Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 586(7831): 769-775, 2020 10.
Article in English | MEDLINE | ID: mdl-33057200

ABSTRACT

Myeloproliferative neoplasms (MPNs) are blood cancers that are characterized by the excessive production of mature myeloid cells and arise from the acquisition of somatic driver mutations in haematopoietic stem cells (HSCs). Epidemiological studies indicate a substantial heritable component of MPNs that is among the highest known for cancers1. However, only a limited number of genetic risk loci have been identified, and the underlying biological mechanisms that lead to the acquisition of MPNs remain unclear. Here, by conducting a large-scale genome-wide association study (3,797 cases and 1,152,977 controls), we identify 17 MPN risk loci (P < 5.0 × 10-8), 7 of which have not been previously reported. We find that there is a shared genetic architecture between MPN risk and several haematopoietic traits from distinct lineages; that there is an enrichment for MPN risk variants within accessible chromatin of HSCs; and that increased MPN risk is associated with longer telomere length in leukocytes and other clonal haematopoietic states-collectively suggesting that MPN risk is associated with the function and self-renewal of HSCs. We use gene mapping to identify modulators of HSC biology linked to MPN risk, and show through targeted variant-to-function assays that CHEK2 and GFI1B have roles in altering the function of HSCs to confer disease risk. Overall, our results reveal a previously unappreciated mechanism for inherited MPN risk through the modulation of HSC function.


Subject(s)
Genetic Predisposition to Disease/genetics , Hematopoietic Stem Cells/pathology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasms/genetics , Neoplasms/pathology , Cell Lineage/genetics , Cell Self Renewal , Checkpoint Kinase 2/genetics , Female , Humans , Leukocytes/pathology , Male , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Risk , Telomere Homeostasis
2.
Gynecol Oncol ; 185: 202-211, 2024 06.
Article in English | MEDLINE | ID: mdl-38834399

ABSTRACT

OBJECTIVE: To report long-term efficacy and safety of selinexor maintenance therapy in adults with TP53 wild-type (TP53wt) stage IV or recurrent endometrial cancer (EC) who achieved partial remission (PR) or complete remission (CR) following chemotherapy. METHODS: Analysis of the prespecified, exploratory subgroup of patients with TP53wt EC from the phase 3 SIENDO study was performed. Progression-free survival (PFS) benefit in patients with TP53wt EC and across other patient subgroups were exploratory endpoints. Safety and tolerability were also assessed. RESULTS: Of the 263 patients enrolled in the SIENDO trial, 113 patients had TP53wt EC; 70/113 (61.9%) had TP53wt/proficient mismatch repair (pMMR) EC, and 29/113 (25.7%) had TP53wt/deficient mismatch repair (dMMR) EC. As of April 1, 2024, the median PFS (mPFS) for TP53wt patients who received selinexor compared with placebo was 28.4 versus 5.2 months (36.8-month follow-up, HR 0.44; 95% CI 0.27-0.73). A benefit in mPFS was seen with selinexor versus placebo regardless of MMR status (patients with TP53wt/pMMR EC: 39.5 vs 4.9 months, HR 0.36; 95% CI 0.19-0.71; patients with TP53wt/dMMR EC: 13.1 vs 3.7 months, HR 0.49; 95% CI 0.18-1.34). Selinexor treatment was generally manageable, with no new safety signals identified. CONCLUSION: In the phase 3 SIENDO study, selinexor maintenance therapy showed a promising efficacy signal and a manageable safety profile in the prespecified subgroup of patients with TP53wt EC who achieved a PR or CR following chemotherapy. These results are being further evaluated in an ongoing randomized phase 3 trial (NCT05611931).


Subject(s)
Endometrial Neoplasms , Hydrazines , Neoplasm Recurrence, Local , Triazoles , Tumor Suppressor Protein p53 , Humans , Female , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/therapeutic use , Middle Aged , Hydrazines/adverse effects , Hydrazines/administration & dosage , Hydrazines/therapeutic use , Aged , Tumor Suppressor Protein p53/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Adult , Follow-Up Studies , Progression-Free Survival , Aged, 80 and over , Maintenance Chemotherapy/methods , Neoplasm Staging
3.
Proc Natl Acad Sci U S A ; 117(42): 26340-26346, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33020282

ABSTRACT

Balanced rearrangements involving the KMT2A gene, located at 11q23, are among the most frequent chromosome aberrations in acute myeloid leukemia (AML). Because of numerous fusion partners, the mutational landscape and prognostic impact of specific 11q23/KMT2A rearrangements are not fully understood. We analyzed clinical features of 172 adults with AML and recurrent 11q23/KMT2A rearrangements, 141 of whom had outcome data available. We compared outcomes of these patients with outcomes of 1,097 patients without an 11q23/KMT2A rearrangement categorized according to the 2017 European LeukemiaNet (ELN) classification. Using targeted next-generation sequencing, we investigated the mutational status of 81 leukemia/cancer-associated genes in 96 patients with 11q23/KMT2A rearrangements with material for molecular studies available. Patients with 11q23/KMT2A rearrangements had a low number of additional gene mutations (median, 1; range 0 to 6), which involved the RAS pathway (KRAS, NRAS, and PTPN11) in 32% of patients. KRAS mutations occurred more often in patients with t(6;11)(q27;q23)/KMT2A-AFDN compared with patients with the other 11q23/KMT2A subsets. Specific gene mutations were too infrequent in patients with specific 11q23/KMT2A rearrangements to assess their associations with outcomes. We demonstrate that younger (age <60 y) patients with t(9;11)(p22;q23)/KMT2A-MLLT3 had better outcomes than patients with other 11q23/KMT2A rearrangements and those without 11q23/KMT2A rearrangements classified in the 2017 ELN intermediate-risk group. Conversely, outcomes of older patients (age ≥60 y) with t(9;11)(p22;q23) were poor and comparable to those of the ELN adverse-risk group patients. Our study shows that patients with an 11q23/KMT2A rearrangement have distinct mutational patterns and outcomes depending on the fusion partner.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Jacobsen Distal 11q Deletion Syndrome/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Adolescent , Adult , Aged , Chromosome Aberrations , Female , Gene Rearrangement/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jacobsen Distal 11q Deletion Syndrome/metabolism , Karyotyping , Male , Middle Aged , Mutation/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Translocation, Genetic/genetics , Treatment Outcome
4.
Haematologica ; 107(5): 1034-1044, 2022 05 01.
Article in English | MEDLINE | ID: mdl-34261293

ABSTRACT

Expression levels of long non-coding RNA (lncRNA) have been shown to associate with clinical outcome of patients with cytogenetically normal acute myeloid leukemia (CN-AML). However, the frequency and clinical significance of genetic variants in the nucleotide sequences of lncRNA in AML patients is unknown. Herein, we analyzed total RNA sequencing data of 377 younger adults (aged <60 years) with CN-AML, who were comprehensively characterized with regard to clinical outcome. We used available genomic databases and stringent filters to annotate genetic variants unequivocally located in the non-coding transcriptome of AML patients. We detected 981 variants, which are recurrently present in lncRNA that are expressed in leukemic blasts. Among these variants, we identified a cytosine-to-thymidine variant in the lncRNA RP5-1074L1.4 and a cytosine-to-thymidine variant in the lncRNA SNHG15, which independently associated with longer survival of CN-AML patients. The presence of the SNHG15 cytosine-to-thymidine variant was also found to associate with better outcome in an independent dataset of CN-AML patients, despite differences in treatment protocols and RNA sequencing techniques. In order to gain biological insights, we cloned and overexpressed both wild-type and variant versions of the SNHG15 lncRNA. In keeping with its negative prognostic impact, overexpression of the wild-type SNHG15 associated with higher proliferation rate of leukemic blasts when compared with the cytosine-to-thymidine variant. We conclude that recurrent genetic variants of lncRNA that are expressed in the leukemic blasts of CN-AML patients have prognostic and potential biological significance.


Subject(s)
Leukemia, Myeloid, Acute , RNA, Long Noncoding , Transcriptome , Adult , Cytosine , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Middle Aged , Mutation , Prognosis , RNA, Long Noncoding/genetics , Thymidine
5.
Haematologica ; 105(3): 721-729, 2020 03.
Article in English | MEDLINE | ID: mdl-31413100

ABSTRACT

Leukemia stem cells (LSC) are more resistant to standard chemotherapy and their persistence during remission can cause relapse, which is still one of the major clinical challenges in the treatment of acute myeloid leukemia (AML). A better understanding of the mutational patterns and the prognostic impact of molecular markers associated with stemness could lead to better clinical management and improve patients' outcomes. We applied a previously described 17-gene expression score comprising genes differently expressed between LSC and leukemic bulk blasts, for 934 adult patients with de novo AML, and studied associations of the 17-gene LSC score with clinical data and mutation status of 81 genes recurrently mutated in cancer and leukemia. We found that patients with a high 17-gene score were older and had more mutations. The 17-gene score was found to have a prognostic impact in both younger (aged <60 years) and older (aged ≥60 years) patients with AML. We also analyzed the 17-gene LSC score in the context of the 2017 European LeukemiaNet genetic-risk classification and found that for younger patients the score refined the classification, and identified patients currently classified in the European LeukemiaNet Favorable-risk category who had a worse outcome.


Subject(s)
Leukemia, Myeloid, Acute , Adult , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Middle Aged , Mutation , Prognosis , Stem Cells , Treatment Outcome
6.
Proc Natl Acad Sci U S A ; 114(36): 9629-9634, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827320

ABSTRACT

Activating mutations in BRAF are found in 50% of melanomas and although treatment with BRAF inhibitors (BRAFi) is effective, resistance often develops. We now show that recently discovered NRAS isoform 2 is up-regulated in the setting of BRAF inhibitor resistance in melanoma, in both cell lines and patient tumor tissues. When isoform 2 was overexpressed in BRAF mutant melanoma cell lines, melanoma cell proliferation and in vivo tumor growth were significantly increased in the presence of BRAFi treatment. shRNA-mediated knockdown of isoform 2 in BRAFi resistant cells restored sensitivity to BRAFi compared with controls. Signaling analysis indicated decreased mitogen-activated protein kinase (MAPK) pathway signaling and increased phosphoinositol-3-kinase (PI3K) pathway signaling in isoform 2 overexpressing cells compared with isoform 1 overexpressing cells. Immunoprecipitation of isoform 2 validated a binding affinity of this isoform to both PI3K and BRAF/RAF1. The addition of an AKT inhibitor to BRAFi treatment resulted in a partial restoration of BRAFi sensitivity in cells expressing high levels of isoform 2. NRAS isoform 2 may contribute to resistance to BRAFi by facilitating PI3K pathway activation.


Subject(s)
GTP Phosphohydrolases/genetics , Melanoma/drug therapy , Melanoma/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Drug Resistance, Neoplasm/genetics , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/metabolism , Gene Knockdown Techniques , Humans , Indoles/therapeutic use , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Melanoma/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Skin Neoplasms/metabolism , Sulfonamides/therapeutic use , Up-Regulation , Vemurafenib
8.
Proc Natl Acad Sci U S A ; 112(49): E6744-51, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26582795

ABSTRACT

The B-Raf proto-oncogene serine/threonine kinase (BRAF) gene is the most frequently mutated gene in malignant melanoma (MM) and papillary thyroid cancer (PTC) and is causally involved in malignant cell transformation. Mutated BRAF is associated with an aggressive disease phenotype, thus making it a top candidate for targeted treatment strategies in MM and PTC. We show that BRAF mutations in both MM and PTC drive increased expression of oncomiR-3151, which is coactivated by the SP1/NF-κB complex. Knockdown of microRNA-3151 (miR-3151) with short hairpin RNAs reduces cell proliferation and increases apoptosis of MM and PTC cells. Using a targeted RNA sequencing approach, we mechanistically determined that miR-3151 directly targets TP53 and other members of the TP53 pathway. Reducing miR-3151's abundance increases TP53's mRNA and protein expression and favors its nuclear localization. Consequently, knockdown of miR-3151 also leads to caspase-3-dependent apoptosis. Simultaneous inhibition of aberrantly activated BRAF and knockdown of miR-3151 potentiates the effects of sole BRAF inhibition with the BRAF inhibitor vemurafenib and may provide a novel targeted therapeutic approach in BRAF-mutated MM and PTC patients. In conclusion, we identify miR-3151 as a previously unidentified player in MM and PTC pathogenesis, which is driven by BRAF-dependent and BRAF-independent mechanisms. Characterization of TP53 as a downstream effector of miR-3151 provides evidence for a causal link between BRAF mutations and TP53 inactivation.


Subject(s)
Carcinoma/genetics , Melanoma/genetics , MicroRNAs/physiology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Tumor Suppressor Protein p53/physiology , Active Transport, Cell Nucleus , Carcinoma, Papillary , Humans , Indoles/therapeutic use , Melanoma/drug therapy , NF-kappa B/physiology , Proto-Oncogene Mas , Sulfonamides/therapeutic use , Thyroid Cancer, Papillary , Vemurafenib
10.
Adv Exp Med Biol ; 943: 99-118, 2017.
Article in English | MEDLINE | ID: mdl-27910066

ABSTRACT

Molecular studies of endometrial cancer have evolved with the tools available to researchers: the methods for measuring nucleic acids, protein expression, and combinations thereof. Today "molecular genetic analysis" implies a broad range of indirect and direct tests that yield molecular phenotypes or genotypes, immunotypes, or signatures that were not conceived of when the histologic and biologic heterogeneity was first fully acknowledged.We will provide a historical perspective on molecular genetic studies of endometrial cancers focusing on candidate genes and how early foundational research shaped both our understanding of the disease and current research directions. Examples of direct tests (mutation, DNA methylation, and/or protein expression) will be provided along with examples of indirect tests that have been and continue to be central to endometrial cancer molecular biology, such as DNA content or microsatellite instability analysis. We will highlight clinically relevant examples of molecular phenotyping and direct evaluation of candidate genes that integrate direct and indirect testing as part of routine patient care. This is not intended to be an exhaustive review but rather an overview of the progress that has been made and how early work is shaping current molecular, clinical, and biologic studies of endometrial cancer.


Subject(s)
DNA Repair/genetics , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Genetic Testing/methods , Mutation , DNA, Neoplasm/genetics , Female , Humans , Ploidies , Tumor Suppressor Proteins/genetics
11.
Proc Natl Acad Sci U S A ; 111(11): 4179-84, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24586049

ABSTRACT

Neuroblastoma rat sarcoma (RAS) viral oncogene homolog (NRAS), a small GTPase, is one of the most thoroughly studied oncogenes that controls cell growth, differentiation, and survival by facilitating signal transduction. Here, we identify four novel naturally occurring NRAS isoforms (isoforms 2-5) in addition to the canonical isoform (isoform 1). Expression analyses performed on a panel of several different human malignancies and matching normal tissue revealed distinct isoform expression patterns. Two of the novel isoforms were found in the nucleus and cytoplasm, whereas the others were exclusively cytoplasmic. The isoforms varied in their binding affinities to known downstream targets and differentially regulated the RAS signaling pathway. Strikingly, forced expression of isoform 5, which encodes only a 20-aa peptide, led to increased cell proliferation and to transformation by activation of known NRAS targets. These discoveries open new avenues in the study of NRAS.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Transformation, Neoplastic/genetics , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/genetics , Protein Isoforms/genetics , Signal Transduction/genetics , Animals , Base Sequence , Blotting, Western , COS Cells , Chlorocebus aethiops , Cloning, Molecular , DNA Primers/genetics , Humans , Immunoprecipitation , Mice , Microscopy, Confocal , Molecular Sequence Data , NIH 3T3 Cells , Sequence Analysis, DNA , Statistics, Nonparametric
12.
Hum Mutat ; 37(10): 1004-12, 2016 10.
Article in English | MEDLINE | ID: mdl-27346418

ABSTRACT

Next-generation sequencing has revolutionized cancer genetics, but accurately detecting mutations in repetitive DNA sequences, especially mononucleotide runs, remains a challenge. This is a particular concern for tumors with defective mismatch repair (MMR) that accumulate strand-slippage mutations. We developed MonoSeq to improve indel mutation detection in mononucleotide runs, and used MonoSeq to investigate strand-slippage mutations in endometrial cancers, a tumor type that has frequent loss of MMR. We performed extensive Sanger sequencing to validate both clonal and subclonal MonoSeq mutation calls. Eighty-one regions containing mononucleotide runs were sequenced in 540 primary endometrial cancers (223 with defective MMR). Our analyses revealed that the overall mutation rate in MMR-deficient tumors was 20-30-fold higher than in MMR-normal tumors. MonoSeq analysis identified several previously unreported mutations, including a novel hotspot in an A7 run in the terminal exon of ARID5B.The ARID5B indel mutations were seen in both MMR-deficient and MMR-normal tumors, suggesting biologic selection. The analysis of tumor mRNAs revealed the presence of mutant transcripts that could result in translation of neopeptides. Improved detection of mononucleotide run strand-slippage mutations has clear implications for comprehensive mutation detection in tumors with defective MMR. Indel frameshift mutations and the resultant antigenic peptides could help guide immunotherapy strategies.


Subject(s)
DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , INDEL Mutation , Sequence Analysis, DNA/methods , Transcription Factors/genetics , Algorithms , DNA Mismatch Repair , Female , Frameshift Mutation , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Humans
13.
Gynecol Oncol ; 141(2): 312-317, 2016 05.
Article in English | MEDLINE | ID: mdl-26957478

ABSTRACT

BACKGROUND: We sought to validate the prognostic significance of estrogen receptor alpha (ERα) expression and to investigate the relationship between ESR1 mutation status and outcomes in a large cohort of patients with endometrial cancer. We also investigated the predictive value of ERα for lymph node involvement in a large surgically staged cohort. METHODS: A tumor microarray (TMA) was constructed including only pure endometrioid adenocarcinomas, stained with ER50 monoclonal antibody, and assessed using digital image analysis. For mutation analysis, somatic DNA was extracted and sequenced for ESR1 gene hotspot regions. Differences in patient and tumor characteristics, recurrence and survival between ERα positive and negative, mutated and wild-type tumors were evaluated. RESULTS: Sixty (18.6%) tumors were negative for ERα. Absence of ERα was significantly associated with stage and grade, but not with disease-free or overall survival. ERα was a strong predictor of lymph node involvement (RR: 2.37, 95% CI: 1.12-5.02). Nineteen of 1034 tumors (1.8%) had an ESR1 hotspot mutation; twelve in hotspot 537Y, four in 538D and three in 536L. Patients with an ESR1 mutation had a significantly lower BMI, but were comparable in age, stage and grade, and progression-free survival. CONCLUSION: Patients with ERα negative endometrioid endometrial cancer are more often diagnosed with higher grade and advanced stage disease. Lymph node involvement is more common with lack of ERα expression, and may be able to help triage which patients should undergo lymphadenectomy. Mutations in ESR1 might explain why some low risk women with low BMI develop endometrial cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Estrogen Receptor alpha/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , Humans , Lymphatic Metastasis , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Staging , Prognosis , Tissue Array Analysis
14.
Blood ; 122(11): 1923-34, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23926298

ABSTRACT

FTY720 (Fingolimod, Gilenya) is a sphingosine analog used as an immunosuppressant in multiple sclerosis patients. FTY720 is also a potent protein phosphatase 2A (PP2A)-activating drug (PAD). PP2A is a tumor suppressor found inactivated in different types of cancer. We show here that PP2A is inactive in polycythemia vera (PV) and other myeloproliferative neoplasms characterized by the expression of the transforming Jak2(V617F) oncogene. PP2A inactivation occurs in a Jak2(V617F) dose/kinase-dependent manner through the PI-3Kγ-PKC-induced phosphorylation of the PP2A inhibitor SET. Genetic or PAD-mediated PP2A reactivation induces Jak2(V617F) inactivation/downregulation and impairs clonogenic potential of Jak2(V617F) cell lines and PV but not normal CD34(+) progenitors. Likewise, FTY720 decreases leukemic allelic burden, reduces splenomegaly, and significantly increases survival of Jak2(V617F) leukemic mice without adverse effects. Mechanistically, we show that in Jak2(V617F) cells, FTY720 antileukemic activity requires neither FTY720 phosphorylation (FTY720-P) nor SET dimerization or ceramide induction but depends on interaction with SET K209. Moreover, we show that Jak2(V617F) also utilizes an alternative sphingosine kinase-1-mediated pathway to inhibit PP2A and that FTY720-P, acting as a sphingosine-1-phosphate-receptor-1 agonist, elicits signals leading to the Jak2-PI-3Kγ-PKC-SET-mediated PP2A inhibition. Thus, PADs (eg, FTY720) represent suitable therapeutic alternatives for Jak2(V617F) MPNs.


Subject(s)
Janus Kinase 2/metabolism , Leukemia/drug therapy , Propylene Glycols/pharmacology , Protein Phosphatase 2/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase , DNA-Binding Proteins , Enzyme Activation/drug effects , Fingolimod Hydrochloride , Histone Chaperones , Humans , Immunoblotting , Immunosuppressive Agents/pharmacology , Janus Kinase 2/genetics , Kaplan-Meier Estimate , Leukemia/genetics , Leukemia/pathology , Mice , Mice, SCID , Mutation , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Phosphatase 2/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingosine/pharmacology , Treatment Outcome
15.
Blood ; 122(17): 3034-44, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-23970380

ABSTRACT

As tyrosine kinase inhibitors (TKIs) fail to induce long-term response in blast crisis chronic myelogenous leukemia (CML-BC) and Philadelphia chromosome-positive (Ph(+)) acute lymphoblastic leukemia (ALL), novel therapies targeting leukemia-dysregulated pathways are necessary. Exportin-1 (XPO1), also known as chromosome maintenance protein 1, regulates cell growth and differentiation by controlling the nucleocytoplasmic trafficking of proteins and RNAs, some of which are aberrantly modulated in BCR-ABL1(+) leukemias. Using CD34(+) progenitors from CML, B-ALL, and healthy individuals, we found that XPO1 expression was markedly increased, mostly in a TKI-sensitive manner, in CML-BC and Ph(+) B-ALL. Notably, XPO1 was also elevated in Ph(-) B-ALL. Moreover, the clinically relevant XPO1 inhibitor KPT-330 strongly triggered apoptosis and impaired the clonogenic potential of leukemic, but not normal, CD34(+) progenitors, and increased survival of BCR-ABL1(+) mice, 50% of which remained alive and, mostly, became BCR-ABL1 negative. Moreover, KPT-330 compassionate use in a patient with TKI-resistant CML undergoing disease progression significantly reduced white blood cell count, blast cells, splenomegaly, lactate dehydrogenase levels, and bone pain. Mechanistically, KPT-330 altered the subcellular localization of leukemia-regulated factors including RNA-binding heterogeneous nuclear ribonucleoprotein A1 and the oncogene SET, thereby inducing reactivation of protein phosphatase 2A tumor suppressor and inhibition of BCR-ABL1 in CML-BC cells. Because XPO1 is important for leukemic cell survival, KPT-330 may represent an alternative therapy for TKI-refractory Ph(+) leukemias.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/pharmacology , Adult , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , DNA-Binding Proteins , Drug Evaluation, Preclinical , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Histone Chaperones/metabolism , Humans , Karyopherins/genetics , Karyopherins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Transport , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Exportin 1 Protein
16.
Leukemia ; 38(1): 45-57, 2024 01.
Article in English | MEDLINE | ID: mdl-38017103

ABSTRACT

Clinical outcome of patients with acute myeloid leukemia (AML) is associated with demographic and genetic features. Although the associations of acquired genetic alterations with patients' sex have been recently analyzed, their impact on outcome of female and male patients has not yet been comprehensively assessed. We performed mutational profiling, cytogenetic and outcome analyses in 1726 adults with AML (749 female and 977 male) treated on frontline Alliance for Clinical Trials in Oncology protocols. A validation cohort comprised 465 women and 489 men treated on frontline protocols of the German AML Cooperative Group. Compared with men, women more often had normal karyotype, FLT3-ITD, DNMT3A, NPM1 and WT1 mutations and less often complex karyotype, ASXL1, SRSF2, U2AF1, RUNX1, or KIT mutations. More women were in the 2022 European LeukemiaNet intermediate-risk group and more men in adverse-risk group. We found sex differences in co-occurring mutation patterns and prognostic impact of select genetic alterations. The mutation-associated splicing events and gene-expression profiles also differed between sexes. In patients aged <60 years, SF3B1 mutations were male-specific adverse outcome prognosticators. We conclude that sex differences in AML-associated genetic alterations and mutation-specific differential splicing events highlight the importance of patients' sex in analyses of AML biology and prognostication.


Subject(s)
Leukemia, Myeloid, Acute , Sex Characteristics , Adult , Humans , Male , Female , Prognosis , Nucleophosmin , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Mutation , fms-Like Tyrosine Kinase 3/genetics
17.
Front Immunol ; 14: 1275329, 2023.
Article in English | MEDLINE | ID: mdl-37954586

ABSTRACT

Competent T-cells with sufficient levels of fitness combat cancer formation and progression. In multiple myeloma (MM), T-cell exhaustion is caused by several factors including tumor burden, constant immune activation due to chronic disease, age, nutritional status, and certain MM treatments such as alkylating agents and proteasome inhibitors. Many currently used therapies, including bispecific T-cell engagers, anti-CD38 antibodies, proteasome inhibitors, and CART-cells, directly or indirectly depend on the anti-cancer activity of T-cells. Reduced T-cell fitness not only diminishes immune defenses, increasing patient susceptibility to opportunistic infections, but can impact effectiveness MM therapy effectiveness, bringing into focus sequencing strategies that could modulate T-cell fitness and potentially optimize overall benefit and clinical outcomes. Certain targeted agents used to treat MM, such as selective inhibitors of nuclear export (SINE) compounds, have the potential to mitigate T-cell exhaustion. Herein referred to as XPO1 inhibitors, SINE compounds inhibit the nuclear export protein exportin 1 (XPO1), which leads to nuclear retention and activation of tumor suppressor proteins and downregulation of oncoprotein expression. The XPO1 inhibitors selinexor and eltanexor reduced T-cell exhaustion in cell lines and animal models, suggesting their potential role in revitalizating these key effector cells. Additional clinical studies are needed to understand how T-cell fitness is impacted by diseases and therapeutic factors in MM, to potentially facilitate the optimal use of available treatments that depend on, and impact, T-cell function. This review summarizes the importance of T-cell fitness and the potential to optimize treatment using T-cell engaging therapies with a focus on XPO1 inhibitors.


Subject(s)
Multiple Myeloma , Animals , Humans , Multiple Myeloma/drug therapy , Proteasome Inhibitors/therapeutic use , Cell Line, Tumor , T-Lymphocytes , Exportin 1 Protein
18.
Blood Adv ; 7(12): 2926-2937, 2023 06 27.
Article in English | MEDLINE | ID: mdl-36827679

ABSTRACT

Selinexor (KPT-330) is a small molecule inhibitor of XPO1, which mediates the transport of tumor suppressor proteins, oncogene messenger RNAs, and other proteins involved in governing cell growthfrom the cell nucleus to the cytoplasm. It is overexpressed in many cancer types. Because eukaryotic translation initiator factor 4E (eIF4E) plays a critical role in protein translation in cancer cells in multiple myeloma (MM), we evaluated the effectiveness of combined inhibition of protein translation and nuclear export in MM. Selinexor, an inhibitor of nuclear protein export, dose-dependently decreased eIF4E, IKZF1, and c-MYC protein levels. Using a doxycycline-inducible-pLKO-Tet-On vector, knockdown of eIF4E significantly enhanced the antiproliferative effects of selinexor, sensitized resistant MM cells to selinexor, and increased apoptosis in MM cells. Immunofluorescent analysis of MM cells showed that the combined treatment increased the localization of residual eIF4E to the nucleus compared with selinexor-only treatment. The overexpression of eIF4E at least partially rescued the effects of selinexor in MM cells by reducing G1 cell cycle arrest and increasing the selinexor-IC50 10-fold. Moreover, the combination of selinexor with pharmacologic inhibitors of protein translation showed synergistic anti-MM effects. These results suggest a synergistic anti-MM effect of selinexor combined with eIF4E inhibitors in vitro. Our work provides a better understanding of the potential mechanism of resistance to selinexor and a rationale for combining selinexor with eIF4E inhibitors for the treatment of MM.


Subject(s)
Karyopherins , Multiple Myeloma , Humans , Active Transport, Cell Nucleus , Karyopherins/metabolism , Karyopherins/pharmacology , Karyopherins/therapeutic use , Eukaryotic Initiation Factor-4E/metabolism , Apoptosis , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Protein Biosynthesis
19.
Endocrinology ; 164(6)2023 04 17.
Article in English | MEDLINE | ID: mdl-37170651

ABSTRACT

Approximately 70% of human breast cancers express estrogen receptor-α (ERα), providing a potential target for endocrine therapy. However, 30% to 40% of patients with ER+ breast cancer still experience recurrence and metastasis, with a 5-year relative overall survival rate of 24%. In this study, we identified nicotinamide phosphoribosyltransferase (NAMPT), an important enzyme in nicotinamide adenine dinucleotide (NAD+) metabolism, to be increased in metastatic breast cancer (MBC) cells treated with fulvestrant (Fulv). We tested whether the blockade of NAD+ production via inhibition of NAMPT synergizes with standard-of-care therapies for ER+ MBC in vitro and in vivo. A synergistic effect was not observed when KPT-9274 was combined with palbociclib or tamoxifen or when Fulv was combined with other metabolic inhibitors. We show that NAMPT inhibitor KPT-9274 and Fulv works synergistically to reduce metastatic tumor burden. RNA-sequencing analysis showed that NAMPT inhibitor in combination with Fulv reversed the expression of gene sets associated with more aggressive tumor phenotype, and metabolomics analysis showed that NAMPT inhibition reduced the abundance of metabolites associated with several key tumor metabolic pathways. Targeting metabolic adaptations in endocrine-resistant MBC is a novel strategy, and alternative approaches aimed at improving the therapeutic response of metastatic ER+ tumors are needed. Our findings uncover the role of ERα-NAMPT crosstalk in MBC and the utility of NAMPT inhibition and antiestrogen combination therapy in reducing tumor burden and metastasis, potentially leading to new avenues of MBC treatment.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Humans , Female , Estrogen Receptor alpha/genetics , NAD/metabolism , Breast Neoplasms/genetics , Acrylamides , Cytokines/metabolism , Cell Line, Tumor
20.
Exp Hematol Oncol ; 12(1): 78, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37715291

ABSTRACT

Leiomyosarcoma (LMS) is a rare soft tissue sarcoma (STS) that begins in smooth muscle tissue and most often initiates in the abdomen or uterus. Compared with other uterine cancers, uterine LMS (ULMS) is an aggressive tumor with poor prognosis and a high risk of recurrence and death, regardless of the stage at presentation. Selinexor is a first-in-class selective inhibitor of nuclear export (SINE) compound that reversibly binds to exportin 1 (XPO1), thereby reactivating tumor suppressor proteins and downregulating the expression of oncogenes and DNA damage repair (DDR) proteins. In this study, we evaluated the effects of selinexor in combination with doxorubicin and eribulin in the LMS tumor model in vitro and in vivo. Treatment of selinexor combined with eribulin showed synergistic effects on tumor growth inhibition in SK-UT1 LMS-derived xenografts. Immunohistochemical assessment of the tumor tissues showed a significantly reduced expression of proliferation (Ki67) and XPO1 markers following combination therapy compared to the control group. Global transcriptome analyses on tumor tissue revealed that the combination therapy regulates genes from several key cancer-related pathways that are differentially expressed in ULMS tumors. To our knowledge, this is the first preclinical study demonstrating the anti-cancer therapeutic potential of using a combination of selinexor and eribulin in vivo. Results from this study further warrant clinical testing a combination of chemotherapy agents with selinexor to reduce the morbidity and mortality from ULMS.

SELECTION OF CITATIONS
SEARCH DETAIL