1
|
Rahman MM, Estifanos B, Glenn HL, Gutierrez-Jensen AD, Kibler K, Li Y, Jacobs B, McFadden G, Hogue BG. Effect of Exportin 1/XPO1 Nuclear Export Pathway Inhibition on Coronavirus Replication. Viruses 2025; 17:284. [PMID: 40007039 PMCID: PMC11860411 DOI: 10.3390/v17020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
The nucleocytoplasmic transport of proteins using XPO1 (exportin 1) plays a vital role in cell proliferation and survival. Many viruses also exploit this pathway to promote infection and replication. Thus, inhibiting the XPO1-mediated nuclear export pathway with selective inhibitors has a diverse effect on virus replication by regulating antiviral, proviral, and anti-inflammatory pathways. The XPO1 inhibitor Selinexor is an FDA-approved anticancer drug predicted to have antiviral or proviral functions against viruses. Here, we observed that the pretreatment of cultured cell lines from human or mouse origin with the nuclear export inhibitor Selinexor significantly enhanced the protein expression and replication of mouse hepatitis virus (MHV), a mouse coronavirus. The knockdown of cellular XPO1 protein expression also significantly enhanced the replication of MHV in human cells. However, for SARS-CoV-2, Selinexor treatment had diverse effects on virus replication in different cell lines. These results indicate that XPO1-mediated nuclear export pathway inhibition might affect coronavirus replication depending on cell types and virus origin.
Collapse
Affiliation(s)
- Masmudur M. Rahman
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA;
- School of Life Sciences Microbiology Graduate Program, Arizona State University, Tempe, AZ 85287, USA;
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (H.L.G.); (Y.L.); (B.J.); (G.M.)
| | - Bereket Estifanos
- School of Life Sciences Microbiology Graduate Program, Arizona State University, Tempe, AZ 85287, USA;
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Honor L. Glenn
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (H.L.G.); (Y.L.); (B.J.); (G.M.)
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Ami D. Gutierrez-Jensen
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA;
| | - Karen Kibler
- Center for ASU-Banner Neurodegenerative Disease Research, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA;
| | - Yize Li
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (H.L.G.); (Y.L.); (B.J.); (G.M.)
- Center for ASU-Banner Neurodegenerative Disease Research, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA;
| | - Bertram Jacobs
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (H.L.G.); (Y.L.); (B.J.); (G.M.)
- Center for ASU-Banner Neurodegenerative Disease Research, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA;
| | - Grant McFadden
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (H.L.G.); (Y.L.); (B.J.); (G.M.)
| | - Brenda G. Hogue
- School of Life Sciences Microbiology Graduate Program, Arizona State University, Tempe, AZ 85287, USA;
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (H.L.G.); (Y.L.); (B.J.); (G.M.)
- Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
2
|
Sokolova V, Gruber R, Pammer LM, Kocher F, Klieser E, Amann A, Pichler R, Günther M, Ormanns S, Neureiter D, Seeber A. Prognostic and functional role of the nuclear export receptor 1 (XPO1) in gastrointestinal cancers: a potential novel target? Mol Biol Rep 2024; 52:87. [PMID: 39729162 PMCID: PMC11680630 DOI: 10.1007/s11033-024-10169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
In the last decades the survival of metastatic gastrointestinal (GI) cancer patients could have been significantly extended due to the introduction of targeted- and immunotherapy. However, only the minority of patients will experience long-lasting survival. Hence, novel therapeutics are clearly necessary for GI cancer patients. Molecular high-throughput profiling techniques have revealed potential novel targetable molecular alterations, emphasizing the necessity for tailored therapeutic approaches. Nuclear export proteins, particularly Exportin-1 (XPO1), have emerged as promising targets in cancer therapy due to their crucial role in cellular homeostasis and regulation of key cellular functions. Dysregulation of XPO1-mediated nuclear export leads to the functional loss of tumor suppressors and pro-apoptotic factors, facilitating cancer progression. Selinexor, a XPO1 inhibitor, has shown promising activity in preclinical and clinical studies, particularly in hematological malignancies. However, its efficacy in GI cancers remains underexplored. This review aims to elucidate the functional and pathophysiological role of XPO1 in GI cancers. Despite the potential of XPO1 inhibitors in suppressing cell proliferation and inducing apoptosis, comprehensive molecular landscape data and validation of selective inhibitors in GI cancers are lacking. Targeting XPO1 presents a significant therapeutic potential for the treatment of GI cancer patients. Further research is necessary to fully elucidate the molecular landscape according to XPO1 expression in GI tumors and to validate the efficacy of selective XPO1 inhibitors. These efforts are expected to contribute to the development of more effective and personalized therapeutic strategies for GI cancer patients.
Collapse
Affiliation(s)
- Viktorija Sokolova
- Department of Nuclear Medicine, Provincial Hospital of Bolzano (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University, Bolzano-Bozen, Italy
| | - Rebecca Gruber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Lorenz M Pammer
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kocher
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Eckhard Klieser
- Institute of Pathology, University Clinics Salzburg, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Arno Amann
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Günther
- Institute of Pathology, INNPATH GmbH, Tirol Kliniken Innsbruck, Innsbruck, Austria
| | - Steffen Ormanns
- Institute of Pathology, INNPATH GmbH, Tirol Kliniken Innsbruck, Innsbruck, Austria
| | - Daniel Neureiter
- Institute of Pathology, University Clinics Salzburg, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Andreas Seeber
- Department of Nuclear Medicine, Provincial Hospital of Bolzano (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University, Bolzano-Bozen, Italy.
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
3
|
Li S, Lin Y, Gao X, Zeng D, Cen W, Su Y, Su J, Zeng C, Huang Z, Zeng H, Huang S, Tang M, Li X, Luo M, Huang Z, Liang R, Ye J. Integrative multi-omics analysis reveals a novel subtype of hepatocellular carcinoma with biological and clinical relevance. Front Immunol 2024; 15:1517312. [PMID: 39712016 PMCID: PMC11659151 DOI: 10.3389/fimmu.2024.1517312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a highly heterogeneous tumor, and the development of accurate predictive models for prognosis and drug sensitivity remains challenging. Methods We integrated laboratory data and public cohorts to conduct a multi-omics analysis of HCC, which included bulk RNA sequencing, proteomic analysis, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics sequencing (ST-seq), and genome sequencing. We constructed a tumor purity (TP) and tumor microenvironment (TME) prognostic risk model. Proteomic analysis validated the TP-TME-related signatures. Joint analysis of scRNA-seq and ST-seq revealed characteristic clusters associated with TP high-risk subtypes, and immunohistochemistry confirmed the expression of key genes. We conducted functional enrichment analysis, transcription factor activity inference, cell-cell interaction, drug efficacy analysis, and mutation information analysis to identify a novel subtype of HCC. Results Our analyses constructed a robust HCC prognostic risk prediction model. The patients with TP-TME high-risk subtypes predominantly exhibit hypoxia and activation of the Wnt/beta-catenin, Notch, and TGF-beta signaling pathways. Furthermore, we identified a novel subtype, XPO1+Epithelial. This subtype expresses signatures of the TP risk subtype and aligns with the biological behavior of high-risk patients. Additional analyses revealed that XPO1+Epithelial is influenced primarily by fibroblasts via ligand-receptor interactions, such as FN1-(ITGAV+ITGB1), and constitute a significant component of the TP-TME subtype. Moreover, XPO1+Epithelial interact with monocytes/macrophages, T/NK cells, and endothelial cells through ligand-receptor pairs, including MIF-(CD74+CXCR4), MIF-(CD74+CD44), and VEGFA-VEGFR1R2, respectively, thereby promoting the recruitment of immune-suppressive cells and angiogenesis. The ST-seq cohort treated with Tyrosine Kinase Inhibitors (TKIs) and Programmed Cell Death Protein 1 (PD-1) presented elevated levels of TP and TME risk subtype signature genes, as well as XPO1+Epithelial, T-cell, and endothelial cell infiltration in the treatment response group. Drug sensitivity analyses indicated that TP-TME high-risk subtypes, including sorafenib and pembrolizumab, were associated with sensitivity to multiple drugs. Further exploratory analyses revealed that CTLA4, PDCD1, and the cancer antigens MSLN, MUC1, EPCAM, and PROM1 presented significantly increase expression levels in the high-risk subtype group. Conclusions This study constructed a robust prognostic model for HCC and identified novel subgroups at the single-cell level, potentially assisting in the assessment of prognostic risk for HCC patients and facilitating personalized drug therapy.
Collapse
Affiliation(s)
- Shizhou Li
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Xing Gao
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Dandan Zeng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Weijie Cen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Yuejiao Su
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Jingting Su
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Can Zeng
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Zhenbo Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Haoyu Zeng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Shilin Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Minchao Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Xiaoqing Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Min Luo
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Zhihu Huang
- Department of Clinical Laboratory, Minzu Hospital Guangxi Zhuang Autonomous Region, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, Guangxi, ;China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| |
Collapse
|
4
|
Blunt MD, Fisher H, Schittenhelm RB, Mbiribindi B, Fulton R, Khan S, Espana-Serrano L, Graham LV, Bastidas-Legarda L, Burns D, Khakoo SM, Mansour S, Essex JW, Ayala R, Das J, Purcell AW, Khakoo SI. The nuclear export protein XPO1 provides a peptide ligand for natural killer cells. SCIENCE ADVANCES 2024; 10:eado6566. [PMID: 39178254 PMCID: PMC11343027 DOI: 10.1126/sciadv.ado6566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024]
Abstract
XPO1 (Exportin-1/CRM1) is a nuclear export protein that is frequently overexpressed in cancer and functions as a driver of oncogenesis. Currently small molecules that target XPO1 are being used in the clinic as anticancer agents. We identify XPO1 as a target for natural killer (NK) cells. Using immunopeptidomics, we have identified a peptide derived from XPO1 that can be recognized by the activating NK cell receptor KIR2DS2 in the context of human leukocyte antigen-C. The peptide can be endogenously processed and presented to activate NK cells specifically through this receptor. Although high XPO1 expression in cancer is commonly associated with a poor prognosis, we show that the outcome of specific cancers, such as hepatocellular carcinoma, can be substantially improved if there is concomitant evidence of NK cell infiltration. We thus identify XPO1 as a bona fide tumor antigen recognized by NK cells that offers an opportunity for a personalized approach to NK cell therapy for solid tumors.
Collapse
Affiliation(s)
- Matthew D. Blunt
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hayden Fisher
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Ralf B. Schittenhelm
- Monash Proteomics & Metabolomics Platform, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Berenice Mbiribindi
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Rebecca Fulton
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sajida Khan
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Laura Espana-Serrano
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Lara V. Graham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Leidy Bastidas-Legarda
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Monash Proteomics & Metabolomics Platform, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, and The Department of Pediatrics, Pelotonia Institute for Immuno-Oncology, Ohio State University, Columbus, OH, USA
| | - Daniel Burns
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sophie M.S. Khakoo
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Salah Mansour
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jonathan W. Essex
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Rochelle Ayala
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, and The Department of Pediatrics, Pelotonia Institute for Immuno-Oncology, Ohio State University, Columbus, OH, USA
| | - Anthony W. Purcell
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Salim I. Khakoo
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
5
|
Schütt J, Brinkert K, Plis A, Schenk T, Brioli A. Unraveling the complexity of drug resistance mechanisms to SINE, T cell-engaging therapies and CELMoDs in multiple myeloma: a comprehensive review. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:26. [PMID: 39050883 PMCID: PMC11267153 DOI: 10.20517/cdr.2024.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
Despite significant advances in the understanding of multiple myeloma (MM) biology and the development of novel treatment strategies in the last two decades, MM is still an incurable disease. Novel drugs with alternative mechanisms of action, such as selective inhibitors of nuclear export (SINE), modulators of the ubiquitin pathway [cereblon E3 ligase modulatory drugs (CELMoDs)], and T cell redirecting (TCR) therapy, have led to significant improvement in patient outcomes. However, resistance still emerges, posing a major problem for the treatment of myeloma patients. This review summarizes current data on treatment with SINE, TCR therapy, and CELMoDs and explores their mechanism of resistance. Understanding these resistance mechanisms is critical for developing strategies to overcome treatment failure and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Jacqueline Schütt
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Kerstin Brinkert
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Andrzej Plis
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| | - Tino Schenk
- Clinic of Internal Medicine 2, Department of Hematology and Medical Oncology, Jena University Hospital, Jena 07741, Germany
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena 07741, Germany
| | - Annamaria Brioli
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| |
Collapse
|
6
|
Andrés CMC, Pérez de la Lastra JM, Bustamante Munguira E, Andrés Juan C, Pérez-Lebeña E. Michael Acceptors as Anti-Cancer Compounds: Coincidence or Causality? Int J Mol Sci 2024; 25:6099. [PMID: 38892287 PMCID: PMC11172677 DOI: 10.3390/ijms25116099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Michael acceptors represent a class of compounds with potential anti-cancer properties. They act by binding to nucleophilic sites in biological molecules, thereby disrupting cancer cell function and inducing cell death. This mode of action, as well as their ability to be modified and targeted, makes them a promising avenue for advancing cancer therapy. We are investigating the molecular mechanisms underlying Michael acceptors and their interactions with cancer cells, in particular their ability to interfere with cellular processes and induce apoptosis. The anti-cancer properties of Michael acceptors are not accidental but are due to their chemical structure and reactivity. The electrophilic nature of these compounds allows them to selectively target nucleophilic residues on disease-associated proteins, resulting in significant therapeutic benefits and minimal toxicity in various diseases. This opens up new perspectives for the development of more effective and precise cancer drugs. Nevertheless, further studies are essential to fully understand the impact of our discoveries and translate them into clinical practice.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | | | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | | |
Collapse
|
7
|
Lai C, Xu L, Dai S. The nuclear export protein exportin-1 in solid malignant tumours: From biology to clinical trials. Clin Transl Med 2024; 14:e1684. [PMID: 38783482 PMCID: PMC11116501 DOI: 10.1002/ctm2.1684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Exportin-1 (XPO1), a crucial protein regulating nuclear-cytoplasmic transport, is frequently overexpressed in various cancers, driving tumor progression and drug resistance. This makes XPO1 an attractive therapeutic target. Over the past few decades, the number of available nuclear export-selective inhibitors has been increasing. Only KPT-330 (selinexor) has been successfully used for treating haematological malignancies, and KPT-8602 (eltanexor) has been used for treating haematologic tumours in clinical trials. However, the use of nuclear export-selective inhibitors for the inhibition of XPO1 expression has yet to be thoroughly investigated in clinical studies and therapeutic outcomes for solid tumours. METHODS We collected numerous literatures to explain the efficacy of XPO1 Inhibitors in preclinical and clinical studies of a wide range of solid tumours. RESULTS In this review, we focus on the nuclear export function of XPO1 and results from clinical trials of its inhibitors in solid malignant tumours. We summarized the mechanism of action and therapeutic potential of XPO1 inhibitors, as well as adverse effects and response biomarkers. CONCLUSION XPO1 inhibition has emerged as a promising therapeutic strategy in the fight against cancer, offering a novel approach to targeting tumorigenic processes and overcoming drug resistance. SINE compounds have demonstrated efficacy in a wide range of solid tumours, and ongoing research is focused on optimizing their use, identifying response biomarkers, and developing effective combination therapies. KEY POINTS Exportin-1 (XPO1) plays a critical role in mediating nucleocytoplasmic transport and cell cycle. XPO1 dysfunction promotes tumourigenesis and drug resistance within solid tumours. The therapeutic potential and ongoing researches on XPO1 inhibitors in the treatment of solid tumours. Additional researches are essential to address safety concerns and identify biomarkers for predicting patient response to XPO1 inhibitors.
Collapse
Affiliation(s)
- Chuanxi Lai
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| | - Lingna Xu
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| | - Sheng Dai
- Department of Colorectal SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Biotherapy of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
8
|
Newell S, van der Watt PJ, Leaner VD. Therapeutic targeting of nuclear export and import receptors in cancer and their potential in combination chemotherapy. IUBMB Life 2024; 76:4-25. [PMID: 37623925 PMCID: PMC10952567 DOI: 10.1002/iub.2773] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/03/2023] [Indexed: 08/26/2023]
Abstract
Systemic modalities are crucial in the management of disseminated malignancies and liquid tumours. However, patient responses and tolerability to treatment are generally poor and those that enter remission often return with refractory disease. Combination therapies provide a methodology to overcome chemoresistance mechanisms and address dose-limiting toxicities. A deeper understanding of tumorigenic processes at the molecular level has brought a targeted therapy approach to the forefront of cancer research, and novel cancer biomarkers are being identified at a rapid rate, with some showing potential therapeutic benefits. The Karyopherin superfamily of proteins is soluble receptors that mediate nucleocytoplasmic shuttling of proteins and RNAs, and recently, nuclear transport receptors have been recognized as novel anticancer targets. Inhibitors against nuclear export have been approved for clinical use against certain cancer types, whereas inhibitors against nuclear import are in preclinical stages of investigation. Mechanistically, targeting nucleocytoplasmic shuttling has shown to abrogate oncogenic signalling and restore tumour suppressor functions through nuclear sequestration of relevant proteins and mRNAs. Hence, nuclear transport inhibitors display broad spectrum anticancer activity and harbour potential to engage in synergistic interactions with a wide array of cytotoxic agents and other targeted agents. This review is focussed on the most researched nuclear transport receptors in the context of cancer, XPO1 and KPNB1, and highlights how inhibitors targeting these receptors can enhance the therapeutic efficacy of standard of care therapies and novel targeted agents in a combination therapy approach. Furthermore, an updated review on the therapeutic targeting of lesser characterized karyopherin proteins is provided and resistance to clinically approved nuclear export inhibitors is discussed.
Collapse
Affiliation(s)
- Stella Newell
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health SciencesUniversity of Cape TownCape TownSouth Africa
| | - Pauline J. van der Watt
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health SciencesUniversity of Cape TownCape TownSouth Africa
- Institute of Infectious Diseases and Molecular Medicine, University of Cape TownCape TownSouth Africa
| | - Virna D. Leaner
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health SciencesUniversity of Cape TownCape TownSouth Africa
- UCT/SAMRC Gynaecological Cancer Research CentreUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
9
|
ÖZDAŞ T, ÖZDAŞ S, CANATAR İ, ÇOŞKUN E, ŞENYURT EB, GÖRGÜLÜ O. CRM1 expression: association with high prognostic value in laryngeal cancer. Turk J Med Sci 2023; 53:909-923. [PMID: 38031942 PMCID: PMC10760544 DOI: 10.55730/1300-0144.5655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/18/2023] [Accepted: 02/03/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Laryngeal cancer is a very common malignant tumor of the head and neck. While laryngeal cancer does not show any obvious early symptoms, it tends to have a poor prognosis in advanced clinical stages. Chromosome region maintenance 1 (CRM1) mediates the nuclear export of some RNAs, major and tumor suppressor proteins and has been associated with the pathogenesis of many tumors. However, the clinicopathological significance of CRM1 gene expression in laryngeal cancer has not been clarified yet. Therefore, this study aims to detect the expression of CRM1 in laryngeal cancer and to investigate its relationship with clinicopathological parameters and prognosis. METHODS CRM1 expression in matched tumor and normal tissues obtained from 43 laryngeal cancer patients were evaluated intracellular for protein and mRNA levels by immunohistochemical staining (IHC), western-blot, and quantitative real-time RT-PCR (qRT-PCR), respectively. RESULTS IHC, western-blot, and qRT-PCR analyses showed that CRM1 expression was significantly increased in laryngeal cancer tissue compared to normal tissue. Increased expression of CRM1 has been associated with poor prognostic clinicopathological features, including advanced tumor stage, increased tumor invasion, larger tumor size, positive lymph node metastasis, distant metastasis, and invasive histological type by IHC, western-blot, and qRT-PCR. Kaplan-Meier survival analysis showed that patients with high expression of CRM1 exhibited lower overall survival compared to those with low expression (Log-rank = 7.16, p = 0.007). According to the The Cancer Genome Atlas (TCGA) datasets, elevated CRM1 expression in head and neck cancer including cases of squamous cell laryngeal origin is associated with advanced tumor stage and histological grade (p > 0.05, for all). DISCUSSION Consequently, CRM1 plays an important role in laryngeal cancer and may serve as an indicator and prognostic factor for poor overall survival in laryngeal cancer patients.
Collapse
Affiliation(s)
- Talih ÖZDAŞ
- Department of Otorhinolaryngology, Adana City Training and Research Hospital, Health Science University, Adana,
Turkiye
| | - Sibel ÖZDAŞ
- Department of Bioengineering, Faculty of Engineering Sciences, Adana Alparslan Türkeş Science and Technology University, Adana,
Turkiye
| | - İpek CANATAR
- Department of Bioengineering, Faculty of Engineering Sciences, Adana Alparslan Türkeş Science and Technology University, Adana,
Turkiye
| | - Erdal ÇOŞKUN
- Genomics Team, Microsoft Research, Redmond, WA,
USA
| | - Elif Burcu ŞENYURT
- Department of Otorhinolaryngology, Adana City Training and Research Hospital, Health Science University, Adana,
Turkiye
| | - Orhan GÖRGÜLÜ
- Department of Otorhinolaryngology, Adana City Training and Research Hospital, Health Science University, Adana,
Turkiye
| |
Collapse
|
10
|
Zhang L, Hong J, Chen W, Zhang W, Liu X, Lu J, Tang H, Yang Z, Zhou K, Xie H, Jia C, Jiang D, Zheng S. DBF4 Dependent Kinase Inhibition Suppresses Hepatocellular Carcinoma Progression and Potentiates Anti-Programmed Cell Death-1 Therapy. Int J Biol Sci 2023; 19:3412-3427. [PMID: 37497004 PMCID: PMC10367558 DOI: 10.7150/ijbs.80351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/13/2023] [Indexed: 07/28/2023] Open
Abstract
The progression of hepatocellular carcinoma (HCC) remains a huge clinical challenge, and elucidation of the underlying molecular mechanisms is critical to develop effective therapeutic strategy. Dumbbell former 4 (DBF4) complexes with cell division cycle 7 (CDC7) to form DBF4-dependent kinase (DDK), playing instrumental roles in tumor cell survival, whereas its roles in HCC remain elusive. This study revealed that DBF4 expression was upregulated in HCC and constituted an independent prognostic factor of patient survival. We identified p65 as an upstream inducer which increased DBF4 expression by directly binding to its promoter. DBF4 accelerated HCC cell proliferation and tumorigenesis in vitro and in vivo. Mechanistically, DBF4 complexed with CDC7 to bind to the coiled coil domain of STAT3 and activate STAT3 signaling through XPO1-mediated nuclear exportation. Notably, p65 enhanced the nuclear transport of DDK and DDK-STAT3 interaction by transcriptionally upregulating XPO1. DBF4 expression positively correlated with activated STAT3 and XPO1 in HCC tissues. Furthermore, combining DDK inhibitor XL413 with anti-PD-1 immunotherapy dramatically suppressed HCC growth and prolonged the survival of HCC-bearing mouse. Our findings reveal that DDK activates STAT3 pathway and facilitates HCC progression, and demonstrate the proof of the concept of targeting DDK to improve the efficacy of HCC immunotherapy.
Collapse
Affiliation(s)
- Liang Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Jiawei Hong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
| | - Wei Zhang
- Department of Medical Oncology, Sir Runrun Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xi Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Jiahua Lu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Hong Tang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Zhentao Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Ke Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Changku Jia
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
| | - Donghai Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| |
Collapse
|
11
|
Udi Y, Zhang W, Stein ME, Ricardo-Lax I, Pasolli HA, Chait BT, Rout MP. A general method for quantitative fractionation of mammalian cells. J Cell Biol 2023; 222:213941. [PMID: 36920247 PMCID: PMC10040634 DOI: 10.1083/jcb.202209062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/11/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Subcellular fractionation in combination with mass spectrometry-based proteomics is a powerful tool to study localization of key proteins in health and disease. Here we offered a reliable and rapid method for mammalian cell fractionation, tuned for such proteomic analyses. This method proves readily applicable to different cell lines in which all the cellular contents are accounted for, while maintaining nuclear and nuclear envelope integrity. We demonstrated the method's utility by quantifying the effects of a nuclear export inhibitor on nucleoplasmic and cytoplasmic proteomes.
Collapse
Affiliation(s)
- Yael Udi
- Laboratory of Cellular and Structural Biology, The Rockefeller University , New York, NY, USA
| | - Wenzhu Zhang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University , New York, NY, USA
| | - Milana E Stein
- Laboratory of Cellular and Structural Biology, The Rockefeller University , New York, NY, USA
| | - Inna Ricardo-Lax
- Laboratory of Virology and Infectious Disease, The Rockefeller University , New York, NY, USA
| | - Hilda A Pasolli
- Electron Microscopy Resource Center, The Rockefeller University , New York, NY, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University , New York, NY, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University , New York, NY, USA
| |
Collapse
|
12
|
Rahman MM, van Oosterom F, Enow JA, Hossain M, Gutierrez-Jensen AD, Cashen M, Everts A, Lowe K, Kilbourne J, Daggett-Vondras J, Karr TL, McFadden G. Nuclear Export Inhibitor Selinexor Enhances Oncolytic Myxoma Virus Therapy against Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:952-968. [PMID: 37377603 PMCID: PMC10234290 DOI: 10.1158/2767-9764.crc-22-0483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/08/2023] [Accepted: 05/11/2023] [Indexed: 06/29/2023]
Abstract
Oncolytic viruses exploited for cancer therapy have been developed to selectively infect, replicate, and kill cancer cells to inhibit tumor growth. However, in some cancer cells, oncolytic viruses are often limited in completing their full replication cycle, forming progeny virions, and/or spreading in the tumor bed because of the heterogeneous cell types within the tumor bed. Here, we report that the nuclear export pathway regulates oncolytic myxoma virus (MYXV) infection and cytoplasmic viral replication in a subclass of human cancer cell types where viral replication is restricted. Inhibition of the XPO-1 (exportin 1) nuclear export pathway with nuclear export inhibitors can overcome this restriction by trapping restriction factors in the nucleus and allow significantly enhanced viral replication and killing of cancer cells. Furthermore, knockdown of XPO-1 significantly enhanced MYXV replication in restrictive human cancer cells and reduced the formation of antiviral granules associated with RNA helicase DHX9. Both in vitro and in vivo, we demonstrated that the approved XPO1 inhibitor drug selinexor enhances the replication of MYXV and kills diverse human cancer cells. In a xenograft tumor model in NSG mice, combination therapy with selinexor plus MYXV significantly reduced the tumor burden and enhanced the survival of animals. In addition, we performed global-scale proteomic analysis of nuclear and cytosolic proteins in human cancer cells to identify the host and viral proteins that were upregulated or downregulated by different treatments. These results indicate, for the first time, that selinexor in combination with oncolytic MYXV can be used as a potential new therapy. Significance We demonstrated that a combination of nuclear export inhibitor selinexor and oncolytic MYXV significantly enhanced viral replication, reduced cancer cell proliferation, reduced tumor burden, and enhanced the overall survival of animals. Thus, selinexor and oncolytic MYXV can be used as potential new anticancer therapy.
Collapse
Affiliation(s)
- Masmudur M. Rahman
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Fleur van Oosterom
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Junior A. Enow
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Maksuda Hossain
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Ami D. Gutierrez-Jensen
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Mackenzie Cashen
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Anne Everts
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Kenneth Lowe
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Jacquelyn Kilbourne
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Juliane Daggett-Vondras
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Timothy L. Karr
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Grant McFadden
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
13
|
Jing CB, Prutsch N, He S, Zimmerman MW, Landesman Y, Look AT. Synthetic lethal targeting of TET2-mutant haematopoietic stem and progenitor cells by XPO1 inhibitors. Br J Haematol 2023; 201:489-501. [PMID: 36746437 PMCID: PMC10121884 DOI: 10.1111/bjh.18667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 02/08/2023]
Abstract
TET2 inactivating mutations serve as initiating genetic lesions in the transformation of haematopoietic stem and progenitor cells (HSPCs). In this study, we analysed known drugs in zebrafish embryos for their ability to selectively kill tet2-mutant HSPCs in vivo. We found that the exportin 1 (XPO1) inhibitors, selinexor and eltanexor, selectively kill tet2-mutant HSPCs. In serial replating colony assays, these small molecules were selectively active in killing murine Tet2-deficient Lineage-, Sca1+, Kit+ (LSK) cells, and also TET2-inactivated human acute myeloid leukaemia (AML) cells. Selective killing of TET2-mutant HSPCs and human AML cells by these inhibitors was due to increased levels of apoptosis, without evidence of DNA damage based on increased γH2AX expression. The finding that TET2 loss renders HSPCs and AML cells selectively susceptible to cell death induced by XPO1 inhibitors provides preclinical evidence of the selective activity of these drugs, justifying further clinical studies of these small molecules for the treatment of TET2-mutant haematopoietic malignancies, and to suppress clonal expansion in age-related TET2-mutant clonal haematopoiesis.
Collapse
Affiliation(s)
- Chang-Bin Jing
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School
| | - Nicole Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School
| | - Shuning He
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School
| | - Mark W. Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School
| | | | - A. Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Division of Pediatric Hematology/Oncology Boston Children’s Hospital, Boston, Massachusetts
| |
Collapse
|
14
|
Saibu OA, Hammed SO, Oladipo OO, Odunitan TT, Ajayi TM, Adejuyigbe AJ, Apanisile BT, Oyeneyin OE, Oluwafemi AT, Ayoola T, Olaoba OT, Alausa AO, Omoboyowa DA. Protein-protein interaction and interference of carcinogenesis by supramolecular modifications. Bioorg Med Chem 2023; 81:117211. [PMID: 36809721 DOI: 10.1016/j.bmc.2023.117211] [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: 10/25/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023]
Abstract
Protein-protein interactions (PPIs) are essential in normal biological processes, but they can become disrupted or imbalanced in cancer. Various technological advancements have led to an increase in the number of PPI inhibitors, which target hubs in cancer cell's protein networks. However, it remains difficult to develop PPI inhibitors with desired potency and specificity. Supramolecular chemistry has only lately become recognized as a promising method to modify protein activities. In this review, we highlight recent advances in the use of supramolecular modification approaches in cancer therapy. We make special note of efforts to apply supramolecular modifications, such as molecular tweezers, to targeting the nuclear export signal (NES), which can be used to attenuate signaling processes in carcinogenesis. Finally, we discuss the strengths and weaknesses of using supramolecular approaches to targeting PPIs.
Collapse
Affiliation(s)
- Oluwatosin A Saibu
- Department of Environmental Toxicology, Universitat Duisburg-Essen, NorthRhine-Westphalia, Germany
| | - Sodiq O Hammed
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oladapo O Oladipo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Tope T Odunitan
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria; Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Temitope M Ajayi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Aderonke J Adejuyigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Boluwatife T Apanisile
- Department of Nutrition and Dietetics, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oluwatoba E Oyeneyin
- Theoretical and Computational Chemistry Unit, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Adenrele T Oluwafemi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Tolulope Ayoola
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Olamide T Olaoba
- Department of Molecular Pathogenesis and Therapeutics, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Abdullahi O Alausa
- Department of Molecular Biology and Biotechnology, ITMO University, St Petersburg, Russia
| | - Damilola A Omoboyowa
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| |
Collapse
|
15
|
Morgan KJ, Doggett K, Geng F, Mieruszynski S, Whitehead L, Smith KA, Hogan BM, Simons C, Baillie GJ, Molania R, Papenfuss AT, Hall TE, Ober EA, Stainier DYR, Gong Z, Heath JK. ahctf1 and kras mutations combine to amplify oncogenic stress and restrict liver overgrowth in a zebrafish model of hepatocellular carcinoma. eLife 2023; 12:73407. [PMID: 36648336 PMCID: PMC9897728 DOI: 10.7554/elife.73407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
The nucleoporin (NUP) ELYS, encoded by AHCTF1, is a large multifunctional protein with essential roles in nuclear pore assembly and mitosis. Using both larval and adult zebrafish models of hepatocellular carcinoma (HCC), in which the expression of an inducible mutant kras transgene (krasG12V) drives hepatocyte-specific hyperplasia and liver enlargement, we show that reducing ahctf1 gene dosage by 50% markedly decreases liver volume, while non-hyperplastic tissues are unaffected. We demonstrate that in the context of cancer, ahctf1 heterozygosity impairs nuclear pore formation, mitotic spindle assembly, and chromosome segregation, leading to DNA damage and activation of a Tp53-dependent transcriptional programme that induces cell death and cell cycle arrest. Heterozygous expression of both ahctf1 and ranbp2 (encoding a second nucleoporin), or treatment of heterozygous ahctf1 larvae with the nucleocytoplasmic transport inhibitor, Selinexor, completely blocks krasG12V-driven hepatocyte hyperplasia. Gene expression analysis of patient samples in the liver hepatocellular carcinoma (LIHC) dataset in The Cancer Genome Atlas shows that high expression of one or more of the transcripts encoding the 10 components of the NUP107-160 subcomplex, which includes AHCTF1, is positively correlated with worse overall survival. These results provide a strong and feasible rationale for the development of novel cancer therapeutics that target ELYS function and suggest potential avenues for effective combinatorial treatments.
Collapse
Affiliation(s)
- Kimberly J Morgan
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Karen Doggett
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Fansuo Geng
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Stephen Mieruszynski
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| | - Lachlan Whitehead
- Department of Medical Biology, University of MelbourneParkvilleAustralia
- Centre for Dynamic Imaging, Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Kelly A Smith
- Department of Physiology, University of MelbourneParkvilleAustralia
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
| | - Benjamin M Hogan
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
- Peter MacCallum Cancer CentreMelbourneAustralia
| | - Cas Simons
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
- Murdoch Children's Research InstituteParkvilleAustralia
| | - Gregory J Baillie
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
| | - Ramyar Molania
- Department of Medical Biology, University of MelbourneParkvilleAustralia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Anthony T Papenfuss
- Department of Medical Biology, University of MelbourneParkvilleAustralia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
| | - Thomas E Hall
- Institute for Molecular Biosciences, University of QueenslandQueenslandAustralia
| | - Elke A Ober
- Danish Stem Cell Center, University of CopenhagenCopenhagenDenmark
| | - Didier YR Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Zhiyuan Gong
- Department of Biological Science, National University of SingaporeSingaporeSingapore
| | - Joan K Heath
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, University of MelbourneParkvilleAustralia
| |
Collapse
|
16
|
Zhao C, Yang ZY, Zhang J, Li O, Liu SL, Cai C, Shu YJ, Pan LJ, Gong W, Dong P. Inhibition of XPO1 with KPT-330 induces autophagy-dependent apoptosis in gallbladder cancer by activating the p53/mTOR pathway. J Transl Med 2022; 20:434. [PMID: 36180918 PMCID: PMC9524043 DOI: 10.1186/s12967-022-03635-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gallbladder cancer (GBC) is a highly aggressive malignant cancer in the biliary system with poor prognosis. XPO1 (chromosome region maintenance 1 or CRM1) mediates the nuclear export of several proteins, mainly tumor suppressors. Thus, XPO1 functions as a pro-oncogenic factor. KPT-330 (Selinexor) is a United States Food and Drug Administration approved selective inhibitor of XPO1 that demonstrates good therapeutic effects in hematologic cancers. However, the function of XPO1 and the effect of KPT-330 have not been reported in GBC. METHODS We analyzed the correlation between XPO1 expression levels by q-PCR and clinical features of GBC patients. Cell proliferation assays were used to analyze the in vitro antitumor effects of XPO1 inhibitor KPT-330. mRNA sequencing was used to explore the underlying mechanisms. Western blot was performed to explore the relationship between apoptosis and autophagy. The in vivo antitumor effect of KPT-330 was investigated in a nude mouse model of gallbladder cancer. RESULTS We found that high expression of XPO1 was related to poor prognosis of GBC patients. We observed that XPO1 inhibitor KPT-330 inhibited the proliferation of GBC cells in vitro. Furthermore, XPO1 inhibitor KPT-330 induced apoptosis by reducing the mitochondrial membrane potential and triggering autophagy in NOZ and GBC-SD cells. Indeed, XPO1 inhibitor KPT-330 led to nuclear accumulation of p53 and activated the p53/mTOR pathway to regulate autophagy-dependent apoptosis. Importantly, KPT-330 suppressed tumor growth with no obvious toxic effects in vivo. CONCLUSION XPO1 may be a promising prognostic indicator for GBC, and KPT-330 appears to be a potential drug for treating GBC effectively and safely.
Collapse
Affiliation(s)
- Cheng Zhao
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Zi-yi Yang
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Jian Zhang
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Ou Li
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Shi-lei Liu
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Chen Cai
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Yi-jun Shu
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Li-jia Pan
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Wei Gong
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| | - Ping Dong
- Laboratory of General Surgery and Department of General Surgery, Xinhua Hospital Affiliated With Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092 China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai, 200092 China
| |
Collapse
|
17
|
English KG, Reid AL, Samani A, Coulis GJF, Villalta SA, Walker CJ, Tamir S, Alexander MS. Next-Generation SINE Compound KPT-8602 Ameliorates Dystrophic Pathology in Zebrafish and Mouse Models of DMD. Biomedicines 2022; 10:2400. [PMID: 36289662 PMCID: PMC9598711 DOI: 10.3390/biomedicines10102400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive, X-linked childhood neuromuscular disorder that results from loss-of-function mutations in the DYSTROPHIN gene. DMD patients exhibit muscle necrosis, cardiomyopathy, respiratory failure, and loss of ambulation. One of the major driving forces of DMD disease pathology is chronic inflammation. The current DMD standard of care is corticosteroids; however, there are serious side effects with long-term use, thus identifying novel anti-inflammatory and anti-fibrotic treatments for DMD is of high priority. We investigated the next-generation SINE compound, KPT-8602 (eltanexor) as an oral therapeutic to alleviate dystrophic symptoms. We performed pre-clinical evaluation of the effects of KPT-8602 in DMD zebrafish (sapje) and mouse (D2-mdx) models. KPT-8602 improved dystrophic skeletal muscle pathologies, muscle architecture and integrity, and overall outcomes in both animal models. KPT-8602 treatment ameliorated DMD pathology in D2-mdx mice, with increased locomotor behavior and improved muscle histology. KPT-8602 altered the immunological profile of the dystrophic mice, and reduced circulating osteopontin serum levels. These findings demonstrate KPT-8602 as an effective therapeutic in DMD through by promotion of an anti-inflammatory environment and overall improvement of DMD pathological outcomes.
Collapse
Affiliation(s)
- Katherine G. English
- Department of Pediatrics, Division of Neurology at Children’s of Alabama the University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Andrea L. Reid
- Department of Pediatrics, Division of Neurology at Children’s of Alabama the University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Adrienne Samani
- Department of Pediatrics, Division of Neurology at Children’s of Alabama the University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Gerald J. F. Coulis
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA
- Institute for Immunology, University of California-Irvine, Irvine, CA 92967, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA
- Institute for Immunology, University of California-Irvine, Irvine, CA 92967, USA
| | | | | | - Matthew S. Alexander
- Department of Pediatrics, Division of Neurology at Children’s of Alabama the University of Alabama at Birmingham, Birmingham, AL 35233, USA
- UAB Center for Exercise Medicine (UCEM), Birmingham, AL 35205, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- UAB Civitan International Research Center (CIRC), Birmingham, AL 35233, USA
- UAB Center for Neurodegeneration and Experimental Therapeutics (CNET), Birmingham, AL 35294, USA
| |
Collapse
|
18
|
The efficacy of selinexor (KPT-330), an XPO1 inhibitor, on non-hematologic cancers: a comprehensive review. J Cancer Res Clin Oncol 2022; 149:2139-2155. [PMID: 35941226 DOI: 10.1007/s00432-022-04247-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/01/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE Selinexor is a novel XPO1 inhibitor which inhibits the export of tumor suppressor proteins and oncoprotein mRNAs, leading to cell-cycle arrest and apoptosis in cancer cells. While selinexor is currently FDA approved to treat multiple myeloma, compelling preclinical and early clinical studies reveal selinexor's efficacy in treating hematologic and non-hematologic malignancies, including sarcoma, gastric, bladder, prostate, breast, ovarian, skin, lung, and brain cancers. Current reviews of selinexor primarily highlight its use in hematologic malignancies; however, this review seeks to summarize the recent evidence of selinexor treatment in solid tumors. METHODS Pertinent literature searches in PubMed and the Karyopharm Therapeutics website for selinexor and non-hematologic malignancies preclinical and clinical trials. RESULTS This review provides evidence that selinexor is a promising agent used alone or in combination with other anticancer medications in non-hematologic malignancies. CONCLUSION Further clinical investigation of selinexor treatment for solid malignancies is warranted.
Collapse
|
19
|
Sellin M, Berg S, Hagen P, Zhang J. The molecular mechanism and challenge of targeting XPO1 in treatment of relapsed and refractory myeloma. Transl Oncol 2022; 22:101448. [PMID: 35660848 PMCID: PMC9166471 DOI: 10.1016/j.tranon.2022.101448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/14/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022] Open
Abstract
Significant progress has been made on the treatment of MM during past two decades. Acquired drug-resistance continues to drive early relapse in primary refractory MM. XPO1 over-expression and cargo mislocalization are associated with drug-resistance. XPO1 inhibitor selinexor restores drug sensitivity to subsets of RR-MM cells.
Multiple myeloma (MM) treatment regimens have vastly improved since the introduction of immunomodulators, proteasome inhibitors, and anti-CD38 monoclonal antibodies; however, MM is considered an incurable disease due to inevitable relapse and acquired drug resistance. Understanding the molecular mechanism by which drug resistance is acquired will help create novel strategies to prevent relapse and help develop novel therapeutics to treat relapsed/refractory (RR)-MM patients. Currently, only homozygous deletion/mutation of TP53 gene due to “double-hits” on Chromosome 17p region is consistently associated with a poor prognosis. The exciting discovery of XPO1 overexpression and mislocalization of its cargos in the RR-MM cells has led to a novel treatment options. Clinical studies have demonstrated that the XPO1 inhibitor selinexor can restore sensitivity of RR-MM to PIs and dexamethasone. We will elaborate on the problems of MM treatment strategies and discuss the mechanism and challenges of using XPO1 inhibitors in RR-MM therapies while deliberating potential solutions.
Collapse
Affiliation(s)
- Mark Sellin
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Loyola University Chicago, USA
| | - Stephanie Berg
- Loyola University Chicago, Department of Cancer Biology and Internal Medicine, Cardinal Bernardin Cancer Center, Stritch School of Medicine, Maywood, IL, USA.
| | - Patrick Hagen
- Department of Medicine, Division of Hematology/Oncology, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, USA
| |
Collapse
|
20
|
Guo Y, Liu Z, Duan L, Shen H, Ding K, Fu R. Selinexor synergizes with azacitidine to eliminate myelodysplastic syndrome cells through p53 nuclear accumulation. Invest New Drugs 2022; 40:738-746. [PMID: 35576022 DOI: 10.1007/s10637-022-01251-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
Myelodysplastic syndromes (MDS) are clonal malignancies of multipotent hematopoietic stem cells, characterized by ineffective hematopoiesis leading to cytopenia. Hypomethylating agents, including azacitidine, have been used for treating MDS with some success; however, the overall survival rate remains poor and, therefore, finding new therapies is necessary. Selinexor, which exerts anticancer effects against some hematologic tumors, is a nuclear export protein inhibitor that blocks cell proliferation and induces apoptosis in various cancer cell lines. We investigated the effects of combined selinexor and azacitidine administration on two MDS cell lines, namely SKM-1 and MUTZ-1. Cells were subjected to a proliferation assay, and the effects of each drug alone, and in combination, were compared. Changes in apoptosis and the cell cycle between groups were also analyzed. Western blotting was conducted to identify the underlying mechanism of action of combined selinexor and azacitidine therapy. The results revealed that the combination of selinexor and azacitidine synergistically inhibited MDS cell proliferation and arrested the cell cycle at the G2/M phase. This combination also promoted MDS cell apoptosis and enhanced p53 accumulation in the nucleus, thereby allowing p53 to be activated and to function as a tumor suppressor. Overall, our results indicate that the combination of selinexor and azacitidine may be a promising approach for treating MDS.
Collapse
Affiliation(s)
- Yixuan Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lixiang Duan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Hematology, Yuncheng Central Hospital, Shanxi, China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
21
|
Yu H, Wu S, Liu S, Li X, Gai Y, Lin H, Wang Y, Edwards H, Ge Y, Wang G. Venetoclax enhances DNA damage induced by XPO1 inhibitors: A novel mechanism underlying the synergistic antileukaemic effect in acute myeloid leukaemia. J Cell Mol Med 2022; 26:2646-2657. [PMID: 35355406 PMCID: PMC9077288 DOI: 10.1111/jcmm.17274] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a highly heterogeneous haematologic malignancy with poor prognosis. We previously showed synergistic antileukaemic interaction between exportin 1 (XPO1) inhibitor KPT-330 (Selinexor) and Bcl-2 inhibitor venetoclax (ABT-199) in preclinical models of AML, which was partially meditated by Mcl-1, although the full mechanism of action remains unknown. In this study, using real-time RT-PCR and Western blot analysis, we show that inhibition of XPO1 via KPT-330 or KPT-8602 (Eltanexor) decreases the mRNA and protein levels of c-Myc, CHK1, WEE1, RAD51 and RRM2. KPT-330 and KPT-8602 induce DNA damage, as determined by alkaline comet assay. In addition, we demonstrate that venetoclax enhances KPT-330- and KPT-8602-induced DNA damage, likely through inhibition of DNA damage repair. This study provides new insight into the molecular mechanism underlying the synergistic antileukaemic activity between venetoclax and XPO1 inhibitors against AML. Our data support the clinical evaluation of this promising combination therapy for the treatment of AML.
Collapse
Affiliation(s)
- Hanxi Yu
- National Engineering Laboratory for AIDS VaccineKey Laboratory for Molecular Enzymology and Engineeringthe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunChina
| | - Shuangshuang Wu
- National Engineering Laboratory for AIDS VaccineKey Laboratory for Molecular Enzymology and Engineeringthe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunChina
| | - Shuang Liu
- National Engineering Laboratory for AIDS VaccineKey Laboratory for Molecular Enzymology and Engineeringthe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunChina
| | - Xinyu Li
- National Engineering Laboratory for AIDS VaccineKey Laboratory for Molecular Enzymology and Engineeringthe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunChina
| | - Yuqing Gai
- National Engineering Laboratory for AIDS VaccineKey Laboratory for Molecular Enzymology and Engineeringthe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunChina
| | - Hai Lin
- Department of Hematology and Oncologythe First Hospital of Jilin UniversityChangchunChina
| | - Yue Wang
- Department of Pediatric Hematology and Oncologythe First Hospital of Jilin UniversityChangchunChina
| | - Holly Edwards
- Department of Oncology and Molecular Therapeutics ProgramBarbara Ann Karmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Yubin Ge
- Department of Oncology and Molecular Therapeutics ProgramBarbara Ann Karmanos Cancer InstituteWayne State University School of MedicineDetroitMichiganUSA
| | - Guan Wang
- National Engineering Laboratory for AIDS VaccineKey Laboratory for Molecular Enzymology and Engineeringthe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunChina
| |
Collapse
|
22
|
Mead BE, Hattori K, Levy L, Imada S, Goto N, Vukovic M, Sze D, Kummerlowe C, Matute JD, Duan J, Langer R, Blumberg RS, Ordovas-Montanes J, Yilmaz ÖH, Karp JM, Shalek AK. Screening for modulators of the cellular composition of gut epithelia via organoid models of intestinal stem cell differentiation. Nat Biomed Eng 2022; 6:476-494. [PMID: 35314801 PMCID: PMC9046079 DOI: 10.1038/s41551-022-00863-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
Abstract
The cellular composition of barrier epithelia is essential to organismal homoeostasis. In particular, within the small intestine, adult stem cells establish tissue cellularity, and may provide a means to control the abundance and quality of specialized epithelial cells. Yet, methods for the identification of biological targets regulating epithelial composition and function, and of small molecules modulating them, are lacking. Here we show that druggable biological targets and small-molecule regulators of intestinal stem cell differentiation can be identified via multiplexed phenotypic screening using thousands of miniaturized organoid models of intestinal stem cell differentiation into Paneth cells, and validated via longitudinal single-cell RNA-sequencing. We found that inhibitors of the nuclear exporter Exportin 1 modulate the fate of intestinal stem cells, independently of known differentiation cues, significantly increasing the abundance of Paneth cells in the organoids and in wild-type mice. Physiological organoid models of the differentiation of intestinal stem cells could find broader utility for the screening of biological targets and small molecules that can modulate the composition and function of other barrier epithelia.
Collapse
Grants
- R01 DK088199 NIDDK NIH HHS
- Howard Hughes Medical Institute
- P30 CA014051 NCI NIH HHS
- DP2 GM119419 NIGMS NIH HHS
- R01 DE013023 NIDCR NIH HHS
- U54 CA217377 NCI NIH HHS
- P30 DK034854 NIDDK NIH HHS
- R01 HL095722 NHLBI NIH HHS
- T32 GM087237 NIGMS NIH HHS
- R01 CA034992 NCI NIH HHS
- R01 CA211184 NCI NIH HHS
- The National Science Foundation graduate research fellowship program and the Massachusetts Institute of Technology – GlaxoSmithKline (MIT-GSK) Gertrude B. Elion Postdoctoral fellowship.
- Fellowships from The Japanese Biochemical Society (The Osamu Hayaishi Memorial Scholarship for Study Abroad), Mochida Memorial Foundation for Medical and Pharmaceutical Research, and The Uehara Memorial Foundation.
- NIH (DE013023)
- NIH (DK088199)
- New York Stem Cell Foundation – Robertson Investigator, the Richard and Susan Smith Family Foundation, the HHMI Damon Runyon Cancer Research Foundation Fellowship (DRG-2274-16), the AGA Research Foundation’s AGA-Takeda Pharmaceuticals Research Scholar Award in IBD – AGA2020-13-01, the HDDC Pilot and Feasibility P30 DK034854, the Food Allergy Science Initiative, and The New York Stem Cell Foundation.
- NIH (R01CA211184, R01CA034992); Pew-Stewart Trust scholar award; the Kathy and Curt Marble Cancer Research Award; a Bridge grant; and the MIT Stem Cell Initiative through Fondation MIT.
- the Kenneth Rainin Foundation Innovator and Breakthrough awards, the Crohn’s and Colitis Foundation (#624458),the NIH (HL095722), and the Harvard Digestive Disease Center and NIH grant P30DK034854.
- the Beckman Young Investigator Program, the Pew-Stewart Scholars Program for Cancer Research, a Sloan Fellowship in Chemistry, the NIH (1DP2GM119419, 1U54CA217377), the Koch Institute Support (core) Grant P30-CA14051 from the National Cancer Institute, and the MIT Stem Cell Initiative through Fondation MIT.
Collapse
Affiliation(s)
- Benjamin E Mead
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Kazuki Hattori
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren Levy
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shinya Imada
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Norihiro Goto
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Marko Vukovic
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology Boston Children's Hospital, Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Daphne Sze
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Conner Kummerlowe
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Juan D Matute
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Neonatology, Department of Pediatrics, MGH Harvard Medical School, Boston, MA, USA
| | - Jinzhi Duan
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert Langer
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jose Ordovas-Montanes
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology Boston Children's Hospital, Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Ömer H Yilmaz
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Pathology, MGH, Harvard Medical School, Boston, MA, USA
| | - Jeffrey M Karp
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Alex K Shalek
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA.
- Department of Chemistry, MIT, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
23
|
Zhao L, Luo B, Wang L, Chen W, Jiang M, Zhang N. Pan-cancer analysis reveals the roles of XPO1 in predicting prognosis and tumorigenesis. Transl Cancer Res 2022; 10:4664-4679. [PMID: 35116322 PMCID: PMC8797940 DOI: 10.21037/tcr-21-1646] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Background Exportin 1 (XPO1), a nuclear export protein, participates in many biological processes, including mRNA transport, nucleocytoplasmic transport, nuclear protein export, regulation of mRNA stability, and drug response. XPO1 plays key roles in many cancer types and may serve as a potential biomarker. It is significant to systematically elucidate the roles of XPO1 in various cancer types in terms of function, molecular biology, immunology, and clinical relevance. Methods Data from UCSC Xena, CCLE, and CBioPortal were analyzed for the investigation of the differential expression of XPO1 across multiple cancer types. Clinical data were acquired to analyze the influence of XPO1 on the clinical characteristics of patients, such as survival outcome and clinical stage. The roles of XPO1 in the onset and progression of multiple cancers were expounded in terms of genetic changes at the molecular level [including tumor mutational burden (TMB), microsatellite instability (MSI), copy number variation (CNV), methylation, and gene co-expression], biological pathway changes, and the immune microenvironment. Results XPO1 was overexpressed in various tumor types, which may be related to CNV. Clinical data analysis revealed that XPO1 may serve as a risk factor in tumors, such as adrenocortical carcinoma, liver hepatocellular carcinoma, and low-grade glioma, thereby affecting patient prognosis. XPO1 in multiple tumor types was also substantially correlated with clinical stage, patient gender, and patient age. In certain tumors, the expression level of XPO1 exerted a greater influence on TMB and MSI. It was also found that XPO1 inhibited the activity of immune cells in the tumor immune microenvironment, such as CD8+ T cells, and affected biological pathways, such as the cell cycle and oxidative phosphorylation, and drove the expression of cancer driver genes, immune checkpoint genes, and highly mutated genes. Conclusions XPO1 is a potential pan-cancer risk factor as it may jointly promote tumor onset and progression by inhibiting the immune response, influencing relevant biological pathways, and promoting mutations in other genes.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Critical Care Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Baiwei Luo
- The First Clinical Medical School of Guangdong Medical University, Zhanjiang, China
| | - Liang Wang
- Department of Hematology, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Wei Chen
- Department of Critical Care Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Manyu Jiang
- The First Clinical Medical School of Guangdong Medical University, Zhanjiang, China
| | - Nengwei Zhang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Kim E, Mordovkina DA, Sorokin A. Targeting XPO1-Dependent Nuclear Export in Cancer. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:S178-S70. [PMID: 35501995 DOI: 10.1134/s0006297922140140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 06/14/2023]
Abstract
Nucleocytoplasmic transport of macromolecules is tightly regulated in eukaryotic cells. XPO1 is a transport factor responsible for the nuclear export of several hundred protein and RNA substrates. Elevated levels of XPO1 and recurrent mutations have been reported in multiple cancers and linked to advanced disease stage and poor survival. In recent years, several novel small-molecule inhibitors of XPO1 were developed and extensively tested in preclinical cancer models and eventually in clinical trials. In this brief review, we summarize the functions of XPO1, its role in cancer, and the latest results of clinical trials of XPO1 inhibitors.
Collapse
Affiliation(s)
- Ekaterina Kim
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Daria A Mordovkina
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Alexey Sorokin
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
25
|
Ou L, Wang X, Cheng S, Zhang M, Cui R, Hu C, Liu S, Tang Q, Peng Y, Chai R, Xie S, Wang S, Huang W, Wang X. Verdinexor, a Selective Inhibitor of Nuclear Exportin 1, Inhibits the Proliferation and Migration of Esophageal Cancer via XPO1/c-Myc/FOSL1 Axis. Int J Biol Sci 2022; 18:276-291. [PMID: 34975332 PMCID: PMC8692140 DOI: 10.7150/ijbs.66612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
Esophageal carcinoma (EC) ranks sixth among cancers in mortality worldwide and effective drugs to reduce EC incidence and mortality are lacking. To explore potential anti-esophageal cancer drugs, we conducted drug screening and discovered that verdinexor, a selective inhibitor of nuclear exportin 1 (XPO1/CRM1), has anti-esophageal cancer effects both in vivo and in vitro. However, the mechanism and role of verdinexor in esophageal cancer remain unknown. In the present study, we observed that verdinexor inhibited the proliferation and migration of EC cells in vitro and suppressed tumor growth in vivo. Additionally, we found that verdinexor induced cleavage of PARP and downregulated XPO1, c-Myc, and FOSL1 expression. RNA-sequence analysis and protein-protein interaction (PPI) analysis revealed that verdinexor regulated the XPO1/c-Myc/FOSL1 axis. The results of immunoprecipitation and proximity ligation assays confirmed that verdinexor disrupted the interaction between XPO1 and c-Myc. Overexpression of c-Myc rescued the inhibition of cell proliferation and cell migration caused by verdinexor. Overexpressed FOSL1 restored the inhibited migration by verdinexor. Taken together, verdinexor inhibited cell proliferation and migration of esophageal cancer via XPO1/c-Myc/FOSL1 axis. Our findings provide a new option for the development of anti-esophageal cancer drugs.
Collapse
Affiliation(s)
- Ling Ou
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Xinyou Wang
- The First District of Gastrointestinal Surgery, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shumin Cheng
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Min Zhang
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Ruiqin Cui
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Chunxia Hu
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Shiyi Liu
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Qian Tang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmacy, Jinan University, Guangzhou 510630, Guangdong, China
| | - Yuying Peng
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmacy, Jinan University, Guangzhou 510630, Guangdong, China
| | - Ruihuan Chai
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Shouxia Xie
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Shaoxiang Wang
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Wei Huang
- Bacteriology & Antibacterial Resistance Surveillance Laboratory, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- School of Pharmacy, Jinan University, Guangzhou 510630, Guangdong, China
| |
Collapse
|
26
|
Özdaş S, Canatar İ. Targeting of nucleo‑cytoplasmic transport factor exportin 1 in malignancy (Review). MEDICINE INTERNATIONAL 2022; 2:2. [PMID: 38938904 PMCID: PMC11208992 DOI: 10.3892/mi.2021.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/03/2021] [Indexed: 06/29/2024]
Abstract
Nuclear pore complexes (NPCs) regulate the entry and exit of molecules from the cell nucleus. Small molecules pass through NPCs by diffusion while large molecules enter and exit the nucleus by karyopherins, which serve as transport factors. Exportin-1 (XPO1) is a protein that is an important member of the karyopherin family and carries macromolecules from the nucleus to the cytoplasm. XPO1 is responsible for nuclear-cytoplasmic transport of protein, ribosomal RNA and certain required mRNAs for ribosomal biogenesis. Furthermore, XPO1-mediated nuclear export is associated with various types of disease, such as cancer, inflammation and viral infection. The key role of XPO1 in carcinogenesis and its potential as a therapeutic target has been demonstrated by previous studies. Clinical use of novel developed generation-specific XPO1 inhibitors and their combination with other agents to block XPO1-mediated nuclear export are a promising new treatment strategy. The aim of the present study was to explain the working mechanism of XPO1 and inhibitors that block XPO1-mediated nuclear export.
Collapse
Affiliation(s)
- Sibel Özdaş
- Department of Bioengineering, Faculty of Engineering Sciences, Adana Alparslan Türkeş Science and Technology University, Adana 01250, Turkey
| | - İpek Canatar
- Department of Bioengineering, Faculty of Engineering Sciences, Adana Alparslan Türkeş Science and Technology University, Adana 01250, Turkey
| |
Collapse
|
27
|
Passirani C, Vessières A, La Regina G, Link W, Silvestri R. Modulating undruggable targets to overcome cancer therapy resistance. Drug Resist Updat 2021; 60:100788. [DOI: 10.1016/j.drup.2021.100788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/03/2022]
|
28
|
von Fallois M, Kosyna FK, Mandl M, Landesman Y, Dunst J, Depping R. Selinexor decreases HIF-1α via inhibition of CRM1 in human osteosarcoma and hepatoma cells associated with an increased radiosensitivity. J Cancer Res Clin Oncol 2021; 147:2025-2033. [PMID: 33856525 PMCID: PMC8164574 DOI: 10.1007/s00432-021-03626-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/30/2021] [Indexed: 10/29/2022]
Abstract
BACKGROUND The nuclear pore complexes (NPCs) are built of about 30 different nucleoporins and act as key regulators of molecular traffic between the cytoplasm and the nucleus for sizeable proteins (> 40 kDa) which must enter the nucleus. Various nuclear transport receptors are involved in import and export processes of proteins through the nuclear pores. The most prominent nuclear export receptor is chromosome region maintenance 1 (CRM1), also known as exportin 1 (XPO1). One of its cargo proteins is the prolyl hydroxylase 2 (PHD2) which is involved in the initiation of the degradation of hypoxia-inducible factors (HIFs) under normoxia. HIFs are proteins that regulate the cellular adaptation under hypoxic conditions. They are involved in many aspects of cell viability and play an important role in the hypoxic microenvironment of cancer. In cancer, CRM1 is often overexpressed thus being a putative target for the development of new cancer therapies. The newly FDA-approved pharmaceutical Selinexor (KPT-330) selectively inhibits nuclear export via CRM1 and is currently tested in additional Phase-III clinical trials. In this study, we investigated the effect of CRM1 inhibition on the subcellular localization of HIF-1α and radiosensitivity. METHODS Human hepatoma cells Hep3B and human osteosarcoma cells U2OS were treated with Selinexor. Intranuclear concentration of HIF-1α protein was measured using immunoblot analysis. Furthermore, cells were irradiated with 2-8 Gy after treatment with Selinexor compared to untreated controls. RESULTS Selinexor significantly reduced the intranuclear level of HIF-1α protein in human hepatoma cells Hep3B and human osteosarcoma cells U2OS. Moreover, we demonstrated by clonogenic survival assays that Selinexor leads to dose-dependent radiosensitization in Hep3B-hepatoma and U2OS-osteosarcoma cells. CONCLUSION Targeting the HIF pathway by Selinexor might be an attractive tool to overcome hypoxia-induced radioresistance.
Collapse
MESH Headings
- Apoptosis
- Bone Neoplasms/drug therapy
- Bone Neoplasms/metabolism
- Bone Neoplasms/pathology
- Bone Neoplasms/radiotherapy
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/radiotherapy
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Humans
- Hydrazines/pharmacology
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Karyopherins/antagonists & inhibitors
- Karyopherins/genetics
- Karyopherins/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/radiotherapy
- Osteosarcoma/drug therapy
- Osteosarcoma/metabolism
- Osteosarcoma/pathology
- Osteosarcoma/radiotherapy
- Radiation Tolerance/drug effects
- Radiation-Sensitizing Agents/pharmacology
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Triazoles/pharmacology
- Tumor Cells, Cultured
- Exportin 1 Protein
Collapse
Affiliation(s)
- Moritz von Fallois
- Universität Zu Lübeck, Institut Für Physiologie, Working Group Hypoxia, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Friederike Katharina Kosyna
- Universität Zu Lübeck, Institut Für Physiologie, Working Group Hypoxia, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Markus Mandl
- Universität Zu Lübeck, Institut Für Physiologie, Working Group Hypoxia, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Yosef Landesman
- Karyopharm Therapeutics, 85 Wells Ave, Newton, MA, 02459, USA
| | - Jürgen Dunst
- Universitätsklinikum Schleswig-Holstein, Campus Kiel-Klinik für Strahlentherapie, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Reinhard Depping
- Universität Zu Lübeck, Institut Für Physiologie, Working Group Hypoxia, Ratzeburger Allee 160, 23562, Lübeck, Germany.
| |
Collapse
|
29
|
Venetoclax response is enhanced by selective inhibitor of nuclear export compounds in hematologic malignancies. Blood Adv 2021; 4:586-598. [PMID: 32045477 DOI: 10.1182/bloodadvances.2019000359] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022] Open
Abstract
The selective inhibitor of nuclear export (SINE) compounds selinexor (KPT-330) and eltanexor (KPT-8602) are from a novel class of small molecules that target exportin-1 (XPO1 [CRM1]), an essential nucleo-cytoplasmic transport protein responsible for the nuclear export of major tumor suppressor proteins and growth regulators such as p53, p21, and p27. XPO1 also affects the translation of messenger RNAs for critical oncogenes, including MYC, BCL2, MCL1, and BCL6, by blocking the export of the translation initiation factor eIF4E. Early trials with venetoclax (ABT-199), a potent, selective inhibitor of BCL2, have revealed responses across a variety of hematologic malignancies. However, many tumors are not responsive to venetoclax. We used models of acute myeloid leukemia (AML) and diffuse large B-cell lymphoma (DLBCL) to determine in vitro and in vivo responses to treatment with venetoclax and SINE compounds combined. Cotreatment with venetoclax and SINE compounds demonstrated loss of viability in multiple cell lines. Further in vitro analyses showed that this enhanced cell death was the result of an increase in apoptosis that led to a loss of clonogenicity in methylcellulose assays, coinciding with activation of p53 and loss of MCL1. Treatment with SINE compounds and venetoclax combined led to a reduction in tumor growth in both AML and DLBCL xenografts. Immunohistochemical analysis of tissue sections revealed that the reduction in tumor cells was partly the result of an induction of apoptosis. The enhanced effects of this combination were validated in primary AML and DLBCL patient cells. Our studies reveal synergy with SINE compounds and venetoclax in aggressive hematologic malignancies and provide a rationale for pursuing this approach in a clinical trial.
Collapse
|
30
|
Liao Y, Ke X, Deng T, Qin Q. The Second-Generation XPO1 Inhibitor Eltanexor Inhibits Human Cytomegalovirus (HCMV) Replication and Promotes Type I Interferon Response. Front Microbiol 2021; 12:675112. [PMID: 34012430 PMCID: PMC8126617 DOI: 10.3389/fmicb.2021.675112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/12/2021] [Indexed: 02/05/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous opportunistic pathogen and can be life-threatening for immunocompromised individuals. There is currently no available vaccine for the prevention of HCMV- associated diseases and most of the available antiviral drugs that target viral DNA synthesis become ineffective in treating HCMV mutants that arise after long-term use in immunocompromised patients. Here, we examined the effects of Eltanexor, a second-generation selective inhibitor of nuclear export (SINE), on HCMV replication. Eltanexor effectively inhibits HCMV replication in human foreskin fibroblasts in a dose-dependent manner. Eltanexor does not significantly inhibit viral entry and nuclear import of viral genomic DNA, but rather suppress the transcript and protein levels of viral immediate-early (IE), early (E) and late (L) genes, and abolishes the production of infectious virions. We further found Eltanexor treatment promotes proteasome-mediated degradation of XPO1, which contributes to the nuclear retention of interferon regulatory factor 3 (IRF-3), resulting in increased expression of type I interferon as well as interferon stimulating genes ISG15 and ISG54. This study reveals a novel antiviral mechanism of Eltanexor which suggests it has potential to inhibit a broad spectrum of viral pathogens.
Collapse
Affiliation(s)
- Yueyan Liao
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, China
| | - Xiangyu Ke
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, China
| | - Tianyi Deng
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, China
| | - Qingsong Qin
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou, China
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou, China
- *Correspondence: Qingsong Qin,
| |
Collapse
|
31
|
Turner JG, Cui Y, Bauer AA, Dawson JL, Gomez JA, Kim J, Cubitt CL, Nishihori T, Dalton WS, Sullivan DM. Melphalan and Exportin 1 Inhibitors Exert Synergistic Antitumor Effects in Preclinical Models of Human Multiple Myeloma. Cancer Res 2020; 80:5344-5354. [PMID: 33023948 PMCID: PMC7718436 DOI: 10.1158/0008-5472.can-19-0677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 08/25/2020] [Accepted: 10/01/2020] [Indexed: 01/22/2023]
Abstract
High-dose chemotherapy with melphalan followed by autologous transplantation is a first-line treatment for multiple myeloma. Here, we present preclinical evidence that this treatment may be significantly improved by the addition of exportin 1 inhibitors (XPO1i). The XPO1i selinexor, eltanexor, and KOS-2464 sensitized human multiple myeloma cells to melphalan. Human 8226 and U266 multiple myeloma cell lines and melphalan-resistant cell lines (8226-LR5 and U266-LR6) were highly sensitized to melphalan by XPO1i. Multiple myeloma cells from newly diagnosed and relapsed/refractory multiple myeloma patients were also sensitized by XPO1i to melphalan. In NOD/SCIDγ mice challenged with either parental 8226 or U266 multiple myeloma and melphalan-resistant multiple myeloma tumors, XPO1i/melphalan combination treatments demonstrated stronger synergistic antitumor effects than single-agent melphalan with minimal toxicity. Synergistic cell death resulted from increased XPO1i/melphalan-induced DNA damage in a dose-dependent manner and decreased DNA repair. In addition, repair of melphalan-induced DNA damage was inhibited by selinexor, which decreased melphalan-induced monoubiquitination of FANCD2 in multiple myeloma cells. Knockdown of FANCD2 was found to replicate the effect of selinexor when used with melphalan, increasing DNA damage (γH2AX) by inhibiting DNA repair. Thus, combination therapies that include selinexor or eltanexor with melphalan may have the potential to improve treatment outcomes of multiple myeloma in melphalan-resistant and newly diagnosed patients. The combination of selinexor and melphalan is currently being investigated in the context of high-dose chemotherapy and autologous transplant (NCT02780609). SIGNIFICANCE: Inhibition of exportin 1 with selinexor synergistically sensitizes human multiple myeloma to melphalan by inhibiting Fanconi anemia pathway-mediated DNA repair.
Collapse
Affiliation(s)
- Joel G Turner
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Yan Cui
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Alexis A Bauer
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jana L Dawson
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Juan A Gomez
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Christopher L Cubitt
- Translational Research Core, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - William S Dalton
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Daniel M Sullivan
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
32
|
Walker JS, Garzon R, Lapalombella R. Selinexor for advanced hematologic malignancies. Leuk Lymphoma 2020; 61:2335-2350. [DOI: 10.1080/10428194.2020.1775210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Janek S. Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
33
|
Azizian NG, Li Y. XPO1-dependent nuclear export as a target for cancer therapy. J Hematol Oncol 2020; 13:61. [PMID: 32487143 PMCID: PMC7268335 DOI: 10.1186/s13045-020-00903-4] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cellular homeostasis requires the proper nuclear-cytoplasmic partitioning of large molecules, which is often deregulated in cancer. XPO1 is an export receptor responsible for the nuclear-cytoplasmic transport of hundreds of proteins and multiple RNA species. XPO1 is frequently overexpressed and/or mutated in human cancers and functions as an oncogenic driver. Suppression of XPO1-mediated nuclear export, therefore, presents a unique therapeutic strategy. In this review, we summarize the physiological functions of XPO1 as well as the development of various XPO1 inhibitors and provide an update on the recent clinical trials of the SINE compounds. We also discuss potential future research directions on the molecular function of XPO1 and the clinical application of XPO1 inhibitors.
Collapse
Affiliation(s)
- Nancy G Azizian
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yulin Li
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
34
|
Ferreira BI, Cautain B, Grenho I, Link W. Small Molecule Inhibitors of CRM1. Front Pharmacol 2020; 11:625. [PMID: 32574233 PMCID: PMC7221118 DOI: 10.3389/fphar.2020.00625] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/20/2020] [Indexed: 12/19/2022] Open
Abstract
The transport through the nuclear pore complex is used by cancer cells to evade tumor-suppressive mechanisms. Several tumor-suppressors have been shown to be excluded from the cell nucleus in cancer cells by the nuclear export receptor CRM1 and abnormal expression of CRM1 is oncogenic. Inhibition of CRM1 has long been postulated as potential approach for the treatment of cancer and to overcome therapy resistance. Furthermore, the nuclear export of viral components mediated by the CRM1 is crucial in various stages of the viral lifecycle and assembly of many viruses from diverse families, including coronavirus. However, the first nuclear export inhibitors failed or never entered into clinical trials. More recently CRM1 reemerged as a cancer target and a successful proof of concept was achieved with the clinical approval of Selinexor. The chemical complexity of natural products is a promising perspective for the discovery of new nuclear export inhibitors with a favorable toxicity profile. Several screening campaigns have been performed and several natural product-based nuclear export inhibitors have been identified. With this review we give an overview over the role of CRM1-mediated nuclear export in cancer and the effort made to identify and develop nuclear export inhibitors in particular from natural sources.
Collapse
Affiliation(s)
- Bibiana I Ferreira
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal.,Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Bastien Cautain
- Fundacion MEDINA Parque tecnológico ciencias de la salud, Granada, Spain.,Evotec France, Toulouse, France
| | - Inês Grenho
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal.,Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| |
Collapse
|
35
|
Aladhraei M, Al-Thobhani AK, Poungvarin N, Suwannalert P. Association of XPO1 Overexpression with NF-κB and Ki67 in Colorectal Cancer. Asian Pac J Cancer Prev 2019; 20:3747-3754. [PMID: 31870117 PMCID: PMC7173379 DOI: 10.31557/apjcp.2019.20.12.3747] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/29/2019] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Exportin 1(XPO1), a nuclear exporter protein, has been gaining recognition in cancer progression and treatment. This study aimed to evaluate the association between the overexpression of XPO1 with NF-κB, Ki67 and clinicopathological characteristics in colorectal cancer (CRC) tissue samples and to explore the anti-proliferative effect of KPT-330, as XPO1 inhibitor, in colorectal cancer cell line. METHODS Forty CRC tissue samples were analyzed by immunostaining for the expressions of XPO1, NF-κB and Ki67 and then the anti-proliferative effect of the KPT-330 was also evaluated in HT29 colorectal cancer cell line. RESULTS XPO1 overexpression was observed in 52.5% of CRC and significantly apparent with strong intensity in tumor cells compared to the normal adjacent epithelium (P<0.001). Regarding to the histopathological characteristics, the XPO1 overexpression significantly associated with advanced tumor stages (P=0.049) and has great tendency towards moderate/poorly differentiated tumors. Although the XPO1 overexpression was strongly associated with high Ki67 expression (P=0.001), only Ki67 expression showed significant association with tumor size (P=0.012). No significant association was detected between the XPO1 overexpression and NF-κB, while the NF-κB positive expression was significantly associated with lymph node metastasis and Ki67 expression at P=0.027 and P= 0.007, respectively. The in vitro experiments showed a great impact of KPT-330, as XPO1 inhibitor, to inhibit cancer growth in dose and time dependent manner and significantly diminished the colony formation (P<0.001) of HT29 cells- associated with the expression of Ki67 (P<0.001). CONCLUSION XPO1 overexpression and NF-κB expression may serve as potential biomarker associated with CRC pathogenesis and proliferation, while the KPT-330 is effectively inhibited-colon cancer growth in vitro. Further studies considering the prognostication role of XPO1 overexpression in CRC are required. .
Collapse
Affiliation(s)
| | - Abdulla Kassem Al-Thobhani
- Clinical Molecular Pathology Laboratory, Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand,
| | - Naravat Poungvarin
- Department of Pathology, Faculty of Medicine and Health Sciences, University of Sana’a, Sana’a, Yemen.
| | | |
Collapse
|
36
|
Liu Y, Azizian NG, Dou Y, Pham LV, Li Y. Simultaneous targeting of XPO1 and BCL2 as an effective treatment strategy for double-hit lymphoma. J Hematol Oncol 2019; 12:119. [PMID: 31752970 PMCID: PMC6868798 DOI: 10.1186/s13045-019-0803-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/07/2019] [Indexed: 12/22/2022] Open
Abstract
Double-hit lymphoma (DHL) is among the most aggressive and chemoresistant lymphoma subtypes. DHLs carry genomic abnormalities in MYC, BCL2, and/or BCL6 oncogenes. Due to the simultaneous overexpression of these driver oncogenes, DHLs are highly resistant to frontline therapies. Most DHLs overexpress both MYC and BCL2 driver oncogenes concurrently. We reasoned that simultaneous suppression of the two driver oncogenes would be more effective in eradicating DHLs than inactivation of single oncogene. XPO1 is a receptor for nuclear cytoplasmic transport of protein and RNA species. Recently, XPO1 inhibition was shown to downregulate MYC expression in several cancer cell lines. We therefore examined the role of XPO1 as a therapeutic target in suppressing MYC function and the potential synergistic effects of simultaneous suppression of XPO1 and BCL2 in the treatment of DHL. Here, we demonstrate that XPO1 inhibition abrogates MYC protein expression and induces massive tumor cell apoptosis. Combined use of XPO1 and BCL2 inhibitors is highly effective in eradicating DHL cells in cell culture. Notably, in a mouse model of DHL bearing primary tumor cells derived from lymphoma patients, combined treatment with XPO1 and BCL2 inhibitors blocks tumor progression, prevents brain metastasis, and extends host survival. Thus, our study confirms the simultaneous targeting of MYC and BCL2 driver oncogenes through the combined use of XPO1 and BCL2 inhibitors as a unique approach for the treatment of DHLs.
Collapse
Affiliation(s)
- Yuanhui Liu
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Nancy G Azizian
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yaling Dou
- Center for Transplantation Immunology, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Lan V Pham
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yulin Li
- Center for Immunotherapy Research, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX, 77030, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
37
|
Chen L, Huang Y, Zhou L, Lian Y, Wang J, Chen D, Wei H, Huang M, Huang Y. Prognostic roles of the transcriptional expression of exportins in hepatocellular carcinoma. Biosci Rep 2019; 39:BSR20190827. [PMID: 31371628 PMCID: PMC6702357 DOI: 10.1042/bsr20190827] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022] Open
Abstract
Aims: A large number of studies have suggested that exportins (XPOs) play a pivotal role in human cancers. In the present study, we analyzed XPO mRNA expression in cancer tissues and explored their prognostic value in hepatocellular carcinoma (HCC).Methods: Transcriptional and survival data related to XPO expression in HCC patients were obtained through the ONCOMINE and UALCAN databases. Survival analysis plots were drawn with Gene Expression Profiling Interactive Analysis (GEPIA). Sequence alteration data for XPOs were obtained from The Cancer Genome Atlas (TCGA) database and c-BioPortal. Gene functional enrichment analyses were performed with Database for Annotation, Visualization and Integrated Discovery (DAVID).Results: Compared with normal liver tissues, significant XPO mRNA overexpression was observed in HCC cancer tissues. There was a trend of higher XPO expression in more advanced clinical stages and lower differentiated pathological grades of HCC. In HCC patients, high expression of XPO1, CSE1L, XPOT, XPO4/5/6 was related to poor overall survival (OS), and XPO1, CSE1L and XPO5/6 were correlated with poor disease-free survival (DFS). The main genetic alterations in XPOs involved mRNA up-regulation, DNA amplification and deletion. General XPO mutations were remarkably associated with worse OS and mostly affected the pathways of RNA transport and oocyte meiosis.Conclusion: High expression of XPOs was associated with a poor prognosis in HCC patients. XPOs may be exploited as good prognostic biomarkers for survival in HCC patients.
Collapse
Affiliation(s)
- Lubiao Chen
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanlin Huang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liang Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifan Lian
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jialiang Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dongmei Chen
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huan Wei
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mingsheng Huang
- Department of Interventional Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuehua Huang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Gruffaz M, Yuan H, Meng W, Liu H, Bae S, Kim JS, Lu C, Huang Y, Gao SJ. CRISPR-Cas9 Screening of Kaposi's Sarcoma-Associated Herpesvirus-Transformed Cells Identifies XPO1 as a Vulnerable Target of Cancer Cells. mBio 2019; 10:e00866-19. [PMID: 31088931 PMCID: PMC6520457 DOI: 10.1128/mbio.00866-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 02/05/2023] Open
Abstract
The abnormal proliferation of cancer cells is driven by deregulated oncogenes or tumor suppressors, among which the cancer-vulnerable genes are attractive therapeutic targets. Targeting mislocalization of oncogenes and tumor suppressors resulting from aberrant nuclear export is effective for inhibiting growth transformation of cancer cells. We performed a clustered regularly interspaced short palindromic repeat (CRISPR)-associated (Cas) screening in a unique model of matched primary and oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV)-transformed cells and identified genes that were growth promoting and growth suppressive for both types of cells, among which exportin XPO1 was demonstrated to be critical for the survival of transformed cells. Using XPO1 inhibitor KPT-8602 and by small interfering RNA (siRNA) knockdown, we confirmed the essential role of XPO1 in cell proliferation and growth transformation of KSHV-transformed cells and in cell lines of other cancers, including gastric cancer and liver cancer. XPO1 inhibition induced cell cycle arrest through p53 activation, but the mechanisms of p53 activation differed among the different types of cancer cells. p53 activation depended on the formation of promyelocytic leukemia (PML) nuclear bodies in gastric cancer and liver cancer cells. Mechanistically, XPO1 inhibition induced relocalization of autophagy adaptor protein p62 (SQSTM1), recruiting p53 for activation in PML nuclear bodies. Taken the data together, we have identified novel growth-promoting and growth-suppressive genes of primary and cancer cells and have demonstrated that XPO1 is a vulnerable target of cancer cells. XPO1 inhibition induces cell arrest through a novel PML- and p62-dependent mechanism of p53 activation in some types of cancer cells.IMPORTANCE Using a model of oncogenic virus KSHV-driven cellular transformation of primary cells, we have performed a genome-wide CRISPR-Cas9 screening to identify vulnerable genes of cancer cells. This screening is unique in that this virus-induced oncogenesis model does not depend on any cellular genetic alterations and has matched primary and KSHV-transformed cells, which are not available for similar screenings in other types of cancer. We have identified genes that are both growth promoting and growth suppressive in primary and transformed cells, some of which could represent novel proto-oncogenes and tumor suppressors. In particular, we have demonstrated that the exportin XPO1 is a critical factor for the survival of transformed cells. Using a XPO1 inhibitor (KPT-8602) and siRNA-mediated knockdown, we have confirmed the essential role of XPO1 in cell proliferation and in growth transformation of KSHV-transformed cells, as well as of gastric and liver cancer cells. XPO1 inhibition induces cell cycle arrest by activating p53, but the mechanisms of p53 activation differed among different types of cancer cells. p53 activation is dependent on the formation of PML nuclear bodies in gastric and liver cancer cells. Mechanistically, XPO1 inhibition induces relocalization of autophagy adaptor protein p62 (SQSTM1), recruiting p53 for activation in PML nuclear bodies. These results illustrate that XPO1 is a vulnerable target of cancer cells and reveal a novel mechanism for blocking cancer cell proliferation by XPO1 inhibition as well as a novel PML- and p62-mediated mechanism of p53 activation in some types of cancer cells.
Collapse
Affiliation(s)
- Marion Gruffaz
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wen Meng
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hui Liu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul, South Korea
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Daejon, South Korea
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Chun Lu
- Nanjing Medical University, Nanjing, China
| | - Yufei Huang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Nanjing Medical University, Nanjing, China
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, China
| |
Collapse
|
39
|
Haider T, Tiwari R, Vyas SP, Soni V. Molecular determinants as therapeutic targets in cancer chemotherapy: An update. Pharmacol Ther 2019; 200:85-109. [PMID: 31047907 DOI: 10.1016/j.pharmthera.2019.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/25/2019] [Indexed: 02/06/2023]
Abstract
It is well known that cancer cells are heterogeneous in nature and very distinct from their normal counterparts. Commonly these cancer cells possess different and complementary metabolic profile, microenvironment and adopting behaviors to generate more ATPs to fulfill the requirement of high energy that is further utilized in the production of proteins and other essentials required for cell survival, growth, and proliferation. These differences create many challenges in cancer treatments. On the contrary, such situations of metabolic differences between cancer and normal cells may be expected a promising strategy for treatment purpose. In this article, we focus on the molecular determinants of oncogene-specific sub-organelles such as potential metabolites of mitochondria (reactive oxygen species, apoptotic proteins, cytochrome c, caspase 9, caspase 3, etc.), endoplasmic reticulum (unfolded protein response, PKR-like ER kinase, C/EBP homologous protein, etc.), nucleus (nucleolar phosphoprotein, nuclear pore complex, nuclear localization signal), lysosome (microenvironment, etc.) and plasma membrane phospholipids, etc. that might be exploited for the targeted delivery of anti-cancer drugs for therapeutic benefits. This review will help to understand the various targets of subcellular organelles at molecular levels. In the future, this molecular level understanding may be combined with the genomic profile of cancer for the development of the molecularly guided or personalized therapeutics for complete eradication of cancer.
Collapse
Affiliation(s)
- Tanweer Haider
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh 470003, India
| | - Rahul Tiwari
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh 470003, India
| | - Suresh Prasad Vyas
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh 470003, India
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
40
|
Zhong T, Wu M, Ma S. Examination of Independent Prognostic Power of Gene Expressions and Histopathological Imaging Features in Cancer. Cancers (Basel) 2019; 11:E361. [PMID: 30871256 PMCID: PMC6468814 DOI: 10.3390/cancers11030361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/04/2019] [Accepted: 03/10/2019] [Indexed: 12/26/2022] Open
Abstract
Cancer prognosis is of essential interest, and extensive research has been conducted searching for biomarkers with prognostic power. Recent studies have shown that both omics profiles and histopathological imaging features have prognostic power. There are also studies exploring integrating the two types of measurements for prognosis modeling. However, there is a lack of study rigorously examining whether omics measurements have independent prognostic power conditional on histopathological imaging features, and vice versa. In this article, we adopt a rigorous statistical testing framework and test whether an individual gene expression measurement can improve prognosis modeling conditional on high-dimensional imaging features, and a parallel analysis is conducted reversing the roles of gene expressions and imaging features. In the analysis of The Cancer Genome Atlas (TCGA) lung adenocarcinoma and liver hepatocellular carcinoma data, it is found that multiple individual genes, conditional on imaging features, can lead to significant improvement in prognosis modeling; however, individual imaging features, conditional on gene expressions, only offer limited prognostic power. Being among the first to examine the independent prognostic power, this study may assist better understanding the "connectedness" between omics profiles and histopathological imaging features and provide important insights for data integration in cancer modeling.
Collapse
Affiliation(s)
- Tingyan Zhong
- SJTU-Yale Joint Center for Biostatistics, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mengyun Wu
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai 200433, China.
| | - Shuangge Ma
- Department of Biostatistics, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
41
|
Jorquera PA, Mathew C, Pickens J, Williams C, Luczo JM, Tamir S, Ghildyal R, Tripp RA. Verdinexor (KPT-335), a Selective Inhibitor of Nuclear Export, Reduces Respiratory Syncytial Virus Replication In Vitro. J Virol 2019; 93:e01684-18. [PMID: 30541831 PMCID: PMC6364025 DOI: 10.1128/jvi.01684-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/27/2018] [Indexed: 01/09/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of hospitalization of infants and young children, causing considerable respiratory disease and repeat infections that may lead to chronic respiratory conditions such as asthma, wheezing, and bronchitis. RSV causes ∼34 million new episodes of lower respiratory tract illness (LRTI) in children younger than 5 years of age, with >3 million hospitalizations due to severe RSV-associated LRTI. The standard of care is limited to symptomatic relief as there are no approved vaccines and few effective antiviral drugs; thus, a safe and efficacious RSV therapeutic is needed. Therapeutic targeting of host proteins hijacked by RSV to facilitate replication is a promising antiviral strategy as targeting the host reduces the likelihood of developing drug resistance. The nuclear export of the RSV M protein, mediated by the nuclear export protein exportin 1 (XPO1), is crucial for RSV assembly and budding. Inhibition of RSV M protein export by leptomycin B correlated with reduced RSV replication in vitro In this study, we evaluated the anti-RSV efficacy of Verdinexor (KPT-335), a small molecule designed to reversibly inhibit XPO1-mediated nuclear export. KPT-335 inhibited XPO1-mediated transport and reduced RSV replication in vitro KPT-335 was effective against RSV A and B strains and reduced viral replication following prophylactic or therapeutic administration. Inhibition of RSV replication by KPT-335 was due to a combined effect of reduced XPO1 expression, disruption of the nuclear export of RSV M protein, and inactivation of the NF-κB signaling pathway.IMPORTANCE RSV is an important cause of LRTI in infants and young children for which there are no suitable antiviral drugs offered. We evaluated the efficacy of KPT-335 as an anti-RSV drug and show that KPT-335 inhibits XPO1-mediated nuclear export, leading to nuclear accumulation of RSV M protein and reduction in RSV levels. KPT-335 treatment also resulted in inhibition of proinflammatory pathways, which has important implications for its effectiveness in vivo.
Collapse
Affiliation(s)
- Patricia A Jorquera
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Cynthia Mathew
- Respiratory Virology Group, Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, Australia
| | - Jennifer Pickens
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Colin Williams
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Jasmina M Luczo
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Sharon Tamir
- Karyopharm Therapeutics, Inc., Newton, Massachusetts, USA
| | - Reena Ghildyal
- Respiratory Virology Group, Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, Australia
| | - Ralph A Tripp
- Animal Health Research Center, Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
42
|
Yan D, Pomicter AD, Tantravahi S, Mason CC, Senina AV, Ahmann JM, Wang Q, Than H, Patel AB, Heaton WL, Eiring AM, Clair PM, Gantz KC, Redwine HM, Swierczek SI, Halverson BJ, Baloglu E, Shacham S, Khorashad JS, Kelley TW, Salama ME, Miles RR, Boucher KM, Prchal JT, O'Hare T, Deininger MW. Nuclear-Cytoplasmic Transport Is a Therapeutic Target in Myelofibrosis. Clin Cancer Res 2018; 25:2323-2335. [PMID: 30563936 DOI: 10.1158/1078-0432.ccr-18-0959] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/31/2018] [Accepted: 12/14/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE Myelofibrosis is a hematopoietic stem cell neoplasm characterized by bone marrow reticulin fibrosis, extramedullary hematopoiesis, and frequent transformation to acute myeloid leukemia. Constitutive activation of JAK/STAT signaling through mutations in JAK2, CALR, or MPL is central to myelofibrosis pathogenesis. JAK inhibitors such as ruxolitinib reduce symptoms and improve quality of life, but are not curative and do not prevent leukemic transformation, defining a need to identify better therapeutic targets in myelofibrosis. EXPERIMENTAL DESIGN A short hairpin RNA library screening was performed on JAK2V617F-mutant HEL cells. Nuclear-cytoplasmic transport (NCT) genes including RAN and RANBP2 were among top candidates. JAK2V617F-mutant cell lines, human primary myelofibrosis CD34+ cells, and a retroviral JAK2V617F-driven myeloproliferative neoplasms mouse model were used to determine the effects of inhibiting NCT with selective inhibitors of nuclear export compounds KPT-330 (selinexor) or KPT-8602 (eltanexor). RESULTS JAK2V617F-mutant HEL, SET-2, and HEL cells resistant to JAK inhibition are exquisitely sensitive to RAN knockdown or pharmacologic inhibition by KPT-330 or KPT-8602. Inhibition of NCT selectively decreased viable cells and colony formation by myelofibrosis compared with cord blood CD34+ cells and enhanced ruxolitinib-mediated growth inhibition and apoptosis, both in newly diagnosed and ruxolitinib-exposed myelofibrosis cells. Inhibition of NCT in myelofibrosis CD34+ cells led to nuclear accumulation of p53. KPT-330 in combination with ruxolitinib-normalized white blood cells, hematocrit, spleen size, and architecture, and selectively reduced JAK2V617F-mutant cells in vivo. CONCLUSIONS Our data implicate NCT as a potential therapeutic target in myelofibrosis and provide a rationale for clinical evaluation in ruxolitinib-exposed patients with myelofibrosis.
Collapse
Affiliation(s)
- Dongqing Yan
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | | | - Srinivas Tantravahi
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - Clinton C Mason
- Department of Pediatrics, The University of Utah, Salt Lake City, Utah
| | - Anna V Senina
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Jonathan M Ahmann
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Qiang Wang
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Department of Hematology, Nanfang Hospital, Southern Medical University
| | - Hein Than
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Department of Haematology, Singapore General Hospital, Singapore
| | - Ami B Patel
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - William L Heaton
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Anna M Eiring
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Phillip M Clair
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Kevin C Gantz
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Hannah M Redwine
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Sabina I Swierczek
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | | | | | | | - Jamshid S Khorashad
- Department of Cellular Pathology, Hammersmith Hospital, Imperial College Health Care NHS Trust, London, United Kingdom
| | - Todd W Kelley
- Department of Pathology, The University of Utah, Salt Lake City, Utah
| | - Mohamed E Salama
- Department of Pathology, The University of Utah, Salt Lake City, Utah
| | - Rodney R Miles
- Department of Pathology, The University of Utah, Salt Lake City, Utah
| | - Kenneth M Boucher
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Josef T Prchal
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - Thomas O'Hare
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - Michael W Deininger
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah. .,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
43
|
Chien W, Sudo M, Ding LW, Sun QY, Wuensche P, Lee KL, Hattori N, Garg M, Xu L, Zheng Y, Gery S, Wongphayak S, Yang H, Baloglu E, Shacham S, Kauffman M, Mori S, Koeffler HP. Functional Genome-wide Screening Identifies Targets and Pathways Sensitizing Pancreatic Cancer Cells to Dasatinib. J Cancer 2018; 9:4762-4773. [PMID: 30588262 PMCID: PMC6299388 DOI: 10.7150/jca.25138] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023] Open
Abstract
This study is an unbiased genomic screen to obtain functional targets for increased effectiveness of dasatinib in pancreatic cancer. Dasatinib, a multi-targeted tyrosine kinase inhibitor, is used in clinical trials for treatment of pancreatic cancer; however, intrinsic and acquired resistance often occurs. We used a dasatinib-resistant pancreatic cancer cell line SU8686 to screen for synthetic lethality that synergizes with dasatinib using a pooled human shRNA library followed by next generation sequencing. Novel genes were identified which when silenced produced a prominent inhibitory effect with dasatinib against the pancreatic cancer cells. Several of these genes are involved in the regulation of epigenetics, as well as signaling pathways of the FOXO and hedgehog families. Small molecule inhibitors of either histone deacetylases or nuclear exporter had marked inhibitory effect with dasatinib in pancreatic cancers, suggesting their potential therapeutic effectiveness in this deadly cancer.
Collapse
Affiliation(s)
- Wenwen Chien
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Hematology-Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Makoto Sudo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ling-Wen Ding
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Qiao-Yang Sun
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Peer Wuensche
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kian Leong Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Norimichi Hattori
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Manoj Garg
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Liang Xu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Yun Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sigal Gery
- Department of Hematology-Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Sarawut Wongphayak
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | | | | | - Seiichi Mori
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Hematology-Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,National University Cancer Institute, National University Hospital, Singapore
| |
Collapse
|
44
|
Subhash VV, Yeo MS, Wang L, Tan SH, Wong FY, Thuya WL, Tan WL, Peethala PC, Soe MY, Tan DSP, Padmanabhan N, Baloglu E, Shacham S, Tan P, Koeffler HP, Yong WP. Anti-tumor efficacy of Selinexor (KPT-330) in gastric cancer is dependent on nuclear accumulation of p53 tumor suppressor. Sci Rep 2018; 8:12248. [PMID: 30115935 PMCID: PMC6095850 DOI: 10.1038/s41598-018-30686-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/06/2018] [Indexed: 12/17/2022] Open
Abstract
Exportin-1 (XPO1) controls the nucleo-cytoplasmic trafficking of several key growth regulatory and tumor suppressor proteins. Nuclear export blockade through XPO1 inhibition is a target for therapeutic inhibition in many cancers. Studies have suggested XPO1 upregulation as an indicator of poor prognosis in gastric cancer. In the current study, we investigated the anti-tumor efficacy of selective inhibitors of nuclear export (SINE) compounds KPT-185, KTP-276 and clinical stage selinexor (KPT-330) in gastric cancer. XPO1 was found to be overexpressed in gastric cancer as compared to adjacent normal tissues and was correlated with poor survival outcomes. Among the 3 SINE compounds, in vitro targeting of XPO1 with selinexor resulted in greatest potency with significant anti-proliferative effects at nano molar concentrations. XPO1 inhibition by selinexor resulted in nuclear accumulation of p53, causing cell cycle arrest and apoptosis. Also, inhibition of XPO1 lead to the cytoplasmic retention of p21 and suppression of survivin. Orally administered selienxor caused significant inhibition of tumor growth in xenograft models of gastric cancer. Furthermore, combination of selinexor with irinotecan exhibited greater anti-tumor effect compared to individual treatment. Taken together, our study underscores the therapeutic utility of XPO1 targeting in gastric cancer and suggests the potential benefits of XPO1 inhibition in-combination with chemotherapy.
Collapse
Affiliation(s)
- Vinod Vijay Subhash
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Mei Shi Yeo
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shi Hui Tan
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Foong Ying Wong
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Win Lwin Thuya
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Woei Loon Tan
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Praveen C Peethala
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mu Yar Soe
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - David S P Tan
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Nisha Padmanabhan
- Department of Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Erkan Baloglu
- Karyopharm Therapeutics Inc, Newton, Massachusetts, USA
| | | | - Patrick Tan
- Department of Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, Yong loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Haematology-Oncology, National University Hospital, Singapore, Singapore.
| |
Collapse
|
45
|
Yue L, Sun ZN, Yao YS, Shen Z, Wang HB, Liu XP, Zhou F, Xiang JY, Yao RY, Niu HT. CRM1, a novel independent prognostic factor overexpressed in invasive breast carcinoma of poor prognosis. Oncol Lett 2018; 15:7515-7522. [PMID: 29725458 PMCID: PMC5920404 DOI: 10.3892/ol.2018.8316] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/21/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer in females globally and is more aggressive at later stages. Chromosome region maintenance 1 (CRM1) is involved in the nuclear export of proteins and RNAs and has been associated with a number of malignancies. However, the clinicopathological significance of its expression in BC remains to be elucidated therefore this was investigated in the present study. CRM1 expression in 280 breast cancer tissues and 60 normal tissues was retrospectively analyzed using immunohistochemistry (IHC) and western blotting. IHC investigation demonstrated that CRM1 expression was significantly increased in BC compared with the normal breast epithelium (P<0.0001). Overexpression of CRM1 was markedly associated with poor prognostic characteristics, including larger tumor size (P=0.024), positive lymph node metastasis (P=0.032), invasive histological type (P=0.004) and distant metastasis (P=0.026). Significant associations were also observed between increased CRM1 expression and the progesterone receptor (P=0.028) and Ki67 (P=0.019). Kaplan-Meier survival analysis demonstrated that patients with high CRM1 expression exhibited a reduced disease-free survival and overall survival compared with those with low CRM1 expression (P=0.013). In the multivariate analysis, CRM1 expression (P=0.011), tumor size (P=0.001) and lymph node metastasis (P<0.001) were independent prognostic markers of BC. In conclusion, CRM1 serves an important role in BC and may serve as a predictive and prognostic factor for a poor outcome in patients with BC.
Collapse
Affiliation(s)
- Lu Yue
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhen-Ni Sun
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ya-Sai Yao
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zan Shen
- Department of Oncology, The Sixth People's Hospital, Medical College of Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Hai-Bo Wang
- Department of Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiang-Ping Liu
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Fang Zhou
- Department of Radiotherapy, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jin-Yu Xiang
- Department of Oncology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Ru-Yong Yao
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Hai-Tao Niu
- Department of Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
46
|
Beck M, Schirmacher P, Singer S. Alterations of the nuclear transport system in hepatocellular carcinoma - New basis for therapeutic strategies. J Hepatol 2017; 67:1051-1061. [PMID: 28673770 DOI: 10.1016/j.jhep.2017.06.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is among the most prevalent human malignancies worldwide with rising incidence in industrialised countries, few therapeutic options and poor prognosis. To expand and improve therapeutic strategies, identification of drug targets involved in several liver cancer-related pathways is crucial. Virtually all signal transduction cascades cross the nuclear envelope and therefore require components of the nuclear transport system (NTS), including nuclear transport receptors (e.g. importins and exportins) and the nuclear pore complex. Accordingly, members of the NTS represent promising targets for therapeutic intervention. Selective inhibitors of nuclear export have already entered clinical trials for various malignancies. Herein, we review the current knowledge regarding alterations of the NTS and their potential for targeted therapy in HCC.
Collapse
Affiliation(s)
- Martin Beck
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Stephan Singer
- European Molecular Biology Laboratory, Heidelberg, Germany; Institute of Pathology, University Hospital Heidelberg, Germany.
| |
Collapse
|
47
|
Garg M, Kanojia D, Mayakonda A, Ganesan TS, Sadhanandhan B, Suresh S, S S, Nagare RP, Said JW, Doan NB, Ding LW, Baloglu E, Shacham S, Kauffman M, Koeffler HP. Selinexor (KPT-330) has antitumor activity against anaplastic thyroid carcinoma in vitro and in vivo and enhances sensitivity to doxorubicin. Sci Rep 2017; 7:9749. [PMID: 28852098 PMCID: PMC5575339 DOI: 10.1038/s41598-017-10325-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is one of the most lethal malignancies having no effective treatment. Exportin-1 (XPO1) is the key mediator of nuclear export of many tumor suppressor proteins and is overexpressed in human cancers. In this study, we examined the therapeutic potential of selinexor (XPO1 inhibitor) against human ATC cells both in vitro and in vivo. Here, we showed that XPO1 is robustly expressed in primary ATC samples and human ATC cell lines. Silencing of XPO1 by either shRNA or selinexor significantly reduced cellular growth and induced cell cycle arrest, apoptosis of ATC cells by altering the protein expression of cancer-related genes. Moreover, selinexor significantly inhibited tumor growth of ATC xenografts. Microarray analysis showed enrichment of DNA replication, cell cycle, cell cycle checkpoint and TNF pathways in selinexor treated ATC cells. Importantly, selinexor decreased AXL and GAS6 levels in CAL62 and HTH83 cells and suppressed the phosphorylation of downstream targets of AXL signaling such as AKT and P70S6K. Finally, a combination of selinexor with doxorubicin demonstrated a synergistic decrease in the cellular proliferation of several ATC cells. These results provide a rationale for investigating the efficacy of combining selinexor and doxorubicin therapy to improve the outcome of ATC patients.
Collapse
Affiliation(s)
- Manoj Garg
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore, Singapore.
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar, Chennai, India.
| | - Deepika Kanojia
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore, Singapore
| | - Anand Mayakonda
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore, Singapore
| | - Trivadi S Ganesan
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar, Chennai, India
| | - Bindhya Sadhanandhan
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar, Chennai, India
| | - Sidhanth Suresh
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar, Chennai, India
| | - Sneha S
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar, Chennai, India
| | - Rohit P Nagare
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar, Chennai, India
| | - Jonathan W Said
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ngan B Doan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ling-Wen Ding
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore, Singapore
| | | | | | | | - H Phillip Koeffler
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California Los Angeles, School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
48
|
Arango NP, Yuca E, Zhao M, Evans KW, Scott S, Kim C, Gonzalez-Angulo AM, Janku F, Ueno NT, Tripathy D, Akcakanat A, Naing A, Meric-Bernstam F. Selinexor (KPT-330) demonstrates anti-tumor efficacy in preclinical models of triple-negative breast cancer. Breast Cancer Res 2017; 19:93. [PMID: 28810913 PMCID: PMC5557476 DOI: 10.1186/s13058-017-0878-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/07/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Selinexor (KPT-330) is an oral agent that has been shown to inhibit the nuclear exporter XPO1. Given the pressing need for novel therapies for triple-negative breast cancer (TNBC), we sought to determine the antitumor effects of selinexor in vitro and in vivo. METHODS Twenty-six breast cancer cell lines of different breast cancer subtypes were treated with selinexor in vitro. Cell proliferation assays were used to measure the half-maximal inhibitory concentration (IC50) and to test the effects in combination with chemotherapy. In vivo efficacy was tested both as a single agent and in combination therapy in TNBC patient-derived xenografts (PDXs). RESULTS Selinexor demonstrated growth inhibition in all 14 TNBC cell lines tested; TNBC cell lines were more sensitive to selinexor (median IC50 44 nM, range 11 to 550 nM) than were estrogen receptor (ER)-positive breast cancer cell lines (median IC50 > 1000 nM, range 40 to >1000 nM; P = 0.017). In multiple TNBC cell lines, selinexor was synergistic with paclitaxel, carboplatin, eribulin, and doxorubicin in vitro. Selinexor as a single agent reduced tumor growth in vivo in four of five different TNBC PDX models, with a median tumor growth inhibition ratio (T/C: treatment/control) of 42% (range 31 to 73%) and demonstrated greater antitumor efficacy in combination with paclitaxel or eribulin (average T/C ratios of 27% and 12%, respectively). CONCLUSIONS Collectively, these findings strongly suggest that selinexor is a promising therapeutic agent for TNBC as a single agent and in combination with standard chemotherapy.
Collapse
Affiliation(s)
- Natalia Paez Arango
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA
| | - Erkan Yuca
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Ming Zhao
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Kurt W Evans
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Stephen Scott
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Charissa Kim
- Department of Genetics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Ana Maria Gonzalez-Angulo
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Argun Akcakanat
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA
| | - Funda Meric-Bernstam
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA. .,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 455, Houston, TX, 77030, USA. .,The Sheikh Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, 1400 Pressler Boulevard, Unit 455, Houston, TX, 77030, USA. .,Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Boulevard, Unit 455, Houston, TX, 77030, USA.
| |
Collapse
|
49
|
Azmi AS, Li Y, Muqbil I, Aboukameel A, Senapedis W, Baloglu E, Landesman Y, Shacham S, Kauffman MG, Philip PA, Mohammad RM. Exportin 1 (XPO1) inhibition leads to restoration of tumor suppressor miR-145 and consequent suppression of pancreatic cancer cell proliferation and migration. Oncotarget 2017; 8:82144-82155. [PMID: 29137251 PMCID: PMC5669877 DOI: 10.18632/oncotarget.19285] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/18/2017] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer related deaths in the United States with a majority of these patients dying from aggressively invasive and metastatic disease. There is growing evidence that suggests an important role for microRNAs (miRNAs) in the pathobiology of aggressive PDAC. In this study, we found that the expression of miR-145 was significantly lower in PDAC cells when compared to normal pancreatic duct epithelial cells. Here we show that inhibition of the nuclear exporter protein exportin 1 (XPO1; also known as chromosome maintenance region 1 [CRM1]) by siRNA knockdown or by the Selective Inhibitor of Nuclear Export (SINE) compound (KPT-330; selinexor) increases miR-145 expression in PDAC cells resulting in the decreased cell proliferation and migration capacities. A similar result was obtained with forced expression of miR-145 in PDAC cells. To this end, SINE compound treatment mediated the down-regulation of known miR-145 targets genes including EGFR, MMP1, MT-MMP, c-Myc, Pak4 and Sox-2. In addition, selinexor induced the expression of two important tumor suppressive miRNAs miR-34c and let-7d leading to the up-regulation of p21WAF1. These results are the first to report that targeted inhibition of the nuclear export machinery could restore tumor suppressive miRNAs in PDAC that warrants further clinical investigations.
Collapse
Affiliation(s)
- Asfar S Azmi
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yiwei Li
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Irfana Muqbil
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Amro Aboukameel
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Erkan Baloglu
- Karyopharm Therapeutics Inc., Newton Centre, MA, USA
| | | | | | | | - Philip A Philip
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramzi M Mohammad
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
50
|
Mathew C, Ghildyal R. CRM1 Inhibitors for Antiviral Therapy. Front Microbiol 2017; 8:1171. [PMID: 28702009 PMCID: PMC5487384 DOI: 10.3389/fmicb.2017.01171] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/08/2017] [Indexed: 12/22/2022] Open
Abstract
Infectious diseases are a major global concern and despite major advancements in medical research, still cause significant morbidity and mortality. Progress in antiviral therapy is particularly hindered by appearance of mutants capable of overcoming the effects of drugs targeting viral components. Alternatively, development of drugs targeting host proteins essential for completion of viral lifecycle holds potential as a viable strategy for antiviral therapy. Nucleocytoplasmic trafficking pathways in particular are involved in several pathological conditions including cancer and viral infections, where hijacking or alteration of function of key transporter proteins, such as Chromosome Region Maintenance1 (CRM1) is observed. Overexpression of CRM1-mediated nuclear export is evident in several solid and hematological malignancies. Interestingly, CRM1-mediated nuclear export of viral components is crucial in various stages of the viral lifecycle and assembly. This review summarizes the role of CRM1 in cancer and selected viruses. Leptomycin B (LMB) is the prototypical inhibitor of CRM1 potent against various cancer cell lines overexpressing CRM1 and in limiting viral infections at nanomolar concentrations in vitro. However, the irreversible shutdown of nuclear export results in high cytotoxicity and limited efficacy in vivo. This has prompted search for synthetic and natural CRM1 inhibitors that can potentially be developed as broadly active antivirals, some of which are summarized in this review.
Collapse
Affiliation(s)
| | - Reena Ghildyal
- Respiratory Virology Group, Centre for Research in Therapeutic Solutions, Health Research Institute, University of CanberraCanberra, ACT, Australia
| |
Collapse
|