Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Klin Padiatr ; 226(6-7): 338-43, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25431866

RESUMEN

BACKGROUND: Recently, the UK CCLG and COG reported that an intrachromosomal amplification of chromosome 21 (iAMP21) in acute lymphoblastic leukemia (ALL) loses its adverse prognostic impact with intensified therapy. PATIENT AND METHODS: We evaluated the prognosis of iAMP21 among patients from the ALL-BFM (Berlin-Frankfurt-Münster) 2000 trial with 46 of 2 637 (2%) patients iAMP21+. RESULTS: 8-year event-free-survival (EFS, 64 ± 8% vs. 81 ± 1%, p=0.0026) and cumulative incidence of relapse (CIR, 29 ± 8% vs. 14 ± 1%, p=0.008) of the iAMP21 cases were significantly worse compared with non-iAMP21 patients. Within the MRD low-risk group, iAMP21 cases (n=14) had an inferior 8-year EFS (76 ± 12% vs. 92 ± 1%, p=0.0081), but no increased CIR (10 ± 10% vs. 6 ± 1%, p=0.624). Within the MRD intermediate-risk group, iAMP21 cases (n=27) had a worse 8-year EFS (56 ± 11% vs. 78 ± 2%, p=0.0077) and CIR (44 ± 11% vs. 20 ± 2%, p=0.003) with 6/10 relapses occurring after 2 years. CONCLUSIONS: Conclusively, we believe that there is no necessity for enrolling all iAMP21 patients into the high-risk arm of ongoing ALL-BFM trials because MRD low-risk patients have a moderate relapse risk under current therapy. Whether the increased relapse risk in MRD intermediate-risk patients can be avoided by late treatment intensification remains to be answered by the AIEOP-BFM ALL 2009 trial randomly using protracted pegylated L-asparaginase during delayed intensification and early maintenance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cromosomas Humanos Par 21/genética , Amplificación de Genes/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Adolescente , Niño , Preescolar , Estudios de Cohortes , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Neoplasia Residual/tratamiento farmacológico , Neoplasia Residual/genética , Neoplasia Residual/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidad , Proteínas Proto-Oncogénicas c-ets/genética , Recurrencia , Proteínas Represoras/genética , Proteína ETS de Variante de Translocación 6
2.
Klin Padiatr ; 225 Suppl 1: S30-3, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23700053

RESUMEN

Recurrent non-random chromosome abnormalities, including numerical or structural changes such as translocations, inversions, insertions or deletions within the leukemia cell nucleus, have been discovered in approximately 80% of patients with a malignant hematological disease. These reciprocal translocations correlate with specific cellular subtypes of hematopoeisis at the stage of their maturation arrest and are therefore important for diagnosis. Some of these aberrations are independent prognostic indicators and help to stratify patients into different risk-adapted therapy groups. Owing to new laboratory methods such as the fluorescence in situ hybridization (FISH) and modified polymerase chain reaction (RT-PCR) the chromosomal breakpoints can be investigated and the rearrangements of genes which produce the abnormal proteins can be identified. Due to the high sensitivity of these available data a new prognostic factor, the "minimal residual disease" (MRD), can be investigated at diagnosis and at intervals during the treatment period. Since we now know which oncoproteins are involved, a target-directed therapy with inhibitors might be possible in the future.Standard cytogenetic and molecular genetic analysis of the leukemia karyotype is of the utmost importance for classification (WHO), therapy and therapy results in the acute childhood leukemias.


Asunto(s)
Aberraciones Cromosómicas , Leucemia/genética , Translocación Genética/genética , Antineoplásicos/uso terapéutico , Niño , Rotura Cromosómica , Sistemas de Liberación de Medicamentos , Reordenamiento Génico/genética , Hematopoyesis/genética , Humanos , Hibridación Fluorescente in Situ , Leucemia/clasificación , Leucemia/diagnóstico , Leucemia/tratamiento farmacológico , Neoplasia Residual/diagnóstico , Neoplasia Residual/genética , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Klin Padiatr ; 225 Suppl 1: S62-72, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23700060

RESUMEN

Between 1981 and 2000, 6 609 children (<18 years of age) were treated in 5 consecutive trials of the Berlin-Frankfurt-Münster (BFM) study group for childhood acute lymphoblastic leukemia (ALL). Patients were treated in up to 82 centers in Germany, Austria, and Switzerland. Probability of 10-year event-free survival (survival) improved from 65% (77%) in study ALL-BFM 81-78% (85%) in ALL-BFM 95. In parallel to relapse reduction, major efforts focused on reducing acute and late toxicity through advanced risk adaptation of treatment. The major findings derived from these ALL-BFM trials were as follows: 1) preventive cranial radiotherapy could be safely reduced to 12 Gy in T-ALL and high-risk ALL patients and eliminated in non-high-risk non-T-ALL patients, if it was replaced by high-dose and intrathecal methotrexate; 2) omission of delayed reintensification severely impaired outcome of low-risk patients; 3) 6 months less maintenance therapy caused an increase in systemic relapses; 4) slow response to an initial 7-day prednisone window was identified as adverse prognostic factor; 5) condensed induction therapy resulted in a significant improvement of outcome; 6) the daunorubicin dose in induction could be safely reduced in low-risk patients; 7) intensification of consolidation/reintensification treatment led to considerable improvement of outcome in high-risk patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/historia , Oncología Médica/historia , Pediatría/historia , Leucemia-Linfoma Linfoblástico de Células Precursoras/historia , Ensayos Clínicos Controlados Aleatorios como Asunto/historia , Asparaginasa/historia , Niño , Ciclofosfamida/historia , Citarabina/historia , Daunorrubicina/historia , Europa (Continente) , Alemania , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Mercaptopurina/historia , Metotrexato/historia , Prednisona/historia , Vincristina/historia
4.
Leukemia ; 21(8): 1739-51, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17541401

RESUMEN

The chromosomal translocation t(8;14) is the hallmark of Burkitt's-lymphoma (BL) and fuses the proto-oncogene c-MYC to the IGH locus. We analyzed the genomic structure of MYC/IGH fusions derived from a large series of 78 patients with t(8;14) and asked (i) whether distinct breakpoint clusters exist within the MYC gene and (ii) whether any pairwise association between particular IGH and MYC breakpoints exist. Identification of such associations will help elucidate the etiology of the breaks on the MYC locus. Scan statistic analyses revealed two distinct, but large clusters within c-MYC containing 60/78 (77%) of the breakpoints. Clusters 1 and 2 were 560 and 779 bp in length within a 4555 bp breakpoint cluster region. Breaks within IGH switch mu and joining region did not differ with respect to their corresponding MYC breakpoints. However, there was a highly significant correlation between breakpoints 5' of MYC cluster 1 and fusions to IGH switch gamma region and breakpoints downstream of MYC cluster 2 and fusions to IGH switch alpha region (chi(2)-test: P<0.005). Chromatin changes governing choice of IGH-Fc region recombination may parallel changes in the MYC gene 5' region chromatin leading to some degree of coordinated ontological specificity in breakpoint location.


Asunto(s)
Linfoma de Burkitt/genética , Rotura Cromosómica , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 8/genética , Genes myc , Cadenas Pesadas de Inmunoglobulina/genética , Translocación Genética/genética , Adolescente , Niño , Preescolar , ADN de Neoplasias/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Masculino , Datos de Secuencia Molecular , Proto-Oncogenes Mas , Secuencias Repetitivas de Ácidos Nucleicos , Células Tumorales Cultivadas
5.
Leukemia ; 20(5): 820-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16541142

RESUMEN

The response to initial glucocorticoid therapy in childhood acute lymphoblastic leukaemia (ALL) reliably predicts the response to multiagent chemotherapy. Patients resistant to glucocorticoids (prednisone poor responders (PPR)) have a poorer event-free survival compared to glucocorticoid-sensitive patients (prednisone good responders (PGR)). A case-control study was performed to investigate differential protein expression in leukaemic blasts from PGR and PPR childhood ALL patients. Two-dimensional gel electrophoresis (2-DE) was used for an unsupervised screening and surface enhanced laser desorption/ionisation-time of flight mass spectrometry (SELDI-TOF MS) for the characterisation of protein spots. In difference maps of average gels for the proteomes of each responder group, differentially expressed proteins were identified after tryptic digestion and spotting onto H4-SELDI-TOF-MS chips. Proteins overexpressed in PPR were Catalase, RING finger protein 22 alpha, Valosin-containing protein (VCP) and a G-protein-coupled receptor. Proteins overexpressed in PGR were protein kinase C and malate dehydrogenase. Valosin-containing protein was chosen for validation and quantification by Western blot analysis in a second case-control group of ALL patients. In this second independent cohort, median VCP expression (P25-P75) was 0.15 (0.11-0.28) in PGR and 0.34 (0.14-0.99) in PPR patients (P = 0.04). We conclude that high VCP expression is associated with poor prednisone response in childhood ALL patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Ciclo Celular/análisis , Resistencia a Antineoplásicos , Glucocorticoides/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Prednisona/uso terapéutico , Proteoma/análisis , Adenosina Trifosfatasas , Western Blotting , Estudios de Casos y Controles , Catalasa/biosíntesis , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/efectos de los fármacos , Niño , Preescolar , Electroforesis en Gel Bidimensional , Femenino , Glucocorticoides/uso terapéutico , Humanos , Lactante , Recién Nacido , Malato Deshidrogenasa/biosíntesis , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Prednisona/farmacología , Proteína Quinasa C/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Sensibilidad y Especificidad , Proteína que Contiene Valosina
6.
Leukemia ; 20(8): 1422-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16738692

RESUMEN

Deletions of chromosome 6q have been reported in several hematological malignancies, but data are not conclusive regarding their biological and prognostic impact. Therefore, we focused on pediatric patients diagnosed with T-cell lymphoblastic lymphoma (T-LBL) treated uniformly according to the NHL-BFM95 protocol. We used loss-of-heterozygosity (LOH) analysis of 25 microsatellite markers located on chromosome 6q14-q24. Fragment-length analysis was performed on ABI-PRISM3100 Genetic-Analyzer. Eligibility criterion was > or =3 informative markers. Between April 1995 and March 2003, 185 T-LBL patients were treated according to the NHL-BFM95 protocol. Five-year event-free (EFS) and disease-free survival (DFS) were 79+/-3 and 87+/-3% (median follow-up 4.7 [1.2-10.1] years). Sixty-one patients were evaluable for LOH analysis, including 18 out of 23 patients with relapse. EFS and DFS were 67+/-6 and 69+/-6% for these 61 patients. Testing of 853 markers in the 61 patients identified the presence of LOH in 19 patients (31%): 13 of the 18 relapse patients and five of the 41 in complete remission (odds ratio 18.7, 95% confidence interval 4.7-75.3). One LOH-positive patient died from treatment-related toxicity. We conclude that LOH on chromosome 6q14-q24 may have conferred a high risk of relapse on our group of children with T-LBL treated according to the NHL-BFM95 protocol.


Asunto(s)
Cromosomas Humanos Par 6 , Leucemia-Linfoma de Células T del Adulto/genética , Pérdida de Heterocigocidad , Adolescente , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia-Linfoma de Células T del Adulto/mortalidad , Masculino
7.
Leukemia ; 19(4): 557-63, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15744345

RESUMEN

This study characterized the additional chromosomal abnormalities (ACA) associated with 11q23 rearrangements in 450 infants and children with acute lymphoblastic leukemia (ALL) and examined the impact of these ACA on survival. Overall, 213 (47%) cases had ACA but the incidence varied according to patient age and 11q23 subgroup. Infants and patients with t(4;11)(q21;q23) had the lowest incidence of ACA (50/182 (27%) and 57/216 (26%) respectively), whereas patients with del(11)(q23) had the highest incidence (66/93 (71%)). Del(11)(q23) abnormalities were heterogeneous and occasionally secondary to t(9;22)(q34;q11.2). Thus, patients with del(11)(q23) comprised a separate biological entity, which was clearly distinct from those with an 11q23 translocation. The most frequent specific ACA were trisomy X (n = 38), abnormal 12p (n = 32), abnormal 9p (n = 28) and del(6q) (n = 19). The presence of ACA did not change the 5 year event-free survival estimates among children (56% (95% Cl 46-65%) vs 62% (54-69%)) or infants (22% (15-29%) vs 18% (9-29%)), nor when the different 11q23 subgroups were analyzed separately. This study has conclusively demonstrated that there is no prognostic effect of secondary chromosomal changes in association with 11q23 abnormalities in childhood ALL. However, characterization of these ACA is important to determine their potential role in initiation of MLL driven leukemogenesis.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 11 , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Niño , Preescolar , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Incidencia , Lactante , Masculino , Pronóstico
8.
Oncogene ; 3(6): 691-5, 1988 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2577871

RESUMEN

A breakpoint cluster region (T-ALLbcr) has been previously described on 11p13 for T-ALL carrying t(11;14)(p13;q11). One further T-ALL breakpoint is described bringing to 5 out of 6 such translocations which are found to break within a maximum of 6.7 kb on chromosome 11p13. Studies of somatic cell hybrids derived from t(11;14)(p13;q11) T-ALL placed the T-ALLbcr between the genes for catalase (CAT) and the beta-subunit of follicle stimulating hormone (FSHB). This suggested a link between the T-ALLbcr and the Wilms' tumour predisposition locus (WT) since constitutional 11p13 deletions predispose to Wilms' tumour. Utilising somatic cell hybrids from patients with Wilms' tumours and aniridia, we show that while the T-ALLbcr maps distal to the catalase gene at 11p13, it maps outside the shortest region of overlap of a series of 11p13 deletions associated with Wilms'-Aniridia. The data suggest the order of genes at 11p13 to be: centromere-CAT-T-ALLbcr-WT-aniridia-FSHB-telomere. Therefore, the T-ALLbcr must lie very close to but may be distinct from the Wilms' predisposition locus at 11p13.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 14 , Neoplasias Renales/genética , Leucemia-Linfoma de Células T del Adulto/genética , Translocación Genética , Tumor de Wilms/genética , Aniridia/genética , Bandeo Cromosómico , Mapeo Cromosómico , Eliminación de Gen , Predisposición Genética a la Enfermedad , Humanos , Células Híbridas , Familia de Multigenes , Oncogenes , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcr , Proto-Oncogenes
9.
Oncogene ; 14(2): 195-202, 1997 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-9010221

RESUMEN

We report the cloning and characterization of the entire AFX gene which fuses to MLL in acute leukemias with a t(X;ll)(q13;q23). AFX consists of two exons and encodes for a protein of 501 amino acids. We found that normal B- and T-cells contain similar levels of AFX mRNA and that both the MLL/AFX as well as the AFX/MLL fusion transcripts are present in the cell line and the ANLL sample with a t(X;11)(q13;q23). The single intron of the AFX gene consists of 3706 nucleotides. It contains five simple sequence repeats with lengths of at least 12 bps, a chi-like octamer sequence (GCA/TGGA/TGG) and several immunoglobulin heptamer-like sequences (GATAGTG) that are distributed throughout the entire AFX intron sequence. In the KARPAS 45 cell line the breakpoints occur at nucleotides 2913/2914 of the AFX intron and at nucleotides 4900/4901 of the breakpoint cluster region of the MLL gene. The AFX protein belongs to the forkhead protein family. It is highly homologous to the human FKHR protein, the gene of which is disrupted by the t(2;13)(q35;q14), a chromosome rearrangement characteristic of alveolar rhabdomyosarcomas. It is noteworthy that the t(X;11)(q13;q23) in the KARPAS 45 cell line and in one acute nonlymphoblastic leukemia (ANLL) disrupts the forkhead domain of the AFX protein exactly at the same amino acids as does the t(2;13)(q35;q14) in case of the FKHR protein. In addition, the 5'-part of the AFX protein contains a conserved hexapeptide motif (QIYEWM) that is homologous to the functionally important conserved hexapeptide QIYPWM upstream of the homeobox domain in Hox proteins. This motif mediates the co-operative DNA binding of Pbx family members and Hox proteins and, therefore, plays an important role in physiologic and oncogenic processes. In acute leukemias with a t(X;11)(q13;q23), this hexapeptide motif is separated from the remaining forkhead domain within the AFX protein. The predicted amino acid sequence of AFX differs significantly from the partial AFX protein sequence published previously (Genes, Chromosomes and Cancer, 1994, 11, 79-84). This discrepancy can be explained by the occurrence of two sequencing errors in the earlier work at nucleotide number 783 and 844 (loss of a cytosine residue or guanosine residue, respectively) that lead to two reading frame shifts.


Asunto(s)
Proteínas Sanguíneas/genética , Cromosomas Humanos Par 11 , Genes/genética , Intrones/genética , Leucemia/genética , Proto-Oncogenes , Factores de Transcripción , Translocación Genética , Cromosoma X , Enfermedad Aguda , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas de Ciclo Celular , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Datos de Secuencia Molecular , Proteína de la Leucemia Mieloide-Linfoide , Células Tumorales Cultivadas
10.
Oncogene ; 18(33): 4663-71, 1999 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-10467413

RESUMEN

Some chromosomal translocations involved in the origin of leukemias and lymphomas are due to malfunctions of the recombinatorial machinery of immunoglobulin and T-cell receptor-genes. This mechanism has also been proposed for translocations t(4;11)(q21;q23), which are regularly associated with acute pro-B cell leukemias in early childhood. Here, reciprocal chromosomal breakpoints in primary biopsy material of fourteen t(4;11)-leukemia patients were analysed. In all cases, duplications, deletions and inversions of less than a few hundred nucleotides indicative of malfunctioning DNA repair mechanisms were observed. We concluded that these translocation events were initiated by several DNA strand breaks on both participating chromosomes and subsequent DNA repair by 'error-prone-repair' mechanisms, but not by the action of recombinases of the immune system.


Asunto(s)
Daño del ADN , Reparación del ADN , Leucemia de Células B/genética , Proto-Oncogenes , Factores de Transcripción , Translocación Genética , Adolescente , Adulto , Secuencia de Bases , Linfoma de Burkitt/etiología , Linfoma de Burkitt/genética , Niño , Preescolar , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 4 , Proteínas de Unión al ADN/genética , Femenino , N-Metiltransferasa de Histona-Lisina , Humanos , Lactante , Leucemia de Células B/etiología , Masculino , Datos de Secuencia Molecular , Proteína de la Leucemia Mieloide-Linfoide , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Factores de Elongación Transcripcional
11.
J Clin Oncol ; 15(6): 2247-53, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9196137

RESUMEN

BACKGROUND: In the last two decades, it has become evident that secondary leukemias after Hodgkin's disease (HD) are mainly caused by the treatment with alkylating agents, especially mechlorethamine. Since 1978, the German-Austrian trials for childhood HD have used combined chemoradiotherapy without mechlorethamine. PATIENTS AND METHODS: The risk of secondary hematologic malignancies (SHM) was assessed in the total cohort of 667 children treated in four consecutive German-Austrian trials between 1978 and 1990. Primary chemotherapy for stages IA/B and IIA consisted of two cycles of vincristine, procarbazine, prednisone, and doxorubicin (OPPA) or OPA (without procarbazine) and, for more advanced stages, of two cycles of OPPA or OPA plus two, four, or six cycles of COPP or COMP (C, cyclophosphamide; M, methotrexate). Radiotherapy was given in the first study to extended fields, and in later trials to involved fields only. In 591 patients, only primary therapy was given; 76 patients (11%) needed additional salvage therapy. The actuarial survival rate at 15 years is 94%. RESULTS: SHM developed in 5 of 667 patients: four acute myeloid leukemias (AMLs) and one myelodysplastic syndrome (MDS). The estimated cumulative risk for SHM at 15 years is 1.1% (95% CI, 0.0% to 2.2%). Salvage therapy was a significant risk factor for SHM (relative risk, 7.25; P = .03), whereas age, sex, stage of HD, splenectomy, and amount of alkylating agents were not. CONCLUSION: The observed risk of SHM is smaller than in other studies (adults and children) in which chemotherapy with mechlorethamine, vincristine, procarbazine, and prednisone (MOPP) was given. This difference can be attributed to the lower cumulative doses of alkylating agents, the absence of mechlorethamine in the chemotherapy, and the small number of patients who needed salvage therapy in the presented cohort. In general, differences in the incidence of SHM after HD reflect complex differences between treatment strategies.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Enfermedad de Hodgkin/tratamiento farmacológico , Leucemia/inducido químicamente , Mecloretamina/efectos adversos , Adolescente , Niño , Preescolar , Estudios de Cohortes , Terapia Combinada , Ciclofosfamida/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Enfermedad de Hodgkin/radioterapia , Humanos , Lactante , Masculino , Neoplasias Primarias Secundarias/inducido químicamente , Prednisona/administración & dosificación , Prednisona/efectos adversos , Procarbazina/administración & dosificación , Procarbazina/efectos adversos , Factores de Riesgo , Terapia Recuperativa , Tasa de Supervivencia , Vincristina/administración & dosificación , Vincristina/efectos adversos
12.
Leukemia ; 12(5): 758-63, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9593275

RESUMEN

Chromosomal rearrangements constitute a significant feature of leukemogenesis and malignant transformation in general. Nucleotide patterns in the immediate vicinity of the break point may provide important information about the underlying causalities, eg illegitimate recombination events mediated by topoisomerase II, Alu repeats, or VDJ recombinase. In order to facilitate the determination of those DNA patterns, we developed a new fingerprint approach. In a first step, two DNA fragments were independently amplified by long distance PCR: the genomic region carrying the break point and the normal nonrearranged counterpart. Subsequently, both PCR products were digested with restriction enzymes, end-labelled with a fluorescent dye, and subjected to high resolution polyacrylamide gel electrophoresis. By comparing the restriction patterns of the rearranged and the nonrearranged PCR fragments, the break points could be easily localized within a size range coverable by a single sequencing reaction. Finally, the exact DNA sequence across the break point was directly determined. The 'fingerprint' technique is fast, reliable and enables the assay of multiple samples in parallel.


Asunto(s)
Dermatoglifia del ADN/métodos , ADN/genética , Proto-Oncogenes , Factores de Transcripción , Translocación Genética , Secuencia de Bases , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 4 , Cromosomas Humanos Par 9 , ADN/metabolismo , Proteínas de Unión al ADN/genética , Reordenamiento Génico , Genoma Humano , N-Metiltransferasa de Histona-Lisina , Humanos , Datos de Secuencia Molecular , Proteína de la Leucemia Mieloide-Linfoide , Reacción en Cadena de la Polimerasa
13.
Leukemia ; 12(5): 823-7, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9593287

RESUMEN

Balanced translocations of 11q23 are associated with specific clinical features and a poor outcome, but the relevance of deletions involving 11q23 is not clear. Fifty-seven patients with this deletion were collected by the Workshop, 30 had terminal and 27 had interstitial deletions. Twenty-seven patients had acute lymphoblastic leukemia (ALL), 16 had acute myeloid leukemia (AML), one had acute biphenotypic leukemia, one had acute undifferentiated leukemia and 12 had myelodysplastic syndrome (MDS). ALL patients had a median age of 7 years, median white blood cell count (WBC) of 15 x 10(9)/l, and 10/24 had common ALL. AML patients had a median age of 23 years, a median WBC of 49 x 10(9)/l, and 9/16 had M4 or M5. MDS patients were all adult, median age of 69 years, median WBC of 3 x 10(9)/l, and 7/12 had refractory anemia. The clinical outcome depended on diagnosis: children with ALL had a better prognosis (4/16 relapsed, one died) than AML patients; all adults and children with AML and 5/12 MDS patients died. Fluorescence in situ hybridization (FISH) identified 3 del(11q23) as translocations or insertions. Molecular studies revealed a MLL rearrangement in 8/10 patients. Because the involvement of MLL might be of prognostic relevance, identification of a del(11q23) should be an indication for FISH and molecular studies.


Asunto(s)
Cromosomas Humanos Par 11 , Eliminación de Gen , Leucemia Mieloide/genética , Síndromes Mielodisplásicos/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Enfermedad Aguda , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Cariotipificación , Masculino , Persona de Mediana Edad , Pronóstico
14.
Leukemia ; 17(6): 1130-6, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12764380

RESUMEN

The fusion transcript AML1/ETO corresponding to translocation t(8;21)(q22;q22) can be found in approximately 7-12% of childhood de novo AML. Despite the favorable prognosis, some of these patients relapse. Most of MRD studies so far were performed on adults treated not uniformly. Therefore, we analyzed the follow-up of 15 AML1/ETO-positive children using real-time quantitative reverse transcription PCR (RQ-RT-PCR), all enrolled in the multicenter therapy trial AML-BFM 98. AML1/ETO copy numbers were normalized to the control gene ABL and the results were expressed in copy numbers AML1/ETO per 10 000 copies ABL. At diagnosis, a median of 10 789 copies AML1/ETO was found. A linear decrease to about 10 copies (2-4 log) could be seen in most of the children by the start of consolidation. In the majority of cases they remained positive at this low level during the ongoing therapy. Four children relapsed and two of them had a decrease of less than 2 log before starting consolidation. Three of the relapsed children showed, prior to relapse, an increase of the AML1/ETO fusion transcript at 6, 9, and 11 weeks, respectively. These results suggest that monitoring of minimal residual disease using RQ-RT-PCR could be helpful in detecting patients with a higher risk of relapse.


Asunto(s)
Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Adolescente , Antineoplásicos/uso terapéutico , Niño , Preescolar , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Cartilla de ADN/química , Femenino , Dosificación de Gen , Humanos , Cariotipificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Masculino , Recurrencia Local de Neoplasia , Neoplasia Residual , Pronóstico , ARN Mensajero/genética , ARN Neoplásico/genética , Proteína 1 Compañera de Translocación de RUNX1 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Translocación Genética
15.
Leukemia ; 3(6): 431-9, 1989 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2725059

RESUMEN

We have studied the immunophenotypic and genotypic features in 35 infants aged less than 1 year with acute lymphoblastic leukemia (ALL) or acute undifferentiated leukemia (AUL). A CD10 (common ALL antigen)-negative, CD19-positive pre-pre-B ALL phenotype was observed in 24 infants. Seventeen of them had blast cells coexpressing myeloid-associated markers such as CD15A (VIM-D5, MZ17) and/or VIM-2, but neither myeloperoxidase nor platelet peroxidase was detected in five of these cases analyzed by electron microscopy. Five patients showed a typical common ALL, five a pre-B ALL phenotype, and one infant was unclassifiable by surface-marker and morphologic analysis. Cytogenetic data, available in 21 of these patients, revealed chromosomal abnormalities involving 11q23 in 10 infants with a CD10-negative pre-pre-B ALL. Immunoglobulin (Ig) and T cell receptor (TCR) gamma, beta and delta gene analysis of 31 infants showed Ig heavy-chain gene rearrangement in all but one patient with evidence for clonal evolution in six and kappa-light-chain rearrangement in three infants. TCR beta-chain and TCR gamma-chain rearrangement occurred in six and five patients respectively, while TCR delta-chain rearrangement was identified in 15 patients. Our data indicate that ALL in infancy may present with heterogeneous immunophenotypic and genotypic features. The high frequency of coexpression of B-lineage and myeloid surface markers as well as of chromosomal rearrangement involving 11q23 suggests that the clonogenic cell of infant ALL may relate to a multipotent progenitor cell in most cases.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Anticuerpos Monoclonales , Antígenos de Superficie/análisis , Aberraciones Cromosómicas , Cromosomas Humanos Par 11 , Femenino , Reordenamiento Génico de Linfocito B , Reordenamiento Génico de Linfocito T , Genotipo , Humanos , Lactante , Recién Nacido , Cariotipificación , Masculino , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Preleucemia/genética
16.
Leukemia ; 3(10): 708-14, 1989 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2779287

RESUMEN

We have studied the immunophenotypic and genotypic characteristic of acute nonlymphoblastic leukemias (ANLL) in infants aged less than one year. Sixty-four percent of cases (16/25) showed a myeloid or myelomonocytic differentiation pattern and 10 of these were classified as FAB M5 (7 M5a, 3 M5b). Only seven of the latter cases expressed the CD14 antigen. Acute megakaryocytic leukemia with a high number of glycoprotein IIb/IIIa or IIIa positive blasts were identified in five patients. Erythroleukemia with a high percentage of rather mature glycophorin A positive erythroblasts were diagnosed in two infants. Cytogenetic studies were successfully performed in all 20 cases investigated. Abnormalities involving chromosome 11 were present in 10 of 17 patients with an abnormal karyotype including five cases with a t(9;11)(p21;q23). Immunoglobulin (Ig) and T cell receptor (TCR) gene analyses were performed in 20 patients. A rearrangement of Ig heavy chain sequences was detected in five cases (20%), one of whom exhibited multiple rearranged fragments. Three of these patients showed additional TCR delta-chain gene rearrangements, while Ig kappa, TCR beta- as well as TCR gamma-chain genes showed a germline configuration in all cases analyzed. Our study confirms the high incidence of myelomonocytic and monoblastic subtypes in infants with ANLL, which are particularly closely associated with chromosome 11 abnormalities. However, we also observed an unexpected high frequency of megakaryoblastic leukemias as well as erythroleukemias. As previously reported for ALL in infants, ANLL of infancy shows a similar heterogeneity regarding phenotypic and genotypic features.


Asunto(s)
Leucemia Mieloide Aguda/inmunología , Antígenos de Diferenciación/análisis , Aberraciones Cromosómicas , Femenino , Reordenamiento Génico de Linfocito T , Genes de Inmunoglobulinas , Genotipo , Humanos , Lactante , Recién Nacido , Leucemia Mieloide Aguda/genética , Masculino , Fenotipo
17.
Leukemia ; 14(12): 2205-22, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11187912

RESUMEN

Four thousand, four hundred and forty eligible children of up to 18 years of age were treated in four consecutive trials between 1981 and 1995 with the treatment protocols of the Berlin-Frankfurt-Münster (BFM) study group for childhood acute lymphoblastic leukemia (ALL). The probability for event-free survival (pEFS) at 8 years improved from 65.8% in study ALL-BFM 81 to 75.9% in study ALL-BFM 90. The cumulative incidence of recurrences with CNS involvement was 10.1% and 9.3% in studies ALL-BFM 81 and 83, but was reduced to less than 5% in study ALL-BFM 90 (for isolated CNS relapses from 5.3% in study ALL-BFM 81 to 1.1% in study ALL-BFM 90). Four major findings were derived from this series of trials performed by 37 to 96 centers in Germany, Austria, and Switzerland: (1) Reintensification is a crucial part of treatment, even in low risk patients; (2) presymptomatic cranial radiotherapy can be safely reduced to 12 Gy, or even be eliminated if it is replaced by early intensive systemic and intrathecal methotrexate applied; (3) maintenance therapy given a total of 24 months from diagnosis provides a lower rate of systemic relapses than treatment for 18 months; (4) inadequate response to an initial 7-day prednisone window (combined with one intrathecal injection of methotrexate on day 1) defines about 10% of the patients with a very high risk of relapse. For patients with adequate early response (90% of all) an 8-year pEFS of 80% has been achieved in the most recent trial ALL-BFM 90. While it has proven so far to be impossible to improve the outcome for the small group of high risk patients, the number of recurrences could be effectively reduced for the large group of patients responding adequately to the prednisone in vivo sensitivity test. Apart from inadequate prednisone response, patients with hyperleukocytosis, age <1 year, or the presence of the Philadelphia-chromosome (Ph+ ALL) are at a particularly high risk of failure.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adolescente , Niño , Preescolar , Terapia Combinada , Supervivencia sin Enfermedad , Humanos , Lactante , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/radioterapia
18.
Leukemia ; 14(7): 1232-8, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10914547

RESUMEN

Translocations involving 11q23 are among the most common genetic abnormalities in hematologic malignancies, occurring in approximately 5-10% of acute lymphoblastic leukemia (ALL) and 5% of acute myeloblastic leukemia (AML). In 11q23 translocations, the mixed lineage leukemia (MLL) gene on chromosome 11, band q23, is usually disrupted. The human homologue of the rat NG2 chondroitin sulfate proteoglycan molecule, as detected by the monoclonal antibody (moab) 7.1, was shown to be expressed on leukemic cells with MLL rearrangements of children with acute leukemia. We further investigated the reactivity of the moab 7.1 on 533 cell samples of adults (n = 215) and children (n = 318) with acute leukemias (271 AML, 217 B-lineage ALL, 37 T-lineage ALL, eight CD7+ CD56+ myeloid/natural killer cell precursor acute leukemias) by flow cytometry. In AML, 38 samples were positive for moab 7.1 ('20%-cut-off-level'). These moab 7.1-positive AML cases revealed a myelomonocytic-differentiated immunophenotype with coexpression of the NK cell marker CD56 in 33 of 38 cases. Two of eight cell samples of the recently described CD7+ CD56+ myeloid/natural killer cell precursor acute leukemia entity reacted with moab 7.1. In ALL, 35 samples mostly of the pro-B-ALL subtype (33 pro-B-ALL, one common-ALL, one pre-B-ALL) were positive for moab 7.1. 58 (81%) of 72 samples with MLL rearrangements were positive for moab 7.1 including 28/31 with a t(4;11), 16/17 with a t(9;11), 3/5 with a t(11;19), and 2/6 with a del(11)(q23). All moab 7.1-positive ALL (n = 34) and childhood AML (n = 17) cases revealed MLL rearrangements as detected by Southern blot analysis and RT-PCR. However, 11 adults with AML, and one adult with moab 7.1-positive CD7+ CD56+ myeloid/natural killer cell precursor acute leukemia were negative for MLL rearrangements as proved by Southern blot analysis. We conclude that moab 7.1 is a sensitive but not entirely specific marker for the identification of 11q23-associated AML and ALL by flow cytometry in children and adults.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Biomarcadores de Tumor/genética , Cromosomas Humanos Par 11/genética , Proteínas de Unión al ADN/genética , Citometría de Flujo , Reordenamiento Génico , Leucemia/patología , Células Madre Neoplásicas/química , Proto-Oncogenes , Factores de Transcripción , Translocación Genética , Enfermedad Aguda , Adulto , Animales , Especificidad de Anticuerpos , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/inmunología , Southern Blotting , Niño , Cromosomas Humanos Par 11/ultraestructura , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/inmunología , N-Metiltransferasa de Histona-Lisina , Humanos , Inmunofenotipificación , Cariotipificación , Leucemia/clasificación , Leucemia/genética , Proteína de la Leucemia Mieloide-Linfoide , Células Madre Neoplásicas/ultraestructura , Valor Predictivo de las Pruebas , Ratas , Sensibilidad y Especificidad
19.
Leukemia ; 16(9): 1685-90, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12200682

RESUMEN

We used karyotyping, fluorescence in situ hybridization (FISH), Southern blotting, and RT-PCR in order to analyze prospectively 77 infants (less than 1 year of age) with acute lymphoblastic leukemia for the occurrence of 11q23/MLL rearrangements and/or other cytogenetic abnormalities. Out of the 69 informative samples we found an 11q23/MLL rearrangement in 42 cases (61%). Regarding only pro-B ALL cases, the incidence of 11q23/MLL rearranged cases, however, reached more than 90% The infants were treated within the therapy studies ALL-BFM90, ALL-BFM95 and CoALL-05-92. For patients with an adequate follow-up of 4 years the event-free survival of the 11q23/MLL-positive and 11q23/MLL-negative group was 0.2 or 0.64, respectively (P = 0.024). The monoclonal antibody 7.1. (moab 7.1) does not react with normal hematopoetic precursors or mature blood cells but was shown to specifically react with leukemic cells bearing a rearrangement of chromosome 11q23 or the MLL gene, respectively. We, therefore, specifically addressed the question whether the reactivity of moab 7.1, as determined by flow cytometry, may substitute for molecular testing of an 11q23/MLL rearrangement in this cohort of infant ALLs. Reactivity of moab 7.1 indicated a 11q23/MLL rearrangement with a specificity of 100%. However, five of the 11q23/MLL-positive cases did not react with moab 7.1 indicating a sensitivity of 84% only. Three of these five moab 7.1-negative but 11q23/MLL-positive cases could be identified by their unique expression pattern of CD65s and/or CD15. Thus, 95% of all 11q23/MLL-positive ALL cases in infancy may be identified by flow cytometry based on their expression of CD15, CD65s and/or moab 7.1.


Asunto(s)
Antígenos CD/genética , Cromosomas Humanos Par 11/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Southern Blotting , Médula Ósea/química , Aberraciones Cromosómicas , Supervivencia sin Enfermedad , Reordenamiento Génico , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Lactante , Recién Nacido , Cariotipificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico , Estudios Prospectivos , Sensibilidad y Especificidad , Resultado del Tratamiento
20.
Leukemia ; 8(4): 542-8, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7908708

RESUMEN

The t(4;11)(q21;q23) characterizes a distinct clinical entity of childhood and adult acute lymphoblastic leukemia (ALL) with a pre-pre-B-phenotype, monocytoid features, coexpression of CD15 and/or CDw65 and a dismal prognosis. The molecular correlate of the t(4;11) has been identified as a fusion transcript of HRX, a gene on 11q23 with homology to drosophila trithorax gene, and FEL, a serine-proline-rich gene on 4q21 of unknown function. The aim of the current study was to establish a reverse transcription-polymerase chain reaction (RT-PCR) approach for the rapid and sensitive detection of the HRX-FEL fusion transcript associated with the t(4;11). For this purpose, two groups of patients were studied: group A comprised cases with cytogenetically proven t(4;11) including three infant and four adult pre-pre-B-ALL, as well as the two cell lines RS4;11 and MV4;11. Group B consisted of ten adult pre-pre-B-ALL with the identical phenotype, but without cytogenetic confirmation of t(4;11). Using primers complementary to HRX and FEL cDNA sequences 300 to 500 bp 5' and 3' of published breakpoints, respectively, specific amplification products were obtained in all nine cases of group A and in nine of the ten cases of group B. Three different types of fusion transcripts were identified by sequence analysis with HRX breakpoints at nucleotides 4086 and 4218 and FEL breakpoints at nucleotides 1413, 1416, and 1458. These data indicate that RT-PCR allows the detection of HRX-FEL fusion transcripts in the vast majority of cytogenetically proven and immunophenotypically suspected t(4;11) ALL. Hence, this technique may allow identification of a further subset of high risk ALL and may also be useful for the monitoring of minimal residual disease in t(4;11) ALL.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 4 , Proteínas de Neoplasias/análisis , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Translocación Genética , Adulto , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Antígenos de Neoplasias/análisis , Secuencia de Bases , Niño , Humanos , Antígeno Lewis X , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA