Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 272
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 24(19): 11100-11110, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32889753

RESUMEN

Primary myelofibrosis (PMF) is a Ph-negative myeloproliferative neoplasm (MPN), characterized by advanced bone marrow fibrosis and extramedullary haematopoiesis. The bone marrow fibrosis results from excessive proliferation of fibroblasts that are influenced by several cytokines in the microenvironment, of which transforming growth factor-ß (TGF-ß) is the most important. Micromechanics related to the niche has not yet been elucidated. In this study, we hypothesized that mechanical stress modulates TGF-ß signalling leading to further activation and subsequent proliferation and invasion of bone marrow fibroblasts, thus showing the important role of micromechanics in the development and progression of PMF, both in the bone marrow and in extramedullary sites. Using three PMF-derived fibroblast cell lines and transforming growth factor-ß receptor (TGFBR) 1 and 2 knock-down PMF-derived fibroblasts, we showed that mechanical stress does stimulate the collagen synthesis by the fibroblasts in patients with myelofibrosis, through the TGFBR1, which however seems to be activated through alternative pathways, other than TGFBR2.


Asunto(s)
Progresión de la Enfermedad , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Fenómenos Biomecánicos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/diagnóstico por imagen , Ratones Desnudos , Modelos Biológicos , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/patología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Estrés Mecánico
2.
Ann Hematol ; 99(12): 2779-2785, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32862283

RESUMEN

We retrospectively investigated a cohort of 176 myelofibrosis patients (128 primary-PMF; 48 secondary-SMF) from five hematology centers. The presence of chronic kidney disease (CKD) was determined in addition to other clinical characteristics. CKD was present in 26.1% of MF patients and was significantly associated with older age (P < 0.001), higher WBC (P = 0.015), and its subsets (neutrophil, monocyte, and basophil counts), higher platelets (P = 0.001), lower albumin (P = 0.018), higher serum uric acid (P = 0.001), higher LDH (P = 0.022), and the presence of CV risk factors (P = 0.011). There was no significant association with driver mutations, degree of bone marrow fibrosis, PMF/SMF, or DIPSS risk categories (P > 0.05 for all analyses). The presence of CKD was significantly associated with shorter time to arterial (HR = 3.49; P = 0.041) and venous thrombosis (HR = 7.08; P = 0.030) as well as with shorter overall survival (HR 2.08; P = 0.009). In multivariate analyses, CKD (HR = 1.8; P = 0.014) was associated with shorter survival independently of the DIPSS (HR = 2.7; P < 0.001); its effect being more pronounced in lower (HR = 3.56; P = 0.036) than higher DIPSS categories (HR = 2.07; P = 0.023). MF patients with CKD should be candidates for active management aimed at the improvement of renal function. Prospective studies defining the optimal therapeutic approach are highly needed.


Asunto(s)
Riñón/fisiología , Mielofibrosis Primaria/mortalidad , Insuficiencia Renal Crónica/mortalidad , Trombosis/mortalidad , Anciano , Estudios de Cohortes , Femenino , Tasa de Filtración Glomerular/fisiología , Humanos , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/fisiopatología , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/fisiopatología , Estudios Retrospectivos , Tasa de Supervivencia/tendencias , Trombosis/diagnóstico , Trombosis/fisiopatología
3.
Blood ; 129(6): 667-679, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28028029

RESUMEN

The genetic landscape of classical myeloproliferative neoplasm (MPN) is in large part elucidated. The MPN-restricted driver mutations, including those in JAK2, calreticulin (CALR), and myeloproliferative leukemia virus (MPL), abnormally activate the cytokine receptor/JAK2 pathway and their downstream effectors, more particularly the STATs. The most frequent mutation, JAK2V617F, activates the 3 main myeloid cytokine receptors (erythropoietin receptor, granulocyte colony-stimulating factor receptor, and MPL) whereas CALR or MPL mutants are restricted to MPL activation. This explains why JAK2V617F is associated with polycythemia vera, essential thrombocythemia (ET), and primary myelofibrosis (PMF) whereas CALR and MPL mutants are found in ET and PMF. Other mutations in genes involved in epigenetic regulation, splicing, and signaling cooperate with the 3 MPN drivers and play a key role in the PMF pathogenesis. Mutations in epigenetic regulators TET2 and DNMT3A are involved in disease initiation and may precede the acquisition of JAK2V617F. Other mutations in epigenetic regulators such as EZH2 and ASXL1 also play a role in disease initiation and disease progression. Mutations in the splicing machinery are predominantly found in PMF and are implicated in the development of anemia or pancytopenia. Both heterogeneity of classical MPNs and prognosis are determined by a specific genomic landscape, that is, type of MPN driver mutations, association with other mutations, and their order of acquisition. However, factors other than somatic mutations play an important role in disease initiation as well as disease progression such as germ line predisposition, inflammation, and aging. Delineation of these environmental factors will be important to better understand the precise pathogenesis of MPN.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Mutación , Policitemia Vera/genética , Mielofibrosis Primaria/genética , Trombocitemia Esencial/genética , Calreticulina/genética , Calreticulina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Policitemia Vera/metabolismo , Policitemia Vera/fisiopatología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/fisiopatología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Trombocitemia Esencial/metabolismo , Trombocitemia Esencial/fisiopatología
4.
Ann Hematol ; 98(7): 1611-1616, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31093708

RESUMEN

Recent evidence suggests that renal dysfunction may be a direct consequence of primary myelofibrosis (PMF). We performed a retrospective analysis of 100 patients with previously untreated PMF, receiving frontline treatment with single agent ruxolitinib, and compared them to 105 patients, receiving frontline treatment with a non-ruxolitinib-based therapy, matched by age, sex, DIPSS plus, and estimated glomerular filtration rate (eGFR). Use of ruxolitinib associated with a significantly higher rate of renal improvement (RI) > 10% (73% vs 50%, p = 0.01) confirmed on multivariate analysis (MVA) [odds ratio 3, 95% confidence interval (CI) 1.6-5.5, p < 0.001]. After a median follow-up of 41 months (range, 1-159 months), median failure-free survival (FFS) was 14 months (range, 1-117 months). Achievement of a RI > 10% maintained its independent association with prolonged FFS on MVA (hazard ratio 1.4, 95% CI 1.1-2, p = 0.02). Ruxolitinib can significantly improve renal function in patients with PMF, significantly impacting failure-free survival.


Asunto(s)
Bases de Datos Factuales , Tasa de Filtración Glomerular/efectos de los fármacos , Riñón , Mielofibrosis Primaria , Pirazoles/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Humanos , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Persona de Mediana Edad , Nitrilos , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/fisiopatología , Pirimidinas , Estudios Retrospectivos , Tasa de Supervivencia
5.
Health Qual Life Outcomes ; 17(1): 61, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975150

RESUMEN

BACKGROUND: The goal of the research reported here was to understand the patient experience of living with myelofibrosis (MF) and establish content validity of the Modified Myeloproliferative Neoplasm Symptom Assessment Diary (MPN-SD). METHODS: Qualitative interviews were performed in patients with MF, including both concept elicitation and cognitive debriefing. Patients with MF were asked to spontaneously report on their signs, symptoms, and impacts of MF, as well as their understanding of the MPN-SD content, and use of the tool on an electronic platform. A supplementary literature review and meetings with MF experts were also performed. RESULTS: Twenty-three patients with MF participated in qualitative interviews. Signs and symptoms most commonly reported by ruxolitinib-experienced patients (n = 16) were: fatigue and/or tiredness (n = 16, 100%), shortness of breath (n = 11, 69%), pain below the ribs on the left side and/or stomach pain and/or abdominal pain (n = 9, 56%), and enlarged spleen (n = 9, 56%) and for ruxolitinib-naïve patients (n = 7) were: fatigue and/or tiredness (n = 6, 86%), pain below the ribs on the left side (n = 6, 86%), enlarged spleen (n = 4, 57%), full quickly/filling up quickly (n = 4, 57%), night sweats and/or general sweats (n = 4, 57%), and itching (n = 4, 57%). Patients demonstrated that they were able to read, understand, and provide meaningful responses to the MPN-SD. The final version of the MPN-SD includes the 10 most commonly reported concepts from the MF patient interviews. CONCLUSIONS: The findings demonstrate the comprehensiveness of the MPN-SD in assessing MF symptoms in both ruxolitinib-experienced and ruxolitinib-naïve patients, while remaining easy for patients to understand and complete.


Asunto(s)
Mielofibrosis Primaria/psicología , Calidad de Vida , Evaluación de Síntomas/normas , Adulto , Anciano , Anciano de 80 o más Años , Fatiga/etiología , Femenino , Humanos , Entrevistas como Asunto , Masculino , Persona de Mediana Edad , Nitrilos , Mielofibrosis Primaria/fisiopatología , Pirazoles/uso terapéutico , Pirimidinas , Investigación Cualitativa , Índice de Severidad de la Enfermedad
6.
Expert Opin Emerg Drugs ; 23(1): 37-49, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29480036

RESUMEN

INTRODUCTION: Myelofibrosis (MF) is a Philadelphia chromosome-negative myeloproliferative neoplasm (MPN). It can be sub-categorized into primary myelofibrosis, post polycythemia vera myelofibrosis and post essential thrombocythemia myelofibrosis. MF is a life-threatening hematologic malignancy characterized by dysregulation of the Janus associated kinase (JAK)/signal transducer and activator of transcription (STAT) signaling network and a heightened inflammatory state. Areas covered: We cover the pathogenesis, clinical features, new prognostic models, current treatment of MF and discuss agents in development. We also cover market review and health care costs associated with some of these therapies. Expert opinion: There are many ongoing clinical trials evaluating novel therapeutic approaches, including selective JAK inhibitors, histone deacetylase/DNA methyltransferase inhibitors, PI3K-inhibitors, Hedgehog/mammalian target of rapamycin (MTOR) inhibitors, anti-fibrotic agents, immunomodulators, monoclonal antibodies and immune checkpoint inhibitors. Ruxolitinib, a potent oral JAK1/JAK2 inhibitor remains the only Food and Drug Administration (FDA)-approved medicinal therapy for the treatment of MF. Unmet needs include alleviation of limiting thrombocytopenia and anemia, halting disease progression to acute leukemia, and extending survival. The development of biomarker driven clinical trials of mechanism based novel therapeutics will usher in a new era of advances in the treatment of this chronic and progressive myeloid malignancy.


Asunto(s)
Diseño de Fármacos , Inhibidores de las Cinasas Janus/uso terapéutico , Mielofibrosis Primaria/tratamiento farmacológico , Biomarcadores/metabolismo , Progresión de la Enfermedad , Humanos , Inhibidores de las Cinasas Janus/farmacología , Nitrilos , Mielofibrosis Primaria/fisiopatología , Pirazoles/farmacología , Pirazoles/uso terapéutico , Pirimidinas
7.
Int J Mol Sci ; 19(3)2018 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-29562644

RESUMEN

Myelofibrosis (MF) is a clinical manifestation of chronic BCR-ABL1-negative chronic myeloproliferative neoplasms. Splenomegaly is one of the major clinical manifestations of MF and is directly linked to splenic extramedullary hematopoiesis (EMH). EMH is associated with abnormal trafficking patterns of clonal hematopoietic cells due to the dysregulated bone marrow (BM) microenvironment leading to progressive splenomegaly. Several recent data have emphasized the role of several cytokines for splenic EMH. Alteration of CXCL12/CXCR4 pathway could also lead to splenic EMH by migrated clonal hematopoietic cells from BM to the spleen. Moreover, low Gata1 expression was found to be significantly associated with the EMH. Several gene mutations were found to be associated with significant splenomegaly in MF. In recent data, JAK2V617F homozygous mutation was associated with a larger spleen size. In other data, CALR mutations in MF were signigicantly associated with longer larger splenomegaly-free survivals than others. In addition, MF patients with ≥1 mutations in AZXL1, EZH1 or IDH1/2 had significantly low spleen reduction response in ruxolitinib treatment. Developments of JAK inhibitors, such as ruxolitinib, pacritinib, momelotinib, and febratinib enabled the effective management in MF patients. Especially, significant spleen reduction responses of the drugs were demonstrated in several randomized clinical studies, although those could not eradicate allele burdens of MF.


Asunto(s)
Hematopoyesis Extramedular/fisiología , Quinasas Janus/genética , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Esplenomegalia/genética , Médula Ósea/patología , Movimiento Celular , Quimiocina CXCL12/metabolismo , Hematopoyesis Extramedular/efectos de los fármacos , Humanos , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Mutación , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/fisiopatología , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores CXCR4/metabolismo , Bazo/patología , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/fisiopatología
8.
Intern Med J ; 47(3): 262-268, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28260257

RESUMEN

Ruxolitinib is a dual janus kinase 1 (JAK1)/JAK2 inhibitor used to treat splenomegaly and symptoms associated with myelofibrosis (MF). Current therapeutic options for symptomatic MF include supportive care, myelosuppressive therapy (such as hydroxycarbamide) and janus kinase (JAK) inhibitors (in particular ruxolitinib). Allogeneic stem cell transplantation remains the only potentially curative treatment for MF, and younger transplant-eligible patients should still be considered for allogeneic stem cell transplantation; however, this is applicable only to a small proportion of patients. There is now increasing and extensive experience of the efficacy and safety of ruxolitinib in MF, both in clinical trials and in 'real-world' practice. The drug has been shown to be of benefit in intermediate-1 risk patients with symptomatic splenomegaly or other MF-related symptoms, and higher risk disease. Optimal use of the drug is required to maximise clinical benefit, requiring an understanding of the balance between dose-dependent responses and dose-limiting toxicities. There is also increasing experience in the use of ruxolitinib in the pre-transplantation setting. This paper aims to utilise several 'real-life' cases to illustrate several strategies that may help to optimise clinical practice.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/uso terapéutico , Esplenomegalia/tratamiento farmacológico , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nitrilos , Guías de Práctica Clínica como Asunto , Mielofibrosis Primaria/fisiopatología , Pirimidinas , Esplenomegalia/etiología , Trasplante Autólogo , Resultado del Tratamiento
9.
Medicina (Kaunas) ; 53(1): 34-39, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28237691

RESUMEN

BACKGROUND AND OBJECTIVE: Data from the literature indicate the relationship between the bone marrow microvessel density and the blood parameters of angiogenesis. The aim of this study was to evaluate selected parameters of angiogenesis (VEGF-A, sVEGFR-1, and sVEGFR-2) and their correlations with white blood cells, platelets, and red blood cells. MATERIALS AND METHODS: The study included 72 patients (mean age, 61.84 years) with myeloproliferative neoplasms (MPNs): essential thrombocythemia (ET) (n=46), polycythemia vera (PV) (n=19), and primary myelofibrosis (PMF) (n=7). Serum VEGF-A, sVEGFR-1, and sVEGFR-2 were determined using the ELISA assay. RESULTS: We observed a significantly higher level of VEGF-A and reduced concentrations of sVEGFR-1 and sVEGFR-2 in the whole group of patients with MPNs as compared to controls. Detailed analysis confirmed significantly higher level of VEGF-A and lower concentration of sVEGFR-2 in each subgroups of MPNs patients. However, sVEGFR-1 concentrations were significantly lower only in PV and ET patients. CONCLUSIONS: The study showed an increased level of VEGF-A, which may indicate the intensity of neoangiogenesis in the bone marrow. Decreased sVEGFR-1 and sVEGFR-2 in the blood of patients with MPNs may reflect consumption of these soluble receptors.


Asunto(s)
Neovascularización Patológica/sangre , Policitemia Vera/fisiopatología , Mielofibrosis Primaria/fisiopatología , Trombocitemia Esencial/fisiopatología , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre , Anciano , Recuento de Células Sanguíneas , Médula Ósea/irrigación sanguínea , Médula Ósea/patología , Análisis Mutacional de ADN , Femenino , Fibrinógeno/análisis , Proteínas de Fusión bcr-abl/genética , Humanos , Masculino , Persona de Mediana Edad , Policitemia Vera/diagnóstico , Policitemia Vera/genética , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/genética , Estadísticas no Paramétricas , Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/genética
10.
J Cell Physiol ; 231(2): 314-27, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26247172

RESUMEN

Endothelial progenitor cells (EPCs) are mobilized either from the bone marrow and/or the arterial to replace dysfunctional endothelial cells and rescue blood perfusion in ischemic tissues. In addition, they may contribute to the angiogenic switch, thereby sustaining tumour growth and metastatization. Understanding the molecular mechanisms utilized by vascular endothelial growth factor (VEGF) to stimulate EPCs might unveil novel targets to enhance their clinical outcome in regenerative medicine and to adverse tumour vascularisation. VEGF stimulates peripheral blood-derived EPCs to undergo repetitive Ca(2+) oscillations shaped by the interaction between inositol-1,4,5-trisphosphate (InsP3 )-dependent Ca(2+) release and store-operated Ca(2+) entry (SOCE). However, the Ca(2+) machinery underlying VEGF-induced Ca(2+) spikes changes in umbilical cord blood-derived EPCs, which require TRPC3-mediated Ca(2+) entry to trigger the interplay between InsP3 and SOCE. Surprisingly, VEGF fails to elicit pro-angiogenic Ca(2+) signals when EPCs derive from renal cellular carcinoma patients, thus questioning the suitability of VEGFR-2 as a target for anti-angiogenic treatments in these individuals. The lack of response to VEGF is likely due to the dramatic rearrangement of the Ca(2+) toolkit occurring in RCC-derived EPCs. Finally, primary myelofibrosis-derived EPCs display a further pattern of reorganization of the Ca(2+) machinery and proliferate independently of SOCE. Thus, the Ca(2+) machinery in human ECFCs is extremely plastic and may change depending on the physio-pathological background of the donor. As a consequence, the Ca(2+) toolkit could properly be used to enhance the regenerative outcome of cell-based therapy or adverse tumor vascularisation.


Asunto(s)
Señalización del Calcio , Células Progenitoras Endoteliales/fisiología , Animales , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/fisiopatología , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Progenitoras Endoteliales/citología , Sangre Fetal/citología , Humanos , Neoplasias Renales/patología , Neoplasias Renales/fisiopatología , Modelos Biológicos , Neovascularización Patológica , Neovascularización Fisiológica , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología
11.
Cancer ; 122(12): 1888-96, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27070130

RESUMEN

BACKGROUND: Patients with myeloproliferative neoplasms (MPNs) including polycythemia vera, essential thrombocythemia, and myelofibrosis, are faced with oppressive symptom profiles that compromise daily functioning and quality of life. Among these symptoms, sexuality-related symptoms have emerged as particularly prominent and largely unaddressed. In the current study, the authors evaluated how sexuality symptoms from MPN relate to other patient characteristics, disease features, treatments, and symptoms. METHODS: A total of 1971 patients with MPN (827 with essential thrombocythemia, 682 with polycythemia vera, 456 with myelofibrosis, and 6 classified as other) were prospectively evaluated and patient responses to the Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF) and the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC-QLQ C30) were collected, along with information regarding individual disease characteristics and laboratory data. Sexuality scores were compared with an age-matched, healthy control population. RESULTS: Overall, patients with MPN were found to have greater sexual dysfunction compared with the healthy population (MPN-SAF score of 3.6 vs 2.0; P<.001), with 64% of patients with MPN describing some degree of sexual dysfunction and 43% experiencing severe symptoms. The presence of sexual symptoms correlated closely with all domains of patient functionality (physical, social, cognitive, emotional, and role functioning) and were associated with a reduced quality of life. Sexual problems also were found to be associated with other MPN symptoms, particularly depression and nocturnal and microvascular-related symptoms. Sexual dysfunction was more severe in patients aged >65 years and in those with cytopenias and transfusion requirements, and those receiving certain therapies such as immunomodulators or steroids. CONCLUSIONS: The results of the current study identify the topic of sexuality as a prominent issue for the MPN population, and this area would appear to benefit from additional investigation and management. Cancer 2016;122:1888-96. © 2016 American Cancer Society.


Asunto(s)
Trastornos Mieloproliferativos/fisiopatología , Trastornos Mieloproliferativos/psicología , Disfunciones Sexuales Fisiológicas/etiología , Disfunciones Sexuales Psicológicas/etiología , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Policitemia Vera/fisiopatología , Policitemia Vera/psicología , Mielofibrosis Primaria/fisiopatología , Mielofibrosis Primaria/psicología , Calidad de Vida , Conducta Sexual , Sexualidad , Encuestas y Cuestionarios , Trombocitemia Esencial/fisiopatología , Trombocitemia Esencial/psicología
12.
Ter Arkh ; 88(1): 89-95, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-26978616

RESUMEN

Patients with myeloproliferative diseases (MPD) are noted to be at high risk for portal thromboses. This problem gives rise to disability if it is untimely treated or resistant to therapy. The paper gives the experience of the Outpatient Department of the Hematology Research Center, Ministry of Health of the Russian Federation, in using antithrombin III in MPD patients (3 patients with primary myelofibrosis, 3 with essential thrombocythemia) and acute and subacute portal vein thromboses resistant to therapy with direct anticoagulants. In all 5 cases, the use of antithrombin III in combination with low-molecular-weight heparin showed a positive clinical effect as rapid relief of pain syndrome and comparatively early (3-week to 1.5-2-month) recanalization of thrombosed vessels. Three clinical cases are described in detail.


Asunto(s)
Antitrombina III/administración & dosificación , Síndrome de Budd-Chiari , Heparina de Bajo-Peso-Molecular/administración & dosificación , Mielofibrosis Primaria , Trombocitemia Esencial , Adulto , Coagulación Sanguínea/efectos de los fármacos , Pruebas de Coagulación Sanguínea/métodos , Síndrome de Budd-Chiari/sangre , Síndrome de Budd-Chiari/diagnóstico , Síndrome de Budd-Chiari/etiología , Síndrome de Budd-Chiari/fisiopatología , Síndrome de Budd-Chiari/terapia , Monitoreo de Drogas/métodos , Femenino , Fibrinolíticos/administración & dosificación , Humanos , Sistema Porta/diagnóstico por imagen , Sistema Porta/fisiopatología , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/fisiopatología , Mielofibrosis Primaria/terapia , Trombocitemia Esencial/sangre , Trombocitemia Esencial/complicaciones , Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/fisiopatología , Trombocitemia Esencial/terapia , Resultado del Tratamiento , Ultrasonografía , Grado de Desobstrucción Vascular/efectos de los fármacos
13.
J Cell Mol Med ; 19(11): 2564-74, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26176817

RESUMEN

JAK2 inhibition therapy is used to treat patients suffering from myeloproliferative neoplasms (MPN). Conflicting data on this therapy are reported possibly linked to the types of inhibitors or disease type. Therefore, we decided to compare in mice the effect of a JAK2 inhibitor, Fedratinib, in MPN models of increasing severity: polycythemia vera (PV), post-PV myelofibrosis (PPMF) and rapid post-essential thrombocythemia MF (PTMF). The models were generated through JAK2 activation by the JAK2(V617F) mutation or MPL constant stimulation. JAK2 inhibition induced a correction of splenomegaly, leucocytosis and microcytosis in all three MPN models. However, the effects on fibrosis, osteosclerosis, granulocytosis, erythropoiesis or platelet counts varied according to the disease severity stage. Strikingly, complete blockade of fibrosis and osteosclerosis was observed in the PPMF model, linked to correction of MK hyper/dysplasia, but not in the PTMF model, suggesting that MF development may also become JAK2-independent. Interestingly, we originally found a decreased in the JAK2(V617F) allele burden in progenitor cells from the spleen but not in other cell types. Overall, this study shows that JAK2 inhibition has different effects according to disease phenotypes and can (i) normalize platelet counts, (ii) prevent the development of marrow fibrosis/osteosclerosis at an early stage and (iii) reduce splenomegaly through blockage of stem cell mobilization in the spleen.


Asunto(s)
Janus Quinasa 2/antagonistas & inhibidores , Policitemia Vera/tratamiento farmacológico , Mielofibrosis Primaria/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirrolidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Trombocitemia Esencial/tratamiento farmacológico , Animales , Progresión de la Enfermedad , Ratones , Recuento de Plaquetas , Policitemia Vera/sangre , Policitemia Vera/fisiopatología , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/fisiopatología , Esplenomegalia/tratamiento farmacológico , Trombocitemia Esencial/sangre , Trombocitemia Esencial/fisiopatología
14.
Oncologist ; 20(10): 1154-60, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26304912

RESUMEN

Primary myelofibrosis is a stem cell-derived clonal malignancy characterized by unchecked proliferation of myeloid cells, resulting in bone marrow fibrosis, osteosclerosis, and pathologic angiogenesis. Bone marrow fibrosis (BMF) plays a central role in the pathophysiology of the disease. This review describes current issues regarding BMF in primary myelofibrosis, including the pathophysiology and impact of abnormal deposition of excess collagen and reticulin fibers in bone marrow spaces, the modified Bauermeister and the European Consensus grading systems of BMF, and the prognostic impact of BMF on the overall outcome of patients with primary myelofibrosis. The impact of novel therapeutic strategies, including JAK-STAT inhibitors and allogeneic stem cell transplant, on BMF is discussed.


Asunto(s)
Células de la Médula Ósea/patología , Mielofibrosis Primaria , Busulfano/uso terapéutico , Fibrosis/diagnóstico , Fibrosis/patología , Fibrosis/terapia , Humanos , Quinasas Janus/antagonistas & inhibidores , Nitrilos , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/fisiopatología , Mielofibrosis Primaria/terapia , Pronóstico , Pirazoles/uso terapéutico , Pirimidinas , Factores de Transcripción STAT/antagonistas & inhibidores , Trasplante de Células Madre/métodos
15.
Expert Opin Emerg Drugs ; 20(4): 663-78, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26153237

RESUMEN

INTRODUCTION: Myelofibrosis (MF) is a myeloproliferative neoplasm associated with significant disease burden composed of splenomegaly, constitutional symptoms and a reduced life expectancy. The advent of targeted treatments has provided new means by which to improve MF associated splenomegaly, symptoms, health-related quality of life and even mortality. AREAS COVERED: We discuss the spectrum of targeted treatments currently under investigation for MF. We furthermore compare their effects on improving anemia, reducing fibrosis and splenomegaly and enhancing symptom control. EXPERT OPINION: MF is a complex disorder, partly attributable to its heterogeneity. Although the severity of patient symptoms correlates with risk category, high symptom burden may also be observed in low-risk patients. Serial use of PRO tools allows clinicians to objectively evaluate the MF symptom burden, compare efficacy of therapies and adjust medications to improve symptom control. Novel targeted agents have proven superior to historic treatment regimens for symptom management. Promising treatment categories include JAK2 inhibitors, histone deacetylase inhibitors, hypomethylating agents, heat shock protein-90 inhibitors, hedgehog inhibitors, PI3-AKT-mTOR inhibitors, antifibrosing agents and telomerase inhibitors. The majority of therapies remain under investigation, either alone or in combination with other treatments. It is anticipated that these agents will be increasingly integrated into standard treatment algorithms for MF symptom management.


Asunto(s)
Diseño de Fármacos , Mielofibrosis Primaria/tratamiento farmacológico , Calidad de Vida , Algoritmos , Animales , Humanos , Esperanza de Vida , Terapia Molecular Dirigida , Mielofibrosis Primaria/fisiopatología , Índice de Severidad de la Enfermedad , Esplenomegalia/etiología
16.
Value Health ; 18(6): 846-55, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26409613

RESUMEN

BACKGROUND: Utility values are required for economic evaluation using cost-utility analyses. Often, generic measures such as the EuroQol five-dimensional questionnaire are used, but this may not appropriately reflect the health-related quality of life of patients with cancer including myelofibrosis. OBJECTIVE: To derive a condition-specific preference-based measure for myelofibrosis using appropriate existing measures, the Myelofibrosis-Symptom Assessment Form and the European Organisation for Research and Treatment of Cancer Quality of Life 30 Questionnaire. METHODS: Data from the Controlled Myelofibrosis Study with Oral JAK Inhibitor Treatment trial (n = 309) were used to derive the health state classification system. Psychometric and factor analyses were used to determine the dimensions of the classification system. Psychometric and Rasch analyses were then used to select an item to represent each dimension. Item selection was validated with experts. A selection of health states was valued by members of the general population using time trade-off. Finally, health state values were modeled using regression analysis to produce utility values for every state. RESULTS: The Myelofibrosis 8 dimensions has eight dimensions: physical functioning, emotional functioning, fatigue, itchiness, pain under ribs on the left side, abdominal discomfort, bone or muscle pain, and night sweats. Regression models were estimated using time trade-off data from 246 members of the general population valuing a total of 33 states. The best performing model was a random effects maximum likelihood model producing utility values ranging from 0.089 to 1. CONCLUSIONS: The Myelofibrosis 8 dimensions is a condition-specific preference-based measure for myelofibrosis. This measure can be used to generate utility values for myelofibrosis for any data set containing the Myelofibrosis-Symptom Assessment Form and the European Organisation for Research and Treatment of Cancer Quality of Life 30 Questionnaire data.


Asunto(s)
Prioridad del Paciente , Mielofibrosis Primaria/tratamiento farmacológico , Proteínas Quinasas/uso terapéutico , Calidad de Vida , Encuestas y Cuestionarios , Adulto , Anciano , Anciano de 80 o más Años , Ensayos Clínicos Fase III como Asunto , Análisis Factorial , Femenino , Investigación sobre Servicios de Salud , Estado de Salud , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Estudios Multicéntricos como Asunto , Valor Predictivo de las Pruebas , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/fisiopatología , Mielofibrosis Primaria/psicología , Proteínas Quinasas/efectos adversos , Psicometría , Años de Vida Ajustados por Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto , Factores de Tiempo , Resultado del Tratamiento
17.
Mediators Inflamm ; 2015: 145293, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26538820

RESUMEN

Myeloproliferative neoplasms (MPNs) are a heterogeneous group of clonal diseases characterized by the excessive and chronic production of mature cells from one or several of the myeloid lineages. Recent advances in the biology of MPNs have greatly facilitated their molecular diagnosis since most patients present with mutation(s) in the JAK2, MPL, or CALR genes. Yet the roles played by these mutations in the pathogenesis and main complications of the different subtypes of MPNs are not fully elucidated. Importantly, chronic inflammation has long been associated with MPN disease and some of the symptoms and complications can be linked to inflammation. Moreover, the JAK inhibitor clinical trials showed that the reduction of symptoms linked to inflammation was beneficial to patients even in the absence of significant decrease in the JAK2-V617F mutant load. These observations suggested that part of the inflammation observed in patients with JAK2-mutated MPNs may not be the consequence of JAK2 mutation. The aim of this paper is to review the different aspects of inflammation in MPNs, the molecular mechanisms involved, the role of specific genetic defects, and the evidence that increased production of certain cytokines depends or not on MPN-associated mutations, and to discuss possible nongenetic causes of inflammation.


Asunto(s)
Inflamación/fisiopatología , Trastornos Mieloproliferativos/fisiopatología , Neoplasias/fisiopatología , Calreticulina/genética , Citocinas/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Síndrome Hipereosinofílico/genética , Síndrome Hipereosinofílico/fisiopatología , Inflamación/genética , Janus Quinasa 2/genética , Leucemia , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/fisiopatología , Mastocitosis/genética , Mastocitosis/fisiopatología , Mutación , Trastornos Mieloproliferativos/genética , Fenotipo , Policitemia Vera/genética , Policitemia Vera/fisiopatología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/fisiopatología , Receptores de Trombopoyetina/genética , Trombocitemia Esencial/genética , Trombocitemia Esencial/fisiopatología
18.
Blood ; 120(1): 166-72, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22627765

RESUMEN

In the current model of the pathogenesis of polycythemia vera (PV), the JAK2V617F mutation arises in hematopoietic stem cells (HSCs) that maintain the disease, while erythroid precursor populations expand, resulting in excessive red blood cell production. We examined the role of these specific cell populations using a conditional Jak2V617F knockin murine model. We demonstrate that the most immature long-term (LT) HSCs are solely responsible for initiating and maintaining the disease in vivo and that Jak2V617F mutant LT-HSCs dominate hematopoiesis over time. When we induced Jak2V617F expression in erythropoietin receptor expressing precursor cells, the mice developed elevated hematocrit, expanded erythroid precursors, and suppressed erythropoietin levels. However, the disease phenotype was significantly attenuated compared with mice expressing Jak2V617F in LT-HSCs. In addition to developing a PV phenotype, all mice transplanted with Jak2V617F LT-HSCs underwent myelofibrotic transformation over time. These findings recapitulate the development of post-PV myelofibrosis in human myeloproliferative neoplasms. In aggregate, these results demonstrate the distinct roles of LT-HSCs and erythroid precursors in the pathogenesis of PV.


Asunto(s)
Células Eritroides/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Janus Quinasa 2/genética , Policitemia Vera/patología , Policitemia Vera/fisiopatología , Animales , Trasplante de Médula Ósea , Linaje de la Célula/fisiología , Células Clonales/citología , Modelos Animales de Enfermedad , Janus Quinasa 2/metabolismo , Ratones , Ratones Mutantes , Fenotipo , Mutación Puntual/fisiología , Policitemia Vera/genética , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/fisiopatología
19.
Haematologica ; 99(2): 292-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23911705

RESUMEN

Prior to Janus kinase inhibitors, available therapies for myelofibrosis were generally supportive and did not improve survival. This analysis compares efficacy outcomes of patients with myelofibrosis in the control arms (placebo [n=154] and best available therapy [n=73]) from the two phase 3 COntrolled MyeloFibrosis study with ORal JAK inhibitor Treatment (COMFORT) studies. Spleen volume was assessed by magnetic resonance imaging/computed tomography at baseline and every 12 weeks through week 72; spleen length was assessed by palpation at each study visit. Health-related quality of life and symptoms were assessed using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 Items at baseline and in weeks 4, 8, 12, 16 and 24 in COMFORT-I and in weeks 8, 16, 24 and 48 in COMFORT-II. The demographic and baseline characteristics were similar between the control arms of the two studies. One patient who received placebo and no patients who received best available therapy had a ≥35% reduction in spleen volume from baseline at week 24. At 24 weeks, neither placebo nor best available therapy had produced clinically meaningful changes in global quality of life or symptom scales. Non-hematologic adverse events were mostly grade 1/2; the most frequently reported adverse events in each group were abdominal pain, fatigue, peripheral edema and diarrhea. These data suggest that non-Janus kinase inhibitor therapies provide little improvement in splenomegaly, symptoms or quality of life as compared with placebo. Both COMFORT-I (NCT00952289) and COMFORT-II (NCT00934544) studies have been appropriately registered with clinicaltrials.gov.


Asunto(s)
Inhibidores de Proteínas Quinasas/administración & dosificación , Calidad de Vida , Bazo , Adulto , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/fisiopatología , Bazo/patología , Bazo/fisiopatología , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/patología , Esplenomegalia/fisiopatología , Factores de Tiempo
20.
J Pediatr Hematol Oncol ; 36(4): 319-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23619118

RESUMEN

Primary myelofibrosis is rare in children. Many causes have been described for secondary myelofibrosis including vitamin D deficiency. Here, we describe a patient with myelofibrosis secondary to vitamin D deficiency, as diagnosed by laboratory evidence. The patient also developed resultant extramedullary hematopoiesis with secondary development of ascites as a result of myelofibrosis. These are findings rarely reported in association with vitamin D deficiency. Potential mechanisms are also discussed.


Asunto(s)
Hematopoyesis Extramedular , Hígado , Mielofibrosis Primaria , Deficiencia de Vitamina D , Ascitis/etiología , Ascitis/metabolismo , Ascitis/patología , Ascitis/fisiopatología , Femenino , Humanos , Lactante , Hígado/metabolismo , Hígado/patología , Hígado/fisiopatología , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/fisiopatología , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/metabolismo , Deficiencia de Vitamina D/patología , Deficiencia de Vitamina D/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA