151
|
Gravina GL, Mancini A, Colapietro A, Marampon F, Sferra R, Pompili S, Biordi LA, Iorio R, Flati V, Argueta C, Landesman Y, Kauffman M, Shacham S, Festuccia C. Pharmacological treatment with inhibitors of nuclear export enhances the antitumor activity of docetaxel in human prostate cancer. Oncotarget 2017; 8:111225-111245. [PMID: 29340049 PMCID: PMC5762317 DOI: 10.18632/oncotarget.22760] [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: 06/29/2017] [Accepted: 11/13/2017] [Indexed: 01/08/2023] Open
Abstract
Background and aims Docetaxel (DTX) modestly increases patient survival of metastatic castration-resistant prostate cancer (mCRPC) due to insurgence of pharmacological resistance. Deregulation of Chromosome Region Maintenance (CRM-1)/ exportin-1 (XPO-1)-mediated nuclear export may play a crucial role in this phenomenon. Material and methods Here, we evaluated the effects of two Selective Inhibitor of Nuclear Export (SINE) compounds, selinexor (KPT-330) and KPT-251, in association with DTX by using 22rv1, PC3 and DU145 cell lines with their. DTX resistant derivatives. Results and conclusions We show that DTX resistance may involve overexpression of β-III tubulin (TUBB3) and P-glycoprotein as well as increased cytoplasmic accumulation of Foxo3a. Increased levels of XPO-1 were also observed in DTX resistant cells suggesting that SINE compounds may modulate DTX effectiveness in sensitive cells as well as restore the sensitivity to DTX in resistant ones. Pretreatment with SINE compounds, indeed, sensitized to DTX through increased tumor shrinkage and apoptosis by preventing DTX-induced cell cycle arrest. Basally SINE compounds induce FOXO3a activation and nuclear accumulation increasing the expression of FOXO-responsive genes including p21, p27 and Bim causing cell cycle arrest. SINE compounds-catenin and survivin supporting apoptosis. βdown-regulated Cyclin D1, c-myc, Nuclear sequestration of p-Foxo3a was able to reduce ABCB1 and TUBB3 H2AX levels, prolonged γ expression. Selinexor treatment increased DTX-mediated double strand breaks (DSB), and reduced the levels of DNA repairing proteins including DNA PKc and Topo2A. Our results provide supportive evidence for the therapeutic use of SINE compounds in combination with DTX suggesting their clinical use in mCRPC patients.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy, University of L'Aquila, L'Aquila, Italy
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Roberta Sferra
- Department of Biotechnological and Applied Clinical Sciences, Division of Human Anatomy, University of L'Aquila, L'Aquila, Italy
| | - Simona Pompili
- Department of Biotechnological and Applied Clinical Sciences, Division of Human Anatomy, University of L'Aquila, L'Aquila, Italy
| | - Leda Assunta Biordi
- Department of Biotechnological and Applied Clinical Sciences, Division of Molecular Pathology, University of L'Aquila, L'Aquila, Italy
| | - Roberto Iorio
- Department of Biotechnological and Applied Clinical Sciences, Division of Applied Biology, University of L'Aquila, L'Aquila, Italy
| | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, Division of Molecular Pathology, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
152
|
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: 4.4] [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
|
153
|
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: 5.4] [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
|
154
|
Tan Y, Li QM, Huang N, Cheng S, Zhao GJ, Chen H, Chen S, Tang ZH, Zhang WQ, Huang Q, Cheng Y. Upregulation of DACT2 suppresses proliferation and enhances apoptosis of glioma cell via inactivation of YAP signaling pathway. Cell Death Dis 2017; 8:e2981. [PMID: 28796248 PMCID: PMC5596571 DOI: 10.1038/cddis.2017.385] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/02/2017] [Accepted: 07/07/2017] [Indexed: 01/01/2023]
Abstract
DACT2, one of the Dact gene family members, was shown to function as a tumor suppressor. However, its function in gliomas remains largely unknown. In this study, we investigated the role of DACT2, underlying molecular mechanisms and its clinical significance in glioma patients. Downexpression of DACT2 in gliomas compared with adjacent normal brain tissues was correlated with glioma grade and poor survival. Cox regression analysis revealed that the DACT2 is an independent prognostic indicator for glioma patients. Overexpression of DACT2 in glioma cells inhibited proliferation, cell cycle and enhanced apoptosis, sensitivity to temozolomide in vitro and suppressed tumor growth in vivo. Whereas knockdown of DACT2 induce opposite reaction. Mechanistically, overexpression of DACT2 resulted in upregulation of important signaling molecules such as p-YAP and p-β-catenin, and prevent YAP translocating into nucleus and sequestering in the cytoplasm to degrade. The study further proved that DACT2 can suppress YAP through Wnt/β-catenin signaling pathway. Collectively, these data indicate that DACT2 has a tumor suppressor function via inactivation of YAP pathway, providing a promising target for the treatment of gliomas.
Collapse
Affiliation(s)
- Ying Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiu-Meng Li
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si Cheng
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guan-Jian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhao-Hua Tang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen-Qian Zhang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
155
|
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: 7.3] [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
|
156
|
Brun S, Abella N, Berciano MT, Tapia O, Jaumot M, Freire R, Lafarga M, Agell N. SUMO regulates p21Cip1 intracellular distribution and with p21Cip1 facilitates multiprotein complex formation in the nucleolus upon DNA damage. PLoS One 2017; 12:e0178925. [PMID: 28582471 PMCID: PMC5459497 DOI: 10.1371/journal.pone.0178925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/22/2017] [Indexed: 01/06/2023] Open
Abstract
We previously showed that p21Cip1 transits through the nucleolus on its way from the nucleus to the cytoplasm and that DNA damage inhibits this transit and induces the formation of p21Cip1-containing intranucleolar bodies (INoBs). Here, we demonstrate that these INoBs also contain SUMO-1 and UBC9, the E2 SUMO-conjugating enzyme. Furthermore, whereas wild type SUMO-1 localized in INoBs, a SUMO-1 mutant, which is unable to conjugate with proteins, does not, suggesting the presence of SUMOylated proteins at INoBs. Moreover, depletion of the SUMO-conjugating enzyme UBC9 or the sumo hydrolase SENP2 changed p21Cip1 intracellular distribution. In addition to SUMO-1 and p21Cip1, cell cycle regulators and DNA damage checkpoint proteins, including Cdk2, Cyclin E, PCNA, p53 and Mdm2, and PML were also detected in INoBs. Importantly, depletion of UBC9 or p21Cip1 impacted INoB biogenesis and the nucleolar accumulation of the cell cycle regulators and DNA damage checkpoint proteins following DNA damage. The impact of p21Cip1 and SUMO-1 on the accumulation of proteins in INoBs extends also to CRM1, a nuclear exportin that is also important for protein translocation from the cytoplasm to the nucleolus. Thus, SUMO and p21Cip1 regulate the transit of proteins through the nucleolus, and that disruption of nucleolar export by DNA damage induces SUMO and p21Cip1 to act as hub proteins to form a multiprotein complex in the nucleolus.
Collapse
Affiliation(s)
- Sonia Brun
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Neus Abella
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Maria T. Berciano
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Olga Tapia
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Montserrat Jaumot
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Tenerife, Spain
| | - Miguel Lafarga
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Neus Agell
- Departament Biomedicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| |
Collapse
|
157
|
Yuan H, Zhang S, Zhao B, Weng Y, Zhu X, Li S, Zhang L, Zhang Y. Enzymatic Reactor with Trypsin Immobilized on Graphene Oxide Modified Polymer Microspheres To Achieve Automated Proteome Quantification. Anal Chem 2017; 89:6324-6329. [DOI: 10.1021/acs.analchem.7b00682] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Huiming Yuan
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Shen Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yejing Weng
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xudong Zhu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Senwu Li
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yukui Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R & A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| |
Collapse
|
158
|
Selective inhibition of nuclear export with selinexor in patients with non-Hodgkin lymphoma. Blood 2017; 129:3175-3183. [PMID: 28468797 DOI: 10.1182/blood-2016-11-750174] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 04/26/2017] [Indexed: 12/19/2022] Open
Abstract
Patients with relapsed or refractory (R/R) non-Hodgkin lymphoma (NHL) have a poor prognosis and limited treatment options. We evaluated selinexor, an orally bioavailable, first-in-class inhibitor of the nuclear export protein XPO1, in this phase 1 trial to assess safety and determine a recommended phase 2 dose (RP2D). Seventy-nine patients with various NHL histologies, including diffuse large B-cell lymphoma, Richter's transformation, mantle cell lymphoma, follicular lymphoma, and chronic lymphocytic leukemia, were enrolled. In the dose-escalation phase, patients received 3 to 80 mg/m2 of selinexor in 3- or 4-week cycles and were assessed for toxicities, pharmacokinetics, and antitumor activity. In the dose-expansion phase, patients were treated with selinexor at 35 or 60 mg/m2 The most common grade 3 to 4 drug-related adverse events were thrombocytopenia (47%), neutropenia (32%), anemia (27%), leukopenia (16%), fatigue (11%), and hyponatremia (10%). Tumor biopsies showed decreases in cell-signaling pathways (Bcl-2, Bcl-6, c-Myc), reduced proliferation (Ki67), nuclear localization of XPO1 cargos (p53, PTEN), and increased apoptosis after treatment. Twenty-two (31%) of the 70 evaluable patients had an objective responses, including 4 complete responses and 18 partial responses, which were observed across a spectrum of NHL subtypes. A dose of 35 mg/m2 (60 mg) was identified as the RP2D. These findings suggest that inhibition of XPO1 with oral selinexor at 35 mg/m2 is a safe therapy with encouraging and durable anticancer activity in patients with R/R NHL. The trial was registered at www.clinicaltrials.gov as #NCT01607892.
Collapse
|
159
|
Tammam SN, Azzazy HM, Lamprecht A. The effect of nanoparticle size and NLS density on nuclear targeting in cancer and normal cells; impaired nuclear import and aberrant nanoparticle intracellular trafficking in glioma. J Control Release 2017; 253:30-36. [DOI: 10.1016/j.jconrel.2017.02.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/26/2017] [Indexed: 10/20/2022]
|
160
|
Pauty J, Couturier AM, Rodrigue A, Caron MC, Coulombe Y, Dellaire G, Masson JY. Cancer-causing mutations in the tumor suppressor PALB2 reveal a novel cancer mechanism using a hidden nuclear export signal in the WD40 repeat motif. Nucleic Acids Res 2017; 45:2644-2657. [PMID: 28158555 PMCID: PMC5389658 DOI: 10.1093/nar/gkx011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 12/31/2016] [Accepted: 01/24/2017] [Indexed: 12/22/2022] Open
Abstract
One typical mechanism to promote genomic instability, a hallmark of cancer, is to inactivate tumor suppressors, such as PALB2. It has recently been reported that mutations in PALB2 increase the risk of breast cancer by 8-9-fold by age 40 and the life time risk is ∼3-4-fold. To date, predicting the functional consequences of PALB2 mutations has been challenging as they lead to different cancer risks. Here, we performed a structure-function analysis of PALB2, using PALB2 truncated mutants (R170fs, L531fs, Q775X and W1038X), and uncovered a new mechanism by which cancer cells could drive genomic instability. Remarkably, the PALB2 W1038X mutant, harboring a mutation in its C-terminal domain, is still proficient in stimulating RAD51-mediated recombination in vitro, although it is unusually localized to the cytoplasm. After further investigation, we identified a hidden NES within the WD40 domain of PALB2 and found that the W1038X truncation leads to the exposure of this NES to CRM1, an export protein. This concept was also confirmed with another WD40-containing protein, RBBP4. Consequently, our studies reveal an unreported mechanism linking the nucleocytoplasmic translocation of PALB2 mutants to cancer formation.
Collapse
Affiliation(s)
- Joris Pauty
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology; Laval University Cancer Research Center, Laval University, Québec City, QC G1V 0A6, Canada
| | - Anthony M. Couturier
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology; Laval University Cancer Research Center, Laval University, Québec City, QC G1V 0A6, Canada
| | - Amélie Rodrigue
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology; Laval University Cancer Research Center, Laval University, Québec City, QC G1V 0A6, Canada
| | - Marie-Christine Caron
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology; Laval University Cancer Research Center, Laval University, Québec City, QC G1V 0A6, Canada
| | - Yan Coulombe
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology; Laval University Cancer Research Center, Laval University, Québec City, QC G1V 0A6, Canada
| | - Graham Dellaire
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Québec Research Center, HDQ Pavilion, Oncology Axis, 9 McMahon, Québec City, QC G1R 2J6, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology; Laval University Cancer Research Center, Laval University, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
161
|
Farren MR, Hennessey RC, Shakya R, Elnaggar O, Young G, Kendra K, Landesman Y, Elloul S, Crochiere M, Klebanov B, Kashyap T, Burd CE, Lesinski GB. The Exportin-1 Inhibitor Selinexor Exerts Superior Antitumor Activity when Combined with T-Cell Checkpoint Inhibitors. Mol Cancer Ther 2017; 16:417-427. [PMID: 28148715 PMCID: PMC5407496 DOI: 10.1158/1535-7163.mct-16-0498] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/02/2016] [Accepted: 12/20/2016] [Indexed: 01/26/2023]
Abstract
Selinexor, a selective inhibitor of nuclear export (SINE) compound targeting exportin-1, has previously been shown to inhibit melanoma cell growth in vivo We hypothesized that combining selinexor with antibodies that block or disrupt T-cell checkpoint molecule signaling would exert superior antimelanoma activity. In vitro, selinexor increased PDCD1 and CTLA4 gene expression in leukocytes and induced CD274 gene expression in human melanoma cell lines. Mice bearing syngeneic B16F10 melanoma tumors demonstrated a significant reduction in tumor growth rate in response to the combination of selinexor and anti-PD-1 or anti-PD-L1 antibodies (P < 0.05). Similar results were obtained in B16F10-bearing mice treated with selinexor combined with anti-CTLA4 antibody. Immunophenotypic analysis of splenocytes by flow cytometry revealed that selinexor alone or in combination with anti-PD-L1 antibody significantly increased the frequency of both natural killer cells (P ≤ 0.050) and CD4+ T cells with a Th1 phenotype (P ≤ 0.050). Further experiments indicated that the antitumor effect of selinexor in combination with anti-PD-1 therapy persisted under an alternative dosing schedule but was lost when selinexor was administered daily. These data indicate that the efficacy of selinexor against melanoma may be enhanced by disrupting immune checkpoint activity. Mol Cancer Ther; 16(3); 417-27. ©2017 AACRSee related article by Tyler et al., p. 428.
Collapse
Affiliation(s)
- Matthew R Farren
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Rebecca C Hennessey
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio
| | - Reena Shakya
- Target Validation Shared Resource, The Ohio State University, Columbus, Ohio
| | - Omar Elnaggar
- Division of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Gregory Young
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Kari Kendra
- Division of Internal Medicine, The Ohio State University, Columbus, Ohio
| | | | | | | | | | | | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia.
| |
Collapse
|
162
|
Garg M, Kanojia D, Mayakonda A, Said JW, Doan NB, Chien W, Ganesan TS, Huey LSC, Venkatachalam N, Baloglu E, Shacham S, Kauffman M, Koeffler HP. Molecular mechanism and therapeutic implications of selinexor (KPT-330) in liposarcoma. Oncotarget 2017; 8:7521-7532. [PMID: 27893412 PMCID: PMC5352339 DOI: 10.18632/oncotarget.13485] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023] Open
Abstract
Exportin-1 mediates nuclear export of multiple tumor suppressor and growth regulatory proteins. Aberrant expression of exportin-1 is noted in human malignancies, resulting in cytoplasmic mislocalization of its target proteins. We investigated the efficacy of selinexor against liposarcoma cells both in vitro and in vivo. Exportin-1 was highly expressed in liposarcoma samples and cell lines as determined by immunohistochemistry, western blot, and immunofluorescence assay. Knockdown of endogenous exportin-1 inhibited proliferation of liposarcoma cells. Selinexor also significantly decreased cell proliferation as well as induced cell cycle arrest and apoptosis of liposarcoma cells. The drug also significantly decreased tumor volumes and weights of liposarcoma xenografts. Importantly, selinexor inhibited insulin-like growth factor 1 (IGF1) activation of IGF-1R/AKT pathway through upregulation of insulin-like growth factor binding protein 5 (IGFBP5). Further, overexpression and knockdown experiments showed that IGFBP5 acts as a tumor suppressor and its expression was restored upon selinexor treatment of liposarcoma cells. Selinexor decreased aurora kinase A and B levels in these cells and inhibitors of these kinases suppressed the growth of the liposarcoma cells. Overall, our study showed that selinexor treatment restored tumor suppressive function of IGFBP5 and inhibited aurora kinase A and B in liposarcoma cells supporting the usefulness of selinexor as a potential therapeutic strategy for the treatment of this cancer.
Collapse
Affiliation(s)
- Manoj Garg
- Cancer Science Institute (CSI) of Singapore, National University of 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
| | - Anand Mayakonda
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore
| | - 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
| | - Wenwen Chien
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore
| | - Trivadi S Ganesan
- Department of Medical Oncology and Clinical Research, Cancer Institute (WIA), Adyar Chennai, India
| | | | | | | | | | | | - H. Phillip Koeffler
- Cancer Science Institute (CSI) of Singapore, National University of Singapore, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California Los Angeles, School of Medicine, Los Angeles, CA, USA
- National University Cancer Institute, National University Hospital, Singapore, Singapore
| |
Collapse
|
163
|
Wang S, Song Y, Yan F, Liu D. Mechanisms of resistance to third-generation EGFR tyrosine kinase inhibitors. Front Med 2016; 10:383-388. [PMID: 27770386 DOI: 10.1007/s11684-016-0488-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/18/2016] [Indexed: 12/14/2022]
|
164
|
Ranganathan P, Kashyap T, Yu X, Meng X, Lai TH, McNeil B, Bhatnagar B, Shacham S, Kauffman M, Dorrance AM, Blum W, Sampath D, Landesman Y, Garzon R. XPO1 Inhibition using Selinexor Synergizes with Chemotherapy in Acute Myeloid Leukemia by Targeting DNA Repair and Restoring Topoisomerase IIα to the Nucleus. Clin Cancer Res 2016; 22:6142-6152. [PMID: 27358488 PMCID: PMC5161584 DOI: 10.1158/1078-0432.ccr-15-2885] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 05/19/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE Selinexor, a selective inhibitor of XPO1, is currently being tested as single agent in clinical trials in acute myeloid leukemia (AML). However, considering the molecular complexity of AML, it is unlikely that AML can be cured with monotherapy. Therefore, we asked whether adding already established effective drugs such as topoisomerase (Topo) II inhibitors to selinexor will enhance its anti-leukemic effects in AML. EXPERIMENTAL DESIGN The efficacy of combinatorial drug treatment using Topo II inhibitors (idarubicin, daunorubicin, mitoxantrone, etoposide) and selinexor was evaluated in established cellular and animal models of AML. RESULTS Concomitant treatment with selinexor and Topo II inhibitors resulted in therapeutic synergy in AML cell lines and patient samples. Using a xenograft MV4-11 AML mouse model, we show that treatment with selinexor and idarubicin significantly prolongs survival of leukemic mice compared with each single therapy. CONCLUSIONS Aberrant nuclear export and cytoplasmic localization of Topo IIα has been identified as one of the mechanisms leading to drug resistance in cancer. Here, we show that in a subset of patients with AML that express cytoplasmic Topo IIα, selinexor treatment results in nuclear retention of Topo IIα protein, resulting in increased sensitivity to idarubicin. Selinexor treatment of AML cells resulted in a c-MYC-dependent reduction of DNA damage repair genes (Rad51 and Chk1) mRNA and protein expression and subsequent inhibition of homologous recombination repair and increased sensitivity to Topo II inhibitors. The preclinical data reported here support further clinical studies using selinexor and Topo II inhibitors in combination to treat AML. Clin Cancer Res; 22(24); 6142-52. ©2016 AACR.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Nucleus/drug effects
- DNA Damage/drug effects
- DNA Repair/drug effects
- DNA Topoisomerases, Type II/metabolism
- Drug Resistance, Neoplasm/drug effects
- Female
- Humans
- Hydrazines/pharmacology
- Karyopherins/antagonists & inhibitors
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mice, SCID
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Topoisomerase II Inhibitors/pharmacology
- Triazoles/pharmacology
- Exportin 1 Protein
Collapse
Affiliation(s)
| | | | - Xueyan Yu
- The Ohio State University, Columbus, Ohio
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Festuccia C. Investigational serine/threonine kinase inhibitors against prostate cancer metastases. Expert Opin Investig Drugs 2016; 26:25-34. [PMID: 27892725 DOI: 10.1080/13543784.2016.1266337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Androgen deprivation therapy (ADT) is used as first therapeutic approach in prostate cancer (PCa) although castration resistant disease (CRPC) develops with high frequency. CRPC is the consequence of lack of apoptotic responses to ADT. Alternative targeting of the androgen axis with abiraterone and enzalutamide, as well as taxane-based chemotherapy were used in CRPC. Serine/threonine protein kinases (STKs) regulate different molecular pathways of normal and neoplastic cells and participate to development of CRPC as well as to the progression towards a bone metastatic disease (mCRPC). Areas covered: The present review provide data on STK expression and activity in the development of CRPC as well as summarize recent reports of different strategies to block STK activity for the control of PCa progression. Expert Opinion: Inhibitors for different STKs have been developed but clinical trials in PCa are comparatively rare and few exhibit satisfactory 'drug-like' properties. It is, however, necessary to intensify, when possible, the number of clinical trials with these drugs in order to insert new therapies or combinations with standard hormone- and chemo-therapies in the treatment guidelines of the mPCA.
Collapse
Affiliation(s)
- Claudio Festuccia
- a Department of Biotechnological and Applied Clinical Sciences , University of L'Aquila , L'Aquila , Italy
| |
Collapse
|
166
|
Carrà G, Crivellaro S, Taulli R, Guerrasio A, Saglio G, Morotti A. Mechanisms of p53 Functional De-Regulation: Role of the IκB-α/p53 Complex. Int J Mol Sci 2016; 17:ijms17121997. [PMID: 27916821 PMCID: PMC5187797 DOI: 10.3390/ijms17121997] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/23/2016] [Accepted: 11/24/2016] [Indexed: 02/06/2023] Open
Abstract
TP53 is one of the most frequently-mutated and deleted tumor suppressors in cancer, with a dramatic correlation with dismal prognoses. In addition to genetic inactivation, the p53 protein can be functionally inactivated in cancer, through post-transductional modifications, changes in cellular compartmentalization, and interactions with other proteins. Here, we review the mechanisms of p53 functional inactivation, with a particular emphasis on the interaction between p53 and IκB-α, the NFKBIA gene product.
Collapse
Affiliation(s)
- Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Sabrina Crivellaro
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Riccardo Taulli
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Angelo Guerrasio
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Turin, Italy.
| |
Collapse
|
167
|
Sun HL, Cui R, Zhou J, Teng KY, Hsiao YH, Nakanishi K, Fassan M, Luo Z, Shi G, Tili E, Kutay H, Lovat F, Vicentini C, Huang HL, Wang SW, Kim T, Zanesi N, Jeon YJ, Lee TJ, Guh JH, Hung MC, Ghoshal K, Teng CM, Peng Y, Croce CM. ERK Activation Globally Downregulates miRNAs through Phosphorylating Exportin-5. Cancer Cell 2016; 30:723-736. [PMID: 27846390 PMCID: PMC5127275 DOI: 10.1016/j.ccell.2016.10.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 04/01/2016] [Accepted: 10/03/2016] [Indexed: 02/05/2023]
Abstract
MicroRNAs (miRNA) are mostly downregulated in cancer. However, the mechanism underlying this phenomenon and the precise consequence in tumorigenesis remain obscure. Here we show that ERK suppresses pre-miRNA export from the nucleus through phosphorylation of exportin-5 (XPO5) at T345/S416/S497. After phosphorylation by ERK, conformation of XPO5 is altered by prolyl isomerase Pin1, resulting in reduction of pre-miRNA loading. In liver cancer, the ERK-mediated XPO5 suppression reduces miR-122, increases microtubule dynamics, and results in tumor development and drug resistance. Analysis of clinical specimens further showed that XPO5 phosphorylation is associated with poor prognosis for liver cancer patients. Our study reveals a function of ERK in miRNA biogenesis and suggests that modulation of miRNA export has potential clinical implications.
Collapse
Affiliation(s)
- Hui-Lung Sun
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA; Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Ri Cui
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - JianKang Zhou
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Kun-Yu Teng
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| | - Yung-Hsuan Hsiao
- Department of Human Sciences, Human Nutrition Program, College of Education and Human Ecology, Ohio State University, Columbus, OH 43210, USA
| | - Kotaro Nakanishi
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Matteo Fassan
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA; ARC-NET Research Centre, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Zhenghua Luo
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Guqin Shi
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA
| | - Esmerina Tili
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA; Department of Anesthesiology, Ohio State University, Columbus, OH 43210, USA
| | - Huban Kutay
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| | - Francesca Lovat
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Caterina Vicentini
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Han-Li Huang
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Taewan Kim
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicola Zanesi
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Young-Jun Jeon
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Tae Jin Lee
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, Taipei 10051, Taiwan
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 40402, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Kalpana Ghoshal
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| | - Che-Ming Teng
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Yong Peng
- Department of Thoracic Surgery, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
168
|
Liu X, Chong Y, Tu Y, Liu N, Yue C, Qi Z, Liu H, Yao Y, Liu H, Gao S, Niu M, Yu R. CRM1/XPO1 is associated with clinical outcome in glioma and represents a therapeutic target by perturbing multiple core pathways. J Hematol Oncol 2016; 9:108. [PMID: 27733172 PMCID: PMC5059893 DOI: 10.1186/s13045-016-0338-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/06/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Malignant gliomas are associated with a high mortality rate, and effective treatment options are limited. Thus, the development of novel targeted treatments to battle this deadly disease is imperative. METHODS In this study, we investigated the in vitro effects of the novel reversible chromosomal region maintenance 1 (CRM1) inhibitor S109 on cell proliferation in human gliomas. S109 was also evaluated in an intracranial glioblastoma xenograft model. RESULTS We found that high expression of CRM1 in glioma is a predictor of short overall survival and poor patient outcome. Our data demonstrate that S109 significantly inhibits the proliferation of human glioma cells by inducing cell cycle arrest at the G1 phase. Notably, we observed that high-grade glioma cells are more sensitive to S109 treatment compared with low-grade glioma cells. In an intracranial mouse model, S109 significantly prolonged the survival of tumor-bearing animals without causing any obvious toxicity. Mechanistically, S109 treatment simultaneously perturbed the three core pathways (the RTK/AKT/Foxos signaling pathway and the p53 and Rb1 tumor-suppressor pathways) implicated in human glioma cells by promoting the nuclear retention of multiple tumor-suppressor proteins. CONCLUSIONS Taken together, our study highlights the potential role of CRM1 as an attractive molecular target for the treatment of human glioma and indicates that CRM1 inhibition by S109 might represent a novel treatment approach.
Collapse
Affiliation(s)
- Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Brain Hospital, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yulong Chong
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Nanjing Durm Tower Hospital Group, Suqian City People's Hospital, Suqian, Jiangsu, China
| | - Yiming Tu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ning Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chenglong Yue
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhenglei Qi
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huize Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yao Yao
- Jiangsu Key Laboratory of Bone Marrow Stem Cell, Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongmei Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Brain Hospital, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shangfeng Gao
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Brain Hospital, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingshan Niu
- Jiangsu Key Laboratory of Bone Marrow Stem Cell, Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Brain Hospital, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
169
|
Wrobel K, Zhao YC, Kulkoyluoglu E, Chen KLA, Hieronymi K, Holloway J, Li S, Ray T, Ray PS, Landesman Y, Lipka AE, Smith RL, Madak-Erdogan Z. ERα-XPO1 Cross Talk Controls Tamoxifen Sensitivity in Tumors by Altering ERK5 Cellular Localization. Mol Endocrinol 2016; 30:1029-1045. [PMID: 27533791 PMCID: PMC5045498 DOI: 10.1210/me.2016-1101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/12/2016] [Indexed: 12/22/2022] Open
Abstract
Most breast cancer deaths occur in women with recurrent, estrogen receptor (ER)-α(+), metastatic tumors. There is a critical need for therapeutic approaches that include novel, targetable mechanism-based strategies by which ERα (+) tumors can be resensitized to endocrine therapies. The objective of this study was to validate a group of nuclear transport genes as potential biomarkers to predict the risk of endocrine therapy failure and to evaluate the inhibition of XPO1, one of these genes as a novel means to enhance the effectiveness of endocrine therapies. Using advanced statistical methods, we found that expression levels of several of nuclear transport genes including XPO1 were associated with poor survival and predicted recurrence of tamoxifen-treated breast tumors in human breast cancer gene expression data sets. In mechanistic studies we showed that the expression of XPO1 determined the cellular localization of the key signaling proteins and the response to tamoxifen. We demonstrated that combined targeting of XPO1 and ERα in several tamoxifen-resistant cell lines and tumor xenografts with the XPO1 inhibitor, Selinexor, and tamoxifen restored tamoxifen sensitivity and prevented recurrence in vivo. The nuclear transport pathways have not previously been implicated in the development of endocrine resistance, and given the need for better strategies for selecting patients to receive endocrine modulatory reagents and improving therapy response of relapsed ERα(+) tumors, our findings show great promise for uncovering the role these pathways play in reducing cancer recurrences.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Biological Transport/drug effects
- Biological Transport/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- MCF-7 Cells
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mitogen-Activated Protein Kinase 7/genetics
- Mitogen-Activated Protein Kinase 7/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Tamoxifen/pharmacology
- Exportin 1 Protein
Collapse
Affiliation(s)
- Kinga Wrobel
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Yiru Chen Zhao
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Eylem Kulkoyluoglu
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Karen Lee Ann Chen
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Kadriye Hieronymi
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Jamie Holloway
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Sarah Li
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Tania Ray
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Partha Sarathi Ray
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Yosef Landesman
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Alexander Edward Lipka
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Rebecca Lee Smith
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| |
Collapse
|
170
|
Leisibach P, Schneiter D, Soltermann A, Yamada Y, Weder W, Jungraithmayr W. Prognostic value of immunohistochemical markers in malignant thymic epithelial tumors. J Thorac Dis 2016; 8:2580-2591. [PMID: 27747012 DOI: 10.21037/jtd.2016.08.82] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Thymic epithelial tumors (TET) are rare neoplasms with inconsistent treatment strategies. When researching for molecular pathways to find new therapies, the correlation between specific molecular markers and outcome has only rarely been investigated. The aim of this study was to investigate the correlation between survival, metastatic potential and invasiveness of aggressive subtypes of TET and immunohistochemical markers. METHODS Overall survival (OS), disease-free survival (DFS), progression-free survival (PFS) and metastasis-free survival (MFS) of patients with WHO type B2/B3 mixed type thymoma (MT), thymoma type B3 (B3) and thymic carcinoma (TC), undergoing surgery [1998-2013] were determined. Tumor specimens were stained using a tissue microarray (TMA) (CD117, CD5, p63, p40, p21, p27, p53, Bcl-2, Ki67, podoplanin, synaptophysin, PTEN and Pax8). Invasive behavior of primary tumors and the presence of extrathoracic metastases were assessed. RESULTS We found in 23 patients included into this study (four MT, ten B3, nine TC) that (I) p21 expression in the cytoplasm significantly correlated with a decrease of OS (P=0.016), PFS (P=0.034) and MFS (P=0.005); (II) MFS was significantly shorter when the combination of p21-low p27-low p53-high was present (P=0.029); and (III) nuclear p27 (P=0.042), Ki-67 (P=0.024) and podoplanin (P=0.05) expression correlated with the presence of extrathoracic metastases. CONCLUSIONS The main finding of this study is that cytoplasmic p21 expression negatively influences the outcome of malignant TETs and correlates with metastatic activity. Additionally, selected immunohistochemical markers correlate with the distant metastatic potential of TETs. These results may contribute to the stratification of diagnosis and improvement of treatment strategies for thymic malignancies.
Collapse
Affiliation(s)
- Priska Leisibach
- Division of Thoracic Surgery, University Hospital Zurich, Zurich 8091, Switzerland
| | - Didier Schneiter
- Division of Thoracic Surgery, University Hospital Zurich, Zurich 8091, Switzerland
| | - Alex Soltermann
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Yoshi Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich 8091, Switzerland
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich 8091, Switzerland
| | | |
Collapse
|
171
|
Jardin F, Pujals A, Pelletier L, Bohers E, Camus V, Mareschal S, Dubois S, Sola B, Ochmann M, Lemonnier F, Viailly PJ, Bertrand P, Maingonnat C, Traverse-Glehen A, Gaulard P, Damotte D, Delarue R, Haioun C, Argueta C, Landesman Y, Salles G, Jais JP, Figeac M, Copie-Bergman C, Molina TJ, Picquenot JM, Cornic M, Fest T, Milpied N, Lemasle E, Stamatoullas A, Moeller P, Dyer MJS, Sundstrom C, Bastard C, Tilly H, Leroy K. Recurrent mutations of the exportin 1 gene (XPO1) and their impact on selective inhibitor of nuclear export compounds sensitivity in primary mediastinal B-cell lymphoma. Am J Hematol 2016; 91:923-30. [PMID: 27312795 DOI: 10.1002/ajh.24451] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 06/11/2016] [Accepted: 06/14/2016] [Indexed: 02/01/2023]
Abstract
Primary mediastinal B-cell lymphoma (PMBL) is an entity of B-cell lymphoma distinct from the other molecular subtypes of diffuse large B-cell lymphoma (DLBCL). We investigated the prevalence, specificity, and clinical relevance of mutations of XPO1, which encodes a member of the karyopherin-β nuclear transporters, in a large cohort of PMBL. PMBL cases defined histologically or by gene expression profiling (GEP) were sequenced and the XPO1 mutational status was correlated to genetic and clinical characteristics. The XPO1 mutational status was also assessed in DLBCL, Hodgkin lymphoma (HL) and mediastinal gray-zone lymphoma (MGZL).The biological impact of the mutation on Selective Inhibitor of Nuclear Export (SINE) compounds (KPT-185/330) sensitivity was investigated in vitro. XPO1 mutations were present in 28/117 (24%) PMBL cases and in 5/19 (26%) HL cases but absent/rare in MGZL (0/20) or DLBCL (3/197). A higher prevalence (50%) of the recurrent codon 571 variant (p.E571K) was observed in GEP-defined PMBL and was associated with shorter PFS. Age, International Prognostic Index and bulky mass were similar in XPO1 mutant and wild-type cases. KPT-185 induced a dose-dependent decrease in cell proliferation and increased cell-death in PMBL cell lines harboring wild type or XPO1 E571K mutant alleles. Experiments in transfected U2OS cells further confirmed that the XPO1 E571K mutation does not have a drastic impact on KPT-330 binding. To conclude the XPO1 E571K mutation represents a genetic hallmark of the PMBL subtype and serves as a new relevant PMBL biomarker. SINE compounds appear active for both mutated and wild-type protein. Am. J. Hematol. 91:923-930, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fabrice Jardin
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Anais Pujals
- Departement of Hematology, Inserm U955 Team 09, APHP Hospital Henri Mondor, Créteil, France
| | - Laura Pelletier
- Departement of Hematology, Inserm U955 Team 09, APHP Hospital Henri Mondor, Créteil, France
| | - Elodie Bohers
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Vincent Camus
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Sylvain Mareschal
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Sydney Dubois
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Brigitte Sola
- Departement of Hematology, Normandie Univ, UNICAEN, Caen, EA4652, France
| | - Marlène Ochmann
- Departement of Hematology, Inserm U917, CHU Pontchaillou, Rennes, France
| | - François Lemonnier
- Departement of Hematology, Inserm U955 Team 09, APHP Hospital Henri Mondor, Créteil, France
| | | | - Philippe Bertrand
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | | | | | - Philippe Gaulard
- Departement of Hematology, Inserm U955 Team 09, APHP Hospital Henri Mondor, Créteil, France
| | - Diane Damotte
- Departement of Hematology, Hospices Civils De Lyon, Lyon-1 University, Pierre Benite, CNRS UMR5239, France
| | - Richard Delarue
- Department of Pathology, Hôpitaux Universitaires, Paris Centre, Team « Cancer, Immune Control, and Escape » INSERM U1138, Cordeliers Research Center, Paris, France
| | - Corinne Haioun
- Departement of Hematology, Inserm U955 Team 09, APHP Hospital Henri Mondor, Créteil, France
| | | | - Yosef Landesman
- Department of Hematology, Necker Hospital, AP-HP, Paris, France
| | | | | | - Martin Figeac
- Departement of Genomics, Functional Genomic Platforms, IRCL, Lille, France
| | | | | | | | - Marie Cornic
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Thierry Fest
- Departement of Hematology, Inserm U917, CHU Pontchaillou, Rennes, France
| | - Noel Milpied
- Department of Hematology, CHU De Bordeaux, France
| | - Emilie Lemasle
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | | | - Peter Moeller
- Department of Pathology, Institute of Pathology, University of Ulm, Germany
| | - Martin J S Dyer
- Department of Hematology, Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom
| | | | - Christian Bastard
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Hervé Tilly
- Departement of Hematology, Inserm U918, Centre Henri Becquerel, Rouen, France
| | - Karen Leroy
- Departement of Hematology, Inserm U955 Team 09, APHP Hospital Henri Mondor, Créteil, France
| |
Collapse
|
172
|
Sun Q, Chen X, Zhou Q, Burstein E, Yang S, Jia D. Inhibiting cancer cell hallmark features through nuclear export inhibition. Signal Transduct Target Ther 2016; 1:16010. [PMID: 29263896 PMCID: PMC5661660 DOI: 10.1038/sigtrans.2016.10] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/28/2016] [Accepted: 05/31/2016] [Indexed: 02/05/2023] Open
Abstract
Treating cancer through inhibition of nuclear export is one of the best examples of basic research translation into clinical application. Nuclear export factor chromosomal region maintenance 1 (CRM1; Xpo1 and exportin-1) controls cellular localization and function of numerous proteins that are critical for the development of many cancer hallmarks. The diverse actions of CRM1 are likely to explain the broad ranging anti-cancer potency of CRM1 inhibitors observed in pre-clinical studies and/or clinical trials (phase I–III) on both advanced-stage solid and hematological tumors. In this review, we compare and contrast the mechanisms of action of different CRM1 inhibitors, and discuss the potential benefit of unexplored non-covalent CRM1 inhibitors. This emerging field has uncovered that nuclear export inhibition is well poised as an attractive target towards low-toxicity broad-spectrum potent anti-cancer therapy.
Collapse
Affiliation(s)
- Qingxiang Sun
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xueqin Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ezra Burstein
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Da Jia
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,West China 2nd University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
173
|
Hong AL, Tseng YY, Cowley GS, Jonas O, Cheah JH, Kynnap BD, Doshi MB, Oh C, Meyer SC, Church AJ, Gill S, Bielski CM, Keskula P, Imamovic A, Howell S, Kryukov GV, Clemons PA, Tsherniak A, Vazquez F, Crompton BD, Shamji AF, Rodriguez-Galindo C, Janeway KA, Roberts CWM, Stegmaier K, van Hummelen P, Cima MJ, Langer RS, Garraway LA, Schreiber SL, Root DE, Hahn WC, Boehm JS. Integrated genetic and pharmacologic interrogation of rare cancers. Nat Commun 2016; 7:11987. [PMID: 27329820 PMCID: PMC4917959 DOI: 10.1038/ncomms11987] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023] Open
Abstract
Identifying therapeutic targets in rare cancers remains challenging due to the paucity of established models to perform preclinical studies. As a proof-of-concept, we developed a patient-derived cancer cell line, CLF-PED-015-T, from a paediatric patient with a rare undifferentiated sarcoma. Here, we confirm that this cell line recapitulates the histology and harbours the majority of the somatic genetic alterations found in a metastatic lesion isolated at first relapse. We then perform pooled CRISPR-Cas9 and RNAi loss-of-function screens and a small-molecule screen focused on druggable cancer targets. Integrating these three complementary and orthogonal methods, we identify CDK4 and XPO1 as potential therapeutic targets in this cancer, which has no known alterations in these genes. These observations establish an approach that integrates new patient-derived models, functional genomics and chemical screens to facilitate the discovery of targets in rare cancers.
Collapse
Affiliation(s)
- Andrew L. Hong
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Yuen-Yi Tseng
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Glenn S. Cowley
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Oliver Jonas
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Jaime H. Cheah
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Bryan D. Kynnap
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Mihir B. Doshi
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Coyin Oh
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Stephanie C. Meyer
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Alanna J. Church
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Shubhroz Gill
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Craig M. Bielski
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paula Keskula
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Alma Imamovic
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Sara Howell
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Gregory V. Kryukov
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Paul A. Clemons
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Aviad Tsherniak
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Francisca Vazquez
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Brian D. Crompton
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Alykhan F. Shamji
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Carlos Rodriguez-Galindo
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Katherine A. Janeway
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Charles W. M. Roberts
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Kimberly Stegmaier
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paul van Hummelen
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Michael J. Cima
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Robert S. Langer
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Levi A. Garraway
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Stuart L. Schreiber
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - David E. Root
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - William C. Hahn
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Jesse S. Boehm
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
174
|
MicroRNA-1301-Mediated RanGAP1 Downregulation Induces BCR-ABL Nuclear Entrapment to Enhance Imatinib Efficacy in Chronic Myeloid Leukemia Cells. PLoS One 2016; 11:e0156260. [PMID: 27228340 PMCID: PMC4881950 DOI: 10.1371/journal.pone.0156260] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/11/2016] [Indexed: 12/18/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease. Imatinib (IM), the first line treatment for CML, is excessively expensive and induces various side effects in CML patients. Therefore, it is essential to investigate a new strategy for improving CML therapy. Our immunoblot data revealed that RanGTPase activating protein 1 (RanGAP1) protein levels increased by approximately 30-fold in K562 cells compared with those in normal cells. RanGAP1 is one of the important components of RanGTPase system, which regulates the export of nuclear protein. However, whether RanGAP1 level variation influences BCR-ABL nuclear export is still unknown. In this report, using shRNA to downregulate RanGAP1 expression level augmented K562 cell apoptosis by approximately 40% after treatment with 250 nM IM. Immunofluorescence assay also indicated that three-fold of nuclear BCR-ABL was detected. These data suggest that BCR-ABL nuclear entrapment induced by RanGAP1 downregulation can be used to improve IM efficacy. Moreover, our qRT-PCR data indicated a trend of inverse correlation between the RanGAP1 and microRNA (miR)-1301 levels in CML patients. MiR-1301, targeting the RanGAP1 3' untranslated region, decreased by approximately 100-fold in K562 cells compared with that in normal cells. RanGAP1 downregulation by miR-1301 transfection impairs BCR-ABL nuclear export to increase approximately 60% of cell death after treatment of 250 nM IM. This result was almost the same as treatment with 1000 nM IM alone. Furthermore, immunofluorescence assay demonstrated that Tyr-99 of nuclear P73 was phosphorylated accompanied with nuclear entrapment of BCR-ABL after transfection with RanGAP1 shRNA or miR-1301 in IM-treated K562 cells. Altogether, we demonstrated that RanGAP1 downregulation can mediate BCR-ABL nuclear entrapment to activate P73-dependent apoptosis pathway which is a novel strategy for improving current IM treatment for CML.
Collapse
|
175
|
Proteome-wide search for functional motifs altered in tumors: Prediction of nuclear export signals inactivated by cancer-related mutations. Sci Rep 2016; 6:25869. [PMID: 27174732 PMCID: PMC4865848 DOI: 10.1038/srep25869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/22/2016] [Indexed: 12/15/2022] Open
Abstract
Large-scale sequencing projects are uncovering a growing number of missense mutations in human tumors. Understanding the phenotypic consequences of these alterations represents a formidable challenge. In silico prediction of functionally relevant amino acid motifs disrupted by cancer mutations could provide insight into the potential impact of a mutation, and guide functional tests. We have previously described Wregex, a tool for the identification of potential functional motifs, such as nuclear export signals (NESs), in proteins. Here, we present an improved version that allows motif prediction to be combined with data from large repositories, such as the Catalogue of Somatic Mutations in Cancer (COSMIC), and to be applied to a whole proteome scale. As an example, we have searched the human proteome for candidate NES motifs that could be altered by cancer-related mutations included in the COSMIC database. A subset of the candidate NESs identified was experimentally tested using an in vivo nuclear export assay. A significant proportion of the selected motifs exhibited nuclear export activity, which was abrogated by the COSMIC mutations. In addition, our search identified a cancer mutation that inactivates the NES of the human deubiquitinase USP21, and leads to the aberrant accumulation of this protein in the nucleus.
Collapse
|
176
|
Ortega JF, de Conti A, Tryndyak V, Furtado KS, Heidor R, Horst MA, Fernandes LHG, Tavares PELM, Pogribna M, Shpyleva S, Beland FA, Pogribny IP, Moreno FS. Suppressing activity of tributyrin on hepatocarcinogenesis is associated with inhibiting the p53-CRM1 interaction and changing the cellular compartmentalization of p53 protein. Oncotarget 2016; 7:24339-47. [PMID: 27013579 PMCID: PMC5029705 DOI: 10.18632/oncotarget.8248] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 02/28/2016] [Indexed: 12/02/2022] Open
Abstract
Hepatocellular carcinoma (HCC), an aggressive and the fastest growing life-threatening cancer worldwide, is often diagnosed at intermediate or advanced stages of the disease, which substantially limits therapeutic approaches for its successful treatment. This indicates that the prevention of hepatocarcinogenesis is probably the most promising approach to reduce both the HCC incidence and cancer-related mortality. In previous studies, we demonstrated a potent chemopreventive effect of tributyrin, a butyric acid prodrug, on experimental hepatocarcinogenesis. The cancer-inhibitory effect of tributyrin was linked to the suppression of sustained cell proliferation and induction of apoptotic cell death driven by an activation of the p53 apoptotic signaling pathway. The goal of the present study was to investigate the underlying molecular mechanisms linked to tributyrin-mediated p53 activation. Using in vivo and in vitro models of liver cancer, we demonstrate that an increase in the level of p53 protein in nuclei, a decrease in the level of cytoplasmic p53, and, consequently, an increase in the ratio of nuclear/cytoplasmic p53 in rat preneoplastic livers and in rat and human HCC cell lines caused by tributyrin or sodium butyrate treatments was associated with a marked increase in the level of nuclear chromosome region maintenance 1 (CRM1) protein. Mechanistically, the increase in the level of nuclear p53 protein was associated with a substantially reduced binding interaction between CRM1 and p53. The results demonstrate that the cancer-inhibitory activity of sodium butyrate and its derivatives on liver carcinogenesis may be attributed to retention of p53 and CRM1 proteins in the nucleus, an event that may trigger activation of p53-mediated apoptotic cell death in neoplastic cells.
Collapse
Affiliation(s)
- Juliana F. Ortega
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline de Conti
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - Volodymyr Tryndyak
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - Kelly S. Furtado
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Renato Heidor
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Aderuza Horst
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Laura Helena Gasparini Fernandes
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paulo Eduardo Latorre Martins Tavares
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marta Pogribna
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - Svitlana Shpyleva
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - Frederick A. Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - Igor P. Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, USA
| | - Fernando Salvador Moreno
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
177
|
Xie QL, Liu Y, Zhu Y. Chromosome region maintenance 1 expression and its association with clinical pathological features in primary carcinoma of the liver. Exp Ther Med 2016; 12:59-68. [PMID: 27347018 PMCID: PMC4907041 DOI: 10.3892/etm.2016.3283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 02/19/2016] [Indexed: 12/18/2022] Open
Abstract
Liver cancer is the third leading cause of cancer-associated mortality worldwide. Recurrence and metastasis are the major factors affecting the prognosis; thus, investigation of the underlying molecular mechanisms of invasion and metastasis, and detection of novel drug target may improve the mortality rate of liver cancer patients. Chromosome region maintenance 1 (CRM1) recognizes specific leucine-rich nuclear export signal sequences, and its overexpression is associated with tumor-suppressor gene inactivation, proliferation, invasion and resistance to chemotherapy. The aim of the present study was to examine the association of CRM1 expression with the clinical and pathological features of primary liver cancer. In total, 152 cases diagnosed with liver cancer were included. CRM1 expression was detected in cancer tissues and adjacent normal tissues by immunohistochemical assay. No statistically significant difference was found between the CRM1 expression levels in tumor and adjacent normal tissues (P=0.106). However, CRM1 expression in adjacent normal tissues was higher compared with that in tumor tissues in the negative hepatitis B envelope antigen (HBeAg; P=0.029) and low differentiation (P=0.004) groups. In tumor tissues, CRM1 expression was significantly correlated with differentiation (P=0.045), whereas in adjacent normal tissues, CRM1 expression was significantly correlated with the tumor diameter (P=0.004). Therefore, it can be concluded that CRM1 is highly expressed in both tumor and adjacent normal tissues. Furthermore, CRM1 expression is associated with the tumor differentiation degree and diameter. Lower differentiation and larger tumor diameter resulted in higher CRM1 expression in adjacent normal tissues, and higher tendency for invasion and metastasis. In addition, the risk of invasion and metastasis remains in chronic hepatitis B patients with negative HBeAg.
Collapse
Affiliation(s)
- Qiao-Ling Xie
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Yue Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Ying Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| |
Collapse
|
178
|
Mechanisms of Nuclear Export in Cancer and Resistance to Chemotherapy. Cancers (Basel) 2016; 8:cancers8030035. [PMID: 26985906 PMCID: PMC4810119 DOI: 10.3390/cancers8030035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/03/2016] [Accepted: 03/08/2016] [Indexed: 01/14/2023] Open
Abstract
Tumour suppressor proteins, such as p53, BRCA1, and ABC, play key roles in preventing the development of a malignant phenotype, but those that function as transcriptional regulators need to enter the nucleus in order to function. The export of proteins between the nucleus and cytoplasm is complex. It occurs through nuclear pores and exported proteins need a nuclear export signal (NES) to bind to nuclear exportin proteins, including CRM1 (Chromosomal Region Maintenance protein 1), and the energy for this process is provided by the RanGTP/RanGDP gradient. Due to the loss of DNA repair and cell cycle checkpoints, drug resistance is a major problem in cancer treatment, and often an initially successful treatment will fail due to the development of resistance. An important mechanism underlying resistance is nuclear export, and a number of strategies that can prevent nuclear export may reverse resistance. Examples include inhibitors of CRM1, antibodies to the nuclear export signal, and alteration of nuclear pore structure. Each of these are considered in this review.
Collapse
|
179
|
Wu J, Savooji J, Liu D. Second- and third-generation ALK inhibitors for non-small cell lung cancer. J Hematol Oncol 2016; 9:19. [PMID: 26951079 PMCID: PMC4782349 DOI: 10.1186/s13045-016-0251-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022] Open
Abstract
Crizotinib as the first-generation ALK inhibitor has shown significant activity in ALK-mutated non-small cell lung cancer (NSCLC). Second- and third-generation ALK inhibitors are entering clinical applications for ALK+ NSCLC. In addition, a third-generation ALK inhibitor, lorlatinib (PF-06463922), was reported to resensitize NSCLC to crizotinib. This review provided a summary of clinical development of alectinib, ceritinib, brigatinib (AP26113), and lorlatinib.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - John Savooji
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY, 10595, USA
| | - Delong Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
180
|
Liu X, Chong Y, Liu H, Han Y, Niu M. CRM1 inhibitor S109 suppresses cell proliferation and induces cell cycle arrest in renal cancer cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:161-8. [PMID: 26937212 PMCID: PMC4770106 DOI: 10.4196/kjpp.2016.20.2.161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/30/2015] [Accepted: 07/14/2015] [Indexed: 02/02/2023]
Abstract
Abnormal localization of tumor suppressor proteins is a common feature of renal cancer. Nuclear export of these tumor suppressor proteins is mediated by chromosome region maintenance-1 (CRM1). Here, we investigated the antitumor eff ects of a novel reversible inhibitor of CRM1 on renal cancer cells. We found that S109 inhibits the CRM1-mediated nuclear export of RanBP1 and reduces protein levels of CRM1. Furthermore, the inhibitory eff ect of S109 on CRM1 is reversible. Our data demonstrated that S109 signifi cantly inhibits proliferation and colony formation of renal cancer cells. Cell cycle assay showed that S109 induced G1-phase arrest, followed by the reduction of Cyclin D1 and increased expression of p53 and p21. We also found that S109 induces nuclear accumulation of tumor suppressor proteins, Foxo1 and p27. Most importantly, mutation of CRM1 at Cys528 position abolished the eff ects of S109. Taken together, our results indicate that CRM1 is a therapeutic target in renal cancer and the novel reversible CRM1 inhibitor S109 can act as a promising candidate for renal cancer therapy.
Collapse
Affiliation(s)
- Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou 221002, Jiangsu, China.; Brain Hospital, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | - Yulong Chong
- Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | - Huize Liu
- Insititute of Nervous System Diseases, Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | - Yan Han
- Dalian Center for Disease Control and Prevention, Dalian 116002, Liaoning, China
| | - Mingshan Niu
- Blood Disease Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, China.; Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
181
|
Macrophages and endothelial cells orchestrate tumor-associated angiogenesis in oral cancer via hedgehog pathway activation. Tumour Biol 2016; 37:9233-41. [DOI: 10.1007/s13277-015-4763-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/29/2015] [Indexed: 12/16/2022] Open
|
182
|
Pitt SC, Hernandez RA, Nehs MA, Gawande AA, Moore FD, Ruan DT, Cho NL. Identification of Novel Oncogenic Mutations in Thyroid Cancer. J Am Coll Surg 2016; 222:1036-1043.e2. [PMID: 27010584 DOI: 10.1016/j.jamcollsurg.2015.12.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/29/2015] [Accepted: 12/21/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Thyroid cancer patients frequently have favorable outcomes. However, a small subset develops aggressive disease refractory to traditional treatments. Therefore, we sought to characterize oncogenic mutations in thyroid cancers to identify novel therapeutic targets that may benefit patients with advanced, refractory disease. STUDY DESIGN Data on 239 thyroid cancer specimens collected between January 2009 and September 2014 were obtained from the Dana Farber/Brigham and Women's Cancer Center. The tumors were analyzed with the OncoMap-4 or OncoPanel high-throughput genotyping platforms that survey up to 275 cancer genes and 91 introns for DNA rearrangement. RESULTS Of the 239 thyroid cancer specimens, 128 (54%) had oncogenic mutations detected. These 128 tumors had 351 different mutations detected in 129 oncogenes or tumor suppressors. Examination of the 128 specimens demonstrated that 55% (n = 70) had 1 oncogenic mutation, and 45% (n = 48) had more than 1 mutation. The 351 oncogenic mutations were in papillary (85%), follicular (4%), medullary (7%), and anaplastic (4%) thyroid cancers. Analysis revealed that 2.3% (n = 3 genes) of the somatic gene mutations were novel. These included AR (n = 1), MPL (n = 2), and EXT2 (n = 1), which were present in 4 different papillary thyroid cancer specimens. New mutations were found in an additional 13 genes known to have altered protein expression in thyroid cancer: BLM, CBL, CIITA, EP300, GSTM5, LMO2, PRAME, SBDS, SF1, TET2, TNFAIP3, XPO1, and ZRSR2. CONCLUSIONS This analysis revealed that several previously unreported oncogenic gene mutations exist in thyroid cancers and may be targets for the development of future therapies. Further investigation into the role of these genes is warranted.
Collapse
Affiliation(s)
- Susan C Pitt
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| | - Roland A Hernandez
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Matthew A Nehs
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Atul A Gawande
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Francis D Moore
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daniel T Ruan
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Nancy L Cho
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
183
|
Crochiere ML, Baloglu E, Klebanov B, Donovan S, del Alamo D, Lee M, Kauffman M, Shacham S, Landesman Y. A method for quantification of exportin-1 (XPO1) occupancy by Selective Inhibitor of Nuclear Export (SINE) compounds. Oncotarget 2016; 7:1863-77. [PMID: 26654943 PMCID: PMC4811503 DOI: 10.18632/oncotarget.6495] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/18/2015] [Indexed: 12/13/2022] Open
Abstract
Selective Inhibitor of Nuclear Export (SINE) compounds are a family of small-molecules that inhibit nuclear export through covalent binding to cysteine 528 (Cys528) in the cargo-binding pocket of Exportin 1 (XPO1/CRM1) and promote cancer cell death. Selinexor is the lead SINE compound currently in phase I and II clinical trials for advanced solid and hematological malignancies. In an effort to understand selinexor-XPO1 interaction and to establish whether cancer cell response is a function of drug-target engagement, we developed a quantitative XPO1 occupancy assay. Biotinylated leptomycin B (b-LMB) was utilized as a tool compound to measure SINE-free XPO1. Binding to XPO1 was quantitated from SINE compound treated adherent and suspension cells in vitro, dosed ex vivo human peripheral blood mononuclear cells (PBMCs), and PBMCs from mice dosed orally with drug in vivo. Evaluation of a panel of selinexor sensitive and resistant cell lines revealed that resistance was not attributed to XPO1 occupancy by selinexor. Administration of a single dose of selinexor bound XPO1 for minimally 72 hours both in vitro and in vivo. While XPO1 inhibition directly correlates with selinexor pharmacokinetics, the biological outcome of this inhibition depends on modulation of pathways downstream of XPO1, which ultimately determines cancer cell responsiveness.
Collapse
MESH Headings
- Acrylamides/chemistry
- Acrylamides/pharmacology
- Acrylates/chemistry
- Acrylates/pharmacology
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacokinetics
- Antibiotics, Antineoplastic/pharmacology
- Biotinylation
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cell Survival/drug effects
- Cells, Cultured
- Drug Evaluation, Preclinical/methods
- Fatty Acids, Unsaturated/chemistry
- Fatty Acids, Unsaturated/pharmacokinetics
- Fatty Acids, Unsaturated/pharmacology
- HCT116 Cells
- Humans
- Hydrazines/chemistry
- Hydrazines/pharmacokinetics
- Hydrazines/pharmacology
- Karyopherins/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Mice
- Molecular Structure
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reproducibility of Results
- Thiazoles/chemistry
- Thiazoles/pharmacology
- Triazoles/chemistry
- Triazoles/pharmacokinetics
- Triazoles/pharmacology
- Exportin 1 Protein
Collapse
Affiliation(s)
- Marsha L. Crochiere
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Erkan Baloglu
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Boris Klebanov
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Scott Donovan
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Diego del Alamo
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Margaret Lee
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Michael Kauffman
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Sharon Shacham
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| | - Yosef Landesman
- All authors are current or former employees of Karyopharm Therapeutics Inc., Newton, MA, 02459 U.S.A
| |
Collapse
|
184
|
Zinzani PL, Bonthapally V, Huebner D, Lutes R, Chi A, Pileri S. Panoptic clinical review of the current and future treatment of relapsed/refractory T-cell lymphomas: Peripheral T-cell lymphomas. Crit Rev Oncol Hematol 2016; 99:214-27. [PMID: 26811013 DOI: 10.1016/j.critrevonc.2015.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/11/2015] [Accepted: 12/23/2015] [Indexed: 12/11/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) tend to be aggressive and chemorefractory, with about 70% of patients developing relapsed/refractory disease. Prior to 2009, chemotherapies were the only options for relapsed/refractory PTCL, other than hematopoietic transplants. However, chemotherapy only improves survival by about 1 month compared with palliation. Four drugs are now approved in the US to treat relapsed/refractory PTCL: pralatrexate, romidepsin, belinostat, and brentuximab vedotin (for systemic anaplastic large cell lymphoma [sALCL]). Response rates with pralatrexate, romidepsin, and belinostat range from 25 to 54% in mixed relapsed/refractory PTCL populations, while 86% of sALCL patients respond to brentuximab vedotin. Here, we critically evaluate the evidence supporting the current drug treatment of relapsed/refractory PTCL, and look to the future to see how the treatment panorama may change with the advent of new targeted therapies, some of which (e.g., alisertib in PTCL and mogamulizumab in CCR4-positive adult T-cell leukemia/lymphoma) are already in phase 3 trials.
Collapse
Affiliation(s)
- Pier Luigi Zinzani
- Institute of Hematology 'L. e A. Seràgnoli', Policlinico Sant'Orsola-Malpighi, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Vijayveer Bonthapally
- Global Outcomes and Epidemiology Research (GOER), Millennium Pharmaceuticals Inc., 40 Lansdowne Street, Cambridge, MA 02139, USA(1)
| | - Dirk Huebner
- Oncology Clinical Research, Millennium Pharmaceuticals Inc., 35 Lansdowne Street, Cambridge, MA 02139, USA(1)
| | - Richard Lutes
- Oncology Clinical Research, Millennium Pharmaceuticals Inc., 35 Lansdowne Street, Cambridge, MA 02139, USA(1)
| | - Andy Chi
- Department of Biostatistics, Millennium Pharmaceuticals Inc., 40 Lansdowne Street, Cambridge, MA 02139, USA(1)
| | - Stefano Pileri
- Department of Experimental, Diagnostic, and Specialty Medicine, Bologna University School of Medicine, Via Massarenti 8, 40138 Bologna, Italy; Unit of Hematopathology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
185
|
Gravina GL, Mancini A, Sanita P, Vitale F, Marampon F, Ventura L, Landesman Y, McCauley D, Kauffman M, Shacham S, Festuccia C. KPT-330, a potent and selective exportin-1 (XPO-1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin [corrected] in prostate cancer models. BMC Cancer 2015; 15:941. [PMID: 26620414 PMCID: PMC4666032 DOI: 10.1186/s12885-015-1936-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND AIMS Increased expression of Chromosome Region Maintenance (CRM-1)/exportin-1 (XPO-1) has been correlated with poor prognosis in several aggressive tumors, making it an interesting therapeutic target. Selective Inhibitor of Nuclear Export (SINE) compounds bind to XPO-1 and block its ability to export cargo proteins. Here, we investigated the effects of a new class of SINE compounds in models of prostate cancer. MATERIAL AND METHODS We evaluated the expression of XPO-1 in human prostate cancer tissues and cell lines. Next, six SINE (KPT-127, KPT-185, KPT-205, KPT-225, KPT-251 and KPT-330) compounds having different potency with broad-spectrum, tumor-selective cytotoxicity, tolerability and pharmacokinetic profiles were tested in a panel of prostate cancer cells representing distinct differentiation/progression states of disease and genotypes. Two SINE candidates for clinical trials (KPT-251 and KPT-330) were also tested in vivo in three cell models of aggressive prostate cancer engrafted in male nude mice. RESULTS AND CONCLUSIONS XPO-1 is overexpressed in prostate cancer compared to normal or hyperplastic tissues. Increased XPO-1 expression, mainly in the nuclear compartment, was associated with increased Gleason score and bone metastatic potential supporting the use of SINEs in advanced prostate cancer. SINE compounds inhibited proliferation and promoted apoptosis of tumor cells, but did not affect immortalized non-transformed prostate epithelial cells. Nuclei from SINE treated cells showed increased protein localization of XPO-1, survivin and cyclin D1 followed by degradation of these proteins leading to cell cycle arrest and apoptosis. Oral administration of KPT-251 and KPT-330 in PC3, DU145 and 22rv1 tumor-bearing nude mice reduced tumor cell proliferation, angiogenesis and induced apoptosis. Our results provide supportive evidence for the therapeutic use of SINE compounds in advanced/castration resistant prostate cancers and warrants further clinical investigation.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Patrizia Sanita
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Flora Vitale
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Luca Ventura
- Pathology Division, San Salvatore Hospital, L'Aquila, Italy.
| | | | | | | | | | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
186
|
Mucke HA. Drug Repurposing Patent Applications April–June 2015. Assay Drug Dev Technol 2015; 13:654-60. [DOI: 10.1089/adt.2015.29030.pq2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
187
|
Cang S, Iragavarapu C, Savooji J, Song Y, Liu D. ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J Hematol Oncol 2015; 8:129. [PMID: 26589495 PMCID: PMC4654800 DOI: 10.1186/s13045-015-0224-3] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/10/2015] [Indexed: 12/21/2022] Open
Abstract
With the advent of new agents targeting CD20, Bruton's tyrosine kinase, and phosphoinositol-3 kinase for chronic lymphoid leukemia (CLL), more treatment options exist than ever before. B-cell lymphoma-2 (BCL-2) plays a major role in cellular apoptosis and is a druggable target. Small molecule inhibitors of BCL-2 are in active clinical studies. ABT-199 (venetoclax, RG7601, GDC-0199) has been granted breakthrough designation by FDA for relapsed or refractory CLL with 17p deletion. In this review, we summarized the latest clinical development of ABT-199/venetoclax and other novel agents targeting the BCL-2 proteins.
Collapse
Affiliation(s)
- Shundong Cang
- Department of Oncology, The Henan Province People's Hospital, Zhengzhou, China
| | - Chaitanya Iragavarapu
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla, New York, 10595, USA
| | - John Savooji
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla, New York, 10595, USA
| | - Yongping Song
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Delong Liu
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
188
|
Crochiere M, Kashyap T, Kalid O, Shechter S, Klebanov B, Senapedis W, Saint-Martin JR, Landesman Y. Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitor of Nuclear Export (SINE) compounds. BMC Cancer 2015; 15:910. [PMID: 26573568 PMCID: PMC4647283 DOI: 10.1186/s12885-015-1790-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Exportin 1 (XPO1) is a well-characterized nuclear export protein whose expression is up-regulated in many types of cancers and functions to transport key tumor suppressor proteins (TSPs) from the nucleus. Karyopharm Therapeutics has developed a series of small-molecule Selective Inhibitor of Nuclear Export (SINE) compounds, which have been shown to block XPO1 function both in vitro and in vivo. The drug candidate, selinexor (KPT-330), is currently in Phase-II/IIb clinical trials for treatment of both hematologic and solid tumors. The present study sought to decipher the mechanisms that render cells either sensitive or resistant to treatment with SINE compounds, represented by KPT-185, an early analogue of KPT-330. METHODS Using the human fibrosarcoma HT1080 cell line, resistance to SINE was acquired over a period of 10 months of constant incubation with increasing concentration of KPT-185. Cell viability was assayed by MTT. Immunofluorescence was used to compare nuclear export of TSPs. Fluorescence activated cell sorting (FACS), quantitative polymerase chain reaction (qPCR), and immunoblots were used to measure effects on cell cycle, gene expression, and cell death. RNA from naïve and drug treated parental and resistant cells was analyzed by Affymetrix microarrays. RESULTS Treatment of HT1080 cells with gradually increasing concentrations of SINE resulted in >100 fold decrease in sensitivity to SINE cytotoxicity. Resistant cells displayed prolonged cell cycle, reduced nuclear accumulation of TSPs, and similar changes in protein expression compared to parental cells, however the magnitude of the protein expression changes were more significant in parental cells. Microarray analyses comparing parental to resistant cells indicate that a number of key signaling pathways were altered in resistant cells including expression changes in genes involved in adhesion, apoptosis, and inflammation. While the patterns of changes in transcription following drug treatment are similar in parental and resistant cells, the extent of response was more robust in the parental cells. CONCLUSIONS These results suggest that SINE resistance is conferred by alterations in signaling pathways downstream of XPO1 inhibition. Modulation of these pathways could potentially overcome the resistance to nuclear export inhibitors.
Collapse
Affiliation(s)
- Marsha Crochiere
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Trinayan Kashyap
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Ori Kalid
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Sharon Shechter
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Boris Klebanov
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - William Senapedis
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | | | - Yosef Landesman
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| |
Collapse
|
189
|
Ohkoshi S, Yano M, Matsuda Y. Oncogenic role of p21 in hepatocarcinogenesis suggests a new treatment strategy. World J Gastroenterol 2015; 21:12150-6. [PMID: 26576099 PMCID: PMC4641132 DOI: 10.3748/wjg.v21.i42.12150] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/30/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
A well-known tumor suppressor, p21, acts paradoxically by promoting tumor growth in some cellular conditions. These conflicting functions have been demonstrated in association with the HBx gene and in hepatocarcinogenesis. The molecular behavior of p21 depends on its subcellular localization. Nuclear p21 may inhibit cell proliferation and be proapoptotic, while cytoplasmic p21 may have oncogenic and anti-apoptotic functions. Because most typical tumor suppressive proteins also have different effects according to subcellular localization, elucidating the regulatory mechanisms underlying nucleo-cytoplasmic transport of these proteins would be significant and may lead to a new strategy for anti-hepatocellular carcinoma (HCC) therapy. Chromosome region maintenance 1 (CRM1) is a major nuclear export receptor involved in transport of tumor suppressors from nucleus to cytoplasm. Expression of CRM1 is enhanced in a variety of malignancies and in vitro studies have shown the efficacy of specific inhibition of CRM1 against cancer cell lines. Interestingly, interferon may keep p21 in the nucleus; this is one of the mechanisms of its anti-hepatocarcinogenic function. Here we review the oncogenic property of p21, which depends on its subcellular localization, and discuss the rationale underlying a new strategy for HCC treatment and prevention.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Drug Design
- Humans
- Karyopherins/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Molecular Targeted Therapy
- Oncogene Proteins/metabolism
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/metabolism
- Exportin 1 Protein
Collapse
|
190
|
Matsuura Y. Mechanistic Insights from Structural Analyses of Ran-GTPase-Driven Nuclear Export of Proteins and RNAs. J Mol Biol 2015; 428:2025-39. [PMID: 26519791 DOI: 10.1016/j.jmb.2015.09.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/28/2015] [Indexed: 12/13/2022]
Abstract
Understanding how macromolecules are rapidly exchanged between the nucleus and the cytoplasm through nuclear pore complexes is a fundamental problem in biology. Exportins are Ran-GTPase-dependent nuclear transport factors that belong to the karyopherin-β family and mediate nuclear export of a plethora of proteins and RNAs, except for bulk mRNA nuclear export. Exportins bind cargo macromolecules in a Ran-GTP-dependent manner in the nucleus, forming exportin-cargo-Ran-GTP complexes (nuclear export complexes). Transient weak interactions between exportins and nucleoporins containing characteristic FG (phenylalanine-glycine) repeat motifs facilitate nuclear pore complex passage of nuclear export complexes. In the cytoplasm, nuclear export complexes are disassembled, thereby releasing the cargo. GTP hydrolysis by Ran promoted in the cytoplasm makes the disassembly reaction virtually irreversible and provides thermodynamic driving force for the overall export reaction. In the past decade, X-ray crystallography of some of the exportins in various functional states coupled with functional analyses, single-particle electron microscopy, molecular dynamics simulations, and small-angle solution X-ray scattering has provided rich insights into the mechanism of cargo binding and release and also begins to elucidate how exportins interact with the FG repeat motifs. The knowledge gained from structural analyses of nuclear export is being translated into development of clinically useful inhibitors of nuclear export to treat human diseases such as cancer and influenza.
Collapse
Affiliation(s)
- Yoshiyuki Matsuura
- Division of Biological Science and Structural Biology Research Center, Graduate School of Science, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
191
|
Tabe Y, Kojima K, Yamamoto S, Sekihara K, Matsushita H, Davis RE, Wang Z, Ma W, Ishizawa J, Kazuno S, Kauffman M, Shacham S, Fujimura T, Ueno T, Miida T, Andreeff M. Ribosomal Biogenesis and Translational Flux Inhibition by the Selective Inhibitor of Nuclear Export (SINE) XPO1 Antagonist KPT-185. PLoS One 2015; 10:e0137210. [PMID: 26340096 PMCID: PMC4560410 DOI: 10.1371/journal.pone.0137210] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 08/13/2015] [Indexed: 01/01/2023] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma characterized by the aberrant expression of several growth-regulating, oncogenic effectors. Exportin 1 (XPO1) mediates the nucleocytoplasmic transport of numerous molecules including oncogenic growth-regulating factors, RNAs, and ribosomal subunits. In MCL cells, the small molecule KPT-185 blocks XPO1 function and exerts anti-proliferative effects. In this study, we investigated the molecular mechanisms of this putative anti-tumor effect on MCL cells using cell growth/viability assays, immunoblotting, gene expression analysis, and absolute quantification proteomics. KPT-185 exhibited a p53-independent anti-lymphoma effect on MCL cells, by suppression of oncogenic mediators (e.g., XPO1, cyclin D1, c-Myc, PIM1, and Bcl-2 family members), repression of ribosomal biogenesis, and downregulation of translation/chaperone proteins (e.g., PIM2, EEF1A1, EEF2, and HSP70) that are part of the translational/transcriptional network regulated by heat shock factor 1. These results elucidate a novel mechanism in which ribosomal biogenesis appears to be a key component through which XPO1 contributes to tumor cell survival. Thus, we propose that the blockade of XPO1 could be a promising, novel strategy for the treatment of MCL and other malignancies overexpressing XPO1.
Collapse
Affiliation(s)
- Yoko Tabe
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
- Department of Clinical Laboratory Medicine, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kensuke Kojima
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| | - Shinichi Yamamoto
- Department of Clinical Laboratory Medicine, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Leading Center for the Development and Research of Cancer Medicine, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kazumasa Sekihara
- Department of Clinical Laboratory Medicine, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Leading Center for the Development and Research of Cancer Medicine, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hiromichi Matsushita
- Department of Laboratory Medicine, Tokai University of Medicine, Kanagawa, Japan
| | - Richard Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| | - Zhiqiang Wang
- Department of Lymphoma and Myeloma, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| | - Wencai Ma
- Department of Lymphoma and Myeloma, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Michael Kauffman
- Karyopharm Therapeutics Inc., Natick, MA, United States of America
| | - Sharon Shacham
- Karyopharm Therapeutics Inc., Natick, MA, United States of America
| | - Tsutomu Fujimura
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takashi Ueno
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Biomedical Research Center Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
192
|
Non-invasive approaches to monitor EGFR-TKI treatment in non-small-cell lung cancer. J Hematol Oncol 2015; 8:95. [PMID: 26227959 PMCID: PMC4521383 DOI: 10.1186/s13045-015-0193-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/20/2015] [Indexed: 01/10/2023] Open
Abstract
Tyrosine kinase inhibitors of epidermal growth factor receptor (EGFR-TKIs) are standard treatments for advanced non-small-cell lung cancer (NSCLC) patients harboring activating epidermal growth factor receptor (EGFR) mutations. Nowadays, tumor tissues acquired by surgery or biopsy are the routine materials for EGFR mutation analysis. However, the accessibility of tumor tissues is not always satisfactory in advanced NSCLC. Moreover, a high proportion of NSCLC patients will eventually develop resistance to EGFR-TKIs. Invasive procedures, such as surgery or biopsy, are impractical to be performed repeatedly to assess the evolution of EGFR-TKI resistance. Thus, exploring some convenient and less invasive techniques to monitor EGFR-TKI treatment is urgently needed. Circulating cell-free tumor DNA (ctDNA) has a high degree of specificity to detect EGFR mutations in NSCLC. Besides, ctDNA is capable of monitoring the disease progression during EGFR-TKI treatment. Certain serum microRNAs that correlate with EGFR signaling pathway, such as miR-21 and miR-10b, have been demonstrated to be helpful in evaluating the efficiency of EGFR-TKI therapeutics. A commercialized serum-based proteomic test, named VeriStrat test, has shown an outstanding ability to predict the clinical outcome of NSCLC patients receiving EGFR-TKIs. Analysis of EGFR mutations in circulating tumor cells (CTCs) is feasible, and CTCs represent a promising material to predict EGFR-TKI-treatment efficacy and resistance. These evidences suggested that non-invasive techniques based on serum or plasma samples had a great potential for monitoring EGFR-TKI treatment in NSCLC. In this review, we summarized these non-invasive approaches and considered their possible applications in EGFR-TKI-treatment monitoring.
Collapse
|
193
|
Li B, Huang MZ, Wang XQ, Tao BB, Zhong J, Wang XH, Zhang WC, Li ST. TMEM140 is associated with the prognosis of glioma by promoting cell viability and invasion. J Hematol Oncol 2015. [PMID: 26198430 PMCID: PMC4511541 DOI: 10.1186/s13045-015-0187-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Gliomas are the most common types of primary brain tumors in the adult central nervous system. TMEM140 is identified as an amplified gene in the human gastric cancer genome. However, the function of TMEM140 in gliomas has not been thoroughly elucidated. The aim of the current study was to determine the clinical significance of TMEM140 expression in patients with gliomas and its effect on tumor cell malignant phenotypes. Methods Immunohistochemical analysis and real-time reverse transcription PCR were performed to detect the expression levels of TMEM140 in 70 glioma brain tissue samples. Next, the correlation between the TMEM140 expression levels and the clinical characteristics and outcomes of glioma patients was statistically analyzed. TMEM140 expression was inhibited in two glioma cell lines (i.e., U87 and U373) using a knockdown method with small interfering RNA. Cell Counting Kit-8 and Transwell assays were used to investigate TMEM140 function during cell proliferation, invasion, and migration, respectively. Using flow cytometry and Western blot analysis, we subsequently determined the cell cycle and apoptosis profile of the TMEM140-silenced cells. Results TMEM140 protein expression was significantly higher in gliomas than in normal brain tissues (p < 0.0001). TMEM140 overexpression was strongly correlated with tumor size, histologic grade, and overall survival time (P < 0.05). TMEM140 decreased cell viability in vitro and dramatically decreased tumor volume in vivo. This phenomenon might be caused by G1 phase cell cycle arrest and cell apoptosis. TMEM140 silencing could suppress the viability, migration, and invasion of glioma cells. Conclusions Our results suggest that TMEM140 expression is a prognostic factor that might play an important role in the viability, migration, and invasion of glioma cells. This study highlights the importance of TMEM140 as a novel prognostic marker and as an attractive therapeutic target for gliomas.
Collapse
Affiliation(s)
- Bin Li
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Ming-Zhu Huang
- Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiao-Qiang Wang
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Bang-Bao Tao
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jun Zhong
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Xu-Hui Wang
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Wen-Chuan Zhang
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Shi-Ting Li
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
194
|
Das A, Wei G, Parikh K, Liu D. Selective inhibitors of nuclear export (SINE) in hematological malignancies. Exp Hematol Oncol 2015; 4:7. [PMID: 25745591 PMCID: PMC4350974 DOI: 10.1186/s40164-015-0002-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/03/2015] [Indexed: 11/10/2022] Open
Abstract
Regulated nucleo-cytoplasmic transport plays a major role in maintaining cellular homeostasis. CRM1 (chromosome region maintenance 1 or exportin 1 or XPO 1) is responsible for the nucleo-cytoplasmic transport of more than 200 proteins, including most of the tumor suppressor proteins (TSP). CRM1 is overexpressed in pancreatic cancer, osteosarcoma, glioma, cervical and hematological malignancies. This inspired the development of novel agents that selectively inhibit nuclear exportins (SINEs). In this review we focus on the significance of CRM1 in carcinogenesis and review the new development of SINE inhibitiors in hematological malignancies. Selinexor (KPT-330) as the first-in-human SINE agent represents this novel class of anti-cancer agents.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Medicine, Westchester Medical Center, Valhalla, NY 10595 USA
| | - Guoqing Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaushal Parikh
- Department of Medicine, Westchester Medical Center, Valhalla, NY 10595 USA
| | - Delong Liu
- Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|