Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Leukemia ; 31(8): 1706-1714, 2017 08.
Article in English | MEDLINE | ID: mdl-27909306

ABSTRACT

Chromosomal region 1p22 is deleted in ⩾20% of multiple myeloma (MM) patients, suggesting the presence of an unidentified tumor suppressor. Using high-resolution genomic profiling, we delimit a 58 kb minimal deleted region (MDR) on 1p22.1 encompassing two genes: ectopic viral integration site 5 (EVI5) and ribosomal protein L5 (RPL5). Low mRNA expression of EVI5 and RPL5 was associated with worse survival in diagnostic cases. Patients with 1p22 deletion had lower mRNA expression of EVI5 and RPL5, however, 1p22 deletion status is a bad predictor of RPL5 expression in some cases, suggesting that other mechanisms downregulate RPL5 expression. Interestingly, RPL5 but not EVI5 mRNA levels were significantly lower in relapsed patients responding to bortezomib and; both in newly diagnosed and relapsed patients, bortezomib treatment could overcome their bad prognosis by raising their progression-free survival to equal that of patients with high RPL5 expression. In conclusion, our genetic data restrict the MDR on 1p22 to EVI5 and RPL5 and although the role of these genes in promoting MM progression remains to be determined, we identify RPL5 mRNA expression as a biomarker for initial response to bortezomib in relapsed patients and subsequent survival benefit after long-term treatment in newly diagnosed and relapsed patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Chromosome Deletion , Chromosomes, Human, Pair 1 , Multiple Myeloma/genetics , Ribosomal Proteins/genetics , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Humans , MDS1 and EVI1 Complex Locus Protein , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Mutation , Proto-Oncogenes/genetics , RNA, Messenger/analysis , Transcription Factors/genetics
3.
Am J Transplant ; 16(2): 414-25, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26780579

ABSTRACT

The molecular pathogenesis of posttransplant diffuse large B cell lymphoma (PT-DLBCL) is largely unknown. We have recently shown that Epstein-Barr virus-positive (EBV(+)) and -negative (EBV(-)) PT-DLBCL have distinct gene expression profiles, and the transcriptomic profile of EBV(-) PT-DLBCL is similar to that of DLBCL in immunocompetent individuals (IC-DLBCL). To validate these observations at the genomic level, we performed array-comparative genome hybridization (aCGH) analysis of 21 EBV(+) PT-DLBCL, 6 EBV(-) PT-DLBCL, and 11 control IC-DLBCL, and subsequently combined genomic and transcriptomic data. The analysis showed that EBV(+) and EBV(-) PT-DLBCL have distinct aCGH profiles and shared only one recurrent imbalance. EBV(-) PT-DLBCL, however, displayed at least 10 aberrations recurrent in IC-DLBCL, among which characteristic gain of 3/3q and 18q, and loss of 6q23/TNFAIP3 as well as 9p21/CDKN2A. The most prevalent aberration in EBV(+) PT-DLBCL was gain/amplification of 9p24.1 targeting PDCD1LG2/PDL2. Our data indicate that the FOXP1 oncogene and the tumor suppressor CDKNA2 implicated in EBV(-) DLBCL, do not play a critical role in the pathogenesis of EBV(+) PT-DLBCL. Altogether, genomic profiling of PT-/IC-DLBCL confirms that EBV(-) and EBV(+) PT-DLBCL are distinct entities, while EBV(-) PT-DLBCL has features in common with IC-DLBCL. These findings support the hypothesis that EBV(-) PT-DLBCL are de novo lymphomas in transplant recipients.


Subject(s)
Biomarkers, Tumor/genetics , Epstein-Barr Virus Infections/genetics , Gene Expression Profiling , Genomics/methods , Lymphoma, Large B-Cell, Diffuse/genetics , Postoperative Complications/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Child , Child, Preschool , Comparative Genomic Hybridization , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Female , Follow-Up Studies , Graft Rejection , Graft Survival , Herpesvirus 4, Human , Humans , Immunoenzyme Techniques , In Situ Hybridization , Lymphoma, Large B-Cell, Diffuse/surgery , Lymphoma, Large B-Cell, Diffuse/virology , Male , Middle Aged , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
4.
Leukemia ; 29(6): 1390-401, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25533804

ABSTRACT

Although anaplastic large-cell lymphomas (ALCL) carrying anaplastic lymphoma kinase (ALK) have a relatively good prognosis, aggressive forms exist. We have identified a novel translocation, causing the fusion of the TRAF1 and ALK genes, in one patient who presented with a leukemic ALK+ ALCL (ALCL-11). To uncover the mechanisms leading to high-grade ALCL, we developed a human patient-derived tumorgraft (hPDT) line. Molecular characterization of primary and PDT cells demonstrated the activation of ALK and nuclear factor kB (NFkB) pathways. Genomic studies of ALCL-11 showed the TP53 loss and the in vivo subclonal expansion of lymphoma cells, lacking PRDM1/Blimp1 and carrying c-MYC gene amplification. The treatment with proteasome inhibitors of TRAF1-ALK cells led to the downregulation of p50/p52 and lymphoma growth inhibition. Moreover, a NFkB gene set classifier stratified ALCL in distinct subsets with different clinical outcome. Although a selective ALK inhibitor (CEP28122) resulted in a significant clinical response of hPDT mice, nevertheless the disease could not be eradicated. These data indicate that the activation of NFkB signaling contributes to the neoplastic phenotype of TRAF1-ALK ALCL. ALCL hPDTs are invaluable tools to validate the role of druggable molecules, predict therapeutic responses and implement patient specific therapies.


Subject(s)
Drug Resistance, Neoplasm , Lymphoma, Large-Cell, Anaplastic/genetics , NF-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/genetics , TNF Receptor-Associated Factor 1/genetics , Translocation, Genetic/genetics , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Flow Cytometry , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , In Situ Hybridization, Fluorescence , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/mortality , Mice , Mice, Inbred NOD , NF-kappa B/genetics , Positive Regulatory Domain I-Binding Factor 1 , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TNF Receptor-Associated Factor 1/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
5.
Am J Transplant ; 13(5): 1305-16, 2013 May.
Article in English | MEDLINE | ID: mdl-23489474

ABSTRACT

Posttransplant patients are at risk of developing a potentially life-threatening posttransplantation lymphoproliferative disorder (PTLD), most often of diffuse large B cell lymphoma (DLBCL) morphology and associated with Epstein-Barr Virus (EBV) infection. The aim of this study was to characterize the clinicopathological and molecular-genetic characteristics of posttransplant DLBCL and to elucidate whether EBV(+) and EBV(-) posttransplant DLBCL are biologically different. We performed gene expression profiling studies on 48 DLBCL of which 33 arose posttransplantation (PT-DLBCL; 72% EBV+) and 15 in immunocompetent hosts (IC-DLBCL; none EBV+). Unsupervised hierarchical analysis showed clustering of samples related to EBV-status rather than immune status. Except for decreased T cell signaling these cases were inseparable from EBV(-) IC-DLBCL. In contrast, a viral response signature clearly segregated EBV(+) PT-DLBCL from EBV(-) PT-DLBCL and IC-DLBCL cases that were intermixed. The broad EBV latency profile (LMP1+/EBNA2+) was expressed in 59% of EBV(+) PT-DLBCL and associated with a more elaborate inflammatory response compared to intermediate latency (LMP1+/EBNA2-). Inference analysis revealed a role for innate and tolerogenic immune responses (including VSIG4 and IDO1) in EBV(+) PT-DLBCL. In conclusion we can state that the EBV signature is the most determining factor in the pathogenesis of EBV(+) PT-DLBCL.


Subject(s)
Epstein-Barr Virus Infections/complications , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Lymphoproliferative Disorders/genetics , Organ Transplantation , Viral Proteins/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Female , Follow-Up Studies , Humans , In Situ Hybridization , Lymphoproliferative Disorders/complications , Lymphoproliferative Disorders/metabolism , Male , Middle Aged , Retrospective Studies , Viral Proteins/genetics , Virus Latency , Young Adult
6.
Leukemia ; 24(8): 1487-97, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20520640

ABSTRACT

Mucosa-associated lymphoid tissue (MALT) lymphoma is characterized by t(11;18)(q21;q21)/API2-MALT1, t(1;14)(p22;q32)/BCL10-IGH and t(14;18)(q32;q21)/IGH-MALT1, which commonly activate the nuclear factor (NF)-kappaB pathway. Gastric MALT lymphomas harboring such translocations usually do not respond to Helicobacter pylori eradication, while most of those without translocation can be cured by antibiotics. To understand the molecular mechanism of these different MALT lymphoma subgroups, we performed gene expression profiling analysis of 21 MALT lymphomas (13 translocation-positive, 8 translocation-negative). Gene set enrichment analysis (GSEA) of the NF-kappaB target genes and 4394 additional gene sets covering various cellular pathways, biological processes and molecular functions have shown that translocation-positive MALT lymphomas are characterized by an enhanced expression of NF-kappaB target genes, particularly toll like receptor (TLR)6, chemokine, CC motif, receptor (CCR)2, cluster of differentiation (CD)69 and B-cell CLL/lymphoma (BCL)2, while translocation-negative cases were featured by active inflammatory and immune responses, such as interleukin-8, CD86, CD28 and inducible T-cell costimulator (ICOS). Separate analyses of the genes differentially expressed between translocation-positive and -negative cases and measurement of gene ontology term in these differentially expressed genes by hypergeometric test reinforced the above findings by GSEA. Finally, expression of TLR6, in the presence of TLR2, enhanced both API2-MALT1 and BCL10-mediated NF-kappaB activation in vitro. Our findings provide novel insights into the molecular mechanism of MALT lymphomas with and without translocation, potentially explaining their different clinical behaviors.


Subject(s)
Lymphoma, B-Cell, Marginal Zone/genetics , NF-kappa B/metabolism , Translocation, Genetic , Adaptor Proteins, Signal Transducing/genetics , B-Cell CLL-Lymphoma 10 Protein , Gene Expression Profiling , Humans , Immunohistochemistry , Lymphoma, B-Cell, Marginal Zone/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 6/genetics
8.
Leukemia ; 23(1): 125-33, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18923437

ABSTRACT

Episomes with the NUP214-ABL1 fusion gene have been observed in 6% of T-ALL. In this multicentric study we collected 27 cases of NUP214-ABL1-positive T-ALL. Median age was 15 years with male predominance. Outcome was poor in 12 patients. An associated abnormality involving TLX1 or TLX3 was found in all investigated cases. Fluorescent in situ hybridization revealed a heterogeneous pattern of NUP214-ABL1 amplification. Multiple episomes carrying the fusion were detected in 24 patients. Episomes were observed in a significant number of nuclei in 18 cases, but in only 1-5% of nuclei in 6. In addition, intrachromosomal amplification (small hsr) was identified either as the only change or in association with episomes in four cases and two T-ALL cell lines (PEER and ALL-SIL). One case showed insertion of apparently non-amplified NUP214-ABL1 sequences at 14q12. The amplified sequences were analyzed using array-based CGH.These findings confirm that the NUP214-ABL1 gene requires amplification for oncogenicity; it is part of a multistep process of leukemogenesis; and it can be a late event present only in subpopulations. Data also provide in vivo evidence for a model of episome formation, amplification and optional reintegration into the genome. Implications for the use of kinase inhibitors are discussed.


Subject(s)
Gene Amplification , Leukemia-Lymphoma, Adult T-Cell/genetics , Oncogene Proteins, Fusion/genetics , Adolescent , Adult , Cell Line, Tumor , Child , Child, Preschool , Female , Homeodomain Proteins/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/etiology , Male , Middle Aged , Plasmids , Proto-Oncogene Proteins/genetics , Sex Factors , Treatment Outcome , Young Adult
9.
Leukemia ; 21(7): 1532-44, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17495977

ABSTRACT

The biologic and pathologic features of B-cell malignancies bearing a translocation t(14;19)(q32;q13) leading to a fusion of IGH and BCL3 are still poorly described. Herein we report the results of a comprehensive cytogenetic, fluorescence in situ hybridization (FISH), molecular and histopathological survey of a large series of B-cell malignancies with t(14;19) or variant translocations. A total of 56 B-cell malignancies with a FISH-proven BCL3 involvement were identified with the translocation partners being IGH (n=51), IGL (n=2), IGK (n=2) and a non-IG locus (n=1). Hierarchical clustering of chromosomal changes associated with the t(14;19) indicated the presence of two different groups of IG/BCL3-positive lymphatic neoplasias. The first group included 26 B-cell malignancies of various histologic subtypes containing a relatively high number of chromosomal changes and mostly mutated IgVH genes. This cluster displayed three cytogenetic branches, one with rearrangements in 7q, another with deletions in 17p and a third one with rearrangements in 1q and deletions in 6q and 13q. The second group included 19 cases, mostly diagnosed as B-cell chronic lymphocytic leukemia (B-CLL), and characterized by few additional chromosomal changes (e.g. trisomy 12) and unmutated IgVH genes. In conclusion, our study indicates that BCL3 translocations are not restricted to B-CLL but present in a heterogeneous group of B-cell malignancies.


Subject(s)
Leukemia, B-Cell/genetics , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic , Adult , Aged , B-Cell Lymphoma 3 Protein , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 19 , Cytogenetic Analysis , Female , Gene Rearrangement , Genes, Immunoglobulin , Histocytochemistry , Humans , In Situ Hybridization, Fluorescence , Leukemia, B-Cell/classification , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/classification , Lymphoma, B-Cell/pathology , Male , Middle Aged
11.
J Pathol ; 210(2): 155-62, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16874743

ABSTRACT

There is a diagnostic grey zone between classic Hodgkin lymphoma (cHL) and some non-Hodgkin lymphoma (NHL), including primary mediastinal B cell lymphoma, diffuse large B cell lymphoma, and anaplastic large cell lymphoma. They all have some morphological and/or phenotypic features in common. To investigate this, we undertook an expression profiling study of these lymphomas using comparative expressed sequence hybridization. This technique detects chromosomal regions that are differentially expressed between a test and a reference tissue in a manner similar to comparative genomic hybridization, and is particularly suitable when the number of informative biopsies is limited. Using this approach, we identified a unique expression profile for all lymphoma types investigated. Unsupervised hierarchical cluster analysis of the acquired data showed that cHL separates from all investigated NHLs, including ALCL-like HL. Moreover, anaplastic lymphoma kinase (ALK)-negative ALCL clustered in a separate branch together with ALCL-like HL. Thus, analysing the neoplastic cells concurrently with their microenvironment, ALK-negative ALCL and ALCL-like HL seem to be related to each other, while cHL constitutes a separate lymphoma entity.


Subject(s)
Hodgkin Disease/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Cluster Analysis , DNA, Neoplasm/genetics , Diagnosis, Differential , Female , Gene Expression Profiling/methods , Hodgkin Disease/diagnosis , Hodgkin Disease/pathology , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Neoplasm Staging , Nucleic Acid Hybridization/methods
12.
Leukemia ; 19(8): 1299-305, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15944719

ABSTRACT

The transcription factor Forkhead box protein P1 (FOXP1) is highly expressed in a proportion of diffuse large B-cell lymphoma (DLBCL). In this report, we provide cytogenetic and fluorescence in situ hybridization (FISH) data showing that FOXP1 (3p13) is recurrently targeted by chromosome translocations. The genomic rearrangement of FOXP1 was identified by FISH in three cases with a t(3;14)(p13;q32) involving the immunoglobulin heavy chain (IGH) locus, and in one case with a variant t(2;3) affecting sequences at 2q36. These aberrations were associated with strong expression of FOXP1 protein in tumor cells, as demonstrated by immunohistochemistry (IHC). The cases with t(3p13) were diagnosed as DLBCL ( x 1), gastric MALT lymphoma ( x 1) and B-cell non-Hodgkin's lymphoma, not otherwise specified ( x 2). Further IHC and FISH studies performed on 98 cases of DLBCL and 93 cases of extranodal marginal zone lymphoma showed a high expression of FOXP1 in approximately 13 and 12% of cases, respectively. None of these cases showed, however, FOXP1 rearrangements by FISH. However, over-representation of the FOXP1 locus found in one additional case of DLBCL may represent another potential mechanism underlying an increased expression of this gene.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/genetics , Repressor Proteins/genetics , Translocation, Genetic , Aged , Chromosome Aberrations , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 3 , Female , Forkhead Transcription Factors , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Humans , Lymphoma, B-Cell/genetics , Male , Middle Aged , Retrospective Studies
13.
Histopathology ; 46(4): 431-41, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15810955

ABSTRACT

AIMS: To evaluate the HER-2/neu status at the mRNA and DNA level of breast carcinomas and to compare it with HER-2/neu receptor overexpression by immunohistochemistry (IHC). METHODS AND RESULTS: In 32 invasive breast carcinomas, frozen tissue was available for real-time detection of HER-2/neu mRNA levels by reverse transcription-polymerase chain reaction (RT-PCR). Corresponding paraffin sections were examined by IHC and fluorescence in-situ hybridization (FISH). Thereby, different IHC and FISH procedures were compared. Using microwave epitope retrieval, all 32 cases scored 3+ on IHC, whereas only 28 out of 32 cases scored IHC 3+ using water bath epitope retrieval. All of these 28 cases showed increased levels of HER-2/neu mRNA. Dual-colour FISH analysis showed corresponding gene amplification in all 28 cases, with two cases showing a peculiar amplification pattern. In the remaining four cases, scoring IHC 2+ using water bath epitope retrieval, mRNA levels were not elevated. Three cases did not have gene amplification and one case showed low-level HER-2/neu gene amplification. All four carcinomas showed chromosome 17 polysomy. CONCLUSIONS: Real-time RT-PCR is accurate in selecting breast carcinoma cases scoring 3+ by IHC with high-level gene amplification. Results obtained by dual-colour FISH suggest that mechanisms leading to HER-2/neu receptor overexpression may be different between carcinomas scoring 2+ and 3+ on IHC, with polysomy 17 found in the former and gene amplification in the latter.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/genetics , Breast Neoplasms/pathology , Female , Humans , In Situ Hybridization, Fluorescence/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reagent Kits, Diagnostic/standards , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods
14.
Leukemia ; 19(1): 27-30, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15496975

ABSTRACT

We report the cloning of a novel PDGFRB fusion gene partner in a patient with a chronic myeloproliferative disorder characterized by t(5;14)(q33;q32), who responded to treatment with imatinib mesylate. Fluorescence in situ hybridization demonstrated that PDGFRB was involved in the translocation. Long distance inversion PCR identified KIAA1509 as the PDGFRB fusion partner. KIAA1509 is an uncharacterized gene with a predicted coiled-coil oligomerization domain with homology to the HOOK family of proteins. The predicted KIAA1509-PDGFRbeta fusion protein contains the KIAA1509 coiled-coil domain fused to the cytoplasmic domain of PDGFRbeta that includes the tyrosine kinase domain. Imatinib therapy resulted in rapid normalization of the patient's blood counts, and subsequent bone marrow biopsies and karyotypic analysis were consistent with sustained complete remission.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 5 , Myeloproliferative Disorders/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/genetics , Translocation, Genetic , Adult , Amino Acid Sequence , Base Sequence , Benzamides , Cloning, Molecular , DNA Primers , Humans , Imatinib Mesylate , In Situ Hybridization, Fluorescence , Male , Myeloproliferative Disorders/genetics
15.
Leukemia ; 19(1): 77-82, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15510210

ABSTRACT

Although reciprocal chromosomal translocations are not typical for B-cell chronic lymphocytic leukemia (B-CLL), we identified the novel t(1;6)(p35.3;p25.2) in eight patients with this disorder. Interestingly, all cases showed lack of somatically mutated IgV(H). Clinical, morphological, immunologic, and genetic features of these patients are described. Briefly, the age ranged from 33 to 81 years (median: 62.5 years) and the sex ratio was 6M:2F. Most of the patients (6/8) presented with advanced clinical stage. Therapy was required in seven cases. After a median follow-up of 28 months, five patients are alive and three died from disease evolution. Three cases developed transformation into diffuse large B-cell lymphoma. Translocation t(1;6) was found as the primary karyotypic abnormality in three patients. Additional chromosomal aberrations included changes frequently found in unmutated B-CLL, that is, del(11)(q), trisomy 12 and 17p aberrations. Fluorescence in situ hybridization analysis performed in seven cases allowed us to map the t(1;6) breakpoints to the 1p35.3 and 6p25.2 chromosomal bands, respectively. The latter breakpoint was located in the genomic region coding for MUM1/IRF4, one of the key regulators of lymphocyte development and proliferation, suggesting involvement of this gene in the t(1;6). Molecular characterization of the t(1;6)(p35.3;p25.2), exclusively found in unmutated subtype of B-CLL, is in progress.


Subject(s)
Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 6 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Translocation, Genetic , Humans , In Situ Hybridization, Fluorescence , Karyotyping
16.
Nat Genet ; 36(10): 1084-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15361874

ABSTRACT

In T-cell acute lymphoblastic leukemia (T-ALL), transcription factors are known to be deregulated by chromosomal translocations, but mutations in protein tyrosine kinases have only rarely been identified. Here we describe the extrachromosomal (episomal) amplification of ABL1 in 5 of 90 (5.6%) individuals with T-ALL, an aberration that is not detectable by conventional cytogenetics. Molecular analyses delineated the amplicon as a 500-kb region from chromosome band 9q34, containing the oncogenes ABL1 and NUP214 (refs. 5,6). We identified a previously undescribed mechanism for activation of tyrosine kinases in cancer: the formation of episomes resulting in a fusion between NUP214 and ABL1. We detected the NUP214-ABL1 transcript in five individuals with the ABL1 amplification, in 5 of 85 (5.8%) additional individuals with T-ALL and in 3 of 22 T-ALL cell lines. The constitutively phosphorylated tyrosine kinase NUP214-ABL1 is sensitive to the tyrosine kinase inhibitor imatinib. The recurrent cryptic NUP214-ABL1 rearrangement is associated with increased HOX expression and deletion of CDKN2A, consistent with a multistep pathogenesis of T-ALL. NUP214-ABL1 expression defines a new subgroup of individuals with T-ALL who could benefit from treatment with imatinib.


Subject(s)
Genes, abl , Leukemia-Lymphoma, Adult T-Cell/genetics , Nuclear Pore Complex Proteins/genetics , Plasmids/genetics , Amino Acid Sequence , Artificial Gene Fusion , Base Sequence , Benzamides , Cell Line, Tumor , Chromosomes, Human, Pair 9/genetics , DNA, Neoplasm/genetics , Enzyme Inhibitors/therapeutic use , Gene Amplification , Humans , Imatinib Mesylate , In Situ Hybridization, Fluorescence , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/enzymology , Molecular Sequence Data , Piperazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrimidines/therapeutic use
18.
Leukemia ; 18(10): 1705-10, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15306823

ABSTRACT

Classical t(11;14)(q13;q32) involving IGH-CCND1 is typically associated with aggressive CD5-positive mantle cell lymphoma (MCL). Recently, we identified the IGK variant of this translocation, t(2;11)(p11;q13), in three patients with a leukemic small-cell B-non-Hodgkin lymphoma. In all cases, rearrangements of the IGK and CCND1 genes were demonstrated by fluorescence in situ hybridization. Moreover, we mapped the 11q13 breakpoint of this variant translocation in the 3' region of CCND1 which contrasts with the 5' breakpoints in a standard t(11;14)(q13;q32). Expression of cyclin D1 was shown in two cases analyzed either at diagnosis or during disease progression. All three patients were asymptomatic at presentation and no initial therapy was required. One patient died of a progressive disease 58 months from diagnosis, and two patients showed stable disease after 12 months of follow-up. In two analyzed cases, mutated IGVH genes were identified. Our findings indicate that variant t(2;11)(p11;q13) does not typify a classical MCL but possibly a more indolent leukemic lymphoma originating from an antigen experienced (mutated) B cell.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 2/genetics , Cyclin D1/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Immunoglobulins/genetics , Lymphoma, Non-Hodgkin/genetics , Translocation, Genetic/genetics , Adult , DNA, Neoplasm/analysis , Disease Progression , Female , Genetic Variation , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia/genetics , Leukemia/immunology , Leukemia/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/pathology , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology
19.
Ann Hematol ; 83(9): 578-83, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15138714

ABSTRACT

A patient with mantle cell lymphoma (MCL) of the pleomorphic blastoid subtype is reported. The disease was clinically aggressive and refractory to chemotherapy, and the patient survived only 2 months. Cytogenetically, a t(11;19;14)(q13;q13;q32) was found. Fluorescent in situ hybridization (FISH) and molecular analyses demonstrated involvement of the BCL1/CCND1 locus in a three-way translocation. In addition, subclonal abnormalities of the region 8q24 manifested either as a t(8;22)(q24;q11)/CMYC rearrangement or trisomy 8 were identified. The pathogenetic impact of this very uncommon association of BCL1/CCND1 and CMYC rearrangements in MCL is discussed and the literature is reviewed.


Subject(s)
Cyclin D1/genetics , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Proto-Oncogene Proteins c-myc/genetics , Translocation, Genetic/genetics , Trisomy/genetics , Aged , Chromosomes, Human/genetics , DNA, Neoplasm/genetics , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Treatment Outcome
20.
Leukemia ; 18(4): 734-42, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14973504

ABSTRACT

Detection of the FIP1L1-PDGFRA fusion gene or the corresponding cryptic 4q12 deletion supports the diagnosis of chronic eosinophilic leukemia (CEL) in patients with chronic hypereosinophilia. We retrospectively characterized 17 patients fulfilling WHO criteria for idiopathic hypereosinophilic syndrome (IHES) or CEL, using nested RT-PCR and interphase fluorescence in situ hybridization (FISH). Eight had FIP1L1-PDGFRA (+) CEL, three had FIP1L1-PDGFRA (-) CEL and six had IHES. FIP1L1-PDGFRA (+) CEL responded poorly to steroids, hydroxyurea or interferon-alpha, and had a high probability of eosinophilic endomyocarditis (n=4) and disease-related death (n=4). In FIP1L1-PDGFRA (+) CEL, palpable splenomegaly was present in 5/8 cases, serum vitamin B(12) was always markedly increased, and marrow biopsies revealed a distinctively myeloproliferative aspect. Imatinib induced rapid complete hematological responses in 4/4 treated FIP1L1-PDGFRA (+) cases, including one female, and complete molecular remission in 2/3 evaluable cases. In the female patient, 1 log reduction of FIP1L1-PDGFRA copy number was reached as by real-time quantitative PCR (RQ-PCR). Thus, correlating IHES/CEL genotype with phenotype, FIP1L1-PDGFRA (+) CEL emerges as a homogeneous clinicobiological entity, where imatinib can induce molecular remission. While RT-PCR and interphase FISH are equally valid diagnostic tools, the role of marrow biopsy in diagnosis and of RQ-PCR in disease and therapy monitoring needs further evaluation.


Subject(s)
Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Adult , Benzamides , Chromosomes, Human, Pair 4 , Clone Cells/pathology , Female , Humans , Hypereosinophilic Syndrome/complications , Hypereosinophilic Syndrome/drug therapy , Imatinib Mesylate , In Situ Hybridization, Fluorescence , Male , Middle Aged , Oncogene Proteins, Fusion , Phenotype , Piperazines/therapeutic use , Pyrimidines/therapeutic use , RNA, Messenger/analysis , Receptor, Platelet-Derived Growth Factor alpha/analysis , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , mRNA Cleavage and Polyadenylation Factors/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...