1
|
Mars JC, Culjkovic-Kraljacic B, Borden KL. eIF4E orchestrates mRNA processing, RNA export and translation to modify specific protein production. Nucleus 2024; 15:2360196. [PMID: 38880976 PMCID: PMC11185188 DOI: 10.1080/19491034.2024.2360196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
The eukaryotic translation initiation factor eIF4E acts as a multifunctional factor that simultaneously influences mRNA processing, export, and translation in many organisms. Its multifactorial effects are derived from its capacity to bind to the methyl-7-guanosine cap on the 5'end of mRNAs and thus can act as a cap chaperone for transcripts in the nucleus and cytoplasm. In this review, we describe the multifactorial roles of eIF4E in major mRNA-processing events including capping, splicing, cleavage and polyadenylation, nuclear export and translation. We discuss the evidence that eIF4E acts at two levels to generate widescale changes to processing, export and ultimately the protein produced. First, eIF4E alters the production of components of the mRNA processing machinery, supporting a widescale reprogramming of multiple mRNA processing events. In this way, eIF4E can modulate mRNA processing without physically interacting with target transcripts. Second, eIF4E also physically interacts with both capped mRNAs and components of the RNA processing or translation machineries. Further, specific mRNAs are sensitive to eIF4E only in particular mRNA processing events. This selectivity is governed by the presence of cis-acting elements within mRNAs known as USER codes that recruit relevant co-factors engaging the appropriate machinery. In all, we describe the molecular bases for eIF4E's multifactorial function and relevant regulatory pathways, discuss the basis for selectivity, present a compendium of ~80 eIF4E-interacting factors which play roles in these activities and provide an overview of the relevance of its functions to its oncogenic potential. Finally, we summarize early-stage clinical studies targeting eIF4E in cancer.
Collapse
Affiliation(s)
- Jean-Clément Mars
- Institute of Research in Immunology and Cancer, Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada
| | - Biljana Culjkovic-Kraljacic
- Institute of Research in Immunology and Cancer, Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada
| | - Katherine L.B. Borden
- Institute of Research in Immunology and Cancer, Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
2
|
Mikhina EA, Stepanycheva DV, Maksimova VP, Sineva ON, Markelova NN, Grebenkina LE, Lesovaya EA, Yakubovskaya MG, Matveev AV, Zhidkova EM. Synthesis of Alkyl/Aryloxymethyl Derivatives of 1,2,4-Triazole-3-Carboxamides and Their Biological Activities. Molecules 2024; 29:4808. [PMID: 39459177 PMCID: PMC11509950 DOI: 10.3390/molecules29204808] [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: 09/16/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Ribavirin and its analogues exhibit an in vitro antiproliferative effect in cancer cells. In this work, we studied the biological activities of a number of alkyl/aryloxymethyl derivatives of ribavirin's aglycon-1,2,4-triazole-3-carboxamide. Alkyl/arylxymethyl derivatives of 1,2,4-triazole-3-carboxamide with substitutions at the fifth or first position of the triazole ring, were synthesized and their antiproliferative and antimicrobial effects were assessed. For both series, the presence of an antiproliferative effect was investigated, and 1-alkyl/aryloxymethyl derivatives were shown an antimicrobial potential against a Gram-positive bacteria Micrococcus luteus and Gram-negative bacterium Pseudomonas aeruginosa. The obtained results showed that the n-decyloxymethyl derivatives induced leukemia cell death at low micromolar concentrations. We confirmed that n-decyloxymethyl derivatives of ribavirin inhibited the cell cycle progression and induced an accumulation of leukemia cells in the subG1-phase. The molecular docking results suggest that alkyl/aryloxymethyl derivatives may act by inhibiting translation initiation, due to interference with eIF4E assembly. The outcome results revealed that active derivatives (1- or 5-n-decyloxymethyl-1,2,4-triazole-3-carboxamides) can be considered as a lead compound for anticancer treatments.
Collapse
Affiliation(s)
- Ekaterina A. Mikhina
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 86 Vernadsky Prospekt, Moscow 119571, Russia; (E.A.M.); (L.E.G.); (A.V.M.)
| | - Daria V. Stepanycheva
- Department of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (D.V.S.); (V.P.M.); (E.A.L.); (M.G.Y.)
| | - Varvara P. Maksimova
- Department of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (D.V.S.); (V.P.M.); (E.A.L.); (M.G.Y.)
| | - Olga N. Sineva
- Gause Institute of New Antibiotics, 11 Bolshaya Pirogovskaya St., Moscow 119021, Russia; (O.N.S.); (N.N.M.)
| | - Natalia N. Markelova
- Gause Institute of New Antibiotics, 11 Bolshaya Pirogovskaya St., Moscow 119021, Russia; (O.N.S.); (N.N.M.)
| | - Lyubov E. Grebenkina
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 86 Vernadsky Prospekt, Moscow 119571, Russia; (E.A.M.); (L.E.G.); (A.V.M.)
| | - Ekaterina A. Lesovaya
- Department of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (D.V.S.); (V.P.M.); (E.A.L.); (M.G.Y.)
- Faculty of Oncology, I.P. Pavlov Ryazan State Medical University, Ministry of Health of Russia, 9 Vysokovol’tnaya St., Ryazan 390026, Russia
- Laboratory of Single Cell Biology, Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow 117198, Russia
| | - Marianna G. Yakubovskaya
- Department of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (D.V.S.); (V.P.M.); (E.A.L.); (M.G.Y.)
| | - Andrey V. Matveev
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 86 Vernadsky Prospekt, Moscow 119571, Russia; (E.A.M.); (L.E.G.); (A.V.M.)
| | - Ekaterina M. Zhidkova
- Department of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (D.V.S.); (V.P.M.); (E.A.L.); (M.G.Y.)
| |
Collapse
|
3
|
Mikhel IB, Bakhrushina EO, Petrusevich DA, Nedorubov AA, Appolonova SA, Moskaleva NE, Demina NB, Kosenkova SI, Parshenkov MA, Krasnyuk II, Krasnyuk II. Development of an Intranasal In Situ System for Ribavirin Delivery: In Vitro and In Vivo Evaluation. Pharmaceutics 2024; 16:1125. [PMID: 39339163 PMCID: PMC11435039 DOI: 10.3390/pharmaceutics16091125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/29/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Recently, ribavirin has demonstrated effectiveness in treating glioblastoma through intranasal administration utilizing the nose-to-brain delivery route. Enhancing ribavirin's bioavailability can be achieved by utilizing intranasal stimuli-responsive systems that create a gel on the nasal mucosa. The research examined thermosensitive, pH-sensitive, and ion-selective polymers in various combinations and concentrations, chosen in line with the current Quality by Design (QbD) approach in pharmaceutical development. Following a thorough assessment of key parameters, the optimal composition of gellan gum at 0.5%, Poloxamer 124 at 2%, and purified water with ribavirin concentration at 100 mg/mL was formulated and subjected to in vivo testing. Through experiments on male rats, the nose-to-brain penetration mechanism of the active pharmaceutical ingredient (API) was elucidated, showcasing drug accumulation in the olfactory bulbs and brain.
Collapse
Affiliation(s)
- Iosif B. Mikhel
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Elena O. Bakhrushina
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Danila A. Petrusevich
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Andrey A. Nedorubov
- Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia;
| | - Svetlana A. Appolonova
- Centre of Biopharmaceutical Analysis and Metabolomics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (S.A.A.); (N.E.M.)
| | - Natalia E. Moskaleva
- Centre of Biopharmaceutical Analysis and Metabolomics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (S.A.A.); (N.E.M.)
| | - Natalia B. Demina
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Svetlana I. Kosenkova
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Mikhail A. Parshenkov
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Ivan I. Krasnyuk
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| | - Ivan I. Krasnyuk
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119048, Russia; (E.O.B.); (D.A.P.); (N.B.D.); (S.I.K.); (M.A.P.); (I.I.K.J.); (I.I.K.)
| |
Collapse
|
4
|
Tang WD, Tang HL, Peng HR, Ren RW, Zhao P, Zhao LJ. Inhibition of tick-borne encephalitis virus in cell cultures by ribavirin. Front Microbiol 2023; 14:1182798. [PMID: 37378295 PMCID: PMC10291047 DOI: 10.3389/fmicb.2023.1182798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Tick-borne encephalitis virus (TBEV) belonging to arboviruses is a major member of zoonotic pathogens. TBEV infection causes severe human encephalitis without specific antiviral drugs. Due to its use of antiviral drug against a wide range of viruses, we investigated antiviral effect of ribavirin against TBEV in susceptible human cell lines A549 and SH-SY5Y. Ribavirin displayed minor cytotoxicity on multiple cell lines. Ribavirin obviously impaired TBEV replication and protected the infected cells from cytopathic effect. Importantly, ribavirin markedly inhibited TBEV propagation, as evidenced by impairment of TBEV production and viral RNA replication. Treatment with ribavirin (co-treatment and post-treatment) led to a dose-dependent reduction in TBEV titers as well as the viral RNA levels. Antiviral protein myxovirus resistance A mRNA expression was significantly up-regulated and signal transducer and activator of transcription 3 was activated in TBEV-infected A549 cells upon the ribavirin treatment. Induction of inflammatory cytokine tumor necrosis factor alpha by TBEV was decreased in A549 cells with the treatment of ribavirin, whereas interleukin 1 beta release appeared to be unaffected. These results suggest that ribavirin might represent a promising safe and effective antiviral drug against TBEV.
Collapse
Affiliation(s)
- Wan-Da Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Hai-Lin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Hao-Ran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Rui-Wen Ren
- Center for Disease Control and Prevention of Southern Theater Command, Guangzhou, China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Lan-Juan Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
5
|
Montiel-Dávalos A, Ayala Y, Hernández G. The dark side of mRNA translation and the translation machinery in glioblastoma. Front Cell Dev Biol 2023; 11:1086964. [PMID: 36994107 PMCID: PMC10042294 DOI: 10.3389/fcell.2023.1086964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Among the different types of cancer affecting the central nervous system (CNS), glioblastoma (GB) is classified by the World Health Organization (WHO) as the most common and aggressive CNS cancer in adults. GB incidence is more frequent among persons aged 45–55 years old. GB treatments are based on tumor resection, radiation, and chemotherapies. The current development of novel molecular biomarkers (MB) has led to a more accurate prediction of GB progression. Moreover, clinical, epidemiological, and experimental studies have established genetic variants consistently associated with the risk of suffering GB. However, despite the advances in these fields, the survival expectancy of GB patients is still shorter than 2 years. Thus, fundamental processes inducing tumor onset and progression remain to be elucidated. In recent years, mRNA translation has been in the spotlight, as its dysregulation is emerging as a key cause of GB. In particular, the initiation phase of translation is most involved in this process. Among the crucial events, the machinery performing this phase undergoes a reconfiguration under the hypoxic conditions in the tumor microenvironment. In addition, ribosomal proteins (RPs) have been reported to play translation-independent roles in GB development. This review focuses on the research elucidating the tight relationship between translation initiation, the translation machinery, and GB. We also summarize the state-of-the-art drugs targeting the translation machinery to improve patients’ survival. Overall, the recent advances in this field are shedding new light on the dark side of translation in GB.
Collapse
|
6
|
Zhu S, Han X, Yang R, Tian Y, Zhang Q, Wu Y, Dong S, Zhang B. Metabolomics study of ribavirin in the treatment of orthotopic lung cancer based on UPLC-Q-TOF/MS. Chem Biol Interact 2023; 370:110305. [PMID: 36529159 DOI: 10.1016/j.cbi.2022.110305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Ribavirin is a common antiviral drug, especially for patients with hepatitis C. Our recent studies demonstrated that ribavirin showed anti-tumor activity in colorectal cancer and hepatocellular carcinoma, but its effects on lung cancer remains unclear. This study aimed to evaluate the anti-tumor activity of ribavirin against lung cancer and elucidate the underlying mechanism. We established orthotopic mouse model of lung cancer (LLC and GLC-82) and employed an ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS)-based metabolomics approach. We found that ribavirin significantly inhibited the proliferation and colony formation of lung cancer cells. Tumor sizes of orthotopic lung cancer in ribavirin-treated groups were also significantly lower than those in control groups. Metabolomics analysis revealed that ribavirin mainly affected 5 metabolic pathways in orthotopic lung tumor models, taurine and hypotaurine metabolism, nicotinate and nicotinamide metabolism, linoleic acid metabolism, arginine biosynthesis and arachidonic acid metabolism. Furthermore, we identified 5 upregulated metabolites including β-nicotinamide adenine dinucleotide (NAD+), nicotinamide (NAM), taurine, ornithine and citrulline, and 7 downregulated metabolites including 1-methylnicotinamide (MNAM), S-adenosyl-l-homocysteine (SAH), N1-Methyl-2-pyridone-5-carboxamide (2PY), homocysteine (Hcy), linoleic acid, arachidonic acid (AA) and argininosuccinic acid in ribavirin-treated groups. These results provide new insight into the anti-tumor mechanism of ribavirin for lung cancer.
Collapse
Affiliation(s)
- Shihao Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Xiang Han
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Ruiying Yang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yizhen Tian
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qingqing Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yongjie Wu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Shuhong Dong
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Baolai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
7
|
Zhang G, Liu H, Xue T, Kong X, Tian D, Luo L, Yang Y, Xu K, Wei Y, Zhuang Z. Ribavirin extends the lifespan of Caenorhabditis elegans through AMPK-TOR Signaling. Eur J Pharmacol 2023; 946:175548. [PMID: 36706801 DOI: 10.1016/j.ejphar.2023.175548] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
Aging is a process accompanied by widespread degenerative changes which are a major cause of human disease and disability. One goal of aging research is to develop interventions or drugs that can extend organism lifespan and treat age-related diseases. Here, we report the identification of a broad spectrum anti-viral agent, ribavirin, as a potential pharmacological aging intervention. Ribavirin extended the lifespan and healthspan of Caenorhabditis elegans by inhibiting Target of Rapamycin (TOR) signaling and activating AMP-activated protein kinase (AMPK). Moreover, our data indicate that ribavirin activated AMPK by reducing the levels of adenosine triphosphate (ATP) and lysosomal v-ATPase-Ragulator-AXIN Complex. Thus, our studies successfully identify ribavirin as a potential anti-aging drug, and indicate that its anti-aging effect is mediated via AMPK-TOR signaling.
Collapse
Affiliation(s)
- Ganlan Zhang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, 213164, China
| | - Hui Liu
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, 213164, China
| | - Ting Xue
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, 213164, China
| | - Xiangming Kong
- Changzhou Railway Higher Vocational and Technical School, Changzhou, 213011, China
| | - Dongmei Tian
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, 213164, China
| | - Libo Luo
- Changzhou Traditional Chinese Medicine Hospital, Changzhou, 213004, China
| | - Yanhua Yang
- Changzhou No.7 People's Hospital, Changzhou, 213011, China
| | - Keqing Xu
- Changzhou No.7 People's Hospital, Changzhou, 213011, China
| | - Youheng Wei
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Ziheng Zhuang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, 213164, China; Changzhou Traditional Chinese Medicine Hospital, Changzhou, 213004, China.
| |
Collapse
|
8
|
Hosseinalizadeh H, Ebrahimi A, Tavakoli A, Monavari SH. Glioblastoma as a Novel Drug Repositioning Target: Updated State. Anticancer Agents Med Chem 2023; 23:1253-1264. [PMID: 36733195 DOI: 10.2174/1871520623666230202163112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 02/04/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of adult brain tumor that can arise from a low-grade astrocytoma. In recent decades, several new conventional therapies have been developed that have significantly improved the prognosis of patients with GBM. Nevertheless, most patients have a limited long-term response to these treatments and survive < 1 year. Therefore, innovative anti-cancer drugs that can be rapidly approved for patient use are urgently needed. One way to achieve accelerated approval is drug repositioning, extending the use of existing drugs for new therapeutic purposes, as it takes less time to validate their biological activity as well as their safety in preclinical models. In this review, a comprehensive analysis of the literature search was performed to list drugs with antiviral, antiparasitic, and antidepressant properties that may be effective in GBM and their putative anti-tumor mechanisms in GBM cells.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ammar Ebrahimi
- Department of Biomedical Sciences, University of Lausanne, Rue Du Bugnon, Lausanne, Switzerland
| | - Ahmad Tavakoli
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
9
|
Ali M, Wani SUD, Masoodi MH, Khan NA, Shivakumar HG, Osmani RMA, Khan KA. Global Effect of COVID-19 Pandemic on Cancer Patients and its Treatment: A Systematic Review. CLINICAL COMPLEMENTARY MEDICINE AND PHARMACOLOGY 2022; 2:100041. [PMID: 36377228 PMCID: PMC9035683 DOI: 10.1016/j.ccmp.2022.100041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 01/11/2023]
Abstract
Background At a global level, the COVID-19 disease outbreak has had a major impact on health services and has induced disruption in routine care of health institutions, exposing cancer patients to severe risks. To provide uninterrupted tumor treatment throughout a pandemic lockdown is a major obstacle. Coronavirus disease (COVID-19) and its causative virus, SARS-CoV-2, stance considerable challenges for the management of oncology patients. COVID-19 presents particularly severe respiratory and systemic infection in aging and immunosuppressed individuals, including patients with cancer. Objective In the present review, we focused on emergent evidence from cancer sufferers that have been contaminated with COVID-19 and cancer patients who were at higher risk of severe COVID-19, and indicates that anticancer treatment may either rise COVID-19 susceptibility or have a duple therapeutic impact on cancer as well as COVID-19; moreover, how SARS-CoV-2 infection impacts cancer cells. Also, to assess the global effect of the COVID-19 disease outbreak on cancer and its treatment. Methods A literature survey was conducted using PubMed, Web of Science (WOS), Embase, Cochrane Library, China National Knowledge Infrastructure (CNKI), and VIral Protein domain DataBase (VIP DB) between Dec 1, 2019 and Sep 23, 2021, for studies on anticancer treatments in patients with COVID-19. The characteristics of the patients, treatment types, mortality, and other additional outcomes were extracted and pooled for synthesis. Results This disease has a huge effect on sufferers who have cancer(s). Sufferers of COVID-19 have a greater percentage of tumor diagnoses than the rest of the population. Likewise, cancer and highest proportion is lung cancer sufferers are more susceptible to COVID-19 constriction than the rest of the population. Conclusion Sufferers who have both COVID-19 and tumor have a considerably elevated death risk than single COVID-19 positive patients overall. During the COVID-19 pandemic, there was a reduction in the screening of cancer and detection, and also deferral of routine therapies, which may contribute to an increase in cancer mortality there in future.
Collapse
Affiliation(s)
- Mohammad Ali
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore 560001, India
| | - Shahid Ud Din Wani
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar 190006, India
| | - Mubashir Hussain Masoodi
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar 190006, India
| | - Nisar Ahmad Khan
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar 190006, India
| | - H G Shivakumar
- College of Pharmacy, JSS Academy of Technical Education, Noida 201301, India
| | - Riyaz M Ali Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Khalid Ahmed Khan
- Assistant Drugs Controller, Drugs Control Department, Government of Karnataka, Bengaluru, Karnataka 560004, India
| |
Collapse
|
10
|
Zhang Q, Yang R, Tian Y, Ge S, Nan X, Zhu S, Dong S, Zhang B. Ribavirin inhibits cell proliferation and metastasis and prolongs survival in soft tissue sarcomas by downregulating both protein arginine methyltransferases 1 and 5. Basic Clin Pharmacol Toxicol 2022; 131:18-33. [PMID: 35470570 DOI: 10.1111/bcpt.13736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 04/10/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022]
Abstract
Protein arginine methyltransferases 1 and 5 (PRMT1 and PRMT5) are frequently overexpressed in diverse types of cancers and correlate with poor prognosis, thus making these enzymes potential therapeutic targets. The aim of this study was to assess and elucidate the anti-tumour effect and epigenetic regulatory mechanism of ribavirin in soft tissue sarcomas (STS). We showed that ribavirin inhibited growth and metastasis and prolonged survival in animals bearing STS cells by downregulating the mRNA and protein levels of PRMT1/PRMT5 and attenuating the accumulation of asymmetric and symmetric di-methylation of arginine (ADMA and SDMA). Furthermore, ribavirin lowered the permeability of the peritoneum in KM mice bearing S180 ascites via decreasing the level of vascular endothelial growth factor (VEGF). Ribavirin was a potent inhibitor of cell proliferation and metastasis in STS cells through downregulation of both type I PRMT1 and type II PRMT5. Ribavirin could be used to enhance the efficacy of doxorubicin in STS allograft tumour models.
Collapse
Affiliation(s)
- Qingqing Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ruiying Yang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yizhen Tian
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Suyin Ge
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaojuan Nan
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Shihao Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Shuhong Dong
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Baolai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
11
|
Guerra G, Kachuri L, Wendt G, Hansen HM, Mack SJ, Molinaro AM, Rice T, Bracci P, Wiencke JK, Kasahara N, Eckel-Passow JE, Jenkins RB, Wrensch M, Francis SS. The immunogenetics of viral antigen response is associated with subtype-specific glioma risk and survival. Am J Hum Genet 2022; 109:1105-1116. [PMID: 35550063 PMCID: PMC9247888 DOI: 10.1016/j.ajhg.2022.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/18/2022] [Indexed: 12/14/2022] Open
Abstract
Glioma is a highly fatal cancer with prognostically significant molecular subtypes and few known risk factors. Multiple studies have implicated infections in glioma susceptibility, but evidence remains inconsistent. Genetic variants in the human leukocyte antigen (HLA) region modulate host response to infection and have been linked to glioma risk. In this study, we leveraged genetic predictors of antibody response to 12 viral antigens to investigate the relationship with glioma risk and survival. Genetic reactivity scores (GRSs) for each antigen were derived from genome-wide-significant (p < 5 × 10-8) variants associated with immunoglobulin G antibody response in the UK Biobank cohort. We conducted parallel analyses of glioma risk and survival for each GRS and HLA alleles imputed at two-field resolution by using data from 3,418 glioma-affected individuals subtyped by somatic mutations and 8,156 controls. Genetic reactivity scores to Epstein-Barr virus (EBV) ZEBRA and EBNA antigens and Merkel cell polyomavirus (MCV) VP1 antigen were associated with glioma risk and survival (Bonferroni-corrected p < 0.01). GRSZEBRA and GRSMCV were associated in opposite directions with risk of IDH wild-type gliomas (ORZEBRA = 0.91, p = 0.0099/ORMCV = 1.11, p = 0.0054). GRSEBNA was associated with both increased risk for IDH mutated gliomas (OR = 1.09, p = 0.040) and improved survival (HR = 0.86, p = 0.010). HLA-DQA1∗03:01 was significantly associated with decreased risk of glioma overall (OR = 0.85, p = 3.96 × 10-4) after multiple testing adjustment. This systematic investigation of the role of genetic determinants of viral antigen reactivity in glioma risk and survival provides insight into complex immunogenomic mechanisms of glioma pathogenesis. These results may inform applications of antiviral-based therapies in glioma treatment.
Collapse
Affiliation(s)
- Geno Guerra
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | - Linda Kachuri
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - George Wendt
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Steven J Mack
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Paige Bracci
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA; Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Nori Kasahara
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, USA
| | | | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen S Francis
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, USA.
| |
Collapse
|
12
|
Ribavirin inhibits the growth and ascites formation of hepatocellular carcinoma through downregulation of type I CARM1 and type II PRMT5. Toxicol Appl Pharmacol 2021; 435:115829. [PMID: 34919946 DOI: 10.1016/j.taap.2021.115829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Type I co-activator-associated arginine methyltransferase 1 (CARM1) and type II protein arginine methyltransferase 5 (PRMT5) are highly expressed in multiple cancers including liver cancer and their overexpression contributes to poor prognosis, thus making them promising therapeutic targets. Here, we evaluated anti-tumor activity of ribavirin in hepatocellular carcinoma (HCC). We found that ribavirin significantly inhibited the proliferation of HCC cells in a time- and dose-dependent manner. Furthermore, ribavirin suppressed the growth of subcutaneous and orthotopic xenograft of HCC in mice, decreased vascular endothelial growth factor (VEGF) and peritoneal permeability to reduce ascites production, and prolonged the survival of mice in HCC ascites tumor models. Mechanistically, ribavirin potently down-regulated global protein expression of CARM1 and PRMT5, and concurrently decreased accumulation of H3R17me2a and H3R8me2s/H4R3me2s. However, ribavirin did not affect the activity and mRNA levels of both CARM1 and PRMT5 in vivo and in vitro HCC cells. In addition, our ChIP results shown that ribavirin inhibited CARM1 which in turn decreased the H3R17me2a, binds to the eukaryotic translation initiation factor 4E (eIF4E) and VEGF promoter region, and reduced the relative mRNA expression level of eIF4E and VEGF in HCC cells. Our findings suggested a potential therapeutic strategy for patients with HCC through inhibition of the abnormal activation/expression of both CARM1 and PRMT5.
Collapse
|
13
|
Drug Repurposing for Glioblastoma and Current Advances in Drug Delivery-A Comprehensive Review of the Literature. Biomolecules 2021; 11:biom11121870. [PMID: 34944514 PMCID: PMC8699739 DOI: 10.3390/biom11121870] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/19/2021] [Accepted: 12/03/2021] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults with an extremely poor prognosis. There is a dire need to develop effective therapeutics to overcome the intrinsic and acquired resistance of GBM to current therapies. The process of developing novel anti-neoplastic drugs from bench to bedside can incur significant time and cost implications. Drug repurposing may help overcome that obstacle. A wide range of drugs that are already approved for clinical use for the treatment of other diseases have been found to target GBM-associated signaling pathways and are being repurposed for the treatment of GBM. While many of these drugs are undergoing pre-clinical testing, others are in the clinical trial phase. Since GBM stem cells (GSCs) have been found to be a main source of tumor recurrence after surgery, recent studies have also investigated whether repurposed drugs that target these pathways can be used to counteract tumor recurrence. While several repurposed drugs have shown significant efficacy against GBM cell lines, the blood–brain barrier (BBB) can limit the ability of many of these drugs to reach intratumoral therapeutic concentrations. Localized intracranial delivery may help to achieve therapeutic drug concentration at the site of tumor resection while simultaneously minimizing toxicity and side effects. These strategies can be considered while repurposing drugs for GBM.
Collapse
|
14
|
Sheikhi M, Shahab S, Balali E, Alnajjar R, Kaviani S, Khancheuski M, Al Saud S. Study of the Ribavirin drug adsorption on the surfaces of carbon nanotube and graphene nanosheet using density functional theory calculations. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Siyamak Shahab
- Belarusian State University, ISEI BSU Minsk Belarus
- Institute of Physical Organic Chemistry National Academy of Sciences of Belarus Minsk Belarus
- Institute of Chemistry of New Materials National Academy of Sciences of Belarus Minsk Belarus
| | - Ebrahim Balali
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry Tehran Medical Sciences, Islamic Azad University Tehran Iran
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science University of Benghazi Benghazi Libya
- Department of Chemistry University of Cape Town Rondebosch South Africa
| | - Sadegh Kaviani
- Research Center for Modeling and Computational Sciences, Department of Chemistry, Faculty of Science Ferdowsi University of Mashhad Mashhad Iran
| | | | | |
Collapse
|
15
|
Lehman SL, Wilson ED, Camphausen K, Tofilon PJ. Translation Initiation Machinery as a Tumor Selective Target for Radiosensitization. Int J Mol Sci 2021; 22:ijms221910664. [PMID: 34639005 PMCID: PMC8508945 DOI: 10.3390/ijms221910664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 01/04/2023] Open
Abstract
Towards improving the efficacy of radiotherapy, one approach is to target the molecules and processes mediating cellular radioresponse. Along these lines, translational control of gene expression has been established as a fundamental component of cellular radioresponse, which suggests that the molecules participating in this process (i.e., the translational machinery) can serve as determinants of radiosensitivity. Moreover, the proteins comprising the translational machinery are often overexpressed in tumor cells suggesting the potential for tumor specific radiosensitization. Studies to date have shown that inhibiting proteins involved in translation initiation, the rate-limiting step in translation, specifically the three members of the eIF4F cap binding complex eIF4E, eIF4G, and eIF4A as well as the cap binding regulatory kinases mTOR and Mnk1/2, results in the radiosensitization of tumor cells. Because ribosomes are required for translation initiation, inhibiting ribosome biogenesis also appears to be a strategy for radiosensitization. In general, the radiosensitization induced by targeting the translation initiation machinery involves inhibition of DNA repair, which appears to be the consequence of a reduced expression of proteins critical to radioresponse. The availability of clinically relevant inhibitors of this component of the translational machinery suggests opportunities to extend this approach to radiosensitization to patient care.
Collapse
|
16
|
Raj D, Agrawal P, Gaitsch H, Wicks E, Tyler B. Pharmacological strategies for improving the prognosis of glioblastoma. Expert Opin Pharmacother 2021; 22:2019-2031. [PMID: 34605345 PMCID: PMC8603465 DOI: 10.1080/14656566.2021.1948013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022]
Abstract
Introduction: Treatments for brain cancer have radically evolved in the past decade due to a better understanding of the interplay between the immune system and tumors of the central nervous system (CNS). However, glioblastoma multiforme (GBM) remains the most common and lethal CNS malignancy affecting adults.Areas covered: The authors review the literature on glioblastoma pharmacologic therapies with a focus on trials of combination chemo-/immunotherapies and drug delivery platforms from 2015 to 2021.Expert opinion: Few therapeutic advances in GBM treatment have been made since the Food and Drug Administration (FDA) approval of the BCNU-eluting wafer, Gliadel, in 1996 and oral temozolomide (TMZ) in 2005. Recent advances in our understanding of GBM have promoted a wide assortment of new therapeutic approaches including combination therapy, immunotherapy, vaccines, and Car T-cell therapy along with developments in drug delivery. Given promising preclinical data, these novel pharmacotherapies for the treatment of GBM are currently being evaluated in various stages of clinical trials.
Collapse
Affiliation(s)
- Divyaansh Raj
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Pranjal Agrawal
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Hallie Gaitsch
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Elizabeth Wicks
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
17
|
Repurposing of Antimicrobial Agents for Cancer Therapy: What Do We Know? Cancers (Basel) 2021; 13:cancers13133193. [PMID: 34206772 PMCID: PMC8269327 DOI: 10.3390/cancers13133193] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
The substantial costs of clinical trials, the lengthy timelines of new drug discovery and development, along the high attrition rates underscore the need for alternative strategies for finding quickly suitable therapeutics agents. Given that most approved drugs possess more than one target tightly linked to other diseases, it encourages promptly testing these drugs in patients. Over the past decades, this has led to considerable attention for drug repurposing, which relies on identifying new uses for approved or investigational drugs outside the scope of the original medical indication. The known safety of approved drugs minimizes the possibility of failure for adverse toxicology, making them attractive de-risked compounds for new applications with potentially lower overall development costs and shorter development timelines. This latter case is an exciting opportunity, specifically in oncology, due to increased resistance towards the current therapies. Indeed, a large body of evidence shows that a wealth of non-cancer drugs has beneficial effects against cancer. Interestingly, 335 drugs are currently being evaluated in different clinical trials for their potential activities against various cancers (Redo database). This review aims to provide an extensive discussion about the anti-cancer activities exerted by antimicrobial agents and presents information about their mechanism(s) of action and stage of development/evaluation.
Collapse
|
18
|
An Alternative Pipeline for Glioblastoma Therapeutics: A Systematic Review of Drug Repurposing in Glioblastoma. Cancers (Basel) 2021; 13:cancers13081953. [PMID: 33919596 PMCID: PMC8073966 DOI: 10.3390/cancers13081953] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Glioblastoma is a devastating malignancy that has continued to prove resistant to a variety of therapeutics. No new systemic therapy has been approved for use against glioblastoma in almost two decades. This observation is particularly disturbing given the amount of money invested in identifying novel therapies for this disease. A relatively rapid and economical pipeline for identification of novel agents is drug repurposing. Here, a comprehensive review detailing the state of drug repurposing in glioblastoma is provided. We reveal details on studies that have examined agents in vitro, in animal models and in patients. While most agents have not progressed beyond the initial stages, several drugs, from a variety of classes, have demonstrated promising results in early phase clinical trials. Abstract The treatment of glioblastoma (GBM) remains a significant challenge, with outcome for most pa-tients remaining poor. Although novel therapies have been developed, several obstacles restrict the incentive of drug developers to continue these efforts including the exorbitant cost, high failure rate and relatively small patient population. Repositioning drugs that have well-characterized mechanistic and safety profiles is an attractive alternative for drug development in GBM. In ad-dition, the relative ease with which repurposed agents can be transitioned to the clinic further supports their potential for examination in patients. Here, a systematic analysis of the literature and clinical trials provides a comprehensive review of primary articles and unpublished trials that use repurposed drugs for the treatment of GBM. The findings demonstrate that numerous drug classes that have a range of initial indications have efficacy against preclinical GBM models and that certain agents have shown significant potential for clinical benefit. With examination in randomized, placebo-controlled trials and the targeting of particular GBM subgroups, it is pos-sible that repurposing can be a cost-effective approach to identify agents for use in multimodal anti-GBM strategies.
Collapse
|
19
|
Huq S, Kannapadi NV, Casaos J, Lott T, Felder R, Serra R, Gorelick NL, Ruiz-Cardozo MA, Ding AS, Cecia A, Medikonda R, Ehresman J, Brem H, Skuli N, Tyler BM. Preclinical efficacy of ribavirin in SHH and group 3 medulloblastoma. J Neurosurg Pediatr 2021; 27:482-488. [PMID: 33545678 DOI: 10.3171/2020.8.peds20561] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/24/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Medulloblastoma, the most common pediatric brain malignancy, has Sonic Hedgehog (SHH) and group 3 (Myc driven) subtypes that are associated with the activity of eukaryotic initiation factor 4E (eIF4E), a critical mediator of translation, and enhancer of zeste homolog 2 (EZH2), a histone methyltransferase and master regulator of transcription. Recent drug repurposing efforts in multiple solid and hematologic malignancies have demonstrated that eIF4E and EZH2 are both pharmacologically inhibited by the FDA-approved antiviral drug ribavirin. Given the molecular overlap between medulloblastoma biology and known ribavirin activity, the authors investigated the preclinical efficacy of repurposing ribavirin as a targeted therapeutic in cell and animal models of medulloblastoma. METHODS Multiple in vitro assays were performed using human ONS-76 (a primitive SHH model) and D425 (an aggressive group 3 model) cells. The impacts of ribavirin on cellular growth, death, migration, and invasion were quantified using proliferation and Cell Counting Kit-8 (CCK-8) assays, flow cytometry with annexin V (AnnV) staining, scratch wound assays, and Matrigel invasion chambers, respectively. Survival following daily ribavirin treatment (100 mg/kg) was assessed in vivo in immunodeficient mice intracranially implanted with D425 cells. RESULTS Compared to controls, ribavirin treatment led to a significant reduction in medulloblastoma cell growth (ONS-76 proliferation assay, p = 0.0001; D425 CCK-8 assay, p < 0.0001) and a significant increase in cell death (flow cytometry for AnnV, ONS-76, p = 0.0010; D425, p = 0.0284). In ONS-76 cells, compared to controls, ribavirin significantly decreased cell migration and invasion (Matrigel invasion chamber assay, p = 0.0012). In vivo, ribavirin significantly extended survival in an aggressive group 3 medulloblastoma mouse model compared to vehicle-treated controls (p = 0.0004). CONCLUSIONS The authors demonstrate that ribavirin, a clinically used drug known to inhibit eIF4E and EZH2, has significant antitumor effects in multiple preclinical models of medulloblastoma, including an aggressive group 3 animal model. Ribavirin may represent a promising targeted therapeutic in medulloblastoma.
Collapse
|
20
|
Ge S, Zhang Q, Chen Y, Tian Y, Yang R, Chen X, Li F, Zhang B. Ribavirin inhibits colorectal cancer growth by downregulating PRMT5 expression and H3R8me2s and H4R3me2s accumulation. Toxicol Appl Pharmacol 2021; 415:115450. [PMID: 33577917 DOI: 10.1016/j.taap.2021.115450] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 12/25/2022]
Abstract
Eukaryotic translation initiation factor 4E (eIF4E) and protein arginine methyltransferase 5 (PRMT5) are frequently overexpressed in colorectal cancer (CRC) tissues and associated with poor prognosis. Ribavirin, the only clinically approved drug known to target eIF4E, is an anti-viral molecule currently used in hepatitis C therapy. The potential of ribavirin to treat CRC remains largely unknown. Ribavirin treatment in CRC cell lines drastically inhibited cell proliferation and colony formation, induced S phase arrest and reduced cyclin D1, cyclin A/E and proliferating cell nuclear antigen (PCNA) levels in vitro, and suppressed tumorigenesis in mouse model of colitis-associated CRC. Mechanistically, ribavirin treatment significantly reduced PRMT5 and eIF4E protein levels and the accumulation of symmetric dimethylation of histone 3 at arginine 8 (H3R8me2s) and that of histone 4 at arginine 3 (H4R3me2s). Importantly, inhibition of PRMT5 by ribavirin resulted in promoted H3R8 methylation in eIF4E promoter region. Our results demonstrate the anti-cancer efficacy of ribavirin in CRC and suggest that the anti-cancer efficacy of ribavirin may be mediated by downregulating PRMT5 levels but not its enzymatic activity.
Collapse
Affiliation(s)
- Suyin Ge
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Qingqing Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Yonglin Chen
- Department of Pathology, First Hospital, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Yizhen Tian
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Ruiying Yang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Xu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Fang Li
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Baolai Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
21
|
The immuno-oncological challenge of COVID-19. ACTA ACUST UNITED AC 2020; 1:946-964. [DOI: 10.1038/s43018-020-00122-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
|
22
|
Ochiai Y, Sumi K, Sano E, Yoshimura S, Yamamuro S, Ogino A, Ueda T, Suzuki Y, Nakayama T, Hara H, Katayama Y, Yoshino A. Antitumor effects of ribavirin in combination with TMZ and IFN-β in malignant glioma cells. Oncol Lett 2020; 20:178. [PMID: 32934745 PMCID: PMC7475644 DOI: 10.3892/ol.2020.12039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
The prognosis of gioblastoma, the standard chemotherapy agent for which is temozolomide (TMZ), remains poor despite recent advances in multimodal treatments. Therefore, it is necessary to identify and develop novel therapeutics for this malignant disease. Ribavirin, an anti-viral agent which is one of the standard agents for treatment of chronic hepatitis C in combination with interferon (IFN), was recently revealed to have an antitumor potential towards various tumor cells, including malignant glioma cells. The aim of the present study was to examine the antitumor effect of ribavirin in combination with TMZ and IFN-β on glioma cells and to evaluate the possibility that such combinations might represent a novel candidate for glioblastoma therapy. The combination of ribavirin with TMZ and IFN-β displayed a significant cell growth inhibitory effect with a ribavirin dose-dependency, including a relatively low concentration of ribavirin, on not only TMZ-sensitive but also TMZ-resistant malignant glioma cells. The antitumor efficacy of such a combination further indicated a synergistic interaction when assessed by the Chou-Talalay method. Furthermore, flow cytometry analysis suggested that apoptosis induction was one of the possible biological processes underlying the synergistic antitumor effect of these triple combination treatments. Therefore, such combinations may be potentially important in the clinical setting for glioblastoma treatment, although further detailed studies, e.g. on the adverse effects, are required.
Collapse
Affiliation(s)
- Yushi Ochiai
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Koichiro Sumi
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Emiko Sano
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Sodai Yoshimura
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shun Yamamuro
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Akiyoshi Ogino
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Tomohiro Nakayama
- Division of Companion Diagnostics, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroyuki Hara
- Division of Functional Morphology, Department of Functional Morphology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoichi Katayama
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan.,Center for Brain and Health Sciences, Aomori University, Aomori 038-0003, Japan
| | - Atsuo Yoshino
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
23
|
Inhibition of eIF4E signaling by ribavirin selectively targets lung cancer and angiogenesis. Biochem Biophys Res Commun 2020; 529:519-525. [PMID: 32736668 DOI: 10.1016/j.bbrc.2020.05.127] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/22/2022]
Abstract
Although the introduction of immune- and targeted-therapy has improved the clinical response and outcomes, lung cancer remains a therapeutic challenge. Developing new therapeutics is necessary to improve the treatment of lung cancer. Here, we show that ribavirin, a clinically available anti-viral drug, is an attractive candidate for lung cancer treatment. We show that ribavirin is active against a panel of lung cancer cell lines regardless of molecular and cellular heterogeneity. Notably, the effective concentrations of ribavirin are clinically achievable, display minimal toxicity to normal cells and synergistic effect with paclitaxel. Its potent efficacy and synergism with chemotherapy on cancer cell, and minimal toxicity on normal cells are observed in lung xenograft mouse model. Ribavirin is also an angiogenesis inhibitor as it inhibits capillary network formation, growth and survival of human lung tumor-associated endothelial cell (HLT-EC). The mechanism studies demonstrate that ribavirin acts on lung cancer cells via suppressing eIF4E and mTOR signaling, leading to the subsequent inhibition of eIF4E-mediated protein translation. Our work suggests that ribavirin has advantage than many anti-cancer agents by targeting both tumor cells and angiogenesis. Our work also highlights the therapeutic potential of ribavirin for the treatment of lung cancer.
Collapse
|
24
|
Huq S, Casaos J, Serra R, Peters M, Xia Y, Ding AS, Ehresman J, Kedda JN, Morales M, Gorelick NL, Zhao T, Ishida W, Perdomo-Pantoja A, Cecia A, Ji C, Suk I, Sidransky D, Brait M, Brem H, Skuli N, Tyler B. Repurposing the FDA-Approved Antiviral Drug Ribavirin as Targeted Therapy for Nasopharyngeal Carcinoma. Mol Cancer Ther 2020; 19:1797-1808. [PMID: 32606016 DOI: 10.1158/1535-7163.mct-19-0572] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/09/2019] [Accepted: 06/09/2020] [Indexed: 11/16/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a squamous cell carcinoma with a proclivity for systemic dissemination, leading many patients to present with advanced stage disease and fail available treatments. There is a notable lack of targeted therapies for NPC, despite working knowledge of multiple proteins with integral roles in NPC cancer biology. These proteins include EZH2, Snail, eIF4E, and IMPDH, which are all overexpressed in NPC and correlated with poor prognosis. These proteins are known to be modulated by ribavirin, an FDA-approved hepatitis C antiviral that has recently been repurposed as a promising therapeutic in several solid and hematologic malignancies. Here, we investigated the potential of ribavirin as a targeted anticancer agent in five human NPC cell lines. Using cellular growth assays, flow cytometry, BrdU cell proliferation assays, scratch wound assays, and invasion assays, we show in vitro that ribavirin decreases NPC cellular proliferation, migration, and invasion and promotes cell-cycle arrest and cell death. Modulation of EZH2, Snail, eIF4E, IMPDH, mTOR, and cyclin D1 were observed in Western blots and enzymatic activity assays in response to ribavirin treatment. As monotherapy, ribavirin reduced flank tumor growth in multiple NPC xenograft models in vivo Most importantly, we demonstrate that ribavirin enhanced the effects of radiotherapy, a central component of NPC treatment, both in vitro and in vivo Our work suggests that NPC responds to ribavirin-mediated EZH2, Snail, eIF4E, IMPDH, and mTOR changes and positions ribavirin for clinical evaluation as a potential addition to our NPC treatment armamentarium.
Collapse
Affiliation(s)
- Sakibul Huq
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joshua Casaos
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Riccardo Serra
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Peters
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuanxuan Xia
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andy S Ding
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeff Ehresman
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jayanidhi N Kedda
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Manuel Morales
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noah L Gorelick
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tianna Zhao
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Wataru Ishida
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander Perdomo-Pantoja
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Arba Cecia
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chenchen Ji
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ian Suk
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David Sidransky
- Head and Neck Cancer Research Laboratory, Department of Otolaryngology and Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mariana Brait
- Head and Neck Cancer Research Laboratory, Department of Otolaryngology and Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henry Brem
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Departments of Biomedical Engineering, Oncology, and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicolas Skuli
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Betty Tyler
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
25
|
Ciliberto G, Mancini R, Paggi MG. Drug repurposing against COVID-19: focus on anticancer agents. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:86. [PMID: 32398164 PMCID: PMC7214852 DOI: 10.1186/s13046-020-01590-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
Background The very limited time allowed to face the COVID-19 pandemic poses a pressing challenge to find proper therapeutic approaches. However, synthesis and full investigation from preclinical studies to phase III trials of new medications is a time-consuming procedure, and not viable in a global emergency, such as the one we are facing. Main Body Drug repurposing/repositioning, a strategy effectively employed in cancer treatment, can represent a valid alternative. Most drugs considered for repurposing/repositioning in the therapy of the COVID-19 outbreak are commercially available and their dosage and toxicity in humans is well known, due to years (or even decades) of clinical use. This can allow their fast-track evaluation in phase II–III clinical trials, or even within straightforward compassionate use. Several drugs being re-considered for COVID-19 therapy are or have been used in cancer therapy. Indeed, virus-infected cells are pushed to enhance the synthesis of nucleic acids, protein and lipid synthesis and boost their energy metabolism, in order to comply to the “viral program”. Indeed, the same features are seen in cancer cells, making it likely that drugs interfering with specific cancer cell pathways may be effective as well in defeating viral replication. Short Conclusion To our knowledge, cancer drugs potentially suitable for facing SARS-CoV-2 infection have not been carefully reviewed. We present here a comprehensive analysis of available information on potential candidate cancer drugs that can be repurposed for the treatment of COIVD-19.
Collapse
Affiliation(s)
- Gennaro Ciliberto
- Scientific Director, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Marco G Paggi
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
26
|
Petković B, Kesić S, Pešić V. Critical View on the Usage of Ribavirin in Already Existing Psychostimulant-Use Disorder. Curr Pharm Des 2020; 26:466-484. [PMID: 31939725 PMCID: PMC8383468 DOI: 10.2174/1381612826666200115094642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022]
Abstract
Substance-use disorder represents a frequently hidden non-communicable chronic disease. Patients with intravenous drug addiction are at high risk of direct exposure to a variety of viral infections and are considered to be the largest subpopulation infected with the hepatitis C virus. Ribavirin is a synthetic nucleoside analog that has been used as an integral component of hepatitis C therapy. However, ribavirin medication is quite often associated with pronounced psychiatric adverse effects. It is not well understood to what extent ribavirin per se contributes to changes in drug-related neurobehavioral disturbances, especially in the case of psychostimulant drugs, such as amphetamine. It is now well-known that repeated amphetamine usage produces psychosis in humans and behavioral sensitization in animals. On the other hand, ribavirin has an affinity for adenosine A1 receptors that antagonistically modulate the activity of dopamine D1 receptors, which play a critical role in the development of behavioral sensitization. This review will focus on the current knowledge of neurochemical/ neurobiological changes that exist in the psychostimulant drug-addicted brain itself and the antipsychotic-like efficiency of adenosine agonists. Particular attention will be paid to the potential side effects of ribavirin therapy, and the opportunities and challenges related to its application in already existing psychostimulant-use disorder.
Collapse
Affiliation(s)
- Branka Petković
- Address correspondence to this author at the Department of Neurophysiology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana Blvd. 142, 11060, Belgrade, Serbia; Tel: +381-11-20-78-300; Fax: +381-11-27-61-433; E-mail:
| | | | | |
Collapse
|
27
|
Marina D, Arnaud L, Paul Noel L, Felix S, Bernard R, Natacha C. Relevance of Translation Initiation in Diffuse Glioma Biology and its Therapeutic Potential. Cells 2019; 8:E1542. [PMID: 31795417 PMCID: PMC6953081 DOI: 10.3390/cells8121542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer cells are continually exposed to environmental stressors forcing them to adapt their protein production to survive. The translational machinery can be recruited by malignant cells to synthesize proteins required to promote their survival, even in times of high physiological and pathological stress. This phenomenon has been described in several cancers including in gliomas. Abnormal regulation of translation has encouraged the development of new therapeutics targeting the protein synthesis pathway. This approach could be meaningful for glioma given the fact that the median survival following diagnosis of the highest grade of glioma remains short despite current therapy. The identification of new targets for the development of novel therapeutics is therefore needed in order to improve this devastating overall survival rate. This review discusses current literature on translation in gliomas with a focus on the initiation step covering both the cap-dependent and cap-independent modes of initiation. The different translation initiation protagonists will be described in normal conditions and then in gliomas. In addition, their gene expression in gliomas will systematically be examined using two freely available datasets. Finally, we will discuss different pathways regulating translation initiation and current drugs targeting the translational machinery and their potential for the treatment of gliomas.
Collapse
Affiliation(s)
- Digregorio Marina
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; (D.M.); (L.A.); (L.P.N.); (S.F.); (R.B.)
| | - Lombard Arnaud
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; (D.M.); (L.A.); (L.P.N.); (S.F.); (R.B.)
- Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium
| | - Lumapat Paul Noel
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; (D.M.); (L.A.); (L.P.N.); (S.F.); (R.B.)
| | - Scholtes Felix
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; (D.M.); (L.A.); (L.P.N.); (S.F.); (R.B.)
- Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium
| | - Rogister Bernard
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; (D.M.); (L.A.); (L.P.N.); (S.F.); (R.B.)
- Department of Neurology, CHU of Liège, 4000 Liège, Belgium
| | - Coppieters Natacha
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; (D.M.); (L.A.); (L.P.N.); (S.F.); (R.B.)
| |
Collapse
|
28
|
Casaos J, Gorelick NL, Huq S, Choi J, Xia Y, Serra R, Felder R, Lott T, Kast RE, Suk I, Brem H, Tyler B, Skuli N. The Use of Ribavirin as an Anticancer Therapeutic: Will It Go Viral? Mol Cancer Ther 2019; 18:1185-1194. [DOI: 10.1158/1535-7163.mct-18-0666] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/25/2019] [Accepted: 05/08/2019] [Indexed: 11/16/2022]
|
29
|
Ravichandran L, Venkatesan A, Febin Prabhu Dass J. Epitope-based immunoinformatics approach on RNA-dependent RNA polymerase (RdRp) protein complex of Nipah virus (NiV). J Cell Biochem 2019; 120:7082-7095. [PMID: 30417438 DOI: 10.1002/jcb.27979] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/04/2018] [Indexed: 01/24/2023]
Abstract
Persistent outbreaks of Nipah virus (NiV) with severe case fatality throw a major challenge on researchers to develop a drug or vaccine to combat the disease. With little knowledge of its molecular mechanisms, we utilized the proteome data of NiV to evaluate the potency of three major proteins (phosphoprotein, polymerase, and nucleocapsid protein) in the RNA-dependent RNA polymerase complex to count as a possible candidate for epitope-based vaccine design. Profound computational analysis was used on the above proteins individually to explore the T-cell immune properties like antigenicity, immunogenicity, binding to major histocompatibility complex class I and class II alleles, conservancy, toxicity, and population coverage. Based on these predictions the peptide 'ELRSELIGY' of phosphoprotein and 'YPLLWSFAM' of nulceocapsid protein were identified as the best-predicted T-cell epitopes and molecular docking with human leukocyte antigen-C (HLA-C*12:03) molecule was effectuated followed by validation with molecular dynamics simulation. The B-cell epitope predictions suggest that the sequence positions 421 to 471 in phosphoprotein, 606 to 640 in polymerase and 496 to 517 in nucleocapsid protein are the best-predicted regions for B-cell immune response. However, the further experimental circumstance is required to test and validate the efficacy of the subunit peptide for potential candidacy against NiV.
Collapse
Affiliation(s)
- Lavanya Ravichandran
- Department of Integrative Biology, School of BioSciences and Technology (SBST), VIT, Vellore, India
| | - Arthi Venkatesan
- Department of Integrative Biology, School of BioSciences and Technology (SBST), VIT, Vellore, India
| | - J Febin Prabhu Dass
- Department of Integrative Biology, School of BioSciences and Technology (SBST), VIT, Vellore, India
| |
Collapse
|
30
|
Jin J, Xiang W, Wu S, Wang M, Xiao M, Deng A. Targeting eIF4E signaling with ribavirin as a sensitizing strategy for ovarian cancer. Biochem Biophys Res Commun 2019; 510:580-586. [PMID: 30739792 DOI: 10.1016/j.bbrc.2019.01.117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/26/2019] [Indexed: 12/20/2022]
Abstract
The essential roles of eukaryotic translation initiation factor 4E (eIF4E) have been shown in various cancers, including ovarian cancer. In this work, we demonstrate that eIF4E inhibition in ovarian cancer can be achieved by ribavirin, a FDA-approved antiviral drug. We show that ribavirin at clinically relevant doses significantly inhibits growth and survival in multiple ovarian cancer cell lines, regardless of morphological and molecular subtypes. Mechanistically, ribavirin suppresses Akt/mTOR and eIF4E/p70S6K signaling pathways in ovarian cancer cells. We confirm that eIF4E is the critical molecular target of ribavirin, and furthermore that this is dependent on phosphorylation at S209. Notably, using both in vitro cell culture system and in vivo xenograft mouse model, we show that the combination of ribavirin with cisplatin (standard of care for patients with ovarian cancer) results in significantly greater efficacy than cisplatin alone in ovarian cancer. Interestingly, the sensitivity to ribavirin varies among a panel of ovarian cancer cell lines, mostly likely due to their differential expression level of eIF4E and dependency to eIF4E inhibition. The differential expression level is further observed in ovarian cancer tissues, with the higher level of eIF4E in the majority of ovarian cancer tissues compared to normal ovary tissues. Our work suggests that eIF4E expression varies among ovarian cancer. Additionally, ribavirin is a useful addition to ovarian cancer treatment, particularly to those with high dependency on eIF4E.
Collapse
Affiliation(s)
- Jing Jin
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Wei Xiang
- Department of Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Shuang Wu
- Department of Obstetrics and Gynecology, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Min Wang
- Department of Obstetrics and Gynecology, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Meifang Xiao
- Department of Clinical Laboratory, Hainan Provincial Women and Children Hospital, Haikou, Hainan Province, China
| | - Ali Deng
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| |
Collapse
|
31
|
Natsumeda M, Liu Y, Nakata S, Miyahara H, Hanaford A, Ahsan S, Stearns D, Skuli N, Kahlert UD, Raabe EH, Rodriguez FJ, Eberhart CG. Inhibition of enhancer of zest homologue 2 is a potential therapeutic target for high-MYC medulloblastoma. Neuropathology 2019; 39:71-77. [PMID: 30632221 DOI: 10.1111/neup.12534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 01/03/2023]
Abstract
MYC amplification is common in Group 3 medulloblastoma and is associated with poor survival. Group 3 and Group 4 medulloblastomas are also known to have elevated levels of histone H3-lysine 27-tri-methylation (H3K27me3), at least in part due to high expression of the H3K27 methyltransferase enhancer of zest homologue 2 (EZH2), which can be regulated by MYC. We therefore examined whether MYC expression is associated with elevated EZH2 and H3K27me3 in medulloblastoma, and if high-MYC medulloblastomas are particularly sensitive to pharmacological EZH2 blockade. Western blot analysis of low (DAOY, UW228, CB SV40) and high (DAOY-MYC, UW228-MYC, CB-MYC, D425) MYC cell lines showed that higher levels of EZH2 and H3K27me3 were associated with elevated MYC. In fixed medulloblastoma samples examined using immunohistochemistry, most MYC positive tumors also had high H3K27me3, but many MYC negative ones did as well, and the correlation was not statistically significant. All high MYC lines tested were sensitive to the EZH2 inhibitor EPZ6438. Many low MYC lines also grew more slowly in the presence of EPZ6438, although DAOY-MYC cells responded more strongly than parent DAOY cultures with lower MYC levels. We find that higher MYC levels are associated with increased EZH2, and pharmacological blockade of EZH2 is a potential therapeutic strategy for aggressive medulloblastoma with elevated MYC.
Collapse
Affiliation(s)
- Manabu Natsumeda
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yang Liu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Satoshi Nakata
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hiroaki Miyahara
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, Oita University Faculty of Medicine, Oita, Japan
| | - Allison Hanaford
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sama Ahsan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Duncan Stearns
- Department of Pediatric Hematology-Oncology, University Hospitals Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicolas Skuli
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Ulf D Kahlert
- Department of Neurosurgery, University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Eric H Raabe
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Targeting EIF4E signaling with ribavirin in infant acute lymphoblastic leukemia. Oncogene 2018; 38:2241-2262. [PMID: 30478448 PMCID: PMC6440839 DOI: 10.1038/s41388-018-0567-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/17/2018] [Accepted: 10/11/2018] [Indexed: 01/02/2023]
Abstract
The poor outcomes in infant acute lymphoblastic leukemia (ALL) necessitate new treatments. Here we discover that EIF4E protein is elevated in most cases of infant ALL and test EIF4E targeting by the repurposed antiviral agent ribavirin, which has anticancer properties through EIF4E inhibition, as a potential treatment. We find that ribavirin treatment of actively dividing infant ALL cells on bone marrow stromal cells (BMSCs) at clinically achievable concentrations causes robust proliferation inhibition in proportion with EIF4E expression. Further, we find that ribavirin treatment of KMT2A-rearranged (KMT2A-R) infant ALL cells and the KMT2A-AFF1 cell line RS4:11 inhibits EIF4E, leading to decreases in oncogenic EIF4E-regulated cell growth and survival proteins. In ribavirin-sensitive KMT2A-R infant ALL cells and RS4:11 cells, EIF4E-regulated proteins with reduced levels of expression following ribavirin treatment include MYC, MCL1, NBN, BCL2 and BIRC5. Ribavirin-treated RS4:11 cells exhibit impaired EIF4E-dependent nuclear to cytoplasmic export and/or translation of the corresponding mRNAs, as well as reduced phosphorylation of the p-AKT1, p-EIF4EBP1, p-RPS6 and p-EIF4E signaling proteins. This leads to an S-phase cell cycle arrest in RS4:11 cells corresponding to the decreased proliferation. Ribavirin causes nuclear EIF4E to re-localize to the cytoplasm in KMT2A-AFF1 infant ALL and RS4:11 cells, providing further evidence for EIF4E inhibition. Ribavirin slows increases in peripheral blasts in KMT2A-R infant ALL xenograft-bearing mice. Ribavirin cooperates with chemotherapy, particularly L-asparaginase, in reducing live KMT2A-AFF1 infant ALL cells in BMSC co-cultures. This work establishes that EIF4E is broadly elevated across infant ALL and that clinically relevant ribavirin exposures have preclinical activity and effectively inhibit EIF4E in KMT2A-R cases, suggesting promise in EIF4E targeting using ribavirin as a means of treatment.
Collapse
|
33
|
Kai J, Wang Y, Xiong F, Wang S. Genetic and pharmacological inhibition of eIF4E effectively targets esophageal cancer cells and augments 5-FU's efficacy. J Thorac Dis 2018; 10:3983-3991. [PMID: 30174840 DOI: 10.21037/jtd.2018.06.43] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Aberrant activation of eIF4E is critically involved in the progression and chemoresistance of various cancers. Elevated expression of eIF4E has also been documented in human cancerous esophageal tissues. However, the role of eIF4E in esophageal cancer is unclear. Methods We analysed the levels of eIF4E expression and eIF4E function in a number of normal and cancerous esophageal cancer cell lines, and studied its underlying mechanism. Results We observed that eIF4E expression varies in different esophageal cancer cell lines but was significantly elevated in all tested esophageal cell lines as compared to the control cell lines. We demonstrated that eIF4E inhibition via genetic and pharmacological approaches inhibits cancer cell growth and survival. This inhibition also augments 5-flurouracil's (5-FU's) efficacy as demonstrated with both the in vitro esophageal cancer culture system and our in vivo xenograft mouse model. Of note, the sensitivity of esophageal cancer cells to ribavirin or eIF4E knockdown correlates well with the expression levels of eIF4E, demonstrating that esophageal cells with higher eIF4E expression are more sensitive to eIF4E inhibition. We further confirmed that the mechanism of action of ribavirin on esophageal cancer cells was through suppressing the Akt/mTOR/eIF4E and eIF4E-regulated pathways. Conclusions To our knowledge, our work is the first to demonstrate the multiple roles of eIF4E in esophageal cancer. eIF4E was shown to promote cancer cell growth and survival, and protected the cells from chemotherapy. Our work also demonstrated that ribavirin is an attractive candidate for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Jindan Kai
- Department of Thoracic Surgery, Hubei Cancer Hospital, Wuhan 430079, China
| | - Yiqiao Wang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Wuhan 430072, China
| | - Fei Xiong
- Department of Thoracic Surgery, Hubei Cancer Hospital, Wuhan 430079, China
| | - Sheng Wang
- Department of Thoracic Surgery, Hubei Cancer Hospital, Wuhan 430079, China
| |
Collapse
|
34
|
Yang Y, Lei T, Du S, Tong R, Wang H, Yang J, Huang J, Sun M, Wang Y, Dong Z. Nuclear GSK3β induces DNA double-strand break repair by phosphorylating 53BP1 in glioblastoma. Int J Oncol 2018; 52:709-720. [PMID: 29328365 PMCID: PMC5807039 DOI: 10.3892/ijo.2018.4237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is the most malignant and lethal subtype brain tumors with high risk of recurrence and therapeutic resistance. Emerging evidence has indicated that glycogen synthesis kinase 3 (GSK3)β plays oncogenic roles in multiple tumor types; however, the underlying mechanisms remain largely unknown. It has also been demonstrated that p53 binding protein 1 (53BP1) plays a central role in DNA double-strand break (DSB) repair. This study aimed to reveal the significance of GSK3β translocation from the cytoplasm to the nucleus, and to determine whether GSK3β induces DNA DSB repair in the nuclei of glioblastoma cells via phospho-53BP1. By performing in vitro experiments, we found that GSK3β translocated from the cytoplasm to the nucleus, and it then bound to 53BP1 following exposure to IR (IR). In addition, 53BP1-mediated DNA DSB repair was observed to be abrogated by the inhibition of GSK3β. Further experiments on the phosphorylation site of 53BP1 by GSK3β revealed that the S/T-Q motif may play a critical role. Importantly, our in vivo and in vitro data clearly indicated that GSK3β induced the phosphorylation of 53BP1 at the Ser166 site. Moreover, brain tumor xenograft models revealed that following exposure to IR plus SB216763, a specific GSK3β inhibitor, tumor growth was markedly inhibited and the survival of mice markedly increased. Based on these results, we concluded that the phosphorylation of 53BP1 by GSK3β was indispensable for DNA DSB repair. Our study also suggested that the inhibition of GSK3β by SB216763 significantly inhibited the proliferation and induced the apoptosis of glioblastoma cells. Taken together, our data indicate that GSK3β, a key phosphorylation protein for 53BP1, may be a potential target for enhancing the sensitivity of glioblastoma cells to radiation.
Collapse
Affiliation(s)
- Yong Yang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610072
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054
| | - Tiantian Lei
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054
| | - Suya Du
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610072
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054
| | - Hailian Wang
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610072
| | - Jiao Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054
| | - Juan Huang
- Department of Pharmacy, Medical Center Hospital of Qionglai, Qionglai, Sichuan 611500
| | - Minghan Sun
- Department of Gynecology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yi Wang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610072
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054
| | - Zhi Dong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016
| |
Collapse
|
35
|
Casaos J, Huq S, Lott T, Felder R, Choi J, Gorelick N, Peters M, Xia Y, Maxwell R, Zhao T, Ji C, Simon T, Sesen J, Scotland SJ, Kast RE, Rubens J, Raabe E, Eberhart CG, Jackson EM, Brem H, Tyler B, Skuli N. Ribavirin as a potential therapeutic for atypical teratoid/rhabdoid tumors. Oncotarget 2018; 9:8054-8067. [PMID: 29487714 PMCID: PMC5814281 DOI: 10.18632/oncotarget.23883] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 11/11/2017] [Indexed: 11/25/2022] Open
Abstract
Atypical teratoid/rhabdoid tumors (AT/RT) are highly aggressive, malignant tumors and are the most common malignant brain tumor in children under 6 months of age. Currently, there is no standard treatment for AT/RT. Recent studies have reported potential anti-tumoral properties of ribavirin, a guanosine analog and anti-viral molecule approved by the Food and Drug Administration for treatment of hepatitis C. We previously demonstrated that ribavirin inhibited glioma cell growth in vitro and in vivo. Based on these results and the fact that no pre-clinical model of ribavirin in AT/RT exists, we decided to investigate the effect of ribavirin on several human AT/RT cell lines (BT12, BT16, and BT37) both in vitro and in vivo. We provide evidence that ribavirin has a significant impact on AT/RT cell growth and increases cell cycle arrest and cell death, potentially through modulation of the eIF4E and/or EZH2 pathways. Interestingly, using scratch wound and transwell Boyden chamber assays, we observed that ribavirin also impairs AT/RT cell migration, invasion, and adhesion. Finally, we demonstrate that ribavirin significantly improves the survival of mice orthotopically implanted with BT12 cells. Our work establishes that ribavirin is effective against AT/RT by decreasing tumoral cell growth and dissemination and could represent a new therapeutic option for children with this deadly disease.
Collapse
Affiliation(s)
- Joshua Casaos
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Sakibul Huq
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Tarik Lott
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Raphael Felder
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - John Choi
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Noah Gorelick
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Michael Peters
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Yuanxuan Xia
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Russell Maxwell
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Tianna Zhao
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Chenchen Ji
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Thomas Simon
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Julie Sesen
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA.,INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, CRCT, 31100 Toulouse, France
| | - Sarah J Scotland
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | - Jeffrey Rubens
- Pathology Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Eric Raabe
- Pathology Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Charles G Eberhart
- Pathology Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Eric M Jackson
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Henry Brem
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Betty Tyler
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Nicolas Skuli
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA.,INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, CRCT, 31100 Toulouse, France
| |
Collapse
|
36
|
Ochiai Y, Sano E, Okamoto Y, Yoshimura S, Makita K, Yamamuro S, Ohta T, Ogino A, Tadakuma H, Ueda T, Nakayama T, Hara H, Yoshino A, Katayama Y. Efficacy of ribavirin against malignant glioma cell lines: Follow-up study. Oncol Rep 2017; 39:537-544. [PMID: 29251333 PMCID: PMC5783620 DOI: 10.3892/or.2017.6149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/07/2017] [Indexed: 01/25/2023] Open
Abstract
Ribavirin, a nucleic acid analog, has been employed as an antiviral agent against RNA and DNA viruses and has become the standard agent used for chronic hepatitis C in combination with interferon-α2a. Furthermore, the potential antitumor efficacy of ribavirin has attracted increasing interest. Recently, we demonstrated a dose-dependent antitumor effect of ribavirin for seven types of malignant glioma cell lines. However, the mechanism underlying the antitumor effect of ribavirin has not yet been fully elucidated. Therefore, the main aim of the present study was to provide further relevant data using two types of malignant glioma cell lines (U-87MG and U-138MG) with different expression of MGMT. Dotted accumulations of γH2AX were found in the nuclei and increased levels of ATM and phosphorylated ATM protein expression were also observed following ribavirin treatment (10 µM of ribavirin, clinical relevant concentration) in both the malignant glioma cells, indicating double-strand breaks as one possible mechanism underlying the antitumor effect of ribavirin. In addition, based on assessements using FACS, ribavirin treatment tended to increase the G0/G1 phase, with a time-lapse, indicating the induction of G0/G1-phase arrest. Furthermore, an increased phosphorylated p53 and p21 protein expression was confirmed in both glioma cells. Additionally, analysis by FACS indicated that apoptosis was induced following ribavirin treatment and caspase cascade, downstream of the p53 pathway, which indicated the activation of both exogenous and endogenous apoptosis in both malignant glioma cell lines. These findings may provide an experimental basis for the clinical treatment of glioblastomas with ribavirin.
Collapse
Affiliation(s)
- Yushi Ochiai
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Emiko Sano
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Yutaka Okamoto
- Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Sodai Yoshimura
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kotaro Makita
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shun Yamamuro
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takashi Ohta
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Akiyoshi Ogino
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hisashi Tadakuma
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Tomohiro Nakayama
- Division of Companion Diagnostics, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroyuki Hara
- Department of Functional Morphology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Atsuo Yoshino
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoichi Katayama
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
37
|
Targeting eIF4E inhibits growth, survival and angiogenesis in retinoblastoma and enhances efficacy of chemotherapy. Biomed Pharmacother 2017; 96:750-756. [DOI: 10.1016/j.biopha.2017.10.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/09/2017] [Accepted: 10/09/2017] [Indexed: 01/09/2023] Open
|
38
|
Tan J, Ye J, Song M, Zhou M, Hu Y. Ribavirin augments doxorubicin's efficacy in human hepatocellular carcinoma through inhibiting doxorubicin-induced eIF4E activation. J Biochem Mol Toxicol 2017; 32. [PMID: 29112301 DOI: 10.1002/jbt.22007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/02/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023]
Abstract
Activation of eukaryotic translation initiation factor 4E (eIF4E) is a cellular survival mechanism in response to chemotherapy in cancers. In this work, we demonstrate that targeting eIF4E by ribavirin sensitizes hepatocellular carcinoma (HCC) cell response to doxorubicin. Ribavirin inhibits growth and survival of HCC cells, and to a greater extent than in normal liver cells. Its combination with doxorubicin achieves greater efficacy than single drug in vitro and in vivo. Ribavirin suppresses phosphorylation of molecules involved in Akt/mTOR/eIF4E pathway. Overexpression of the phosphomimetic form (S209D) but not the nonphosphorylatable form (S209A) eIF4E significantly reverses the inhibitory effects of ribavirin. Interestingly, doxorubicin significantly increases p-eIF4E(S209) level in a dose- and time-dependent manner, suggesting that doxorubicin induces eIF4E activation in HCC cells. In addition, eIF4E activation induced by doxorubicin in HCC cells is inhibited by ribavirin. Our work demonstrates the greater efficacy of ribavirin and doxorubicin combination and its underlying mechanisms.
Collapse
Affiliation(s)
- Jun Tan
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo 315010, People's Republic of China
| | - Jingfen Ye
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo 315010, People's Republic of China
| | - Meijun Song
- Department of Respiratory Medicine, Ningbo Medical Treatment Center Li Huili Hospital, Ningbo 315041, People's Republic of China
| | - Mi Zhou
- School of Medicine, Ningbo University, Ningbo 315211, People's Republic of China
| | - Yaoren Hu
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo 315010, People's Republic of China
| |
Collapse
|
39
|
Bell JB, Eckerdt F, Dhruv HD, Finlay D, Peng S, Kim S, Kroczynska B, Beauchamp EM, Alley K, Clymer J, Goldman S, Cheng SY, James CD, Nakano I, Horbinski C, Mazar AP, Vuori K, Kumthekar P, Raizer J, Berens ME, Platanias LC. Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation. Mol Cancer Res 2017; 16:32-46. [PMID: 29042487 DOI: 10.1158/1541-7786.mcr-17-0397] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/13/2017] [Accepted: 10/11/2017] [Indexed: 12/16/2022]
Abstract
Mesenchymal (MES) and proneural (PN) are two distinct glioma stem cell (GSC) populations that drive therapeutic resistance in glioblastoma (GBM). We screened a panel of 650 small molecules against patient-derived GBM cells to discover compounds targeting specific GBM subtypes. Arsenic trioxide (ATO), an FDA-approved drug that crosses the blood-brain barrier, was identified as a potent PN-specific compound in the initial screen and follow-up validation studies. Furthermore, MES and PN GSCs exhibited differential sensitivity to ATO. As ATO has been shown to activate the MAPK-interacting kinase 1 (MNK1)-eukaryotic translation initiation factor 4E (eIF4E) pathway and subsequent mRNA translation in a negative regulatory feedback manner, the mechanistic role of ATO resistance in MES GBM was explored. In GBM cells, ATO-activated translation initiation cellular events via the MNK1-eIF4E signaling axis. Furthermore, resistance to ATO in intracranial PDX tumors correlated with high eIF4E phosphorylation. Polysomal fractionation and microarray analysis of GBM cells were performed to identify ATO's effect on mRNA translation and enrichment of anti-apoptotic mRNAs in the ATO-induced translatome was found. Additionally, it was determined that MNK inhibition sensitized MES GSCs to ATO in neurosphere and apoptosis assays. Finally, examination of the effect of ATO on patients from a phase I/II clinical trial of ATO revealed that PN GBM patients responded better to ATO than other subtypes as demonstrated by longer overall and progression-free survival.Implications: These findings raise the possibility of a unique therapeutic approach for GBM, involving MNK1 targeting to sensitize MES GSCs to drugs like arsenic trioxide. Mol Cancer Res; 16(1); 32-46. ©2017 AACR.
Collapse
Affiliation(s)
- Jonathan B Bell
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Frank Eckerdt
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Harshil D Dhruv
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Darren Finlay
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Sen Peng
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Seungchan Kim
- Integrated Cancer Genomics Division, The Translational Genomics Research Institute, Phoenix, Arizona.,Department of Electrical and Computer Engineering, Roy G. Perry College of Engineering, Prairie View A&M University, Prairie View, Texas
| | - Barbara Kroczynska
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Elspeth M Beauchamp
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Medicine, Jesse Brown VA Medical Center, Chicago, Illinois
| | - Kristen Alley
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jessica Clymer
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Division of Hematology/Oncology/Stem Cell Transplantation, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Stewart Goldman
- Division of Hematology/Oncology/Stem Cell Transplantation, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Shi-Yuan Cheng
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - C David James
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ichiro Nakano
- Department of Neurosurgery and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew P Mazar
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Developmental Therapeutics Core, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois
| | - Kristiina Vuori
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Priya Kumthekar
- Division of Neuro-Oncology, Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jeffrey Raizer
- Division of Neuro-Oncology, Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Michael E Berens
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. .,Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Medicine, Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
40
|
Kast RE, Skuli N, Cos S, Karpel-Massler G, Shiozawa Y, Goshen R, Halatsch ME. The ABC7 regimen: a new approach to metastatic breast cancer using seven common drugs to inhibit epithelial-to-mesenchymal transition and augment capecitabine efficacy. BREAST CANCER-TARGETS AND THERAPY 2017; 9:495-514. [PMID: 28744157 PMCID: PMC5513700 DOI: 10.2147/bctt.s139963] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Breast cancer metastatic to bone has a poor prognosis despite recent advances in our understanding of the biology of both bone and breast cancer. This article presents a new approach, the ABC7 regimen (Adjuvant for Breast Cancer treatment using seven repurposed drugs), to metastatic breast cancer. ABC7 aims to defeat aspects of epithelial-to-mesenchymal transition (EMT) that lead to dissemination of breast cancer to bone. As add-on to current standard treatment with capecitabine, ABC7 uses ancillary attributes of seven already-marketed noncancer treatment drugs to stop both the natural EMT process inherent to breast cancer and the added EMT occurring as a response to current treatment modalities. Chemotherapy, radiation, and surgery provoke EMT in cancer generally and in breast cancer specifically. ABC7 uses standard doses of capecitabine as used in treating breast cancer today. In addition, ABC7 uses 1) an older psychiatric drug, quetiapine, to block RANK signaling; 2) pirfenidone, an anti-fibrosis drug to block TGF-beta signaling; 3) rifabutin, an antibiotic to block beta-catenin signaling; 4) metformin, a first-line antidiabetic drug to stimulate AMPK and inhibit mammalian target of rapamycin, (mTOR); 5) propranolol, a beta-blocker to block beta-adrenergic signaling; 6) agomelatine, a melatonergic antidepressant to stimulate M1 and M2 melatonergic receptors; and 7) ribavirin, an antiviral drug to prevent eIF4E phosphorylation. All these block the signaling pathways - RANK, TGF-beta, mTOR, beta-adrenergic receptors, and phosphorylated eIF4E - that have been shown to trigger EMT and enhance breast cancer growth and so are worthwhile targets to inhibit. Agonism at MT1 and MT2 melatonergic receptors has been shown to inhibit both breast cancer EMT and growth. This ensemble was designed to be safe and augment capecitabine efficacy. Given the expected outcome of metastatic breast cancer as it stands today, ABC7 warrants a cautious trial.
Collapse
Affiliation(s)
| | - Nicolas Skuli
- INSERM, Centre de Recherches en Cancérologie de Toulouse - CRCT, UMR1037 Inserm/Université Toulouse III - Paul Sabatier, Toulouse, France
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), Santander, Spain
| | | | - Yusuke Shiozawa
- Department of Cancer Biology, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ran Goshen
- Eliaso Consulting Ltd., Tel Aviv-Yafo, Israel
| | | |
Collapse
|
41
|
Kast RE, Hill QA, Wion D, Mellstedt H, Focosi D, Karpel-Massler G, Heiland T, Halatsch ME. Glioblastoma-synthesized G-CSF and GM-CSF contribute to growth and immunosuppression: Potential therapeutic benefit from dapsone, fenofibrate, and ribavirin. Tumour Biol 2017; 39:1010428317699797. [PMID: 28459367 DOI: 10.1177/1010428317699797] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Increased ratio of circulating neutrophils to lymphocytes is a common finding in glioblastoma and other cancers. Data reviewed establish that any damage to brain tissue tends to cause an increase in G-CSF and/or GM-CSF (G(M)-CSF) synthesized by the brain. Glioblastoma cells themselves also synthesize G(M)-CSF. G(M)-CSF synthesized by brain due to damage by a growing tumor and by the tumor itself stimulates bone marrow to shift hematopoiesis toward granulocytic lineages away from lymphocytic lineages. This shift is immunosuppressive and generates the relative lymphopenia characteristic of glioblastoma. Any trauma to brain-be it blunt, sharp, ischemic, infectious, cytotoxic, tumor encroachment, or radiation-increases brain synthesis of G(M)-CSF. G(M)-CSF are growth and motility enhancing factors for glioblastomas. High levels of G(M)-CSF contribute to the characteristic neutrophilia and lymphopenia of glioblastoma. Hematopoietic bone marrow becomes entrained with, directed by, and contributes to glioblastoma pathology. The antibiotic dapsone, the lipid-lowering agent fenofibrate, and the antiviral drug ribavirin are Food and Drug Administration- and European Medicines Agency-approved medicines that have potential to lower synthesis or effects of G(M)-CSF and thus deprive a glioblastoma of some of the growth promoting contributions of bone marrow and G(M)-CSF.
Collapse
Affiliation(s)
| | - Quentin A Hill
- 2 Department of Haematology, St James's University Hospital, Leeds Teaching Hospitals, Leeds, UK
| | - Didier Wion
- 3 INSERM U1205, Centre de Recherche Biomédicale Edmond J. Safra, Grenoble, France
| | - Håkan Mellstedt
- 4 Department of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Daniele Focosi
- 5 North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | | | - Tim Heiland
- 6 Department of Neurosurgery, University of Ulm, Ulm, Germany
| | | |
Collapse
|