Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Cancer Cell ; 39(2): 209-224.e11, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33450196

ABSTRACT

The methylthioadenosine phosphorylase (MTAP) gene is located adjacent to the cyclin-dependent kinase inhibitor 2A (CDKN2A) tumor-suppressor gene and is co-deleted with CDKN2A in approximately 15% of all cancers. This co-deletion leads to aggressive tumors with poor prognosis that lack effective, molecularly targeted therapies. The metabolic enzyme methionine adenosyltransferase 2α (MAT2A) was identified as a synthetic lethal target in MTAP-deleted cancers. We report the characterization of potent MAT2A inhibitors that substantially reduce levels of S-adenosylmethionine (SAM) and demonstrate antiproliferative activity in MTAP-deleted cancer cells and tumors. Using RNA sequencing and proteomics, we demonstrate that MAT2A inhibition is mechanistically linked to reduced protein arginine methyltransferase 5 (PRMT5) activity and splicing perturbations. We further show that DNA damage and mitotic defects ensue upon MAT2A inhibition in HCT116 MTAP-/- cells, providing a rationale for combining the MAT2A clinical candidate AG-270 with antimitotic taxanes.


Subject(s)
DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , Methionine Adenosyltransferase/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Purine-Nucleoside Phosphorylase/genetics , RNA Splicing/drug effects , RNA, Messenger/genetics , Animals , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16 , DNA Damage/genetics , Gene Deletion , HCT116 Cells , HEK293 Cells , Humans , Methionine Adenosyltransferase/genetics , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Neoplasms/genetics , RNA Splicing/genetics , S-Adenosylmethionine/metabolism
4.
Nature ; 547(7661): 109-113, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28658205

ABSTRACT

Activation of the PTEN-PI3K-mTORC1 pathway consolidates metabolic programs that sustain cancer cell growth and proliferation. Here we show that mechanistic target of rapamycin complex 1 (mTORC1) regulates polyamine dynamics, a metabolic route that is essential for oncogenicity. By using integrative metabolomics in a mouse model and human biopsies of prostate cancer, we identify alterations in tumours affecting the production of decarboxylated S-adenosylmethionine (dcSAM) and polyamine synthesis. Mechanistically, this metabolic rewiring stems from mTORC1-dependent regulation of S-adenosylmethionine decarboxylase 1 (AMD1) stability. This novel molecular regulation is validated in mouse and human cancer specimens. AMD1 is upregulated in human prostate cancer with activated mTORC1. Conversely, samples from a clinical trial with the mTORC1 inhibitor everolimus exhibit a predominant decrease in AMD1 immunoreactivity that is associated with a decrease in proliferation, in line with the requirement of dcSAM production for oncogenicity. These findings provide fundamental information about the complex regulatory landscape controlled by mTORC1 to integrate and translate growth signals into an oncogenic metabolic program.


Subject(s)
Adenosylmethionine Decarboxylase/metabolism , Multiprotein Complexes/metabolism , Polyamines/metabolism , Prostatic Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenosylmethionine Decarboxylase/immunology , Animals , Cell Proliferation , Enzyme Activation , Everolimus/therapeutic use , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Metabolomics , Mice , Multiprotein Complexes/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Stability , S-Adenosylmethionine/analogs & derivatives , S-Adenosylmethionine/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
5.
Cell Rep ; 15(3): 574-587, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27068473

ABSTRACT

Homozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.


Subject(s)
Adenosine/analogs & derivatives , Antigens, Neoplasm/metabolism , Gene Deletion , Methionine Adenosyltransferase/metabolism , Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Signal Transduction , Thionucleosides/metabolism , Adenosine/metabolism , Genomics , HCT116 Cells , Humans , Multiprotein Complexes/metabolism , Neoplasms/metabolism , Purine-Nucleoside Phosphorylase/deficiency , RNA, Small Interfering/metabolism
6.
Chem Biol Drug Des ; 84(3): 270-81, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24641677

ABSTRACT

Phospholipase D enzymes cleave lipid substrates to produce phosphatidic acid, an important precursor for many essential cellular molecules. Phospholipase D is a target to modulate cancer-cell invasiveness. This study reports synthesis of a new class of phospholipase D inhibitors based on 1,3-disubstituted-4-amino-pyrazolopyrimidine core structure. These molecules were synthesized and used to perform initial screening for the inhibition of purified bacterial phospholipase D, which is highly homologous to the human PLD1 . Initially tested with the bacterial phospholipase D enzyme, then confirmed with the recombinant human PLD1 and PLD2 enzymes, the molecules presented here exhibited inhibition of phospholipase D activity (IC50 ) in the low-nanomolar to low-micromolar range with both monomeric substrate diC4 PC and phospholipid vesicles and micelles. The data strongly indicate that these inhibitory molecules directly block enzyme/vesicle substrate binding. Preliminary activity studies using recombinant human phospholipase Ds in in vivo cell assays measuring both transphosphatidylation and head-group cleavage indicate inhibition in the mid- to low-nanomolar range for these potent inhibitory novel molecules in a physiological environment.


Subject(s)
Enzyme Inhibitors/chemistry , Phospholipase D/antagonists & inhibitors , Pyrazoles/chemistry , Pyrimidines/chemistry , Binding Sites , Catalytic Domain , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Kinetics , Micelles , Molecular Dynamics Simulation , Phospholipase D/biosynthesis , Phospholipase D/genetics , Phospholipase D/metabolism , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Structure-Activity Relationship
7.
PLoS One ; 8(6): e66982, 2013.
Article in English | MEDLINE | ID: mdl-23785514

ABSTRACT

Glucocorticoid-induced TNF receptor (GITR) plays a crucial role in modulating immune response and inflammation, however the role of GITR in human cancers is poorly understood. In this study, we demonstrated that GITR is inactivated during tumor progression in Multiple Myeloma (MM) through promoter CpG island methylation, mediating gene silencing in primary MM plasma cells and MM cell lines. Restoration of GITR expression in GITR deficient MM cells led to inhibition of MM proliferation in vitro and in vivo and induction of apoptosis. These findings were supported by the presence of induction of p21 and PUMA, two direct downstream targets of p53, together with modulation of NF-κB in GITR-overexpressing MM cells. Moreover, the unbalanced expression of GITR in clonal plasma cells correlated with MM disease progression, poor prognosis and survival. These findings provide novel insights into the pivotal role of GITR in MM pathogenesis and disease progression.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/metabolism , Multiple Myeloma/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , CpG Islands , DNA Methylation , Disease Progression , Gene Expression Regulation, Neoplastic , Glucocorticoid-Induced TNFR-Related Protein/genetics , Humans , Mice , Models, Biological , Multiple Myeloma/genetics , Multiple Myeloma/mortality , NF-kappa B/metabolism , Prognosis , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics
8.
Blood ; 120(8): 1678-86, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22797699

ABSTRACT

miR-155 acts as an oncogenic miR in B-cell lymphoproliferative disorders, including Waldenstrom macroglobulinemia (WM) and chronic lymphocytic leukemia, and is therefore a potential target for therapeutic intervention. However, efficient targeting of miRs in tumor cells in vivo remains a significant challenge for the development of miR-155-based therapeutics for the treatment of B-cell malignancies. In the present study, we show that an 8-mer locked nucleic acid anti-miR-155 oligonucleotide targeting the seed region of miR-155 inhibits WM and chronic lymphocytic leukemia cell proliferation in vitro. Moreover, anti-miR-155 delivered systemically showed uptake in the BM CD19(+) cells of WM-engrafted mice, resulting in the up-regulation of several miR-155 target mRNAs in these cells, and decreased tumor growth significantly in vivo. We also found miR-155 levels to be elevated in stromal cells from WM patients compared with control samples. Interestingly, stromal cells from miR-155-knockout mice led to significant inhibition of WM tumor growth, indicating that miR-155 may also contribute to WM proliferation through BM microenvironmental cells. The results of the present study highlight the therapeutic potential of anti-miR-155-mediated inhibition of miR-155 in the treatment of WM.


Subject(s)
Lymphoma, B-Cell/genetics , MicroRNAs/genetics , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Waldenstrom Macroglobulinemia/genetics , Animals , Cell Proliferation , Female , Gene Silencing , Genetic Therapy , Humans , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Oligonucleotides/genetics , Oligonucleotides, Antisense/genetics , Tumor Cells, Cultured , Waldenstrom Macroglobulinemia/pathology , Waldenstrom Macroglobulinemia/therapy
9.
Blood ; 119(24): 5782-94, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22394600

ABSTRACT

The spread of multiple myeloma (MM) involves (re)circulation into the peripheral blood and (re)entrance or homing of MM cells into new sites of the BM. Hypoxia in solid tumors was shown to promote metastasis through activation of proteins involved in the epithelial-mesenchymal transition (EMT) process. We hypothesized that MM-associated hypoxic conditions activate EMT-related proteins and promote metastasis of MM cells. In the present study, we have shown that hypoxia activates EMT-related machinery in MM cells, decreases the expression of E-cadherin, and, consequently, decreases the adhesion of MM cells to the BM and enhances egress of MM cells to the circulation. In parallel, hypoxia increased the expression of CXCR4, consequently increasing the migration and homing of circulating MM cells to new BM niches. Further studies to manipulate hypoxia to regulate tumor dissemination as a therapeutic strategy are warranted.


Subject(s)
Epithelial-Mesenchymal Transition , Multiple Myeloma/pathology , Animals , Bone Marrow/pathology , Cadherins/metabolism , Cell Adhesion , Cell Hypoxia , Cell Line, Tumor , Chemotaxis , Disease Progression , Humans , Male , Mice , Mice, SCID , Multiple Myeloma/blood , Neoplasm Proteins/metabolism , Receptors, CXCR4/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment
10.
Blood ; 119(6): 1468-78, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22096244

ABSTRACT

Interactions between multiple myeloma (MM) cells and the BM microenvironment play a critical role in the pathogenesis of MM and in the development of drug resistance by MM cells. Selectins are involved in extravasation and homing of leukocytes to target organs. In the present study, we focused on adhesion dynamics that involve P-selectin glycoprotein ligand-1 (PSGL-1) on MM cells and its interaction with selectins in the BM microenvironment. We show that PSGL-1 is highly expressed on MM cells and regulates the adhesion and homing of MM cells to cells in the BM microenvironment in vitro and in vivo. This interaction involves both endothelial cells and BM stromal cells. Using loss-of-function studies and the small-molecule pan-selectin inhibitor GMI-1070, we show that PSGL-1 regulates the activation of integrins and downstream signaling. We also document that this interaction regulates MM-cell proliferation in coculture with BM microenvironmental cells and the development of drug resistance. Furthermore, inhibiting this interaction with GMI-1070 enhances the sensitization of MM cells to bortezomib in vitro and in vivo. These data highlight the critical contribution of PSGL-1 to the regulation of growth, dissemination, and drug resistance in MM in the context of the BM microenvironment.


Subject(s)
Bone Marrow/metabolism , Membrane Glycoproteins/metabolism , Multiple Myeloma/metabolism , P-Selectin/metabolism , Animals , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Glycolipids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Membrane Glycoproteins/genetics , Mice , Mice, SCID , Microscopy, Confocal , Multiple Myeloma/genetics , Multiple Myeloma/pathology , P-Selectin/genetics , Protein Binding/drug effects , RNA Interference , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment/drug effects
11.
Clin Cancer Res ; 18(1): 91-104, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22010211

ABSTRACT

PURPOSE: The ephrin receptors (Eph) are found in a wide range of cancers and correlate with metastasis. In this study, we characterized the role of Eph-B2 receptor in the interaction of Waldenstrom's macroglobulinemia (WM) cells with the bone marrow microenvironment. EXPERIMENTAL DESIGN: We screened the activity of different receptor tyrosine kinases in WM patients and found that Eph-B2 was overexpressed compared with control. Also, we tested the expression of ephrin-B2 ligand on endothelial cells and bone marrow stromal cells (BMSC) isolated from WM patients. We then tested the role of Eph-B2/Ephrin-B2 interaction in the adhesion of WM cells to endothelial cells and BMSCs; the cell signaling induced by the coculture in both the WM cells and the endothelial cells; WM cell proliferation, apoptosis, and cell cycle in vitro and tumor progression in vivo; and in angiogenesis. RESULTS: Eph-B2 receptor was found to be activated in WM patients compared with control, with a 5-fold increase in CD19(+) WM cells, and activated cell adhesion signaling, including focal adhesion kinase, Src, P130, paxillin, and cofilin, but decreased WM cell chemotaxis. Ephrin-B2 ligand was highly expressed on endothelial cells and BMSCs isolated from WM patients and on human umbilical vein endothelial cells and induced signaling in the endothelial cells promoting adhesion and angiogenesis. Blocking of ephrin-B2 or Eph-B2 inhibited adhesion, cytoskeletal signaling, proliferation, and cell cycle in WM cells, which was induced by coculture with endothelial cells and decreased WM tumor progression in vivo. CONCLUSION: Ephrin-B2/Eph-B2 axis regulates adhesion, proliferation, cell cycle, and tumor progression in vivo through the interaction of WM with the cells in the bone marrow microenvironment.


Subject(s)
Cell Adhesion , Cell Proliferation , Ephrin-B2/metabolism , Receptors, Eph Family/metabolism , Waldenstrom Macroglobulinemia/pathology , Waldenstrom Macroglobulinemia/prevention & control , Animals , Apoptosis , Blotting, Western , Bone Marrow/metabolism , Bone Marrow/pathology , Case-Control Studies , Cell Cycle , Cell Movement , Cells, Cultured , Ephrin-B2/antagonists & inhibitors , Ephrin-B2/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Immunoenzyme Techniques , Immunoprecipitation , Mice , Mice, SCID , Neovascularization, Physiologic , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Eph Family/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Waldenstrom Macroglobulinemia/metabolism
12.
Blood ; 118(26): 6860-70, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22045983

ABSTRACT

Mammalian target of rapamycin (mTOR) is a downstream serine/threonine kinase of the PI3K/Akt pathway that integrates signals from the tumor microenvironment to regulate multiple cellular processes. Rapamycin and its analogs have not shown significant activity in multiple myeloma (MM), likely because of the lack of inhibition of TORC2. In the present study, we investigated the baseline activity of the PI3K/Akt/mTOR pathway TORC1/2 in MM cell lines with different genetic abnormalities. TORC1/2 knock-down led to significant inhibition of the proliferation of MM cells, even in the presence of BM stromal cells. We also tested INK128, a dual TORC1/2 inhibitor, as a new therapeutic agent against these MM cell lines. We showed that dual TORC1/2 inhibition is much more active than TORC1 inhibition alone (rapamycin), even in the presence of cytokines or stromal cells. In vitro and in vivo studies showed that p-4EBP1 and p-Akt inhibition could be predictive markers of TORC2 inhibition in MM cell lines. Dual TORC1/2 inhibition showed better inhibition of adhesion to BM microenvironmental cells and inhibition of homing in vivo. These studies form the basis for further clinical testing of TORC1/2 inhibitors in MM.


Subject(s)
Multiple Myeloma/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
13.
Clin Lymphoma Myeloma Leuk ; 11 Suppl 1: S65-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22035751

ABSTRACT

Waldenstrom macroglobulinemia (WM) is a low-grade B-cell lymphoma characterized by widespread involvement of the bone marrow with lymphoplasmacytic cells. In approximately 20% of patients, the malignant clone also involves the lymph nodes and induces hepatosplenomegaly. The mechanisms by which the tumor cells home to the bone marrow and preferentially reside in the marrow niches are not fully elucidated. In this review, we examine the role of the bone marrow microenvironment in the regulation of cell growth, survival and cell dissemination in WM. We also summarize specific regulators of niche-dependent tumor proliferation in WM. These include chemokines, adhesion molecules, Src/PI3K/Akt/mTOR signaling, NF-kB activation, and micro-RNA regulation in WM. Targeting these pathways in clinical trials could lead to significant responses in this rare disease.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/pathology , Waldenstrom Macroglobulinemia/drug therapy , Waldenstrom Macroglobulinemia/pathology , Animals , Bone Marrow/metabolism , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Signal Transduction , Tumor Microenvironment , Waldenstrom Macroglobulinemia/metabolism
14.
Clin Lymphoma Myeloma Leuk ; 11(1): 109-11, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21454206

ABSTRACT

Epigenetics represent heritable changes in gene expression that are not due to any alteration in the DNA sequence. One of the best-known epigenetic markers is histone acetylation, which has been shown to be deregulated in neoplastic diseases, including B-cell malignancies, such as Waldenström's Macroglobulinemia (WM), a low-grade B-cell lymphoma characterized by the presence of lymphoplasmacytic cells in the bone marrow and a serum monoclonal immunoglobulin M in the circulation. It has been recently demonstrated that microRNAs may be responsible for modulating histone acetylation in WM cells, thus providing the preclinical evidences for using microRNA-based therapeutic strategies in this disease.


Subject(s)
MicroRNAs/genetics , Waldenstrom Macroglobulinemia/genetics , Epigenomics , Female , Humans , Immunophenotyping , Male , Waldenstrom Macroglobulinemia/pathology
15.
Clin Lymphoma Myeloma Leuk ; 11(1): 118-20, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21454209

ABSTRACT

The widespread involvement of the bone marrow with tumor cells indicates that there is continuous cell trafficking of WM cells in and out of the bone marrow leading to cell dissemination in the bone marrow and in the lymph nodes in many patients with WM. The interaction of the WM cells with the bone marrow is critical for the regulation of cell proliferation, cell cycle, drug resistance as well as cell dissemination and trafficking. Advances in understanding the interaction of the tumor clone with the BM microenvironment have led to the development of therapeutic agents that not only target the tumor clone but also regulate the bone marrow microenvironment. Here, we review the role of the cellular and liquid bone marrow compartments in the regulation of cell proliferation and dissemination in WM.


Subject(s)
Bone Marrow/pathology , Waldenstrom Macroglobulinemia/pathology , Cell Growth Processes/physiology , Female , Humans , Male
16.
Clin Cancer Res ; 17(13): 4389-99, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21521775

ABSTRACT

PURPOSE: There is no standard of therapy for the treatment of Waldenström macroglobulinemia (WM), therefore there is a need for the development of new agents. Fibroblast growth factor receptor 3 (FGFR3) was shown to play a major role in several types in cancer. Dovitinib, an inhibitor of FGFR3, was effective in hematologic malignancies. In this study, we tested FGFR3 as a therapeutic target in WM and tested the effect of dovitinib on cell proliferation and apoptosis of WM cells in the context of BM microenvironment. METHODS: The expression of FGFR3 in WM cells was tested using immunofluorescence and flow cytometry. Cell signaling in response to stimulation with FGF3 and stromal cells, and its inhibition by dovitinib was performed using immunoblotting. Cell survival and cell proliferation were assessed by MTT and BrdU assays. Apoptosis was measured by detection of APO-2.7 and cleavage of caspase-3 using flow cytometry. Cell cycle was performed by PI staining of cells and flow cytometry. The combinatory effect of dovitinib with other drugs was analyzed using Calcusyn software. The effect of dovitinib was tested in vivo. RESULTS: FGFR3 was overexpressed in WM cells and its activation induced cell proliferation. Inhibition of FGFR3 with dovitinib decreased cell survival, increased apoptosis, and induced cell cycle arrest. Inhibition of FGFR3 by dovitinib reduced the interaction of WM to bone marrow components, and reversed its proliferative effect. Dovitinib had an additive effect with other drugs. Moreover, dovitinib reduced WM tumor progression in vivo. CONCLUSION: We report that FGFR3 is a novel therapeutic target in WM, and suggest dovitinib for future clinical trial the treatment of patients with WM.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation, Neoplastic , Protein Kinase Inhibitors/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Stromal Cells/drug effects , Waldenstrom Macroglobulinemia/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Enzyme Activation/drug effects , Humans , Mice , Mice, SCID , Receptor, Fibroblast Growth Factor, Type 3/genetics , Stromal Cells/metabolism , Tumor Microenvironment/drug effects , Waldenstrom Macroglobulinemia/genetics
17.
Clin Cancer Res ; 17(7): 1753-64, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21355079

ABSTRACT

PURPOSE: Primary Waldenstrom's Macroglobulinemia (WM) cells present with a significantly higher level of the immunoproteasome compared with the constitutive proteasome. It has been demonstrated that selective inhibition of the chymotrypsin-like (CT-L) activity of constitutive-(c20S) and immuno-(i20S) proteasome represents a valid strategy to induce antineoplastic effect in hematologic tumors. We therefore evaluated carfilzomib, a potent selective, irreversible inhibitor of the CT-L activity of the i20S and c20S in WM cells. EXPERIMENTAL DESIGN: We tested the effect of carfilzomib on survival and proliferation of primary WM cells, as well as of other IgM-secreting lymphoma cell lines. Carfilzomib-dependent mechanisms of induced apoptosis in WM cells, and its effect on WM cells in the context of bone marrow (BM) microenvironment have been also evaluated. Moreover, the combinatory effect of carfilzomib and bortezomib has been investigated. In vivo studies have been performed. RESULTS: We demonstrated that carfilzomib targeted the CT-L activity of both i20S and c20S, which led to the induction of toxicity in primary WM cells, as well as in other IgM-secreting lymphoma cells. Importantly, carfilzomib targeted WM cells even in the context of BM milieu. In addition, carfilzomib induced apoptosis through c-jun-N-terminal-kinase activation, caspase cleavage, and initiation of unfolded protein response. Importantly, the combination of carfilzomib and bortezomib synergistically inhibited CT-L activity, as well as caspase-, PARP-cleavage and GRP94 expression. Antitumor activity of carfilzomib has been validated in vivo. CONCLUSIONS: These findings suggest that targeting i20S and c20S CT-L activity by carfilzomib represents a valid antitumor strategy in WM and other IgM-secreting lymphomas.


Subject(s)
Antineoplastic Agents/pharmacology , Chymotrypsin/antagonists & inhibitors , Oligopeptides/pharmacology , Proteasome Inhibitors , Serine Proteinase Inhibitors/pharmacology , Waldenstrom Macroglobulinemia/drug therapy , Animals , Apoptosis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Boronic Acids/pharmacology , Bortezomib , Caspases/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chemokine CXCL12/pharmacology , Coculture Techniques , DNA Fragmentation , Drug Synergism , Enzyme Activation , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lymphoma/pathology , Mice , Mice, SCID , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Pyrazines/pharmacology , Unfolded Protein Response , Waldenstrom Macroglobulinemia/enzymology , Waldenstrom Macroglobulinemia/pathology
18.
Ther Adv Hematol ; 2(4): 267-72, 2011 Aug.
Article in English | MEDLINE | ID: mdl-23556094

ABSTRACT

Waldenstrom macroglobulinemia (WM) is a low-grade B-cell lymphoproliferative disorder characterized primarily by specific homing and growth of tumor cells within the bone marrow niches. The progressive growth of tumor cells throughout the bone marrow indicates that the tumor cells are capable of homing and adhering to specific niches that allow growth, survival and drug resistance. In this review we highlight the interaction of the tumor cells in WM and the bone marrow microenvironment including bone marrow stromal cells, endothelial cells and mast cells. Migration, adhesion and downstream activation of signaling pathways leads to cell trafficking and cell dissemination in WM. Future therapeutic agents need to target not only the tumor clone, but also its close interaction with the bone marrow microenvironment.

19.
Blood ; 116(9): 1506-14, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20519629

ABSTRACT

Waldenström macroglobulinemia (WM) cells present with increased expression of microRNA-206 (miRNA-206) and reduced expression of miRNA-9*. Predicted miRNA-206- and -9*-targeted genes include histone deacetylases (HDACs) and histone acetyl transferases (HATs), indicating that these miRNAs may play a role in regulating histone acetylation. We were able to demonstrate that primary WM cells are characterized by unbalanced expression of HDACs and HATs, responsible for decreased acetylated histone-H3 and -H4, and increased HDAC activity. We next examined whether miRNA-206 and -9* modulate the aberrant expression of HDAC and HATs in WM cells leading to increased transcriptional activity. We found that restoring miRNA-9* levels induced toxicity in WM cells, supported by down-modulation of HDAC4 and HDAC5 and up-regulation of acetyl-histone-H3 and -H4. These, together with inhibited HDAC activity, led to induction of apoptosis and autophagy in WM cells. To further confirm that miRNA-9*-dependent modulation of histone acetylation is responsible for induction of WM cytotoxicity, a novel class of HDAC inhibitor (LBH589) was used; we confirmed that inhibition of HDAC activity leads to toxicity in this disease. These findings confirm that histone-modifying genes and HDAC activity are deregulated in WM cells, partially driven by the aberrant expression of miRNA-206 and -9* in the tumor clone.


Subject(s)
Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , MicroRNAs/physiology , Repressor Proteins/metabolism , Waldenstrom Macroglobulinemia/metabolism , Waldenstrom Macroglobulinemia/pathology , Acetylation , Apoptosis , Autophagy , Blotting, Western , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Histone Acetyltransferases/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histones/metabolism , Humans , RNA, Messenger/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Waldenstrom Macroglobulinemia/genetics
20.
Blood ; 115(20): 4051-60, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20110419

ABSTRACT

Proteasome inhibition represents a valid antitumor approach and its use has been validated in Waldenström macroglobulinemia (WM), where bortezomib has been successfully tested in clinical trials. Nevertheless, a significant fraction of patients relapses, and many present toxicity due to its off-target effects. Selective inhibition of the chymotrypsin-like (CT-L) activity of constitutive proteasome 20S (c20S) and immunoproteasome 20S (i20S) represents a sufficient and successful strategy to induce antineoplastic effect in hematologic tumors. We therefore studied ONX0912, a novel selective, irreversible inhibitor of the CT-L activity of i20S and c20S. Primary WM cells express higher level of i20S compared with c20S, and that ONX0912 inhibited the CT-L activity of both i20S and c20S, leading to induction of toxicity in primary WM cells, as well as of apoptosis through c-Jun N-terminal kinase activation, nuclear factor kappaB (NF-kappaB) inhibition, caspase cleavage, and initiation of the unfolded protein response. Importantly, ONX0912 exerted toxicity in WM cells, by reducing bone marrow (BM)-derived interleukin-6 (IL-6) and insulin-like growth factor 1 (IGF-1) secretion, thus inhibiting BM-induced p-Akt and phosphorylated extracellular signal-related kinase (p-ERK) activation in WM cells. These findings suggest that targeting i20S and c20S CT-L activity by ONX0912 represents a valid antitumor therapy in WM.


Subject(s)
Chymotrypsin/antagonists & inhibitors , Dipeptides/pharmacology , Lymphoma/enzymology , Lymphoma/immunology , Proteasome Inhibitors , Thiazoles/pharmacology , Waldenstrom Macroglobulinemia/enzymology , Apoptosis , Chymotrypsin/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Insulin-Like Growth Factor I/metabolism , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lymphoma/drug therapy , NF-kappa B/genetics , NF-kappa B/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Waldenstrom Macroglobulinemia/drug therapy , Waldenstrom Macroglobulinemia/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...