1
|
Lee Y, Lee E, Roh TH, Kim SH. Association between Levetiracetam Use and Survival in Patients with Glioblastoma: A Nationwide Population-Based Study. Cancer Res Treat 2025; 57:369-377. [PMID: 39265622 PMCID: PMC12016850 DOI: 10.4143/crt.2024.355] [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: 04/09/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
PURPOSE This study aimed to investigate whether levetiracetam (LEV), the most used antiepileptic drug, influences survival in patients with glioblastoma (GBM), using a national database. MATERIALS AND METHODS This study used data from the Korea Health Insurance Review and Assessment database. Patients diagnosed with GBM between 2007-2018 treated with standard therapy were included. The study population was divided into long-term (≥ 60 days) and short-term (< 30 days) LEV groups. A separate long-term valproic acid (VPA) group (≥ 60 days) was identified for comparison. Demographics, disease characteristics, and treatment parameters were collected. Kaplan-Meier method and Cox regression were used to compare survival outcomes between the groups. RESULTS Overall, 2,971 patients were included, with 1,319 and 1,652 in the short-term and long-term LEV groups, respectively. The median overall survival (OS) for the entire population was 19.15 months post-surgery. Kaplan-Meier analysis revealed a significantly longer median OS in the long-term LEV group versus the short-term LEV group. After adjusting for confounders, Cox proportional hazard analysis revealed an association of long-term LEV use with improved survival, which was also observed in a subgroup analysis of patients without preoperative seizure history. The long-term LEV group demonstrated longer median OS, compared with the long-term VPA group. CONCLUSION Our nationwide population-based study found an association between long-term LEV use and improved survival in patients with GBM, regardless of preoperative seizure history. Prospective studies are needed to validate these findings and investigate the potential impact of LEV on the survival outcomes of patients with GBM.
Collapse
Affiliation(s)
- Yeonhu Lee
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea
| | - Eunyoung Lee
- Department of Neurology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Tae Hoon Roh
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea
| | - Se-Hyuk Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
2
|
Khalili BF, Walbert T, Horbinski C, Dixit K, Gururangan K, Thio H, Tate MC, Stupp R, Lukas RV, Templer JW. Levetiracetam and valproic acid in glioma: antiseizure and potential antineoplastic effects. Future Oncol 2025; 21:483-491. [PMID: 39786974 PMCID: PMC11812422 DOI: 10.1080/14796694.2025.2450215] [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: 09/03/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Seizures are a frequent complication in glioma. Incidence of brain tumor-related epilepsy (BTRE) in high-grade glioma (HGG) is an estimated > 25% and in low-grade glioma (LGG) is approximately 72%. Two first-line antiseizure medications (ASMs) for BTRE include levetiracetam (LEV) and valproic acid (VPA). Use of VPA has decreased because of a broader side effect profile, potential interaction with chemotherapeutic drugs, and availability of newer generation agents. In refractory BTRE, LEV and VPA may be prescribed together to enhance seizure control. VPA and LEV have gained attention for their purported antineoplastic effects and synergistic role with temozolomide. VPA is suggested to modulate anticancer activity in vitro through multiple mechanisms. In addition, retrospective studies indicate increased overall survival in patients with epileptogenic HGGs who are managed with LEV or VPA rather than other ASMs. However, these studies have numerous limitations. It is also reported that patients with glioma and a seizure history have a longer survival. This extended survival, if one exists, may be only observed in certain gliomas with corresponding patient characteristics. We provide a brief overview of the management of BTRE, VPA and LEV as anticonvulsants and antineoplastics, and the factors that may be associated with survival in epileptogenic glioma.
Collapse
Affiliation(s)
| | - Tobias Walbert
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Craig Horbinski
- Department of Pathology, Northwestern University, Chicago, IL, USA
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
| | - Karan Dixit
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Kapil Gururangan
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Helen Thio
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Matthew C. Tate
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
- Department of Neurology, Northwestern University, Chicago, IL, USA
- Section of Hematology & Oncology, Northwestern University, Chicago, IL, USA
| | - Rimas V. Lukas
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Jessica W. Templer
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
- Department of Neurology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
3
|
Vychopen M, Güresir A, Basaran AE, Güresir E, Wach J. Impact of levetiracetam use in glioblastoma: an individual patient-level meta-analysis assessing overall survival. Neurosurg Rev 2024; 47:897. [PMID: 39653818 PMCID: PMC11628436 DOI: 10.1007/s10143-024-03137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/15/2024] [Accepted: 11/30/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Levetiracetam (Lev), an antiepileptic drug (AED), enhances alkylating chemotherapy sensitivity in glioblastoma (GB) by inhibiting MGMT expression. This meta-analysis evaluates Lev's impact on GB treatment by analyzing overall survival of individual patient data (IPD) from published studies. METHODS IPD was reconstructed using the R package IPDfromKM. Pooled IPD Kaplan-Meier charts of survival stratified by Lev therapy were created using the R package Survminer. One- and two-stage meta-analyses of Lev treatment regarding survival was performed. RESULTS Three articles covering 825 patients were included out of 3567 screened records. Lev usage prevalence was 0.36. IPD from 590 IDH wild-type glioblastomas, with a median follow-up of 16.1 months, were utilized. Pooled data revealed median survival times of 19.2 months (95%CI: 16.4-22.0) for Lev users versus 16.5 months (95%CI: 15.2-17.8) for partial/no use (p = 0.006). One-stage meta-analysis indicated a significant association between Lev use and survival in IDH wild-type GB (HR: 1.33, 95%CI: 1.08-1.64, p = 0.007). Two-stage meta-analysis confirmed these results. CONCLUSIONS This meta-analysis highlights that Lev use may prolong survival in IDH wild-type GB patients. Further randomized trials are needed to confirm these findings and identify subgroups benefiting most from Lev treatment.
Collapse
Affiliation(s)
- Martin Vychopen
- Department of Neurosurgery, University Hospital Leipzig, Liebigstraße 20, Leipzig, 04103, Germany
| | - Agi Güresir
- Department of Neurosurgery, University Hospital Leipzig, Liebigstraße 20, Leipzig, 04103, Germany
| | - Alim Emre Basaran
- Department of Neurosurgery, University Hospital Leipzig, Liebigstraße 20, Leipzig, 04103, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Leipzig, Liebigstraße 20, Leipzig, 04103, Germany
| | - Johannes Wach
- Department of Neurosurgery, University Hospital Leipzig, Liebigstraße 20, Leipzig, 04103, Germany.
| |
Collapse
|
4
|
Martens L, Babalola O, Aslam A, Ashraf R. Vanishing bile duct syndrome: a sequela of temozolomide and levetiracetam-induced cholestatic liver injury. BMJ Case Rep 2024; 17:e260830. [PMID: 39542501 PMCID: PMC11574768 DOI: 10.1136/bcr-2024-260830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Temozolomide (TMZ)-levetiracetam (LEV) combination therapy in glioblastoma management is gradually becoming a mainstay treatment given its superior effect compared with TMZ monotherapy. While there have been previous cases of hepatotoxicity, there are no prior reports of vanishing bile duct syndrome (VBDS) associated with TMZ-LEV combination use. This case report details a male in his 50s who had recently completed TMZ and LEV for right frontal lobe glioblastoma. He presented 3 days later with painless jaundice, dark urine and pale stools. Laboratory evaluation was remarkable for marked hyperbilirubinemia and transaminitis. Extensive work up for hepatic and extra-hepatic causes of jaundice was of no yield, thus necessitating a liver biopsy. Liver pathology showed a non-specific histomorphology pattern suggesting drug-induced liver injury and cholestasis with severe ductopenia. VBDS due to TMZ and LEV was diagnosed. The patient followed with the gastroenterology clinic over 6 months for persistently elevated liver function tests before suffering a fatal cardiac arrest.
Collapse
Affiliation(s)
- Lindsey Martens
- Medical School, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Olawale Babalola
- Internal Medicine, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Awais Aslam
- Essentia Health West Region, Fargo, North Dakota, USA
| | - Rabiah Ashraf
- Essentia Health Fargo Hospital, Fargo, North Dakota, USA
| |
Collapse
|
5
|
Nguyen TTT, Greene LA, Mnatsakanyan H, Badr CE. Revolutionizing Brain Tumor Care: Emerging Technologies and Strategies. Biomedicines 2024; 12:1376. [PMID: 38927583 PMCID: PMC11202201 DOI: 10.3390/biomedicines12061376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive forms of brain tumor, characterized by a daunting prognosis with a life expectancy hovering around 12-16 months. Despite a century of relentless research, only a select few drugs have received approval for brain tumor treatment, largely due to the formidable barrier posed by the blood-brain barrier. The current standard of care involves a multifaceted approach combining surgery, irradiation, and chemotherapy. However, recurrence often occurs within months despite these interventions. The formidable challenges of drug delivery to the brain and overcoming therapeutic resistance have become focal points in the treatment of brain tumors and are deemed essential to overcoming tumor recurrence. In recent years, a promising wave of advanced treatments has emerged, offering a glimpse of hope to overcome the limitations of existing therapies. This review aims to highlight cutting-edge technologies in the current and ongoing stages of development, providing patients with valuable insights to guide their choices in brain tumor treatment.
Collapse
Affiliation(s)
- Trang T. T. Nguyen
- Ronald O. Perelman Department of Dermatology, Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Lloyd A. Greene
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Hayk Mnatsakanyan
- Department of Neurology, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA; (H.M.); (C.E.B.)
| | - Christian E. Badr
- Department of Neurology, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA; (H.M.); (C.E.B.)
| |
Collapse
|
6
|
Lee PY, Wei YT, Chao KSC, Chu CN, Chung WH, Wang TH. Anti-epileptic drug use during adjuvant chemo-radiotherapy is associated with poorer survival in patients with glioblastoma: A nationwide population-based cohort study. J Cancer Res Ther 2024; 20:555-562. [PMID: 38687925 DOI: 10.4103/jcrt.jcrt_750_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 10/31/2022] [Indexed: 05/02/2024]
Abstract
INTRODUCTION There are emerging but inconsistent evidences about anti-epileptic drugs (AEDs) as radio- or chemo-sensitizers to improve survival in glioblastoma patients. We conducted a nationwide population-based study to evaluate the impact of concurrent AED during post-operative chemo-radiotherapy on outcome. MATERIAL AND METHODS A total of 1057 glioblastoma patients were identified by National Health Insurance Research Database and Cancer Registry in 2008-2015. Eligible criteria included those receiving surgery, adjuvant radiotherapy and temozolomide, and without other cancer diagnoses. Survival between patients taking concurrent AED for 14 days or more during chemo-radiotherapy (AED group) and those who did not (non-AED group) were compared, and subgroup analyses for those with valproic acid (VPA), levetiracetam (LEV), or phenytoin were performed. Multivariate analyses were used to adjust for confounding factors. RESULTS There were 642 patients in the AED group, whereas 415 in the non-AED group. The demographic data was balanced except trend of more patients in the AED group had previous drug history of AEDs (22.6% vs. 18%, P 0.078). Overall, the AED group had significantly increased risk of mortality (HR = 1.18, P 0.016) compared to the non-AED group. Besides, an adverse dose-dependent relationship on survival was also demonstrated in the AED group (HR = 1.118, P 0.0003). In subgroup analyses, the significant detrimental effect was demonstrated in VPA group (HR = 1.29,P 0.0002), but not in LEV (HR = 1.18, P 0.079) and phenytoin (HR = 0.98, P 0.862). CONCLUSIONS Improved survival was not observed in patients with concurrent AEDs during chemo-radiotherapy. Our real-world data did not support prophylactic use of AEDs for glioblastoma patients.
Collapse
Affiliation(s)
- Peng-Yi Lee
- Department of Radiation Oncology, Show-Chwan Memorial Hospital, No. 542, Section 1 CHUNG-SHAN Road, Changhua, Taiwan
- Department of Radiation Oncology, Lin Shin Hospital, No. 36, Section 3 Huizhong Road, Taichung, Taiwan
- Department of Radiation Oncology, China Medical University Beigang Hospital, Yunlin, Taiwan
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Ting Wei
- Division of Family Medicine, Department of Community Medicine, China Medical University Hospital, Taichung, Taiwan
- Division of Occupational Medicine, Department of Community Medicine, China Medical University Hospital, Taichung, Taiwan
| | | | - Chin-Nan Chu
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
| | - Wen-Hui Chung
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ti-Hao Wang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
7
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
8
|
Chen P, Lou L, Sharma B, Li M, Xie C, Yang F, Wu Y, Xiao Q, Gao L. Recent Advances on PKM2 Inhibitors and Activators in Cancer Applications. Curr Med Chem 2024; 31:2955-2973. [PMID: 37455458 DOI: 10.2174/0929867331666230714144851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 05/28/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
Metabolic reprogramming of cells, from the normal mode of glucose metabolism named glycolysis, is a pivotal characteristic of impending cancerous cells. Pyruvate kinase M2 (PKM2), an important enzyme that catalyzes the final rate-limiting stage during glycolysis, is highly expressed in numerous types of tumors and aids in development of favorable conditions for the survival of tumor cells. Increasing evidence has suggested that PKM2 is one of promising targets for innovative drug discovery, especially for the developments of antitumor therapeutics. Herein, we systematically summarize the recent advancement on PKM2 modulators including inhibitors and activators in cancer applications. We also discussed the classifications of pyruvate kinases in mammals and the biological functions of PKM2 in this review. We do hope that this review would provide a comprehensive understanding of the current research on PKM2 modulators, which may benefit the development of more potent PKM2-related drug candidates to treat PKM2-associated diseases including cancers in future.
Collapse
Affiliation(s)
- Peng Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Liang Lou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Bigyan Sharma
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Mengchu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Chengliang Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Fen Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| |
Collapse
|
9
|
Joghataei MT, Bakhtiarzadeh F, Dehghan S, Ketabforoush AHME, Golab F, Zarbakhsh S, Ahmadirad N. The role of neurotransmitters in glioblastoma multiforme-associated seizures. Int J Dev Neurosci 2023; 83:677-690. [PMID: 37563091 DOI: 10.1002/jdn.10294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
GBM, or glioblastoma multiforme, is a brain tumor that poses a great threat to both children and adults, being the primary cause of death related to brain tumors. GBM is often associated with epilepsy, which can be debilitating. Seizures and the development of epilepsy are the primary symptoms that have a severe impact on the quality of life for GBM patients. It is increasingly apparent that the nervous system plays an essential role in the tumor microenvironment for all cancer types, including GBM. In recent years, there has been a growing understanding of how neurotransmitters control the progression of gliomas. Evidence suggests that neurotransmitters and neuromodulators found in the tumor microenvironment play crucial roles in the excitability, proliferation, quiescence, and differentiation of neurons, glial cells, and neural stem cells. The involvement of neurotransmitters appears to play a significant role in various stages of GBM. In this review, the focus is on presenting updated knowledge and emerging ideas regarding the interplay between neurotransmitters and neuromodulators, such as glutamate, GABA, norepinephrine, dopamine, serotonin, adenosine, and their relationship with GBM and the seizures induced by this condition. The review aims to explore the current understanding and provide new insights into the complex interactions between these neurotransmitters and neuromodulators in the context of GBM-related seizures.
Collapse
Affiliation(s)
| | - Fatemeh Bakhtiarzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Samaneh Dehghan
- Eye Research Center, The Five Senses Institute, Rasool Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sam Zarbakhsh
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Nooshin Ahmadirad
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Aroosa M, Malik JA, Ahmed S, Bender O, Ahemad N, Anwar S. The evidence for repurposing anti-epileptic drugs to target cancer. Mol Biol Rep 2023; 50:7667-7680. [PMID: 37418080 PMCID: PMC10460753 DOI: 10.1007/s11033-023-08568-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/31/2023] [Indexed: 07/08/2023]
Abstract
Antiepileptic drugs are versatile drugs with the potential to be used in functional drug formulations with drug repurposing approaches. In the present review, we investigated the anticancer properties of antiepileptic drugs and interlinked cancer and epileptic pathways. Our focus was primarily on those drugs that have entered clinical trials with positive results and those that provided good results in preclinical studies. Many contributing factors make cancer therapy fail, like drug resistance, tumor heterogeneity, and cost; exploring all alternatives for efficient treatment is important. It is crucial to find new drug targets to find out new antitumor molecules from the already clinically validated and approved drugs utilizing drug repurposing methods. The advancements in genomics, proteomics, and other computational approaches speed up drug repurposing. This review summarizes the potential of antiepileptic drugs in different cancers and tumor progression in the brain. Valproic acid, oxcarbazepine, lacosamide, lamotrigine, and levetiracetam are the drugs that showed potential beneficial outcomes against different cancers. Antiepileptic drugs might be a good option for adjuvant cancer therapy, but there is a need to investigate further their efficacy in cancer therapy clinical trials.
Collapse
Affiliation(s)
- Mir Aroosa
- Department of Pharmacology, Jamia Hamdard, New Delhi, India
| | - Jonaid Ahmad Malik
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Ropar, Ropar, India
| | - Sakeel Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Onur Bender
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan lagoon selatan, Petaling Jaya, Selangor, DE, Malaysia.
| | - Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Saudi Arabia.
| |
Collapse
|
11
|
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: 5] [Impact Index Per Article: 2.5] [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
|
12
|
Pellegrino M, Ricci E, Ceraldi R, Nigro A, Bonofiglio D, Lanzino M, Morelli C. From HDAC to Voltage-Gated Ion Channels: What's Next? The Long Road of Antiepileptic Drugs Repositioning in Cancer. Cancers (Basel) 2022; 14:cancers14184401. [PMID: 36139561 PMCID: PMC9497059 DOI: 10.3390/cancers14184401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although in the last decades the clinical outcome of cancer patients considerably improved, the major drawbacks still associated with chemotherapy are the unwanted side effects and the development of drug resistance. Therefore, a continuous effort in trying to discover new tumor markers, possibly of diagnostic, prognostic and therapeutic value, is being made. This review is aimed at highlighting the anti-tumor activity that several antiepileptic drugs (AEDs) exert in breast, prostate and other types of cancers, mainly focusing on their ability to block the voltage-gated Na+ and Ca++ channels, as well as to inhibit the activity of histone deacetylases (HDACs), all well-documented tumor markers and/or molecular targets. The existence of additional AEDs molecular targets is highly suspected. Therefore, the repurposing of already available drugs as adjuvants in cancer treatment would have several advantages, such as reductions in dose-related toxicity CVs will be sent in a separate mail to the indicated address of combined treatments, lower production costs, and faster approval for clinical use. Abstract Cancer is a major health burden worldwide. Although the plethora of molecular targets identified in the last decades and the deriving developed treatments, which significantly improved patients’ outcome, the occurrence of resistance to therapies remains the major cause of relapse and mortality. Thus, efforts in identifying new markers to be exploited as molecular targets in cancer therapy are needed. This review will first give a glance on the diagnostic and therapeutic significance of histone deacetylase (HDAC) and voltage gated ion channels (VGICs) in cancer. Nevertheless, HDAC and VGICs have also been reported as molecular targets through which antiepileptic drugs (AEDs) seem to exert their anticancer activity. This should be claimed as a great advantage. Indeed, due to the slowness of drug approval procedures, the attempt to turn to off-label use of already approved medicines would be highly preferable. Therefore, an updated and accurate overview of both preclinical and clinical data of commonly prescribed AEDs (mainly valproic acid, lamotrigine, carbamazepine, phenytoin and gabapentin) in breast, prostate, brain and other cancers will follow. Finally, a glance at the emerging attempt to administer AEDs by means of opportunely designed drug delivery systems (DDSs), so to limit toxicity and improve bioavailability, is also given.
Collapse
Affiliation(s)
| | | | | | | | | | - Marilena Lanzino
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| | - Catia Morelli
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| |
Collapse
|
13
|
Kumar TS, Afnan WM, Chan CY, Audrey C, Fong SL, Rajandram R, Lim KS, Narayanan V. Impact of seizures and antiseizure medication on survival in patients with glioma. J Neurooncol 2022; 159:657-664. [PMID: 36036318 DOI: 10.1007/s11060-022-04108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Seizures are a common presenting symptom among patients with low- and high-grade glioma. However, the impact and inter-relationship between the presence of seizures, anti-seizure medication (ASM) and survival are unclear. We retrospectively analyzed the incidence of seizures and identified the pattern and relationship of anti-seizure medication on survival in our cohort of patients with glioma. METHODS We evaluated all glioma patients who underwent treatment at the University of Malaya Medical Centre (UMMC) between 2008 and 2020. Demographic and clinical data of seizures and pattern of ASM administration in comparison to overall survival were analyzed. RESULTS A total of 235 patients were studied, with a minimum of one year clinical follow-up post-treatment. The median survival for low-grade glioma was 38 months whereas high-grade glioma was 15 months. One-third of our glioma patients (n = 74) presented with seizures. All patients with seizures and a further 31% of patients without seizures were started on anti-seizure medication preoperatively. Seizure and Levetiracetam (LEV) were significantly associated with OS on univariate analysis. However, only LEV (HR 0.49; 95% CI 0.23-0.87; p=0.02) was significantly associated with improving overall survival (OS) on multivariate analysis. Once ASM was adjusted for relevant factors and each other, LEV was associated with improved survival in all grade gliomas (HR 0.52; 95% CI 0.31-0.88; p=0.02) and specifically high-grade gliomas (HR 0.53; 95% CI 0.30-0.94; p=0.03). CONCLUSIONS Pre-operative seizures among patients with glioma indicated a better overall prognosis. The administration of ASM, specifically LEV was associated with a significant survival advantage in our retrospective cohort of patients.
Collapse
Affiliation(s)
- Thinisha Sathis Kumar
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Wan Muhammad Afnan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Chet-Ying Chan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Christine Audrey
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Si-Lei Fong
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Retnagowri Rajandram
- Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Kheng-Seang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia
| | - Vairavan Narayanan
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia.
- Hospital Tanah Merah Kelantan, Tanah Merah, Malaysia.
| |
Collapse
|
14
|
Ntafoulis I, Koolen SLW, Leenstra S, Lamfers MLM. Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma. Cancers (Basel) 2022; 14:3705. [PMID: 35954371 PMCID: PMC9367381 DOI: 10.3390/cancers14153705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma (GBM) remains one of the most difficult tumors to treat. The mean overall survival rate of 15 months and the 5-year survival rate of 5% have not significantly changed for almost 2 decades. Despite progress in understanding the pathophysiology of the disease, no new effective treatments to combine with radiation therapy after surgical tumor debulking have become available since the introduction of temozolomide in 1999. One of the main reasons for this is the scarcity of compounds that cross the blood-brain barrier (BBB) and reach the brain tumor tissue in therapeutically effective concentrations. In this review, we focus on the role of the BBB and its importance in developing brain tumor treatments. Moreover, we discuss drug repurposing, a drug discovery approach to identify potential effective candidates with optimal pharmacokinetic profiles for central nervous system (CNS) penetration and that allows rapid implementation in clinical trials. Additionally, we provide an overview of repurposed candidate drug currently being investigated in GBM at the preclinical and clinical levels. Finally, we highlight the importance of phase 0 trials to confirm tumor drug exposure and we discuss emerging drug delivery technologies as an alternative route to maximize therapeutic efficacy of repurposed candidate drug.
Collapse
Affiliation(s)
- Ioannis Ntafoulis
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands;
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Sieger Leenstra
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Martine L. M. Lamfers
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| |
Collapse
|
15
|
Cucchiara F, Luci G, Giannini N, Giorgi FS, Orlandi P, Banchi M, Di Paolo A, Pasqualetti F, Danesi R, Bocci G. Association of plasma levetiracetam concentration, MGMT methylation and sex with survival of chemoradiotherapy-treated glioblastoma patients. Pharmacol Res 2022; 181:106290. [DOI: 10.1016/j.phrs.2022.106290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 12/27/2022]
|
16
|
Morales DE, Mousa SA. Intranasal Delivery in Glioblastoma treatment: Prospective Molecular Treatment Modalities. Heliyon 2022; 8:e09517. [PMID: 35647354 PMCID: PMC9136349 DOI: 10.1016/j.heliyon.2022.e09517] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/09/2022] [Accepted: 05/18/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is rare and fatal glioma with limited treatment options. Treatments provide minimal improvement in prognosis and only 6.8% of GBM patients have a life expectancy greater than five years. Surgical resection of this malignant glioma is difficult due to its highly invasive nature and follow-up radiotherapy with concomitant temozolomide, the currently approved standard of care, and will only extend the life of patients by a few months. It has been nearly two decades since the approval of temozolomide and there have been no clinically relevant major breakthroughs since, painting a dismal picture for patients with GBM. Although the future of GBM management seems bleak, there are many new treatment options on the horizon that propose methods of delivery to circumvent current limitations in the standard of care, i.e., the blood brain barrier and treatment resistance mechanisms. The nose is a highly accessible non-invasive route of delivery that has been incorporated into many investigational studies within the past five years and potentially paves the path to a brighter future for the management of GBM. Intranasal administration has its limitations however, as drugs can be degraded and/or fail to reach the site of action. This has prompted many studies for implementation of nanoparticle systems to overcome these limitations and to accurately deliver drugs to the site of action. This review highlights the advances in intranasal therapy delivery and impact of nanotechnology in the management of GBM and discusses potential treatment modalities that show promise for further investigation.
Collapse
|
17
|
Sun M, Huang N, Tao Y, Wen R, Zhao G, Zhang X, Xie Z, Cheng Y, Mao J, Liu G. The efficacy of temozolomide combined with levetiracetam for glioblastoma (GBM) after surgery: a study protocol for a double-blinded and randomized controlled trial. Trials 2022; 23:234. [PMID: 35346332 PMCID: PMC8962067 DOI: 10.1186/s13063-022-06168-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background Temozolomide is applied as the standard chemotherapy agent in patients with glioblastoma (GBM) after surgery. However, the benefit of this treatment for patients is limited by the invasive growth of gliomas and drug resistance. There are indications from fundamental experimental and retrospective studies that levetiracetam has the potential to improve the survival rate of patients with GBM. However, it has yet to be determined whether the combination of temozolomide and levetiracetam is more effective than standard temozolomide chemotherapy. Therefore, we designed a randomized clinical trial to investigate the therapeutic effect of the new combined regime for treating GBM. Methods/design This is a double-blind and randomized clinical trial conducted in a single center. One hundred forty-two patients will be recruited and screened for the inclusion and exclusion criteria. Then, eligible participants will be randomly assigned to an experimental group or a control group in a 1:1 ratio. Based on the administration of radiation therapy (RT), participants in the experimental group will be prescribed levetiracetam plus temozolomide chemotherapy for 34 weeks while participants in the control group will receive placebo tablets plus temozolomide for the same duration. A 3-year follow-up will be conducted on all patients after intervention. Accordingly, the primary outcome will be progression-free survival (PFS). The secondary endpoints include overall survival (OS), the Karnofsky Performance Status (KPS), the objective response rate (ORR), and adverse event incidence. Discussion It is expected that the results of this trial will provide high-level evidence regarding the clinical benefits of levetiracetam and temozolomide combined in the treatment of GBM. Trial registration Chinese Clinical Trial Registry, ChiCTR2100049941. Registered on 14 August 2021 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06168-1.
Collapse
Affiliation(s)
- Maoyuan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Rong Wen
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Xiang Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Jinning Mao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China. .,Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA, 02114, USA. .,Neuroscience Program, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
18
|
The effect of levetiracetam treatment on survival in patients with glioblastoma: a systematic review and meta-analysis. J Neurooncol 2022; 156:257-267. [PMID: 34982371 DOI: 10.1007/s11060-021-03940-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/28/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Levetiracetam (LEV) is an anti-epileptic drug (AED) that sensitizes glioblastoma (GBM) to temozolomide (TMZ) chemotherapy by inhibiting O6-methylguanine-DNA methyltransferase (MGMT) expression. Adding LEV to the standard of care (SOC) for GBM may improve TMZ efficacy. This study aimed to pool the existing evidence in the literature to quantify LEV's effect on GBM survival and characterize its safety profile to determine whether incorporating LEV into the SOC is warranted. METHOD A search of CINAHL, Embase, PubMed, and Web of Science from inception to May 2021 was performed to identify relevant articles. Hazard ratios (HR), median overall survival, and adverse events were pooled using random-effect models. Meta-regression, funnel plots, and the Newcastle-Ottawa Scale were utilized to identify sources of heterogeneity, bias, and statistical influence. RESULTS From 20 included studies, 5804 GBM patients underwent meta-analysis, of which 1923 (33%) were treated with LEV. Administration of LEV did not significantly improve survival in the entire patient population (HR 0.89, p = 0.094). Significant heterogeneity was observed during pooling of HRs (I2 = 75%, p < 0.01). Meta-regression determined that LEV treatment effect decreased with greater rates of MGMT methylation (RC = 0.03, p = 0.02) and increased with greater proportions of female patients (RC = - 0.05, p = 0.002). Concurrent LEV with the SOC for GBM did not increase odds of adverse events relative to other AEDs. CONCLUSIONS Levetiracetam treatment may not be effective for all GBM patients. Instead, LEV may be better suited for treating specific molecular profiles of GBM. Further studies are necessary to identify optimal GBM candidates for LEV.
Collapse
|
19
|
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: 12] [Impact Index Per Article: 3.0] [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
|
20
|
Pallud J, Huberfeld G, Dezamis E, Peeters S, Moiraghi A, Gavaret M, Guinard E, Dhermain F, Varlet P, Oppenheim C, Chrétien F, Roux A, Zanello M. Effect of Levetiracetam Use Duration on Overall Survival of Isocitrate Dehydrogenase Wildtype Glioblastoma in Adults: An Observational Study. Neurology 2021; 98:e125-e140. [PMID: 34675100 DOI: 10.1212/wnl.0000000000013005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/15/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES The association between Levetiracetam and survival of Isocitrate Dehydrogenase (IDH) wildtype glioblastomas is controversial. We investigated whether the duration of Levetiracetam use during the standard chemoradiation protocol impacts overall survival of IDH-wildtype glioblastoma patients. METHODS Observational single-institution cohort study (2010-2018). Inclusion criteria were: 1) patients ≥18 years old; 2) newly diagnosed supratentorial tumor; 3) histomolecular diagnosis of IDH-wildtype glioblastoma; 4) standard chemoradiation protocol. To assess the survival benefit of Levetiracetam use during the standard chemoradiation protocol (whole duration, part time, and never subgroups), a Cox proportional hazard model was constructed. We performed a case-matched analysis (1:1) between patients with Levetiracetam use during the whole duration of the standard chemoradiation protocol and patients with Levetiracetam use part time or never according to the following criteria: sex, age, epileptic seizures at diagnosis, RTOG-RPA class, tumor location, preoperative volume, extent of resection, and O6-Methylguanine-DNA methyltransferase promoter methylation status. Patients with unavailable O6-Methylguanine-DNA methyltransferase promoter methylation status (48.5%) were excluded. RESULTS 460 patients were included. The median overall survival was longer in the 116 patients with Levetiracetam use during the whole duration of the standard chemoradiation protocol (21.0 months; 95%CI, 17.2-24.0) than in the 126 patients with part time Levetiracetam use (16.8 months; 95%CI, 12.4-19.0], and in the 218 patients who never received Levetiracetam (16.0 months; 95%CI, 15.5-19.4; p=0.027). Levetiracetam use during the whole duration of the standard chemoradiation protocol (adjusted Hazard Ratio (aHR) 0.69; 95%CI, 0.52-0.93; p=0.014), O6-Methylguanine-DNA methyltransferase promoter methylation (aHR 0.53; 95%CI, 0.39-0.71; p<0.001), and gross total tumor resection (aHR 0.57; 95%CI, 0.44-0.74; p<0.001) were independent predictors of a longer overall survival. After case matching (n=54 per group), a longer overall survival was found for Levetiracetam use during the whole duration of the standard chemoradiation protocol (HR=0.63; 95%CI, 0.42-0.94, p=0.023). DISCUSSION Levetiracetam use during the whole standard chemoradiation protocol possibly improves overall survival of IDH-wildtype glioblastoma patients. It should be considered in the anti-tumor strategy of future multicentric trials. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that in individuals with IDH-wildtype glioblastoma, levetiracetam use throughout the duration of standard chemotherapy is associated with longer median overall survival.
Collapse
Affiliation(s)
- Johan Pallud
- Department of Neurosurgery, GHU Paris - Sainte-Anne Hospital, Paris, France .,Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Gilles Huberfeld
- Neurology Department, Hopital Fondation Adolphe de Rothschild, 29 rue Main, 75019 Paris, France.,Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL Paris, France
| | - Edouard Dezamis
- Department of Neurosurgery, GHU Paris - Sainte-Anne Hospital, Paris, France.,Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Sophie Peeters
- Department of Neurosurgery, University of California, Los Angeles - Los Angeles, CA, USA
| | - Alessandro Moiraghi
- Department of Neurosurgery, GHU Paris - Sainte-Anne Hospital, Paris, France.,Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Martine Gavaret
- Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France.,Department of Neurophysiology, GHU Paris - Sainte-Anne Hospital, Paris, France
| | - Eléonore Guinard
- Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France.,Department of Neurophysiology, GHU Paris - Sainte-Anne Hospital, Paris, France
| | - Frédéric Dhermain
- Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France
| | - Pascale Varlet
- Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France.,Department of Neuropathology, GHU Paris - Sainte-Anne Hospital, Paris, France
| | - Catherine Oppenheim
- Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France.,Department of Neuroradiology, GHU Paris - Sainte-Anne Hospital, Paris, France
| | - Fabrice Chrétien
- Université de Paris, Sorbonne Paris Cité, Paris, France.,Department of Neuropathology, GHU Paris - Sainte-Anne Hospital, Paris, France
| | - Alexandre Roux
- Department of Neurosurgery, GHU Paris - Sainte-Anne Hospital, Paris, France.,Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Marc Zanello
- Department of Neurosurgery, GHU Paris - Sainte-Anne Hospital, Paris, France.,Université de Paris, Sorbonne Paris Cité, Paris, France.,Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| |
Collapse
|
21
|
Ulasov IV, Borovjagin A, Laevskaya A, Kamynina M, Timashev P, Cerchia L, Rozhkova EA. The IL13α 2R paves the way for anti-glioma nanotherapy. Genes Dis 2021; 10:89-100. [PMID: 37013057 PMCID: PMC10066331 DOI: 10.1016/j.gendis.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive (grade IV) gliomas characterized by a high rate of recurrence, resistance to therapy and a grim survival prognosis. The long-awaited improvement in GBM patients' survival rates essentially depends on advances in the development of new therapeutic approaches. Recent preclinical studies show that nanoscale materials could greatly contribute to the improvement of diagnosis and management of brain cancers. In the current review, we will discuss how specific features of glioma pathobiology can be employed for designing efficient targeting approaches. Moreover, we will summarize the main evidence for the potential of the IL-13R alpha 2 receptor (IL13α2R) targeting in GBM early diagnosis and experimental therapy.
Collapse
Affiliation(s)
- Ilya V. Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Corresponding author.
| | - Anton Borovjagin
- Department of BioMedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, 4 Kosygin St, Moscow 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow 119991, Russia
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples 80131, Italy
| | - Elena A. Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, Argonne, IL 60439, USA
| |
Collapse
|
22
|
Tomar MS, Kumar A, Srivastava C, Shrivastava A. Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochim Biophys Acta Rev Cancer 2021; 1876:188616. [PMID: 34419533 DOI: 10.1016/j.bbcan.2021.188616] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/25/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023]
Abstract
Temozolomide (TMZ) is a first-choice alkylating agent inducted as a gold standard therapy for glioblastoma multiforme (GBM) and astrocytoma. A majority of patients do not respond to TMZ during the course of their treatment. Activation of DNA repair pathways is the principal mechanism for this phenomenon that detaches TMZ-induced O-6-methylguanine adducts and restores genomic integrity. Current understanding in the domain of oncology adds several other novel mechanisms of resistance such as the involvement of miRNAs, drug efflux transporters, gap junction's activity, the advent of glioma stem cells as well as upregulation of cell survival autophagy. This review describes a multifaceted account of different mechanisms responsible for the intrinsic and acquired TMZ-resistance. Here, we summarize different strategies that intensify the TMZ effect such as MGMT inhibition, development of novel imidazotetrazine analog, and combination therapy; with an aim to incorporate a successful treatment and increased overall survival in GBM patients.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, Madhya Pradesh, India
| | - Chhitij Srivastava
- Department of Neurosurgery, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow 226003, Uttar Pradesh, India.
| |
Collapse
|
23
|
Fan Q, Li L, Wang TL, Emerson RE, Xu Y. A Novel ZIP4-HDAC4-VEGFA Axis in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13153821. [PMID: 34359722 PMCID: PMC8345154 DOI: 10.3390/cancers13153821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/25/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Despite tremendous research efforts, epithelial ovarian cancer (EOC) remains one of the most difficult cancers to detect early and treat successfully for >5-year survival. We have recently shown that ZIP4, a zinc transporter, is a novel cancer stem cell (CSC) marker and a therapeutic target for EOC. The current work focuses on developing new strategies to target ZIP4 and inhibit its CSC activities in EOC. We found that cells expressing high levels of ZIP4 were supersensitive to a group of inhibitors called HDACis. One of the major targets of these inhibitors is a protein called HDAC4. We revealed the new molecular bases for the ZIP4-HDAC4 axis and tested the efficacies of targeting this axis in the lab and in mouse models. Our study provides a new mechanistic-based targeting strategy for EOC. Abstract We have recently identified ZIP4 as a novel cancer stem cell (CSC) marker in high-grade serous ovarian cancer (HGSOC). While it converts drug-resistance to cisplatin (CDDP), we unexpectedly found that ZIP4 induced sensitization of HGSOC cells to histone deacetylase inhibitors (HDACis). Mechanistically, ZIP4 selectively upregulated HDAC IIa HDACs, with little or no effect on HDACs in other classes. HDAC4 knockdown (KD) and LMK-235 inhibited spheroid formation in vitro and tumorigenesis in vivo, with hypoxia inducible factor-1 alpha (HIF1α) and endothelial growth factor A (VEGFA) as functional downstream mediators of HDAC4. Moreover, we found that ZIP4, HDAC4, and HIF1α were involved in regulating secreted VEGFA in HGSOC cells. Furthermore, we tested our hypothesis that co-targeting CSC via the ZIP4-HDAC4 axis and non-CSC using CDDP is necessary and highly effective by comparing the effects of ZIP4-knockout/KD, HDAC4-KD, and HDACis, in the presence or absence of CDDP on tumorigenesis in mouse models. Our results showed that the co-targeting strategy was highly effective. Finally, data from human HGSOC tissues showed that ZIP4 and HDAC4 were upregulated in a subset of recurrent tumors, justifying the clinical relevance of the study. In summary, our study provides a new mechanistic-based targeting strategy for HGSOC.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut St. R2-E380, Indianapolis, IN 46202, USA;
| | - Lihong Li
- Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 600 North Wolfe St., Baltimore, MD 21287, USA;
| | - Tian-Li Wang
- Department of Gynecology, Oncology, and Pathology, Johns Hopkins Medical Institutions, 1550 Orleans Street, Baltimore, MD 21231, USA;
| | - Robert E. Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana University Health Pathology Laboratory, 350 W. 11th Street, Room 4010, Indianapolis, IN 46202, USA;
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut St. R2-E380, Indianapolis, IN 46202, USA;
- Correspondence: ; Tel.: +1-(317)-274-3972
| |
Collapse
|
24
|
Terrié E, Déliot N, Benzidane Y, Harnois T, Cousin L, Bois P, Oliver L, Arnault P, Vallette F, Constantin B, Coronas V. Store-Operated Calcium Channels Control Proliferation and Self-Renewal of Cancer Stem Cells from Glioblastoma. Cancers (Basel) 2021; 13:cancers13143428. [PMID: 34298643 PMCID: PMC8307764 DOI: 10.3390/cancers13143428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Glioblastoma is a high-grade primary brain tumor that contains a subpopulation of cells called glioblastoma stem cells, which are responsible for tumor initiation, growth and recurrence after treatment. Recent transcriptomic studies have highlighted that calcium pathways predominate in glioblastoma stem cells. Calcium channels have the ability to transduce signals from the microenvironment and are therefore ideally placed to control cellular behavior. Using multiple approaches, we demonstrate in five different primary cultures, previously derived from surgical specimens, that glioblastoma stem cells express store-operated channels (SOC) that support calcium entry into these cells. Pharmacological inhibition of SOC dramatically reduces cell proliferation and stem cell self-renewal in these cultures. By identifying SOC as a critical mechanism involved in the maintenance of the stem cell population in glioblastoma, our study will contribute to the framework for the identification of new therapies against this deadly tumor. Abstract Glioblastoma is the most frequent and deadly form of primary brain tumors. Despite multimodal treatment, more than 90% of patients experience tumor recurrence. Glioblastoma contains a small population of cells, called glioblastoma stem cells (GSC) that are highly resistant to treatment and endowed with the ability to regenerate the tumor, which accounts for tumor recurrence. Transcriptomic studies disclosed an enrichment of calcium (Ca2+) signaling transcripts in GSC. In non-excitable cells, store-operated channels (SOC) represent a major route of Ca2+ influx. As SOC regulate the self-renewal of adult neural stem cells that are possible cells of origin of GSC, we analyzed the roles of SOC in cultures of GSC previously derived from five different glioblastoma surgical specimens. Immunoblotting and immunocytochemistry experiments showed that GSC express Orai1 and TRPC1, two core SOC proteins, along with their activator STIM1. Ca2+ imaging demonstrated that SOC support Ca2+ entries in GSC. Pharmacological inhibition of SOC-dependent Ca2+ entries decreased proliferation, impaired self-renewal, and reduced expression of the stem cell marker SOX2 in GSC. Our data showing the ability of SOC inhibitors to impede GSC self-renewal paves the way for a strategy to target the cells considered responsible for conveying resistance to treatment and tumor relapse.
Collapse
Affiliation(s)
- Elodie Terrié
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Nadine Déliot
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Yassine Benzidane
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Thomas Harnois
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Laëtitia Cousin
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Patrick Bois
- EA 4379, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France;
| | - Lisa Oliver
- CRCINA-UMR 1232 INSERM, Université de Nantes, CEDEX 01, 44007 Nantes, France; (L.O.); (F.V.)
| | - Patricia Arnault
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - François Vallette
- CRCINA-UMR 1232 INSERM, Université de Nantes, CEDEX 01, 44007 Nantes, France; (L.O.); (F.V.)
- CNRS GDR3697, Micronit “Microenvironment of Tumor Niches”, 37000 Tours, France
| | - Bruno Constantin
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
- CNRS GDR3697, Micronit “Microenvironment of Tumor Niches”, 37000 Tours, France
| | - Valérie Coronas
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
- CNRS GDR3697, Micronit “Microenvironment of Tumor Niches”, 37000 Tours, France
- Correspondence: ; Tel.: +33-(0)5-49-45-36-55
| |
Collapse
|
25
|
Kong FC, Ma CL, Zhong MK. Epigenetic Effects Mediated by Antiepileptic Drugs and their Potential Application. Curr Neuropharmacol 2020; 18:153-166. [PMID: 31660836 PMCID: PMC7324883 DOI: 10.2174/1570159x17666191010094849] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/01/2019] [Accepted: 10/03/2019] [Indexed: 12/20/2022] Open
Abstract
An epigenetic effect mainly refers to a heritable modulation in gene expression in the short term but does not involve alterations in the DNA itself. Epigenetic molecular mechanisms include DNA methylation, histone modification, and untranslated RNA regulation. Antiepileptic drugs have drawn attention to biological and translational medicine because their impact on epigenetic mechanisms will lead to the identification of novel biomarkers and possible therapeutic strategies for the prevention and treatment of various diseases ranging from neuropsychological disorders to cancers and other chronic conditions. However, these transcriptional and posttranscriptional alterations can also result in adverse reactions and toxicity in vitro and in vivo. Hence, in this review, we focus on recent findings showing epigenetic processes mediated by antiepileptic drugs to elucidate their application in medical experiments and shed light on epigenetic research for medicinal purposes.
Collapse
Affiliation(s)
- Fan-Cheng Kong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Chun-Lai Ma
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Ni XR, Guo CC, Yu YJ, Yu ZH, Cai HP, Wu WC, Ma JX, Chen FR, Wang J, Chen ZP. Combination of levetiracetam and IFN-α increased temozolomide efficacy in MGMT-positive glioma. Cancer Chemother Pharmacol 2020; 86:773-782. [PMID: 33074386 DOI: 10.1007/s00280-020-04169-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Glioma, especially glioblastoma (GBM), is the most aggressive malignant brain tumor and its standard therapy is often ineffective because of temozolomide (TMZ) resistance. Reversal of the TMZ resistance might improve the prognosis of glioma patients. We previously found that interferon-α (IFN-α) and anti-epileptic drug levetiracetam (LEV) could sensitize glioma to TMZ, respectively. In this study, we further investigated the efficiency of combining of LEV and IFN-α for improving the efficacy of TMZ. METHODS We evaluated whether LEV and IFN-α could increase TMZ efficacy using colony formation assay and cell viability assay with MGMT-positive and MGMT-negative glioma cell lines in vitro. Subcutaneous xenografts and orthotopic xenografts mice models were used in vivo to observe the tumor growth and mice survival upon treatments with TMZ, TMZ + IFN-α, TMZ + LEV, or TMZ + LEV + IFN-α. The expression levels of MGMT, markers of pro-apoptotic and anti-apoptotic in tumor samples were analyzed by Western blotting. RESULTS The combinational use of IFN-α, LEV, and TMZ showed the best anti-tumor activity in MGMT-positive cell lines (U138, GSC-1, U118, and T98 G). TMZ + LEV + IFN-α further obviously increased TMZ + LEV or TMZ + IFN-α efficiency in MGMT-positive cell lines, while not in negative cell lines (SKMG-4, U87, U373, and U251) in vitro, which were also observed in subcutaneous mice models (U138, GSC-1 compared to SKMG-4, U87) and orthotopic models (GSC-1) in vivo. Strikingly, the combination of LEV and IFN-α together with TMZ significantly prolonged the survival of mice with orthotopic GSC-1 glioma. Furthermore, we confirmed that the combination of LEV and IFN-α enhanced the inhibition of MGMT and the activation of apoptosis in U138 tumor on the basis of TMZ treatment. CONCLUSIONS The combination use of LEV and IFN-α could be an optimal method to overcome TMZ resistance through obvious MGMT inhibition in MGMT-positive glioma.
Collapse
Affiliation(s)
- Xiang-Rong Ni
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Cheng-Cheng Guo
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Yan-Jiao Yu
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Zhi-Hui Yu
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Hai-Ping Cai
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Wei-Chi Wu
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Jun-Xiao Ma
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Fu-Rong Chen
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China
| | - Jing Wang
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China.
| | - Zhong-Ping Chen
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Rd East, Guangdong, 510060, People's Republic of China.
| |
Collapse
|
27
|
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: 26] [Impact Index Per Article: 5.2] [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
|
28
|
Zhu H, Zhu H, Tian M, Wang D, He J, Xu T. DNA Methylation and Hydroxymethylation in Cervical Cancer: Diagnosis, Prognosis and Treatment. Front Genet 2020; 11:347. [PMID: 32328088 PMCID: PMC7160865 DOI: 10.3389/fgene.2020.00347] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022] Open
Abstract
Recent discoveries have led to the development of novel ideas and techniques that have helped elucidate the correlation between epigenetics and tumor biology. Nowadays, the field of tumor genetics has evolved to include a new type of regulation by epigenetics. An increasing number of studies have demonstrated the importance of DNA methylation and hydroxymethylation in specific genes in the progression of cervical cancer. Determining the methylation and hydroxymethylation profiles of these genes will help in the early prevention and diagnosis, monitoring recurrence, prognosis, and treatment of patients with cervical cancer. In this review, we focus on the significance of aberrant DNA methylation and hydroxymethylation in cervical cancer and the use of these epigenetic signatures in clinical settings.
Collapse
Affiliation(s)
- Hongming Zhu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - He Zhu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Miao Tian
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Dongying Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jiaxing He
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Tianmin Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
29
|
Antitumorigenic Effects of Inhibiting Ephrin Receptor Kinase Signaling by GLPG1790 against Colorectal Cancer Cell Lines In Vitro and In Vivo. JOURNAL OF ONCOLOGY 2020; 2020:9342732. [PMID: 32184826 PMCID: PMC7063197 DOI: 10.1155/2020/9342732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Erythropoietin-producing hepatocellular receptors (Eph) promote the onset and sustain the progression of cancers such as colorectal cancer (CRC), in which the A2 subtype of Eph receptor expression has been shown to correlate with a poor prognosis and has been identified as a promising therapeutic target. Herein, we investigated, in vitro and in vivo, the effects of treatment with GLPG1790, a potent pan-Eph inhibitor. The small molecule has selective activity against the EphA2 isoform in human HCT116 and HCT15 CRC cell lines expressing a constitutively active form of RAS concurrently with a wild-type or mutant form of p53, respectively. GLPG1790 reduced EPHA2 phosphorylation/activation and induced G1/S cell-cycle growth arrest by downregulating the expression of cyclin E and PCNA, while upregulating p21Waf1/Cip1 and p27Cip/Kip. The inhibition of ephrin signaling induced quiescence in HCT15 and senescence in HCT116 cells. While investigating the role of CRC-related, pro-oncogenic p53 and RAS pathways, we found that GLPG1790 upregulated p53 expression and that silencing p53 or inhibiting RAS (human rat sarcoma)/ERKs (extracellular signal-regulated kinase) signaling restrained the ability of GLPG1790 to induce senescence in HCT116 cells. On the other hand, HCT15 silencing of p53 predisposed cells to GLPG1790-induced senescence, whilst no effects of ERK inhibition were observed. Finally, GLPG1790 hindered the epithelial-mesenchymal transition, reduced the migratory capacities of CRC, and affected tumor formation in xenograft models in vivo more efficiently using HCT116 than HCT15 for xenografts. Taken together, our data suggest the therapeutic potential of GLPG1790 as a signal transduction-based therapeutic strategy in to treat CRC.
Collapse
|
30
|
Salvati M, Bruzzaniti P, Relucenti M, Nizzola M, Familiari P, Giugliano M, Scafa AK, Galletta S, Li X, Chen R, Barbaranelli C, Frati A, Santoro A. Retrospective and Randomized Analysis of Influence and Correlation of Clinical and Molecular Prognostic Factors in a Mono-Operative Series of 122 Patients with Glioblastoma Treated with STR or GTR. Brain Sci 2020; 10:brainsci10020091. [PMID: 32050461 PMCID: PMC7071604 DOI: 10.3390/brainsci10020091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma is a solid, infiltrating, and the most frequent highly malignant primary brain tumor. Our aim was to find the correlation between sex, age, preoperative Karnofsky performance status (KPS), presenting with seizures, and extent of resection (EOR) with overall survival (OS), progression-free survival (PFS), and postoperative KPS, along with the prognostic value of IDH1, MGMT, ATRX, EGFR, and TP53 genes mutations and of Ki67 through the analysis of a single-operator series in order to avoid the biases of a multi-operator series, such as the lack of homogeneity in surgical and adjuvant nonsurgical treatments. A randomized retrospective analysis of 122 patients treated by a single first operator at Sapienza University of Rome was carried out. After surgery, patients followed standard Stupp protocol treatment. Exclusion criteria were: (1) patients with primary brainstem and spinal cord gliomas and (2) patients who underwent partial resections (resection < 90%) or a biopsy exclusively for diagnostic purposes. Statistical analysis with a simultaneous regression model was carried out through the use of SPSS 25® (IBM). Results showed statistically significant survival increase in four groups: (1) patients treated with gross total resection (GTR) (p < 0.030); (2) patients with mutation of IDH1 (p < 0.0161); (3) patients with methylated MGMT promoter (p < 0.005); (4) patients without EGFR amplification or EGFRvIII mutation (p < 0.035). Higher but not statistically significant survival rates were also observed in: patients <75 years, patients presenting with seizures at diagnosis, patients affected by lesions in noneloquent areas, as well as in patients with ATRX gene mutation and Ki-67 < 10%.
Collapse
Affiliation(s)
- Maurizio Salvati
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Placido Bruzzaniti
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
- Correspondence: ; Tel.: +39-3349753520
| | - Michela Relucenti
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Science, “La Sapienza” University of Rome, 00161 Rome, Italy;
| | - Mariagrazia Nizzola
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Pietro Familiari
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Marco Giugliano
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Anthony Kevin Scafa
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Santi Galletta
- UOSD of Neurophysiopathology and DISMOV, AOU G Martino, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing 210009, China; (X.L.); (R.C.)
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing 210009, China; (X.L.); (R.C.)
| | - Claudio Barbaranelli
- Department of Psychology, Faculty of Medicine and Psychology “La Sapienza” University of Rome, 00189 Rome, Italy;
| | - Alessandro Frati
- Department of Neurosurgery, IRCCS Neuromed Pozzilli IS, 86077 Isernia, Italy;
| | - Antonio Santoro
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| |
Collapse
|
31
|
Kim SK, Moon J, Cho JM, Kim KH, Kim SH, Kim YI, Kim YZ, Kim HS, Dho YS, Park JS, Park JE, Seo Y, Sung KS, Song JH, Wee CW, Yoon WS, Yoon HI, Lee SH, Lim DH, Im JH, Chang JH, Han MH, Hong JB, Hwang K, Park CK, Lee YS, Gwak HS. A National Consensus Survey for Current Practice in Brain Tumor Management I: Antiepileptic Drug and Steroid Usage. Brain Tumor Res Treat 2020; 8:1-10. [PMID: 32390348 PMCID: PMC7221462 DOI: 10.14791/btrt.2020.8.e5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/02/2020] [Accepted: 03/16/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The Guideline Working Group of the Korean Society for Neuro-Oncology (KSNO) conducted a nationwide questionnaire survey for diverse queries faced in the treatment of brain tumors. As part I of the survey, the aim of this study is to evaluate national patterns of clinical practice about antiepileptic drug (AED) and steroid usage for management of brain tumors. METHODS A web-based survey was sent to all members of the KSNO by email. The survey included 9 questions of AED usage and 5 questions of steroid usage for brain tumor patients. All questions were developed by consensus of the Guideline Working Group. RESULTS The overall response rate was 12.8% (54/423). Regarding AED usage, the majority of respondents (95.2%) routinely prescribed prophylactic AEDs for patients with seizure at the peri/postoperative period. However, as many as 72.8% of respondents prescribed AED routinely for seizure-naïve patients, and others prescribed AED as the case may be. The duration of AED prophylaxis showed wide variance according to the epilepsy status and the location of tumor. Levetiracetam (82.9%) was the most preferred AED for epilepsy prophylaxis. Regarding steroid usage, 90.5% of respondents use steroids in perioperative period, including 34.2% of them as a routine manner. Presence of peritumoral edema (90.9%) was considered as the most important factor determining steroid usage followed by degree of clinical symptoms (60.6%). More than half of respondents (51.2%) replied to discontinue the steroids within a week after surgery if there are no specific medical conditions, while 7.3% preferred slow tapering up to a month after surgery. CONCLUSION The survey demonstrated the prevailing practice patterns on AED and steroid usage in neuro-oncologic field among members of the KSNO. This information provides a point of reference for establishing a practical guideline in the management of brain tumor patients.
Collapse
Affiliation(s)
- Sung Kwon Kim
- Department of Neurosurgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Korea
| | - Jangsup Moon
- Department of Neurology, Rare Disease Center, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Mo Cho
- Department of Neurosurgery, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Korea
| | - Kyung Hwan Kim
- Department of Neurosurgery, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Il Kim
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Young Zoon Kim
- Division of Neurooncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yun Sik Dho
- Department of Neurosurgery, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Jae Sung Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Youngbeom Seo
- Department of Neurosurgery, Yeungnam University Hospital, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyoung Su Sung
- Department of Neurosurgery, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Korea
| | - Jin Ho Song
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan Woo Wee
- Department of Radiation Oncology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Wan Soo Yoon
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Se Hoon Lee
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Ho Im
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Myung Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Je Beom Hong
- Department of Neurosurgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kihwan Hwang
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Chul Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| | - Youn Soo Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Ho Shin Gwak
- Department of Cancer Control, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
| | | |
Collapse
|
32
|
Ryu JY, Min KL, Chang MJ. Effect of anti-epileptic drugs on the survival of patients with glioblastoma multiforme: A retrospective, single-center study. PLoS One 2019; 14:e0225599. [PMID: 31790459 PMCID: PMC6886804 DOI: 10.1371/journal.pone.0225599] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a lethal and aggressive malignant tumor of the central nervous system. The World Health Organization classifies it as a grade IV astrocytoma. Controlling seizures is essential during GBM treatment because they are often present and closely associated with the quality of life of GBM patients. Some antiepileptic drugs (AEDs) exhibit antitumor effects and could decrease the mortality of patients with GBM. In this retrospective cohort study, we examined 418 patients treated with surgery, radiotherapy, and chemotherapy with temozolomide (TMZ) at Severance Hospital in South Korea, per the current protocol. Median overall survival (OS) was 21 months [95% confidence interval (CI): 18.1-23.9] in the levetiracetam (LEV) treatment group, whereas it was 16 months [95% CI: 14.1-17.9] in the group without LEV, exhibiting a statistically significant difference between the two groups (P < 0.001). Of nine AED groups, only LEV treatment [P = 0.001; hazard ratio (HR), 0.65; 95% CI: 0.51-0.83] exhibited a statistically significant difference in the OS, in the univariate analysis. In the risk analysis of the baseline characteristics, age, administration of LEV, and O6-methylguanine-DNA methyltransferase (MGMT) promoter status correlated with OS. The use of LEV in the group with a methylated MGMT promoter resulted in a positive impact on the OS [P = 0.006; HR, 0.174; 95% CI: 0.050-0.608], but the effect of LEV on the OS was not statistically significant in the unmethylated MGMT promoter group (P = 0.623). This study suggests that, compared with other AEDs, the administration of LEV may prolong the survival period in GBM patients with methylated MGMT promoters, who are undergoing chemotherapy with TMZ.
Collapse
Affiliation(s)
- Jae Yeoul Ryu
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Kyoung Lok Min
- Department of Pharmaceutical Medicines and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Min Jung Chang
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
- Department of Pharmaceutical Medicines and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
- * E-mail:
| |
Collapse
|
33
|
Su Q, Luo S, Tan Q, Deng J, Zhou S, Peng M, Tao T, Yang X. The role of pyruvate kinase M2 in anticancer therapeutic treatments. Oncol Lett 2019; 18:5663-5672. [PMID: 31788038 PMCID: PMC6865080 DOI: 10.3892/ol.2019.10948] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer cells are characterized by a high glycolytic rate, which leads to energy regeneration and anabolic metabolism; a consequence of this is the abnormal expression of pyruvate kinase isoenzyme M2 (PKM2). Multiple studies have demonstrated that the expression levels of PKM2 are upregulated in numerous cancer types. Consequently, the mechanism of action of certain anticancer drugs is to downregulate PKM2 expression, indicating the significance of PKM2 in a chemotherapeutic setting. Furthermore, it has previously been highlighted that the downregulation of PKM2 expression, using either inhibitors or short interfering RNA, enhances the anticancer effect exerted by THP treatment on bladder cancer cells, both in vitro and in vivo. The present review summarizes the detailed mechanisms and therapeutic relevance of anticancer drugs that inhibit PKM2 expression. In addition, the relationship between PKM2 expression levels and drug resistance were explored. Finally, future directions, such as the targeting of PKM2 as a strategy to explore novel anticancer agents, were suggested. The current review explored and highlighted the important role of PKM2 in anticancer treatments.
Collapse
Affiliation(s)
- Qiongli Su
- Department of Pharmacy, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Shengping Luo
- Department of Pharmacy, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Qiuhong Tan
- Department of Pharmacy, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Jun Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Sichun Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ting Tao
- Department of Pharmacy, Yueyang Maternal-Child Medicine Health Hospital, Yueyang, Hunan 414000, P.R. China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
34
|
Marampon F, Di Nisio V, Pietrantoni I, Petragnano F, Fasciani I, Scicchitano BM, Ciccarelli C, Gravina GL, Festuccia C, Del Fattore A, Tombolini M, De Felice F, Musio D, Cecconi S, Tini P, Maddalo M, Codenotti S, Fanzani A, Polimeni A, Maggio R, Tombolini V. Pro-differentiating and radiosensitizing effects of inhibiting HDACs by PXD-101 (Belinostat) in in vitro and in vivo models of human rhabdomyosarcoma cell lines. Cancer Lett 2019; 461:90-101. [DOI: 10.1016/j.canlet.2019.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/08/2019] [Accepted: 07/13/2019] [Indexed: 12/11/2022]
|
35
|
Bahadur S, Sahu AK, Baghel P, Saha S. Current promising treatment strategy for glioblastoma multiform: A review. Oncol Rev 2019; 13:417. [PMID: 31410248 PMCID: PMC6661528 DOI: 10.4081/oncol.2019.417] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/29/2019] [Indexed: 01/15/2023] Open
Abstract
Glioblastoma multiform (GBM) is a heterogeneous group of primary neoplasm resistant to conventional therapies. Due to their infiltrative nature it not fully isolated by aggressive surgery, radiation and chemotherapy showing poor prognosis in glioma patients. Unfortunately, diagnosed patients die within 1.5-2 year treatment schedule. Currently temozolomide (TMZ) is the first choice for the prognosis of GBM patients. TMZ metabolites methyl triazen imidazol carboxamide form complex with alkyl guanine alkyl transferase (O6 MGMT- DNA repair protein) induced DNA damage following resistance properties of TMZ and inhibit the overall survival of the patients. Last few decades different TMZ conjugated strategy is developed to overcome the resistance and enhance the chemotherapy efficacy. The main aim of this review is to introduce the new promising pharmaceutical candidates that significantly influence the therapeutic response of the TMZ in context of targeted therapy of glioblastoma patients. It is hoped that this proposed strategy are highly effective to overcome the current resistance limitations of TMZ in GBM patients and enhance the survival rate of the patients.
Collapse
Affiliation(s)
| | - Arvind Kumar Sahu
- Department of Pharmaceutics, Columbia Institute of Pharmacy, Near Vidhan Sabha, Raipur, Chhattisgarh, India
| | | | | |
Collapse
|
36
|
Pathological and Molecular Features of Glioblastoma and Its Peritumoral Tissue. Cancers (Basel) 2019; 11:cancers11040469. [PMID: 30987226 PMCID: PMC6521241 DOI: 10.3390/cancers11040469] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive and lethal human brain tumors. At present, GBMs are divided in primary and secondary on the basis of the mutational status of the isocitrate dehydrogenase (IDH) genes. In addition, IDH1 and IDH2 mutations are considered crucial to better define the prognosis. Although primary and secondary GBMs are histologically indistinguishable, they retain distinct genetic alterations that account for different evolution of the tumor. The high invasiveness, the propensity to disperse throughout the brain parenchyma, and the elevated vascularity make these tumors extremely recidivist, resulting in a short patient median survival even after surgical resection and chemoradiotherapy. Furthermore, GBM is considered an immunologically cold tumor. Several studies highlight a highly immunosuppressive tumor microenvironment that promotes recurrence and poor prognosis. Deeper insight into the tumor immune microenvironment, together with the recent discovery of a conventional lymphatic system in the central nervous system (CNS), led to new immunotherapeutic strategies. In the last two decades, experimental evidence from different groups proved the existence of cancer stem cells (CSCs), also known as tumor-initiating cells, that may play an active role in tumor development and progression. Recent findings also indicated the presence of highly infiltrative CSCs in the peritumoral region of GBM. This region appears to play a key role in tumor growing and recurrence. However, until recently, few studies investigated the biomolecular characteristics of the peritumoral tissue. The aim of this review is to recapitulate the pathological features of GBM and of the peritumoral region associated with progression and recurrence.
Collapse
|
37
|
Marampon F, Codenotti S, Megiorni F, Del Fattore A, Camero S, Gravina GL, Festuccia C, Musio D, De Felice F, Nardone V, Santoro AN, Dominici C, Fanzani A, Pirtoli L, Fioravanti A, Tombolini V, Cheleschi S, Tini P. NRF2 orchestrates the redox regulation induced by radiation therapy, sustaining embryonal and alveolar rhabdomyosarcoma cells radioresistance. J Cancer Res Clin Oncol 2019; 145:881-893. [PMID: 30701326 DOI: 10.1007/s00432-019-02851-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Tumor cells generally exhibit higher levels of reactive oxygen species (ROS), however, when stressed, tumor cells can undergo a process of 'Redox Resetting' to acquire a new redox balance with stronger antioxidant systems that enable cancer cells to become resistant to radiation therapy (RT). Here, we describe how RT affects the oxidant/antioxidant balance in human embryonal (RD) and alveolar (RH30) rhabdomyosarcoma (RMS) cell lines, investigating on the molecular mechanisms involved. METHODS Radiations were delivered using an x-6 MV photon linear accelerator and their effects were assessed by vitality and clonogenic assays. The expression of specific antioxidant-enzymes, such as Superoxide Dismutases (SODs), Catalase (CAT) and Glutathione Peroxidases 4 (GPx4), miRNAs (miR-22, -126, -210, -375, -146a, -34a) and the transcription factor NRF2 was analyzed by quantitative polymerase chain reaction (q-PCR) and western blotting. RNA interference experiments were performed to evaluate the role of NRF2. RESULTS Doses of RT higher than 2 Gy significantly affected RMS clonogenic ability by increasing ROS production. RMS rapidly and efficiently brought back ROS levels by up-regulating the gene expression of antioxidant enzymes, miRNAs as well as of NRF2. Silencing of NRF2 restrained the RMS ability to counteract RT-induced ROS accumulation, antioxidant enzyme and miRNA expression and was able to increase the abundance of γ-H2AX, a biomarker of DNA damage, in RT-treated cells. CONCLUSIONS Taken together, our data suggest the strategic role of oxidant/antioxidant balance in restraining the therapeutic efficiency of RT in RMS treatment and identify NRF2 as a new potential molecular target whose inhibition might represent a novel radiosensitizing therapeutic strategy for RMS clinical management.
Collapse
MESH Headings
- Antioxidants/metabolism
- Cell Line, Tumor
- Dose-Response Relationship, Radiation
- Humans
- MicroRNAs/biosynthesis
- MicroRNAs/genetics
- NF-E2-Related Factor 2/biosynthesis
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Oxidation-Reduction/radiation effects
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Radiation Tolerance
- Reactive Oxygen Species/metabolism
- Rhabdomyosarcoma, Alveolar/genetics
- Rhabdomyosarcoma, Alveolar/metabolism
- Rhabdomyosarcoma, Alveolar/radiotherapy
- Rhabdomyosarcoma, Embryonal/genetics
- Rhabdomyosarcoma, Embryonal/metabolism
- Rhabdomyosarcoma, Embryonal/radiotherapy
- Transfection
- Up-Regulation/radiation effects
Collapse
Affiliation(s)
- Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy.
| | - Silvia Codenotti
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Andrea Del Fattore
- Multi-Factorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCCS, Viale di San Paolo 15, 00146, Rome, Italy
| | - Simona Camero
- Department of Pediatrics, "Sapienza" University of Rome, Rome, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Daniela Musio
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Francesca De Felice
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | | | | | - Carlo Dominici
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Alessandro Fanzani
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luigi Pirtoli
- Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Unit of Radiation Oncology, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Istituto Toscano Tumori, Florence, Italy
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | | | - Vincenzo Tombolini
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Sara Cheleschi
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, University of Siena, Policlinico Le Scotte, Siena, Italy
| | - Paolo Tini
- Unit of Radiation Oncology, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Istituto Toscano Tumori, Florence, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Sbarro Health Research Organization, Temple University, Philadelphia, PA, USA
| |
Collapse
|
38
|
Wang Q, He Z, Chen Y. Comprehensive Analysis Reveals a 4-Gene Signature in Predicting Response to Temozolomide in Low-Grade Glioma Patients. Cancer Control 2019; 26:1073274819855118. [PMID: 31167546 PMCID: PMC6558750 DOI: 10.1177/1073274819855118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 11/25/2022] Open
Abstract
Low-grade gliomas (LGGs) are a highly heterogeneous group of slow-growing, lethal, diffusive brain tumors. Temozolomide (TMZ) is a frequently used primary chemotherapeutic agent for LGGs. Currently there is no consensus as to the optimal biomarkers to predict the efficacy of TMZ, which calls for decision-making for each patient while considering molecular profiles. Low-grade glioma data sets were retrieved from The Cancer Genome Atlas. Cox regression and survival analyses were applied to identify clinical features significantly associated with survival. Subsequently, Ordinal logistic regression, co-expression, and Cox regression analyses were applied to identify genes that correlate significantly with response rate, disease-free survival, and overall survival of patients receiving TMZ as primary therapy. Finally, gene expression and methylation analyses were exploited to explain the mechanism between these gene expression and TMZ efficacy in LGG patients. Overall survival was significantly correlated with age, Karnofsky Performance Status score, and histological grade, but not with IDH1 mutation status. Using 3 distinct efficacy end points, regression and co-expression analyses further identified a novel 4-gene signature of ASPM, CCNB1, EXO1, and KIF23 which negatively correlated with response to TMZ therapy. In addition, expression of the 4-gene signature was associated with those of genes involved in homologous recombination. Finally, expression and methylation profiling identified a largely unknown olfactory receptor OR51F2 as potential mediator of the roles of the 4-gene signature in reducing TMZ efficacy. Taken together, these findings propose the 4-gene signature as a novel panel of efficacy predictors of TMZ therapy, as well as potential downstream mechanisms, including homologous recombination, OR51F2, and DNA methylation independent of MGMT.
Collapse
Affiliation(s)
- Qi Wang
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and
Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic
Science and Technology of China, Sichuan, China
| | - Zongze He
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and
Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic
Science and Technology of China, Sichuan, China
| | - Yong Chen
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and
Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic
Science and Technology of China, Sichuan, China
| |
Collapse
|
39
|
Pham DC, Chang YC, Lin SR, Fuh YM, Tsai MJ, Weng CF. FAK and S6K1 Inhibitor, Neferine, Dually Induces Autophagy and Apoptosis in Human Neuroblastoma Cells. Molecules 2018; 23:molecules23123110. [PMID: 30486505 PMCID: PMC6321370 DOI: 10.3390/molecules23123110] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/24/2018] [Accepted: 11/25/2018] [Indexed: 12/13/2022] Open
Abstract
Human neuroblastoma cancer is the most typical extracranial solid tumor. Yet, new remedial treatment therapies are demanded to overcome its sluggish survival rate. Neferine, isolated from the lotus embryos, inhibits the proliferation of various cancer cells. This study aimed to evaluate the anti-cancer activity of neferine in IMR32 human neuroblastoma cells and to expose the concealable molecular mechanisms. IMR32 cells were treated with different concentrations of neferine, followed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to assess cell viability. In an effort to determine the molecular mechanisms in neferine-incubated IMR32 cells, cell cycle arrest, cell migration, and focal adhesion kinase (FAK), the 70-kDa ribosomal S6 kinase 1 (S6K1), poly (ADP-ribose) polymerase (PARP), caspase-3, Beclin-1, and microtubule-associated protein 1A/1B-light chain 3 (LC3) protein expressions were investigated. Neferine strongly disrupted the neuroblastoma cell growth via induction of G2/M phase arrest. Furthermore, neferine provoked autophagy and apoptosis in IMR32 cells, confirmed by p-FAK, and p-S6K1 reduction, LC3-II accumulation, Beclin-1 overexpression, and cleaved caspase-3/PARP improvement. Finally, neferine markedly retarded cell migration of neuroblastoma cancer cells. As a result, our findings for the first time showed an explicit anti-cancer effect of neferine in IMR32 cells, suggesting that neferine might be a potential candidate against human neuroblastoma cells to improve clinical outcomes with further in vivo investigation.
Collapse
Affiliation(s)
- Dinh-Chuong Pham
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Yu-Chuan Chang
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Yuh-Ming Fuh
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - May-Jywan Tsai
- Neural Regeneration Laboratory, Taipei Veterans General Hospital, Taipei 11260, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| |
Collapse
|