Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
2.
Blood ; 116(15): e56-65, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20616218

ABSTRACT

To obtain a comprehensive genomic profile of presenting multiple myeloma cases we performed high-resolution single nucleotide polymorphism mapping array analysis in 114 samples alongside 258 samples analyzed by U133 Plus 2.0 expression array (Affymetrix). We examined DNA copy number alterations and loss of heterozygosity (LOH) to define the spectrum of minimally deleted regions in which relevant genes of interest can be found. The most frequent deletions are located at 1p (30%), 6q (33%), 8p (25%), 12p (15%), 13q (59%), 14q (39%), 16q (35%), 17p (7%), 20 (12%), and 22 (18%). In addition, copy number-neutral LOH, or uniparental disomy, was also prevalent on 1q (8%), 16q (9%), and X (20%), and was associated with regions of gain and loss. Based on fluorescence in situ hybridization and expression quartile analysis, genes of prognostic importance were found to be located at 1p (FAF1, CDKN2C), 1q (ANP32E), and 17p (TP53). In addition, we identified common homozygously deleted genes that have functions relevant to myeloma biology. Taken together, these analyses indicate that the crucial pathways in myeloma pathogenesis include the nuclear factor-κB pathway, apoptosis, cell-cycle regulation, Wnt signaling, and histone modifications. This study was registered at http://isrctn.org as ISRCTN68454111.


Subject(s)
Chromosome Aberrations , Chromosomes, Human/genetics , DNA Copy Number Variations/genetics , Multiple Myeloma/genetics , Aged , Chromosome Deletion , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Loss of Heterozygosity , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prognosis , Translocation, Genetic , Uniparental Disomy
3.
Haematologica ; 95(7): 1221-5, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410185

ABSTRACT

A large series of plasma cell dyscrasias (n=2207) was examined for translocations which deregulate the MAF genes, t(14;20)(q32;q12) and t(14;16)(q32;q23), and their disease behavior was compared to a group characterized by the t(4;14)(p16;q32) where CCND2 is also up-regulated. The t(14;20) showed low prevalence in myeloma (27/1830, 1.5%) and smoldering myeloma (1/148, <1%) with a higher incidence in MGUS (9/193, 5% P=0.005). Strong associations with del(13) (76%), non-hyperdiploidy (83%) and gain of 1q (58%) were seen but no association with an IgA M-protein or absence of bone disease was noted. All three translocations were associated with poor outcome in myeloma, but strikingly all t(14;20) MGUS/smoldering myeloma cases (n=10) had stable, low level disease. In contrast, the 10 t(14;16) and 25 t(4;14) MGUS/smoldering myeloma cases were associated with both evolving and non-evolving disease. None of the associated genetic abnormalities helped to predict for progression from MGUS or smoldering myeloma.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Translocation, Genetic , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Multiple Myeloma/diagnosis , Prognosis
4.
Haematologica ; 94(12): 1708-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19996118

ABSTRACT

BACKGROUND: Multiple myeloma, monoclonal gammopathy of undetermined significance and smoldering multiple myeloma harbor common chromosomal abnormalities but the prevalence and relative association of aberrations in these diagnostic groups remains controversial. We investigated these aspects in a large series of patients. DESIGN AND METHODS: Chromosome 13 deletion (Delta13), deletion of TP53, ploidy status and immunoglobulin heavy chain (IgH) translocations were evaluated by fluorescence in situ hybridization in patients with monoclonal gammopathy of undetermined significance (n=189), smoldering multiple myeloma (n=127) and multiple myeloma (n=400). RESULTS: Overall, Delta13 (25%, 34% and 47%), 16q23 deletions (6%, 8% and 21%) and 17p13 deletions (3%, 1% and 10%) were less frequent in patients with monoclonal gammopathy of undetermined significance and smoldering multiple myeloma than in those with multiple myeloma. When distinct genetic groups were considered, no differences in the prevalence of Delta13 were found with t(4;14)(p16;q32) and t(14;16)(q32;q23) among the three diagnostic groups; in contrast Delta13 was rarer in t(11;14)(q13;q32) in patients with monoclonal gammopathy (1/28) and smoldering myeloma (2/13) than in those with multiple myeloma (40%). Similar results were seen for the few t(6;14)(p21;q32) cases: 0/3 patients with monoclonal gammopathy or smoldering myeloma had the Delta13, whereas 4/6 (67%) patients with multiple myeloma and this translocation also had the deletion. In multiple myeloma patients with both an IgH translocation and Delta13, the proportions of cells affected by the two abnormalities were similar, as was the case for t(4;14) and t(14;16) monoclonal gammopathy patients positive for Delta13. In contrast, in monoclonal gammopathy patients with t(14;20)(q32;q11), the translocation was present in almost all cells, while the Delta13 was present in only a sub-population. CONCLUSIONS: These results indicate that the presence and time of occurrence of Delta13 depends on the presence of specific concurrent abnormalities. The observation that Delta13 was extremely rare in monoclonal gammopathy of undetermined significance and smoldering multiple myeloma with translocations directly involving cyclin D genes (CCND1 and CCND3) suggest a possible role of Delta13 in the progression of the disease specifically in these genetic sub-groups. (clinicaltrials.gov identifier: ISRCTN 68454111; UKCRN ID 1176).


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 13/genetics , Paraproteinemias/genetics , Translocation, Genetic , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 4/genetics , Chromosomes, Human, Pair 6/genetics , Cyclin D/genetics , Gene Rearrangement , Humans , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Paraproteinemias/pathology , Time Factors , Tumor Suppressor Protein p53/genetics
5.
Haematologica ; 94(7): 1024-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19454499

ABSTRACT

We report serial genetic studies on a young female patient initially diagnosed with asymptomatic smouldering myeloma who progressed to symptomatic myeloma 4.5 years after presentation. An unbalanced translocation, der(14)t(4;14)(p16;q32), was initially found in all plasma cells plus deletions of other chromosomal regions as detected by array-based comparative genomic hybridization. Deletion of chromosome 13 was observed in a minor population of plasma cells (<20%) for the first two years, increasing to 100% of plasma cells by the time of multiple myeloma diagnosis. Loss of 1p and a rearrangement of MYC were first observed in a small population of plasma cells one year prior to the clinical diagnosis of multiple myeloma, but these subclones increased rapidly in size to become the major population suggesting that they were directly involved in the transformation process. This case report provides a unique insight into the mechanisms of disease progression from smouldering multiple myeloma to multiple myeloma.


Subject(s)
Chromosome Aberrations , Chromosome Deletion , Chromosomes, Human, Pair 1/ultrastructure , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-myc/genetics , Translocation, Genetic , Adult , Comparative Genomic Hybridization , Disease Progression , Female , Humans , In Situ Hybridization, Fluorescence , Models, Genetic , Time Factors , Treatment Outcome
6.
Genes Chromosomes Cancer ; 48(7): 624-36, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19396865

ABSTRACT

Plasma cell leukemia (PCL) is a rare form of monoclonal gammopathy, which can originate de novo or evolve from multiple myeloma (MM) as a terminal leukemic phase. Previous cytogenetic studies of PCL have reported the presence of complex karyotypes with involvement of multiple unidentified chromosomal regions. We report here the analysis of 12 PCL (10 primary and two secondary) by metaphase and FISH analysis combined with oligonucleotide array data (244 k, Agilent). Interphase-FISH results were compared with those from a series of 861 newly diagnosed patients with MM. Cytogenetic analysis was successful on 11 patients, all of whom showed clonal chromosomal abnormalities. Compared with MM, t(11;14)(q13;q32) (42% versus 15%; P = 0.027) and t(14;16)(q32;q23) (25% versus 4%; P = 0.010) were more frequent in PCL, although neither the specific partner chromosome involved in the IgH translocation nor the ploidy status predicted for survival. Chromosomes 1, 8, 13, and 16 showed the highest number of copy number alterations with 8q24 being the chromosomal region most frequently involved. In eight of 12 patients we found abnormalities (translocations, one amplification, small deletions, and duplications) that directly targeted or were very close to MYC. Only four of these changes were detected by routine FISH analysis using commercial probes with the others exclusively detected by arrays. Quantitative reverse transcription polymerase chain reaction demonstrated that these different abnormalities were associated with increased levels of MYC mRNA. We conclude that MYC dysregulation by complex mechanisms is one of the major molecular events in the oncogenesis of PCL.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, Plasma Cell/genetics , Proto-Oncogene Proteins c-myc/genetics , Aged , Aged, 80 and over , Bone Marrow , Chromosome Aberrations , Chromosomes, Human/genetics , Comparative Genomic Hybridization , DNA Mutational Analysis , Female , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Leukemia, Plasma Cell/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Statistics, Nonparametric , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL