistically eliminate cancer cells. We focused our efforts on the development of new combinations of anticancer agents coupled with 2-DG and its derivatives, targeting glioblastoma, which is in desperate need of novel approaches and therapeutic options and is particularly suited to glycolysis inhibition, due to its reliance on aerobic glycolysis. Herein, we present evidence that a combined treatment of 2-DG analogs and modulation of histone deacetylases (HDAC) activity via HDAC inhibitors (sodium butyrate and sodium valproate) exerts synergistic cytotoxic effects in glioblastoma U-87 and U-251 cells and represents a promising therapeutic strategy.
Collapse
|
11
|
Tresch NS, Fuchs D, Morandi L, Tonon C, Rohrer Bley C, Nytko KJ. Temozolomide is additive with cytotoxic effect of irradiation in canine glioma cell lines. Vet Med Sci 2021; 7:2124-2134. [PMID: 34477324 PMCID: PMC8604143 DOI: 10.1002/vms3.620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Similar to human glioblastoma patients, glial tumours in dogs have high treatment resistance and a guarded prognosis. In human medicine, the addition of temozolomide to radiotherapy leads to a favourable outcome in vivo as well as a higher antiproliferative effect on tumour cells in vitro. OBJECTIVES The aim of the study was to determine the radio- and temozolomide-sensitivity of three canine glial tumour cell lines and to investigate a potential additive cytotoxic effect in combined treatment. Additionally, we wanted to detect the level of MGMT promoter methylation in these cell lines and to investigate a potential association between MGMT promoter methylation and treatment resistance. METHODS Cells were treated with various concentrations of temozolomide and/or irradiated with 4 and 8 Gy. Radiosensitization by temozolomide was evaluated using proliferation assay and clonogenic assay, and MGMT DNA methylation was investigated using bisulfite next-generation sequencing. RESULTS In all tested canine cell lines, clonogenicity was inhibited significantly in combined treatment compared to radiation alone. All canine glial cell lines tested in this study were found to have high methylation levels of MGMT promoter. CONCLUSIONS Hence, an additive effect of combined treatment in MGMT negative canine glial tumour cell lines in vitro was detected. This motivates to further investigate the association between treatment resistance and MGMT, such as MGMT promoter methylation status.
Collapse
Affiliation(s)
- Nina Simona Tresch
- Division of Radiation OncologyVetsuisse Faculty University of ZurichZurichSwitzerland
- Center for Clinical Studies at the Vetsuisse Faculty of the University of ZurichZurichSwitzerland
| | - Daniel Fuchs
- Division of Radiation OncologyVetsuisse Faculty University of ZurichZurichSwitzerland
- Center for Clinical Studies at the Vetsuisse Faculty of the University of ZurichZurichSwitzerland
| | - Luca Morandi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- Functional and Molecular Neuroimaging UnitIRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Caterina Tonon
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- Functional and Molecular Neuroimaging UnitIRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Carla Rohrer Bley
- Division of Radiation OncologyVetsuisse Faculty University of ZurichZurichSwitzerland
- Center for Clinical Studies at the Vetsuisse Faculty of the University of ZurichZurichSwitzerland
| | - Katarzyna J. Nytko
- Division of Radiation OncologyVetsuisse Faculty University of ZurichZurichSwitzerland
- Center for Clinical Studies at the Vetsuisse Faculty of the University of ZurichZurichSwitzerland
| |
Collapse
|
12
|
Tsai HC, Wei KC, Chen PY, Huang CY, Chen KT, Lin YJ, Cheng HW, Chen YR, Wang HT. Valproic Acid Enhanced Temozolomide-Induced Anticancer Activity in Human Glioma Through the p53-PUMA Apoptosis Pathway. Front Oncol 2021; 11:722754. [PMID: 34660288 PMCID: PMC8518553 DOI: 10.3389/fonc.2021.722754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/08/2021] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma (GBM), the most lethal type of brain tumor in adults, has considerable cellular heterogeneity. The standard adjuvant chemotherapeutic agent for GBM, temozolomide (TMZ), has a modest response rate due to the development of drug resistance. Multiple studies have shown that valproic acid (VPA) can enhance GBM tumor control and prolong survival when given in conjunction with TMZ. However, the beneficial effect is variable. In this study, we analyzed the impact of VPA on GBM patient survival and its possible correlation with TMZ treatment and p53 gene mutation. In addition, the molecular mechanisms of TMZ in combination with VPA were examined using both p53 wild-type and p53 mutant human GBM cell lines. Our analysis of clinical data indicates that the survival benefit of a combined TMZ and VPA treatment in GBM patients is dependent on their p53 gene status. In cellular experiments, our results show that VPA enhanced the antineoplastic effect of TMZ by enhancing p53 activation and promoting the expression of its downstream pro-apoptotic protein, PUMA. Our study indicates that GBM patients with wild-type p53 may benefit from a combined TMZ+VPA treatment.
Collapse
Affiliation(s)
- Hong-Chieh Tsai
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan.,Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pin-Yuan Chen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan.,Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ko-Ting Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ya-Jui Lin
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Wei Cheng
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Institute of Pharmacology, College of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Rou Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Doctor Degree Program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
Lavogina D, Laasfeld T, Vardja M, Lust H, Jaal J. Viability fingerprint of glioblastoma cell lines: roles of mitotic, proliferative, and epigenetic targets. Sci Rep 2021; 11:20338. [PMID: 34645858 PMCID: PMC8514540 DOI: 10.1038/s41598-021-99630-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023] Open
Abstract
Despite the use of multimodal treatment combinations, the prognosis of glioblastoma (GB) is still poor. To prevent rapid tumor recurrence, targeted strategies for the treatment of GB are widely sought. Here, we compared the efficacy of focused modulation of a set of signaling pathways in two GB cell lines, U-251 MG and T98-G, using a panel of thirteen compounds targeting cell cycle progression, proliferation, epigenetic modifications, and DNA repair mechanism. In parallel, we tested combinations of these compounds with temozolomide and lomustine, the standard chemotherapy agents used in GB treatment. Two major trends were found: within individual compounds, the lowest IC50 values were exhibited by the Aurora kinase inhibitors, whereas in the case of mixtures, the addition of DNA methyltransferase 1 inhibitor azacytidine to lomustine proved the most beneficial. The efficacy of cell cycle-targeting compounds was further augmented by combination with radiation therapy using two different treatment regimes. The potency of azacytidine and lomustine mixtures was validated using a unique assay pipeline that utilizes automated imaging and machine learning-based data analysis algorithm for assessment of cell number and DNA damage extent. Based on our results, the combination of azacytidine and lomustine should be tested in GB clinical trials.
Collapse
Affiliation(s)
- Darja Lavogina
- grid.10939.320000 0001 0943 7661Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, 50406 Tartu, Estonia ,grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Tõnis Laasfeld
- grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Tartu, Estonia ,grid.10939.320000 0001 0943 7661Department of Computer Science, University of Tartu, Tartu, Estonia
| | - Markus Vardja
- grid.412269.a0000 0001 0585 7044Department of Radiotherapy and Oncological Therapy, Tartu University Hospital, Tartu, Estonia
| | - Helen Lust
- grid.10939.320000 0001 0943 7661Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, 50406 Tartu, Estonia
| | - Jana Jaal
- grid.10939.320000 0001 0943 7661Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, 50406 Tartu, Estonia ,grid.412269.a0000 0001 0585 7044Department of Radiotherapy and Oncological Therapy, Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
14
|
Han W, Guan W. Valproic Acid: A Promising Therapeutic Agent in Glioma Treatment. Front Oncol 2021; 11:687362. [PMID: 34568018 PMCID: PMC8461314 DOI: 10.3389/fonc.2021.687362] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
Glioma, characterized by infiltrative growth and treatment resistance, is regarded as the most prevalent intracranial malignant tumor. Due to its poor prognosis, accumulating investigation has been performed for improvement of overall survival (OS) and progression-free survival (PFS) in glioma patients. Valproic acid (VPA), one of the most common histone deacetylase inhibitors (HDACIs), has been detected to directly or synergistically exert inhibitory effects on glioma in vitro and in vivo. In this review, we generalize the latest advances of VPA in treating glioma and its underlying mechanisms and clinical implications, providing a clearer profile for clinical application of VPA as a therapeutic agent for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
15
|
Haddad AF, Young JS, Amara D, Berger MS, Raleigh DR, Aghi MK, Butowski NA. Mouse models of glioblastoma for the evaluation of novel therapeutic strategies. Neurooncol Adv 2021; 3:vdab100. [PMID: 34466804 PMCID: PMC8403483 DOI: 10.1093/noajnl/vdab100] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM) is an incurable brain tumor with a median survival of approximately 15 months despite an aggressive standard of care that includes surgery, chemotherapy, and ionizing radiation. Mouse models have advanced our understanding of GBM biology and the development of novel therapeutic strategies for GBM patients. However, model selection is crucial when testing developmental therapeutics, and each mouse model of GBM has unique advantages and disadvantages that can influence the validity and translatability of experimental results. To shed light on this process, we discuss the strengths and limitations of 3 types of mouse GBM models in this review: syngeneic models, genetically engineered mouse models, and xenograft models, including traditional xenograft cell lines and patient-derived xenograft models.
Collapse
Affiliation(s)
- Alexander F Haddad
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Dominic Amara
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Corresponding Author: Nicholas A. Butowski, MD, Department of Neurological Surgery, University of California, San Francisco, 400 Parnassus Ave Eighth Floor, San Francisco, CA, 94143, USA ()
| |
Collapse
|
16
|
Yebra M, Bhargava S, Kumar A, Burgoyne AM, Tang CM, Yoon H, Banerjee S, Aguilera J, Cordes T, Sheth V, Noh S, Ustoy R, Li S, Advani SJ, Corless CL, Heinrich MC, Kurzrock R, Lippman SM, Fanta PT, Harismendy O, Metallo C, Sicklick JK. Establishment of Patient-Derived Succinate Dehydrogenase-Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response. Clin Cancer Res 2021; 28:187-200. [PMID: 34426440 DOI: 10.1158/1078-0432.ccr-21-2092] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract, with mutant succinate dehydrogenase (SDH) subunits (A-D) comprising less than 7.5% (i.e., 150-200/year) of new cases annually in the United States. Contrary to GISTs harboring KIT or PDGFRA mutations, SDH-mutant GISTs affect adolescents/young adults, often metastasize, and are frequently resistant to tyrosine kinase inhibitors (TKI). Lack of human models for any SDH-mutant tumors, including GIST, has limited molecular characterization and drug discovery. EXPERIMENTAL DESIGN We describe methods for establishing novel patient-derived SDH-mutant (mSDH) GIST models and interrogated the efficacy of temozolomide on these tumor models in vitro and in clinical trials of patients with mSDH GIST. RESULTS Molecular and metabolic characterization of our patient-derived mSDH GIST models revealed that these models recapitulate the transcriptional and metabolic hallmarks of parent tumors and SDH deficiency. We further demonstrate that temozolomide elicits DNA damage and apoptosis in our mSDH GIST models. Translating our in vitro discovery to the clinic, a cohort of patients with SDH-mutant GIST treated with temozolomide (n = 5) demonstrated a 40% objective response rate and 100% disease control rate, suggesting that temozolomide represents a promising therapy for this subset of GIST. CONCLUSIONS We report the first methods to establish patient-derived mSDH tumor models, which can be readily employed for understanding patient-specific tumor biology and treatment strategies. We also demonstrate that temozolomide is effective in patients with mSDH GIST who are refractory to existing chemotherapeutic drugs (namely, TKIs) in clinic for GISTs, bringing a promising treatment option for these patients to clinic.
Collapse
Affiliation(s)
- Mayra Yebra
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Shruti Bhargava
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Avi Kumar
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Adam M Burgoyne
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California
| | - Chih-Min Tang
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Hyunho Yoon
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Sudeep Banerjee
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Joseph Aguilera
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | - Thekla Cordes
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Vipul Sheth
- Department of Radiology, Stanford University, Palo Alto, Stanford, California
| | - Sangkyu Noh
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Rowan Ustoy
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Sam Li
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Sunil J Advani
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | | | - Michael C Heinrich
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, Portland, Oregon
| | - Razelle Kurzrock
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| | - Scott M Lippman
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| | - Paul T Fanta
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| | - Olivier Harismendy
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Biomedical Informatics, University of California San Diego, San Diego, California
| | - Christian Metallo
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California.,Diabetes and Endocrinology Research Center, University of California San Diego, La Jolla, California.,Institute of Engineering in Medicine, University of California San Diego, La Jolla, California
| | - Jason K Sicklick
- Moores Cancer Center, University of California San Diego, La Jolla, California. .,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| |
Collapse
|
17
|
Warnon C, Bouhjar K, Ninane N, Verhoyen M, Fattaccioli A, Fransolet M, Lambert de Rouvroit C, Poumay Y, Piel G, Mottet D, Debacq-Chainiaux F. HDAC2 and 7 down-regulation induces senescence in dermal fibroblasts. Aging (Albany NY) 2021; 13:17978-18005. [PMID: 34253688 PMCID: PMC8351730 DOI: 10.18632/aging.203304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Originally simply reported to be in a stable and irreversible growth arrest in vitro, senescent cells are now clearly associated with normal and pathological ageing in vivo. They are characterized by several biomarkers and changes in gene expression that may depend on epigenetic factors, such as histone acetylation, involving a balance between histone acetyltransferases (HATs) and histone deacetylases (HDACs). In this study, we investigate the expression and the role of HDACs on the senescent phenotype of dermal fibroblasts. We report that during replicative senescence, most canonical HDACs are less expressed. Moreover, treatment with SAHA, a histone deacetylase inhibitor (HDACi) also known as Vorinostat, or the specific downregulation of HDAC2 or HDAC7 by siRNA, induces the appearance of senescence biomarkers of dermal fibroblasts. Conversely, the ectopic re-expression of HDAC7 by lentiviral transduction in pre-senescent dermal fibroblasts extends their proliferative lifespan. These results demonstrate that HDACs expression can modulate the senescent phenotype, highlighting their pharmaceutical interest in the context of healthy ageing.
Collapse
Affiliation(s)
- Céline Warnon
- URBC, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Karim Bouhjar
- URBC, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Noëlle Ninane
- URBC, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Mathilde Verhoyen
- URBC, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Antoine Fattaccioli
- URBC, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Maude Fransolet
- URBC, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | | | - Yves Poumay
- URPHYM, Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Géraldine Piel
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, University of Liège, Liège, Belgium
| | - Denis Mottet
- University of Liège, GIGA-Molecular Biology of Diseases, Gene Expression and Cancer Laboratory, Liège, Belgium
| | | |
Collapse
|
18
|
Rani V, Prabhu A. Combining Angiogenesis Inhibitors with Radiation: Advances and Challenges in Cancer Treatment. Curr Pharm Des 2021; 27:919-931. [PMID: 33006535 DOI: 10.2174/1381612826666201002145454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/28/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Radiation therapy is a widely employed modality that is used to destroy cancer cells, but it also tends to induce changes in the tumor microenvironment and promote angiogenesis. Radiation, when used as a sole means of therapeutic approach to treat cancer, tends to trigger the angiogenic pathways, leading to the upregulation of several angiogenic growth factors such as VEGF, bFGF, PDGF and angiogenin. This uncontrolled angiogenesis leads to certain angiogenic disorders like vascular outgrowth and an increase in tumor progression that can pose a serious threat to patients. OBJECTIVE This review emphasizes on various components of the tumor microenvironment, angiogenic growth factors and biological effects of radiation on tumors in provoking the relapse. It also describes the angiogenic mechanisms that trigger the tumor relapse after radiation therapy and how angiogenesis inhibitors can help in overcoming this phenomenon. It gives an overview of various angiogenesis inhibitors in pre-clinical as well as in clinical trials. CONCLUSION The review focuses on the beneficial effects of the combinatorial therapeutic approach of anti-angiogenesis therapy and radiation in tumor management.
Collapse
Affiliation(s)
- Vinitha Rani
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore - 575 018, Karnataka, India
| | - Ashwini Prabhu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore - 575 018, Karnataka, India
| |
Collapse
|
19
|
Wei X, Xiao B, Wang L, Zang L, Che F. Potential new targets and drugs related to histone modifications in glioma treatment. Bioorg Chem 2021; 112:104942. [PMID: 33965781 DOI: 10.1016/j.bioorg.2021.104942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
Glioma accounts for 40-50% of craniocerebral tumors, whose outcome rarely improves after standard treatment. The development of new therapeutic targets for glioma treatment has important clinical significance. With the deepening of research on gliomas, recent researchers have found that the occurrence and development of gliomas is closely associated with histone modifications, including methylation, acetylation, phosphorylation, and ubiquitination. Additionally, evidence has confirmed the close relationship between histone modifications and temozolomide (TMZ) resistance. Therefore, histone modification-related proteins have been widely recognized as new therapeutic targets for glioma treatment. In this review, we summarize the potential histone modification-associated targets and related drugs for glioma treatment. We have further clarified how histone modifications regulate the pathogenesis of gliomas and the mechanism of drug action, providing novel insights for the current clinical glioma treatment. Herein, we have also highlighted the limitations of current clinical therapies and have suggested future research directions and expected advances in potential areas of disease prognosis. Due to the complicated glioma pathogenesis, in the present review, we have acknowledged the limitations of histone modification applications in the related clinical treatment.
Collapse
Affiliation(s)
- Xiuhong Wei
- Graduate School, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, China; Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, Shandong, China
| | - Bolian Xiao
- Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Key Laboratory of Neurophysiology, Key Laboratory of Tumor Biology, Linyi, Shandong, China
| | - Liying Wang
- Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Department of Neurology, the Clinical Medical College of Weifang Medical College, Weifang, Shandong, China
| | - Lanlan Zang
- Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Key Laboratory of Neurophysiology, Key Laboratory of Tumor Biology, Linyi, Shandong, China; Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| | - Fengyuan Che
- Graduate School, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, China; Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Key Laboratory of Neurophysiology, Key Laboratory of Tumor Biology, Linyi, Shandong, China.
| |
Collapse
|
20
|
New insights into cytotoxic mechanisms of bozepinib against glioblastoma. Eur J Pharm Sci 2021; 162:105823. [PMID: 33781855 DOI: 10.1016/j.ejps.2021.105823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 02/18/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive brain tumor in adults and the current treatments only have a modest effect on patient survival. Recent studies show that bozepinib (BZP), a purine derivative, has potential applications in cancer treatment. The aim of this study was to evaluate the effect of BZP against GBM cells, specially concerning the purinergic system. Thus, GBM cells (C6 and U138 cell lines) were treated with BZP and cell viability, cell cycle, and annexin/PI assays, and active caspase-3 measurements were carried out. Besides, the effect of BZP over the purinergic system was also evaluated in silico and in vitro. Finally, we evaluate the action of BZP against important markers related to cancer progression, such as Akt, NF-κB, and CD133. We demonstrate here that BZP reduces GBM cell viability (IC50 = 5.7 ± 0.3 µM and 12.7 ± 1.5 µM, in C6 and U138 cells, respectively), inducing cell death through caspase-dependent apoptosis, autophagosome formation, activation of NF-κB, without any change in cell cycle progression or on the Akt pathway. Also, BZP modulates the purinergic system, inducing an increase in CD39 enzyme expression and activity, while inhibiting CD73 activity and adenosine formation, without altering CD73 enzyme expression. Curiously, one cycle of treatment resulted in enrichment of GBM cells expressing NF-κB and CD133+, suggesting resistant cells selection. However, after another treatment round, the resistant cells were eliminated. Altogether, BZP presented in vitro anti-glioma activity, encouraging further in vivo studies in order to better understand its mechanism of action.
Collapse
|
21
|
Gortany NK, Panahi G, Ghafari H, Shekari M, Ghazi-Khansari M. Foretinib induces G2/M cell cycle arrest, apoptosis, and invasion in human glioblastoma cells through c-MET inhibition. Cancer Chemother Pharmacol 2021; 87:827-842. [PMID: 33688998 DOI: 10.1007/s00280-021-04242-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/01/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is one of the most aggressive human cancers. The c-MET receptor tyrosine kinase (RTK) which is frequently deregulated in GBM is considered as a promising target for GBM treatment. The c-MET plays a key role in cell proliferation, cell cycle progression, invasion, angiogenesis, and metastasis. Here, we investigated the anti-tumour activity of foretinib, a c-MET inhibitor, on three human GBM cells (T98G, U87MG and U251). METHODS Anti-proliferative effect of foretinib was determined using MTT, crystal violet staining, and clonogenic assays. PI and Annexin V/PI staining flow cytometry were used to evaluate the effects of foretinib on cell cycle and apoptosis, respectively. Scratch assay, qRT-PCR, western blot, and zymography analyses were applied to elucidate the molecular mechanisms underlying the anti-tumour activity of foretinib. RESULTS Foretinib treatment reduced phosphorylation of c-MET on T98G and U251 cells, but not in U87MG cells. The highest inhibitory effect was observed in T98G cells (IC50 = 4.66 ± 0.29 µM) and the lowest one in U87MG cells (IC50 = 29.99 ± 1.31 µM). The results showed that foretinib inhibited the proliferation of GBM cells through a G2/M cell cycle arrest and mitochondrial-mediated apoptosis in association with alternation in expression of the related genes and protein-regulated G2/M phase and apoptosis. Foretinib diminished GBM cell invasion through downregulation of the proteolytic cascade of MMP2, uPA and uPAR and epithelial-mesenchymal transition (EMT)-related genes. A different GBM cell sensitivity pattern was noticeable in all experiments which demonstrated T98G as a sensitive and U87MG as a resistant phenotype to foretinib treatment. CONCLUSION The results indicated that foretinib might have the therapeutic potential against human GBM which deserve further investigation.
Collapse
Affiliation(s)
- Narges K Gortany
- Cancer Biology Research Center, Cancer Institute of I.R. Iran, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Homanaz Ghafari
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Maryam Shekari
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mahmoud Ghazi-Khansari
- Cancer Biology Research Center, Cancer Institute of I.R. Iran, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.
| |
Collapse
|
22
|
Yamada T, Tsuji S, Nakamura S, Egashira Y, Shimazawa M, Nakayama N, Yano H, Iwama T, Hara H. Riluzole enhances the antitumor effects of temozolomide via suppression of MGMT expression in glioblastoma. J Neurosurg 2021; 134:701-710. [PMID: 32168477 DOI: 10.3171/2019.12.jns192682] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/30/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Glutamatergic signaling significantly promotes proliferation, migration, and invasion in glioblastoma (GBM). Riluzole, a metabotropic glutamate receptor 1 inhibitor, reportedly suppresses GBM growth. However, the effects of combining riluzole with the primary GBM chemotherapeutic agent, temozolomide (TMZ), are unknown. This study aimed to investigate the efficacy of combinatorial therapy with TMZ/riluzole for GBM in vitro and in vivo. METHODS Three GBM cell lines, T98G (human; O6-methylguanine DNA methyltransferase [MGMT] positive), U87MG (human; MGMT negative), and GL261 (murine; MGMT positive), were treated with TMZ, riluzole, or a combination of both. The authors performed cell viability assays, followed by isobologram analysis, to evaluate the effects of combinatorial treatment for each GBM cell line. They tested the effect of riluzole on MGMT, a DNA repair enzyme causing chemoresistance to TMZ, through quantitative real-time reverse transcription polymerase chain reaction in T98G cells. Furthermore, they evaluated the efficacy of combinatorial TMZ/riluzole treatment in an orthotopic mouse allograft model of MGMT-positive GBM using C57BL/6 J mice and GL261 cells. RESULTS Riluzole displayed significant time- and dose-dependent growth-inhibitory effects on all GBM cell lines assessed independently. Riluzole enhanced the antitumor effect of TMZ synergistically in MGMT-positive but not in MGMT-negative GBM cell lines. Riluzole singularly suppressed MGMT expression, and it significantly suppressed TMZ-induced MGMT upregulation (p < 0.01). Furthermore, combinatorial TMZ/riluzole treatment significantly suppressed tumor growth in the intracranial MGMT-positive GBM model (p < 0.05). CONCLUSIONS Riluzole attenuates TMZ-induced MGMT upregulation and enhances the antitumor effect of TMZ in MGMT-positive GBMs. Therefore, combinatorial TMZ/riluzole treatment is a potentially promising novel therapeutic regimen for MGMT-positive GBMs.
Collapse
Affiliation(s)
- Tetsuya Yamada
- 1Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University; and
- 2Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shohei Tsuji
- 1Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University; and
| | - Shinsuke Nakamura
- 1Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University; and
| | - Yusuke Egashira
- 2Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masamitsu Shimazawa
- 1Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University; and
| | - Noriyuki Nakayama
- 2Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirohito Yano
- 2Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toru Iwama
- 2Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hideaki Hara
- 1Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University; and
| |
Collapse
|
23
|
Liu YC, Su CW, Ko PS, Lee RC, Liu CJ, Huang YH, Gau JP, Liu JH. A clinical trial with valproic acid and hydralazine in combination with gemcitabine and cisplatin followed by doxorubicin and dacarbazine for advanced hepatocellular carcinoma. Asia Pac J Clin Oncol 2020; 18:19-27. [PMID: 32964588 DOI: 10.1111/ajco.13443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Survival benefit from chemotherapy in advanced hepatocellular carcinoma (HCC) was limited till now. New chemoregimens with cytotoxicity modulators were explored to improve efficacy. Chemotherapy modulated with valproic acid (VA) as a deacetylation inhibitor of histone and DNA damage response proteins, and hydralazine (HZ) as a DNA hypomethylating agent, hypothetically suppressing DNA repair, were used in phase II trial here for advanced HCC. METHODS Between July 2008 and March 2016, patients with chemo-naive advanced HCC, regardless of previous sorafenib treatment, not amenable to local therapy and with Child Pugh score ≤7, were treated with VA (200 mg thrice per day) and HZ (12.5 mg twice per day) in conjunction with gemcitabine and cisplatin (GCGG): gemcitabine (1000 mg/m2 , D1; 800 mg/m2 D8, 15) and cisplatin (70 mg/m2 , D1) every 28 days till disease progression and then with Dox-DTIC: doxorubicin (45 mg/m2 ) and dacarbazine (450 mg/m2 ) every 28 days. The primary endpoint was overall survival (OS); the secondary endpoints were safety, progression-free survival (PFS) and response rate (RR). RESULTS Thirty-seven patients with 16 sorafenib-experienced, underwent GCGG treatment, and 30 of them underwent the following Dox-DTIC treatment. The median OS was 14.6 months (95% confidence interval: 6.0-23.1). The median PFSs for patients treated with VA- and HZ-combined GCGG and Dox-DTIC were 3.7 and 4.2 months, respectively; the RRs were 10/37 (27.0%) and 7/30 (23.3%); and grade 3/4 neutropenia were 54% and 51%. However, there were no chemotherapy-related deaths. CONCLUSION VA- and HZ-combined sequential chemotherapy was effective in advanced HCC with manageable toxicities.
Collapse
Affiliation(s)
- Yao-Chung Liu
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Wei Su
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Shen Ko
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Rheun-Chuan Lee
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Jen Liu
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Hsiang Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jyh-Pyng Gau
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jin-Hwang Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Hematology and Oncology, Cheng Hsin General Hospital, Taipei, Taiwan.,Chong Hin Loon Cancer and Biotherapy Research Center and Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
24
|
Valproic Acid Sensitizes Glioma Cells to Luteolin Through Induction of Apoptosis and Autophagy via Akt Signaling. Cell Mol Neurobiol 2020; 41:1625-1634. [PMID: 32719967 DOI: 10.1007/s10571-020-00930-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Glioma is a highly malignant type of intracranial tumor with a poor prognosis resulting from traditional chemo-resistance with temozolomide (TMZ). Luteolin has been detected to exert limited anti-tumor effects on gliomas, while valproic acid (VPA) is a common chemotherapy sensitizer in the treatment of tumors. In this study, three glioma cell lines including U251, LN229 and SNB19 were selected for evaluation of combined anti-tumor effects of VPA and luteolin via Cell Counting Kit-8 (CCK-8) assay, colony formation assay, wound-healing assay, flow cytometry and western blot assay. The results disclosed that VPA sensitized glioma cells to luteolin by repressing cell viability, colony formation and migration. Mechanically, VPA boosted cellular apoptosis and cell-cycle arrest by increased level of cleaved caspase-3/caspase-3, cleaved PARP/PARP and Bax/Bcl-2. In addition, VPA also facilitated cellular autophagy via the decline of p62, p-Akt/Akt and the accumulation of LC3-II. These findings suggested that VPA enhanced the anticancer effects of luteolin by strengthening apoptosis and autophagy via Akt signaling, which could be adopted as a novel therapy for glioma.
Collapse
|
25
|
Kuo YJ, Yang YH, Lee IY, Chen PC, Yang JT, Wang TC, Lin MHC, Yang WH, Cheng CY, Chen KT, Huang WC, Lee MH. Effect of valproic acid on overall survival in patients with high-grade gliomas undergoing temozolomide: A nationwide population-based cohort study in Taiwan. Medicine (Baltimore) 2020; 99:e21147. [PMID: 32664146 PMCID: PMC7360242 DOI: 10.1097/md.0000000000021147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High-grade gliomas (HGGs) are a rapidly progressive and highly recurrent group of primary brain tumors. Despite aggressive surgical resection with chemoradiotherapy, prognoses remained poor. Valproic acid (VPA), a histone deacetylase inhibitor has shown the potential to inhibit glioma cell growth in vitro through several diverse mechanisms. However clinical studies regarding the effect of VPA on HGGs are limited. This study aimed to investigate whether using VPA in patients with HGGs under temozolomide (TMZ) would lead to a better overall survival (OS).We used the Taiwan National Health Insurance Research database to conduct this population-based cohort study. A total of 2379 patients with HGGs under TMZ treatment were included and were further classified into VPA (n = 1212, VPA ≥ 84 defined daily dose [DDD]) and non-VPA (n = 1167, VPA < 84 DDD) groups. Each patient was followed from 1998 to 2013 or until death. A Cox proportional hazard regression was performed to evaluate the effect of VPA and OS.The VPA group had a longer mean OS time compared with the non-VPA group (OS: 50.3 ± 41.0 vs 42.0 ± 37.2 months, P < .001). In patients between 18 and 40 years old, the difference is most significant (OS: 70.5 ± 48.7 vs 55.1 ± 46.0, P = .001). The adjusted hazard ratio is 0.81 (95% confidence interval, 0.72-0.91) for the VPA group relative to the non-VPA group.VPA at over 84 DDD improved OS in HGGs TMZ treatment.
Collapse
Affiliation(s)
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital
- Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan
| | - I-Yun Lee
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital
| | - Pau-Chung Chen
- Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei
| | - Jen-Tsung Yang
- Department of Neurosurgery
- Chang Gung University, College of Medicine, Taoyuan
| | | | | | | | | | | | | | - Ming-Hsueh Lee
- Department of Neurosurgery
- Chang Gung University of Science and Technology Chiayi Campus, Chiayi, Taiwan
| |
Collapse
|
26
|
Roh TH, Moon JH, Park HH, Kim EH, Hong CK, Kim SH, Kang SG, Chang JH. Association between survival and levetiracetam use in glioblastoma patients treated with temozolomide chemoradiotherapy. Sci Rep 2020; 10:10783. [PMID: 32612203 PMCID: PMC7330022 DOI: 10.1038/s41598-020-67697-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/29/2020] [Indexed: 12/22/2022] Open
Abstract
This study was conducted to assess whether levetiracetam (LEV) affects the survival of patients with glioblastoma (GBM) treated with concurrent temozolomide (TMZ) chemotherapy. To this end, from 2004 to 2016, 322 patients with surgically resected and pathologically confirmed isocitrate dehydrogenase (IDH)-wildtype GBM who received TMZ-based chemoradiotherapy were analysed. The patients were divided into two groups based on whether LEV was used as an anticonvulsant both at the time of surgery and the first visit thereafter. The median overall survival (OS) and progression-free survival (PFS) were compared between the groups. The OS was 21.1 and 17.5 months in the LEV (+) and LEV (−) groups, respectively (P = 0.003); the corresponding PFS was 12.3 and 11.2 months (P = 0.017). The other prognostic factors included age, extent of resection, O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status, and Karnofsky Performance Status (KPS) score. The multivariate analysis showed age (hazard ratio [HR], 1.02; P < 0.001), postoperative KPS score (HR 0.99; P = 0.002), complete tumour resection (HR 0.52; P < 0.001), MGMT promoter methylation (HR 0.75; P < 0.001), and LEV use (HR 0.72; P = 0.011) were significantly associated with OS. In conclusion, LEV use was associated with prolonged survival in patients with GBM treated with concurrent TMZ chemoradiotherapy.
Collapse
Affiliation(s)
- Tae Hoon Roh
- Department of Neurosurgery, Ajou University Hospital, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ju Hyung Moon
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hun Ho Park
- Department of Neurosurgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chang-Ki Hong
- Department of Neurosurgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
27
|
Petővári G, Dankó T, Krencz I, Hujber Z, Rajnai H, Vetlényi E, Raffay R, Pápay J, Jeney A, Sebestyén A. Inhibition of Metabolic Shift can Decrease Therapy Resistance in Human High-Grade Glioma Cells. Pathol Oncol Res 2020; 26:23-33. [PMID: 31187466 PMCID: PMC7109188 DOI: 10.1007/s12253-019-00677-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
The high-grade brain malignancy, glioblastoma multiforme (GBM), is one of the most aggressive tumours in central nervous system. The developing resistance against recent therapies and the recurrence rate of GBMs are extremely high. In spite several new ongoing trials, GBM therapies could not significantly increase the survival rate of the patients as significantly. The presence of inter- and intra-tumoral heterogeneity of GBMs arise the problem to find both the pre-existing potential resistant clones and the cellular processes which promote the adaptation mechanisms such as multidrug resistance, stem cell-ness or metabolic alterations, etc. In our work, the in situ metabolic heterogeneity of high-grade human glioblastoma cases were analysed by immunohistochemistry using tissue-microarray. The potential importance of the detected metabolic heterogeneity was tested in three glioma cell lines (grade III-IV) using protein expression analyses (Western blot and WES Simple) and therapeutic drug (temozolomide), metabolic inhibitor treatments (including glutaminase inhibitor) to compare the effects of rapamycin (RAPA) and glutaminase inhibitor combinations in vitro (Alamar Blue and SRB tests). The importance of individual differences and metabolic alterations were observed in mono-therapeutic failures, especially the enhanced Rictor expressions after different mono-treatments in correlation to lower sensitivity (temozolomide, doxycycline, etomoxir, BPTES). RAPA combinations with other metabolic inhibitors were the best strategies except for RAPA+glutaminase inhibitor. These observations underline the importance of multi-targeting metabolic pathways. Finally, our data suggest that the detected metabolic heterogeneity (the high mTORC2 complex activity, enhanced expression of Rictor, p-Akt, p-S6, CPT1A, and LDHA enzymes in glioma cases) and the microenvironmental or treatment induced metabolic shift can be potential targets in combination therapy. Therefore, it should be considered to map tissue heterogeneity and alterations with several cellular metabolism markers in biopsy materials after applying recently available or new treatments.
Collapse
Affiliation(s)
- Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Ildikó Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Zoltán Hujber
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Hajnalka Rajnai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Enikő Vetlényi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Regina Raffay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Judit Pápay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - András Jeney
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary.
| |
Collapse
|
28
|
Chen JC, Lee IN, Huang C, Wu YP, Chung CY, Lee MH, Lin MHC, Yang JT. Valproic acid-induced amphiregulin secretion confers resistance to temozolomide treatment in human glioma cells. BMC Cancer 2019; 19:756. [PMID: 31370819 PMCID: PMC6670223 DOI: 10.1186/s12885-019-5843-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/16/2019] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most severe type of primary brain tumor with a high mortality rate. Although extensive treatments for GBM, including resection, irradiation, chemotherapy and immunotherapy, have been tried, the prognosis is still poor. Temozolomide (TMZ), an alkylating agent, is a front-line chemotherapeutic drug for the clinical treatment of GBM; however, its effects are very limited because of the chemoresistance. Valproic acid (VPA), an antiepileptic agent with histone deacetylase inhibitor activity, has been shown to have synergistic effects with TMZ against GBM. The mechanism of action of VPA on TMZ combination therapy is still unclear. Accumulating evidence has shown that secreted proteins are responsible for the cross talking among cells in the tumor microenvironment, which may play a critical role in the regulation of drug responses. METHODS To understand the effect of VPA on secreted proteins in GBM cells, we first used the antibody array to analyze the cell culture supernatant from VPA-treated and untreated GBM cells. The results were further confirmed by lentivirus-mediated knockdown and exogenous recombinant administration. RESULTS Our results showed that amphiregulin (AR) was highly secreted in VPA-treated cells. Knockdown of AR can sensitize GBM cells to TMZ. Furthermore, pretreatment of exogenous recombinant AR significantly increased EGFR activation and conferred resistance to TMZ. To further verify the effect of AR on TMZ resistance, cells pre-treated with AR neutralizing antibody markedly increased sensitivity to TMZ. In addition, we also observed that the expression of AR was positively correlated with the resistance of TMZ in different GBM cell lines. CONCLUSIONS The present study aimed to identify the secreted proteins that contribute to the modulation of drug response. Understanding the full set of secreted proteins present in glial cells might help reveal potential therapeutic opportunities. The results indicated that AR may potentially serve as biomarker and therapeutic approach for chemotherapy regimens in GBM.
Collapse
Affiliation(s)
- Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi City, 60004 Taiwan
| | - I-Neng Lee
- Department of Medical Research, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Cheng Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Earth and Life Sciences, University of Taipei, Taipei, Taiwan
| | - Yu-Ping Wu
- Department of Medical Research, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Chiu-Yen Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Ming-Hsueh Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Martin Hsiu-Chu Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
- College of Medicine, Chang Gung University, Tao-Yuan, 33302 Taiwan
| |
Collapse
|
29
|
Zhu Y, Zhao YF, Liu RS, Xiong YJ, Shen X, Wang Y, Liang ZQ. Olanzapine induced autophagy through suppression of NF-κB activation in human glioma cells. CNS Neurosci Ther 2019; 25:911-921. [PMID: 30955240 PMCID: PMC6698966 DOI: 10.1111/cns.13127] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/06/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022] Open
Abstract
Aims Our laboratory previously reported that olanzapine treatment inhibited growth of glioma cell lines and hypothesized that autophagy may be involved in the proliferation inhibitory effects of olanzapine. However, the mechanisms of olanzapine‐contributed autophagy activation are unclear. Methods The inhibitory effects of olanzapine on glioma cells were evaluated by CCK8 assay, Hoechst 33258 staining and annexin V‐FITC/PI staining. Western blotting, nuclear separation techniques, and immunofluorescence assays were used to investigate the relationship between the inhibition of NF‐κB and autophagy activation by olanzapine. Results In this work, we verified that olanzapine increased autophagic flux and autophagic vesicles. In addition, we confirmed that autophagy was related to NF‐κB inhibition in cancer progression, especially with the nuclear translocation of p65. Furthermore, we demonstrated that autophagy induced by olanzapine could be impaired with TNFα cotreatment. We also found that olanzapine had an inhibitory effect on T98 cells with positive MGMT protein expression, which may involve the inhibition of MGMT through effects on NF‐κB. Conclusions Our findings identify a pathway by which olanzapine induces autophagy by depressing NF‐κB in a glioma cell line, providing evidence which supports the use of olanzapine as a potential anticancer drug.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Fan Zhao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Rui-Si Liu
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ya-Jie Xiong
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiao Shen
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yan Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhong-Qin Liang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
30
|
Gopal J, Lee YM, Shin J, Muthu M, Jung S, Jeong S, Oh J, Oh JW. The graviola impact on human astroglioma cells: functional significance of MUDENG. RSC Adv 2019; 9:8935-8942. [PMID: 35517667 PMCID: PMC9062117 DOI: 10.1039/c8ra10039j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/24/2019] [Indexed: 11/21/2022] Open
Abstract
Graviola (Annona muricate) is a coveted tropical plant that has been found to be effective against many human cancers. Malignant glioblastoma multiformes are the most common and aggressive malignant forms of astrocytoma in the central nervous system. MUDENG (Mu-2-related death-inducing gene, MuD) is involved in cell death signaling. In this study, we investigated the impact of extracts from graviola leaves (from Korea and Africa), fruits and seeds against human astroglioma cells. Interestingly, graviola leaf extract-Korea (GLE-K), graviola leaf extract-Africa (GLE-A) and graviola fruit extract-Africa (GFE-A) exhibited significant cytotoxic effects on the cell proliferation in a dose-dependent manner and altered the MuD expression pattern. Cell cycle analyses revealed that GLE-A and GLE-K triggered no significant induction of apoptosis at concentrations up to 5% in U251-MG cells, while in GLE-K treated cells at 10% concentrations, there were much fewer apoptotic cells (33.64%) compared to those in GLE-A (73.55%) treated cells. In the case of GFE-A treated cells, 5% graviola extract (GE) concentration resulted in predominant cells entering the apoptotic phase (59.31%), whereas almost no significant increase in apoptotic cells was observed in GSE-A treated cells (1.38%) even up to 25% of graviola extract (GE) concentration. While using stable transfectants knock-out (KO)(-)-and overexpressing (OE)-MuD(+), significant and consistent differences in the cell viability (enhanced anti-astroglioma effect of GEs) were observed in KO-MuD(-) cells. This validated the functional consequence of MuD in the anti-astroglioma activity of GEs. Our results confirmed that GFE-A possesses the highest anti-astroglioma activity followed by the leaf extracts (GLE-A/K). This is the first report that highlights the MuD aspect of GEs.
Collapse
Affiliation(s)
- Judy Gopal
- Department of Environmental Health Sciences, Konkuk University Seoul 05029 Korea
| | - Yoon-Mi Lee
- Department of Stem Cell and Regenerative Biotechnology, Department of Animal Biotechnology, Konkuk University 120 Neungdong-ro, Gwangjin-gu Seoul 05029 Korea +82-2-455-1044 +82-2-2049-6271
| | - Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology, Department of Animal Biotechnology, Konkuk University 120 Neungdong-ro, Gwangjin-gu Seoul 05029 Korea +82-2-455-1044 +82-2-2049-6271
| | - Manikandan Muthu
- Department of Environmental Health Sciences, Konkuk University Seoul 05029 Korea
| | - Seunghwa Jung
- Department of Stem Cell and Regenerative Biotechnology, Department of Animal Biotechnology, Konkuk University 120 Neungdong-ro, Gwangjin-gu Seoul 05029 Korea +82-2-455-1044 +82-2-2049-6271
| | - Somi Jeong
- Department of Stem Cell and Regenerative Biotechnology, Department of Animal Biotechnology, Konkuk University 120 Neungdong-ro, Gwangjin-gu Seoul 05029 Korea +82-2-455-1044 +82-2-2049-6271
| | - Jeongheon Oh
- Department of Stem Cell and Regenerative Biotechnology, Department of Animal Biotechnology, Konkuk University 120 Neungdong-ro, Gwangjin-gu Seoul 05029 Korea +82-2-455-1044 +82-2-2049-6271
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Department of Animal Biotechnology, Konkuk University 120 Neungdong-ro, Gwangjin-gu Seoul 05029 Korea +82-2-455-1044 +82-2-2049-6271
| |
Collapse
|
31
|
Lange F, Weßlau K, Porath K, Hörnschemeyer MF, Bergner C, Krause BJ, Mullins CS, Linnebacher M, Köhling R, Kirschstein T. AMPA receptor antagonist perampanel affects glioblastoma cell growth and glutamate release in vitro. PLoS One 2019; 14:e0211644. [PMID: 30716120 PMCID: PMC6361447 DOI: 10.1371/journal.pone.0211644] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/17/2019] [Indexed: 12/17/2022] Open
Abstract
Epileptic seizures are frequent in patients with glioblastoma, and anticonvulsive treatment is often necessary. While clinical guidelines recommend all approved anticonvulsants, so far it is still unclear which of the available drugs is the best therapeutic option for treating glioma-associated seizures, also in view of possible anti-tumorigenic effects. In our study, we employed four patient-derived low-passage cell lines of glioblastoma and three cell lines of brain metastases, and challenged these cultures with four anticonvulsants with different mechanisms of action: levetiracetam, valproic acid, carbamazepine and perampanel. Cell proliferation was determined by bromodeoxyuridine incorporation. To further analyze the effects of perampanel, apoptosis induction was measured by caspase 3/7 activation. Glutamate release was quantified and glucose uptake was determined using 18F-fluorodeoxyglucose. Real-time polymerase chain reaction was employed to assess the expression of genes associated with glutamate release and uptake in brain tumor cells. Of the four anticonvulsants, only perampanel showed systematic inhibitory effects on cell proliferation, whereas all other anticonvulsants failed to inhibit glioma and metastasis cell growth in vitro. Metastasis cells were much more resistant to perampanel than glioblastoma cell lines. Glucose uptake was attenuated in all glioblastoma cells after perampanel exposure, whereas cell death via apoptosis was not induced. Extracellular glutamate levels were found to be significantly higher in glioblastoma cell lines as compared to metastasis cell lines, but could be reduced by perampanel exposure. Incubation with perampanel up-regulated glutamine synthetase expression in glioblastoma cells, whereas treatment with valproic acid and levetiracetam downregulated excitatory amino acid transporter-2 expression. Overall, our data suggest that perampanel acts as an anticonvulsive drug and additionally mediated anti-tumorigenic effects.
Collapse
Affiliation(s)
- Falko Lange
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Konrad Weßlau
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Katrin Porath
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | | | - Carina Bergner
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Bernd Joachim Krause
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | | | | | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| |
Collapse
|
32
|
Petővári G, Hujber Z, Krencz I, Dankó T, Nagy N, Tóth F, Raffay R, Mészáros K, Rajnai H, Vetlényi E, Takács-Vellai K, Jeney A, Sebestyén A. Targeting cellular metabolism using rapamycin and/or doxycycline enhances anti-tumour effects in human glioma cells. Cancer Cell Int 2018; 18:211. [PMID: 30574020 PMCID: PMC6300020 DOI: 10.1186/s12935-018-0710-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022] Open
Abstract
Background Glioma is the most common highly aggressive, primary adult brain tumour. Clinical data show that therapeutic approaches cannot reach the expectations in patients, thus gliomas are mainly incurable diseases. Tumour cells can adapt rapidly to alterations during therapeutic treatments related to their metabolic rewiring and profound heterogeneity in tissue environment. Renewed interests aim to develop effective treatments targeting angiogenesis, kinase activity and/or cellular metabolism. mTOR (mammalian target of rapamycin), whose hyper-activation is characteristic for many tumours, promotes metabolic alterations, macromolecule biosynthesis, cellular growth and survival. Unfortunately, mTOR inhibitors with their lower toxicity have not resulted in appreciable survival benefit. Analysing mTOR inhibitor sensitivity, other metabolism targeting treatments and their combinations could help to find potential agents and biomarkers for therapeutic development in glioma patients. Methods In vitro proliferation assays, protein expression and metabolite concentration analyses were used to study the effects of mTOR inhibitors, other metabolic treatments and their combinations in glioma cell lines. Furthermore, mTOR activity and cellular metabolism related protein expression patterns were also investigated by immunohistochemistry in human biopsies. Temozolomide and/or rapamycin treatments altered the expressions of enzymes related to lipid synthesis, glycolysis and mitochondrial functions as consequences of metabolic adaptation; therefore, other anti-metabolic drugs (chloroquine, etomoxir, doxycycline) were combined in vitro. Results Our results suggest that co-targeting metabolic pathways had tumour cell dependent additive/synergistic effects related to mTOR and metabolic protein expression patterns cell line dependently. Drug combinations, especially rapamycin + doxycycline may have promising anti-tumour effect in gliomas. Additionally, our immunohistochemistry results suggest that metabolic and mTOR activity alterations are not related to the recent glioma classification, and these protein expression profiles show individual differences in patients’ materials. Conclusions Based on these, combinations of different new/old drugs targeting cellular metabolism could be promising to inhibit high adaptation capacity of tumour cells depending on their metabolic shifts. Relating to this, such a development of current therapy needs to find special biomarkers to characterise metabolic heterogeneity of gliomas.
Collapse
Affiliation(s)
- Gábor Petővári
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Zoltán Hujber
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Ildikó Krencz
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Titanilla Dankó
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Noémi Nagy
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Fanni Tóth
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Regina Raffay
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Katalin Mészáros
- 2Hungarian Academy of Sciences-Momentum Hereditary Endocrine Tumours Research Group, Semmelweis University-National Bionics Program Budapest, Üllői út 26, Budapest, 1085 Hungary
| | - Hajnalka Rajnai
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Enikő Vetlényi
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Krisztina Takács-Vellai
- 3Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest, 1117 Hungary
| | - András Jeney
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| | - Anna Sebestyén
- 11st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085 Hungary
| |
Collapse
|
33
|
Ciechomska IA, Marciniak MP, Jackl J, Kaminska B. Pre-treatment or Post-treatment of Human Glioma Cells With BIX01294, the Inhibitor of Histone Methyltransferase G9a, Sensitizes Cells to Temozolomide. Front Pharmacol 2018; 9:1271. [PMID: 30450051 PMCID: PMC6224489 DOI: 10.3389/fphar.2018.01271] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/18/2018] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is a malignant, primary brain tumor, highly resistant to conventional therapies. Temozolomide (TMZ) is a first line therapeutic agent in GBM patients, however, survival of such patients is poor. High level of DNA repair protein, O6-methylguanine-DNA-methyltransferase (MGMT) and occurrence of glioma stem-like cells contribute to GBM resistance to the drug. Here, we explored a possibility of epigenetic reprograming of glioma cells to increase sensitivity to TMZ and restore apoptosis competence. We combined TMZ treatment with BIX01294, an inhibitor of histone methyltransferase G9a, known to be involved in cancerogenesis. Two treatment combinations were tested: BIX01294 was administered to human LN18 and U251 glioma cell cultures 48 h before TMZ or 48 h after TMZ treatment. Despite their different status of the MGMT gene promoter, there was no correlation with the response to TMZ. The analyses of cell viability, appearance of apoptotic alterations in morphology of cells and nuclei, and markers of apoptosis, such as levels of cleaved caspase 3, caspase 7 and PARP, revealed that both pre-treatment and post-treatment with BIX01294 sensitize glioma cells to TMZ. The additive effect was stronger in LN18 cells. Moreover, BIX01294 enhanced the cytotoxic effect of TMZ on glioma stem-like cells, although it was not associated with modulation of the pluripotency markers (NANOG, SOX2, CD133) expression or methylation of NANOG and SOX2 gene promoters. Accordingly, knockdown of methyltransferase G9a augments TMZ-induced cell death in LN18 cells. We found the significant increases of the LC3-II levels in LN18 cells treated with BIX01294 alone and with drug combination that suggests involvement of autophagy in enhancement of anti-tumor effect of TMZ. Treatment with BIX01294 did not affect methylation of the MGMT gene promoter. Altogether, our results suggest that G9a is a potential therapeutic target in malignant glioma and the treatment with the G9a inhibitor reprograms glioma cells and glioma stem-like cells to increase sensitivity to TMZ and restore apoptosis competence.
Collapse
Affiliation(s)
- Iwona Anna Ciechomska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marta Patrycja Marciniak
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Judyta Jackl
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
34
|
da Silva VD, de Faria BM, Colombo E, Ascari L, Freitas GPA, Flores LS, Cordeiro Y, Romão L, Buarque CD. Design, synthesis, structural characterization and in vitro evaluation of new 1,4-disubstituted-1,2,3-triazole derivatives against glioblastoma cells. Bioorg Chem 2018; 83:87-97. [PMID: 30343205 DOI: 10.1016/j.bioorg.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/25/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022]
Abstract
A new series of 1,4-disubstituted-1,2,3-triazole derivatives were synthesized through the copper-catalyzed azide-alkyne 1,3-dipolar cycloaddition (Click chemistry) and their inhibitory activities were evaluated against different human glioblastoma (GBM) cell lines, including highly drug-resistant human cell lines GBM02, GBM95. The most effective compounds were 9d, containing the methylenoxy moiety linked to triazole and the tosyl-hydrazone group, and the symmetrical bis-triazole 10a, also containing methylenoxy moiety linked to triazole. Single crystal X-ray diffraction analysis was employed for structural elucidation of compound 9d. In silico analyses of physicochemical, pharmacokinetic, and toxicological properties suggest that compounds 8a, 8b, 8c, 9d, and 10a are potential candidates for central nervous system-acting drugs.
Collapse
Affiliation(s)
- Veronica D da Silva
- Laboratório de Síntese orgânica, Pontifícia Universidade Católica do Rio de Janeiro, 22451-900 Rio de Janeiro, RJ, Brazil
| | - Bruna M de Faria
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Eduardo Colombo
- Laboratório de Síntese orgânica, Pontifícia Universidade Católica do Rio de Janeiro, 22451-900 Rio de Janeiro, RJ, Brazil
| | - Lucas Ascari
- Faculdade de Farmácia, UFRJ, RJ 21941-902, Brazil
| | - Gabriella P A Freitas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Leonã S Flores
- Laboratório de Difração de raios X, UFJF, MG 36036-900, Brazil
| | | | - Luciana Romão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Camilla D Buarque
- Laboratório de Síntese orgânica, Pontifícia Universidade Católica do Rio de Janeiro, 22451-900 Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
35
|
Kim TW, Lee SY, Kim M, Cheon C, Ko SG. Kaempferol induces autophagic cell death via IRE1-JNK-CHOP pathway and inhibition of G9a in gastric cancer cells. Cell Death Dis 2018; 9:875. [PMID: 30158521 PMCID: PMC6115440 DOI: 10.1038/s41419-018-0930-1] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/11/2018] [Accepted: 07/25/2018] [Indexed: 01/19/2023]
Abstract
Kaempferol, a flavonoid, found in traditional medicine, fruits, and vegetables, and an HDAC inhibitor, is a powerful anti-cancer reagent against various cancer cell lines. However, detailed mechanisms involved in the treatment of gastric cancer (GC) using kaempferol are not fully understood. In our study, we investigated the biological activity and molecular mechanism involved in kaempferol-mediated treatment of GC. Kaempferol promoted autophagy and cell death, and increased LC3-I to LC3-II conversion and the downregulation of p62 in GC. Furthermore, our results showed that kaempferol induces autophagic cell death via the activation of the IRE1-JNK-CHOP signaling, indicating ER stress response. Indeed, the inhibition of ER stress suppressed kaempferol-induced autophagy and conferred prolonged cell survival, indicating autophagic cell death. We further showed that kaempferol mediates epigenetic change via the inhibition of G9a (HDAC/G9a axis) and also activates autophagic cell death. Taken together, our findings indicate that kaempferol activates the IRE1-JNK-CHOP signaling from cytosol to nucleus, and G9a inhibition activates autophagic cell death in GC cells.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Seon Young Lee
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Mia Kim
- Department of Cardiovascular and Neurologic disease (Stroke center), College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Chunhoo Cheon
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea.
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
36
|
Hoja S, Schulze M, Rehli M, Proescholdt M, Herold-Mende C, Hau P, Riemenschneider MJ. Molecular dissection of the valproic acid effects on glioma cells. Oncotarget 2018; 7:62989-63002. [PMID: 27556305 PMCID: PMC5325342 DOI: 10.18632/oncotarget.11379] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/12/2016] [Indexed: 11/25/2022] Open
Abstract
Many glioblastoma patients suffer from seizures why they are treated with antiepileptic agents. Valproic acid (VPA) is a histone deacetylase inhibitor that apart from its anticonvulsive effects in some retrospective studies has been suggested to lead to a superior outcome of glioblastoma patients. However, the exact molecular effects of VPA treatment on glioblastoma cells have not yet been deciphered. We treated glioblastoma cells with VPA, recorded the functional effects of this treatment and performed a global and unbiased next generation sequencing study on the chromatin (ChIP) and RNA level. 1) VPA treatment clearly sensitized glioma cells to temozolomide: A protruding VPA-induced molecular feature in this context was the transcriptional upregulation/reexpression of numerous solute carrier (SLC) transporters that was also reflected by euchromatinization on the histone level and a reexpression of SLC transporters in human biopsy samples after VPA treatment. DNA repair genes were adversely reduced. 2) VPA treatment, however, also reduced cell proliferation in temozolomide-naive cells: On the molecular level in this context we observed a transcriptional upregulation/reexpression and euchromatinization of several glioblastoma relevant tumor suppressor genes and a reduction of stemness markers, while transcriptional subtype classification (mesenchymal/proneural) remained unaltered. Taken together, these findings argue for both temozolomide-dependent and -independent effects of VPA. VPA might increase the uptake of temozolomide and simultaneously lead to a less malignant glioblastoma phenotype. From a mere molecular perspective these findings might indicate a surplus value of VPA in glioblastoma therapy and could therefore contribute an additional ratio for clinical decision making.
Collapse
Affiliation(s)
- Sabine Hoja
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Markus Schulze
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Michael Rehli
- Department of Internal Medicine III, Regensburg University Hospital, Regensburg, Germany.,RCI Regensburg Centre for Interventional Immunology, Regensburg University Hospital, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany.,Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Christel Herold-Mende
- Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Peter Hau
- Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany.,Department of Neurology, Regensburg University, Regensburg, Germany
| | - Markus J Riemenschneider
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany.,Wilhelm Sander Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
37
|
Li Z, Xia Y, Bu X, Yang D, Yuan Y, Guo X, Zhang G, Wang Z, Jiao J. Effects of valproic acid on the susceptibility of human glioma stem cells for TMZ and ACNU. Oncol Lett 2018; 15:9877-9883. [PMID: 29805689 PMCID: PMC5958707 DOI: 10.3892/ol.2018.8551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 02/23/2018] [Indexed: 11/27/2022] Open
Abstract
To investigate the effect of valproic acid (VPA) on the susceptibility of glioma stem cells to temozolomide (TMZ) and nimustine (ACNU), the O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and its expression of MGMT were examined. A total of 3 glioma cell populations were isolated from human glioma tissues, and immunocytochemistry was used to detect the expression of MGMT. VPA inhibition on the growth of the 3 glioma cell populations exposed to various concentrations of TMZ and ACNU was evaluated. Flow cytometry was applied to detect the apoptosis of glioma cells, and a methylation-specific polymerase chain reaction was used to identify methylation of MGMT promoter. Immunocytochemistry results indicated that MGMT was negatively expressed in the G1 population, but positively expressed in the G2 and G3 populations. Cell growth inhibition assays demonstrated that the survival rate in the VPA + TMZ or ACNU groups was decreased compared with that of the TMZ or ACNU alone groups (P<0.05). As for the apoptotic rate, those in the VPA alone group were increased compared with the control group (P<0.05), and the rates in the VPA + TMZ or ACNU groups were increased compared with TMZ or ACNU alone groups (P<0.05). The expression of MGMT remained negative in the G1 population following treatment with VPA, but MGMT expression became negative in the 2 MGMT-positive cell populations (G2 and G3) following VPA treatment. The MGMT promoter in the G1 population was partially methylated in the control group, but was fully methylated following VPA treatment, while the promoters of G2, G3 were unmethylated in the control group and became partially methylated in the VPA treatment group. Taken together, TMZ and ACNU may suppress the growth of glioma stem cells in vitro in a dose-dependent manner. VPA may enhance the inhibitory effects of various concentrations of TMZ and ACNU on the growth of MGMT-negative/positive cells, particularly on MGMT-positive cell populations. VPA itself may induce the apoptosis of glioma cells, and VPA combined with TMZ or ACNU may enhance TMZ/ACNU-induced apoptosis of glioma stem cells. Furthermore, VPA may also promote the methylation of the MGMT promoter to silence MGMT expression in glioma cells, which may be an important mechanism through which VPA enhances the efficacy of TMZ and ACNU in targeting glioma stem cells.
Collapse
Affiliation(s)
- Zhiying Li
- Department of Neurosurgery, Zhengzhou No. 7 People's Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Yun Xia
- Department of Microbiology and Immunology, Zhengzhou Health School, Zhengzhou, Henan 450000, P.R. China
| | - Xingyao Bu
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Dongyi Yang
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yiqiang Yuan
- Department of Neurosurgery, Zhengzhou No. 7 People's Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Xiaohe Guo
- Department of Neurosurgery, Zhengzhou No. 7 People's Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Gangzhong Zhang
- Department of Neurosurgery, Zhengzhou No. 7 People's Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Zhanwei Wang
- Department of Neurosurgery, Zhengzhou No. 7 People's Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Jichao Jiao
- Department of Neurosurgery, Zhengzhou No. 7 People's Hospital, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
38
|
Festuccia C, Mancini A, Colapietro A, Gravina GL, Vitale F, Marampon F, Delle Monache S, Pompili S, Cristiano L, Vetuschi A, Tombolini V, Chen Y, Mehrling T. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma. J Hematol Oncol 2018; 11:32. [PMID: 29486795 PMCID: PMC5830080 DOI: 10.1186/s13045-018-0576-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/14/2018] [Indexed: 12/14/2022] Open
Abstract
Background The use of alkylating agents such as temozolomide in association with radiotherapy (RT) is the therapeutic standard of glioblastoma (GBM). This regimen modestly prolongs overall survival, also if, in light of the still dismal prognosis, further improvements are desperately needed, especially in the patients with O6-methylguanine-DNA-methyltransferase (MGMT) unmethylated tumors, in which the benefit of standard treatment is less. Tinostamustine (EDO-S101) is a first-in-class alkylating deacetylase inhibitor (AK-DACi) molecule that fuses the DNA damaging effect of bendamustine with the fully functional pan-histone deacetylase (HDAC) inhibitor, vorinostat, in a completely new chemical entity. Methods Tinostamustine has been tested in models of GBM by using 13 GBM cell lines and seven patient-derived GBM proliferating/stem cell lines in vitro. U87MG and U251MG (MGMT negative), as well as T98G (MGMT positive), were subcutaneously injected in nude mice, whereas luciferase positive U251MG cells and patient-derived GBM stem cell line (CSCs-5) were evaluated the orthotopic intra-brain in vivo experiments. Results We demonstrated that tinostamustine possesses stronger antiproliferative and pro-apoptotic effects than those observed for vorinostat and bendamustine alone and similar to their combination and irrespective of MGMT expression. In addition, we observed a stronger radio-sensitization of single treatment and temozolomide used as control due to reduced expression and increased time of disappearance of γH2AX indicative of reduced signal and DNA repair. This was associated with higher caspase-3 activation and reduction of RT-mediated autophagy. In vivo, tinostamustine increased time-to-progression (TTP) and this was additive/synergistic to RT. Tinostamustine had significant therapeutic activity with suppression of tumor growth and prolongation of DFS (disease-free survival) and OS (overall survival) in orthotopic intra-brain models that was superior to bendamustine, RT and temozolomide and showing stronger radio sensitivity. Conclusions Our data suggest that tinostamustine deserves further investigation in patients with glioblastoma. Electronic supplementary material The online version of this article (10.1186/s13045-018-0576-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudio Festuccia
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy.
| | - Andrea Mancini
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Luca Gravina
- Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy.,Division of Radiotherapy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Flora Vitale
- Division of Neurosciences, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Francesco Marampon
- Division of Radiotherapy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Simona Delle Monache
- Division of Applied Biology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Simona Pompili
- Division of Human Anatomy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Laboratory of Applied Biology, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonella Vetuschi
- Division of Human Anatomy, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, L'Aquila, Italy
| | - Vincenzo Tombolini
- Division of Radiotherapy, Department of Experimental Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Yi Chen
- Northlake International LLC, Pleasanton, CA, USA
| | | |
Collapse
|
39
|
Toren A, Pismenyuk T, Yalon M, Freedman S, Simon AJ, Fisher T, Moshe I, Reichardt JKV, Constantini S, Mardor Y, Last D, Guez D, Daniels D, Assoulin M, Mehrian-Shai R. Zinc enhances temozolomide cytotoxicity in glioblastoma multiforme model systems. Oncotarget 2018; 7:74860-74871. [PMID: 27556862 PMCID: PMC5342707 DOI: 10.18632/oncotarget.11382] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/19/2016] [Indexed: 11/26/2022] Open
Abstract
Temozolomide (TMZ) is an alkylating agent that has become the mainstay treatment of the most malignant brain cancer, glioblastoma multiforme (GBM). Unfortunately only a limited number of patients positively respond to it. It has been shown that zinc metal reestablishes chemosensitivity but this effect has not been tested with TMZ. Using both in vitro and in vivo experimental approaches, we investigated whether addition of zinc to TMZ enhances its cytotoxicity against GBM. In vitro cell viability analysis showed that the cytotoxic activity of TMZ was substantially increased with addition of zinc and this response was accompanied by an elevation of p21, PUMA, BAX and Caspase-3 expression and a decrease in growth fraction as manifested by low ki67 and lower colony formation. Analysis of GBM as intracranial xenografts in athymic mice and administration of concurrent TMZ and zinc yielded results consistent with those of the in vitro analyses. The co-treatment resulted in significant reduction in tumor volume in TMZ/zinc treated mice relative to treatment with TMZ alone. Our results suggest that zinc may serve as a potentiator of TMZ therapy in GBM patients.
Collapse
Affiliation(s)
- Amos Toren
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Tatyana Pismenyuk
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Michal Yalon
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Shani Freedman
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Amos J Simon
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Tamar Fisher
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Itai Moshe
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | | | - Shlomi Constantini
- Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel-Aviv-Sourasky Medical Center, Israel
| | - Yael Mardor
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - David Last
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - David Guez
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Dianne Daniels
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Moria Assoulin
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ruty Mehrian-Shai
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Tel Hashomer Affiliated to The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Miranda A, Blanco-Prieto MJ, Sousa J, Pais A, Vitorino C. Breaching barriers in glioblastoma. Part II: Targeted drug delivery and lipid nanoparticles. Int J Pharm 2017; 531:389-410. [DOI: 10.1016/j.ijpharm.2017.07.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 02/07/2023]
|
41
|
Wang HH, Chang TY, Lin WC, Wei KC, Shin JW. GADD45A plays a protective role against temozolomide treatment in glioblastoma cells. Sci Rep 2017; 7:8814. [PMID: 28821714 PMCID: PMC5562912 DOI: 10.1038/s41598-017-06851-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive cancers. Despite recent advances in multimodal therapies, high-grade glioma remains fatal. Temozolomide (TMZ) is an alkylating agent used worldwide for the clinical treatment of GBM; however, the innate and acquired resistance of GBM limits its application. Here, we found that TMZ inhibited the proliferation and induced the G2/M arrest of GBM cells. Therefore, we performed microarrays to identify the cell cycle- and apoptosis-related genes affected by TMZ. Notably, GADD45A was found to be up-regulated by TMZ in both cell cycle and apoptosis arrays. Furthermore, GADD45A knockdown (GADD45Akd) enhanced the cell growth arrest and cell death induced by TMZ, even in natural (T98) and adapted (TR-U373) TMZ-resistant cells. Interestingly, GADD45Akd decreased the expression of O6-methylguanine-DNA methyltransferase (MGMT) in TMZ-resistant cells (T98 and TR-U373). In MGMT-deficient/TMZ-sensitive cells (U87 and U373), GADD45Akd decreased TMZ-induced TP53 expression. Thus, in this study, we investigated the genes influenced by TMZ that were important in GBM therapy, and revealed that GADD45A plays a protective role against TMZ treatment which may through TP53-dependent and MGMT-dependent pathway in TMZ-sensitive and TMZ-resistant GBM, respectively. This protective role of GADD45A against TMZ treatment may provide a new therapeutic strategy for GBM treatment.
Collapse
Affiliation(s)
- Hsiao-Han Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsuey-Yu Chang
- Department of Parasitology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Chen Lin
- Department of Parasitology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuo-Chen Wei
- Departments of Neurosurgery, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Jyh-Wei Shin
- Department of Parasitology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
42
|
Histone Deacetylase Inhibitors as Anticancer Drugs. Int J Mol Sci 2017; 18:ijms18071414. [PMID: 28671573 PMCID: PMC5535906 DOI: 10.3390/ijms18071414] [Citation(s) in RCA: 814] [Impact Index Per Article: 116.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/11/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
Carcinogenesis cannot be explained only by genetic alterations, but also involves epigenetic processes. Modification of histones by acetylation plays a key role in epigenetic regulation of gene expression and is controlled by the balance between histone deacetylases (HDAC) and histone acetyltransferases (HAT). HDAC inhibitors induce cancer cell cycle arrest, differentiation and cell death, reduce angiogenesis and modulate immune response. Mechanisms of anticancer effects of HDAC inhibitors are not uniform; they may be different and depend on the cancer type, HDAC inhibitors, doses, etc. HDAC inhibitors seem to be promising anti-cancer drugs particularly in the combination with other anti-cancer drugs and/or radiotherapy. HDAC inhibitors vorinostat, romidepsin and belinostat have been approved for some T-cell lymphoma and panobinostat for multiple myeloma. Other HDAC inhibitors are in clinical trials for the treatment of hematological and solid malignancies. The results of such studies are promising but further larger studies are needed. Because of the reversibility of epigenetic changes during cancer development, the potency of epigenetic therapies seems to be of great importance. Here, we summarize the data on different classes of HDAC inhibitors, mechanisms of their actions and discuss novel results of preclinical and clinical studies, including the combination with other therapeutic modalities.
Collapse
|
43
|
The Effect of Sodium Valproate on the Glioblastoma U87 Cell Line Tumor Development on the Chicken Embryo Chorioallantoic Membrane and on EZH2 and p53 Expression. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28642877 PMCID: PMC5469982 DOI: 10.1155/2017/6326053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Literature data support evidences that glioblastoma (GBM) patients experience prolonged survival due to sodium valproate (NaVP) treatment. The study assessed the human GBM cell U87 xenograft studied in the chicken embryo chorioallantoic membrane (CAM) model evaluating NaVP effect on tumor. Three groups of tumors (each n = 10) were studied: nontreated, treated with 4 mM, and treated with 8 mM of NaVP. The majority of tumors without NaVP treatment during tumor growth destroyed the chorionic epithelium, invaded the mesenchyme, and induced angiogenesis. Incidence of tumor formation on CAM without invasion into the mesenchyme was higher when U87 cells were treated with NaVP; the effect significantly increased with NaVP concentration. Treatment with 8 mM of NaVP did not show clear dynamics of tumor growth during 5 days; at the same time, the angiogenesis failed. With a strong staining of EZH2, p53 in tumors without NaVP treatment was found, and NaVP significantly decreased the expression of EZH2- and p53-positive cells; the effect was significantly higher at its 8 mM concentration. NaVP has a function in blocking the growth, invasion, and angiogenesis of tumor in the CAM model; tumor growth interferes with EZH2 and p53 molecular pathways, supporting the NaVP potential in GBM therapy.
Collapse
|
44
|
Pan H, Wang H, Jia Y, Wang Q, Li L, Wu Q, Chen L. VPA and MEL induce apoptosis by inhibiting the Nrf2-ARE signaling pathway in TMZ-resistant U251 cells. Mol Med Rep 2017; 16:908-914. [PMID: 28560379 DOI: 10.3892/mmr.2017.6621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/23/2017] [Indexed: 11/05/2022] Open
Abstract
Chemoresistance is the primary obstacle to effective treatment of glioblastoma, the most lethal brain tumor. Our previous study demonstrated that Nf-E2 related factor 2 (Nrf2), a traditional cytoprotective transcription factor, was overexpressed in gliomas and promoted malignancy. The present study aimed to investigate the expression levels of Nrf2‑antioxidant response element (ARE) signaling pathway genes in temozolomide (TMZ)‑resistant U251 human glioblastoma cells (U251‑TMZ). Additionally, the effect of valproic acid (VPA) and melatonin (MEL) on Nrf2 expression in U251‑TMZ cells and their association with chemoresistance was investigated. The results of the present study indicated that the expression levels of components of the Nrf2‑ARE signaling pathway were increased in U251‑TMZ cells compared with U251 parent cells. Silencing of Nrf2 by transfection with small interfering RNA restored the chemosensitivity of U251‑TMZ cells. The Nrf2 inhibitors VPA and MEL successfully reduced Nrf2 expression and survival in U251‑TMZ cells treated with TMZ, accompanied by increased reactive oxygen species levels and apoptosis. Therefore, VPA and MEL may be potential chemotherapeutic sensitizers for the treatment of chemoresistant glioblastoma.
Collapse
Affiliation(s)
- Hao Pan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yue Jia
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiang Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Liwen Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qi Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Longbang Chen
- Department of Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
45
|
Akbarnejad Z, Eskandary H, Dini L, Vergallo C, Nematollahi-Mahani SN, Farsinejad A, Abadi MFS, Ahmadi M. Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low-frequency electromagnetic field (100Hz, 100G). Biomed Pharmacother 2017; 92:254-264. [PMID: 28551545 DOI: 10.1016/j.biopha.2017.05.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/09/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain cancer that causes high mortality in humans. It responds poorly to the most common cancer treatments, such as surgery, chemo- and radiation therapy. Temozolomide (TMZ) is an alkylating agent that has been widely used to treat GBM; resistance to this drug is often found. One unexplored possibility for overcoming this resistance is a treatment based on concomitant exposure to electromagnetic fields (EMF) and TMZ. Indeed, many evidences show that EMF affects cancer cells and drug performance. In this study, we evaluated the potential synergistic effect of 100μM TMZ and EMF (100Hz, 100G) on two human glioma cells line, i.e., U87 and T98G above single treatments, TMZ or EMF. Co-treatment synergistically enhanced apoptosis in U87 and T98G cells, by increasing the expression of P53, Bax, and Caspase-3 and decreasing that of Bcl-2 and Cyclin-D1. We also observed an increase in reactive oxygen species (ROS) production and the overexpression of the heme oxygenase-1 (HO-1) gene in comparison to controls. In conclusion, since EMF enhanced the apoptotic effect of TMZ, possibly through a redox regulation mechanism, the TMZ/EMF combination may be effective for glioma cancer treating. Further studies are needed to reveal the action mechanism of this possible novel therapeutic approach.
Collapse
Affiliation(s)
- Zeinab Akbarnejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, 76175-113 Kerman, Iran
| | - Hossein Eskandary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, 76175-113 Kerman, Iran; Afzal Research Institute (NGO), 76175-113 Kerman, Iran.
| | - Luciana Dini
- Department of Biological and Environmental Science and Technology (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy; CNR Nanotec, 73100 Lecce, Italy
| | - Cristian Vergallo
- Department of Biological and Environmental Science and Technology (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy
| | | | - Alireza Farsinejad
- Pathology and Stem Cell Research Center, Afzalipour Medical School, Kerman University of Medical Sciences, 76175-113 Kerman, Iran
| | - Maryam Fekri Soofi Abadi
- Pathology and Stem Cell Research Center, Afzalipour Medical School, Kerman University of Medical Sciences, 76175-113 Kerman, Iran
| | - Meysam Ahmadi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, 76175-113 Kerman, Iran.
| |
Collapse
|
46
|
Xu K, Zhang Z, Pei H, Wang H, Li L, Xia Q. FoxO3a induces temozolomide resistance in glioblastoma cells via the regulation of β-catenin nuclear accumulation. Oncol Rep 2017; 37:2391-2397. [PMID: 28260024 DOI: 10.3892/or.2017.5459] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/30/2017] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common malignant brain tumor, is currently treated with temozolomide (TMZ), but GBM often exhibits resistance to TMZ. Although several mechanisms underlying GBM resistance to TMZ have been identified, these mechanisms are yet to fully explain how GBM gains resistance to TMZ. Our previous work has shown that FoxO3a, a member of the FoxO subfamily of transcription factors, promotes glioma cell proliferation and invasion. In this study, we sought to determine whether FoxO3a participates in TMZ resistance in GBM cells. Parental cell lines (also designated as sensitive cell lines) U87-MG and U251-MG, as well as the corresponding resistant cell lines U87-TR and U251-TR (generated by repeated TMZ treatments), were subjected to western blot analysis. Our results showed that the resistant cells (both U87-TRand U251-TR) exhibited higher levels of FoxO3a and β-catenin relative to their corresponding sensitive counterparts. Depletion of FoxO3a in the resistant cells enhanced the cytotoxic effect of TMZ. Further investigation showed that FoxO3a depletion did not affect the total protein level of β-catenin, but otherwise markedly reduced the nuclear β-catenin level. Taken together, these findings strongly support that FoxO3a renders GBM cells resistant to TMZ treatment, at least in part, through the regulation of β-catenin nuclear accumulation.
Collapse
Affiliation(s)
- Ke Xu
- Department of Immunology, School of Tropical and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571101, P.R. China
| | - Zhenhao Zhang
- Medical Technology Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Hua Pei
- Department of Immunology, School of Tropical and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571101, P.R. China
| | - Huamin Wang
- Department of Immunology, School of Tropical and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571101, P.R. China
| | - Liang Li
- Department of Immunology, School of Tropical and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571101, P.R. China
| | - Qianfeng Xia
- Key Laboratory of Tropical Biomedicine, and Faculty of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571101, P.R. China
| |
Collapse
|
47
|
Chen R, Lee WYW, Zhang XH, Zhang JT, Lin S, Xu LL, Huang B, Yang FY, Liu HL, Wang B, Tsang LL, Willaime-Morawek S, Li G, Chan HC, Jiang X. Epigenetic Modification of the CCL5/CCR1/ERK Axis Enhances Glioma Targeting in Dedifferentiation-Reprogrammed BMSCs. Stem Cell Reports 2017; 8:743-757. [PMID: 28216148 PMCID: PMC5355636 DOI: 10.1016/j.stemcr.2017.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/15/2017] [Accepted: 01/16/2017] [Indexed: 01/07/2023] Open
Abstract
The success of stem cell-mediated gene therapy in cancer treatment largely depends on the specific homing ability of stem cells. We have previously demonstrated that after in vitro induction of neuronal differentiation and dedifferentiation, bone marrow stromal cells (BMSCs) revert to a primitive stem cell population (De-neu-BMSCs) distinct from naive BMSCs. We report here that De-neu-BMSCs express significantly higher levels of chemokines, and display enhanced homing abilities to glioma, the effect of which is mediated by the activated CCL5/CCR1/ERK axis. Intriguingly, we find that the activated chemokine axis in De-neu-BMSCs is epigenetically regulated by histone modifications. On the therapeutic front, we show that De-neu-BMSCs elicit stronger homing and glioma-killing effects together with cytosine deaminase/5-fluorocytosine compared with unmanipulated BMSCs in vivo. Altogether, the current study provides an insight into chemokine regulation in BMSCs, which may have more profound effects on BMSC function and their application in regenerative medicine and cancer targeting.
Collapse
Affiliation(s)
- Rui Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Xiao Hu Zhang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Jie Ting Zhang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Liang Liang Xu
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Biao Huang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Fu Yuan Yang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Hai Long Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Bin Wang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Lai Ling Tsang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | | | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; The Chinese University of Hong Kong, Shenzhen Research Institute, Shenzhen 518057 PR China
| | - Hsiao Chang Chan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; The Chinese University of Hong Kong, Shenzhen Research Institute, Shenzhen 518057 PR China.
| | - Xiaohua Jiang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Epithelial Cell Biology Research Center, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China; The Chinese University of Hong Kong, Shenzhen Research Institute, Shenzhen 518057 PR China.
| |
Collapse
|
48
|
Pinheiro R, Braga C, Santos G, Bronze MR, Perry MJ, Moreira R, Brites D, Falcão AS. Targeting Gliomas: Can a New Alkylating Hybrid Compound Make a Difference? ACS Chem Neurosci 2017; 8:50-59. [PMID: 27665765 DOI: 10.1021/acschemneuro.6b00169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive type of brain tumor in adults. The triazene Temozolomide (TMZ), an alkylating drug, is the classical chemotherapeutic agent for gliomas, but has been disappointing against the highly invasive and resistant nature of GBM. Hybrid compounds may open new horizons within this challenge. The multicomponent therapeutic strategy here used resides on a combination of two repurposing drugs acting by different but potentially synergistic mechanisms, improved efficacy, and lower resistance effects. We synthesized a new hybrid compound (HYBCOM) by covalently binding a TMZ analogue to valproic acid, a histone deacetylase inhibitor drug that was shown to sensitize TMZ-resistant glioma cells. Advantages of this new molecule as compared to TMZ, in terms of chemotherapeutic efficacy, were investigated. Our results evidenced that HYBCOM more efficiently decreased the viability and proliferation of the GL261 glioma cells, while showing to better target the tumor cells than the functionally normal astrocytes. Increased cytotoxicity by HYBCOM may be a consequence of the improved autophagic process observed. Additionally, HYBCOM changed the morphology of GL261 cells into a nonpolar, more rounded shape, impairing cell migration ability. Most interesting, and in opposite to TMZ, cells exposed to HYBCOM did not enhance the expression of drug resistance proteins, a major issue in the treatment of GBM. Overall, our studies indicate that HYBCOM has promising chemotherapeutic benefits over the classical TMZ, and future studies should assess if the treatment translates into efficacy in glioblastoma experimental models and reveal clinical benefits in GBM patients.
Collapse
Affiliation(s)
- Rui Pinheiro
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Cláudia Braga
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Gisela Santos
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria R. Bronze
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria J. Perry
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Rui Moreira
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Dora Brites
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| | - Ana S. Falcão
- Research Institute for
Medicines (iMed.ULisboa), ‡Department of Biochemistry and Human
Biology, §Department
of Pharmaceutical Chemistry and Therapeutics, and ∥Department of Toxicological and Bromatological
Sciences, Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor
Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
49
|
Connelly KE, Martin EC, Dykhuizen EC. CBX Chromodomain Inhibition Enhances Chemotherapy Response in Glioblastoma Multiforme. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2016; 89:431-440. [PMID: 28018136 PMCID: PMC5168823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Glioblastoma multiforme (GBM) lacks effective therapeutic options leaving patients with a survival time of approximately one year. Recently, the alteration of chromatin modulators has been implicated in the pathogenesis and chemoresistance of numerous cancers; in particular, the Polycomb Group Proteins have been shown to play a role in glioblastoma progression and maintenance [1-5]. In this study, we aimed to identify drug combinations that decrease GBM cell viability by combining small molecule inhibitors against the Polycomb family with two standard chemotherapies. We identified dual inhibition of the CBX chromodomain with doxorubicin as a novel therapeutic strategy. While treatment with chromodomain inhibitor is non-toxic to cells alone, it dramatically increased the toxicity of standard chemotherapy drugs. We further validated an increase in DNA damage resulting in a G2/M block and subsequent apoptosis using the dual inhibitor treatment.
Collapse
Affiliation(s)
| | | | - Emily C. Dykhuizen
- Purdue University, West Lafayette, IN,To whom all correspondence should be addressed: Emily C. Dykhuizen, 201 S. University, West Lafayette, IN 47906, , tel: 765-494-4706
| |
Collapse
|
50
|
Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine. Cell Oncol (Dordr) 2016; 40:21-32. [PMID: 27766591 DOI: 10.1007/s13402-016-0301-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2016] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Glioblastoma (GBM) ranks among the deadliest solid cancers worldwide and its prognosis has remained dismal, despite the use of aggressive chemo-irradiation treatment regimens. Limited drug delivery into the brain parenchyma and frequent resistance to currently available therapies are problems that call for a prompt development of novel therapeutic strategies. While only displaying modest efficacies as mono-therapy in pre-clinical settings, histone deacetylase inhibitors (HDACi) have shown promising sensitizing effects to a number of cytotoxic agents. Here, we sought to investigate the sensitizing effect of the HDACi trichostatin A (TSA) to the alkylating agent lomustine (CCNU), which is used in the clinic for the treatment of GBM. METHODS Twelve primary GBM cell cultures grown as neurospheres were used in this study, as well as one established GBM-derived cell line (U87 MG). Histone deacetylase (HDAC) expression levels were determined using quantitative real-time PCR and Western blotting. The efficacy of either CCNU alone or its combination with TSA was assessed using various assays, i.e., cell viability assays (MTT), cell cycle assays (flow cytometry, FACS), double-strand DNA break (DSB) quantification assays (microscopy/immunofluorescence) and expression profiling assays of proteins involved in apoptosis and cell stress (Western blotting and protein array). RESULTS We found that the HDAC1, 3 and 6 expression levels were significantly increased in GBM samples compared to non-neoplastic brain control samples. Additionally, we found that pre-treatment of GBM cells with TSA resulted in an enhancement of their sensitivity to CCNU, possibly via the accumulation of DSBs, decreased cell proliferation and viability rates, and an increased apoptotic rate. CONCLUSION From our data we conclude that the combined administration of TSA and CCNU eradicates GBM cells with a higher efficacy than either drug alone, thereby opening a novel avenue for the treatment of GBM.
Collapse
|