Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 925
Filtrar
1.
Leuk Res ; 112: 106755, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34844140

RESUMEN

Iron deficiency (ID) and iron deficiency anemia (IDA) have many adverse effects on human health. Also, iron deficiency anemia and anemia in general are linked with an increased risk of various cancers, particularly blood cancers. It is known that subjects with IDA as well as smokers have elevated blood levels of toxic divalent cations, particularly cadmium (Cd2+) and lead (Pb2+). Cadmium is a proven carcinogen. Most of the circulating cadmium is bound to transferrin and apart from the target organs of cadmium accumulation, kidney and liver, tissues (cells) which highly express transferrin receptor 1 (TfR1) may also accumulate high levels of circulating cadmium. Density of TfR1, glycoprotein that is expressed on cell surface, is not uniform in bone marrow cells. Namely, megakaryocyte/erythrocyte progenitors and pro-erythroblasts express TfR1 incomparably more than other cell lines within the bone marrow and we hypothesize that the mentioned cell lines will uptake most of the circulating cadmium and lead, and will consequently be most suitable for malignant transformation. In this review, we discuss in detail the mechanisms involved in accumulation of cadmium in particular cell lines of the bone marrow and the consequent occurrence of acute myeloid leukemia (AML).


Asunto(s)
Anemia Ferropénica/sangre , Cadmio/sangre , Transformación Celular Neoplásica , Plomo/sangre , Leucemia Mieloide/sangre , Metales Pesados/sangre , Enfermedad Aguda , Anemia Ferropénica/complicaciones , Anemia Ferropénica/metabolismo , Antígenos CD/metabolismo , Células de la Médula Ósea/metabolismo , Cadmio/metabolismo , Eritroblastos/metabolismo , Humanos , Plomo/metabolismo , Leucemia Mieloide/etiología , Leucemia Mieloide/metabolismo , Metales Pesados/metabolismo , Modelos Biológicos , Receptores de Transferrina/metabolismo
2.
Leukemia ; 35(12): 3352-3360, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34518645

RESUMEN

Children with Down syndrome are at an elevated risk of leukemia, especially myeloid leukemia (ML-DS). This malignancy is frequently preceded by transient abnormal myelopoiesis (TAM), which is self-limited expansion of fetal liver-derived megakaryocyte progenitors. An array of international studies has led to consensus in treating ML-DS with reduced-intensity chemotherapy, leading to excellent outcomes. In addition, studies performed in the past 20 years have revealed many of the genetic and epigenetic features of the tumors, including GATA1 mutations that are arguably associated with all cases of both TAM and ML-DS. Despite these advances in understanding the clinical and biological aspects of ML-DS, little is known about the mechanisms of relapse. Upon relapse, patients face a poor outcome, and there is no consensus on treatment. Future studies need to be focused on this challenging aspect of leukemia in children with DS.


Asunto(s)
Síndrome de Down/complicaciones , Factor de Transcripción GATA1/genética , Leucemia Mieloide/patología , Mutación , Humanos , Leucemia Mieloide/etiología , Leucemia Mieloide/metabolismo
3.
Front Immunol ; 12: 683381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220833

RESUMEN

Natural killer (NK) cells are prominent cytotoxic and cytokine-producing components of the innate immune system representing crucial effector cells in cancer immunotherapy. Presently, various NK cell-based immunotherapies have contributed to the substantial improvement in the reconstitution of NK cells against advanced-staged and high-risk AML. Various NK cell sources, including haploidentical NK cells, adaptive NK cells, umbilical cord blood NK cells, stem cell-derived NK cells, chimeric antigen receptor NK cells, cytokine-induced memory-like NK cells, and NK cell lines have been identified. Devising innovative approaches to improve the generation of therapeutic NK cells from the aforementioned sources is likely to enhance NK cell expansion and activation, stimulate ex vivo and in vivo persistence of NK cells and improve conventional treatment response of myeloid leukemia. The tumor-promoting properties of the tumor microenvironment and downmodulation of NK cellular metabolic activity in solid tumors and hematological malignancies constitute a significant impediment in enhancing the anti-tumor effects of NK cells. In this review, we discuss the current NK cell sources, highlight ongoing interventions in enhancing NK cell function, and outline novel strategies to circumvent immunosuppressive factors in the tumor microenvironment to improve the efficacy of NK cell-based immunotherapy and expand their future success in treating myeloid leukemia.


Asunto(s)
Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Leucemia Mieloide/terapia , Biomarcadores , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Terapia Combinada , Citocinas/metabolismo , Citotoxicidad Inmunológica , Manejo de la Enfermedad , Humanos , Factores Inmunológicos/metabolismo , Memoria Inmunológica , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/etiología , Microambiente Tumoral/inmunología
4.
Blood ; 138(21): 2093-2105, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34125889

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) is associated with increased risk of cancers and inflammation-related diseases. This phenomenon becomes common in persons aged ≥80 years, in whom the implications of CHIP are not well defined. We performed a mutational screening in 1794 persons aged ≥80 years and investigated the relationships between CHIP and associated pathologies. Mutations were observed in one-third of persons aged ≥80 years and were associated with reduced survival. Mutations in JAK2 and splicing genes, multiple mutations (DNMT3A, TET2, and ASXL1 with additional genetic lesions), and variant allele frequency ≥0.096 had positive predictive value for myeloid neoplasms. Combining mutation profiles with abnormalities in red blood cell indices improved the ability of myeloid neoplasm prediction. On this basis, we defined a predictive model that identifies 3 risk groups with different probabilities of developing myeloid neoplasms. Mutations in DNMT3A, TET2, ASXL1, or JAK2 were associated with coronary heart disease and rheumatoid arthritis. Cytopenia was common in persons aged ≥80 years, with the underlying cause remaining unexplained in 30% of cases. Among individuals with unexplained cytopenia, the presence of highly specific mutation patterns was associated with myelodysplastic-like phenotype and a probability of survival comparable to that of myeloid neoplasms. Accordingly, 7.5% of subjects aged ≥80 years with cytopenia had presumptive evidence of myeloid neoplasm. In summary, specific mutational patterns define different risk of developing myeloid neoplasms vs inflammatory-associated diseases in persons aged ≥80 years. In individuals with unexplained cytopenia, mutational status may identify those subjects with presumptive evidence of myeloid neoplasms.


Asunto(s)
Hematopoyesis Clonal , Mutación , Factores de Edad , Anciano de 80 o más Años , Artritis Reumatoide/etiología , Artritis Reumatoide/genética , Enfermedad Coronaria/etiología , Enfermedad Coronaria/genética , Femenino , Humanos , Leucemia Mieloide/etiología , Leucemia Mieloide/genética , Masculino , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/genética
5.
Curr Hematol Malig Rep ; 16(3): 286-303, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33945086

RESUMEN

PURPOSE OF REVIEW: Monocytosis is a distinct but non-specific manifestation of various physiologic and pathologic conditions. Among hematopoietic stem cell neoplasms, depending on the criteria used for disease classification, monocytosis may be a consistent and integral component of diseases such as chronic myelomonocytic leukemia or acute myeloid leukemia with monocytic differentiation, or it may represent an inconsistent finding that often provides a clue to the underlying genetic changes driving the neoplasm. The purpose of this review is to provide the readers with a laboratory-based approach to neoplastic monocytosis. RECENT FINDINGS: In-depth elucidation of the genomic landscape of myeloid neoplasms within the past few years has broadened our understanding of monocytosis and its implications for diagnosis and prognosis. Genetic findings also shed light on potential disease response - or lack thereof - to various therapeutic agents used in the setting of myeloid neoplasms. In this review, we provide our approach to diagnose neoplastic monocytosis in the context of case-based studies while incorporating the most recent literature on this topic.


Asunto(s)
Leucemia Mieloide/diagnóstico , Leucemia Mielomonocítica Crónica/diagnóstico , Factores de Edad , Biomarcadores , Médula Ósea/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Manejo de la Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Leucemia Mieloide/etiología , Leucemia Mieloide/mortalidad , Leucemia Mielomonocítica Crónica/etiología , Leucemia Mielomonocítica Crónica/mortalidad , Monocitos/metabolismo , Monocitos/patología
6.
Curr Hematol Malig Rep ; 16(3): 276-285, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33890194

RESUMEN

PURPOSE OF REVIEW: In this review, we provide a comprehensive and contemporary understanding of malignant monocytosis and provide a framework by which the appropriate diagnosis with malignant monocytosis can be rendered. RECENT FINDINGS: Increasing data support the use of molecular data to refine the diagnostic approach to persistent monocytosis. The absence of a TET2, SRSF2, or ASXL1 mutation has ≥ 90% negative predictive value for a diagnosis of CMML. These data may also reliably differentiate chronic myelomonocytic leukemia, the malignancy that is most associated with mature monocytosis, from several other diseases that can be associated with typically a lesser degree of monocytosis. These include acute myelomonocytic leukemia, acute myeloid leukemia with monocytic differentiation, myelodysplastic syndromes, and myeloproliferative neoplasms driven by BCR-ABL1, PDGFRA, PDGFRB, or FGFR1 rearrangements or PCM1-JAK2 fusions among other rarer aberrations. The combination of monocyte partitioning with molecular data in patients with persistent monocytosis may increase the predictive power for the ultimate development of CMM but has not been prospectively validated. Many conditions, both benign and malignant, can be associated with an increase in mature circulating monocytes. After reasonably excluding a secondary or reactive monocytosis, there should be a concern for and investigation of malignant monocytosis, which includes hematopathologic review of blood and marrow tissues, flow cytometric analysis, and cytogenetic and molecular studies to arrive at an appropriate diagnosis.


Asunto(s)
Susceptibilidad a Enfermedades , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/etiología , Monocitos/metabolismo , Monocitos/patología , Edad de Inicio , Algoritmos , Animales , Biomarcadores de Tumor , Biopsia , Médula Ósea/patología , Toma de Decisiones Clínicas , Manejo de la Enfermedad , Regulación Leucémica de la Expresión Génica , Humanos , Inmunohistoquímica , Leucemia Mieloide/mortalidad , Leucemia Mieloide/terapia , Neoplasias Primarias Secundarias/etiología
7.
Curr Hematol Malig Rep ; 16(3): 267-275, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33880680

RESUMEN

PURPOSE OF REVIEW: Monocytosis is a frequently encountered clinical condition that needs appropriate investigation due to a broad range of differential diagnoses. This review is meant to summarize the latest literature in the diagnostic testing and interpretation and offer a stepwise diagnostic approach for a patient presenting with monocytosis. RECENT FINDINGS: Basic studies have highlighted the phenotypic and functional heterogeneity in the monocyte compartment. Studies, both translational and clinical, have provided insights into why monocytosis occurs and how to distinguish the different etiologies. Flow cytometry studies have illustrated that monocyte repartitioning can distinguish chronic myelomonocytic leukemia, a prototypical neoplasm with monocytosis from other reactive or neoplastic causes. In summary, we provide an algorithmic approach to the diagnosis of a patient presenting with monocytosis and expect this document to serve as a reference guide for clinicians.


Asunto(s)
Leucemia Mieloide/diagnóstico , Biomarcadores de Tumor , Médula Ósea/patología , Evolución Clonal/genética , Evolución Clonal/inmunología , Diagnóstico Diferencial , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Citometría de Flujo , Humanos , Leucemia Mieloide/etiología , Leucemia Mieloide/terapia , Leucemia Mielomonocítica Crónica
10.
Curr Hematol Malig Rep ; 16(3): 256-266, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33728588

RESUMEN

PURPOSE OF REVIEW: For decades, the management of chronic myelomonocytic leukemia (CMML) or juvenile myelomonocytic leukemia (JMML) has been largely inextricable from myelodysplastic syndromes (MDS), myeloproliferative neoplasms, and acute myeloid leukemia. Hallmarks of these diseases have been the emergence of unique genomic signatures and discouraging responses to available therapies. Here, we will critically examine the current options for management and review the rapidly developing opportunities based on advances in CMML and JMML disease biology. RECENT FINDINGS: Few clinical trials have exclusively been done in CMML, and in JMML, the rarity of the disease limits wide scale participation. Recent case series in JMML suggest that hypomethylating agents (HMAs) are a viable option for bridging to curative intent with allogeneic hematopoietic stem cell transplant or as posttransplant maintenance. Emerging evidence has demonstrated targeting the RAS-pathway via MEK inhibition may also be considered. In CMML, treatment with HMAs is largely derived from data inclusive of MDS patients, including a small number of patients with dysplastic CMML variants. Based on CMML disease biology, additional therapeutic targets being investigated include inhibitors of splicing, CD123/dendritic cell axis, inherent GM-CSF progenitor cell hypersensitivity, and targeting the JAK/STAT pathway. Current evidence is also expanding for oral HMAs. The management of CMML and JMML is rapidly evolving and clinicians must be aware of the genetic landscape and expanding treatment options to ensure these rare populations are afforded therapeutic interventions best suited to their needs.


Asunto(s)
Leucemia Mieloide/terapia , Leucemia Mielomonocítica Juvenil/terapia , Factores de Edad , Biomarcadores , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/etiología , Leucemia Mielomonocítica Crónica/diagnóstico , Leucemia Mielomonocítica Crónica/etiología , Leucemia Mielomonocítica Crónica/terapia , Leucemia Mielomonocítica Juvenil/diagnóstico , Leucemia Mielomonocítica Juvenil/etiología , Terapia Molecular Dirigida
15.
Nat Commun ; 11(1): 1659, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32246016

RESUMEN

Oncogenic Ras mutations occur in various leukemias. It was unclear if, besides the direct transforming effect via constant RAS/MEK/ERK signaling, an inflammation-related effect of KRAS contributes to the disease. Here, we identify a functional link between oncogenic KrasG12D and NLRP3 inflammasome activation in murine and human cells. Mice expressing active KrasG12D in the hematopoietic system developed myeloproliferation and cytopenia, which is reversed in KrasG12D mice lacking NLRP3 in the hematopoietic system. Therapeutic IL-1-receptor blockade or NLRP3-inhibition reduces myeloproliferation and improves hematopoiesis. Mechanistically, KrasG12D-RAC1 activation induces reactive oxygen species (ROS) production causing NLRP3 inflammasome-activation. In agreement with our observations in mice, patient-derived myeloid leukemia cells exhibit KRAS/RAC1/ROS/NLRP3/IL-1ß axis activity. Our findings indicate that oncogenic KRAS not only act via its canonical oncogenic driver function, but also enhances the activation of the pro-inflammatory RAC1/ROS/NLRP3/IL-1ß axis. This paves the way for a therapeutic approach based on immune modulation via NLRP3 blockade in KRAS-mutant myeloid malignancies.


Asunto(s)
Inflamasomas/inmunología , Trastornos Mieloproliferativos/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Proliferación Celular , Expresión Génica , Hematopoyesis , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Leucemia Mieloide/etiología , Leucemia Mieloide/genética , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Células Mieloides/metabolismo , Proteínas NLR/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
16.
Ir J Med Sci ; 189(3): 979-984, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32006388

RESUMEN

BACKGROUND: Acute megakaryoblastic leukaemia (AMKL) is a subtype of myeloid leukaemia and is the most common leukaemia type in children with Down syndrome (DS) under 4 years of age. AMKL is often preceded by a transient neonatal pre-leukaemic syndrome, transient myeloproliferative disorder (TMD). Although TMD often spontaneously resolves, 20-30% of these patients subsequently develop AMKL within the first 4 years of life. AIMS: To perform a retrospective consecutive national audit of all documented cases of childhood TMD and AMKL-DS from 1990 to 2018 at Our Lady's Children's Hospital, Crumlin (OLCHC), Ireland. METHODS: All patients with a diagnosis of AMKL treated consecutively at (OLCHC) between 1990 and 2018 were reviewed. Kaplan-Meier survival curves were constructed. RESULTS: Twenty-seven patients with AMKL-DS were identified. A prior neonatal diagnosis of TMD was described in 10 patients (37%). Nineteen patients (70%) are alive and well, in complete remission, at a median follow-up of 11.4 years. Overall survival (OS) of this cohort has risen from 54% from those treated between the years 1990 and 2004 (n = 13) to 93% for those treated between the years 2005 and 2018 (n = 14). CONCLUSION: High cure rates are observed in AMKL-DS using current polychemotherapy protocols. The finding of a low platelet count at time of diagnosis is in keeping with the knowledge that AMKL-DS is a malignancy of platelet progenitor cells.


Asunto(s)
Síndrome de Down/complicaciones , Leucemia Mieloide/etiología , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Irlanda , Masculino , Estudios Retrospectivos
17.
Semin Cancer Biol ; 60: 191-201, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31408723

RESUMEN

The development of a myeloid neoplasm is a step-wise process that originates from leukemic stem cells (LSC) and includes pre-leukemic stages, overt leukemia and a drug-resistant terminal phase. Organ-invasion may occur in any stage, but is usually associated with advanced disease and a poor prognosis. Sometimes, extra-medullary organ invasion shows a metastasis-like or even sarcoma-like destructive growth of neoplastic cells in local tissue sites. Examples are myeloid sarcoma, mast cell sarcoma and localized blast phase of chronic myeloid leukemia. So far, little is known about mechanisms underlying re-distribution and extramedullary dissemination of LSC in myeloid neoplasms. In this article, we discuss mechanisms through which LSC can mobilize out of the bone marrow niche, can transmigrate from the blood stream into extramedullary organs, can invade local tissue sites and can potentially create or support the formation of local stem cell niches. In addition, we discuss strategies to interfere with LSC expansion and organ invasion by targeted drug therapies.


Asunto(s)
Leucemia Mieloide/etiología , Leucemia Mieloide/metabolismo , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral , Animales , Biomarcadores , Médula Ósea/patología , Comunicación Celular , Movimiento Celular , Humanos , Inmunofenotipificación , Leucemia Mieloide/patología , Estadificación de Neoplasias , Células Madre Neoplásicas/patología , Fenotipo , Recurrencia , Migración Transendotelial y Transepitelial/genética , Microambiente Tumoral/genética
19.
Pediatr Blood Cancer ; 66(11): e27938, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31347793

RESUMEN

The therapies used to treat Ewing sarcoma are associated with a risk of second malignant neoplasm (SMN). We conducted a systematic review to pool available evidence on the risks, types, and outcomes after SMN. We obtained 52 articles that met inclusion criteria. Cumulative incidence rates of SMN ranged from 0.9 to 8.4% and 10.1 to 20.5% at 5 and 30 years after initial diagnosis. Of the 327 reported SMNs, 63.6% were solid tumors, although acute myeloid leukemia /myelodysplastic syndrome was the single most commonly diagnosed SMN, with generally poor outcomes. Patients treated for Ewing sarcoma are at substantial risk of SMN, with a broad range of reported secondary cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias Óseas , Neoplasias Inducidas por Radiación/etiología , Neoplasias Primarias Secundarias/etiología , Radioterapia/efectos adversos , Sarcoma de Ewing , Factores de Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/radioterapia , Neoplasias Óseas/cirugía , Carcinoma/epidemiología , Carcinoma/etiología , Carcinoma/terapia , Humanos , Incidencia , Leucemia Mieloide/epidemiología , Leucemia Mieloide/etiología , Leucemia Mieloide/terapia , Linfoma/epidemiología , Linfoma/etiología , Linfoma/terapia , Melanoma/epidemiología , Melanoma/etiología , Melanoma/terapia , Neoplasias Inducidas por Radiación/epidemiología , Neoplasias Inducidas por Radiación/terapia , Neoplasias Primarias Secundarias/epidemiología , Neoplasias Primarias Secundarias/terapia , Riesgo , Sarcoma/epidemiología , Sarcoma/etiología , Sarcoma/terapia , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/radioterapia , Sarcoma de Ewing/cirugía , Factores de Tiempo , Resultado del Tratamiento
20.
Br J Haematol ; 186(4): 524-537, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31236939

RESUMEN

Despite significant advances in the treatment of myeloid malignancies, many patients become resistant to therapy and ultimately succumb to their disease. Accumulating evidence over the past several years has suggested that the inadequacy of many leukaemia therapies results from their failure to target the leukaemic stem cell (LSC). For this reason, the LSC population currently represents the most critical target in the treatment of myeloid malignancies. However, while LSCs are ideal targets in the treatment of these diseases, they are also the most difficult population to target. This is due to both their heterogeneity within the LSC population, and also their phenotypic similarities with normal haematopoietic stem cells. This review will highlight the current landscape surrounding LSC biology in myeloid malignancies, with a focus on altered energy metabolism, and how that knowledge is being translated into clinical advances for the treatment of chronic and acute myeloid leukaemia and myelodysplastic syndromes.


Asunto(s)
Antineoplásicos/farmacología , Metabolismo Energético , Leucemia Mieloide/etiología , Leucemia Mieloide/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos/genética , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/tratamiento farmacológico , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/terapia , Transducción de Señal , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...