Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Transplant ; 15(4): 1091-100, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25736912

RESUMEN

Tailoring treatment by patient strata based on the risk of disease progression and treatment toxicity might improve outcomes of patients with posttransplant lymphoproliferative disorder (PTLD). We analysed the cohort of 70 patients treated in the international, multicenter phase II PTLD-1 trial (NCT01458548) to identify such factors. Of the previously published scoring systems in PTLD, the international prognostic index (IPI), the PTLD prognostic index and the Ghobrial score were predictive for overall survival. None of the scoring systems had a considerable effect on the risk for disease progression. Age and ECOG performance status were the baseline variables with the highest prognostic impact in the different scoring systems. Baseline variables not included in the scoring systems that had an impact on overall survival and disease progression were the type of transplant and the response to rituximab at interim staging. Thoracic organ transplant recipients who did not respond to rituximab monotherapy were at particularly high risk for death from disease progression with subsequent CHOP-based chemotherapy. Patients in complete remission after four courses of rituximab and patients in partial remission with low-risk IPI had a low risk of disease progression. We speculate that chemotherapy might not be necessary in this patient cohort.


Asunto(s)
Antígenos CD20/inmunología , Linfocitos B/inmunología , Trastornos Linfoproliferativos/tratamiento farmacológico , Rituximab/uso terapéutico , Humanos , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/patología , Persona de Mediana Edad , Pronóstico
2.
N Engl J Med ; 367(6): 520-31, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22873532

RESUMEN

BACKGROUND: The long-term prognosis for older patients with mantle-cell lymphoma is poor. Chemoimmunotherapy results in low rates of complete remission, and most patients have a relapse. We investigated whether a fludarabine-containing induction regimen improved the complete-remission rate and whether maintenance therapy with rituximab prolonged remission. METHODS: We randomly assigned patients 60 years of age or older with mantle-cell lymphoma, stage II to IV, who were not eligible for high-dose therapy to six cycles of rituximab, fludarabine, and cyclophosphamide (R-FC) every 28 days or to eight cycles of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) every 21 days. Patients who had a response underwent a second randomization to maintenance therapy with rituximab or interferon alfa, each given until progression. RESULTS: Of the 560 patients enrolled, 532 were included in the intention-to-treat analysis for response, and 485 in the primary analysis for response. The median age was 70 years. Although complete-remission rates were similar with R-FC and R-CHOP (40% and 34%, respectively; P=0.10), progressive disease was more frequent with R-FC (14%, vs. 5% with R-CHOP). Overall survival was significantly shorter with R-FC than with R-CHOP (4-year survival rate, 47% vs. 62%; P=0.005), and more patients in the R-FC group died during the first remission (10% vs. 4%). Hematologic toxic effects occurred more frequently in the R-FC group than in the R-CHOP group, but the frequency of grade 3 or 4 infections was balanced (17% and 14%, respectively). In 274 of the 316 patients who were randomly assigned to maintenance therapy, rituximab reduced the risk of progression or death by 45% (in remission after 4 years, 58%, vs. 29% with interferon alfa; hazard ratio for progression or death, 0.55; 95% confidence interval, 0.36 to 0.87; P=0.01). Among patients who had a response to R-CHOP, maintenance therapy with rituximab significantly improved overall survival (4-year survival rate, 87%, vs. 63% with interferon alfa; P=0.005). CONCLUSIONS: R-CHOP induction followed by maintenance therapy with rituximab is effective for older patients with mantle-cell lymphoma. (Funded by the European Commission and others; ClinicalTrials.gov number, NCT00209209.).


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfoma de Células del Manto/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales de Origen Murino/efectos adversos , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Femenino , Humanos , Quimioterapia de Inducción , Análisis de Intención de Tratar , Linfoma de Células del Manto/mortalidad , Quimioterapia de Mantención , Masculino , Persona de Mediana Edad , Prednisona/efectos adversos , Prednisona/uso terapéutico , Estudios Prospectivos , Inducción de Remisión , Rituximab , Tasa de Supervivencia , Vidarabina/administración & dosificación , Vidarabina/análogos & derivados , Vincristina/efectos adversos , Vincristina/uso terapéutico
3.
Cancer Res ; 55(5): 984-8, 1995 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-7867008

RESUMEN

Deletions of chromosomal band 9p21 have been detected in various tumor types including melanoma, glioma, lung cancer, mesothelioma, and bladder cancer. Recently, the CDKN2 gene (p16INK4A, MTS I, CDK41) has been proposed as a candidate tumor suppressor gene because it is frequently deleted in cell lines derived from multiple tumor types. We performed fluorescence in situ hybridization (FISH) with interphase cells using yeast artificial chromosome clones and a cosmid contig of the CDKN2 region. In 10 cell lines (4 glioma, 2 melanoma, 2 non-small cell lung cancer, 2 bladder cancer) with 9p alterations detected by molecular or cytogenetic analysis, interphase FISH with the CDKN2 cosmid contig detected all 9p deletions previously identified by molecular analysis. Using this probe, FISH analysis of primary glioblastoma tumors revealed homozygous deletions of the CDKN2 region in 6 of 9 tumors (67%) whereas a yeast artificial chromosome probe containing the interferon type I (IFN) gene cluster was deleted in only 4 cases (44%). Thus, it is likely that the CDKN2 region is the target of 9p deletions in gliomas. Interphase FISH will play an important role in defining the clinical significance of 9p deletions in primary tumors because it is especially applicable to clinical samples which may be contaminated by normal cells.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 9 , Quinasas Ciclina-Dependientes , Eliminación de Gen , Genes Supresores de Tumor , Glioma/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas , Carcinoma de Pulmón de Células no Pequeñas/genética , Quinasa 4 Dependiente de la Ciclina , Humanos , Hibridación Fluorescente in Situ , Interfase/fisiología , Neoplasias Pulmonares/genética , Melanoma/genética , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética
4.
Cancer Res ; 57(20): 4608-14, 1997 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9377576

RESUMEN

Mantle cell lymphoma (MCL) has recently become generally accepted as a subentity of malignant lymphomas that is characterized by the chromosomal translocation t(11;14)(q13;q32), resulting in the overexpression of cyclin D1. Cyclin D1 forms a complex with cell cycle-dependent kinase (cdk) 4, which inactivates the retinoblastoma protein (pRB) via phosphorylation. However, in transgenic mice, the overexpression of cyclin D1 alone is not sufficient for the development of malignant lymphoma. To determine whether other members of the pRB pathway contribute to the malignant transformation of MCL, we analyzed 37 cases of MCL that were well characterized by morphology, immunophenotype, and/or interphase cytogenetics [detection of t(11;14)(q13;q32)]. Interphase fluorescence in situ hybridization was performed using a cosmid contig (250 kb) of the CDKN2/p16 region (encoding an inhibitor of the cyclin D1/cdk4 complex) and a phage contig (200 kb) of the Rb region. CDKN2/p16 deletion was detected in 15 cases (41%), including 6 homozygous deletions; Rb was deleted in 15 cases (41%), all of which were hemizygous deletions. Nine cases (24%) had deletions of both CDKN2/p16 and Rb. Further analysis of a subset of 17 MCLs revealed a highly significant correlation between CDKN2/p16 deletion and proliferation index, determined by the rate of Ki67 expression (P = 0.014; t test). No significant correlation was found between CDKN2/p16 deletion and the blastoid variant of MCL (P = 0.23; Fisher's test) or between proliferation index and blastoid morphology (P = 0.51; t test). Deletion of Rb did not have any impact on cell proliferation in addition to CDKN2/p16 deletion (P = 0.76; t test). Additional analysis of 13q14 deletions suggests that these deletions may target another gene telomeric to Rb. We conclude that deletion of CDKN2/p16 occurs in approximately one-half of MCLs and is a more relevant indicator of the proliferative features as compared to morphological criteria. In contrast, although deletions of chromosomal band 13q14 are frequent in MCL, inactivation of Rb seems not to be involved in the pathogenesis of MCL.


Asunto(s)
Aberraciones Cromosómicas , Ciclina D1/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Linfoma no Hodgkin/genética , Linfoma no Hodgkin/patología , Proteína de Retinoblastoma/biosíntesis , Animales , Ciclo Celular , Mapeo Cromosómico , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 14 , Ciclina D1/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Eliminación de Gen , Genes de Retinoblastoma , Genes p16 , Humanos , Inmunofenotipificación , Linfoma no Hodgkin/inmunología , Ratones , Ratones Transgénicos , Translocación Genética
5.
Leukemia ; 14(1): 93-9, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10637482

RESUMEN

The recurring translocation t(10;11)(p13;q14) which is found in acute myeloid leukemia (AML) and in acute lymphoblastic leukemia (ALL) results in the fusion of the putative transcription factor AF10 to CALM encoding a clathrin assembly protein. Previous studies using mainly fluorescence in situ hybridization (FISH) analysis have shown that the CALM/AF10 rearrangement is found in immature acute myeloid leukemia (AML) of subtype M0 and M1 and in T cell ALL. In this study we analyzed the CALM/AF10 and AF10/CALM fusion mRNAs in a series of three patients with AML, one patient with T-ALL and two patients with precusor T lymphoblastic lymphoma. In all six patients the breakpoint in CALM is at the 3' end of the coding region (nt1926/1927 or nt 2091/2092). Three breakpoints could be identified in AF10 (nt 588/589, nt 882/883 and nt 978/979). These data demonstrate that the CALM/AF10 fusions found in patients differ only slightly with respect to the portion of AF10 present and that there is no obvious difference between the fusions found in AML patients compared to those found in patients with lymphoid malignancies. Leukemia (2000) 14, 93-99.


Asunto(s)
Reordenamiento Génico , Leucemia Mieloide/genética , Linfoma no Hodgkin/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos Par 10 , Cromosomas Humanos Par 11 , Cartilla de ADN , ADN de Neoplasias , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Translocación Genética
6.
Clin Cancer Res ; 2(12): 1993-8, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9816158

RESUMEN

The tumor suppressor p16(INK4a) has been shown to be inactivated in numerous cancer lines and primary tumors. Recently, we reported loss of heterozygosity of the region in which p16(INK4a) is located in more than one-half of primary breast tumors. However, mutational analysis of these same tumors revealed mutation of p16(INK4a) to be infrequent. Other possible modes of inactivation, such as de novo methylation and homozygous deletion, have since been shown to occur in numerous neoplasias. Furthering the complexity of this locus, a transcript overlapping the p16(INK4a) coding sequence and encoding a novel peptide with growth-suppressive activity, p19(ARF), has been described. To clearly elucidate the target of aberrations affecting this subchromosomal region and approximate frequency in breast cancer, we performed a comprehensive study including p16 deletion analysis by means of interphase chromosomal fluorescence in situ hybridization, methylation analysis of the first exon encoding p16(INK4a) (exon 1alpha), mutational analysis of exon 1beta by single-strand conformational polymorphism analysis of p19(ARF) transcripts, and expression of both alpha and beta transcripts by reverse transcription PCR. Homozygous deletion of p16, as determined by interphase chromosomal fluorescence in situ hybridization, was observed in 3 of 18 (17%) tumors analyzed, whereas de novo methylation of exon 1alpha was observed in an additional 17% (4 of 23). Reduced expression of p16(INK4a) was observed in 11 tumors (48%), including all those in which homozygous deletion or complete methylation was observed. No mutations of exon 1 beta were detected, and expression of its transcript was variable, with 13% demonstrating decreased expression and 17% demonstrating overexpression. These results further support p16(INK4a) as a target of inactivation in the 9p21 region for breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Proteínas/genética , Metilación de ADN , Análisis Mutacional de ADN , Exones/genética , Femenino , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ , Pérdida de Heterocigocidad , Polimorfismo Conformacional Retorcido-Simple , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p14ARF Supresora de Tumor
7.
Cancer Genet Cytogenet ; 83(1): 46-55, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7656204

RESUMEN

Hemizygous and homozygous deletions of the type I interferon gene cluster (IFN) have been detected in about 20% of acute lymphoblastic leukemias. A putative tumor suppressor gene (TSG) is thought to be located centromeric to the IFN cluster on chromosomal bands 9p21-22. We studied the accuracy of fluorescence in situ hybridization (FISH) for detecting deletions in interphase cells using yeast artificial chromosome (YAC) clones containing all or part of the IFN cluster. FISH probes were generated from YACs (320-1300 kb in size) by a sequence-independent amplification technique (SIA). Fifteen cell lines (nine T-ALL, three B-cell precursor ALL, one B-ALL, one AML, one CML-BC) that had been well characterized by conventional cytogenetic analysis and molecular techniques were analyzed. We were able to detect all numerical changes of the IFN cluster including homozygous and hemizygous deletions accurately and to define subclones of the cell lines. Moreover, in six cell lines we were able to identify subclones. In dilution experiments the detection thresholds for subpopulations with homozygous and hemizygous deletions were determined to be 5% and 7.5%, respectively.


Asunto(s)
Deleción Cromosómica , Leucemia/genética , Secuencia de Bases , Cromosomas Artificiales de Levadura , Sondas de ADN , Humanos , Hibridación Fluorescente in Situ , Metafase , Datos de Secuencia Molecular , Ploidias , Sensibilidad y Especificidad , Células Tumorales Cultivadas
8.
Cytogenet Cell Genet ; 76(3-4): 202-5, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9186524

RESUMEN

We have developed a method to generate FISH probes from small (2-4 kb) nonrepetitive genomic restriction fragments. The probes showed a high hybridization efficiency of up to 90% in metaphase cells from normal peripheral blood. With the use of these probes, homozygous as well as hemizygous 9p deletions were reliably identified in nine leukemia-derived cell lines.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 9 , Sondas de ADN , Hibridación Fluorescente in Situ , Línea Celular , Células HL-60 , Humanos , Mutagénesis Insercional
9.
Blood ; 86(5): 1931-8, 1995 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-7544647

RESUMEN

Deletions of chromosomal band 9p21 have been detected in various tumor types as well as in more than 20% of acute lymphoblastic leukemia (ALL). These deletions frequently include the entire interferon (IFN) gene cluster as well as the methylthioadenosine phosphorylase (MTAP) gene. Recently, the CDKN2 gene (p16INK4A, MTS I, CDK41) was proposed as a candidate tumor-suppressor gene on 9p21 because it is frequently deleted in cell lines derived from multiple tumor types. To determine if CDKN2 or another closely related gene on 9p is the target of 9p deletions in ALL and other hematologic malignancies, we analyzed 20 primary patient samples (13 ALL, 2 acute myeloid leukemias [AML], and 5 non-Hodgkin's lymphomas [NHL]) with 9p rearrangements using Southern blot analysis, fluorescence in situ hybridization (FISH), and single-strand conformation polymorphism (SSCP) for alterations of CDKN2. Homozygous deletions of the CDKN2/CDKN2B (p15) region were detected in 10 cases (50%; 6 ALL, 2 AML, and 2 NHL). In 1 additional case, the intensity of the Southern blot band was significantly reduced, suggesting a CDKN2 deletion in a subpopulation of the malignant cells. No CDKN2 or CDKN2B rearrangements were seen. The IFN gene cluster was homozygously deleted in 2 of 15 (13%) analyzed cases, whereas the MTAP gene was deleted in 6 of 15 cases (40%). In addition, hemizygous deletions of the CDKN2 region were identified in 6 ALL cases using interphase FISH. No point mutation of the coding region of CDKN2 was detected by SSCP in these cases. We conclude that CDKN2 is the most frequently homozygously deleted marker on 9p. The absence of point mutations in the coding region of CDKN2 in cases with hemizygous 9p deletions and the frequent codeletion of MTAP, CDKN2B, and other yet unidentified neighboring genes suggest that the simultaneous deletion of these genes may be necessary for the selective growth advantage of malignant cells.


Asunto(s)
Proteínas Portadoras/genética , Aberraciones Cromosómicas , Deleción Cromosómica , Trastornos de los Cromosomas , Cromosomas Humanos Par 9 , Leucemia Mieloide Aguda/genética , Linfoma no Hodgkin/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Adolescente , Adulto , Anciano , Southern Blotting , Médula Ósea/patología , Células de la Médula Ósea , Proteínas Portadoras/sangre , Células Cultivadas , Preescolar , Mapeo Cromosómico , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Femenino , Eliminación de Gen , Reordenamiento Génico , Humanos , Hibridación Fluorescente in Situ , Interferones/genética , Cariotipificación , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/patología , Linfoma no Hodgkin/sangre , Linfoma no Hodgkin/patología , Masculino , Persona de Mediana Edad , Monocitos/patología , Familia de Multigenes , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Purina-Nucleósido Fosforilasa/genética
10.
Genes Chromosomes Cancer ; 22(1): 72-8, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9591637

RESUMEN

Identifying the various genetic alterations that contribute to lymphomagenesis is key to our improved understanding of the biological behavior of the disease. Recently, we and others have defined a tumor suppressor region on the short arm of chromosome 9 harboring a cluster of genes, including MTAP, CDKN2A (p16INK4a), and CDKN2B (p15INK4B), which is frequently deleted in a variety of tumor types. To determine whether this region is involved in a particular subset of malignant lymphomas, we have examined 16 cases of diffuse large-cell lymphoma (DLCL) (including three cases that evolved from low-grade non-Hodgkin lymphoma (NHL) (transformed DLCL)), and nine cases of low-grade NHL that had subpopulations of large cells with a diffuse growth pattern (seven follicular NHL, one chronic lymphocytic leukemia, one mycosis fungoides). Interphase fluorescence in situ hybridization was performed on these samples using a 250-kb cosmid contig (COSp16), which encompasses MTAP, CDKN2A, and CDKN2B. Six of the 16 DLCLs and one of nine low-grade NHLs had deletions of COSp16. COSp16 was homozygously deleted in four cases; two cases had hemizygous deletions, and one case had a partial homozygous deletion of the cosmid contig. Three of 13 cases of de novo DLCL, all three transformed DLCLs, and one of nine low-grade NHL had COSp16 deletions. Although the numbers are small, COSp16 deletion was associated with transformed DLCL in contrast to de novo DLCL (P < 0.04, Fisher's exact test) or low-grade NHL (P < 0.02). The COSp16 deletion was mostly submicroscopic and was not observed in association with any specific recurring cytogenetic abnormalities. These results suggest that targeted deletion of the CDKN2A region occurs in a subset of non-Hodgkin's lymphomas, and may be associated with transformed lymphomas.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Eliminación de Gen , Linfoma de Células B Grandes Difuso/genética , Linfoma no Hodgkin/genética , Purina-Nucleósido Fosforilasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Transformación Celular Neoplásica/patología , Femenino , Humanos , Hibridación Fluorescente in Situ , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/patología , Linfoma no Hodgkin/enzimología , Linfoma no Hodgkin/patología , Masculino , Persona de Mediana Edad
11.
Genomics ; 24(2): 211-7, 1994 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-7535286

RESUMEN

Deletions of the short arm of chromosome 9 have been observed in a number of malignant cell lines and primary tumor samples using cytogenetic and molecular techniques. These tumors include acute lymphoblastic leukemias, lymphomas, gliomas, melanomas, mesotheliomas, bladder cancer, and lung cancer. The smallest region of overlap (SRO) of these deletions is thought to contain a tumor suppressor gene. A microdissection library was constructed from bands 9p21-p23 to obtain DNA probes that would be useful in further defining the limits of the deletions. Eight single-copy probes were found to be homozygously deleted in at least 1 of the 10 cell lines examined. The mapping of these 8 clones using a panel of cell lines with deletions revealed that 3 probes mapped telomeric to the SRO and 5 clones mapped centromeric to the SRO.


Asunto(s)
Bandeo Cromosómico , Cromosomas Humanos Par 9 , Genes Supresores de Tumor , Secuencia de Bases , Mapeo Cromosómico , Cartilla de ADN , Biblioteca Genómica , Humanos , Interferones/genética , Datos de Secuencia Molecular , Neoplasias/genética , Eliminación de Secuencia , Células Tumorales Cultivadas
12.
Proc Natl Acad Sci U S A ; 93(10): 4804-9, 1996 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-8643484

RESUMEN

The translocation t(10;11)(p13;q14) is a recurring chromosomal abnormality that has been observed in patients with acute lymphoblastic leukemia as well as acute myeloid leukemia. We have recently reported that the monocytic cell line U937 has a t(10;11)(p13;q14) translocation. Using a combination of positional cloning and candidate gene approach, we cloned the breakpoint and were able to show that AF10 is fused to a novel gene that we named CALM (Clathrin Assembly Lymphoid Myeloid leukemia gene) located at 11q14. AF10, a putative transcription factor, had recently been cloned as one of the fusion partners of MLL. CALM has a very high homology in its N-terminal third to the murine ap-3 gene which is one of the clathrin assembly proteins. The N-terminal region of ap-3 has been shown to bind to clathrin and to have a high-affinity binding site for phosphoinositols. The identification of the CALM/AF10 fusion gene in the widely used U937 cell line will contribute to our understanding of the malignant phenotype of this line.


Asunto(s)
Cromosomas Humanos Par 10/genética , Cromosomas Humanos Par 11/genética , Proteínas de Ensamble de Clatrina Monoméricas , Translocación Genética , Proteínas Adaptadoras del Transporte Vesicular , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , Cartilla de ADN/genética , ADN Complementario/genética , Humanos , Hibridación Fluorescente in Situ , Leucemia Mieloide Aguda/genética , Ratones , Datos de Secuencia Molecular , Monocitos/ultraestructura , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/genética , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética , Células Tumorales Cultivadas
13.
Br J Haematol ; 91(4): 865-70, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8547131

RESUMEN

Homozygous deletions of the cyclin-dependent kinase 4 (CDK4) inhibitor gene CDKN2 (p16, MTS1) have been demonstrated to occur frequently in human cancer cell lines of different origin. However, in most primary tumours the frequencies of CDKN2 deletions are not well defined. We studied primary samples of 100 patients with lymphoid leukaemias [B-lineage acute lymphoblastic leukaemia (ALL), n = 23; T-ALL, n = 7; B-cell chronic lymphocytic (B-CLL) or prolymphocytic (B-PLL) leukaemia, n = 50; T-CLL/T-PLL, n = 20] using fluorescence in situ hybridization (FISH) with eight overlapping cosmid clones covering the region on chromosome band 9p21 containing CDKN2. We did not observe any CDKN2 deletions in the 70 patients with chronic lymphoid leukaemias of B- or T-cell origin. Of the 23 patients with B-lineage ALL, one (4%) exhibited a CDKN2 deletion: in this patient, two clones were detected, one exhibiting a hemizygous and the other a homozygous deletion. On chromosome banding analysis, four patients with B-lineage ALL had a 9p aberration, whereas all CDKN2 copies were retained. In contrast, six of the seven (86%) patients with T-ALL exhibited CDKN2 deletions (homozygous, n = 4; hemizygous, n = 2). We conclude that hemizygous or homozygous deletions of the CDKN2 gene occur at high frequency in T-ALL and at low frequency in B-lineage ALL, supporting the role of this gene as a tumour suppressor, especially in T-ALL. However, from our data there is no evidence that CDKN2 is involved in the pathogenesis of chronic lymphoid leukaemias of B- or T-cell origin.


Asunto(s)
Proteínas Portadoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Inhibidores de Proteínas Quinasas , Adolescente , Adulto , Anciano , Niño , Preescolar , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Lactante , Leucemia de Células B/genética , Leucemia de Células T/genética , Persona de Mediana Edad
14.
Genes Chromosomes Cancer ; 26(3): 210-4, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10502318

RESUMEN

Structural aberrations of chromosomal band 13q14 are frequent in B-cell chronic lymphocytic leukemia (B-CLL) and target a putative tumor suppressor gene in the genomic region between the RB1 gene and the genetic marker D13S25. Recently, it has been suggested that alterations of this particular region might also be of relevance for the pathogenesis of mantle cell lymphomas (MCL). We applied dual-color fluorescence in situ hybridization (FISH) using probes for the RB1 and/or D13S25 loci and screened a total of 236 B- and T-cell non-Hodgkin's lymphomas (NHL) for deletions occurring in this genomic region. In MCL, the high rate (12/32; 38%) of hemizygous deletions and especially a deletion pattern similar to B-CLL in four of the cases provide further evidence that a substantial proportion of MCL cases may share a common way of pathogenesis with B-CLL. In other B-cell NHL, the frequency of allelic loss affecting 13q14 was overall low. However, the finding of 13q14 microdeletions in seven cases without detectable alterations of chromosome 13 at G-banding analysis might indicate a possible involvement of this genetic region also for the lymphomagenesis of single cases of B-cell NHL other than B-CLL and MCL. In T-cell NHL, allelic loss at 13q14 was encountered in three of 13 peripheral T-NHL, NOS. Taking into account the very limited cytogenetic data yet available in this entity, our series provides further evidence that 13q14 changes might represent one of the most frequent genetic abnormalities in T-cell NHL.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 13 , Linfoma de Células del Manto/genética , Linfoma de Células T Periférico/genética , Bandeo Cromosómico , Genes de Retinoblastoma/genética , Humanos , Hibridación Fluorescente in Situ
15.
Haematologica ; 86(2): 181-6, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11224488

RESUMEN

BACKGROUND AND OBJECTIVES: Microsatellite instability (MSI) is characterized by tumor-associated alterations in the germline size of microsatellite repeats caused by a reduced efficacy of the DNA mismatch repair machinery. The aim of this study was to investigate the presence of MSI in a number of cases of indolent and aggressive mantle cell lymphoma (MCL) and B-cell chronic lymphocytic leukemia (B-CLL) to determine its possible role in the initial development and progression of these disorders. DESIGN AND METHODS: We examined the presence of MSI in 28 B-CLL, 24 typical and 4 transformed B-CLL (Richter's syndrome) and 29 MCL, 19 typical and 10 blastoid variants by using a panel of 10 microsatellite markers and analyzed them using an AbiPrism 310 DNA sequencer. Fisher's exact test was used to compare categorical variables and Mann-Whitney's U-test for continuous variables. RESULTS: MSI alterations were not observed in any case of MCL or Richter's syndrome and in only three (13%) patients with typical B-CLL. Two of these patients also had loss of heterozygosity in one of the 10 sites examined. These patients presented with a more advanced stage, diffuse bone marrow involvement, and poorer performance status than patients without these alterations. INTERPRETATION AND CONCLUSIONS: These findings indicate that MSI is not involved in the pathogenesis or progression of B-CLL and MCL but may appear in a small subset of patients with advanced B-CLL.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/genética , Linfoma de Células del Manto/genética , Repeticiones de Microsatélite , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , ADN de Neoplasias/genética , Progresión de la Enfermedad , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/etiología , Linfoma de Células del Manto/etiología , Masculino , Persona de Mediana Edad
16.
Proc Natl Acad Sci U S A ; 92(14): 6489-93, 1995 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-7604019

RESUMEN

Many human malignant cells lack methylthioadenosine phosphorylase (MTAP) enzyme activity. The gene (MTAP) encoding this enzyme was previously mapped to the short arm of chromosome 9, band p21-22, a region that is frequently deleted in multiple tumor types. To clone candidate tumor suppressor genes from the deleted region on 9p21-22, we have constructed a long-range physical map of 2.8 megabases for 9p21 by using overlapping yeast artificial chromosome and cosmid clones. This map includes the type IIFN gene cluster, the recently identified candidate tumor suppressor genes CDKN2 (p16INK4A) and CDKN2B (p15INK4B), and several CpG islands. In addition, we have identified other transcription units within the yeast artificial chromosome contig. Sequence analysis of a 2.5-kb cDNA clone isolated from a CpG island that maps between the IFN genes and CDKN2 reveals a predicted open reading frame of 283 amino acids followed by 1302 nucleotides of 3' untranslated sequence. This gene is evolutionarily conserved and shows significant amino acid homologies to mouse and human purine nucleoside phosphorylases and to a hypothetical 25.8-kDa protein in the pet gene (coding for cytochrome bc1 complex) region of Rhodospirillum rubrum. The location, expression pattern, and nucleotide sequence of this gene suggest that it codes for the MTAP enzyme.


Asunto(s)
Deleción Cromosómica , Cromosomas Artificiales de Levadura , Cromosomas Humanos Par 9 , Clonación Molecular/métodos , Genes Supresores de Tumor , Neoplasias/genética , Purina-Nucleósido Fosforilasa/biosíntesis , Purina-Nucleósido Fosforilasa/genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Mapeo Cromosómico , Humanos , Datos de Secuencia Molecular , Mapeo Restrictivo , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
17.
Blood ; 91(12): 4662-7, 1998 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9616163

RESUMEN

The translocation t(10;11)(p13;q14) has been observed in acute lymphoblastic leukemia (ALL) as well as acute myeloid leukemia (AML). A recent study showed a MLL/AF10 fusion in all cases of AML with t(10;11) and various breakpoints on chromosome 11 ranging from q13 to q23. We recently cloned CALM (Clathrin Assembly Lymphoid Myeloid leukemia gene), the fusion partner of AF10 at 11q14 in the monocytic cell line U937. To further define the role of these genes in acute leukemias, 10 cases (9 AML and 1 ALL) with cytogenetically proven t(10;11)(p12-14;q13-21) and well-characterized morphology, immunophenotype, and clinical course were analyzed. Interphase fluorescence in situ hybridization (FISH) was performed with 2 YACs flanking the CALM region, a YAC contig of the MLL region, and a YAC spanning the AF10 breakpoint. Rearrangement of at least one of these genes was detected in all cases with balanced t(10;11). In 4 cases, including 3 AML with immature morphology (1 AML-M0 and 2 AML-M1) and 1 ALL, the signals of the CALM YACS were separated in interphase cells, indicating a translocation breakpoint within the CALM region. MLL was rearranged in 3 AML with myelomonocytic differentiation (2 AML-M2 and 1 AML-M5), including 1 secondary AML. In all 3 cases, a characteristic immunophenotype was identified (CD4+, CD13-, CD33+, CD65s+). AF-10 was involved in 5 of 6 evaluable cases, including 1 case without detectable CALM or MLL rearrangement. In 2 complex translocations, none of the three genes was rearranged. All cases had a remarkably poor prognosis, with a mean survival of 9.6 +/- 6.6 months. For the 7 AML cases that were uniformly treated according to the AMLCG86/92 protocols, disease-free and overall survival was significantly worse than for the overall study group (P = .03 and P = .01, respectively). We conclude that the t(10;11)(p13;q14) indicates CALM and MLL rearrangements in morphologically distinct subsets of acute leukemia and may be associated with a poor prognosis.


Asunto(s)
Cromosomas Humanos Par 10 , Cromosomas Humanos Par 11 , Clatrina/genética , Proteínas de Unión al ADN/genética , Leucemia/genética , Proto-Oncogenes , Factores de Transcripción/genética , Translocación Genética , Enfermedad Aguda , Adulto , Anciano , Preescolar , Femenino , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia/patología , Leucemia/fisiopatología , Subgrupos Linfocitarios/patología , Masculino , Persona de Mediana Edad , Proteína de la Leucemia Mieloide-Linfoide , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA