Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancer Res ; 80(23): 5344-5354, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33023948

ABSTRACT

High-dose chemotherapy with melphalan followed by autologous transplantation is a first-line treatment for multiple myeloma. Here, we present preclinical evidence that this treatment may be significantly improved by the addition of exportin 1 inhibitors (XPO1i). The XPO1i selinexor, eltanexor, and KOS-2464 sensitized human multiple myeloma cells to melphalan. Human 8226 and U266 multiple myeloma cell lines and melphalan-resistant cell lines (8226-LR5 and U266-LR6) were highly sensitized to melphalan by XPO1i. Multiple myeloma cells from newly diagnosed and relapsed/refractory multiple myeloma patients were also sensitized by XPO1i to melphalan. In NOD/SCIDγ mice challenged with either parental 8226 or U266 multiple myeloma and melphalan-resistant multiple myeloma tumors, XPO1i/melphalan combination treatments demonstrated stronger synergistic antitumor effects than single-agent melphalan with minimal toxicity. Synergistic cell death resulted from increased XPO1i/melphalan-induced DNA damage in a dose-dependent manner and decreased DNA repair. In addition, repair of melphalan-induced DNA damage was inhibited by selinexor, which decreased melphalan-induced monoubiquitination of FANCD2 in multiple myeloma cells. Knockdown of FANCD2 was found to replicate the effect of selinexor when used with melphalan, increasing DNA damage (γH2AX) by inhibiting DNA repair. Thus, combination therapies that include selinexor or eltanexor with melphalan may have the potential to improve treatment outcomes of multiple myeloma in melphalan-resistant and newly diagnosed patients. The combination of selinexor and melphalan is currently being investigated in the context of high-dose chemotherapy and autologous transplant (NCT02780609). SIGNIFICANCE: Inhibition of exportin 1 with selinexor synergistically sensitizes human multiple myeloma to melphalan by inhibiting Fanconi anemia pathway-mediated DNA repair.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Karyopherins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Animals , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Hydrazines/administration & dosage , Hydrazines/pharmacology , Karyopherins/metabolism , Melphalan/administration & dosage , Mice, Inbred NOD , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Nestin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Triazoles/administration & dosage , Triazoles/pharmacology , Ubiquitination/drug effects , Xenograft Model Antitumor Assays , Exportin 1 Protein
2.
Clin Cancer Res ; 26(1): 54-60, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31636097

ABSTRACT

PURPOSE: Induction chemotherapy results in complete remission (CR) rates of 20% to 50% among patients with poor-risk AML. Selinexor is an oral selective inhibitor of nuclear export with promising single-agent activity. By inhibiting the primary export protein, XPO1, selinexor localizes and activates tumor suppressor proteins in the nucleus and inhibits DNA damage repair, rationalizing combination with DNA-damaging agents. PATIENTS AND METHODS: This was a single-arm phase I clinical trial of selinexor combined with cytarabine and daunorubicin (7+3). Dose escalation was selinexor alone (3+3) with an expansion at the MTD. Cohorts 1 and 2 received 60 and 80 mg orally, respectively, twice weekly during induction. Consolidation cycles (≤ 2) with selinexor at induction dose plus 5+2 were allowed for patients who achieved CR. MTD and recommended phase II dose of selinexor were the primary endpoints. RESULTS: Twenty-one patients with poor-risk AML were enrolled. All 21 patients were included in the safety evaluations and survival analyses (4 in each of 2 cohorts; 13 in the expansion); 8 (53%) of the 19 patients evaluable for response achieved CR/CRi. MTD was not reached. Selinexor 80 mg (orally, twice weekly) was used in the expansion phase. The most common grade 3/4 nonhematologic treatment-emergent adverse events were febrile neutropenia (67%), diarrhea (29%), hyponatremia (29%), and sepsis (14%). At median follow-up (28.9 months), 38% of patients were alive. Median overall survival was 10.3 months. CONCLUSIONS: Selinexor plus 7+3 is a safe regimen for patients with newly diagnosed poor-risk AML and warrants further investigation in a larger clinical trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Female , Humans , Hydrazines/administration & dosage , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Maximum Tolerated Dose , Middle Aged , Patient Safety , Survival Rate , Tissue Distribution , Treatment Outcome , Triazoles/administration & dosage
3.
Oncotarget ; 7(48): 78896-78909, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27806331

ABSTRACT

Acquired proteasome-inhibitor (PI) resistance is a major obstacle in the treatment of multiple myeloma (MM). We investigated whether the clinical XPO1-inhibitor selinexor, when combined with bortezomib or carfilzomib, could overcome acquired resistance in MM. PI-resistant myeloma cell lines both in vitro and in vivo and refractory myeloma patient biopsies were treated with selinexor/bortezomib or carfilzomib and assayed for apoptosis. Mechanistic studies included NFκB pathway protein expression assays, immunofluorescence microscopy, ImageStream flow-cytometry, and proximity-ligation assays. IκBα knockdown and NFκB activity were measured in selinexor/bortezomib-treated MM cells. We found that selinexor restored sensitivity of PI-resistant MM to bortezomib and carfilzomib. Selinexor/bortezomib treatment inhibited PI-resistant MM tumor growth and increased survival in mice. Myeloma cells from PI-refractory MM patients were sensitized by selinexor to bortezomib and carfilzomib without affecting non-myeloma cells. Immunofluorescence microscopy, Western blot, and ImageStream analyses of MM cells showed increases in total and nuclear IκBα by selinexor/bortezomib. Proximity ligation found increased IκBα-NFκB complexes in treated MM cells. IκBα knockdown abrogated selinexor/bortezomib-induced cytotoxicity in MM cells. Selinexor/bortezomib treatment decreased NFκB transcriptional activity. Selinexor, when used with bortezomib or carfilzomib, has the potential to overcome PI drug resistance in MM. Sensitization may be due to inactivation of the NFκB pathway by IκBα.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bortezomib/pharmacology , Cell Nucleus/drug effects , Drug Resistance, Neoplasm/drug effects , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Multiple Myeloma/drug therapy , NF-KappaB Inhibitor alpha/metabolism , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/pharmacology , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/pathology , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Humans , Karyopherins/metabolism , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , NF-KappaB Inhibitor alpha/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Stability , Proteolysis , RNA Interference , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors , Transcription, Genetic , Transfection , Xenograft Model Antitumor Assays , Exportin 1 Protein
4.
J Hematol Oncol ; 9(1): 73, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27557643

ABSTRACT

BACKGROUND: Acquired drug resistance is the greatest obstacle to the successful treatment of multiple myeloma (MM). Despite recent advanced treatment options such as liposomal formulations, proteasome inhibitors, immunomodulatory drugs, myeloma-targeted antibodies, and histone deacetylase inhibitors, MM is still considered an incurable disease. METHODS: We investigated whether the clinical exportin 1 (XPO1) inhibitor selinexor (KPT-330), when combined with pegylated liposomal doxorubicin (PLD) or doxorubicin hydrochloride, could overcome acquired drug resistance in multidrug-resistant human MM xenograft tumors, four different multidrug-resistant MM cell lines, or ex vivo MM biopsies from relapsed/refractory patients. Mechanistic studies were performed to assess co-localization of topoisomerase II alpha (TOP2A), DNA damage, and siRNA knockdown of drug targets. RESULTS: Selinexor was found to restore sensitivity of multidrug-resistant 8226B25, 8226Dox6, 8226Dox40, and U266PSR human MM cells to doxorubicin to levels found in parental myeloma cell lines. NOD/SCID-γ mice challenged with drug-resistant or parental U266 human MM and treated with selinexor/PLD had significantly decreased tumor growth and increased survival with minimal toxicity. Selinexor/doxorubicin treatment selectively induced apoptosis in CD138/light-chain-positive MM cells without affecting non-myeloma cells in ex vivo-treated bone marrow aspirates from newly diagnosed or relapsed/refractory MM patients. Selinexor inhibited XPO1-TOP2A protein complexes (proximity ligation assay), preventing nuclear export of TOP2A in both parental and multidrug-resistant MM cell lines. Selinexor/doxorubicin treatment significantly increased DNA damage (comet assay/γ-H2AX) in both parental and drug-resistant MM cells. TOP2A knockdown reversed both the anti-tumor effect and significantly reduced DNA damage induced by selinexor/doxorubicin treatment. CONCLUSIONS: The combination of an XPO1 inhibitor and liposomal doxorubicin was highly effective against acquired drug resistance in in vitro MM models, in in vivo xenograft studies, and in ex vivo samples obtained from patients with relapsed/refractory myeloma. This drug combination synergistically induced TOP2A-mediated DNA damage and subsequent apoptosis. In addition, based on our preclinical data, we have initiated a phase I/II study with the XPO1 inhibitor selinexor and PLD (ClinicalTrials.gov NCT02186834). Initial results from both preclinical and clinical trials have shown significant promise for this drug combination for the treatment of MM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm , Multiple Myeloma/drug therapy , Animals , Apoptosis/drug effects , Biopsy , Cell Line, Tumor , DNA Damage/drug effects , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Drug Synergism , Heterografts , Humans , Hydrazines/therapeutic use , Karyopherins/antagonists & inhibitors , Mice , Multiple Myeloma/mortality , Multiple Myeloma/physiopathology , Polyethylene Glycols/therapeutic use , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Survival Rate , Topoisomerase II Inhibitors/therapeutic use , Triazoles/therapeutic use , Tumor Burden/drug effects , Exportin 1 Protein
5.
Semin Cancer Biol ; 27: 62-73, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24631834

ABSTRACT

Nuclear-cytoplasmic trafficking of proteins is a significant factor in the development of cancer and drug resistance. Subcellular localization of exported proteins linked to cancer development include those involved in cell growth and proliferation, apoptosis, cell cycle regulation, transformation, angiogenesis, cell adhesion, invasion, and metastasis. Here, we examined the basic mechanisms involved in the export of proteins from the nucleus to the cytoplasm. All proteins over 40kDa use the nuclear pore complex to gain entry or exit from the nucleus, with the primary nuclear export molecule involved in these processes being chromosome region maintenance 1 (CRM1, exportin 1 or XPO1). Proteins exported from the nucleus must possess a hydrophobic nuclear export signal (NES) peptide that binds to a hydrophobic groove containing an active-site Cys528 in the CRM1 protein. CRM1 inhibitors function largely by covalent modification of the active site Cys528 and prevent binding to the cargo protein NES. In the absence of a CRM1 inhibitor, CRM1 binds cooperatively to the NES of the cargo protein and RanGTP, forming a trimer that is actively transported out of the nucleus by facilitated diffusion. Nuclear export can be blocked by CRM1 inhibitors, NES peptide inhibitors or by preventing post-translational modification of cargo proteins. Clinical trials using the classic CRM1 inhibitor leptomycin B proved too toxic for patients; however, a new generation of less toxic small molecule inhibitors is being used in clinical trials in patients with both hematological malignancies and solid tumors. Additional trials are being initiated using small-molecule CRM1 inhibitors in combination with chemotherapeutics such as pegylated liposomal doxorubicin. In this review, we present evidence that combining the new CRM1 inhibitors with other classes of therapeutics may prove effective in the treatment of cancer. Potential combinatorial therapies discussed include the use of CRM1 inhibitors and the addition of alkylating agents (melphalan), anthracyclines (doxorubicin and daunomycin), BRAF inhibitors, platinum drugs (cisplatin and oxaliplatin), proteosome inhibitors (bortezomib and carfilzomib), or tyrosine-kinase inhibitors (imatinib). Also, the sequence of treatment may be important for combination therapy. We found that the most effective treatment regimen involved first priming the cancer cells with the CRM1 inhibitor followed by doxorubicin, bortezomib, carfilzomib, or melphalan. This order sensitized both de novo and acquired drug-resistant cancer cell lines.


Subject(s)
Active Transport, Cell Nucleus , Karyopherins/metabolism , Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm , Humans , Karyopherins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Xenograft Model Antitumor Assays , Exportin 1 Protein
6.
J Cancer ; 4(8): 614-25, 2013.
Article in English | MEDLINE | ID: mdl-24155773

ABSTRACT

Multiple myeloma (MM) remains an incurable disease despite improved treatments, including lenalidomide/pomalidomide and bortezomib/carfilzomib based therapies and high-dose chemotherapy with autologous stem cell rescue. New drug targets are needed to further improve treatment outcomes. Nuclear export of macromolecules is misregulated in many cancers, including in hematological malignancies such as MM. CRM1 (chromosome maintenance protein-1) is a ubiquitous protein that exports large proteins (>40 kDa) from the nucleus to the cytoplasm. We found that small-molecule Selective Inhibitors of Nuclear Export (SINE) prevent CRM1-mediated export of p53 and topoisomerase IIα (topo IIα). SINE's CRM1-inhibiting activity was verified by nuclear-cytoplasmic fractionation and immunocytochemical staining of the CRM1 cargoes p53 and topo IIα in MM cells. We found that SINE molecules reduced cell viability and induced apoptosis when used as both single agents in the sub-micromolar range and when combined with doxorubicin, bortezomib, or carfilzomib but not lenalidomide, melphalan, or dexamethasone. In addition, CRM1 inhibition sensitized MM cell lines and patient myeloma cells to doxorubicin, bortezomib, and carfilzomib but did not affect peripheral blood mononuclear or non-myeloma bone marrow mononuclear cells as shown by cell viability and apoptosis assay. Drug resistance induced by co-culture of myeloma cells with bone marrow stroma cells was circumvented by the addition of SINE molecules. These results support the continued development of SINE for patients with MM.

7.
Blood ; 122(13): 2251-61, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23908471

ABSTRACT

Alloreactive donor T cells against host minor histocompatibility antigens (mHAs) cause graft-versus-host disease (GVHD) after marrow transplantation from HLA-identical siblings. We sought to identify and expand regulatory CD4 T cells (Tregs) specific for human mHAs in numbers and potency adequate for clinical testing. Purified Tregs from normal donors were stimulated by dendritic cells (DCs) from their HLA-matched siblings in the presence of interleukin 2, interleukin 15, and rapamycin. Male-specific Treg clones against H-Y antigens DBY, UTY, or DFFRY-2 suppressed conventional CD4 T cell (Tconv) response to the specific antigen. In the blood of 16 donors, we found a 24-fold (range, 8-fold to 39-fold) excess Tconvs over Tregs reactive against sibling mHAs. We expanded mHA-specific Tregs from 4 blood samples and 4 leukaphereses by 155- to 405-fold. Cultured Tregs produced allospecific suppression, maintained demethylation of the Treg-specific Foxp3 gene promoter, Foxp3 expression, and transforming growth factor ß production. The rare CD4 T conv and CD8 T cells in the end product were anergic. This is the first report of detection and expansion of potent mHA-specific Tregs from HLA-matched siblings in sufficient numbers for application in human transplant trials.


Subject(s)
Cell Culture Techniques/methods , Graft vs Host Disease/prevention & control , Minor Histocompatibility Antigens/immunology , T-Lymphocytes, Regulatory/immunology , Coculture Techniques , Female , Flow Cytometry , Humans , Male , Siblings , T-Lymphocytes, Regulatory/cytology , Transplantation, Homologous
8.
Biochem Pharmacol ; 83(8): 1021-32, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22209898

ABSTRACT

The intracellular location of a protein is crucial to its normal functioning in a cell. Cancer cells utilize the normal processes of nuclear-cytoplasmic transport through the nuclear pore complex of a cell to effectively evade anti-neoplastic mechanisms. CRM1-mediated export is increased in various cancers. Proteins that are exported in cancer include tumor-suppressive proteins such as retinoblastoma, APC, p53, BRAC1, FOXO proteins, INI1/hSNF5, galectin-3, Bok, nucleophosmin, RASSF2, Merlin, p21(CIP), p27(KIP1), N-WASP/FAK, estradiol receptor and Tob, drug targets topoisomerase I and IIα and BCR-ABL, and the molecular chaperone protein Hsp90. Here, we review in detail the current processes and known structures involved in the export of a protein through the nuclear pore complex. We also discuss the export receptor molecule CRM1 and its binding to the leucine-rich nuclear export signal of the cargo protein and the formation of a nuclear export trimer with RanGTP. The therapeutic potential of various CRM1 inhibitors will be addressed, including leptomycin B, ratjadone, KOS-2464, and specific small molecule inhibitors of CRM1, N-azolylacrylate analogs, FOXO export inhibitors, valtrate, acetoxychavicol acetate, CBS9106, and SINE inhibitors. We will also discuss examples of how drug resistance may be reversed by targeting the exported proteins topoisomerase IIα, BCR-ABL, and galectin-3. As effective and less toxic CRM1 export inhibitors become available, they may be used as both single agents and in combination with current chemotherapeutic drugs. We believe that the future development of low-toxicity, small-molecule CRM1 inhibitors may provide a new approach to treating cancer.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Antineoplastic Agents/pharmacology , Aminopyridines/pharmacology , Antigens, Neoplasm/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Fatty Acids, Unsaturated/pharmacology , Female , Galectin 3/metabolism , Humans , Karyopherins/antagonists & inhibitors , Karyopherins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proteins/metabolism , Pyrones/pharmacology , Pyrrolidinones/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
9.
Cancer Res ; 69(17): 6899-905, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19690141

ABSTRACT

Topoisomerase IIalpha (topo IIalpha) is exported from the nucleus of human myeloma cells by a CRM1-dependent mechanism at cellular densities similar to those found in patient bone marrow. When topo IIalpha is trafficked to the cytoplasm, it is not in contact with the DNA; thus, topo IIalpha inhibitors are unable to induce DNA-cleavable complexes and cell death. Using a CRM1 inhibitor or a CRM1-specific small interfering RNA (siRNA), we were able to block nuclear export of topo IIalpha as shown by immunofluorescence microscopy. Human myeloma cell lines and patient myeloma cells isolated from bone marrow were treated with a CRM1 inhibitor or CRM1-specific siRNA and exposed to doxorubicin or etoposide at high cell densities. CRM1-treated cell lines or myeloma patient cells were 4-fold more sensitive to topo II poisons as determined by an activated caspase assay. Normal cells were not significantly affected by CRM1-topo II inhibitor combination treatment. Cell death was correlated with increased DNA double-strand breaks as shown by the comet assay. Band depletion assays of CRM1 inhibitor-exposed myeloma cells showed increased topo IIalpha covalently bound to DNA. Topo IIalpha knockdown by a topo IIalpha-specific siRNA abrogated the CRM1-topo II therapy synergistic effect. These results suggest that blocking topo IIalpha nuclear export sensitizes myeloma cells to topo II inhibitors. This method of sensitizing myeloma cells suggests a new therapeutic approach to multiple myeloma.


Subject(s)
Bone Marrow Neoplasms/drug therapy , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Karyopherins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Topoisomerase II Inhibitors , Antibiotics, Antineoplastic/pharmacology , Antigens, Neoplasm/metabolism , Bone Marrow Neoplasms/enzymology , Bone Marrow Neoplasms/pathology , Cell Count , Cell Death/drug effects , Cell Line, Tumor , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Humans , Karyopherins/genetics , Karyopherins/metabolism , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Protein Binding , Protein Transport/drug effects , Pyrones/pharmacology , RNA, Small Interfering , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
10.
Mol Cancer Ther ; 8(4): 794-801, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372552

ABSTRACT

Histone deacetylases (HDAC) may have a prominent role in the development of cancer and the response to anticancer therapy. However, the therapeutic relevance and tissue specificity of individual HDAC enzymes remain largely unknown. HDAC inhibitors may function as sensitizing agents to chemotherapies that target DNA through their effects on chromatin structure and plasticity. Here, we report a new role for HDAC2 as a regulator of chromatin compaction status and the mediator of HDAC inhibitor-induced sensitization to chemotherapy. The selective depletion of HDAC2 by small interfering RNA led to reduced expression of heterochromatin maintenance proteins and morphologic changes indicative of chromatin decondensation. Furthermore, depletion of HDAC2 but not HDAC1 or HDAC6 was sufficient to sensitize breast cancer cells to topoisomerase inhibitor-induced apoptosis. The levels of HDAC2 expression appear to correlate with the degree of HDAC inhibitor-induced histone acetylation in a surrogate tissue in patients. These data suggest that HDAC2 may be a relevant pharmacologic and biological target for combination therapy involving drugs that target DNA.


Subject(s)
Breast Neoplasms/genetics , Chromatin/metabolism , DNA, Neoplasm/metabolism , DNA/genetics , Histone Deacetylases/physiology , Repressor Proteins/physiology , Acetylation , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA/metabolism , Enzyme Inhibitors/pharmacology , Epirubicin/pharmacology , Gene Expression Profiling , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylase 6 , Histones , Humans , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
11.
Curr Med Chem ; 15(26): 2648-55, 2008.
Article in English | MEDLINE | ID: mdl-18991627

ABSTRACT

Expression levels of intact tumor suppressor proteins and molecular targets of anti-neoplastic agents are critical in defining cancer cell drug sensitivity; however, the intracellular location of a specific protein may be as important. Many tumor suppressor proteins must be present in the cell nucleus to perform their policing activities or for the cell to respond to chemotherapeutic agents. Nuclear proteins needed to prevent cancer initiation or progression or to optimize chemotherapeutic response include the tumor suppressor proteins p53, APC/beta-catenin, and FOXO family genes; negative regulators of cell cycle progression and survival such as p21(CIP1) and p27(KIP1;) and chemotherapeutic targets such as DNA topoisomerases I and IIalpha. Mislocalization of a nuclear protein into the cytoplasm can render it ineffective as a tumor suppressor or as a target for chemotherapy. Blocking nuclear export of any or all of these proteins may restore tumor suppression or apoptosis or, for topoisomerases I and IIalpha, reverse drug resistance to inhibitors of these enzymes. During disease progression or in response to the tumor environment, cancer cells appear to acquire intracellular mechanisms to export anti-cancer nuclear proteins. These mechanisms generally involve modification of nuclear proteins, causing the proteins to reveal leucine-rich nuclear export signal protein sequences. Subsequent export is mediated by CRM1. This review defines the general processes involved in nuclear export mediated by CRM1/RanGTP (exportin/XPO1), examines the functions of individual tumor suppressor nuclear proteins and nuclear targets of chemotherapy, and explores potential mechanisms of cancer cells to induce export of these proteins. Novel drugs that could potentially counteract nuclear export of specific proteins are also discussed.


Subject(s)
Drug Resistance, Neoplasm , Karyopherins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Humans , Molecular Sequence Data , Exportin 1 Protein
12.
Blood ; 108(12): 3881-9, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16917002

ABSTRACT

We investigated the role of the breast cancer resistance protein (BCRP/ABCG2) in drug resistance in multiple myeloma (MM). Human MM cell lines, and MM patient plasma cells isolated from bone marrow, were evaluated for ABCG2 mRNA expression by quantitative polymerase chain reaction (PCR) and ABCG2 protein, by Western blot analysis, immunofluorescence microscopy, and flow cytometry. ABCG2 function was determined by measuring topotecan and doxorubicin efflux using flow cytometry, in the presence and absence of the specific ABCG2 inhibitor, tryprostatin A. The methylation of the ABCG2 promoter was determined using bisulfite sequencing. We found that ABCG2 expression in myeloma cell lines increased after exposure to topotecan and doxorubicin, and was greater in logphase cells when compared with quiescent cells. Myeloma patients treated with topotecan had an increase in ABCG2 mRNA and protein expression after treatment with topotecan, and at relapse. Expression of ABCG2 is regulated, at least in part, by promoter methylation both in cell lines and in patient plasma cells. Demethylation of the promoter increased ABCG2 mRNA and protein expression. These findings suggest that ABCG2 is expressed and functional in human myeloma cells, regulated by promoter methylation, affected by cell density, up-regulated in response to chemotherapy, and may contribute to intrinsic drug resistance.


Subject(s)
ATP-Binding Cassette Transporters/genetics , DNA Methylation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Multiple Myeloma/genetics , Neoplasm Proteins/genetics , Promoter Regions, Genetic , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blotting, Western/methods , Cell Line, Tumor , DNA Methylation/drug effects , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Flow Cytometry/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Microscopy, Fluorescence/methods , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Proteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Topotecan/pharmacology , Topotecan/therapeutic use , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
13.
Clin Cancer Res ; 11(23): 8467-75, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322310

ABSTRACT

BACKGROUND: DNA topoisomerase II inhibitors and poisons are among the most efficacious drugs for the treatment of cancer. Sensitivity of cancer cells to the cytotoxic effects of topoisomerase II targeting agents is thought to depend on the expression of the topoisomerase IIalpha isoform, and drug resistance is often associated with loss or mutation of topoisomerase IIalpha. Histone deacetylase inhibitors (HDACi) are a novel class of compounds that potentiate the antitumor effects of topoisomerase II-targeting agents. METHODS: The interaction between HDACi and topoisomerase II-targeting agents in cancer cells was evaluated as a function of topoisomerase IIalpha and topoisomerase IIbeta expression. Topoisomerase II isoforms were selectively depleted using small interfering RNA and antisense. Drug-induced formation of cleavable complexes involving topoisomerase IIalpha and topoisomerase IIbeta was evaluated by trapped-in-agarose DNA immunostaining and band depletion assays in the presence and absence of HDACi. RESULTS: Preexposure to HDACi increased the cytotoxicity of topoisomerase II poisons. This was associated with a down-regulation of topoisomerase IIalpha expression but had no effects on topoisomerase IIbeta. In the setting of HDACi-induced chromatin decondensation and topoisomerase IIalpha depletion, topoisomerase II poison cytotoxicity was mediated through topoisomerase IIbeta cleavable complex formation. The HDACi-induced sensitization was also observed in cells with target-specific resistance to topoisomerase II poisons. CONCLUSIONS: The recruitment of topoisomerase IIbeta as a target may overcome primary or emergent drug resistance to topoisomerase II-targeting agents and hence may broaden the applicability of this important class of anticancer agents.


Subject(s)
DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Neoplasms/drug therapy , Topoisomerase II Inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Butyrates/pharmacology , Chromatin/metabolism , DNA Topoisomerases, Type II/metabolism , DNA, Antisense/pharmacology , Drug Interactions , Drug Synergism , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Hydroxamic Acids/pharmacology , Mitoxantrone/pharmacology , Neoplasms/enzymology , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , Teniposide/poisoning , Tumor Cells, Cultured , Valproic Acid/pharmacology
14.
Mol Carcinog ; 43(4): 225-36, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15864800

ABSTRACT

Tumor progression is a multistep process, which enables cells to evolve from benign to malignant tumors. This progression has been suggested to depend on six essential characteristics identified as the "hallmarks of cancer," which include: self-sufficiency in growth signals, insensitivity to growth-inhibitory signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. Osteopontin (OPN) is an integrin-binding protein that has been shown to be associated with the progression of several cancer types, and to play an important functional role in various aspects of malignancy, particularly tissue invasion and metastasis. Here we studied genes regulated by OPN in a model of human breast cancer using oligonucleotide microarray technology by comparing the gene-expression profiles of 21NT mammary carcinoma cells transfected to overexpress OPN versus mock-transfected control cells. From over 12,000 human genes, we identified 99 known human genes differentially regulated by OPN whose expression changed by at least 1.5-fold and showed statistically significant differences in mean expression levels between groups. Functional classification of these genes into the hallmarks of cancer categories showed that OPN can affect the expression of genes involved in all six categories in this model. Furthermore, we were able to validate the expression of 18/19 selected candidate genes by quantitative real-time PCR, further supporting our microarray findings. This study provides the first evidence that OPN can lead to numerous gene expression changes that influence multiple aspects of tumor progression and malignant growth.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Sialoglycoproteins/pharmacology , Cell Line, Tumor , Disease Progression , Down-Regulation/drug effects , Down-Regulation/genetics , Humans , Models, Biological , Osteopontin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
15.
J Cell Sci ; 117(Pt 14): 3061-71, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15173319

ABSTRACT

Resistance to chemotherapeutic drugs is a major obstacle in the treatment of leukemia and multiple myeloma. We have previously found that myeloma and leukemic cells in transition from low-density log phase conditions to high-density plateau phase conditions export substantial amounts of endogenous topoisomerase II alpha from the nucleus to the cytoplasm. In order for topoisomerase-targeted chemotherapy to function, the topoisomerase target must have access to the nuclear DNA. Therefore, the nuclear export of topoisomerase II alpha may contribute to drug resistance, and defining this mechanism may lead to methods to preclude this avenue of resistance. We have identified nuclear export signals for topoisomerase II alpha at amino acids 1017-1028 and 1054-1066, using FITC-labeled BSA-export signal peptide conjugates microinjected into the nuclei of HeLa cells. Functional confirmation of both signals (1017-1028 and 1054-1066) was provided by transfection of human myeloma cells with plasmids containing the gene for a full-length human FLAG-topoisomerase fusion protein, mutated at hydrophobic amino acid residues in the export signals. Of the six putative export signals tested, the two sites above were found to induce export into the cytoplasm. Export by both signals was blocked by treatment of the cells with leptomycin B, indicating that a CRM-1-dependent pathway mediates export. Site-directed mutagenesis of two central hydrophobic residues in either export signal in full-length human topoisomerase blocked export of recombinant FLAG-topoisomerase II alpha, indicating that both signals may be required for export. Interestingly, this pair of nuclear export signals (1017-1028 and 1054-1066) also defines a dimerization domain of the topoisomerase II alpha molecule.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Nucleus/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Karyopherins/physiology , Nuclear Localization Signals , Receptors, Cytoplasmic and Nuclear/physiology , Active Transport, Cell Nucleus , Amino Acid Sequence , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cytoplasm/metabolism , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Humans , Microinjections , Molecular Sequence Data , Mutagenesis, Site-Directed , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...