Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Blood ; 115(3): 559-69, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19965685

ABSTRACT

We have previously shown clinical activity of a mammalian target of rapamycin (mTOR) complex 1 inhibitor in Waldenstrom macroglobulinemia (WM). However, 50% of patients did not respond to therapy. We therefore examined mechanisms of activation of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR in WM, and mechanisms of overcoming resistance to therapy. We first demonstrated that primary WM cells show constitutive activation of the PI3K/Akt pathway, supported by decreased expression of phosphate and tensin homolog tumor suppressor gene (PTEN) at the gene and protein levels, together with constitutive activation of Akt and mTOR. We illustrated that dual targeting of the PI3K/mTOR pathway by the novel inhibitor NVP-BEZ235 showed higher cytotoxicity on WM cells compared with inhibition of the PI3K or mTOR pathways alone. In addition, NVP-BEZ235 inhibited both rictor and raptor, thus abrogating the rictor-induced Akt phosphorylation. NVP-BEZ235 also induced significant cytotoxicity in WM cells in a caspase-dependent and -independent manner, through targeting the Forkhead box transcription factors. In addition, NVP-BEZ235 targeted WM cells in the context of bone marrow microenvironment, leading to significant inhibition of migration, adhesion in vitro, and homing in vivo. These studies therefore show that dual targeting of the PI3K/mTOR pathway is a better modality of targeted therapy for tumors that harbor activation of the PI3K/mTOR signaling cascade, such as WM.


Subject(s)
Imidazoles/therapeutic use , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Oncogene Protein v-akt/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinolines/therapeutic use , Waldenstrom Macroglobulinemia/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cells, Cultured , Drug Delivery Systems/methods , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Inhibitors/therapeutic use , Humans , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred BALB C , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Quinolines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases
13.
Clin Cancer Res ; 15(19): 6035-41, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19755386

ABSTRACT

PURPOSE: Waldenstrom macroglobulinemia is a lymphoplasmacytic lymphoma characterized by widespread involvement of the bone marrow. Despite different options of therapy, Waldenstrom macroglobulinemia is still incurable. Src tyrosine kinase has been shown to play a central role in the regulation of a variety of biological processes, such as cell proliferation, migration, adhesion, and survival in solid tumors. We sought to determine whether the protein tyrosine kinase Src regulates adhesion, migration, and survival in Waldenstrom macroglobulinemia. EXPERIMENTAL DESIGN: We tested the expression of Src tyrosine kinase in Waldenstrom macroglobulinemia and normal cells, and the effect of the specific Src inhibitor AZD0530 on the adhesion, migration, cell cycle, and survival of a Waldenstrom macroglobulinemia cell line and patient samples. Moreover, we tested the effect of AZD0530 on cytoskeletal and cell cycle signaling in Waldenstrom macroglobulinemia. RESULTS: We show that Src is overexpressed in Waldenstrom macroglobulinemia cells compared with control B cells, and that the use of the Src inhibitor AZD0530 led to significant inhibition of adhesion, migration, and cytoskeletal signaling induced by SDF1. Moreover, inhibition of Src activity induced G(1) cell cycle arrest; however, it had minimal effect on survival of Waldenstrom macroglobulinemia cells, and no significant effect on survival of normal cells. CONCLUSIONS: Taken together, these results delineate the role of Src kinase activity in Waldenstrom macroglobulinemia and provide the framework for future clinical trials using Src inhibitors in combination with other drugs to improve the outcome of patients with Waldenstrom macroglobulinemia.


Subject(s)
Chemotaxis , Oncogene Protein pp60(v-src)/physiology , Waldenstrom Macroglobulinemia/pathology , Benzodioxoles/pharmacology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Cytotoxins/pharmacology , Drug Evaluation, Preclinical , Humans , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Waldenstrom Macroglobulinemia/metabolism
14.
Blood ; 114(3): 619-29, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19443661

ABSTRACT

The interaction of multiple myeloma (MM) cells with the bone marrow (BM) milieu plays a crucial role in MM pathogenesis. Stromal cell-derived factor-1 (SDF1) regulates homing of MM cells to the BM. In this study, we examined the role of RhoA and Rac1 GTPases in SDF1-induced adhesion and chemotaxis of MM. We found that both RhoA and Rac1 play key roles in SDF1-induced adhesion of MM cells to BM stromal cells, whereas RhoA was involved in chemotaxis and motility. Furthermore, both ROCK and Rac1 inhibitors reduced SDF1-induced polymerization of actin and activation of LIMK, SRC, FAK, and cofilin. Moreover, RhoA and Rac1 reduced homing of MM cells to BM niches. In conclusion, we characterized the role of RhoA and Rac1 GTPases in SDF1-induced adhesion, chemotaxis, and homing of MM cells to the BM, providing the framework for targeting RhoA and Rac1 GTPases as novel MM therapy.


Subject(s)
Cell Adhesion , Chemokine CXCL12/physiology , Chemotaxis , Multiple Myeloma/pathology , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/physiology , Animals , Bone Marrow , Cytoskeletal Proteins/metabolism , Humans , Mice , Mice, SCID , Stromal Cells , Tumor Cells, Cultured
15.
Clin Lymphoma Myeloma ; 9(1): 94-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19362985

ABSTRACT

The paradigm for the treatment of monoclonal gammophaties has dramatically changed: based on the understanding of the complex interaction between tumor cells and bone marrow microenvironment and the signaling pathways that are deregulated in this process, a number of novel therapeutic agents are now available. For example, 3 novel agents with a targeted anti-multiple myeloma activity, have been FDA approved for the treatment of this disease, namely bortezomib, thalidomide, and lenalidomide. The success of targeted therapy in myeloma has led to the development and investigation of more than 30 new compounds in this disease and in other plasma cell dyscrasias such as Waldenström's macroglobulinemia (WM), both in the preclinical settings and as part of clinical trials. Among them the role of proteasome inhibitors has been widely dissected providing the preclinical basis for clinical trials of combinations of proteasome inhibitors in WM.


Subject(s)
Boronic Acids/therapeutic use , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Pyrazines/therapeutic use , Waldenstrom Macroglobulinemia/drug therapy , Bortezomib , Humans , Waldenstrom Macroglobulinemia/enzymology
16.
Blood ; 113(26): 6669-80, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19401561

ABSTRACT

Detailed genomic studies have shown that cytogenetic abnormalities contribute to multiple myeloma (MM) pathogenesis and disease progression. Nevertheless, little is known about the characteristics of MM at the epigenetic level and specifically how microRNAs regulate MM progression in the context of the bone marrow milieu. Therefore, we performed microRNA expression profiling of bone marrow derived CD138(+) MM cells versus their normal cellular counterparts and validated data by qRT-PCR. We identified a MM-specific microRNA signature characterized by down-expression of microRNA-15a/-16 and overexpression of microRNA-222/-221/-382/-181a/-181b (P < .01). We investigated the functional role of microRNA-15a and -16 and showed that they regulate proliferation and growth of MM cells in vitro and in vivo by inhibiting AKT serine/threonine-protein-kinase (AKT3), ribosomal-protein-S6, MAP-kinases, and NF-kappaB-activator MAP3KIP3. Moreover, miRNA-15a and -16 exerted their anti-MM activity even in the context of the bone marrow milieu in vitro and in vivo. These data indicate that microRNAs play a pivotal role in the biology of MM and represent important targets for novel therapies in MM.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Multiple Myeloma/pathology , RNA, Neoplasm/physiology , Angiogenesis Inhibitors/physiology , Animals , Cell Adhesion , Cell Division/physiology , Clinical Trials, Phase II as Topic/statistics & numerical data , Coculture Techniques , Endothelial Cells/cytology , Gene Expression Profiling , Humans , Mice , Mice, SCID , MicroRNAs/biosynthesis , MicroRNAs/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6/metabolism , Stromal Cells/cytology
17.
Blood ; 113(18): 4341-51, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19139079

ABSTRACT

The interaction of multiple myeloma (MM) cells with their microenvironment in the bone marrow (BM) provides a protective environment and resistance to therapeutic agents. We hypothesized that disruption of the interaction of MM cells with their BM milieu would lead to their sensitization to therapeutic agents such as bortezomib, melphalan, doxorubicin, and dexamethasone. We report that the CXCR4 inhibitor AMD3100 induces disruption of the interaction of MM cells with the BM reflected by mobilization of MM cells into the circulation in vivo, with kinetics that differed from that of hematopoietic stem cells. AMD3100 enhanced sensitivity of MM cell to multiple therapeutic agents in vitro by disrupting adhesion of MM cells to bone marrow stromal cells (BMSCs). Moreover, AMD3100 increased mobilization of MM cells to the circulation in vivo, increased the ratio of apoptotic circulating MM cells, and enhanced the tumor reduction induced by bortezomib. Mechanistically, AMD3100 significantly inhibited Akt phosphorylation and enhanced poly(ADP-ribose) polymerase (PARP) cleavage as a result of bortezomib, in the presence of BMSCs in coculture. These experiments provide a proof of concept for the use of agents that disrupt interaction with the microenvironment for enhancement of efficacy of cytotoxic agents in cancer therapy.


Subject(s)
Anti-HIV Agents/pharmacology , Antineoplastic Agents/pharmacology , Bone Marrow/metabolism , Heterocyclic Compounds/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Receptors, CXCR4/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzylamines , Boronic Acids/pharmacology , Bortezomib , Cell Adhesion/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Coculture Techniques , Colony-Forming Units Assay , Cyclams , Drug Resistance, Neoplasm , Fibronectins/metabolism , Flow Cytometry , Humans , Immunoblotting , Immunoenzyme Techniques , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lentivirus/genetics , Male , Mice , Mice, SCID , Pyrazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Stromal Cells/metabolism , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Blood ; 113(3): 626-34, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18981296

ABSTRACT

Waldenstrom macroglobulinemia (WM) is an incurable low-grade lymphoma characterized by bone marrow (BM) involvement of IgM secreting lymphoplasmacytic cells. The induction of unfolded protein response (UPR) genes ("physiologic" UPR) enables cells to differentiate into professional secretory cells capable of production of high amounts of endoplasmic reticulum (ER)-processed proteins, such as immunoglobulins. Ultimately, the initially cytoprotective UPR triggers an apoptotic cascade if ER stress is not corrected, called proapoptotic/terminal UPR. We show that WM cells inherently express the physiologic UPR machinery compared with normal BM cells, and that increased ER stress leads to proapoptotic/terminal UPR in WM cells. We therefore examined tunicamycin, ER stress inducer, for potential antitumor effects in WM. Tunicamycin induced significant cytotoxicity, apoptosis and cell-cycle arrest, and inhibited DNA synthesis in WM cell lines and primary BM CD19(+) cells from patients with WM with an inhibitory concentration (IC(50)) of 0.5 microg/mL to 1 microg/mL, but not in healthy donor cells. Importantly, coculture of WM cells in the context of the BM microenvironment did not inhibit tunicamycin-induced cytotoxicity. Finally, we demonstrate that ER stress inducer synergizes with other agents used in the treatment of WM. These preclinical studies provide a framework for further evaluation of ER stress inducing agents as therapeutic agents in WM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Tunicamycin/pharmacology , Waldenstrom Macroglobulinemia/drug therapy , Apoptosis/physiology , B-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endoplasmic Reticulum/pathology , Flow Cytometry , Gene Expression , Humans , Immunoblotting , Protein Folding/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/pathology
19.
Blood ; 113(18): 4391-402, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19074725

ABSTRACT

Multilevel genetic characterization of Waldenström macroglobulinemia (WM) is required to improve our understanding of the underlying molecular changes that lead to the initiation and progression of this disease. We performed microRNA-expression profiling of bone marrow-derived CD19(+) WM cells, compared with their normal cellular counterparts and validated data by quantitative reverse-transcription-polymerase chain reaction (qRT-PCR). We identified a WM-specific microRNA signature characterized by increased expression of microRNA-363*/-206/-494/-155/-184/-542-3p, and decreased expression of microRNA-9* (ANOVA; P < .01). We found that microRNA-155 regulates proliferation and growth of WM cells in vitro and in vivo, by inhibiting MAPK/ERK, PI3/AKT, and NF-kappaB pathways. Potential microRNA-155 target genes were identified using gene-expression profiling and included genes involved in cell-cycle progression, adhesion, and migration. Importantly, increased expression of the 6 miRNAs significantly correlated with a poorer outcome predicted by the International Prognostic Staging System for WM. We further demonstrated that therapeutic agents commonly used in WM alter the levels of the major miRNAs identified, by inducing downmodulation of 5 increased miRNAs and up-modulation of patient-downexpressed miRNA-9*. These data indicate that microRNAs play a pivotal role in the biology of WM; represent important prognostic marker; and provide the basis for the development of new microRNA-based targeted therapies in WM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , MicroRNAs/genetics , Neoplasms, Experimental/pathology , Waldenstrom Macroglobulinemia/drug therapy , Waldenstrom Macroglobulinemia/genetics , Aged , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Bone Marrow/metabolism , Bone Marrow/pathology , Boronic Acids/administration & dosage , Bortezomib , Case-Control Studies , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/metabolism , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms, Experimental/drug therapy , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rituximab , Signal Transduction , Waldenstrom Macroglobulinemia/metabolism , Waldenstrom Macroglobulinemia/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Br J Haematol ; 142(5): 775-85, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18537966

ABSTRACT

Waldenstrom macroglobulinaemia (WM) is an incurable lymphoplasmacytic lymphoma with secretion of serum monoclonal immunoglobulin M (IgM). We previously showed that patients receiving cholesterol-lowering statins, had the lowest IgM value in a large cohort of patients with WM. Simvastatin, a 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitor, induced inhibition of proliferation, cytotoxic effect and apoptosis in IgM secreting cell lines as well as in primary CD19(+) WM cells. Interestingly, those effects were reversed by addition of mevalonate and geranylgeranylpyrophosphate, demonstrating that simvastatin inhibited cell growth, survival and IgM secretion on BCWM.1 WM cells by inhibition of geranylgeranylated proteins. Furthermore, simvastatin overcame tumour cell growth induced by co-culture of WM cells with bone-marrow stromal cells. Simvastatin also decreased IgM secretion by BCWM.1 cells at an early time-point that had not affected cell survival. Simvastatin-induced cytotoxicity was preceded by a decrease in Akt (protein kinase B, PKB) and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) pathways at 18 h. In addition, simvastatin induced an increase in stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) MAPK followed by caspase-8, -9, -3 and poly(ADP-ribose) polymerase (PARP) cleavages at 18 h, leading to apoptosis. Furthermore, simvastatin enhanced the cytotoxicity induced by bortezomib, fludarabine and dexamethasone. Our studies therefore support our earlier observation of statin-mediated anti-WM activity and provide the framework for future clinical trials testing simvastatin in WM.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Waldenstrom Macroglobulinemia/drug therapy , Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoglobulin M/metabolism , In Vitro Techniques , Proto-Oncogene Proteins c-akt/metabolism , Waldenstrom Macroglobulinemia/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...