Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 143(11): 953-966, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38096358

RESUMEN

ABSTRACT: Relapse after complete remission (CR) remains the main cause of mortality after allogeneic stem cell transplantation for hematological malignancies and, therefore, improved biomarkers for early prediction of relapse remains a critical goal toward development and assessment of preemptive relapse treatment. Because the significance of cancer stem cells as a source of relapses remains unclear, we investigated whether mutational screening for persistence of rare cancer stem cells would enhance measurable residual disease (MRD) and early relapse prediction after transplantation. In a retrospective study of patients who relapsed and patients who achieved continuous-CR with myelodysplastic syndromes and related myeloid malignancies, combined flow cytometric cell sorting and mutational screening for persistence of rare relapse-initiating stem cells was performed in the bone marrow at multiple CR time points after transplantation. In 25 CR samples from 15 patients that later relapsed, only 9 samples were MRD-positive in mononuclear cells (MNCs) whereas flowcytometric-sorted hematopoietic stem and progenitor cells (HSPCs) were MRD-positive in all samples, and always with a higher variant allele frequency than in MNCs (mean, 97-fold). MRD-positivity in HSPCs preceded MNCs in multiple sequential samples, in some cases preceding relapse by >2 years. In contrast, in 13 patients in long-term continuous-CR, HSPCs remained MRD-negative. Enhanced MRD sensitivity was also observed in total CD34+ cells, but HSPCs were always more clonally involved (mean, 8-fold). In conclusion, identification of relapse-initiating cancer stem cells and mutational MRD screening for their persistence consistently enhances MRD sensitivity and earlier prediction of relapse after allogeneic stem cell transplantation.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Humanos , Trasplante Homólogo , Estudios Retrospectivos , Recurrencia Local de Neoplasia , Respuesta Patológica Completa , Enfermedad Crónica , Células Madre Neoplásicas/patología , Recurrencia , Neoplasia Residual/diagnóstico , Neoplasia Residual/patología , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/terapia
2.
J Intern Med ; 296(1): 53-67, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38654517

RESUMEN

BACKGROUND: The Molecular International Prognostic Scoring System (IPSS-M) is the new gold standard for diagnostic outcome prediction in patients with myelodysplastic syndromes (MDS). This study was designed to assess the additive prognostic impact of dynamic transfusion parameters during early follow-up. METHODS: We retrieved complete transfusion data from 677 adult Swedish MDS patients included in the IPSS-M cohort. Time-dependent erythrocyte transfusion dependency (E-TD) was added to IPSS-M features and analyzed regarding overall survival and leukemic transformation (acute myeloid leukemia). A multistate Markov model was applied to assess the prognostic value of early changes in transfusion patterns. RESULTS: Specific clinical and genetic features were predicted for diagnostic and time-dependent transfusion patterns. Importantly, transfusion state both at diagnosis and within the first year strongly predicts outcomes in both lower (LR) and higher-risk (HR) MDSs. In multivariable analysis, 8-month landmark E-TD predicted shorter survival independently of IPSS-M (p < 0.001). A predictive model based on IPSS-M and 8-month landmark E-TD performed significantly better than a model including only IPSS-M. Similar trends were observed in an independent validation cohort (n = 218). Early transfusion patterns impacted both future transfusion requirements and outcomes in a multistate Markov model. CONCLUSION: The transfusion requirement is a robust and available clinical parameter incorporating the effects of first-line management. In MDS, it provides dynamic risk information independently of diagnostic IPSS-M and, in particular, clinical guidance to LR MDS patients eligible for potentially curative therapeutic intervention.


Asunto(s)
Síndromes Mielodisplásicos , Humanos , Síndromes Mielodisplásicos/terapia , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/mortalidad , Femenino , Pronóstico , Masculino , Anciano , Persona de Mediana Edad , Suecia , Cadenas de Markov , Anciano de 80 o más Años , Transfusión de Eritrocitos , Transfusión Sanguínea , Adulto
3.
Blood ; 130(7): 881-890, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28634182

RESUMEN

Mutations in the RNA splicing gene SF3B1 are found in >80% of patients with myelodysplastic syndrome with ring sideroblasts (MDS-RS). We investigated the origin of SF3B1 mutations within the bone marrow hematopoietic stem and progenitor cell compartments in patients with MDS-RS. Screening for recurrently mutated genes in the mononuclear cell fraction revealed mutations in SF3B1 in 39 of 40 cases (97.5%), combined with TET2 and DNMT3A in 11 (28%) and 6 (15%) patients, respectively. All recurrent mutations identified in mononuclear cells could be tracked back to the phenotypically defined hematopoietic stem cell (HSC) compartment in all investigated patients and were also present in downstream myeloid and erythroid progenitor cells. While in agreement with previous studies, little or no evidence for clonal (SF3B1 mutation) involvement could be found in mature B cells, consistent involvement at the pro-B-cell progenitor stage was established, providing definitive evidence for SF3B1 mutations targeting lymphomyeloid HSCs and compatible with mutated SF3B1 negatively affecting lymphoid development. Assessment of stem cell function in vitro as well as in vivo established that only HSCs and not investigated progenitor populations could propagate the SF3B1 mutated clone. Upon transplantation into immune-deficient mice, SF3B1 mutated MDS-RS HSCs differentiated into characteristic ring sideroblasts, the hallmark of MDS-RS. Our findings provide evidence of a multipotent lymphomyeloid HSC origin of SF3B1 mutations in MDS-RS patients and provide a novel in vivo platform for mechanistically and therapeutically exploring SF3B1 mutated MDS-RS.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Linfocitos/metabolismo , Mutación/genética , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Células Mieloides/metabolismo , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Anciano , Anciano de 80 o más Años , Animales , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Empalmosomas/metabolismo
4.
Br J Haematol ; 180(4): 526-533, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29377069

RESUMEN

The mechanisms underlying lenalidomide-resistance of del(5q) MDS stem cells remain to be elucidated and may include cell-intrinsic as well as microenvironmental causes. Abnormal hypolobated megakaryocytes constitute one of the hallmarks of del(5q) MDS. We hypothesized that these cells have potential implications for the regulation of haematopoietic stem cells (HSC) similarly to what has recently been described for megakaryocytes in the murine system. Therefore, we conducted a study to determine the response of abnormal hypolobated megakaryocytes to lenalidomide therapy. We studied lenalidomide-treated patients in the MDS-004 trial as well as a cohort seen at our institution. Morphological evaluation at time of complete cytogenetic remission (CCyR) demonstrated the persistence of hypolobated megakaryocytes in all evaluable patients (n = 9). Furthermore, we provide evidence that the abnormal hypolobated morphology is restricted to del(5q) megakaryocytes, both at diagnosis and during CCyR. Using fluorescence in situ hybridisation analysis on flow-sorted stem- and progenitor populations, we observed a similar degree of clonal involvement in megakaryocyte-erythroid-progenitors as in HSC. Taken together, our findings suggest that megakaryocyte morphology might aid in the evaluation of patients where discontinuation of lenalidomide is considered and offers interesting hypotheses for further investigation of lenalidomide resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Deleción Cromosómica , Cromosomas Humanos Par 5 , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/genética , Megacariocitos/metabolismo , Talidomida/análogos & derivados , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Médula Ósea/patología , Evolución Clonal , Análisis Citogenético , Neoplasias Hematológicas/diagnóstico , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Lenalidomida , Células Progenitoras de Megacariocitos y Eritrocitos/metabolismo , Células Progenitoras de Megacariocitos y Eritrocitos/patología , Megacariocitos/patología , Inducción de Remisión , Talidomida/administración & dosificación , Talidomida/efectos adversos , Talidomida/uso terapéutico , Resultado del Tratamiento
5.
Blood ; 126(2): 233-41, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25957392

RESUMEN

Refractory anemia with ring sideroblasts (RARS) is a myelodysplastic syndrome (MDS) characterized by isolated erythroid dysplasia and 15% or more bone marrow ring sideroblasts. Ring sideroblasts are found also in other MDS subtypes, such as refractory cytopenia with multilineage dysplasia and ring sideroblasts (RCMD-RS). A high prevalence of somatic mutations of SF3B1 was reported in these conditions. To identify mutation patterns that affect disease phenotype and clinical outcome, we performed a comprehensive mutation analysis in 293 patients with myeloid neoplasm and 1% or more ring sideroblasts. SF3B1 mutations were detected in 129 of 159 cases (81%) of RARS or RCMD-RS. Among other patients with ring sideroblasts, lower prevalence of SF3B1 mutations and higher prevalence of mutations in other splicing factor genes were observed (P < .001). In multivariable analyses, patients with SF3B1 mutations showed significantly better overall survival (hazard ratio [HR], .37; P = .003) and lower cumulative incidence of disease progression (HR = 0.31; P = .018) compared with SF3B1-unmutated cases. The independent prognostic value of SF3B1 mutation was retained in MDS without excess blasts, as well as in sideroblastic categories (RARS and RCMD-RS). Among SF3B1-mutated patients, coexisting mutations in DNA methylation genes were associated with multilineage dysplasia (P = .015) but had no effect on clinical outcome. TP53 mutations were frequently detected in patients without SF3B1 mutation, and were associated with poor outcome. Thus, SF3B1 mutation identifies a distinct MDS subtype that is unlikely to develop detrimental subclonal mutations and is characterized by indolent clinical course and favorable outcome.


Asunto(s)
Anemia Sideroblástica/genética , Mutación , Síndromes Mielodisplásicos/clasificación , Síndromes Mielodisplásicos/genética , Fosfoproteínas/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/epidemiología , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/epidemiología , Pronóstico , Factores de Empalme de ARN , Adulto Joven
6.
Haematologica ; 102(3): 498-508, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27884971

RESUMEN

A high proportion of patients with lower-risk del(5q) myelodysplastic syndromes will respond to treatment with lenalidomide. The median duration of transfusion-independence is 2 years with some long-lasting responses, but almost 40% of patients progress to acute leukemia by 5 years after starting treatment. The mechanisms underlying disease progression other than the well-established finding of small TP53-mutated subclones at diagnosis remain unclear. We studied a longitudinal cohort of 35 low- and intermediate-1-risk del(5q) patients treated with lenalidomide (n=22) or not (n=13) by flow cytometric surveillance of hematopoietic stem and progenitor cell subsets, targeted sequencing of mutational patterns, and changes in the bone marrow microenvironment. All 13 patients with disease progression were identified by a limited number of mutations in TP53, RUNX1, and TET2, respectively, with PTPN11 and SF3B1 occurring in one patient each. TP53 mutations were found in seven of nine patients who developed acute leukemia, and were documented to be present in the earliest sample (n=1) and acquired during lenalidomide treatment (n=6). By contrast, analysis of the microenvironment, and of hematopoietic stem and progenitor cells by flow cytometry was of limited prognostic value. Based on our data, we advocate conducting a prospective study aimed at investigating, in a larger number of cases of del(5q) myelodysplastic syndromes, whether the detection of such mutations before and after lenalidomide treatment can guide clinical decision-making.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 5 , Mutación , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Anciano , Anciano de 80 o más Años , Biomarcadores , Biología Computacional/métodos , Progresión de la Enfermedad , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Lenalidomida , Masculino , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Síndromes Mielodisplásicos/terapia , Pronóstico , Nicho de Células Madre , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Resultado del Tratamiento
7.
Br J Haematol ; 171(4): 478-90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26255870

RESUMEN

Refractory anaemia with ring sideroblasts (RARS) is distinguished by hyperplastic inefficient erythropoiesis, aberrant mitochondrial ferritin accumulation and anaemia. Heterozygous mutations in the spliceosome gene SF3B1 are found in a majority of RARS cases. To explore the link between SF3B1 mutations and anaemia, we studied mutated RARS CD34(+) marrow cells with regard to transcriptome sequencing, splice patterns and mutational allele burden during erythroid differentiation. Transcriptome profiling during early erythroid differentiation revealed a marked up-regulation of genes involved in haemoglobin synthesis and in the oxidative phosphorylation process, and down-regulation of mitochondrial ABC transporters compared to normal bone marrow. Moreover, mis-splicing of genes involved in transcription regulation, particularly haemoglobin synthesis, was confirmed, indicating a compromised haemoglobinization during RARS erythropoiesis. In order to define the phase during which erythroid maturation of SF3B1 mutated cells is most affected, we assessed allele burden during erythroid differentiation in vitro and in vivo and found that SF3B1 mutated erythroblasts showed stable expansion until late erythroblast stage but that terminal maturation to reticulocytes was significantly reduced. In conclusion, SF3B1 mutated RARS progenitors display impaired splicing with potential downstream consequences for genes of key importance for haemoglobin synthesis and terminal erythroid differentiation.


Asunto(s)
Anemia Refractaria/genética , Anemia Sideroblástica/genética , Eritropoyesis/genética , Hemoglobinas/biosíntesis , Fosfoproteínas/genética , Empalme del ARN/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Anciano , Anciano de 80 o más Años , Anemia Refractaria/sangre , Anemia Sideroblástica/sangre , Transporte Biológico/genética , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Heterogeneidad Genética , Humanos , Hierro/metabolismo , Fosfoproteínas/fisiología , Isoformas de Proteínas/genética , Factores de Empalme de ARN , ARN Mensajero/genética , Ribonucleoproteína Nuclear Pequeña U2/fisiología , Análisis de Secuencia de ARN , Transducción de Señal/genética
8.
Cancer Res ; 84(2): 211-225, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-37921711

RESUMEN

Myelodysplastic syndromes with ring sideroblasts (MDS-RS) commonly develop from hematopoietic stem cells (HSC) bearing mutations in the splicing factor SF3B1 (SF3B1mt). Direct studies into MDS-RS pathobiology have been limited by a lack of model systems that fully recapitulate erythroid biology and RS development and the inability to isolate viable human RS. Here, we combined successful direct RS isolation from patient samples, high-throughput multiomics analysis of cells encompassing the SF3B1mt stem-erythroid continuum, and functional assays to investigate the impact of SF3B1mt on erythropoiesis and RS accumulation. The isolated RS differentiated, egressed into the blood, escaped traditional nonsense-mediated decay (NMD) mechanisms, and leveraged stress-survival pathways that hinder wild-type hematopoiesis through pathogenic GDF15 overexpression. Importantly, RS constituted a contaminant of magnetically enriched CD34+ cells, skewing bulk transcriptomic data. Mis-splicing in SF3B1mt cells was intensified by erythroid differentiation through accelerated RNA splicing and decreased NMD activity, and SF3B1mt led to truncations in several MDS-implicated genes. Finally, RNA mis-splicing induced an uncoupling of RNA and protein expression, leading to critical abnormalities in proapoptotic p53 pathway genes. Overall, this characterization of erythropoiesis in SF3B1mt RS provides a resource for studying MDS-RS and uncovers insights into the unexpectedly active biology of the "dead-end" RS. SIGNIFICANCE: Ring sideroblast isolation combined with state-of-the-art multiomics identifies survival mechanisms underlying SF3B1-mutant erythropoiesis and establishes an active role for erythroid differentiation and ring sideroblasts themselves in SF3B1-mutant myelodysplastic syndrome pathogenesis.


Asunto(s)
Síndromes Mielodisplásicos , Fosfoproteínas , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Empalme del ARN/genética , Mutación , Factores de Transcripción/metabolismo , ARN/metabolismo
9.
Clin Cancer Res ; 29(20): 4256-4267, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37498312

RESUMEN

PURPOSE: Ring sideroblasts (RS) define the low-risk myelodysplastic neoplasm (MDS) subgroup with RS but may also reflect erythroid dysplasia in higher risk myeloid neoplasm. The benign behavior of MDS with RS (MDSRS+) is limited to SF3B1-mutated cases without additional high-risk genetic events, but one third of MDSRS+ carry no SF3B1 mutation, suggesting that different molecular mechanisms may underlie RS formation. We integrated genomic and transcriptomic analyses to evaluate whether transcriptome profiles may improve current risk stratification. EXPERIMENTAL DESIGN: We studied a prospective cohort of MDSRS+ patients irrespective of World Health Organization (WHO) class with regard to somatic mutations, copy-number alterations, and bone marrow CD34+ cell transcriptomes to assess whether transcriptome profiles add to prognostication and provide input on disease classification. RESULTS: SF3B1, SRSF2, or TP53 multihit mutations were found in 89% of MDSRS+ cases, and each mutation category was associated with distinct clinical outcome, gene expression, and alternative splicing profiles. Unsupervised clustering analysis identified three clusters with distinct hemopoietic stem and progenitor (HSPC) composition, which only partially overlapped with mutation groups. IPSS-M and the transcriptome-defined proportion of megakaryocyte/erythroid progenitors (MEP) independently predicted survival in multivariable analysis. CONCLUSIONS: These results provide essential input on the molecular basis of SF3B1-unmutated MDSRS+ and propose HSPC quantification as a prognostic marker in myeloid neoplasms with RS.


Asunto(s)
Genómica , Neoplasias , Humanos , Factores de Empalme de ARN/genética , Estudios Prospectivos , Medición de Riesgo , Perfilación de la Expresión Génica , Mutación , Fosfoproteínas/genética , Pronóstico
10.
Nat Cell Biol ; 24(3): 299-306, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35292784

RESUMEN

Transfer RNA-derived fragments (tRFs) are emerging small noncoding RNAs that, although commonly altered in cancer, have poorly defined roles in tumorigenesis1. Here we show that pseudouridylation (Ψ) of a stem cell-enriched tRF subtype2, mini tRFs containing a 5' terminal oligoguanine (mTOG), selectively inhibits aberrant protein synthesis programmes, thereby promoting engraftment and differentiation of haematopoietic stem and progenitor cells (HSPCs) in patients with myelodysplastic syndrome (MDS). Building on evidence that mTOG-Ψ targets polyadenylate-binding protein cytoplasmic 1 (PABPC1), we employed isotope exchange proteomics to reveal critical interactions between mTOG and functional RNA-recognition motif (RRM) domains of PABPC1. Mechanistically, this hinders the recruitment of translational co-activator PABPC1-interacting protein 1 (PAIP1)3 and strongly represses the translation of transcripts sharing pyrimidine-enriched sequences (PES) at the 5' untranslated region (UTR), including 5' terminal oligopyrimidine tracts (TOP) that encode protein machinery components and are frequently altered in cancer4. Significantly, mTOG dysregulation leads to aberrantly increased translation of 5' PES messenger RNA (mRNA) in malignant MDS-HSPCs and is clinically associated with leukaemic transformation and reduced patient survival. These findings define a critical role for tRFs and Ψ in difficult-to-treat subsets of MDS characterized by high risk of progression to acute myeloid leukaemia (AML).


Asunto(s)
Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Células Madre Hematopoyéticas/metabolismo , Humanos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Factores de Iniciación de Péptidos/metabolismo , Seudouridina , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Proteínas de Unión al ARN/genética
11.
Haematologica ; 96(7): 963-71, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21719884

RESUMEN

BACKGROUND: Patients with chromosome 5 abnormalities and high-risk myelodysplastic syndromes or acute myeloid leukemia have a poor outcome. We hypothesized that increasing doses of lenalidomide may benefit this group of patients by inhibiting the tumor clone, as assessed by fluorescence in situ hybridization for del(5q31). DESIGN AND METHODS: Twenty-eight patients at diagnosis or with relapsed disease and not eligible for standard therapy (16 with acute myeloid leukemia, 12 with intermediate-risk 2 or high-risk myelodysplastic syndrome) were enrolled in this prospective phase II multicenter trial and treated with lenalidomide up to 30 mg daily for 16 weeks. Three patients had isolated del(5q), six had del(5q) plus one additional aberration, 14 had del(5q) and a complex karyotype, four had monosomy 5, and one had del(5q) identified by fluorescence in situ hybridization only. RESULTS: Major and minor cytogenetic responses, assessed by fluorescence in situ hybridization, were achieved in 5/26 (19%) and 2/26 (8%) patients, respectively, who received one or more dose of lenalidomide, while two patients achieved only a bone marrow response. Nine of all 26 patients (35%) and nine of the ten who completed the 16 weeks of trial responded to treatment. Using the International Working Group criteria for acute myeloid leukemia and myelodysplastic syndrome the overall response rate in treated patients with acute myeloid leukemia was 20% (3/15), while that for patients with myelodysplastic syndrome was 36% (4/11). Seven patients stopped therapy due to progressive disease and nine because of complications, most of which were disease-related. Response rates were similar in patients with isolated del(5q) and in those with additional aberrations. Interestingly, patients with TP53 mutations responded less well than those without mutations (2/13 versus 5/9, respectively; P=0.047). No responses were observed among 11 cases with deleterious TP53 mutations. CONCLUSIONS: Our data support a role for higher doses of lenalidomide in poor prognosis patients with myelodysplastic syndrome and acute myeloid leukemia with deletion 5q. (Clinicaltrials.gov identifier NCT00761449).


Asunto(s)
Antineoplásicos/administración & dosificación , Aberraciones Cromosómicas , Cromosomas Humanos Par 5/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Talidomida/análogos & derivados , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Secuencia de Bases , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Femenino , Regulación Leucémica de la Expresión Génica , Humanos , Lenalidomida , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Mutación/genética , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/mortalidad , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Talidomida/administración & dosificación , Talidomida/efectos adversos , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
12.
Blood Adv ; 5(4): 1003-1016, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33591326

RESUMEN

Although copy number alterations (CNAs) and translocations constitute the backbone of the diagnosis and prognostication of acute myeloid leukemia (AML), techniques used for their assessment in routine diagnostics have not been reconsidered for decades. We used a combination of 2 next-generation sequencing-based techniques to challenge the currently recommended conventional cytogenetic analysis (CCA), comparing the approaches in a series of 281 intensively treated patients with AML. Shallow whole-genome sequencing (sWGS) outperformed CCA in detecting European Leukemia Net (ELN)-defining CNAs and showed that CCA overestimated monosomies and suboptimally reported karyotype complexity. Still, the concordance between CCA and sWGS for all ELN CNA-related criteria was 94%. Moreover, using in silico dilution, we showed that 1 million reads per patient would be enough to accurately assess ELN-defining CNAs. Total genomic loss, defined as a total loss ≥200 Mb by sWGS, was found to be a better marker for genetic complexity and poor prognosis compared with the CCA-based definition of complex karyotype. For fusion detection, the concordance between CCA and whole-transcriptome sequencing (WTS) was 99%. WTS had better sensitivity in identifying inv(16) and KMT2A rearrangements while showing limitations in detecting lowly expressed PML-RARA fusions. Ligation-dependent reverse transcription polymerase chain reaction was used for validation and was shown to be a fast and reliable method for fusion detection. We conclude that a next-generation sequencing-based approach can replace conventional CCA for karyotyping, provided that efforts are made to cover lowly expressed fusion transcripts.


Asunto(s)
Leucemia Mieloide Aguda , Aberraciones Cromosómicas , Análisis Citogenético , Variaciones en el Número de Copia de ADN , Humanos , Cariotipificación , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética
14.
Leukemia ; 34(1): 271-282, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31375745

RESUMEN

Established cell culture systems have failed to accurately recapitulate key features of terminal erythroid maturation, hampering our ability to in vitro model and treat diseases with impaired erythropoiesis such as myelodysplastic syndromes with ring sideroblasts (MDS-RS). We developed an efficient and robust three-dimensional (3D) scaffold culture model supporting terminal erythroid differentiation from both mononuclear (MNC) or CD34+-enriched primary bone marrow cells from healthy donors and MDS-RS patients. While CD34+ cells did not proliferate beyond two weeks in 2D suspension cultures, the 3D scaffolds supported CD34+ and MNC erythroid proliferation over four weeks demonstrating the importance of the 3D environment. CD34+ cells cultured in 3D facilitated the highest expansion and maturation of erythroid cells, including generation of erythroblastic islands and enucleated erythrocytes, while MNCs supported multi-lineage hemopoietic differentiation and cytokine secretion relevant for MDS-RS. Importantly, MDS-RS 3D-cultures supported de novo generation of ring sideroblasts and maintenance of the mutated clone. The 3D cultures effectively model a clonal disease characterized by terminal erythroid failure and can be used to assess therapeutic compounds.


Asunto(s)
Técnicas de Cultivo de Célula , Eritropoyesis , Síndromes Mielodisplásicos , Antígenos CD34 , Diferenciación Celular/fisiología , Células Cultivadas , Células Precursoras Eritroides/citología , Humanos , Células Madre Mesenquimatosas/citología
15.
Oncol Rep ; 22(1): 137-42, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19513515

RESUMEN

High-dose therapy (HDT) followed by autologous stem cell transplantation (ASCT) is the most common treatment for patients under 65 years of age with multiple myeloma (MM). In this study, we present a retrospective analysis of the prognostic impact of different factors in patients who have received this treatment as first line therapy in our centre. Abnormalities in chromosome 13 were identified by fluorescence in situ hybridization at the time of diagnosis. The median overall survival (OS) and progression-free survival (PFS) from transplantation time in the whole group of 193 patients were 90 and 48 months respectively. The median follow-up was 65 months (range: 6-186 months). The complete remission (CR) rate in patients with and without del(13) was 31 and 40% respectively whereas the median OS in patients with del(13) was 58 months but not reached in patients without del(13) (p=0.006). The PFS was 26 months in patients with del(13) and 84 months in those without del(13) (p=0.001). The transplantation related mortality was 2.5% both in the absence and presence of del(13). Patients who achieved CR following ASCT had longer OS and PFS when compared to those who only achieved partial remission. Thus, this study confirms the role of del(13) as a marker of poor prognosis. Multivariate analysis showed that the existence of del(13) was the only single independent factor effecting survival (p=0.001). In patients without del(13), the prognostic impact was even stronger when combined with the plasma cell load in the bone marrow (p=0.020), whereas the plasma cell load had no effect on survival of patients with del(13). Overall, the absence of del(13) in combination with low plasma cell infiltration at diagnosis predicts the best survival.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 13 , Mieloma Múltiple/cirugía , Células Plasmáticas/inmunología , Trasplante de Células Madre , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Melfalán/administración & dosificación , Persona de Mediana Edad , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Mieloma Múltiple/mortalidad , Agonistas Mieloablativos/administración & dosificación , Selección de Paciente , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/mortalidad , Sobrevivientes , Factores de Tiempo , Acondicionamiento Pretrasplante/métodos , Trasplante Autólogo , Resultado del Tratamiento
16.
Oncotarget ; 8(17): 28812-28825, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-28427179

RESUMEN

Azacitidine (Aza) is first-line treatment for patients with high-risk myelodysplastic syndromes (MDS), although its precise mechanism of action is unknown. We performed the first study to globally evaluate the epigenetic effects of Aza on MDS bone marrow progenitor cells assessing gene expression (RNA seq), DNA methylation (Illumina 450k) and the histone modifications H3K18ac and H3K9me3 (ChIP seq). Aza induced a general increase in gene expression with 924 significantly upregulated genes but this increase showed no correlation with changes in DNA methylation or H3K18ac, and only a weak association with changes in H3K9me3. Interestingly, we observed activation of transcripts containing 15 endogenous retroviruses (ERVs) confirming previous cell line studies. DNA methylation decreased moderately in 99% of all genes, with a median ß-value reduction of 0.018; the most pronounced effects seen in heterochromatin. Aza-induced hypomethylation correlated significantly with change in H3K9me3. The pattern of H3K18ac and H3K9me3 displayed large differences between patients and healthy controls without any consistent pattern induced by Aza. We conclude that the marked induction of gene expression only partly could be explained by epigenetic changes, and propose that activation of ERVs may contribute to the clinical effects of Aza in MDS.


Asunto(s)
Antineoplásicos/uso terapéutico , Azacitidina/uso terapéutico , Células de la Médula Ósea/efectos de los fármacos , Retrovirus Endógenos/genética , Histonas/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Antígenos CD34/metabolismo , Antineoplásicos/farmacología , Azacitidina/farmacología , Células de la Médula Ósea/fisiología , Células Cultivadas , Inmunoprecipitación de Cromatina , Biología Computacional , Metilación de ADN/efectos de los fármacos , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Síndromes Mielodisplásicos/genética , Análisis de Secuencia de ARN , Transcriptoma , Resultado del Tratamiento
17.
Exp Hematol ; 33(3): 380-7, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15730862

RESUMEN

OBJECTIVE: The present study was designed to investigate the influence of the administration sequence of busulphan (Bu) and cyclophosphamide (Cy) during conditioning regimen on myeloablative and immunosuppressive effects and on engraftment. METHODS: Female Balb/C mice were treated with either Bu-Cy or Cy-Bu (assigned order of administration). Bu was administered as 8.75 mg/kg/day x 4 and Cy as 100 mg/kg/day x 2. The control consisted of untreated animals. Bone marrow and spleen were harvested during the conditioning regimen and for up to 19 days after treatment. Colony-forming unit granulocyte macrophage assay was performed on marrow cells. Immunological analyses were performed using spleen cells. Liver status was determined using aspartate amino transferase (AST), alanine amino transferase (ALT), and bilirubin. Animals assigned for engraftment study were conditioned as above and transplanted using sca-1 cells from male Balb/C donors. Engraftment was followed using fluorescence in situ hybridization up to 30 days posttransplantation. RESULTS: No significant difference in myeloablative effect was observed between treatments. Immunosuppressive activity expressed as CD3+/CD19+ and CD4+/CD8+ was also similar. Levels of cytokines interleukin 2, tumor necrosis factor alpha, and interferon gamma at the end of the conditioning regimen were lower in the Cy-Bu group, while liver enzymes were higher after the Bu-Cy regimen. Engraftment in bone marrow was reached faster within the first 20 days after conditioning with Cy-Bu compared to Bu-Cy. However, no difference in chimerism was observed at 30 days. CONCLUSION: Cy-Bu treatment resulted in lower levels of cytokines, faster bone marrow engraftment, and lower values of liver enzymes compared to Bu-Cy regimen, which may benefit stem cell transplantation outcomes.


Asunto(s)
Trasplante de Médula Ósea , Busulfano/administración & dosificación , Ciclofosfamida/administración & dosificación , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/administración & dosificación , Acondicionamiento Pretrasplante , Animales , Antígenos CD/sangre , Células de la Médula Ósea/fisiología , Citocinas/sangre , Quimioterapia Combinada , Femenino , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos BALB C , Bazo/fisiología , Acondicionamiento Pretrasplante/métodos
18.
Oncotarget ; 7(16): 22103-15, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-26959885

RESUMEN

Early therapeutic decision-making is crucial in patients with higher-risk MDS. We evaluated the impact of clinical parameters and mutational profiles in 134 consecutive patients treated with azacitidine using a combined cohort from Karolinska University Hospital (n=89) and from King's College Hospital, London (n=45). While neither clinical parameters nor mutations had a significant impact on response rate, both karyotype and mutational profile were strongly associated with survival from the start of treatment. IPSS high-risk cytogenetics negatively impacted overall survival (median 20 vs 10 months; p<0.001), whereas mutations in histone modulators (ASXL1, EZH2) were associated with prolonged survival (22 vs 12 months, p=0.01). This positive association was present in both cohorts and remained highly significant in the multivariate cox model. Importantly, patients with mutations in histone modulators lacking high-risk cytogenetics showed a survival of 29 months compared to only 10 months in patients with the opposite pattern. While TP53 was negatively associated with survival, neither RUNX1-mutations nor the number of mutations appeared to influence survival in this cohort. We propose a model combining histone modulator mutational screening with cytogenetics in the clinical decision-making process for higher-risk MDS patients eligible for treatment with azacitidine.


Asunto(s)
Biomarcadores/análisis , Histonas/genética , Síndromes Mielodisplásicos/genética , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/uso terapéutico , Análisis Mutacional de ADN , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Resultado del Tratamiento
19.
Transplantation ; 79(11): 1607-14, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15940052

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSC) are progenitors of mesenchymal tissues such as bone, cartilage, and adipose. Adult human leukocyte antigen (HLA)-matched MSC have been used in cellular therapies of bone disorders such as osteogenesis imperfecta, with promising results. METHODS: A female fetus with multiple intrauterine fractures, diagnosed as severe osteogenesis imperfecta, underwent transplantation with allogeneic HLA-mismatched male fetal MSC in the 32nd week of gestation. Engraftment analyses of donor cells, immunologic reaction against donor cells, and the well-being of the patient were assessed. RESULTS: At 9 months of age, on slides stained for osteocalcin or osteopontin, a centromeric XY-specific probe revealed 0.3% of XY-positive cells in a bone biopsy specimen. Whole Y genome fluorescent in situ hybridization staining showed a median of 7.4% Y-positive cells (range, 6.8%-16.6%). Bone histology showed regularly arranged and configurated bone trabeculae. Patient lymphocyte proliferation against donor MSC was not observed in co-culture experiments performed in vitro after MSC injection. Complementary bisphosphonate treatment was begun at 4 months. During the first 2 years of life, three fractures were noted. At 2 years of corrected age, psychomotor development was normal and growth followed the same channel, -5 SD. CONCLUSIONS: The authors' findings show that allogeneic fetal MSC can engraft and differentiate into bone in a human fetus even when the recipient is immunocompetent and HLA-incompatible.


Asunto(s)
Trasplante de Tejido Fetal , Osteogénesis Imperfecta/terapia , Complicaciones del Embarazo/terapia , Trasplante de Células Madre , Adulto , Biopsia , Huesos/patología , Colágeno/genética , Colágeno Tipo I , Cartilla de ADN , Femenino , Edad Gestacional , Antígenos HLA-DR/genética , Cadenas HLA-DRB1 , Humanos , Hibridación Fluorescente in Situ , Lactante , Recién Nacido , Masculino , Mesodermo , Embarazo , Quimera por Trasplante
20.
Methods Mol Biol ; 1313: 131-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25947661

RESUMEN

During the past decades, several studies in animals have displayed the ability of cells from the bone marrow (BM) to participate in regeneration of various tissues including skeletal muscle tissue. Studies in mice have demonstrated that regular physical activity is sufficient to induce contribution of BM derived cells to the skeletal muscle tissue, suggesting that this is part of the physiological remodeling of skeletal muscle. To analyze whether BM-derived cells participate in skeletal muscle remodeling in human, we developed a protocol of immunofluorescence in combination with fluorescence in situ hybridization (FISH) that enables the detection of male donor bone marrow cell contribution to female skeletal muscle tissue.


Asunto(s)
Células de la Médula Ósea/metabolismo , Hibridación Fluorescente in Situ , Fibras Musculares Esqueléticas/metabolismo , Cromosoma X , Cromosoma Y , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones , Fibras Musculares Esqueléticas/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA