Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 690
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 78(3): 477-492.e8, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32386542

RESUMEN

Myelofibrosis is a severe myeloproliferative neoplasm characterized by increased numbers of abnormal bone marrow megakaryocytes that induce fibrosis, destroying the hematopoietic microenvironment. To determine the cellular and molecular basis for aberrant megakaryopoiesis in myelofibrosis, we performed single-cell transcriptome profiling of 135,929 CD34+ lineage- hematopoietic stem and progenitor cells (HSPCs), single-cell proteomics, genomics, and functional assays. We identified a bias toward megakaryocyte differentiation apparent from early multipotent stem cells in myelofibrosis and associated aberrant molecular signatures. A sub-fraction of myelofibrosis megakaryocyte progenitors (MkPs) are transcriptionally similar to healthy-donor MkPs, but the majority are disease specific, with distinct populations expressing fibrosis- and proliferation-associated genes. Mutant-clone HSPCs have increased expression of megakaryocyte-associated genes compared to wild-type HSPCs, and we provide early validation of G6B as a potential immunotherapy target. Our study paves the way for selective targeting of the myelofibrosis clone and illustrates the power of single-cell multi-omics to discover tumor-specific therapeutic targets and mediators of tissue fibrosis.


Asunto(s)
Hematopoyesis/fisiología , Megacariocitos/patología , Mielofibrosis Primaria/sangre , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Femenino , Regulación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Megacariocitos/fisiología , Persona de Mediana Edad , Mutación , Receptores Inmunológicos/genética , Análisis de la Célula Individual/métodos
3.
Blood ; 134(10): 814-825, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31270105

RESUMEN

Monocyte-derived fibrocytes recently garnered attention because the novel pathogenesis of myelofibrosis (MF), and suppression of fibrocyte differentiation by serum amyloid P remarkably improved MF. We previously revealed that human fibrocytes highly expressed signaling lymphocytic activation molecule F7 (SLAMF7) compared with macrophages and that SLAMF7high monocytes in the peripheral blood (PB) of MF patients were significantly elevated relative to those in healthy controls (HCs). In this study, we evaluated SLAMF7high monocyte percentage in the PB of HCs, myeloproliferative neoplasm (MPN) patients with MF, and MPN patients without MF by using a cross-sectional approach. We found that MPN patients with MF who harbored JAK2V617F had a significantly elevated SLAMF7high monocyte percentage, which correlated positively with the JAK2V617F allele burden. In addition, the serum concentration of interleukin-1ra (IL-1ra) was significantly correlated with the SLAMF7high monocyte percentage and JAK2V617F allele burden. These findings suggest that both SLAMF7high monocytes and IL-1ra could be useful noninvasive markers of MF onset. Furthermore, the JAK2V617F allele burden of SLAMF7high monocytes was significantly higher than that of SLAMF7low monocytes and could be a potential target of elotuzumab (Elo), an anti-SLAMF7 antibody used for treating multiple myeloma. Elo independently inhibited differentiation of fibrocytes derived not only from HCs but also from MF patients in vitro. Elo also ameliorated MF and splenomegaly induced by romiplostim administration in humanized NOG mice. In conclusion, an increase of SLAMF7high monocytes with higher JAK2V617F allele burden was associated with the onset of MF in MPN patients harboring JAK2V617F, and Elo could be a therapeutic agent for MPN patients with MF who harbor JAK2V617F.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Janus Quinasa 2/genética , Monocitos/patología , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Recuento de Células Sanguíneas , Proliferación Celular , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Monocitos/metabolismo , Mutación Missense , Fenilalanina/genética , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/patología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Valina/genética
4.
Eur J Haematol ; 107(6): 624-633, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34411333

RESUMEN

BACKGROUND: Hydroxyurea (HU) treatment of patients with essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF) (MPNs) normalizes elevated blood cell counts within weeks in the large majority of patients. Studies on the impact of HU upon the kinetics of the JAK2V617F allele burden, leukocyte, and platelet counts over time are scarce. PURPOSE: Using data-driven analysis as a novel tool to model the kinetics of the JAK2V617F allele burden and blood cell counts over time during treatment with HU. MATERIAL AND METHODS: Using serial measurements of JAK2V617F and correlation analysis of routine hematological values (the Hb-concentration, leukocyte count, platelet count, and lactic dehydrogenase), we present a detailed description and analysis of the kinetics of the JAK2V617F, leukocyte, and platelet counts and lactic dehydrogenase in 27 patients (PV = 18; ET = 7; PMF = 2), who were followed in the Danish randomized trial (DALIAH). To further analyze the JAK2V617F kinetics, we use a machine learning clustering algorithm to group the response patterns. RESULTS: Response patterns were highly heterogeneous, with clustering resulting in 3 groups and 3 outliers. In the large majority of patients, HU treatment was initially associated with a modest decline in the JAK2V617F allele burden in concert with a decline in leukocyte and platelet counts. However, HU did not induce a sustained and continuous decrease in the JAK2V617F allele burden. CONCLUSION: Using data-driven analysis of the JAK2V617F allele burden, leukocyte, and platelet kinetics during treatment with HU, we have shown that HU does not induce a sustained decrease in the JAK2V617F allele burden and neither induces sustained normalization of elevated cell counts in MPN patients. Our results may explain why MPN patients during treatment with HU still have a substantially increased risk of thrombosis.


Asunto(s)
Alelos , Antineoplásicos/uso terapéutico , Recuento de Células Sanguíneas , Hidroxiurea/uso terapéutico , Janus Quinasa 2/genética , Policitemia Vera/genética , Mielofibrosis Primaria/genética , Trombocitemia Esencial/genética , Anciano , Antineoplásicos/administración & dosificación , Estudios de Cohortes , Femenino , Humanos , Hidroxiurea/administración & dosificación , Interferón alfa-2/administración & dosificación , Cinética , Masculino , Persona de Mediana Edad , Policitemia Vera/sangre , Policitemia Vera/tratamiento farmacológico , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/tratamiento farmacológico , Trombocitemia Esencial/sangre , Trombocitemia Esencial/tratamiento farmacológico
5.
Arterioscler Thromb Vasc Biol ; 40(10): e262-e272, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32814440

RESUMEN

OBJECTIVE: The risk of thrombosis in myeloproliferative neoplasms, such as primary myelofibrosis varies depending on the type of key driving mutation (JAK2 [janus kinase 2], CALR [calreticulin], and MPL [myeloproliferative leukemia protein or thrombopoietin receptor]) and the accompanying mutations in other genes. In the current study, we sought to examine the propensity for thrombosis, as well as platelet activation properties in a mouse model of primary myelofibrosis induced by JAK2V617F (janus kinase 2 with valine to phenylalanine substitution on codon 617) mutation. Approach and Results: Vav1-hJAK2V617F transgenic mice show hallmarks of primary myelofibrosis, including significant megakaryocytosis and bone marrow fibrosis, with a moderate increase in red blood cells and platelet number. This mouse model was used to study responses to 2 models of vascular injury and to investigate platelet properties. Platelets derived from the mutated mice have reduced aggregation in response to collagen, reduced thrombus formation and thrombus size, as demonstrated using laser-induced or FeCl3-induced vascular injury models, and increased bleeding time. Strikingly, the mutated platelets had a significantly reduced number of dense granules, which could explain impaired ADP secretion upon platelet activation, and a diminished second wave of activation. CONCLUSIONS: Together, our study highlights for the first time the influence of a hyperactive JAK2 on platelet activation-induced ADP secretion and dense granule homeostasis, with consequent effects on platelet activation properties.


Asunto(s)
Coagulación Sanguínea , Plaquetas/enzimología , Traumatismos de las Arterias Carótidas/enzimología , Janus Quinasa 2/sangre , Megacariocitos/enzimología , Activación Plaquetaria , Mielofibrosis Primaria/enzimología , Trombosis/enzimología , Animales , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/genética , Modelos Animales de Enfermedad , Janus Quinasa 2/genética , Ratones Transgénicos , Mutación , Agregación Plaquetaria , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/genética , Trombopoyesis , Trombosis/sangre , Trombosis/genética
6.
Cancer ; 126(19): 4322-4331, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32697338

RESUMEN

BACKGROUND: Circulating blasts (peripheral blood [PB] blasts) ≥1% have long been considered an unfavorable feature for patients with primary myelofibrosis. Whether further quantification of PB blasts and their correlation with bone marrow (BM) blasts have incremental value with regard to patient prognostication is unclear. Similarly, the role of the JAK1/JAK2 inhibitor ruxolitinib (RUX) is not well defined in patients who have increased blasts. METHODS: The authors retrospectively studied 1316 patients with myelofibrosis who presented at their institution between 1984 and 2018 and had available PB and BM blasts. RESULTS: The PB blast percentage influenced overall survival (OS) only among patients who had BM blasts <5%, with a median OS of 64 months for patients with 0% PB blasts, 48 months for those with 1% to 3% PB blasts, and 22 months for those with 4% PB blasts (P < .01). Patients who had 4% PB blasts and 5% to 9% BM/PB blasts had clinical features similar to those of patients who had 10% to 19% blasts. Although the OS of the former patients was longer than in patients who had 10% to 19% blasts, it was not statistically different (median OS: 22, 26, and 13 months, respectively; P > .05). Forty-four percent of patients received RUX throughout their disease course. All patients who had <10% blasts (PB or BM) and received treatment with RUX had superior OS compared with those who did not receive RUX within the same group. PB blasts ≥4% and BM blasts ≥5% were significant for predicting inferior survival in multivariate analysis. CONCLUSIONS: The current results provide comprehensive insight into the role of peripheral blasts in patients with myelofibrosis and indicates that patients who have PB blasts ≥4% have an unfavorable prognosis. RUX provides a survival benefit to patients who have PB blasts <10%.


Asunto(s)
Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nitrilos , Mielofibrosis Primaria/mortalidad , Pronóstico , Pirazoles/farmacología , Pirimidinas , Estudios Retrospectivos , Análisis de Supervivencia , Adulto Joven
7.
Cancer ; 126(6): 1243-1252, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31860137

RESUMEN

BACKGROUND: After discontinuing ruxolitinib, the outcome of patients with myelofibrosis reportedly has been poor. The authors investigated whether disease characteristics before the receipt of ruxolitinib may predict drug discontinuation in patients with myelofibrosis and whether reasons for drug discontinuation, disease phase at discontinuation, and salvage therapies may influence the outcome. METHODS: A centralized electronic clinical database was created in 20 European hematology centers, including clinical and laboratory data for 524 patients who received ruxolitinib for myelofibrosis. RESULTS: At 3 years, 40.8% of patients had stopped ruxolitinib. Baseline predictors of drug discontinuation were: intermediate-2-risk/high-risk category (Dynamic International Prognostic Score System), a platelet count <100 ×109 per liter, transfusion dependency, and unfavorable karyotype. At last contact, 268 patients (51.1%) had discontinued therapy, and the median drug exposure was 17.5 months. Fifty patients (18.7%) died while taking ruxolitinib. The reasons for discontinuation in the remaining 218 patients were the lack (22.9%) or loss (11.9%) of a spleen response, ruxolitinib-related adverse events (27.5%), progression to blast phase (23.4%), ruxolitinib-unrelated adverse events (9.2%), and allogeneic transplantation during response (5.1%). The median survival after ruxolitinib was 13.2 months and was significantly better in the 167 patients who discontinued ruxolitinib in chronic phase (27.5 vs 3.9 months for those who discontinued in blast phase; P < .001). No survival differences were observed among patients who discontinued ruxolitinib in chronic phase because of lack of response, loss of response, or ruxolitinib-related adverse events. The use of investigational agents and/or ruxolitinib rechallenge were associated with improved outcome. CONCLUSIONS: The survival of patients with myelofibrosis after discontinuation of ruxolitinib is poor, particularly for those who discontinue in blast phase. Salvage therapies can improve outcome, emphasizing the need for novel therapies.


Asunto(s)
Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/uso terapéutico , Privación de Tratamiento/estadística & datos numéricos , Adulto , Anciano , Anciano de 80 o más Años , Crisis Blástica , Progresión de la Enfermedad , Transfusión de Eritrocitos , Europa (Continente) , Femenino , Humanos , Cariotipo , Masculino , Persona de Mediana Edad , Nitrilos , Recuento de Plaquetas , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/patología , Pirazoles/efectos adversos , Pirimidinas , Estudios Retrospectivos , Terapia Recuperativa , Bazo/efectos de los fármacos , Esplenomegalia/tratamiento farmacológico , Estadísticas no Paramétricas , Análisis de Supervivencia , Trasplante Homólogo/estadística & datos numéricos , Resultado del Tratamiento , Adulto Joven
8.
Br J Haematol ; 189(5): 888-903, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32017044

RESUMEN

Ruxolitinib is a potent Janus kinase (JAK) 1/JAK2 inhibitor approved for the treatment of myelofibrosis (MF). Ruxolitinib was assessed in JUMP, a large (N = 2233), phase 3b, expanded-access study in MF in countries without access to ruxolitinib outside a clinical trial, which included patients with low platelet counts (<100 × 109 /l) and patients without splenomegaly - populations that have not been extensively studied. The most common adverse events (AEs) were anaemia and thrombocytopenia, but they rarely led to discontinuation (overall, 5·4%; low-platelet cohort, 12·3%). As expected, rates of worsening thrombocytopenia were higher in the low-platelet cohort (all grades, 73·2% vs. 53·5% overall); rates of anaemia were similar (all grades, 52·9% vs. 59·5%). Non-haematologic AEs, including infections, were mainly grade 1/2. Overall, ruxolitinib led to meaningful reductions in spleen length and symptoms, including in patients with low platelet counts, and symptom improvements in patients without splenomegaly. In this trial, the largest study of ruxolitinib in patients with MF to date, the safety profile was consistent with previous reports, with no new safety concerns identified. This study confirms findings from the COMFORT studies and supports the use of ruxolitinib in patients with platelet counts of 50-100 × 109 /l. (ClinicalTrials.gov identifier NCT01493414).


Asunto(s)
Mielofibrosis Primaria/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia/inducido químicamente , Femenino , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/etiología , Masculino , Persona de Mediana Edad , Neoplasias/etiología , Nitrilos , Recuento de Plaquetas , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/complicaciones , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Inhibidores de Proteínas Quinasas/efectos adversos , Pirazoles/efectos adversos , Pirimidinas , Bazo/patología , Esplenomegalia/etiología , Trombocitopenia/inducido químicamente , Adulto Joven
9.
Int J Mol Sci ; 21(9)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403407

RESUMEN

Growing evidence highlights the endocannabinoid (EC) system involvement in cancer progression. Lipid mediators of this system are secreted by hematopoietic cells, including the ECs 2-arachidonoyl-glycerol (2AG) and arachidonoyl-ethanolamide (AEA), the 2AG metabolite 1AG, and members of N-acylethanolamine (NAE) family-palmitoyl-ethanolamide (PEA) and oleoyl-ethanolamide (OEA). However, the relevance of the EC system in myeloproliferative neoplasms (MPN) was never investigated. We explored the EC plasma profile in 55 MPN patients, including myelofibrosis (MF; n = 41), polycythemia vera (PV; n = 9), and essential thrombocythemia (ET; n = 5) subclasses and in 10 healthy controls (HC). AEA, PEA, OEA, 2AG, and 1AG plasma levels were measured by LC-MS/MS. Overall considered, MPN patients displayed similar EC and NAE levels compared to HC. Nonetheless, AEA levels in MPN were directly associated with the platelet count. MF patients showed higher levels of the sum of 2AG and 1AG compared to ET and PV patients, higher OEA/AEA ratios compared to HC and ET patients, and higher OEA/PEA ratios compared to HC. Furthermore, the sum of 2AG and 1AG positively correlated with JAK2V617F variant allele frequency and splenomegaly in MF and was elevated in high-risk PV patients compared to in low-risk PV patients. In conclusion, our work revealed specific alterations of ECs and NAE plasma profile in MPN subclasses and potentially relevant associations with disease severity.


Asunto(s)
Endocannabinoides/sangre , Etanolaminas/sangre , Trastornos Mieloproliferativos/sangre , Policitemia Vera/sangre , Mielofibrosis Primaria/sangre , Trombocitemia Esencial/sangre , Adulto , Anciano , Anciano de 80 o más Años , Amidas/sangre , Ácidos Araquidónicos/sangre , Cromatografía Liquida/métodos , Femenino , Glicéridos/sangre , Humanos , Janus Quinasa 2/genética , Masculino , Persona de Mediana Edad , Mutación Missense , Trastornos Mieloproliferativos/diagnóstico , Trastornos Mieloproliferativos/genética , Ácidos Oléicos/sangre , Ácidos Palmíticos/sangre , Policitemia Vera/diagnóstico , Policitemia Vera/genética , Alcamidas Poliinsaturadas/sangre , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/genética , Espectrometría de Masas en Tándem/métodos , Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/genética
10.
Biol Blood Marrow Transplant ; 25(8): 1536-1543, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30826464

RESUMEN

The choice of alternative donor is a major issue in allogeneic hematopoietic stem cell transplantation (HSCT) for patients with primary myelofibrosis (PMF) without an HLA-matched related donor. We conducted this retrospective study using the Japanese national registry data for 224 PMF patients to compare the outcomes of first allogeneic HSCT from HLA-matched related donor bone marrow (Rtd-BM), HLA-matched related donor peripheral blood stem cells (Rtd-PB), HLA-matched unrelated donor bone marrow (UR-BM), unrelated umbilical cord blood (UR-UCB), and other hematopoietic stem cell grafts. Nonrelapse mortality (NRM) rates at 1 year after Rtd-BM, Rtd-PB, UR-BM, UR-UCB, and other transplantations were 16%, 36%, 30%, 41%, and 48%, respectively. Multivariate analysis identified UR-UCB transplantation, other transplantation, frequent RBC transfusion before transplantation, and frequent platelet (PLT) transfusion before transplantation as predictive of higher NRM. Relapse rates at 1 year after Rtd-BM, Rtd-PB, UR-BM, UR-UCB, and other transplantation were 14%, 17%, 11%, 14%, and 15%, respectively. No specific factor was associated with the incidence of relapse. Overall survival (OS) at 1 and 4 years after Rtd-BM, Rtd-PB, UR-BM, UR-UCB, and other transplantation were 81% and 71%, 58% and 52%, 61% and 46%, 48% and 27%, and 48% and 41%, respectively. Multivariate analysis identified older patient age, frequent RBC transfusion before transplantation, and frequent PLT transfusion before transplantation as predictive of lower OS. In conclusion, UR-UCB transplantation, as well as UR-BM transplantation, can be selected for PMF patients without an HLA-identical related donor. However, careful management is required for patients after UR-UCB transplantation because of the high NRM. Further studies including more patients after HLA-haploidentical related donor and HLA-mismatched unrelated donor transplantation would provide more valuable information for patients with PMF when making decisions regarding the choice of alternative donor.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Células Madre Hematopoyéticas , Trasplante de Células Madre de Sangre Periférica , Mielofibrosis Primaria , Donante no Emparentado , Adulto , Anciano , Aloinjertos , Supervivencia sin Enfermedad , Transfusión de Eritrocitos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/terapia , Sistema de Registros , Estudios Retrospectivos , Tasa de Supervivencia
11.
Haematologica ; 104(4): 659-668, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30872371

RESUMEN

Advances in understanding the pathogenesis and molecular landscape of myelofibrosis have occurred over the last decade. Treating physicians now have access to an ever-evolving armamentarium of novel agents to treat patients, although allogeneic hematopoietic stem cell transplantation remains the only curative approach. Improvements in donor selection, conditioning regimens, disease monitoring and supportive care have led to augmented survival after transplantation. Nowadays, there are comprehensive guidelines concerning allogeneic hematopoietic stem cell transplantation for patients with myelofibrosis. However, it commonly remains difficult for both physicians and patients alike to weigh up the risk-benefit ratio of transplantation given the inherent heterogeneity regarding both clinical course and therapeutic response. In this timely review, we provide an up-to-date synopsis of current transplantation recommendations, discuss usage of JAK inhibitors before and after transplantation, examine donor selection and compare conditioning platforms. Moreover, we discuss emerging data concerning the impact of the myelofibrosis mutational landscape on transplantation outcome, peritransplant management of splenomegaly, poor graft function and prevention/management of relapse.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mielofibrosis Primaria , Acondicionamiento Pretrasplante , Supervivencia sin Enfermedad , Humanos , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/terapia , Recurrencia , Tasa de Supervivencia , Trasplante Homólogo
13.
Ann Hematol ; 98(4): 869-879, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30515541

RESUMEN

Current prognostication in myelofibrosis (MF) is based on clinicopathological features and mutations in a limited number of driver genes. The impact of other genetic mutations remains unclear. We evaluated for mutations in a myeloid panel of 54 genes using next-generation sequencing. Multivariate Cox regression analysis was used to determine prognostic factors for overall survival (OS) and leukaemia-free survival (LFS), based on mutations of these genes and relevant clinical and haematological features. One hundred and one patients (primary MF, N = 70; secondary MF, N = 31) with a median follow-up of 49 (1-256) months were studied. For the entire cohort, inferior OS was associated with male gender (P = 0.04), age > 65 years (P = 0.04), haemoglobin < 10 g/dL (P = 0.001), CUX1 mutation (P = 0.003) and TP53 mutation (P = 0.049); and inferior LFS was associated with male gender (P = 0.03), haemoglobin < 10 g/dL (P = 0.04) and SRSF2 mutations (P = 0.008). In primary MF, inferior OS was associated with male gender (P = 0.03), haemoglobin < 10 g/dL (P = 0.002), platelet count < 100 × 109/L (P = 0.02), TET2 mutation (P = 0.01) and CUX1 mutation (P = 0.01); and inferior LFS was associated with haemoglobin < 10 g/dL (P = 0.02), platelet count < 100 × 109/L (P = 0.02), TET2 mutations (P = 0.01) and CUX1 mutations (P = 0.04). These results showed that clinical and haematological features and genetic mutations should be considered in MF prognostication.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Mutación , Mielofibrosis Primaria , Adulto , Anciano , Anciano de 80 o más Años , Pueblo Asiatico , China/epidemiología , Análisis Mutacional de ADN , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Recuento de Plaquetas , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/mortalidad , Factores Sexuales , Tasa de Supervivencia
15.
Acta Haematol ; 141(3): 158-163, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30799408

RESUMEN

TAFRO syndrome, a rare systemic inflammatory disease, can lead to multiorgan failure without appropriate treatment. Although thrombocytopenia is frequently seen in patients with TAFRO syndrome, little is known about its pathogenesis. Moreover, while recent studies have reported the presence of an anterior mediastinal mass in some patients, the pathological status of this remains unclear. Here, we report a case of fatal bleeding in a patient with TAFRO syndrome accompanied by an anterior mediastinal mass. A 55-year-old female was transferred to our hospital with a 2-week history of fever, epistaxis, and dyspnea. Laboratory tests revealed severe thrombocytopenia, computed tomography (CT) showed pleural effusions, and bone marrow biopsy revealed reticulin myelofibrosis. We suspected TAFRO syndrome, but the CT scan showed an anterior mediastinal mass that required a biopsy to exclude malignancy. She soon developed severe hemorrhagic diathesis and died of intracranial hemorrhage despite intensive treatment. She had multiple autoantibodies against platelets, which caused platelet destruction. An autopsy of the mediastinal mass revealed fibrous thymus tissues with infiltration by plasma cells. Our case suggests that thrombocytopenia could be attributed to antibody-mediated destruction and could be lethal. Hence, immediate treatment is imperative in cases of severe thrombocytopenia, even when accompanied by an anterior mediastinal mass.


Asunto(s)
Autoanticuerpos , Enfermedad de Castleman , Enfermedades del Mediastino , Púrpura Trombocitopénica Idiopática , Tomografía Computarizada por Rayos X , Autopsia , Enfermedad de Castleman/sangre , Enfermedad de Castleman/diagnóstico por imagen , Enfermedad de Castleman/patología , Enfermedad de Castleman/terapia , Resultado Fatal , Femenino , Humanos , Hemorragias Intracraneales/sangre , Hemorragias Intracraneales/diagnóstico por imagen , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/terapia , Enfermedades del Mediastino/sangre , Enfermedades del Mediastino/diagnóstico por imagen , Enfermedades del Mediastino/patología , Enfermedades del Mediastino/terapia , Persona de Mediana Edad , Derrame Pleural/sangre , Derrame Pleural/diagnóstico , Derrame Pleural/patología , Derrame Pleural/terapia , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/diagnóstico por imagen , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/terapia , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/diagnóstico por imagen , Púrpura Trombocitopénica Idiopática/patología , Púrpura Trombocitopénica Idiopática/terapia
16.
J Infect Chemother ; 25(1): 50-53, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30057341

RESUMEN

Mucormycosis generally develops under immunocompromised conditions, including hematological malignancies and solid organ or hematopoietic stem cell transplantation. Although mucormycosis usually affects the lungs and paranasal sinuses, sporadic cases of invasive mucormycosis of the liver have been reported. We hereby report a patient with myelofibrosis who developed hepatic mucormycosis diagnosed by post-mortem examination. An extensive literature review identified 13 reported cases of hepatic mucormycosis, including ours, without lung involvement. Most of the underlying diseases or conditions were hematological malignancies and solid organ transplantation. Three cases had splenic lesions and four had gastrointestinal lesions, suggesting the possibility of translocation to the liver and/or spleen from the gastrointestinal tracts. Hepatic mucormycosis should be recognized as one of the presentations of invasive mucormycosis, especially when hepatic nodules are found in immunocompromised patients such as those with hematological malignancy or recipients of solid organ transplantation.


Asunto(s)
Infecciones Fúngicas Invasoras/complicaciones , Hepatopatías/microbiología , Mucormicosis/complicaciones , Anciano , Anfotericina B/administración & dosificación , Anfotericina B/uso terapéutico , Antifúngicos/administración & dosificación , Antifúngicos/uso terapéutico , Autopsia , Resultado Fatal , Ferritinas/sangre , Galactosa/análogos & derivados , Humanos , Infecciones Fúngicas Invasoras/sangre , Infecciones Fúngicas Invasoras/tratamiento farmacológico , Hepatopatías/diagnóstico , Hepatopatías/tratamiento farmacológico , Masculino , Mananos/sangre , Mucormicosis/sangre , Mucormicosis/tratamiento farmacológico , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/tratamiento farmacológico , Bazo/patología
17.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31801274

RESUMEN

Acid sphingomyelinase (aSMase) is involved in the generation of metabolites that function as part of the sphingolipid signaling pathway. It catalyzes the breakdown of sphingomyelin into ceramide, a bioactive lipid that, among other roles, is involved in regulation of apoptosis. Dry drop blood test (DBS) and colorimetric 2-step enzymatic assay were used to assess the activity of human blood aSMase, beta-galactosidase, and beta-glucosidase, these enzymes are lysosomal hydrolases that catalyze the degradation of related sphingolipids, of sphingolipid signaling molecules. Blood was collected from a group of healthy volunteers and patients that were diagnosed with multiple myeloma (MM) in various stages of the disease. Additionally, activity of those enzymes in patients diagnosed with other hematological cancers was also assessed. We found that aSMase activity in the blood of patients with MM (at the time of diagnosis) was 305.43 pmol/spot*20 h, and this value was significantly lower (p < 0.030) compared to the healthy group 441.88 pmol/spot*20 h. Our collected data suggest a possible role of aSMase in pathogenesis of MM development.


Asunto(s)
Mieloma Múltiple/sangre , Esfingolípidos/sangre , Esfingomielina Fosfodiesterasa/sangre , beta-Galactosidasa/sangre , beta-Glucosidasa/sangre , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Humanos , Leucemia de Células Pilosas/sangre , Leucemia de Células Pilosas/diagnóstico , Leucemia de Células Pilosas/patología , Leucemia Linfocítica Crónica de Células B/sangre , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/patología , Metabolismo de los Lípidos , Linfoma de Células B de la Zona Marginal/sangre , Linfoma de Células B de la Zona Marginal/diagnóstico , Linfoma de Células B de la Zona Marginal/patología , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Estadificación de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/patología
18.
Mod Pathol ; 31(5): 690-704, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29327708

RESUMEN

Myeloproliferative neoplasms arise from hematopoietic stem cells with somatically altered tyrosine kinase signaling. Classification of myeloproliferative neoplasms is based on hematologic, histopathologic and molecular characteristics including the presence of the BCR-ABL1 and JAK2 V617F. Although thought to be mutually exclusive, a number of cases with co-occurring BCR-ABL1 and JAK2 V617F have been identified. To characterize the clinicopathologic features of myeloproliferative neoplasms with concomitant BCR-ABL1 and JAK2 V617F, and define the frequency of co-occurrence, we conducted a retrospective multi-institutional study. Cases were identified using a search of electronic databases over a decade at six major institutions. Of 1570 patients who were tested for both BCR-ABL1 and JAK2 V617F, six were positive for both. An additional five patients were identified via clinical records providing a total of 11 cases for detailed evaluation. For each case, clinical variables, hematologic and genetic data, and bone marrow histomorphologic features were analyzed. The sequence of identification of the genetic abnormalities varied: five patients were initially diagnosed with a JAK2 V617F+ myeloproliferative neoplasm, one patient initially had BCR-ABL1+ chronic myeloid leukemia, while both alterations were identified simultaneously in five patients. Classification of the BCR-ABL1-negative myeloproliferative neoplasms varied, and in some cases, features only became apparent following tyrosine kinase inhibitor therapy. Seven of the 11 patients showed myelofibrosis, in some cases before identification of the second genetic alteration. Our data, reflecting the largest reported study comprehensively detailing clinicopathologic features and response to therapy, show that the co-occurrence of BCR-ABL1 and JAK2 V617F is rare, with an estimated frequency of 0.4%, and most often reflects two distinct ('composite') myeloproliferative neoplasms. Although uncommon, it is important to be aware of this potentially confounding genetic combination, lest these features be misinterpreted to reflect resistance to therapy or disease progression, considerations that could lead to inappropriate management.


Asunto(s)
Neoplasias de la Médula Ósea/genética , Médula Ósea/patología , Proteínas de Fusión bcr-abl/genética , Janus Quinasa 2/genética , Sistemas Multiinstitucionales , Trastornos Mieloproliferativos/genética , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Neoplasias de la Médula Ósea/sangre , Neoplasias de la Médula Ósea/tratamiento farmacológico , Neoplasias de la Médula Ósea/patología , Progresión de la Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Femenino , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/sangre , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Persona de Mediana Edad , Mutación , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/patología , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Estudios Retrospectivos
19.
Transfusion ; 58(12): 2773-2776, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30265752

RESUMEN

BACKGROUND: Donor-specific human leukocyte antigen (HLA) antibodies are a significant risk factor for graft failure in cord blood transplantation (CBT). Although there are several treatments to decrease HLA antibodies, such as platelet transfusion, plasma exchange, rituximab, and bortezomib, their effectiveness has not been established. STUDY DESIGN AND METHODS: We herein report the case of a primary myelofibrosis (PMF) patient with broad HLA antibodies who underwent CBT from an HLA-homozygous donor in which the alleles were matched only in the host-versus-graft direction (homo-to-hetero CBT). RESULTS: The cord blood was killer cell immunoglobulin-like receptor (KIR) ligand matched. She received a reduced-intensity conditioning regimen. We used tacrolimus and mycophenolate mofetil as prophylaxis against graft-versus-host disease (GVHD). The neutrophils engrafted on Day 31. A chimerism analysis with fluorescence in situ hybridization of peripheral blood cells showed 99.9% donor type on Day 33. She developed only mild acute skin GVHD and chronic skin GVHD. CONCLUSION: This case indicates the usefulness of homo-to-hetero CBT in a patient with broad HLA antibodies with a strong mean fluorescence intensity, which is a significant risk factor for graft failure. Further studies are necessary to determine the risk of GVHD and to elucidate the association between KIR ligand incompatibility and graft failure in homo-to-hetero CBT.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Supervivencia de Injerto , Prueba de Histocompatibilidad , Isoanticuerpos , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/terapia , Adulto , Aloinjertos , Femenino , Humanos
20.
Clin Lab ; 64(9): 1373-1383, 2018 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-30274004

RESUMEN

BACKGROUND: Primary myelofibrosis (PMF) is a chronic clonal myeloid disorder. Together with essential thrombocythemia (ET) and polycythemia vera (PV), it belongs to a group of Philadelphia chromosome-negative myeloproliferative neoplasms. Thrombotic events are serious complications negatively influencing the quality and length of these patients' lives. The confirmed risk factors for venous thromboembolism are age over 60 years, a positive history of thromboembolism, presence of common cardiovascular risks, JAK2 V617F mutation and, according to some authors, leukocytosis. Various opinions on the role of thrombocythemia have been published. The present study was undertaken to evaluate the benefit of thrombin generation test and its potential use in predicting the risk of thrombosis in MF patients. METHODS: The analysis included plasma samples obtained from 36 patients diagnosed with MF in our center from 2004 to 2016 (JAK2 V617-positive 53%; CALR-positive 31%; MPL-positive 14%; triple negative 2%) and a control group comprising 20 healthy volunteer blood donors. Thrombin generation was measured in platelet-rich plasma using the TECHNOTHROMBIN® TGA kit (Technoclone, Austria) and the fully automated system Ceveron® Alpha (Technoclone). The results were correlated with clinical and laboratory parameters of the patients. RESULTS: There were differences in thrombin generation as expressed by endogenous thrombin potential (ETP) between patients and healthy controls, with ETP being lower in the patient group (p = 0.0003). Analysis confirmed a significant correlation between thrombin generation and platelet counts, with higher thrombin generation in patients with thrombocythemia > 400 x 109/L (p = 0.04). ETP values were consistently higher in earlier disease stages and lower in CALR-mutated myelofibrosis. CONCLUSIONS: In MF patients, thrombin generation is mainly influenced by platelet counts and, to a lesser extent, by mutation status, activity, and progression of the disease. Thrombin generation test results have confirmed that thrombocythemia is a potential risk factor for thrombotic complications.


Asunto(s)
Pruebas de Coagulación Sanguínea , Coagulación Sanguínea , Mielofibrosis Primaria/diagnóstico , Trombina/metabolismo , Trombosis/diagnóstico , Adulto , Anciano , Biomarcadores/sangre , Calreticulina/genética , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Humanos , Janus Quinasa 2/genética , Masculino , Persona de Mediana Edad , Mutación , Fenotipo , Recuento de Plaquetas , Valor Predictivo de las Pruebas , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/genética , Pronóstico , Receptores de Trombopoyetina/genética , Medición de Riesgo , Factores de Riesgo , Trombosis/sangre , Trombosis/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA