XPO1 (CRM1) Inhibition Represses STAT3 Activation to Drive a Survivin-Dependent Oncogenic Switch in Triple-Negative Breast Cancer

Total Page:16

File Type:pdf, Size:1020Kb

XPO1 (CRM1) Inhibition Represses STAT3 Activation to Drive a Survivin-Dependent Oncogenic Switch in Triple-Negative Breast Cancer Published OnlineFirst January 15, 2014; DOI: 10.1158/1535-7163.MCT-13-0416 Molecular Cancer Cancer Biology and Signal Transduction Therapeutics XPO1 (CRM1) Inhibition Represses STAT3 Activation to Drive a Survivin-Dependent Oncogenic Switch in Triple-Negative Breast Cancer Yan Cheng1, Michael P. Holloway1, Kevin Nguyen1, Dilara McCauley2, Yosef Landesman2, Michael G. Kauffman2, Sharon Shacham2, and Rachel A. Altura1 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. Mol Cancer Ther; 13(3); 675–86. Ó2014 AACR. Introduction they are exported out of the nucleus. Therefore, inhibiting XPO1, one of the seven nuclear exportin proteins XPO1, leading to forced nuclear localization, accumula- required for all mammalian cells, shuttles approximately tion, and activation of tumor suppressor proteins, is 220 diverse proteins and some RNAs from the nucleus to considered a potential therapeutic target for anticancer the cytoplasm (1–4). XPO1 interacts with cargo proteins drug development. through a nuclear export sequence (NES) that is depen- Survivin is a multifunctional protein with its major dent on the small GTPase protein Ran in its GTP-bound oncogenic property being inhibition of caspase-depen- form, which binds to XPO1 together with export cargos dent apoptosis that it accomplishes, in part, by stabilizing (2). NES-containing cargo proteins include multiple tran- X-linked inhibitor of apoptosis (XIAP) in the cytoplasm of scription factors and tumor suppressor proteins that play tumor cells (9). Survivin is highly expressed in breast key roles in the growth and survival of cancer cells. tumor cells and is one of the genes profiled on OncoDx Interestingly, XPO1 is the sole nuclear export protein for and Mammoprint as a predictor of clinical response to the major tumor suppressor proteins (e.g., p53; ref. 5), therapy (10). Survivin export from the nucleus to the STAT3; ref. 6), survivin; ref. 7), FOXO3; ref. 8). Many of cytoplasm is mediated by the XPO1–Ran-GTP complex these proteins lose their tumor suppressor function when (7, 11); cytoplasmic localization is required for survivin’s antiapoptotic and tumor-promoting functions (12–14). Disruption of the survivin NES leads to enhanced sus- Authors' Affiliations: 1Division of Hematology-Oncology, Department of ceptibility to anticancer treatments (12, 14). One mecha- Pediatrics, The Warren Alpert Medical School at Brown University and Rhode Island Hospital, Providence, Rhode Island; and 2Karyopharm Ther- nism of targeting survivin’s selective cytoplasmic func- apeutics Inc., Natick, Massachusetts tion without affecting its antitumor nuclear effects could Note: Supplementary data for this article are available at Molecular Cancer be to inhibit its cytoplasmic export. Therapeutics Online (http://mct.aacrjournals.org/). STAT3 is a member of the Janus-activated kinase Corresponding Author: Rachel A. Altura, Department of Pediatrics, Rhode (JAK)/STAT that is constitutively activated in multiple Island Hospital, 593 Eddy Street, Providence, RI 02903. Phone: 401-444- cancer types (15), including triple-negative breast cancer 5171; Fax: 401-444-8845; E-mail: [email protected] (TNBC). STAT3 activation in these tumors leads to doi: 10.1158/1535-7163.MCT-13-0416 increased expression of antiapoptotic proteins, including Ó2014 American Association for Cancer Research. survivin (16, 17), and other proteins to enhance cell www.aacrjournals.org 675 Downloaded from mct.aacrjournals.org on October 3, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst January 15, 2014; DOI: 10.1158/1535-7163.MCT-13-0416 Cheng et al. proliferation, induce angiogenesis, and suppress immune inhibitors (KPT-185cis, KPT-185trans, KPT-276, KPT- responses. Thus, STAT3 is a potential high-yield target for 330) were obtained from Karyopharm Therapeutics, Inc. drug development to treat TNBC for which there are no A 10-mmol/L stock solution dissolved in dimethyl sulf- currently approved molecular therapies. Although sev- oxide was diluted in medium to the final concentrations eral small-molecule STAT3 inhibitors have been reported indicated. To inhibit protein translation, cells were treated (18–20), thus far none are in clinical trials due to pharma- with 20 mg/mL cyclohexamide (CHX; Sigma-Aldrich). To cokinetic and other problems. Interestingly, STAT3 has inhibit proteasome-mediated degradation, cells were several NES elements through which it binds to XPO1 for treated with 2 mmol/L MG132 (SelleckBio.com) for 18 its nuclear export (21). hours or 40 mmol/L MG132 for 6 hours. To inhibit cas- Leptomycin B (LMB) was the first natural XPO1 inhib- pase-3 and caspase-9, cells were treated with 20 mmol/L of itor discovered that was shown to be a potent anticancer inhibitors for 4 hours. Of note, 6Â MYC-survivin was a gift agent (22). This agent failed in early clinical trials due to from H. Cheung (University of Ottawa). Cells were trans- off-target side effects predictable from animal toxicity fected with siRNA to XPO1 (Ambion/Invitrogen), survi- testing, prompting the development of more selective and vin (Cell Signaling Technology), XIAP (Thermo Scientific less toxic XPO1 inhibitors (23). The recent crystal structure Dharmacon), or control (Thermo Scientific Dharmacon). of LMB and XPO1 shows that LMB binds covalently to Cys-528 in the XPO1 NES-binding groove, occupying the Cell viability and apoptosis assays majority of the groove and undergoing hydrolysis by Cells were seeded at a density of 5,000 cells/well then XPO1 (24). Newer small-molecule, selective inhibitors of treated at the doses indicated. Seventy hours later, cells nuclear export (SINE) XPO1 antagonists developed by were incubated with CellTiter 96 Aqueous One Solution Karyopharm Therapeutics, Inc. bind similarly in the NES Reagent (Promega), and viability was assessed using a groove; however, due to their smaller size, these mole- Synergy Mx Monochromator (BioTek Instrument Inc.). cules occupy less space and are more specific for XPO1, Flow cytometry-based apoptosis assays were performed with no detectable binding to other proteins (24). X-ray with the Annexin V-FITC Apoptosis Kit (Biovision), fol- crystal structures of SINEs bound to XPO1 have been lowing the manufacturer’s instructions. Multiwell plate published and confirm covalent modification of Cys528 apoptosis assays were performed with the Annexin V (24). SINEs have been demonstrated to reduce tumor FITC Assay Kit (Cayman Chemical). growth with good tolerability in several preclinical mod- els of hematologic cancers and solid tumors (25, 26). The IC50 determination exact molecular mechanism of their antitumor effects in Subconfluent cultures were grown in 96-well plates and different cancer subtypes is not yet well defined. treated with KPT compounds for 72 hours before perform- We sought to characterize the effects of XPO1 inhibition ing MTT assays with CellTiter 96 AQueous One Solution on survivin using a breast cancer model and the SINE Cell Proliferation Assay (Promega). Data were graphed compounds. In vitro and in vivo assays demonstrated that and analyzed using Kaleidagraph software (Synergy Soft- these drugs potently inhibit breast cancer growth by ware). Assay values obtained in triplicate were used to blocking proliferation and enhancing cell death path- derive the IC50 by curve fitting to the Hill equation. ways, as demonstrated for other tumor types. Important- ly, here we show that these compounds can dramatically Subcellular fractionation, immunoprecipitation, repress Survivin transcription by blocking STAT3 trans- Western blot analysis, and tricine-SDS-PAGE activation and that they enhance survivin degradation Subcellular fractionation was performed as previously through cleavage by caspase-3. These results identify described (27). Immunocomplexes were resolved by SDS- survivin- and STAT3-dependent mechanisms for the PAGE. Tricine-SDS-PAGE was carried out as described XPO1 inhibitors that may explain their in vivo antitumor (28). Anti-XPO1 (H-300): sc-5595, anti-survivin (D-8), anti- effects. We also note that XPO1 inhibitors force
Recommended publications
  • XPO1E571K Mutation Modifies Exportin 1 Localisation And
    cancers Article XPO1E571K Mutation Modifies Exportin 1 Localisation and Interactome in B-Cell Lymphoma Hadjer Miloudi 1, Élodie Bohers 1,2, François Guillonneau 3 , Antoine Taly 4,5 , Vincent Cabaud Gibouin 6,7 , Pierre-Julien Viailly 1,2 , Gaëtan Jego 6,7 , Luca Grumolato 8 , Fabrice Jardin 1,2 and Brigitte Sola 1,* 1 INSERM U1245, Unicaen, Normandie University, F-14000 Caen, France; [email protected] (H.M.); [email protected] (E.B.); [email protected] (P.-J.V.); [email protected] (F.J.) 2 Centre de lutte contre le Cancer Henri Becquerel, F-76000 Rouen, France 3 Plateforme Protéomique 3P5, Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; [email protected] 4 Laboratoire de Biochimie Théorique, CNRS UPR 9030, Université de Paris, F-75005 Paris, France; [email protected] 5 Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, F-75005 Paris, France 6 INSERM, LNC UMR1231, F-21000 Dijon, France; [email protected] (V.C.G.); [email protected] (G.J.) 7 Team HSP-Pathies, University of Burgundy and Franche-Comtée, F-21000 Dijon, France 8 INSERM U1239, Unirouen, Normandie University, F-76130 Mont-Saint-Aignan, France; [email protected] * Correspondence: [email protected]; Tel.: +33-2-3156-8210 Received: 11 September 2020; Accepted: 28 September 2020; Published: 30 September 2020 Simple Summary: Almost 25% of patients with either primary mediastinal B-cell lymphoma (PMBL) or classical Hodgkin lymphoma (cHL) possess a recurrent mutation of the XPO1 gene encoding the major nuclear export protein.
    [Show full text]
  • 1325.Full-Text.Pdf
    IN THIS ISSUE APC Mutation Position Dictates Effect of Tankyrase Inhibition in Colorectal Cancer • The effects of APC mutations, which in- • New animal models, human cells, and or- • Cases with different mutations in crease WNT signaling in colorectal can- ganoids were used to circumvent issues the same gene should be evaluated cer, can be reversed by TNKS inhibition . with mouse colorectal cancer models . separately for therapeutic response . Hyperactive WNT signaling is tumor growth in vivo. However, whether TNKS inhibition seen in most colorectal cancers, was effective depended on the mechanism of APC disrup- and inactivating mutations in the tion: APC mutants with truncations in the mutation cluster tumor suppressor adenomatous region were still able to regulate β-catenin and responded to polyposis coli (APC)—a scaffold TNKS blockade, whereas this was not the case when there protein mediating the formation were truncations earlier in the sequence. Truncations in the of the destruction complex (DC) mutation cluster region are commonly observed in patients, that facilitates β-catenin degrada- whereas the earlier truncations are present in commonly used tion—is the cause in 80% of such mouse models. Collectively, these results indicate that TNKS cases. Restoring DC activity (and, inhibition can restore control of WNT signaling in some thus, normal WNT signaling) in the context of inactivated APC-mutant cases and illustrate that different mutations in APC is possible through pharmacologic inhibition of tanky- the same gene, even those causing the same phenotype (in rase (TNKS) 1 and 2, which are functionally redundant. Using this case, WNT hyperactivation), can respond differently to APC-mutant animal models, human cells, and ex vivo orga- targeted therapies.
    [Show full text]
  • Inhibition of the Nuclear Export Receptor XPO1 As a Therapeutic Target for Platinum-Resistant Ovarian Cancer Ying Chen1, Sandra Catalina Camacho1, Thomas R
    Published OnlineFirst September 20, 2016; DOI: 10.1158/1078-0432.CCR-16-1333 Cancer Therapy: Preclinical Clinical Cancer Research Inhibition of the Nuclear Export Receptor XPO1 as a Therapeutic Target for Platinum-Resistant Ovarian Cancer Ying Chen1, Sandra Catalina Camacho1, Thomas R. Silvers1, Albiruni R.A. Razak2, Nashat Y. Gabrail3, John F. Gerecitano4, Eva Kalir1, Elena Pereira5, Brad R. Evans1, Susan J. Ramus6, Fei Huang1, Nolan Priedigkeit1, Estefania Rodriguez1, Michael Donovan7, Faisal Khan7, Tamara Kalir7, Robert Sebra1, Andrew Uzilov1, Rong Chen1, Rileen Sinha1, Richard Halpert8, Jean-Noel Billaud8, Sharon Shacham9, Dilara McCauley9, Yosef Landesman9, Tami Rashal9, Michael Kauffman9, Mansoor R. Mirza9, Morten Mau-Sørensen10, Peter Dottino5, and John A. Martignetti1,5,11 Abstract Purpose: The high fatality-to-case ratio of ovarian cancer is Results: XPO1 RNA overexpression and protein nuclear directly related to platinum resistance. Exportin-1 (XPO1) is a localization were correlated with decreased survival and plat- nuclear exporter that mediates nuclear export of multiple tumor inum resistance in ovarian cancer. Targeted XPO1 inhibition suppressors. We investigated possible clinicopathologic correla- decreased cell viability and synergistically restored platinum tions of XPO1 expression levels and evaluated the efficacy of sensitivity in both immortalized ovarian cancer cells and XPO1 inhibition as a therapeutic strategy in platinum-sensitive PDCL. The XPO1 inhibitor–mediated apoptosis occurred and -resistant ovarian cancer. through both p53-dependent and p53-independent signaling Experimental Design: XPO1 expression levels were analyzed to pathways. Selinexor treatment, alone and in combination with define clinicopathologic correlates using both TCGA/GEO data- platinum, markedly decreased tumor growth and prolonged sets and tissue microarrays (TMA).
    [Show full text]
  • KPT-350 (And Other XPO1 Inhibitors)
    Cognitive Vitality Reports® are reports written by neuroscientists at the Alzheimer’s Drug Discovery Foundation (ADDF). These scientific reports include analysis of drugs, drugs-in- development, drug targets, supplements, nutraceuticals, food/drink, non-pharmacologic interventions, and risk factors. Neuroscientists evaluate the potential benefit (or harm) for brain health, as well as for age-related health concerns that can affect brain health (e.g., cardiovascular diseases, cancers, diabetes/metabolic syndrome). In addition, these reports include evaluation of safety data, from clinical trials if available, and from preclinical models. KPT-350 (and other XPO1 inhibitors) Evidence Summary CNS penetrant XPO1 inhibitor with pleiotropic effects. May protect against neuroinflammation and proteotoxic stress, but similar drugs show a poor benefit to side effect profile in cancer. Neuroprotective Benefit: KPT-350 may reduce neuroinflammation and partially alleviate nucleocytoplasmic transport defects, but effects are pleiotropic and dependent on cellular and environmental conditions. Aging and related health concerns: XPO1 inhibition may promote autophagy, but has pleiotropic effects and is only marginally beneficial in cancer. Safety: KPT-350 has not been clinically tested. Tested XPO1 inhibitors are associated with myelosuppression, gastrointestinal events, anorexia, low blood sodium, and neurological events. Poor benefit to side effect ratio in cancer. 1 Availability: In clinical trials and Dose: Oral administration KPT-350 research use Dose not established for KPT-350 Chemical formula: Half-life: Unknown for KPT-350 BBB: KPT-350 is penetrant C18H17F6N5O2 6-7 hours for selinexor MW: 449.3 g/mol SINEs terminal half-life ~24 hours Clinical trials: None completed for Observational studies: Evidence for KPT-350. Phase 1 in ALS is ongoing.
    [Show full text]
  • Renal Medullary Carcinomas Depend Upon SMARCB1 Loss And
    RESEARCH ARTICLE Renal medullary carcinomas depend upon SMARCB1 loss and are sensitive to proteasome inhibition Andrew L Hong1,2,3, Yuen-Yi Tseng3, Jeremiah A Wala3, Won-Jun Kim2, Bryan D Kynnap2, Mihir B Doshi3, Guillaume Kugener3, Gabriel J Sandoval2,3, Thomas P Howard2, Ji Li2, Xiaoping Yang3, Michelle Tillgren2, Mahmhoud Ghandi3, Abeer Sayeed3, Rebecca Deasy3, Abigail Ward1,2, Brian McSteen4, Katherine M Labella2, Paula Keskula3, Adam Tracy3, Cora Connor5, Catherine M Clinton1,2, Alanna J Church1, Brian D Crompton1,2,3, Katherine A Janeway1,2, Barbara Van Hare4, David Sandak4, Ole Gjoerup2, Pratiti Bandopadhayay1,2,3, Paul A Clemons3, Stuart L Schreiber3, David E Root3, Prafulla C Gokhale2, Susan N Chi1,2, Elizabeth A Mullen1,2, Charles WM Roberts6, Cigall Kadoch2,3, Rameen Beroukhim2,3,7, Keith L Ligon2,3,7, Jesse S Boehm3, William C Hahn2,3,7* 1Boston Children’s Hospital, Boston, United States; 2Dana-Farber Cancer Institute, Boston, United States; 3Broad Institute of Harvard and MIT, Cambridge, United States; 4Rare Cancer Research Foundation, Durham, United States; 5RMC Support, North Charleston, United States; 6St. Jude Children’s Research Hospital, Memphis, United States; 7Brigham and Women’s Hospital, Boston, United States Abstract Renal medullary carcinoma (RMC) is a rare and deadly kidney cancer in patients of African descent with sickle cell trait. We have developed faithful patient-derived RMC models and using whole-genome sequencing, we identified loss-of-function intronic fusion events in one SMARCB1 allele with concurrent loss of the other allele. Biochemical and functional characterization of these models revealed that RMC requires the loss of SMARCB1 for survival.
    [Show full text]
  • Anticancer Activity by Inhibition of Nucleocytoplasmic Shuttling Fabio Conforti1, Yisong Wang1,2, Jose A
    Published OnlineFirst August 31, 2015; DOI: 10.1158/1078-0432.CCR-15-0408 Molecular Pathways Clinical Cancer Research Molecular Pathways: Anticancer Activity by Inhibition of Nucleocytoplasmic Shuttling Fabio Conforti1, Yisong Wang1,2, Jose A. Rodriguez3, Anna Teresa Alberobello1, Yu-Wen Zhang1, and Giuseppe Giaccone1 Abstract A dynamic distribution between nucleus and cytoplasm involved in the shuttling process, exportin XPO1, also known as (nucleocytoplasmic shuttling) is one of the control mechanisms chromosome region maintenance 1, appears to play a particularly adapted by normal cells to regulate the activity of a variety of prominent role in pathogenesis of both hematological malignan- molecules. Growing evidence suggests that dysregulation of the cies and solid tumors. Given the importance of nucleocytoplas- nucleocytoplasmic shuttling is involved in promoting abnormal mic shuttling in cancer pathogenesis and the rapidly expanding cell survival, tumor progression, and drug resistance, and is knowledge in this field, attempts have been made to develop associated with poor cancer prognosis. Aberrant nucleocytoplas- compounds able to revert the aberrant nucleocytoplasmic shut- mic shuttling in cancer cells may result from a hyperactive status of tling. A promising new drug, KPT-330 (Selinexor), which belongs diverse signal-transduction pathways, such as the PI3K–AKT and to the class of XPO1 inhibitors called selective inhibitors of MAPK pathways, or from alterations in the general nuclear nuclear export, is now being tested in phase I/II clinical trials. import/export machinery. Among the large number of molecules Clin Cancer Res; 21(20); 1–6. Ó2015 AACR. Background Nucleocytoplasmic shuttling dysregulation in cancer A dynamic subcellular compartmentalization via nucleocyto- Physical separation of the nucleus from the cytoplasm by the plasmic shuttling is one of the regulatory mechanisms used by nuclear envelope is a hallmark of eukaryotic cells.
    [Show full text]
  • A Member of the Ran-Binding Protein Family, Yrb2p, Is Involved in Nuclear Protein Export
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 7427–7432, June 1998 Cell Biology A member of the Ran-binding protein family, Yrb2p, is involved in nuclear protein export TETSUYA TAURA,HEIKE KREBBER, AND PAMELA A. SILVER* Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and The Dana–Farber Cancer Institute, 44 Binney Street, Boston, MA 02115 Communicated by David M. Livingston, Dana–Farber Cancer Institute, Boston, MA, April 27, 1998 (received for review February 18, 1998) ABSTRACT Yeast cells mutated in YRB2, which encodes will be a number of distinct ‘‘export pathways’’ defined by the a nuclear protein with similarity to other Ran-binding pro- type of cargo being transported. teins, fail to export nuclear export signal (NES)-containing The GTPase, Ran (13, 14) (Gsp1p in yeast), critically defines proteins including HIV Rev out of the nucleus. Unlike Xpo1py the movement of macromolecules in both directions across the Crm1pyexportin, an NES receptor, Yrb2p does not shuttle nuclear envelope. The asymmetric distribution of the major between the nucleus and the cytoplasm but instead remains Ran regulators may provide an important key to how Ran inside the nucleus. However, by both biochemical and genetic mediates both import and export. Ran in the GDP bound state criteria, Yrb2p interacts with Xpo1p and not with other is probably high in the cytoplasm because of the cytoplasmic members of the importinykaryopherin b superfamily. More- location of the RanGAP (Rna1p in yeast) (15). Conversely, over, the Yrb2p region containing nucleoporin-like FG repeats Ran-GTP would be the preferred state in the nucleus where is important for NES-mediated protein export.
    [Show full text]
  • Frequent Somatic Mutations in Components of the RNA Processing Machinery in Chronic Lymphocytic Leukemia
    Letters to the Editor 1600 CONFLICT OF INTEREST 2 Sokol L, Loughran TP Jr. Large granular lymphocyte leukemia. Oncologist 2006; 11: WK, CH, TH and SS are part owners of the MLL Munich Leukemia Laboratory. AF and 263–273. VG are employed by the MLL Munich Leukemia Laboratory. 3 Koskela HL, Eldfors S, Ellonen P, van Adrichem AJ, Kuusanmaki H, Andersson EI et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N Engl J Med 2012; 366: 1905–1913. ACKNOWLEDGEMENTS 4 Jerez A, Clemente MJ, Makishima H, Koskela H, Leblanc F, Ng KP et al. STAT3 mutations unify the pathogenesis of chronic lymphoproliferative dis- We thank all clinicians for sending samples to our laboratory for diagnostic purposes, orders of NK cells and T cell large granular lymphocyte leukemia. Blood 2012; 120: and for providing clinical information and follow-up data. In addition, we would like 3048–3057. to thank all co-workers at the MLL Munich Leukemia Laboratory for approaching 5 Kern W, Bacher U, Haferlach C, Alpermann T, Dicker F, Schnittger S et al. together many aspects in the field of leukemia diagnostics and research. In addition, Frequency and prognostic impact of the aberrant CD8 expression in 5,523 we are grateful for the data management support performed by Tamara Alpermann. patients with chronic lymphocytic leukemia. Cytometry B Clin Cytom 2012; 82: 145–150. A Fasan, W Kern, V Grossmann, C Haferlach, 6 van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M, Lavender FL T Haferlach and S Schnittger et al. Design and standardization of PCR primers and protocols for detection of MLL Munich Leukemia Laboratory, Munich, Germany clonal immunoglobulin and T-cell receptor gene recombinations in suspect lym- E-mail: [email protected] phoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936.
    [Show full text]
  • Microduplication in the 2P16.1P15 Chromosomal Region Linked To
    Lovrecic et al. Molecular Cytogenetics (2018) 11:39 https://doi.org/10.1186/s13039-018-0388-y CASE REPORT Open Access Microduplication in the 2p16.1p15 chromosomal region linked to developmental delay and intellectual disability Luca Lovrecic1* , Chiara Gnan2, Federica Baldan3, Alessandra Franzoni2, Sara Bertok4, Giuseppe Damante5, Bertrand Isidor6 and Borut Peterlin1 Abstract Background: Several patients with the 2p16.1p15 microdeletion syndrome have been reported. However, microduplication in the 2p16.1p15 chromosomal region has only been reported in one case, and milder clinical features were present compared to those attributed to 2p16.1p15 microdeletion syndrome. Some additional cases were deposited in DECIPHER database. Case presentation: In this report we describe four further cases of 2p16.1p15 microduplication in four unrelated probands. They presented with mild gross motor delay, delayed speech and language development, and mild dysmorphic features. In addition, two probands have macrocephaly and one a congenital heart anomaly. Newly described cases share several phenotype characteristics with those detailed in one previously reported microduplication case. Conclusion: The common features among patients are developmental delay, speech delay, mild to moderate intellectual disability and unspecific dysmorphic features. Two patients have bilateral clinodactyly of the 5th finger and two have bilateral 2nd-3rd toes syndactyly. Interestingly, as opposed to the deletion phenotype with some cases of microcephaly, 2 patients are reported
    [Show full text]
  • Inhibition of the Nuclear Export Receptor XPO1 As a Therapeutic Target for Platinum-Resistant Ovarian Cancer Ying Chen1, Sandra Catalina Camacho1, Thomas R
    Published OnlineFirst September 20, 2016; DOI: 10.1158/1078-0432.CCR-16-1333 Cancer Therapy: Preclinical Clinical Cancer Research Inhibition of the Nuclear Export Receptor XPO1 as a Therapeutic Target for Platinum-Resistant Ovarian Cancer Ying Chen1, Sandra Catalina Camacho1, Thomas R. Silvers1, Albiruni R.A. Razak2, Nashat Y. Gabrail3, John F. Gerecitano4, Eva Kalir1, Elena Pereira5, Brad R. Evans1, Susan J. Ramus6, Fei Huang1, Nolan Priedigkeit1, Estefania Rodriguez1, Michael Donovan7, Faisal Khan7, Tamara Kalir7, Robert Sebra1, Andrew Uzilov1, Rong Chen1, Rileen Sinha1, Richard Halpert8, Jean-Noel Billaud8, Sharon Shacham9, Dilara McCauley9, Yosef Landesman9, Tami Rashal9, Michael Kauffman9, Mansoor R. Mirza9, Morten Mau-Sørensen10, Peter Dottino5, and John A. Martignetti1,5,11 Abstract Purpose: The high fatality-to-case ratio of ovarian cancer is Results: XPO1 RNA overexpression and protein nuclear directly related to platinum resistance. Exportin-1 (XPO1) is a localization were correlated with decreased survival and plat- nuclear exporter that mediates nuclear export of multiple tumor inum resistance in ovarian cancer. Targeted XPO1 inhibition suppressors. We investigated possible clinicopathologic correla- decreased cell viability and synergistically restored platinum tions of XPO1 expression levels and evaluated the efficacy of sensitivity in both immortalized ovarian cancer cells and XPO1 inhibition as a therapeutic strategy in platinum-sensitive PDCL. The XPO1 inhibitor–mediated apoptosis occurred and -resistant ovarian cancer. through both p53-dependent and p53-independent signaling Experimental Design: XPO1 expression levels were analyzed to pathways. Selinexor treatment, alone and in combination with define clinicopathologic correlates using both TCGA/GEO data- platinum, markedly decreased tumor growth and prolonged sets and tissue microarrays (TMA).
    [Show full text]
  • XPO1 Inhibitor KPT-330 Synergizes with Bcl-Xl Inhibitor to Induce
    Zhu et al. Cell Death and Disease (2019) 10:395 https://doi.org/10.1038/s41419-019-1627-9 Cell Death & Disease ARTICLE Open Access XPO1 inhibitor KPT-330 synergizes with Bcl-xL inhibitor to induce cancer cell apoptosis by perturbing rRNA processing and Mcl-1 protein synthesis Zhi-Chuan Zhu1, Ji-Wei Liu2,CanYang1,3, Miao Zhao4 and Zhi-Qi Xiong1,3,5 Abstract XPO1 (exportin1) mediates nuclear export of proteins and RNAs and is frequently overexpressed in cancers. In this study, we show that the orally bioavailable XPO1 inhibitor KPT-330 reduced Mcl-1 protein level, by which it synergized with Bcl- xL inhibitor A-1331852 to induce apoptosis in cancer cells. KPT-330/A-1331852 combination disrupted bindings of Mcl-1 and Bcl-xL to Bax, Bak, and/or Bim, elicited mitochondrial outer membrane permeabilization, and triggered apoptosis. KPT- 330 generally mitigated mRNA expression and protein synthesis rather than mRNA nuclear export or protein stability of Mcl-1. KPT-330 inhibited mTORC1/4E-BP1 and Mnk1/eIF4E axes, which disrupted the eIF4F translation initiation complex but was dispensable for Mcl-1 reduction and KPT-330/A-1331852 combination-induced apoptosis. Mature rRNAs are integral components of the ribosome that determines protein synthesis ability. KPT-330 impeded nucleolar rRNA processing and reduced total levels of multiple mature rRNAs. Reconstitution of XPO1 by expressing degradation-resistant C528S mutant retained rRNA amount, Mcl-1 expression, and Bcl-xL inhibitor resistance upon KPT-330 treatment. KPT-330/ 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; A-1331852 combination suppressed growth and enhanced apoptosis of non-small cell lung cancer xenografts.
    [Show full text]
  • Renal Medullary Carcinomas Depend Upon SMARCB1 Loss and Are Sensitive to 2 Proteasome Inhibition 3 4 5 Andrew L
    bioRxiv preprint doi: https://doi.org/10.1101/487579; this version posted December 5, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Renal medullary carcinomas depend upon SMARCB1 loss and are sensitive to 2 proteasome inhibition 3 4 5 Andrew L. Hong1,2,3, Yuen-Yi Tseng3, Jeremiah Wala3, Won Jun Kim2, Bryan D. Kynnap2, 6 Mihir B. Doshi3, Guillaume Kugener3, Gabriel J. Sandoval2,3, Thomas P. Howard2, Ji Li2, 7 Xiaoping Yang3, Michelle Tillgren2, Mahmoud Ghandi3, Abeer Sayeed3, Rebecca Deasy3, 8 Abigail Ward1,2, Brian McSteen4, Katherine M. Labella2, Paula Keskula3, Adam Tracy3, 9 Cora Connor5, Catherine M. Clinton1,2, Alanna J. Church1, Brian D. Crompton1,2,3, 10 Katherine A. Janeway1,2, Barbara Van Hare4, David Sandak4, Ole Gjoerup2, 11 Pratiti Bandopadhayay1,2,3, Paul A. Clemons3, Stuart L. Schreiber3, David E. Root3, 12 Prafulla C. Gokhale2, Susan N. Chi1,2, Elizabeth A. Mullen1,2, Charles W. M. Roberts6, 13 Cigall Kadoch2,3, Rameen Beroukhim2,3,7, Keith L. Ligon2,3,7, Jesse S. Boehm3, 14 William C. Hahn2,3,7* 15 Affiliations: 16 1Boston Children’s Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA 17 2Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts, 02215 USA 18 3Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts, 02142 USA 19 4Rare Cancer Research Foundation, 112 S. Duke Street, Suite 101, Durham, NC 27701 USA 20 5RMC Support, P.O. Box 73156, North Charleston, S.C. 29415 USA 21 6St.
    [Show full text]