Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 23(6): 1552-1563, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-27649553

ABSTRACT

Purpose: The high fatality-to-case ratio of ovarian cancer is directly related to platinum resistance. Exportin-1 (XPO1) is a nuclear exporter that mediates nuclear export of multiple tumor suppressors. We investigated possible clinicopathologic correlations of XPO1 expression levels and evaluated the efficacy of XPO1 inhibition as a therapeutic strategy in platinum-sensitive and -resistant ovarian cancer.Experimental Design: XPO1 expression levels were analyzed to define clinicopathologic correlates using both TCGA/GEO datasets and tissue microarrays (TMA). The effect of XPO1 inhibition, using the small-molecule inhibitors KPT-185 and KPT-330 (selinexor) alone or in combination with a platinum agent on cell viability, apoptosis, and the transcriptome was tested in immortalized and patient-derived ovarian cancer cell lines (PDCL) and platinum-resistant mice (PDX). Seven patients with late-stage, recurrent, and heavily pretreated ovarian cancer were treated with an oral XPO1 inhibitor.Results: XPO1 RNA overexpression and protein nuclear localization were correlated with decreased survival and platinum resistance in ovarian cancer. Targeted XPO1 inhibition decreased cell viability and synergistically restored platinum sensitivity in both immortalized ovarian cancer cells and PDCL. The XPO1 inhibitor-mediated apoptosis occurred through both p53-dependent and p53-independent signaling pathways. Selinexor treatment, alone and in combination with platinum, markedly decreased tumor growth and prolonged survival in platinum-resistant PDX and mice. In selinexor-treated patients, tumor growth was halted in 3 of 5 patients, including one with a partial response, and was safely tolerated by all.Conclusions: Taken together, these results provide evidence that XPO1 inhibition represents a new therapeutic strategy for overcoming platinum resistance in women with ovarian cancer. Clin Cancer Res; 23(6); 1552-63. ©2016 AACR.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Karyopherins/genetics , Ovarian Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/genetics , Acrylates/administration & dosage , Active Transport, Cell Nucleus/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Hydrazines/administration & dosage , Karyopherins/antagonists & inhibitors , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Platinum/administration & dosage , Platinum/adverse effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/administration & dosage , Xenograft Model Antitumor Assays , Exportin 1 Protein
2.
J Clin Oncol ; 34(34): 4142-4150, 2016 12.
Article in English | MEDLINE | ID: mdl-26926685

ABSTRACT

Purpose This trial evaluated the safety, pharmacokinetics, pharmacodynamics, and efficacy of selinexor (KPT-330), a novel, oral small-molecule inhibitor of exportin 1 (XPO1/CRM1), and determined the recommended phase II dose. Patients and Methods In total, 189 patients with advanced solid tumors received selinexor (3 to 85 mg/m2) in 21- or 28-day cycles. Pre- and post-treatment levels of XPO1 mRNA in patient-derived leukocytes were determined by reverse transcriptase quantitative polymerase chain reaction, and tumor biopsies were examined by immunohistochemistry for changes in markers consistent with XPO1 inhibition. Antitumor response was assessed according Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 guidelines. Results The most common treatment-related adverse events included fatigue (70%), nausea (70%), anorexia (66%), and vomiting (49%), which were generally grade 1 or 2. Most commonly reported grade 3 or 4 toxicities were thrombocytopenia (16%), fatigue (15%), and hyponatremia (13%). Clinically significant major organ or cumulative toxicities were rare. The maximum-tolerated dose was defined at 65 mg/m2 using a twice-a-week (days 1 and 3) dosing schedule. The recommended phase II dose of 35 mg/m2 given twice a week was chosen based on better patient tolerability and no demonstrable improvement in radiologic response or disease stabilization compared with higher doses. Pharmacokinetics were dose proportional, with no evidence of drug accumulation. Dose-dependent elevations in XPO1 mRNA in leukocytes were demonstrated up to a dose level of 28 mg/m2 before plateauing, and paired tumor biopsies showed nuclear accumulation of key tumor-suppressor proteins, reduction of cell proliferation, and induction of apoptosis. Among 157 patients evaluable for response, one complete and six partial responses were observed (n = 7, 4%), with 27 patients (17%) achieving stable disease for ≥ 4 months. Conclusion Selinexor is a novel and safe therapeutic with broad antitumor activity. Further interrogation into this class of therapy is warranted.


Subject(s)
Hydrazines/pharmacology , Neoplasms/drug therapy , Triazoles/pharmacology , Active Transport, Cell Nucleus/drug effects , Adult , Aged , Aged, 80 and over , Biopsy , Drug Administration Schedule , Female , Humans , Hydrazines/administration & dosage , Immunohistochemistry , Male , Middle Aged , Neoplasms/pathology , Research Design , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome , Triazoles/administration & dosage
4.
Mol Cancer Ther ; 15(1): 60-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26637366

ABSTRACT

Exportin1 (XPO1; also known as chromosome maintenance region 1, or CRM1) controls nucleo-cytoplasmic transport of most tumor suppressors and is overexpressed in many cancers, including multiple myeloma, functionally impairing tumor suppressive function via target mislocalization. Selective inhibitor of nuclear export (SINE) compounds block XPO1-mediated nuclear escape by disrupting cargo protein binding, leading to retention of tumor suppressors, induction of cancer cell death, and sensitization to other drugs. Combined treatment with the clinical stage SINE compound selinexor and the irreversible proteasome inhibitor (PI) carfilzomib induced synergistic cell death of myeloma cell lines and primary plasma cells derived from relapsing/refractory myeloma patients and completely impaired the growth of myeloma cell line-derived tumors in mice. Investigating the details of SINE/PI-induced cell death revealed (i) reduced Bcl-2 expression and cleavage and inactivation of Akt, two prosurvival regulators of apoptosis and autophagy; (ii) intracellular membrane-associated aggregation of active caspases, which depended on caspase-10 protease activity; and (iii) novel association of caspase-10 and autophagy-associated proteins p62 and LC3 II, which may prime activation of the caspase cascade. Overall, our findings provide novel mechanistic rationale behind the potent cell death induced by combining selinexor with carfilzomib and support their use in the treatment of relapsed/refractory myeloma and potentially other cancers.


Subject(s)
Apoptosis/drug effects , Caspase 10/metabolism , Hydrazines/pharmacology , Multiple Myeloma/metabolism , Oligopeptides/pharmacology , Triazoles/pharmacology , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Enzyme Activation/drug effects , Humans , Intracellular Space , Mice , Proteasome Inhibitors/pharmacology , Xenograft Model Antitumor Assays
5.
BMC Cancer ; 15: 941, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620414

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.


Subject(s)
Analgesics/administration & dosage , Cyclin D1/metabolism , Hydrazines/administration & dosage , Inhibitor of Apoptosis Proteins/metabolism , Prostatic Neoplasms/drug therapy , Triazoles/administration & dosage , Adult , Analgesics/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrazines/pharmacology , Male , Mice , Mice, Nude , Neoplasm Grading , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacology , Prostatic Neoplasms/metabolism , Survivin , Triazoles/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 21(14): 3286-97, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25878333

ABSTRACT

PURPOSE: XPO1 inhibitors have shown promise for cancer treatment, and yet the underlying mechanisms for the antitumor effects are not well understood. In this study, we explored the usefulness of selective inhibitors of nuclear export (SINE) compounds that are specific inhibitors of XPO1. EXPERIMENTAL DESIGN: We used proteomic analysis in XPO1 inhibitor-treated ovarian cancer cell lines and examined antitumor effects in ovarian and breast cancer mouse models. We also studied the effects of XPO1 inhibitor in combination with chemotherapeutic agents. RESULTS: XPO1 inhibitor treatment substantially increased the percentage of apoptotic cells (60%) after 72 hours of incubation. XPO1 inhibitor promoted the accumulation of eIF5A in mitochondria, leading to cancer cell death. Topotecan showed the greatest synergistic effect with XPO1 inhibitor. XPO1 inhibitors prevented the translocation of IGF2BP1 from the nucleus to the cytoplasm, thereby permitting the localization of eIF5A in the mitochondria. This process was p53, RB, and FOXO independent. Significant antitumor effects were observed with XPO1 inhibitor monotherapy in orthotopic ovarian (P < 0.001) and breast (P < 0.001) cancer mouse models, with a further decrease in tumor burden observed in combination with topotecan or paclitaxel (P < 0.05). This mitochondrial accumulation of eIF5A was highly dependent on the cytoplasmic IGF2BP1 levels. CONCLUSIONS: We have unveiled a new understanding of the role of eIF5A and IGF2BP1 in XPO1 inhibitor-mediated cell death and support their clinical development for the treatment of ovarian and other cancers. Our data also ascertain the combinations of XPO1 inhibitors with specific chemotherapy drugs for therapeutic trials.


Subject(s)
Antineoplastic Agents/pharmacology , Karyopherins/antagonists & inhibitors , Mammary Neoplasms, Experimental/metabolism , Mitochondria/metabolism , Ovarian Neoplasms/metabolism , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Nude , Proteomics , RNA, Small Interfering , Signal Transduction/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Transfection , Xenograft Model Antitumor Assays , Eukaryotic Translation Initiation Factor 5A , Exportin 1 Protein
7.
Nat Neurosci ; 18(4): 511-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25706475

ABSTRACT

Axonal damage has been associated with aberrant protein trafficking. We examined a newly characterized class of compounds that target nucleo-cytoplasmic shuttling by binding to the catalytic groove of the nuclear export protein XPO1 (also known as CRM1, chromosome region maintenance protein 1). Oral administration of reversible CRM1 inhibitors in preclinical murine models of demyelination significantly attenuated disease progression, even when started after the onset of paralysis. Clinical efficacy was associated with decreased proliferation of immune cells, characterized by nuclear accumulation of cell cycle inhibitors, and preservation of cytoskeletal integrity even in demyelinated axons. Neuroprotection was not limited to models of demyelination, but was also observed in another mouse model of axonal damage (that is, kainic acid injection) and detected in cultured neurons after knockdown of Xpo1, the gene encoding CRM1. A proteomic screen for target molecules revealed that CRM1 inhibitors in neurons prevented nuclear export of molecules associated with axonal damage while retaining transcription factors modulating neuroprotection.


Subject(s)
Axons , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Karyopherins/metabolism , Neuroprotective Agents/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Acrylamides/administration & dosage , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Female , Karyopherins/antagonists & inhibitors , Karyopherins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Proteomics , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Treatment Outcome , Exportin 1 Protein
8.
Neuro Oncol ; 17(5): 697-707, 2015 May.
Article in English | MEDLINE | ID: mdl-25366336

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is poorly responsive to current chemotherapy. The nuclear transporter exportin 1 (XPO1, CRM1) is often highly expressed in GBM, which may portend a poor prognosis. Here, we determine the efficacy of novel selective inhibitors of nuclear export (SINE) specific to XPO1 in preclinical models of GBM. METHODS: Seven patient-derived GBM lines were treated with 3 SINE compounds (KPT-251, KPT-276, and Selinexor) in neurosphere culture conditions. KPT-276 and Selinexor were also evaluated in a murine orthotopic patient-derived xenograft (PDX) model of GBM. Cell cycle effects were assayed by flow cytometry in vitro and immunohistochemistry in vivo. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and caspase 3/7 activity assays. RESULTS: Treatment of GBM neurosphere cultures with KPT-276, Selinexor, and KPT-251 revealed dose-responsive growth inhibition in all 7 GBM lines [range of half-maximal inhibitory concentration (IC50), 6-354 nM]. In an orthotopic PDX model, treatment with KPT-276 and Selinexor demonstrated pharmacodynamic efficacy, significantly suppressed tumor growth, and prolonged animal survival. Cellular proliferation was not altered with SINE treatment. Instead, induction of apoptosis was apparent both in vitro and in vivo with SINE treatment, without overt evidence of neurotoxicity. CONCLUSIONS: SINE compounds show preclinical efficacy utilizing in vitro and in vivo models of GBM, with induction of apoptosis as the mechanism of action. Selinexor is now in early clinical trials in solid and hematological malignancies. Based on these preclinical data and excellent brain penetration, we have initiated clinical trials of Selinexor in patients with relapsed GBM.


Subject(s)
Acrylamides/therapeutic use , Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hydrazines/therapeutic use , Karyopherins/antagonists & inhibitors , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Thiazoles/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hydrazines/pharmacokinetics , Hydrazines/pharmacology , Karyopherins/metabolism , Macaca fascicularis , Male , Mice , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Treatment Outcome , Triazoles/pharmacokinetics , Xenograft Model Antitumor Assays , Exportin 1 Protein
9.
J Hematol Oncol ; 7: 85, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25476752

ABSTRACT

Shuttling of specific proteins out of the nucleus is essential for the regulation of the cell cycle and proliferation of both normal and malignant tissues. Dysregulation of this fundamental process may affect many other important cellular processes such as tumor growth, inflammatory response, cell cycle, and apoptosis. It is known that XPO1 (Exportin-1/Chromosome Region Maintenance 1/CRM1) is the main mediator of nuclear export in many cell types. Nuclear proteins exported to the cytoplasm by XPO1 include the drug targets topoisomerase IIα (topo IIα) and BCR-ABL and tumor suppressor proteins such as Rb, APC, p53, p21, and p27. XPO1 can mediate cell proliferation through several pathways: (i) the sub-cellular localization of NES-containing oncogenes and tumor suppressor proteins, (ii) the control of the mitotic apparatus and chromosome segregation, and (iii) the maintenance of nuclear and chromosomal structures. The XPO1 protein is elevated in ovarian carcinoma, glioma, osteosarcoma, pancreatic and cervical cancer. There is a growing body of research indicating that XPO1 may have an important role as a prognostic marker in solid tumors. Because of this, nuclear export inhibition through XPO1 is a potential target for therapeutic intervention in many cancers. The best understood XPO1 inhibitors are the small molecule nuclear export inhibitors (NEIs; Leptomycin B and derivatives, ratjadones, PKF050-638, valtrate, ACA, CBS9106, selinexor/KPT-330, and verdinexor/KPT-335). Selinexor and verdinexor are orally bioavailable, highly potent, small molecules that are classified as Selective Inhibitors of Nuclear Export (SINE). KPT-330 is the only NEI currently in Phase I/II human clinical trials in hematological and solid cancers. Of all the potential targets in nuclear cytoplasmic transport, the nuclear export receptor XPO1 remains the best understood and most advanced therapeutic target for the treatment of cancer.


Subject(s)
Active Transport, Cell Nucleus/physiology , Antineoplastic Agents/pharmacology , Karyopherins/metabolism , Neoplasms/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Exportin 1 Protein
10.
J Hematol Oncol ; 7: 46, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25284315

ABSTRACT

BACKGROUND: Exportin 1 (XPO1), also called chromosome region maintenance 1 (CRM1), is the sole exportin mediating transport of many multiple tumor suppressor proteins out of the nucleus. AIM AND METHODS: To verify the hypothesis that XPO1 inhibition affects prostate cancer (PCa) metastatic potential, orally available, potent and selective, SINE compounds, Selinexor (KPT- 330) and KPT-251, were tested in preclinical models known to generate bone lesions and systemic tumor spread. RESULTS: In vitro, Selinexor reduced both secretion of proteases and ability to migrate and invade of PCa cells. SINEs impaired secretion of pro-angiogenic and pro-osteolytic cytokines and reduced osteoclastogenesis in RAW264.7 cells. In the intra-prostatic growth model, Selinexor reduced DU145 tumor growth by 41% and 61% at the doses of 4 mg/Kg qd/5 days and 10 mg/Kg q2dx3 weeks, respectively, as well as the incidence of macroscopic visceral metastases. In a systemic metastasis model, following intracardiac injection of PCb2 cells, 80% (8/10) of controls, 10% (1/10) Selinexor- and 20% (2/10) KPT-251-treated animals developed radiographic evidence of lytic bone lesions. Similarly, after intra-tibial injection, the lytic areas were higher in controls than in Selinexor and KPT-251 groups. Analogously, the serum levels of osteoclast markers (mTRAP and type I collagen fragment, CTX), were significantly higher in controls than in Selinexor- and KPT-251-treated animals. Importantly, overall survival and disease-free survival were significantly higher in Selinexor- and KPT-251-treated animals when compared to controls. CONCLUSIONS: Selective blockade of XPO1-dependent nuclear export represents a completely novel approach for the treatment of advanced and metastatic PCa.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/pharmacology , Karyopherins/metabolism , Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement , Humans , Hydrazines/pharmacology , Male , Mice, SCID , Osteoblasts/drug effects , Prostatic Neoplasms/metabolism , Triazoles/pharmacology , Xenograft Model Antitumor Assays , Exportin 1 Protein
11.
BMC Vet Res ; 10: 160, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25022346

ABSTRACT

BACKGROUND: Exportin 1 (XPO1, also known as CRM1), is a chaperone protein responsible for the export of over 200 target proteins out of the nucleus. The expression and activity of XPO1 is upregulated in several human cancers and its expression is also linked to the development of chemotherapy resistance. Recent studies using both human and murine cancer cell lines have demonstrated that XPO1 is a relevant target for therapeutic intervention. The present study sought to characterize the biologic activity of an orally bioavailable selective inhibitor of nuclear export (SINE), KPT-335, against canine melanoma cell lines as a prelude to future clinical trials in dogs with melanoma. RESULTS: We evaluated the effects of KPT-335 on 4 canine malignant melanoma cell lines and found that KPT-335 inhibited proliferation, blocked colony formation, and induced apoptosis of treated cells at biologically relevant concentrations of drug. Additionally, KPT-335 downregulated XPO1 protein while inducing a concomitant increase in XPO1 messenger RNA. Lastly, KPT-335 treatment of cell lines upregulated the expression of both protein and mRNA for the tumor suppressor proteins p53 and p21, and promoted their nuclear localization. CONCLUSIONS: KPT-335 demonstrates biologic activity against canine melanoma cell lines at physiologically relevant doses, suggesting that KPT-335 may represent a viable treatment option for dogs with malignant melanoma.


Subject(s)
Acrylamides/pharmacology , Biological Availability , Dogs , Hydrazines/pharmacology , Melanoma , Acrylamides/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Hydrazines/administration & dosage , Karyopherins/genetics , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Exportin 1 Protein
12.
J Virol ; 88(17): 10228-43, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965445

ABSTRACT

UNLABELLED: Influenza is a global health concern, causing death, morbidity, and economic losses. Chemotherapeutics that target influenza virus are available; however, rapid emergence of drug-resistant strains is common. Therapeutic targeting of host proteins hijacked by influenza virus to facilitate replication is an antiviral strategy to reduce the development of drug resistance. Nuclear export of influenza virus ribonucleoprotein (vRNP) from infected cells has been shown to be mediated by exportin 1 (XPO1) interaction with viral nuclear export protein tethered to vRNP. RNA interference screening has identified XPO1 as a host proinfluenza factor where XPO1 silencing results in reduced influenza virus replication. The Streptomyces metabolite XPO1 inhibitor leptomycin B (LMB) has been shown to limit influenza virus replication in vitro; however, LMB is toxic in vivo, which makes it unsuitable for therapeutic use. In this study, we tested the anti-influenza virus activity of a new class of orally available small-molecule selective inhibitors of nuclear export, specifically, the XPO1 antagonist KPT-335 (verdinexor). Verdinexor was shown to potently and selectively inhibit vRNP export and effectively inhibited the replication of various influenza virus A and B strains in vitro, including pandemic H1N1 virus, highly pathogenic H5N1 avian influenza virus, and the recently emerged H7N9 strain. In vivo, prophylactic and therapeutic administration of verdinexor protected mice against disease pathology following a challenge with influenza virus A/California/04/09 or A/Philippines/2/82-X79, as well as reduced lung viral loads and proinflammatory cytokine expression, while having minimal toxicity. These studies show that verdinexor acts as a novel anti-influenza virus therapeutic agent. IMPORTANCE: Antiviral drugs represent important means of influenza virus control. However, substantial resistance to currently approved influenza therapeutic drugs has developed. New antiviral approaches are required to address drug resistance and reduce the burden of influenza virus-related disease. This study addressed critical preclinical studies for the development of verdinexor (KPT-335) as a novel antiviral drug. Verdinexor blocks progeny influenza virus genome nuclear export, thus effectively inhibiting virus replication. Verdinexor was found to limit the replication of various strains of influenza A and B viruses, including a pandemic H1N1 influenza virus strain, a highly pathogenic H5N1 avian influenza virus strain, and a recently emerging H7N9 influenza virus strain. Importantly, oral verdinexor treatments, given prophylactically or therapeutically, were efficacious in limiting lung virus burdens in influenza virus-infected mice, in addition to limiting lung proinflammatory cytokine expression, pathology, and death. Thus, this study demonstrated that verdinexor is efficacious against influenza virus infection in vitro and in vivo.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antiviral Agents/metabolism , Enzyme Inhibitors/metabolism , Influenza A virus/physiology , Influenza B virus/physiology , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/therapeutic use , Cell Line , Chemoprevention/methods , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Female , Humans , Influenza A virus/drug effects , Influenza B virus/drug effects , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Exportin 1 Protein
13.
PLoS One ; 9(2): e87585, 2014.
Article in English | MEDLINE | ID: mdl-24503695

ABSTRACT

BACKGROUND: The purpose of this study was to evaluate the activity of Selective Inhibitors of Nuclear Export (SINE) compounds that inhibit the function of the nuclear export protein Exportin 1 (XPO1/CRM1) against canine tumor cell lines and perform a Phase I clinical trial of KPT-335 in dogs with spontaneous cancer to provide a preliminary assessment of biologic activity and tolerability. METHODS AND FINDINGS: Canine tumor cell lines derived from non-Hodgkin lymphoma (NHL), mast cell tumor, melanoma and osteosarcoma exhibited growth inhibition and apoptosis in response to nanomolar concentrations of SINE compounds; NHL cells were particularly sensitive with IC50 concentrations ranging from 2-42 nM. A Phase I clinical trial of KPT-335 was performed in 17 dogs with NHL (naive or relapsed), mast cell tumor or osteosarcoma. The maximum tolerated dose was 1.75 mg/kg given orally twice/week (Monday/Thursday) although biologic activity was observed at 1 mg/kg. Clinical benefit (CB) including partial response to therapy (PR, n = 2) and stable disease (SD, n = 7) was observed in 9/14 dogs with NHL with a median time to progression (TTP) for responders of 66 days (range 35-256 days). A dose expansion study was performed in 6 dogs with NHL given 1.5 mg/kg KPT-335 Monday/Wednesday/Friday; CB was observed in 4/6 dogs with a median TTP for responders of 83 days (range 35-354 days). Toxicities were primarily gastrointestinal consisting of anorexia, weight loss, vomiting and diarrhea and were manageable with supportive care, dose modulation and administration of low dose prednisone; hepatotoxicity, anorexia and weight loss were the dose limiting toxicities. CONCLUSIONS: This study provides evidence that the novel orally bioavailable XPO1 inhibitor KPT-335 is safe and exhibits activity in a relevant, spontaneous large animal model of cancer. Data from this study provides critical new information that lays the groundwork for evaluation of SINE compounds in human cancer.


Subject(s)
Acrylamides/pharmacokinetics , Acrylamides/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Nucleus/metabolism , Drug Evaluation, Preclinical , Hydrazines/pharmacokinetics , Hydrazines/therapeutic use , Neoplasms/drug therapy , Neoplasms/veterinary , Acrylamides/administration & dosage , Acrylamides/adverse effects , Active Transport, Cell Nucleus/drug effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biological Availability , Cell Line, Tumor , Cell Nucleus/drug effects , Dogs , Dose-Response Relationship, Drug , Female , Humans , Hydrazines/administration & dosage , Hydrazines/adverse effects , Inhibitory Concentration 50 , Male , Quality of Life
14.
Mol Cancer Ther ; 13(3): 675-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24431073

ABSTRACT

Inhibition of XPO1 (CRM1)-mediated nuclear export of multiple tumor suppressor proteins has been proposed as a novel cancer therapeutic strategy to turn off oncogenic signals and enhance tumor suppression. Survivin is a multifunctional protein with oncogenic properties when expressed in the cytoplasm that requires the XPO1-RanGTP complex for its nuclear export. We investigated the antitumor mechanisms of the drug-like selective inhibitors of nuclear export (SINE) XPO1 antagonists KPT-185, KPT-251 KPT-276, and KPT-330 in estrogen receptor-positive and triple-negative breast cancer (TNBC) cell lines and xenograft models of human breast tumors. KPT compounds significantly inhibited breast cancer cell growth and induced tumor cell death, both in vitro and in vivo. These drugs initially promoted survivin accumulation within tumor cell nuclei. However, their major in vitro effect was to decrease survivin cytoplasmic protein levels, correlating with the onset of apoptosis. XPO1 inhibition repressed Survivin transcription by inhibiting CREB-binding protein-mediated STAT3 acetylation, and blocking STAT3 binding to the Survivin promoter. In addition, caspase-3 was activated to cleave survivin, rendering it unavailable to bind X-linked inhibitor of apoptosis protein and block the caspase cascade. Collectively, these data demonstrate that XPO1 inhibition by SINE compounds represses STAT3 transactivation to block the selective oncogenic properties of survivin and supports their clinical use in TNBC.


Subject(s)
Inhibitor of Apoptosis Proteins/genetics , Karyopherins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , STAT3 Transcription Factor/genetics , Triple Negative Breast Neoplasms/genetics , Active Transport, Cell Nucleus/genetics , Apoptosis/drug effects , Caspase 3 , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Female , Genetic Therapy , Humans , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Survivin , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Exportin 1 Protein
15.
Haematologica ; 98(7): 1098-106, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23403316

ABSTRACT

The nuclear export protein chromosome maintenance region 1, found to be elevated in non-Hodgkin's lymphomas, controls localization of critical tumor suppressor proteins. Nuclear localization of tumor suppressor proteins is necessary for their cell surveillance function. However, their nuclear exclusion by chromosome maintenance region 1 renders them ineffective making this nuclear transporter an attractive therapeutic target. We have identified selective inhibitors of nuclear export that lock tumor suppressor proteins in the cell nucleus leading to apoptosis of lymphoid but not normal cells. Our inhibitors induce tumor suppressor protein nuclear retention-dependent growth inhibition and apoptosis in a panel of non-Hodgkin's lymphoma cell lines. Western blot of nuclear protein fraction and confocal microscopy analysis demonstrated retention of major tumor suppressor proteins in the cell nucleus. Co-immunoprecipitation studies showed disruption of the tumor suppressor protein-chromosome maintenance region 1 interaction. Small inhibitor RNA knockdown of two major tumor suppressor proteins, p53 in wild-type protein-53 and protein 73 in mutant-protein-53, abrogated inhibitor activity. Oral administration of related inhibitor at 75 and 150 mg/kg resulted in 65 and 70% tumor reduction, respectively and subcutaneous injections of inhibitor (25 and 75 mg/kg) resulted in 70 and 74% suppression of non-Hodgkin's lymphoma tumor growth with no toxicity; residual tumors showed activation of the protein 73 pathway. Our study verifies chromosome maintenance region 1 as a therapeutic target in non-Hodgkin's lymphoma, indicating that this nuclear export protein warrants further clinical investigations.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Karyopherins/antagonists & inhibitors , Karyopherins/metabolism , Lymphoma, Non-Hodgkin/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Acrylates/chemistry , Acrylates/pharmacology , Acrylates/therapeutic use , Active Transport, Cell Nucleus/physiology , Animals , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Non-Hodgkin/drug therapy , Mice , Mice, SCID , Treatment Outcome , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/therapeutic use , Tumor Cells, Cultured , Waldenstrom Macroglobulinemia/drug therapy , Waldenstrom Macroglobulinemia/metabolism , Xenograft Model Antitumor Assays/methods , Exportin 1 Protein
16.
Br J Haematol ; 161(1): 117-27, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23373539

ABSTRACT

This study explored the anti-leukaemic efficacy of novel irreversible inhibitors of the major nuclear export receptor, chromosome region maintenance 1 (CRM1, also termed XPO1). We found that these novel CRM1 antagonists, termed SINE (Selective Inhibitors of Nuclear Export), induced rapid apoptosis at low nanomolar concentrations in a panel of 14 human T-cell acute lymphoblastic leukaemia (T-ALL) cell lines representing different molecular subtypes of the disease. To assess in vivo anti-leukaemia cell activity, we engrafted immunodeficient mice intravenously with the human T-ALL MOLT-4 cells, which harbour activating mutations of NOTCH1 and NRAS as well as loss of function of the CDKN2A, PTEN and TP53 tumour suppressors and express a high level of oncogenic transcription factor TAL1. Importantly, we examined the in vivo anti-leukaemic efficacy of the clinical SINE compound KPT-330 against T-ALL and acute myeloid leukaemia (AML) cells. These studies demonstrated striking in vivo activity of KPT-330 against T-ALL and AML cells, with little toxicity to normal murine haematopoietic cells. Taken together, our results show that SINE CRM1 antagonists represent promising 'first-in-class' drugs with a novel mechanism of action and wide therapeutic index, and imply that drugs of this class show promise for the targeted therapy of T-ALL and AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Karyopherins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blood Cells/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Xenograft Model Antitumor Assays , Exportin 1 Protein
17.
Blood ; 120(23): 4621-34, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23034282

ABSTRACT

The nuclear export protein XPO1 is overexpressed in cancer, leading to the cytoplasmic mislocalization of multiple tumor suppressor proteins. Existing XPO1-targeting agents lack selectivity and have been associated with significant toxicity. Small molecule selective inhibitors of nuclear export (SINEs) were designed that specifically inhibit XPO1. Genetic experiments and X-ray structures demonstrate that SINE covalently bind to a cysteine residue in the cargo-binding groove of XPO1, thereby inhibiting nuclear export of cargo proteins. The clinical relevance of SINEs was explored in chronic lymphocytic leukemia (CLL), a disease associated with recurrent XPO1 mutations. Evidence is presented that SINEs can restore normal regulation to the majority of the dysregulated pathways in CLL both in vitro and in vivo and induce apoptosis of CLL cells with a favorable therapeutic index, with enhanced killing of genomically high-risk CLL cells that are typically unresponsive to traditional therapies. More importantly, SINE slows disease progression, and improves overall survival in the Eµ-TCL1-SCID mouse model of CLL with minimal weight loss or other toxicities. Together, these findings demonstrate that XPO1 is a valid target in CLL with minimal effects on normal cells and provide a basis for the development of SINEs in CLL and related hematologic malignancies.


Subject(s)
Acrylates/pharmacology , Karyopherins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Triazoles/pharmacology , Acrylates/chemistry , Acrylates/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Crystallography, X-Ray , Humans , Immunoblotting , Interleukin-10/metabolism , Interleukin-6/metabolism , Karyopherins/chemistry , Karyopherins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Mice, SCID , Mice, Transgenic , Microscopy, Confocal , Models, Molecular , Molecular Structure , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Triazoles/chemistry , Triazoles/metabolism , Exportin 1 Protein
18.
ACS Med Chem Lett ; 2(2): 97-101, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-24900286

ABSTRACT

We have discovered novel benzofuran-based S1P1 agonists with excellent in vitro potency and selectivity. 1-((4-(5-Benzylbenzofuran-2-yl)-3-fluorophenyl)methyl) azetidine-3-carboxylic acid (18) is a potent S1P1 agonist with >1000× selectivity over S1P3. It demonstrated a good in vitro ADME profile and excellent oral bioavailability across species. Dosed orally at 0.3 mg/kg, 18 significantly reduced blood lymphocyte counts 24 h postdose and demonstrated efficacy in a mouse EAE model of relapsing MS.

19.
Cancer Res ; 67(15): 7358-67, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671206

ABSTRACT

Inhibition of platelet derived growth factor (PDGF) can increase the efficacy of other cancer therapeutics, but the cellular mechanism is incompletely understood. We examined the cellular effects on tumor vasculature of a novel DNA oligonucleotide aptamer (AX102) that selectively binds PDGF-B. Treatment with AX102 led to progressive reduction of pericytes, identified by PDGF receptor beta, NG2, desmin, or alpha-smooth muscle actin immunoreactivity, in Lewis lung carcinomas. The decrease ranged from 35% at 2 days, 63% at 7 days, to 85% at 28 days. Most tumor vessels that lacked pericytes at 7 days subsequently regressed. Overall tumor vascularity decreased 79% over 28 days, without a corresponding decrease in tumor size. Regression of pericytes and endothelial cells led to empty basement membrane sleeves, which were visible at 7 days, but only 54% remained at 28 days. PDGF-B inhibition had a less pronounced effect on pancreatic islet tumors in RIP-Tag2 transgenic mice, where pericytes decreased 47%, vascularity decreased 38%, and basement membrane sleeves decreased 21% over 28 days. Taken together, these findings show that inhibition of PDGF-B signaling can lead to regression of tumor vessels, but the magnitude is tumor specific and does not necessarily retard tumor growth. Loss of pericytes in tumors is an expected direct consequence of PDGF-B blockade, but reduced tumor vascularity is likely to be secondary to pericyte regression.


Subject(s)
Aptamers, Nucleotide/pharmacology , Carcinoma, Lewis Lung/drug therapy , Endothelium, Vascular/pathology , Insulinoma/drug therapy , Pericytes/pathology , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , 3T3 Cells , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Insulinoma/blood supply , Insulinoma/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/prevention & control , Pericytes/drug effects , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism
20.
J Cell Physiol ; 207(2): 407-12, 2006 May.
Article in English | MEDLINE | ID: mdl-16419035

ABSTRACT

Platelet-derived growth factor-B (PDGF-B) has been implicated in the pathogenesis of proliferative retinopathies and other scarring disorders in the eye. In this study, we sought to test the therapeutic potential of an aptamer that selectively binds PDGF-B, ARC126, and its PEGylated derivative, ARC127. Both ARC126 and ARC127 blocked PDGF-B-induced proliferation of cultured fibroblasts with an IC50 of 4 nM. Pharmacokinetic studies in rabbits showed similar peak vitreous concentrations of approximately 110 microM after intravitreous injection of 1 mg of either ARC126 or ARC127, but the terminal half-life was longer for ARC127 (98 versus 43 h). Efficacy was tested in rho/PDGF-B transgenic mice that express PDGF-B in photoreceptors and develop severe proliferative retinopathy resulting in retinal detachment. Compared to eyes injected with 20 microg of scrambled aptamer in which five of six developed detachments (three total and two partial), eyes injected with ARC126 (no detachment in five of six and one partial detachment), or ARC127 (no detachment in six of six) had significantly fewer retinal detachments. They also showed a significant reduction in epiretinal membrane formation. These data demonstrate that a single intravitreous injection of an aptamer that specifically binds PDGF-B is able to significantly reduce epiretinal membrane formation and retinal detachment in rho/PDGF-B mice. These striking effects in an aggressive model of proliferative retinopathy suggest that ARC126 and ARC127 should be considered for treatment of diseases in which PDGF-B has been implicated, including ischemic retinopathies such as proliferative diabetic retinopathy, proliferative vitreoretinopathy (PVR), and choroidal neovascularization.


Subject(s)
Aptamers, Nucleotide/pharmacology , Proto-Oncogene Proteins c-sis/genetics , Retinal Diseases/drug therapy , 3T3 Cells , Animals , Aptamers, Nucleotide/pharmacokinetics , Cell Proliferation/drug effects , Disease Models, Animal , Epiretinal Membrane/drug therapy , Eye/drug effects , Eye/metabolism , Eye/pathology , Injections , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , Rabbits , Retina/drug effects , Retina/pathology , Retinal Detachment/drug therapy , Rhodopsin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...