Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 222
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163413

RESUMEN

Neutrophils are an essential component of the innate immune response, but their prolonged activation can lead to chronic inflammation. Consequently, neutrophil homeostasis is tightly regulated through balance between granulopoiesis and clearance of dying cells. The bone marrow is both a site of neutrophil production and the place they return to and die. Myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders characterized by the mutations in three types of molecular markers, with emphasis on Janus kinase 2 gene mutation (JAK2V617F). The MPN bone marrow stem cell niche is a site of chronic inflammation, with commonly increased cells of myeloid lineage, including neutrophils. The MPN neutrophils are characterized by the upregulation of JAK target genes. Additionally, MPN neutrophils display malignant nature, they are in a state of activation, and with deregulated apoptotic machinery. In other words, neutrophils deserve to be placed in the midst of major events in MPN. Our crucial interest in this review is better understanding of how neutrophils die in MPN mirrored by defects in apoptosis and to what possible extent they can contribute to MPN pathophysiology. We tend to expect that reduced neutrophil apoptosis will establish a pathogenic link to chronic inflammation in MPN.


Asunto(s)
Neoplasias Hematológicas/inmunología , Inmunidad Innata , Trastornos Mieloproliferativos/inmunología , Neutrófilos/inmunología , Sustitución de Aminoácidos , Animales , Enfermedad Crónica , Neoplasias Hematológicas/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Mutación Missense , Trastornos Mieloproliferativos/genética
3.
Br J Haematol ; 196(5): 1149-1158, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34618358

RESUMEN

Breakpoint cluster region-Abelson (BCR-ABL) negative myeloproliferative neoplasms (MPNs) are chronic myeloid neoplasms initiated by the acquisition of gene mutation(s) in a haematopoietic stem cell, leading to clonal expansion and over-production of blood cells and their progenitors. MPNs encompass a spectrum of disorders with overlapping but distinct molecular, laboratory and clinical features. This includes polycythaemia vera, essential thrombocythaemia and myelofibrosis. Dysregulation of the immune system is key to the pathology of MPNs, supporting clonal evolution, mediating symptoms and resulting in varying degrees of immunocompromise. Targeting immune dysfunction is an important treatment strategy. In the present review, we focus on the immune landscape in patients with MPNs - the role of inflammation in disease pathogenesis, susceptibility to infection and emerging strategies for therapeutic immune modulation. Further detailed work is required to delineate immune perturbation more precisely in MPNs to determine how and why vulnerability to infection differs between clinical subtypes and to better understand how inflammation results in a competitive advantage for the MPN clone. These studies may help shed light on new designs for disease-modifying therapies.


Asunto(s)
Inmunoterapia , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/terapia , Animales , Proteínas de Fusión bcr-abl/análisis , Humanos , Inmunidad , Inmunoterapia/métodos , Infecciones/inmunología , Infecciones/patología , Infecciones/terapia , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Trastornos Mieloproliferativos/patología , Escape del Tumor
4.
Blood Adv ; 6(1): 200-206, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34555844

RESUMEN

Development of normal blood cells is often suppressed in juvenile myelomonocytic leukemia (JMML), a myeloproliferative neoplasm (MPN) of childhood, causing complications and impacting therapeutic outcomes. However, the mechanism underlying this phenomenon remains uncharacterized. To address this question, we induced the most common mutation identified in JMML (Ptpn11E76K) specifically in the myeloid lineage with hematopoietic stem cells (HSCs) spared. These mice uniformly developed a JMML-like MPN. Importantly, HSCs in the same bone marrow (BM) microenvironment were aberrantly activated and differentiated at the expense of self-renewal. As a result, HSCs lost quiescence and became exhausted. A similar result was observed in wild-type (WT) donor HSCs when co-transplanted with Ptpn11E76K/+ BM cells into WT mice. Co-culture testing demonstrated that JMML/MPN cells robustly accelerated differentiation in mouse and human normal hematopoietic stem/progenitor cells. Cytokine profiling revealed that Ptpn11E76K/+ MPN cells produced excessive IL-1ß, but not IL-6, T NF-α, IFN-γ, IL-1α, or other inflammatory cytokines. Depletion of the IL-1ß receptor effectively restored HSC quiescence, normalized their pool size, and rescued them from exhaustion in Ptpn11E76K/+/IL-1R-/- double mutant mice. These findings suggest IL-1ß signaling as a potential therapeutic target for preserving normal hematopoietic development in JMML.


Asunto(s)
Células Madre Hematopoyéticas , Inflamación , Interleucina-1beta , Leucemia Mielomonocítica Juvenil , Animales , Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/metabolismo , Leucemia Mielomonocítica Juvenil/inmunología , Leucemia Mielomonocítica Juvenil/metabolismo , Leucemia Mielomonocítica Juvenil/patología , Ratones , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Receptores de Interleucina-1/deficiencia , Microambiente Tumoral
5.
Front Immunol ; 12: 750346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867980

RESUMEN

BCR-ABL1-negative myeloproliferative neoplasms are burdened by a reduced life expectancy mostly due to an increased risk of thrombo-hemorrhagic events, fibrotic progression/leukemic evolution, and infectious complications. In these clonal myeloid malignancies, JAK2V617F is the main driver mutation, leading to an aberrant activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway. Therefore, its inhibition represents an attractive therapeutic strategy for these disorders. Several JAK inhibitors have entered clinical trials, including ruxolitinib, the first JAK1/2 inhibitor to become commercially available for the treatment of myelofibrosis and polycythemia vera. Due to interference with the JAK-STAT pathway, JAK inhibitors affect several components of the innate and adaptive immune systems such as dendritic cells, natural killer cells, T helper cells, and regulatory T cells. Therefore, even though the clinical use of these drugs in MPN patients has led to a dramatic improvement of symptoms control, organ involvement, and quality of life, JAK inhibitors-related loss of function in JAK-STAT signaling pathway can be a cause of different adverse events, including those related to a condition of immune suppression or deficiency. This review article will provide a comprehensive overview of the current knowledge on JAK inhibitors' effects on immune cells as well as their clinical consequences, particularly with regards to infectious complications.


Asunto(s)
Antiinflamatorios/uso terapéutico , Inmunosupresores/uso terapéutico , Inhibidores de las Cinasas Janus/uso terapéutico , Trastornos Mieloproliferativos/tratamiento farmacológico , Animales , Antiinflamatorios/efectos adversos , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Humanos , Inmunosupresores/efectos adversos , Infecciones/etiología , Inhibidores de las Cinasas Janus/efectos adversos , Trastornos Mieloproliferativos/inmunología
8.
Front Immunol ; 12: 772408, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804065

RESUMEN

The immune microenvironment is a critical driver and regulator of leukemic progression and hematological disease. Recent investigations have demonstrated that multiple immune components play a central role in regulating hematopoiesis, and dysfunction at the immune cell level significantly contributes to neoplastic disease. Immune cells are acutely sensitive to remodeling by leukemic inflammatory cytokine exposure. Importantly, immune cells are the principal cytokine producers in the hematopoietic system, representing an untapped frontier for clinical interventions. Due to a proinflammatory cytokine environment, dysregulation of immune cell states is a hallmark of hematological disease and neoplasia. Malignant immune adaptations have profound effects on leukemic blast proliferation, disease propagation, and drug-resistance. Conversely, targeting the immune landscape to restore hematopoietic function and limit leukemic expansion may have significant therapeutic value. Despite the fundamental role of the immune microenvironment during the initiation, progression, and treatment response of hematological disease, a detailed examination of how leukemic cytokines alter immune cells to permit, promote, or inhibit leukemia growth is lacking. Here we outline an immune-based model of leukemic transformation and highlight how the profound effect of immune alterations on the trajectory of malignancy. The focus of this review is to summarize current knowledge about the impacts of pro- and anti-inflammatory cytokines on immune cells subsets, their modes of action, and immunotherapeutic approaches with the potential to improve clinical outcomes for patients suffering from hematological myeloid malignancies.


Asunto(s)
Citocinas/inmunología , Neoplasias Hematológicas/inmunología , Trastornos Mieloproliferativos/inmunología , Animales , Humanos , Inmunidad Innata , Transducción de Señal
9.
Cells ; 10(9)2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34571965

RESUMEN

Myeloproliferative Neoplasms (MPN) are acquired clonal disorders of the hematopoietic stem cells and include Essential Thrombocythemia, Polycythemia Vera and Myelofibrosis. MPN are characterized by mutations in three driver genes (JAK2, CALR and MPL) and by a state of chronic inflammation. Notably, MPN patients experience increased risk of thrombosis, disease progression, second neoplasia and evolution to acute leukemia. Extracellular vesicles (EVs) are a heterogeneous population of microparticles with a role in cell-cell communication. The EV-mediated cross-talk occurs via the trafficking of bioactive molecules such as nucleic acids, proteins, metabolites and lipids. Growing interest is focused on EVs and their potential impact on the regulation of blood cancers. Overall, EVs have been suggested to orchestrate the complex interplay between tumor cells and the microenvironment with a pivotal role in "education" and "crafting" of the microenvironment by regulating angiogenesis, coagulation, immune escape and drug resistance of tumors. This review is focused on the role of EVs in MPN. Specifically, we will provide an overview of recent findings on the involvement of EVs in MPN pathogenesis and discuss opportunities for their potential application as diagnostic and prognostic biomarkers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Vesículas Extracelulares/metabolismo , Trastornos Mieloproliferativos/metabolismo , Microambiente Tumoral , Animales , Biomarcadores de Tumor/genética , Vesículas Extracelulares/genética , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/patología , Policitemia Vera/genética , Policitemia Vera/inmunología , Policitemia Vera/metabolismo , Policitemia Vera/patología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/inmunología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Transducción de Señal , Trombocitemia Esencial/genética , Trombocitemia Esencial/inmunología , Trombocitemia Esencial/metabolismo , Trombocitemia Esencial/patología , Microambiente Tumoral/inmunología
10.
Int J Mol Sci ; 22(17)2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34502471

RESUMEN

Philadelphia chromosome negative myeloproliferative neoplasms (MPN) are composed of polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). The clinical picture is determined by constitutional symptoms and complications, including arterial and venous thromboembolic or hemorrhagic events. MPNs are characterized by mutations in JAK2, MPL, or CALR, with additional mutations leading to an expansion of myeloid cell lineages and, in PMF, to marrow fibrosis and cytopenias. Chronic inflammation impacting the initiation and expansion of disease in a major way has been described. Neutrophilic granulocytes play a major role in the pathogenesis of thromboembolic events via the secretion of inflammatory markers, as well as via interaction with thrombocytes and the endothelium. In this review, we discuss the molecular biology underlying myeloproliferative neoplasms and point out the central role of leukocytosis and, specifically, neutrophilic granulocytes in this group of disorders.


Asunto(s)
Neoplasias Hematológicas/inmunología , Trastornos Mieloproliferativos/inmunología , Proteínas de Neoplasias/inmunología , Neutrófilos/inmunología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Humanos , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Proteínas de Neoplasias/genética , Neutrófilos/patología , Cromosoma Filadelfia
12.
J Hematol Oncol ; 14(1): 81, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001183

RESUMEN

BACKGROUND: Safety and immunogenicity of BNT162b2 mRNA vaccine are unknown in hematological patients; both were evaluated prospectively in 42 patients with multiple myeloma (MM) and 50 with myeloproliferative malignancies (MPM) (20 chronic myeloid leukemias and 30 myeloproliferative neoplasms), all of them on active anti-cancer treatment, in comparison with 36 elderly controls not suffering from cancer. Subjects serologically and/or molecularly (by nasal/throat swab) positives at basal for SARS-CoV-2 were excluded. Primary endpoint was to compare titers of neutralizing anti-SARS-CoV-2 IgG and seroprotection rates among the cohorts at 3 and 5 weeks from first dose. METHODS: Titration was done using LIAISON® SARS-CoV-2 S1/S2 IgG test, a quantitative chemiluminescent immunoassay approved by FDA on the basis of robust evidences of concordance (94.4%) between the test at cutoff of 15 AU/mL and the Plaque Reduction Neutralization Test 90% at 1:40 ratio. Cutoff of 15 AU/mL was assumed to discriminate responders to vaccination with a protective titer. Cohorts were compared using Fisher' exact test and the Mann-Whitney test as appropriated. Geometric mean concentrations (GMCs), geometric mean ratios and response rates after 1st and 2nd dose were compared in each cohort by Wilcoxon and McNemar tests, respectively. RESULTS: At 5 weeks, GMC of IgG in elderly controls was 353.3 AU/mL versus 106.7 in MM (p = 0.003) and 172.9 in MPM patients (p = 0.049). Seroprotection rate at cutoff of 15 AU/mL was 100% in controls compared to 78.6% in MM (p = 0.003) and 88% in MPM patients (p = 0.038). In terms of logarithm of IgG titer, in a generalized multivariate linear model, no gender effect was observed (p = 0.913), while there was a significant trend toward lower titers by increasing age (p < 0.001) and in disease cohorts with respect to controls (MM: p < 0.001 and MPM: p < 0.001). An ongoing treatment without daratumumab was associated with higher likelihood of response in MM patients (p = 0.003). No swabs resulted positive on each time point. No safety concerns were observed. CONCLUSIONS: BNT162b2 has demonstrated to be immunogenic at different extent among the cohorts. Response was 88% and robust in MPM patients. MM patients responded significantly less, particularly those on anti-CD38-based treatment. These latter patients should be advised to maintain masks and social distancing regardless of vaccination status, and their cohabiting family members need to be vaccinated in order to reduce the risk of contagion from the family. Additional boosters and titer monitoring could be considered. Trial registration Study was formally approved by the IRCCS Central Ethical Committee of Regione Lazio in January 2021 (Prot. N-1463/21).


Asunto(s)
Vacunas contra la COVID-19/uso terapéutico , COVID-19/prevención & control , Mieloma Múltiple/complicaciones , Trastornos Mieloproliferativos/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Vacuna BNT162 , COVID-19/inmunología , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/inmunología , Femenino , Humanos , Inmunogenicidad Vacunal , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Mieloma Múltiple/inmunología , Trastornos Mieloproliferativos/inmunología , Datos Preliminares , Estudios Prospectivos , SARS-CoV-2/inmunología
13.
Eur J Haematol ; 107(3): 324-332, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34022082

RESUMEN

OBJECTIVE: ABO mismatch between donor and recipient occurs in 40% of allogeneic hematopoietic stem cell transplantations (HCT). Different strategies have been described to reduce isohemagglutinins (IHA) before HCT. We describe the effect of selective ABO immunoadsorption (ABO IA) on erythrocyte transfusion rate and the development of post-transplant pure red cell aplasia (ptPRCA). METHODS: 63 patients with major ABO incompatibility were retrospectively analyzed. Nine patients with major ABO incompatibility and high-IHA titer were treated by ABO IA before HCT. We analyzed the need for transfusion and the occurrence of ptPRCA. We compared the outcome with patients treated by other methods to reduce IHA. RESULTS: In all nine patients treated by ABO IA, IHA decreased in a median four times. PtPRCA occurred in one patient. The median number of transfusions was 8 (range: 0-36) between d0 and d100. In 25 patients with high-IHA titer without treatment or treated by other methods to reduce IHA, the need for transfusions was comparable. No difference in the incidence of ptPRCA was observed. CONCLUSIONS: Selective ABO IA is a feasible, safe, and effective method to reduce IHA before HCT in major ABO incompatibility. No effect on transfusion rate or ptPRCA compared to other strategies could be observed.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia/terapia , Síndromes Mielodisplásicos/terapia , Trastornos Mieloproliferativos/terapia , Plasmaféresis/métodos , Aplasia Pura de Células Rojas/prevención & control , Reacción a la Transfusión/prevención & control , Sistema del Grupo Sanguíneo ABO/inmunología , Incompatibilidad de Grupos Sanguíneos/inmunología , Incompatibilidad de Grupos Sanguíneos/mortalidad , Incompatibilidad de Grupos Sanguíneos/terapia , Transfusión de Eritrocitos/efectos adversos , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Leucemia/inmunología , Leucemia/mortalidad , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/mortalidad , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/mortalidad , Aplasia Pura de Células Rojas/etiología , Aplasia Pura de Células Rojas/inmunología , Aplasia Pura de Células Rojas/mortalidad , Estudios Retrospectivos , Análisis de Supervivencia , Donantes de Tejidos , Reacción a la Transfusión/etiología , Reacción a la Transfusión/inmunología , Reacción a la Transfusión/mortalidad , Trasplante Homólogo , Resultado del Tratamiento
15.
Int J Mol Sci ; 22(4)2021 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-33672997

RESUMEN

The Philadelphia-negative myeloproliferative neoplasms (MPNs) are malignancies of the hematopoietic stem cell (HSC) arising as a consequence of clonal proliferation driven by somatically acquired driver mutations in discrete genes (JAK2, CALR, MPL). In recent years, along with the advances in molecular characterization, the role of immune dysregulation has been achieving increasing relevance in the pathogenesis and evolution of MPNs. In particular, a growing number of studies have shown that MPNs are often associated with detrimental cytokine milieu, expansion of the monocyte/macrophage compartment and myeloid-derived suppressor cells, as well as altered functions of T cells, dendritic cells and NK cells. Moreover, akin to solid tumors and other hematological malignancies, MPNs are able to evade T cell immune surveillance by engaging the PD-1/PD-L1 axis, whose pharmacological blockade with checkpoint inhibitors can successfully restore effective antitumor responses. A further interesting cue is provided by the recent discovery of the high immunogenic potential of JAK2V617F and CALR exon 9 mutations, that could be harnessed as intriguing targets for innovative adoptive immunotherapies. This review focuses on the recent insights in the immunological dysfunctions contributing to the pathogenesis of MPNs and outlines the potential impact of related immunotherapeutic approaches.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inmunoterapia/métodos , Inflamación/inmunología , Trastornos Mieloproliferativos/terapia , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Calreticulina/genética , Calreticulina/inmunología , Calreticulina/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Inflamación/genética , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Janus Quinasa 2/metabolismo , Mutación/inmunología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Cromosoma Filadelfia , Linfocitos T/metabolismo , Microambiente Tumoral/genética
16.
J Immunol Methods ; 493: 113015, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33689807

RESUMEN

BACKGROUND: During eosinophil differentiation, the granule eosinophil major basic protein 1 (eMBP1) is synthesized as a 32-kDa precursor form, referred to as proMBP1, which is processed into the 14-kDa mature form of eMBP1. The prevalence of these two forms of MBP1 in most pathological conditions remains unknown. OBJECTIVE: To develop the immunoassays that differentiate mature eMBP1 and proMBP1 and apply them to analyze their levels in biological fluids from patients with eosinophilia and hematologic disorders. METHODS: We produced a series of monoclonal antibodies and selected pairs capable of discriminating between the two molecular forms of eMBP1. Radioimmunoassay (RIA) was performed to simultaneously quantitate the levels of mature eMBP1 and proMBP1 in secretions from patients with chronic rhinosinusitis (CRS) and sera from patients with hypereosinophilic syndrome (HES) and other myeloproliferative disorders. RESULTS: The novel immunoassays possessed less than 1% crossreactivity between mature eMBP1 and proMBP1. Mature eMBP1, but not proMBP1, was found in nasal secretions of CRS patients. In contrast, elevated serum levels of mature eMBP1 and proMBP1 were observed in approximately 60% and 90% of HES patients, respectively, with proMBP1 present in greater quantities than mature eMBP1. Patients with several myeloproliferative disorders also showed high serum levels of proMBP1 while mature eMBP1 remained at basal levels. CONCLUSION: The novel immunoassays successfully differentiated mature eMBP1 and proMBP1 in human biological fluids. Further studies addressing the clinical correlates of these assays will help to develop biomarkers to diagnose and monitor patients with eosinophilia and myeloproliferative disorders.


Asunto(s)
Proteína Mayor Básica del Eosinófilo/sangre , Eosinofilia/diagnóstico , Inmunoensayo/métodos , Trastornos Mieloproliferativos/diagnóstico , Proteoglicanos/sangre , Anticuerpos Monoclonales/inmunología , Proteína Mayor Básica del Eosinófilo/inmunología , Eosinofilia/inmunología , Humanos , Trastornos Mieloproliferativos/inmunología , Proteoglicanos/inmunología
17.
Int J Mol Sci ; 22(3)2021 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-33498945

RESUMEN

Thrombosis is a major cause of morbimortality in patients with chronic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN). In the last decade, multiple lines of evidence support the role of leukocytes in thrombosis of MPN patients. Besides the increase in the number of cells, neutrophils and monocytes of MPN patients show a pro-coagulant activated phenotype. Once activated, neutrophils release structures composed of DNA, histones, and granular proteins, called extracellular neutrophil traps (NETs), which in addition to killing pathogens, provide an ideal matrix for platelet activation and coagulation mechanisms. Herein, we review the published literature related to the involvement of NETs in the pathogenesis of thrombosis in the setting of MPN; the effect that cytoreductive therapies and JAK inhibitors can have on markers of NETosis, and, finally, the novel therapeutic strategies targeting NETs to reduce the thrombotic complications in these patients.


Asunto(s)
Leucemia/complicaciones , Trastornos Mieloproliferativos/complicaciones , Neutrófilos , Trombosis/etiología , Animales , Humanos , Leucemia/inmunología , Trastornos Mieloproliferativos/inmunología , Trombosis/inmunología
18.
Br J Haematol ; 193(1): 150-154, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33159465

RESUMEN

The seasonal influenza A vaccine is recommended for patients with myeloproliferative neoplasms (MPNs). We hypothesised that immune deregulation associated with MPNs may affect the immune response gained following vaccinations when compared to healthy controls. Using deep immunophenotyping with high-dimensional single-cell analysis and mass cytometry we could demonstrate an altered immune response in MPN patients following vaccination. We found that prior to vaccination, MPN patients had reduced numbers of naive CD4 T cells. Furthermore, at 3-weeks and 3-months post-vaccination there was evidence of both delayed and impaired B- and T-memory cells responses. Thus, although, the immune systems of MPN patients can 'recognise' the Influenza A vaccine, the response appears inferior compared to healthy controls.


Asunto(s)
Inmunidad/efectos de los fármacos , Virus de la Influenza A/inmunología , Gripe Humana/prevención & control , Trastornos Mieloproliferativos/inmunología , Vacunación/efectos adversos , Adulto , Anciano , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Estudios de Casos y Controles , Femenino , Humanos , Inmunidad/inmunología , Memoria Inmunológica/efectos de los fármacos , Inmunofenotipificación/métodos , Gripe Humana/inmunología , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/patología , Neoplasias/diagnóstico , Neoplasias/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
19.
Exp Hematol ; 92: 75-88.e10, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33017633

RESUMEN

Human leukocyte antigen class I (HLA-I) genotype has been found to influence cancer development through the presentation of mutational neoepitopes. However, our understanding of its effect on the development of myeloproliferative neoplasms (MPNs) remains limited. We aimed to elucidate the putative protective role of HLA-I alleles in the development of JAK2 V617F-driven MPNs using a population genetics approach. The variability of the HLA-I genotype had no effect on the presence of JAK2 V617F mutation. However, three alleles were found to be inversely correlated with the presence of JAK2 V617F mutation: HLA-A*02:01 (p = 0.036), HLA-B*35:01 (p = 0.017), and HLA-C*15:02 (p = 0.033). The HLA-B*35:01 allele was predicted to bind to a 9-mer peptide derived from JAK2 V617F mutant protein. Gene expression analysis revealed a lower expression of HLA-A and -B in MPN CD34+ cells compared with normal CD34+ cells, which was modulated by ruxolitinib and interferon-α treatment. In summary, we provide robust evidence that specific HLA-I molecules restrict JAK2 V617F-driven oncogenesis. JAK2 V617F+ stem cells evade immune surveillance through downregulation of the HLA-I expression. Therefore, the presence of specific HLA-I alleles might be a predictive marker for response to certain immunotherapies upregulating HLA-I expression. Finally, our findings have implications in the development of mutational neoepitope-based vaccines in MPNs.


Asunto(s)
Alelos , Carcinogénesis , Neoplasias Hematológicas , Antígenos de Histocompatibilidad Clase I , Janus Quinasa 2 , Mutación Missense , Trastornos Mieloproliferativos , Sustitución de Aminoácidos , Carcinogénesis/genética , Carcinogénesis/inmunología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología
20.
Front Immunol ; 11: 2093, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983162

RESUMEN

Philadelphia-negative myeloproliferative neoplasms (MPN) are malignant bone marrow (BM) disorders, typically arising from a single somatically mutated hematopoietic stem cell. The most commonly mutated genes, JAK2, CALR, and MPL lead to constitutively active JAK-STAT signaling. Common clinical features include myeloproliferation, splenomegaly and constitutional symptoms. This review covers the contributions of cellular components of MPN pathology (e.g., monocytes, megakaryocytes, and mesenchymal stromal cells) as well as cytokines and soluble mediators to the development of myelofibrosis (MF) and highlights recent therapeutic advances. These findings outline the importance of malignant and non-malignant BM constituents to the pathogenesis and treatment of MF.


Asunto(s)
Médula Ósea , Neoplasias Hematológicas , Mutación , Trastornos Mieloproliferativos , Proteínas de Neoplasias , Transducción de Señal , Microambiente Tumoral , Animales , Médula Ósea/inmunología , Médula Ósea/patología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Humanos , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...