Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Blood Cancer J ; 7(3): e549, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28362441

ABSTRACT

Myeloma is heterogeneous at the molecular level with subgroups of patients characterised by features of epigenetic dysregulation. Outcomes for myeloma patients have improved over the past few decades except for molecularly defined high-risk patients who continue to do badly. Novel therapeutic approaches are, therefore, required. A growing number of epigenetic inhibitors are now available including EZH2 inhibitors that are in early-stage clinical trials for treatment of haematological and other cancers with EZH2 mutations or in which overexpression has been correlated with poor outcomes. For the first time, we have identified and validated a robust and independent deleterious effect of high EZH2 expression on outcomes in myeloma patients. Using two chemically distinct small-molecule inhibitors, we demonstrate a reduction in myeloma cell proliferation with EZH2 inhibition, which leads to cell cycle arrest followed by apoptosis. This is mediated via upregulation of cyclin-dependent kinase inhibitors associated with removal of the inhibitory H3K27me3 mark at their gene loci. Our results suggest that EZH2 inhibition may be a potential therapeutic strategy for the treatment of myeloma and should be investigated in clinical studies.


Subject(s)
Cell Cycle Checkpoints/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Apoptosis/genetics , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Epigenesis, Genetic , Gene Expression Profiling , Histones/metabolism , Humans , Kaplan-Meier Estimate , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Phenotype , Prognosis , Proportional Hazards Models , RNA, Messenger/genetics
3.
Leukemia ; 30(5): 1103-15, 2016 05.
Article in English | MEDLINE | ID: mdl-26859080

ABSTRACT

The role of endothelial progenitor cell (EPC)-mediated vasculogenesis in hematological malignancies is not well explored. Here, we showed that EPCs are mobilized from the bone marrow (BM) to the peripheral blood at early stages of multiple myeloma (MM); and recruited to MM cell-colonized BM niches. Using EPC-defective ID1+/- ID3-/- mice, we found that MM tumor progression is dependent on EPC trafficking. By performing RNA-sequencing studies, we confirmed that endothelial cells can enhance proliferation and favor cell-cycle progression only in MM clones that are smoldering-like and have dependency on endothelial cells for tumor growth. We further confirmed that angiogenic dependency occurs early and not late during tumor progression in MM. By using a VEGFR2 antibody with anti-vasculogenic activity, we demonstrated that early targeting of EPCs delays tumor progression, while using the same agent at late stages of tumor progression is ineffective. Thus, although there is significant angiogenesis in myeloma, the dependency of the tumor cells on EPCs and vasculogenesis may actually precede this step. Manipulating vasculogenesis at an early stage of disease may be examined in clinical trials in patients with smoldering MM, and other hematological malignancies with precursor conditions.


Subject(s)
Multiple Myeloma/pathology , Neovascularization, Pathologic/drug therapy , Animals , Antibodies/therapeutic use , Bone Marrow , Cell Movement , Clone Cells/pathology , Disease Progression , Endothelial Cells/pathology , Mice , Multiple Myeloma/blood supply , Multiple Myeloma/drug therapy , Neovascularization, Pathologic/prevention & control , Secondary Prevention , Vascular Endothelial Growth Factor Receptor-2/immunology
4.
Int J Lab Hematol ; 37 Suppl 1: 108-14, 2015 May.
Article in English | MEDLINE | ID: mdl-25976968

ABSTRACT

Multiple myeloma (MM) is a tumor of indolent, bone marrow (BM) localized, isotype-switched plasma cells. Recently, the diagnostic criteria have been amended to include some patients who would previously have been diagnosed with ultra-high-risk smoldering MM and benefit from immediate treatment. Genetically it can be divided into tumors with different recurrent immunoglobulin heavy chain gene translocations (4p16, 11q13, 6p21, 16q23, 20q11) and tumors characterized by hyperdiploidy with multiple trisomies. Recent genomic studies have shown that almost half of untreated patients have a genetic rearrangements of the MYC locus that result in juxtaposition of ectopic super-enhancers adjacent to MYC, as well as somatic mutations that activate the RAS/MAPK pathway (NRAS, KRAS, BRAF, FGFR3). Mutations that result in constitutive activation of the NFkB pathway and that inactivate TP53, CDKN2C, KDM6A, FAM46C, and DIS3 are also recurrent. A major insight from these studies has been the recognition of the high degree of subclonal heterogeneity in MM, which is more frequent in patients with high-risk genetics. The subclones may alternate in dominance under alternating therapeutic pressure, a phenomenon known as 'clonal tides'. The identification of marked subclonal heterogeneity argues in those patients for the use of therapeutic strategies to maximize response, and long-term suppressive therapies to prevent tumor regrowth and development of additional subclones.


Subject(s)
Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Translocation, Genetic , Trisomy , Gene Rearrangement , Humans , Models, Genetic , Mutation , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics
5.
Oncogene ; 33(3): 316-25, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-23318440

ABSTRACT

The phosphatidylinositol 3'-kinase (PI3K) pathway is dysregulated in multiple myeloma (MM); we therefore tested a highly selective class I PI3K inhibitor, GDC-0941, for anti-myeloma activity. Functional and mechanistic studies were first performed in MM cell lines, then extended to primary MM patient samples cultured in vitro. GDC-0941 was then assessed as a single agent and in various combinations in myeloma tumor xenograft models. We show p110 α and ß are the predominant PI3K catalytic subunits in MM and that a highly selective class I PI3K inhibitor, GDC-0941, has robust activity as a single agent to induce cell cycle arrest and apoptosis of both MM cell lines and patient myeloma cells. Mechanistic studies revealed an induction of cell cycle arrest at G0/G1, with decreased phospho-FoxO1/3a levels, decreased cyclin D1 and c-myc expression, and an increase in the cell cycle inhibitor, p27kip. Induction of apoptosis correlated with increased expression of the pro-apoptotic BH3-only protein BIM, cleaved caspase 3 and cleaved poly (ADP-ribose) polymerase (PARP). In vitro, GDC-0941 synergized with dexamethasone (Dex) and lenalidomide (combination index values of 0.3-0.4 and 0.4-0.8, respectively); in vivo GDC-0941 has anti-myeloma activity and significantly increases the activity of the standard of care agents in several murine xenograft tumor models (additional tumor growth inhibition of 37-53% (Dex) and 22-72% (lenalidomide)). These data provide a clear therapeutic hypothesis for the inhibition of PI3K and provide a rationale for clinical development of GDC-0941 in myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Indazoles/pharmacology , Multiple Myeloma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Dexamethasone/administration & dosage , Female , Humans , Indazoles/administration & dosage , Inhibitory Concentration 50 , Lenalidomide , Mice , Mice, SCID , Multiple Myeloma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sulfonamides/administration & dosage , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Tumor Burden/genetics , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 4: e798, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24030150

ABSTRACT

Multiple myeloma (MM) is an incurable malignancy with an unmet need for innovative treatment options. Histone deacetylase inhibitors (HDACi) are a new class of anticancer agent that have demonstrated activity in hematological malignancies. Here, we investigated the efficacy and safety of HDACi (vorinostat, panobinostat, romidepsin) and novel combination therapies using in vitro human MM cell lines and in vivo preclinical screening utilizing syngeneic transplanted Vk*MYC MM. HDACi were combined with ABT-737, which targets the intrinsic apoptosis pathway, recombinant human tumour necrosis factor-related apoptosis-inducing ligand (rhTRAIL/MD5-1), that activates the extrinsic apoptosis pathway or the DNA methyl transferase inhibitor 5-azacytidine. We demonstrate that in vitro cell line-based studies provide some insight into drug activity and combination therapies that synergistically kill MM cells; however, they do not always predict in vivo preclinical efficacy or toxicity. Importantly, utilizing transplanted Vk*MYC MM, we report that panobinostat and 5-azacytidine synergize to prolong the survival of tumor-bearing mice. In contrast, combined HDACi/rhTRAIL-based strategies, while efficacious, demonstrated on-target dose-limiting toxicities that precluded prolonged treatment. Taken together, our studies provide evidence that the transplanted Vk*MYC model of MM is a useful screening tool for anti-MM drugs and should aid in the prioritization of novel drug testing in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/therapeutic use , Biphenyl Compounds/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Nitrophenols/therapeutic use , Sulfonamides/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Azacitidine/pharmacology , Biphenyl Compounds/pharmacology , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Nitrophenols/pharmacology , Panobinostat , Piperazines/pharmacology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Death Domain/metabolism , Recombinant Proteins , Sulfonamides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology
7.
Leukemia ; 27(12): 2357-65, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23752175

ABSTRACT

RNA interference screening identified XPO1 (exportin 1) among the 55 most vulnerable targets in multiple myeloma (MM). XPO1 encodes CRM1, a nuclear export protein. XPO1 expression increases with MM disease progression. Patients with MM have a higher expression of XPO1 compared with normal plasma cells (P<0.04) and to patients with monoclonal gammopathy of undetermined significance/smoldering MM (P<0.0001). The highest XPO1 level was found in human MM cell lines (HMCLs). A selective inhibitor of nuclear export compound KPT-276 specifically and irreversibly inhibits the nuclear export function of XPO1. The viability of 12 HMCLs treated with KTP-276 was significantly reduced. KPT-276 also actively induced apoptosis in primary MM patient samples. In gene expression analyses, two genes of probable relevance were dysregulated by KPT-276: cell division cycle 25 homolog A (CDC25A) and bromodomain-containing protein 4 (BRD4), both of which are associated with c-MYC pathway. Western blotting and reverse transcription-PCR confirm that c-MYC, CDC25A and BRD4 are all downregulated after treatment with KPT-276. KPT-276 reduced monoclonal spikes in the Vk*MYC transgenic MM mouse model, and inhibited tumor growth in a xenograft MM mouse model. A phase I clinical trial of an analog of KPT-276 is ongoing in hematological malignancies including MM.


Subject(s)
Acrylamides/pharmacology , Biological Transport/drug effects , Cell Nucleus/drug effects , Genome-Wide Association Study , Karyopherins/genetics , Multiple Myeloma/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/pharmacology , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Profiling , Humans , Karyopherins/drug effects , Mice , RNA Interference , Receptors, Cytoplasmic and Nuclear/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays , Exportin 1 Protein
8.
Leukemia ; 25(6): 1026-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21468039

ABSTRACT

Events mediating transformation from the pre-malignant monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma (MM) are unknown. We analyzed gene expression data sets generated on the Affymetrix U133 platform from 22 MGUS and 101 MM patients using gene-set enrichment analysis. Genes overexpressed in MM were enriched for cell cycle, proliferation and MYC activation gene sets. Upon dissecting the relationship between MYC and cell-cycle gene sets, we identified and validated an MYC activation signature dissociated from proliferation. Applying this signature, MYC is activated in 67% of myeloma, but not in MGUS. This was further confirmed by immunohistochemistry (IHC) using membrane CD138 and nuclear MYC double staining. We also showed that almost all tumors with RAS mutations expressed the MYC activation signature, and multiple mechanisms may be involved in activating MYC. MYC activation, whether assessed by gene-expression signature or IHC, is associated with hyperdiploid MM and shorter survival even in tumors that are not proliferative. Bortezomib treatment is able to overcome the survival disadvantage in patients with MYC activation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-myc/metabolism , Boronic Acids/therapeutic use , Bortezomib , Cell Cycle/genetics , Gene Expression Profiling , Humans , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/pathology , Mutation , Proto-Oncogene Proteins c-myc/genetics , Pyrazines/therapeutic use , Survival Rate , ras Proteins/genetics
9.
Leukemia ; 23(12): 2210-21, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19798094

ABSTRACT

Myeloma is a malignant proliferation of monoclonal plasma cells. Although morphologically similar, several subtypes of the disease have been identified at the genetic and molecular level. These genetic subtypes are associated with unique clinicopathological features and dissimilar outcome. At the top hierarchical level, myeloma can be divided into hyperdiploid and non-hyperdiploid subtypes. The latter is mainly composed of cases harboring IgH translocations, generally associated with more aggressive clinical features and shorter survival. The three main IgH translocations in myeloma are the t(11;14)(q13;q32), t(4;14)(p16;q32) and t(14;16)(q32;q23). Trisomies and a more indolent form of the disease characterize hyperdiploid myeloma. A number of genetic progression factors have been identified including deletions of chromosomes 13 and 17 and abnormalities of chromosome 1 (1p deletion and 1q amplification). Other key drivers of cell survival and proliferation have also been identified such as nuclear factor- B-activating mutations and other deregulation factors for the cyclin-dependent pathways regulators. Further understanding of the biological subtypes of the disease has come from the application of novel techniques such as gene expression profiling and array-based comparative genomic hybridization. The combination of data arising from these studies and that previously elucidated through other mechanisms allows for most myeloma cases to be classified under one of several genetic subtypes. This paper proposes a framework for the classification of myeloma subtypes and provides recommendations for genetic testing. This group proposes that genetic testing needs to be incorporated into daily clinical practice and also as an essential component of all ongoing and future clinical trials.


Subject(s)
Genetic Testing/methods , Multiple Myeloma/classification , Classification , Cytogenetic Analysis , Gene Expression Profiling , Humans , Multiple Myeloma/genetics , Prognosis
11.
Leukemia ; 20(5): 807-13, 2006 May.
Article in English | MEDLINE | ID: mdl-16511510

ABSTRACT

Chromosomal hyperdiploidy is the defining genetic signature in 40-50% of myeloma (MM) patients. We characterize hyperdiploid-MM (H-MM) in terms of its clinical and prognostic features in a cohort of 220 H-MM patients entered into clinical trials. Hyperdiploid-myeloma is associated with male sex, kappa immunoglobulin subtype, symptomatic bone disease and better survival compared to nonhyperdiploid-MM (median overall survival 48 vs 35 months, log-rank P = 0.023), despite similar response to treatment. Among 108 H-MM cases with FISH studies for common genetic abnormalities, survival is negatively affected by the existence of immunoglobulin heavy chain (IgH) translocations, especially those involving unknown partners, while the presence of chromosome 13 deletion by FISH did not significantly affect survival (median overall survival 50 vs 47 months, log-rank P = 0.47). Hyperdiploid-myeloma is therefore a unique genetic subtype of MM associated with improved outcome with distinct clinical features. The existence of IgH translocations but not chromosome 13 deletion by FISH negatively impacts survival and may allow further risk stratification of this population of MM patients.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 17/genetics , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Polyploidy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosome Aberrations , Female , Follow-Up Studies , Genes, p53/genetics , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Prognosis , Randomized Controlled Trials as Topic/statistics & numerical data , Retrospective Studies , Survival Rate , Translocation, Genetic , Treatment Outcome
12.
Leuk Res ; 30(3): 266-71, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16111750

ABSTRACT

Hyperdiploid and non-hyperdiploid multiple myeloma represents distinct biological entities characterized by different patterns of genetic changes. We sought to determine whether ploidy category (non-hyperdiploid versus hyperdiploid) remains stable over time from diagnosis to progression. Of the 43 patients studied (39 by flow cytometry DNA index and 4 by a FISH-based index), only five (12%) altered their ploidy status at progression. In three of these patients, the change may possibly be attributable to technical artifacts because of the low absolute change in DNA index. For those who retain their ploidy subtypes, the DNA index change minimally (3.75+/-4.87%). It would appear that the initiating genetic events underlying hyperdiploid and non-hyperdiploid MM that marks them out as distinct entities continue to dominate and persist during disease evolution and progression.


Subject(s)
DNA, Neoplasm/genetics , Multiple Myeloma/genetics , Ploidies , DNA, Neoplasm/analysis , Disease Progression , Female , Flow Cytometry , Humans , In Situ Hybridization, Fluorescence , Male , Multiple Myeloma/pathology
14.
Blood ; 97(8): 2413-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290605

ABSTRACT

Translocations involving fibroblast growth factor receptor 3 (fgfr3) have been identified in about 25% of patients with myeloma. To directly examine the oncogenic potential of fgfr3, murine bone marrow (BM) cells were transduced with retroviral vectors containing either wild-type fgfr3 or an activated mutant form of the receptor, fgfr3-TD. Mice transplanted with FGFR3-TD-expressing BM developed a marked leukocytosis and lethal hematopoietic cell infiltration of multiple tissues within 6 weeks of transplantation. Secondary and tertiary recipients of spleen or BM from primary fgfr3-TD mice also developed tumors within 6 to 8 weeks. Analysis of the circulating tumor cells revealed a pre-B-cell phenotype in most mice, although immature T-lymphoid or mature myeloid populations also predominated in some animals. Enhanced lymphoid but not myeloid colony formation was observed in the early posttransplantation period and only interleukin 7 and FGF-responsive pre-B-cell lines could be established from tumors. Cell expansions in primary recipients appeared polyclonal, whereas tumors in later passages exhibited either clonal B- or T-cell receptor gene rearrangements. Mice transplanted with wild-type FGFR3-expressing BM developed delayed pro-B-cell lymphoma/leukemias approximately 1 year after transplantation. These studies confirm that FGFR3 is transforming and can produce lymphoid malignancies in mice.


Subject(s)
Cell Transformation, Neoplastic/genetics , Hematopoietic Stem Cells/cytology , Lymphoproliferative Disorders/etiology , Multiple Myeloma/genetics , Neoplasm Proteins/physiology , Oncogenes , Protein-Tyrosine Kinases , Animals , Bone Marrow Transplantation , Clone Cells/pathology , Female , Fibroblast Growth Factors/pharmacology , Gene Rearrangement, B-Lymphocyte , Gene Rearrangement, T-Lymphocyte , Humans , Interleukin-7/pharmacology , Leukocytosis/etiology , Leukocytosis/genetics , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/genetics , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Radiation Chimera , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , Recombinant Fusion Proteins/physiology , Spleen/transplantation
15.
Blood ; 97(3): 729-36, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157491

ABSTRACT

The t(4;14) translocation occurs frequently in multiple myeloma (MM) and results in the simultaneous dysregulated expression of 2 potential oncogenes, FGFR3 (fibroblast growth factor receptor 3) from der(14) and multiple myeloma SET domain protein/Wolf-Hirschhorn syndrome candidate gene 1 from der(4). It is now shown that myeloma cells carrying a t(4;14) translocation express a functional FGFR3 that in some cases is constitutively activated by the same mutations that cause thanatophoric dysplasia. As with activating mutations of K-ras and N-ras, which are reported in approximately 40% of patients with MM, activating mutations of FGFR3 occur during tumor progression. However, the constitutive activation of ras and FGFR3 does not occur in the same myeloma cells. Thus the activated forms of these proteins appear to share an overlapping role in tumor progression, suggesting that they also share the signaling cascade. Consistent with this prediction, it is shown that activated FGFR3-when expressed at levels similar to those seen in t(4;14) myeloma-is an oncogene that acts through the MAP kinase pathway to transform NIH 3T3 cells, which can then generate tumors in nude mice. Thus, FGFR3, when overexpressed in MM, may be not only oncogenic when stimulated by FGF ligands in the bone marrow microenvironment, but is also a target for activating mutations that enable FGFR3 to play a ras-like role in tumor progression.


Subject(s)
Multiple Myeloma/genetics , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/genetics , 3T3 Cells , Animals , Cell Transformation, Neoplastic , Disease Progression , Gene Expression , Genes, ras , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Models, Genetic , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Mutation , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/metabolism , Transfection , Translocation, Genetic , Tumor Cells, Cultured
16.
Ann Oncol ; 11 Suppl 1: 131-5, 2000.
Article in English | MEDLINE | ID: mdl-10707795

ABSTRACT

BACKGROUND: Chromosome translocations involving the immunoglobulin heavy chain gene (IgH) on 14q32 are a seminal event in the pathogenesis of many B-cell malignancies. Since myeloma is a post-germinal center tumor of mature, isotype switched plasma cells, we hypothesized that 14q32 translocations would usually involve IgH switch regions. MATERIALS AND METHODS: We analyzed a panel of 21 human myeloma cell lines using a Southern blot assay to detect illegitimate rearrangements involving the switch regions. We then cloned the breakpoints, developed probes for FISH analysis, and characterized the oncogenes dysregulated by the translocations. RESULTS: Only half of the cell lines demonstrated a 14q32 abnormality by conventional karyotypic analysis, but we were able to identify translocations involving IgH switch regions in 15 of 21 lines, including all of the lines in which a 14q32 translocations was not identified by conventional karyotypic analysis. Six cell lines have an Ig translocation involving 11q13 with overexpression of cyclin D1. Six cell lines have an Ig translocation involving 16q23 with overexpression of c-maf. Five lines have an Ig translocations involving 4p16 with overexpression of FGFR3 and a novel gene, MMSET. The 4p16 breakpoints occur within the 5' introns of MMSET, and are associated with IgH-MMSET hybrid mRNA transcripts. The remaining five cell lines have translocations involving other loci, including: 6p25 (MUM1), 8q24 (c-myc), and 21q22 (?AML1). CONCLUSIONS: Recurrent Ig translocations identify at least three distinct molecular subtypes of myeloma. Our long-term goal is to determine if there are phenotypic, prognostic and therapeutic differences associated with these molecular subtypes.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 16 , Immunoglobulins/genetics , Multiple Myeloma/genetics , Translocation, Genetic , Blotting, Southern , Humans , In Situ Hybridization, Fluorescence , Sensitivity and Specificity
17.
Blood ; 95(3): 992-8, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10648414

ABSTRACT

The t(4;14) translocation occurs in 25% of multiple myeloma (MM) and results in both the ectopic expression of fibroblast growth factor receptor 3 (FGFR3) from der4 and immunoglobulin heavy chain-MMSET hybrid messenger RNA transcripts from der14. The subsequent selection of activating mutations of the translocated FGFR3 by MM cells indicates an important role for this signaling pathway in tumor development and progression. To investigate the mechanism by which FGFR3 overexpression promotes MM development, interleukin-6 (IL-6)-dependent murine B9 cells were transduced with retroviruses expressing functional wild-type or constitutively activated mutant FGFR3. Overexpression of mutant FGFR3 resulted in IL-6 independence, decreased apoptosis, and an enhanced proliferative response to IL-6. In the presence of ligand, wild-type FGFR3-expressing cells also exhibited enhanced proliferation and survival in comparison to controls. B9 clones expressing either wild-type FGFR3 at high levels or mutant FGFR3 displayed increased phosphorylation of STAT3 and higher levels of bcl-x(L) expression than did parental B9 cells after cytokine withdrawal. The mechanism of the enhanced cell responsiveness to IL-6 is unknown at this time, but does not appear to be mediated by the mitogen-activated protein kinases SAPK, p38, or ERK. These findings provide a rational explanation for the mechanism by which FGFR3 contributes to both the viability and propagation of the myeloma clone and provide a basis for the development of therapies targeting this pathway.


Subject(s)
Apoptosis , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Gene Expression Regulation, Neoplastic , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/metabolism , Myeloma Proteins/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/biosynthesis , Signal Transduction/physiology , Translocation, Genetic/genetics , Cell Division/drug effects , Chromosomes, Human, Pair 14/ultrastructure , Chromosomes, Human, Pair 4/ultrastructure , DNA-Binding Proteins/metabolism , Germinal Center/pathology , Humans , Interleukin-6/pharmacology , MAP Kinase Signaling System , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Myeloma Proteins/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , STAT3 Transcription Factor , Trans-Activators/metabolism , bcl-X Protein
18.
Mol Cell ; 3(1): 119-23, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10024885

ABSTRACT

Oncogenes are often dysregulated in B cell tumors as a result of a reciprocal translocation involving an immunoglobulin locus. The translocations are caused by errors in two developmentally regulated DNA recombination processes: V(D)J and IgH switch recombination. Both processes share the property of joining discontinuous sequences from one chromosome and releasing intervening sequences as circles that are lost from progeny cells. Here we show that these intervening sequences may instead insert in the genome and that during productive IgH mu-epsilon switch recombination in U266 myeloma tumor cells, a portion of the excised IgH switch intervening sequences containing the 3' alpha-1 enhancer has inserted on chromosome 11q13, resulting in overexpression of the adjacent cyclin D1 oncogene.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Switch Region/genetics , Multiple Myeloma/genetics , Base Sequence , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Oncogenes/genetics , Recombination, Genetic/genetics , Sequence Alignment , Translocation, Genetic/genetics , Tumor Cells, Cultured
19.
Blood ; 92(9): 3025-34, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9787135

ABSTRACT

Previously we reported that a karyotypically silent t(4;14)(p16. 3;q32.3) translocation is present in about 25% of multiple myeloma (MM) tumors, and causes overexpression of FGFR3, which is 50 to 100 kb telomeric to the 4p16 breakpoints. Frequent FGFR3 kinase activating mutations in MM with t(4;14) translocations substantiate an oncogenic role for FGFR3. We now report that the 4p16 breakpoints occur telomeric to and within the 5' introns of a novel gene, MMSET (Multiple Myeloma SET domain). In normal tissues, MMSET has a complex pattern of expression with a short form (647 amino acids [aa]) containing an HMG box and hath region, and an alternatively spliced long form (1365 aa) containing the HMG box and hath region plus 4 PHD fingers and a SET domain. Although t(4;14) translocation results in IgH/MMSET hybrid transcripts, overexpression of MMSET also occurs from endogenous promoters on 4p16. Given the homology to HRX/MLL1/ALL1 at 11q23 that is dysregulated by translocations in acute leukemia, we hypothesize that dysregulation of MMSET contributes to neoplastic transformation in MM with t(4;14) translocation. This is the first example of an IgH translocation that simultaneously dysregulates two genes with oncogenic potential: FGFR3 on der(14) and MMSET on der(4).


Subject(s)
Carrier Proteins , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Genes, Immunoglobulin , High Mobility Group Proteins/genetics , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/genetics , Repressor Proteins , Translocation, Genetic , Amino Acid Sequence , Chromosomes, Human, Pair 14/ultrastructure , Chromosomes, Human, Pair 4/ultrastructure , DNA Primers , DNA, Neoplasm/genetics , Exons/genetics , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase , Humans , Karyotyping , Male , Molecular Sequence Data , Multiple Myeloma/pathology , Oncogene Proteins, Fusion/biosynthesis , Poly A/metabolism , Promoter Regions, Genetic , RNA Splicing , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptor, Fibroblast Growth Factor, Type 3 , Testis/metabolism , Thymus Gland/metabolism , Tumor Cells, Cultured
20.
Blood ; 91(12): 4457-63, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9616139

ABSTRACT

Dysregulation of oncogenes by translocation to an IgH (14q32) or IgL (kappa, 2p11 or lambda, 22q11) locus is a frequent event in the pathogenesis of B-cell tumors. Translocations involving an IgH locus and a diverse but nonrandom array of chromosomal loci occur in most multiple myeloma (MM) tumors even though the translocations often are not detected by conventional cytogenetic analysis. In a continuing analysis of translocations in 21 MM lines, we show that the novel, karyotypically silent t(14;16)(q32.3;q23) translocation is present in 5 MM lines, with cloned breakpoints from 4 lines dispersed over an approximately 500-kb region centromeric to the c-maf proto-oncogene at 16q23. Another line has a t(16;22)(q23;q11), with the breakpoint telomeric to c-maf, so that the translocation breakpoints in these 6 lines bracket c-maf. Only these 6 lines overexpress c-maf mRNA. As predicted for dysregulation of c-maf by translocation, there is selective expression of one c-maf allele in 2 informative lines with translocations. This is the first human tumor in which the basic zipper c-maf transcription factor is shown to function as an oncogene.


Subject(s)
Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 16 , DNA-Binding Proteins/genetics , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Proto-Oncogene Proteins/genetics , Translocation, Genetic , Amino Acid Sequence , Chromosomes, Artificial, Yeast , Gene Expression Regulation, Neoplastic , Genes, Immunoglobulin , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Proto-Oncogene Mas , Proto-Oncogene Proteins c-maf , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL