Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Cancer J ; 29(3): 168-178, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37195773

RESUMEN

ABSTRACT: Myelodysplastic syndromes are a heterogeneous group of bone marrow disorders characterized by ineffective hematopoiesis, progressive cytopenias, and an innate capability of progressing to acute myeloid leukemia. The most common causes of morbidity and mortality are complications related to myelodysplastic syndromes rather than progression to acute myeloid leukemia. Although supportive care measures are applicable to all patients with myelodysplastic syndromes, they are especially essential in patients with lower-risk disease who have a better prognosis compared with their higher-risk counterparts and require longer-term monitoring of disease and treatment-related complications. In this review, we will address the most frequent complications and supportive care interventions used in patients with myelodysplastic syndromes, including transfusion support, management of iron overload, antimicrobial prophylaxis, important considerations in the era of COVID-19 (coronavirus infectious disease 2019), role of routine immunizations, and palliative care in the myelodysplastic syndrome population.


Asunto(s)
Síndromes Mielodisplásicos , Cuidados Paliativos , Humanos , Transfusión Sanguínea , Quimioprevención/métodos , COVID-19/epidemiología , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/terapia , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/prevención & control , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/terapia , Cuidados Paliativos/métodos
2.
Microvasc Res ; 140: 104296, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34863990

RESUMEN

Acute myeloid leukemia (AML) has been characterized by the swift development of abnormal cells in the bone marrow. This research aimed to examine the impacts of the miR-185-5p-GPX1 axis on AML progression and differentiation. Findings indicated that miR-185-5p and GPX1 levels were significantly reduced and elevated, respectively. The upregulation of miR-185-5p was observed to restrict the proliferation and invasion abilities of AML cells, and promote differentiate and apoptosis. Moreover, the overexpression of GPX1 was noticed to enhance the growth of AML cells. In conclusion, this research suggested that by targeting GPX1, miR-185-5p inhibited AML progression and downregulated AML cells' proliferation and invasion.


Asunto(s)
Glutatión Peroxidasa/metabolismo , Leucemia Mieloide Aguda/prevención & control , MicroARNs/metabolismo , Animales , Apoptosis , Estudios de Casos y Controles , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Glutatión Peroxidasa/genética , Células HL-60 , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Interferencia de ARN , Transducción de Señal , Glutatión Peroxidasa GPX1
3.
Epidemiol Mikrobiol Imunol ; 70(3): 208-220, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34641695

RESUMEN

Acute leukaemias are malignant diseases of haematopoiesis, traditionally classified according to the affected cell line as acute lymphoblastic and acute myelogenous leukaemia. In terms of incidence, acute leukaemias are rare diseases - in the Czech Republic, only 2-3 new acute myelogenous leukaemia cases/100 000 population are diagnosed annually and less than 1 new case of acute lymphoblastic leukaemia/100 000 residents. The causes of acute leukaemias are still poorly understood. The established risk factors are age, ionizing radiation or Downs syndrome. Moreover, a number of potential risk factors have been described to play a role in development of acute leukaemias and to multiply the risk, such as physical factors, chemicals, genetic and familial predispositions or other diseases. The presented review summarizes the knowledge of the aetiology of acute leukaemias published since 2000. It describes their epidemiological characteristics and risk factors and outlines the possibilities for their prevention.


Asunto(s)
Leucemia Mieloide Aguda , República Checa/epidemiología , Humanos , Incidencia , Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/prevención & control , Factores de Riesgo
4.
Rev. cuba. hematol. inmunol. hemoter ; 37(3): e1428, 2021. tab
Artículo en Español | LILACS, CUMED | ID: biblio-1341401

RESUMEN

Introducción: La enfermedad mínima residual es la permanencia de células leucémicas residuales en niveles subclínicos luego de la remisión de la enfermedad. Esta condición incrementa el riesgo de recaída y mortalidad. Objetivo: Caracterizar factores clínicos y moleculares de pacientes con leucemias agudas y enfermedad mínima residual detectada por citometría de flujo en una institución de alta complejidad de la ciudad de Medellín, Colombia durante los años 2015 - 2017. Metodología: Este es un estudio descriptivo retrospectivo, que incluyó pacientes con leucemia diagnosticada por citometría de flujo. Se realizó un muestreo no probabilístico de casos consecutivos. La información recolectada fue digitada en una base de datos en Excel, y el análisis se realizó a través del programa IBM SPSS Versión 24, empleando según la naturaleza de cada variable frecuencias absolutas y relativas, promedio y desviación estándar o mediana y rangos intercuartílicos según su distribución. Resultados: Se incluyó un total de 60 pacientes con predominio del sexo masculino 63,3 por ciento (38). El diagnóstico más frecuente fue la leucemia linfoide 78,3 por ciento (47). Del total de pacientes incluidos, 36,6 por ciento (22) fue positivo para enfermedad mínima residual; 28,3 por ciento recibió trasplante de médula ósea y el 10 por ciento (6) presentó compromiso de líquido cefalorraquídeo. En la segunda citometría en pacientes con enfermedad mínima residual, 90,9 por ciento (20) expresaba CD45+. El 31,8 por ciento (7) de los pacientes con enfermedad mínima residual presentó recaída. Conclusión: La enfermedad mínima residual es una condición frecuente en pacientes con leucemias agudas que requiere seguimiento y constituye un factor pronóstico relevante(AU)


Introduction: The minimal residual disease is the permanence of residual leukemic cells at subclinical levels after remission of the disease. This condition increases the risk of relapse and mortality. Objective: To characterize the clinical and molecular factors of patients with acute leukemias and minimal residual disease detected by flow cytometry in a highly complex institution in the city of Medellín, Colombia during the years 2015 - 2017. Methodology: This is a retrospective descriptive observational study, which included patients with leukemia diagnosed by flow cytometry. A non-probabilistic sampling of consecutive cases was carried out. The information collected was entered into a database in Excel, and the analysis was carried out through the IBM SPSS Version 24 program, using absolute and relative frequencies, average and standard deviation or median and interquartile ranges, according to the nature of each variable and its distribution. Results: 60 patients were included in which male sex predominated with 63.3 percent (38). The most frequent diagnosis was lymphoid leukemia with 78.3 percent (47). Of the total patients included, 36.6 percent (22) were positive for minimal residual disease; 28.3 percent received a bone marrow transplant and 10 percent (6) had a cerebrospinal fluid compromise. In the second cytometry of the patients with minimal residual disease, 90.9 percent (20) expressed CD45 +. 31.8 percent (7) of the patients with minimal residual disease relapsed. Conclusion: Minimal residual disease is a frequent pathology in patients with acute leukemias that requires follow-up and constitutes a relevant prognostic factor(AU)


Asunto(s)
Humanos , Masculino , Femenino , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/prevención & control , Neoplasia Residual/diagnóstico , Citometría de Flujo/métodos , Epidemiología Descriptiva , Estudios Retrospectivos
5.
Cancer Cell ; 39(7): 958-972.e8, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34048709

RESUMEN

N6-Methyladenosine (m6A) on mRNAs mediates different biological processes and its dysregulation contributes to tumorigenesis. How m6A dictates its diverse molecular and cellular effects in leukemias remains unknown. We found that YTHDC1 is the essential m6A reader in myeloid leukemia from a genome-wide CRISPR screen and that m6A is required for YTHDC1 to undergo liquid-liquid phase separation and form nuclear YTHDC1-m6A condensates (nYACs). The number of nYACs increases in acute myeloid leukemia (AML) cells compared with normal hematopoietic stem and progenitor cells. AML cells require the nYACs to maintain cell survival and the undifferentiated state that is critical for leukemia maintenance. Furthermore, nYACs enable YTHDC1 to protect m6A-mRNAs from the PAXT complex and exosome-associated RNA degradation. Collectively, m6A is required for the formation of a nuclear body mediated by phase separation that maintains mRNA stability and control cancer cell survival and differentiation.


Asunto(s)
Adenosina/análogos & derivados , Núcleo Celular/metabolismo , Metilación de ADN , Leucemia Mieloide Aguda/prevención & control , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Mensajero/metabolismo , Adenosina/química , Adenosina/metabolismo , Animales , Apoptosis , Diferenciación Celular , Núcleo Celular/genética , Proliferación Celular , Femenino , Hematopoyesis , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Extracción Líquido-Líquido , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas del Tejido Nervioso/genética , Transición de Fase , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Empalme de ARN/genética , Estabilidad del ARN , ARN Mensajero/química , ARN Mensajero/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Curr Cancer Drug Targets ; 20(7): 473-489, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32357813

RESUMEN

Over the past decade, our increased understanding of the interactions between the immune system and cancer cells has led to paradigm shifts in the clinical management of solid and hematologic malignancies. The incorporation of immune-targeted strategies into the treatment landscape of acute myeloid leukemia (AML), however, has been challenging. While this is in part due to the inability of the immune system to mount an effective tumor-specific immunogenic response against the heterogeneous nature of AML, the decreased immunogenicity of AML cells also represents a major obstacle in the effort to design effective immunotherapeutic strategies. In fact, AML cells have been shown to employ sophisticated escape mechanisms to evade elimination, such as direct immunosuppression of natural killer cells and decreased surface receptor expression leading to impaired recognition by the immune system. Yet, cellular and humoral immune reactions against tumor-associated antigens (TAA) of acute leukemia cells have been reported and the success of allogeneic stem cell transplantation and monoclonal antibodies in the treatment of AML clearly provides proof that an immunotherapeutic approach is feasible in the management of this disease. This review discusses the recent progress and persisting challenges in cellular immunotherapy for patients with AML.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/prevención & control , Animales , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Humanos , Células Asesinas Naturales/inmunología , Leucemia Mieloide Aguda/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T Citotóxicos/inmunología , Trasplante Homólogo/métodos , Escape del Tumor
7.
Blood ; 136(1): 50-60, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32430504

RESUMEN

Our understanding of the genetics of acute myeloid leukemia (AML) development from myelodysplastic syndrome (MDS) has advanced significantly as a result of next-generation sequencing technology. Although differences in cell biology and maturation exist between MDS and AML secondary to MDS, these 2 diseases are genetically related. MDS and secondary AML cells harbor mutations in many of the same genes and functional categories, including chromatin modification, DNA methylation, RNA splicing, cohesin complex, transcription factors, cell signaling, and DNA damage, confirming that they are a disease continuum. Differences in the frequency of mutated genes in MDS and secondary AML indicate that the order of mutation acquisition is not random during progression. In almost every case, disease progression is associated with clonal evolution, typically defined by the expansion or emergence of a subclone with a unique set of mutations. Monitoring tumor burden and clonal evolution using sequencing provides advantages over using the blast count, which underestimates tumor burden, and could allow for early detection of disease progression prior to clinical deterioration. In this review, we outline advances in the study of MDS to secondary AML progression, with a focus on the genetics of progression, and discuss the advantages of incorporating molecular genetic data in the diagnosis, classification, and monitoring of MDS to secondary AML progression. Because sequencing is becoming routine in the clinic, ongoing research is needed to define the optimal assay to use in different clinical situations and how the data can be used to improve outcomes for patients with MDS and secondary AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Recuento de Células , Evolución Clonal , Metilación de ADN/efectos de los fármacos , Análisis Mutacional de ADN , Progresión de la Enfermedad , Epigénesis Genética , Células Madre Hematopoyéticas/patología , Humanos , Lenalidomida/farmacología , Lenalidomida/uso terapéutico , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/prevención & control , Mutación , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/patología , Mielopoyesis , Células Madre Neoplásicas/patología , Pronóstico , Carga Tumoral , Secuenciación del Exoma , Secuenciación Completa del Genoma
8.
Blood ; 136(6): 674-683, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32285126

RESUMEN

This phase 2 study was designed to compare systemic decitabine exposure, demethylation activity, and safety in the first 2 cycles with cedazuridine 100 mg/decitabine 35 mg vs standard decitabine 20 mg/m2 IV. Adults with International Prognostic Scoring System intermediate-1/2- or high-risk myelodysplastic syndromes (MDS) or chronic myelomonocytic leukemia (CMML) were randomized 1:1 to receive oral cedazuridine/decitabine or IV decitabine in cycle 1, followed by crossover to the other treatment in cycle 2. All patients received oral cedazuridine/decitabine in subsequent cycles. Cedazuridine and decitabine were given initially as separate capsules in a dose-confirmation stage and then as a single fixed-dose combination (FDC) tablet. Primary end points: mean decitabine systemic exposure (geometric least-squares mean [LSM]) of oral/IV 5-day area under curve from time 0 to last measurable concentration (AUClast), percentage long interspersed nuclear element 1 (LINE-1) DNA demethylation for oral cedazuridine/decitabine vs IV decitabine, and clinical response. Eighty patients were randomized and treated. Oral/IV ratios of geometric LSM 5-day AUClast (80% confidence interval) were 93.5% (82.1-106.5) and 97.6% (80.5-118.3) for the dose-confirmation and FDC stages, respectively. Differences in mean %LINE-1 demethylation between oral and IV were ≤1%. Clinical responses were observed in 48 patients (60%), including 17 (21%) with complete response. The most common grade ≥3 adverse events regardless of causality were neutropenia (46%), thrombocytopenia (38%), and febrile neutropenia (29%). Oral cedazuridine/decitabine (100/35 mg) produced similar systemic decitabine exposure, DNA demethylation, and safety vs decitabine 20 mg/m2 IV in the first 2 cycles, with similar efficacy. This study is registered at www.clinicaltrials.gov as #NCT02103478.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Bajo la Curva , Cápsulas , Estudios Cruzados , Metilación de ADN/efectos de los fármacos , ADN-Citosina Metilasas/antagonistas & inhibidores , Decitabina/administración & dosificación , Decitabina/efectos adversos , Decitabina/farmacocinética , Decitabina/farmacología , Progresión de la Enfermedad , Combinación de Medicamentos , Monitoreo de Drogas , Femenino , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Hematológicas/inducido químicamente , Humanos , Estimación de Kaplan-Meier , Análisis de los Mínimos Cuadrados , Leucemia Mieloide Aguda/prevención & control , Elementos de Nucleótido Esparcido Largo/efectos de los fármacos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/antagonistas & inhibidores , Comprimidos , Uridina/administración & dosificación , Uridina/efectos adversos , Uridina/análogos & derivados , Uridina/farmacocinética , Uridina/farmacología
9.
J Mater Chem B ; 8(10): 2063-2081, 2020 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-32068215

RESUMEN

There is currently no effective treatment for acute myeloid leukemia, and surgery is also ineffective as an important treatment for most tumors. Rapidly developing artificial intelligence technology can be applied to different aspects of drug development, and it plays a key role in drug discovery. Based on network pharmacology and virtual screening, candidates were selected from the molecular database. Nine artificial intelligence algorithm models were used to further verify the candidates' potential. The 350 training results of the deep learning model showed higher credibility, and the R-square of the training set and test set of the optimal model reached 0.89 and 0.84, respectively. The random forest model has an R-square of 0.91 and a mean square error of only 0.003. The R-square of the Adaptive Boosting model and the Bagging model reached 0.92 and 0.88, respectively. Molecular dynamics simulation evaluated the stability of the ligand-protein complex and achieved good results. Artificial intelligence models had unearthed the promising candidates for STAT3 inhibitors, and the good performance of most models showed that they still had practical value on small data sets.


Asunto(s)
Inteligencia Artificial , Descubrimiento de Drogas/métodos , Leucemia Mieloide Aguda/tratamiento farmacológico , Bases de Datos de Compuestos Químicos , Humanos , Leucemia Mieloide Aguda/prevención & control , Ligandos , Aprendizaje Automático , Simulación de Dinámica Molecular , Unión Proteica , Factor de Transcripción STAT3/antagonistas & inhibidores
10.
Pediatr Hematol Oncol ; 37(3): 259-268, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32028812

RESUMEN

Relapse of acute myeloblastic leukemia (AML) after first allogenic hematopoietic stem-cell transplantation (allo-HSCT) is a fatal complication. Sixty-five children transplanted for AML were included in a prospective national study from June 2005 to July 2008 to explore the feasibility of preemptive immune modulation based on the monitoring of blood chimerism. Relapse occurred in 23 patients (35%). The median time between the last complete chimerism and relapse was 13.5 days (2-138). Prompt discontinuation of cyclosporin and the administration of donor lymphocyte infusions (DLIs) based on chimerism monitoring failed as a preemptive tool, either for detecting relapse or certifying long-term remission.


Asunto(s)
Ciclosporina/administración & dosificación , Trasplante de Células Madre Hematopoyéticas , Inmunomodulación , Leucemia Mieloide Aguda , Transfusión de Linfocitos , Donantes de Tejidos , Quimera por Trasplante/sangre , Aloinjertos , Niño , Ciclosporina/efectos adversos , Femenino , Humanos , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/prevención & control , Masculino , Estudios Prospectivos , Recurrencia
11.
Blood Cancer J ; 10(2): 16, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029705

RESUMEN

Large-scale chromosomal translocations are frequent oncogenic drivers in acute myeloid leukemia (AML). These translocations often occur in critical transcriptional/epigenetic regulators and contribute to malignant cell growth through alteration of normal gene expression. Despite this knowledge, the specific gene expression alterations that contribute to the development of leukemia remain incompletely understood. Here, through characterization of transcriptional regulation by the RUNX1-ETO fusion protein, we have identified Ras-association domain family member 2 (RASSF2) as a critical gene that is aberrantly transcriptionally repressed in t(8;21)-associated AML. Re-expression of RASSF2 specifically inhibits t(8;21) AML development in multiple models. Through biochemical and functional studies, we demonstrate RASSF2-mediated functions to be dependent on interaction with Hippo kinases, MST1 and MST2, but independent of canonical Hippo pathway signaling. Using proximity-based biotin labeling we define the RASSF2-proximal proteome in leukemia cells and reveal association with Rac GTPase-related proteins, including an interaction with the guanine nucleotide exchange factor, DOCK2. Importantly, RASSF2 knockdown impairs Rac GTPase activation, and RASSF2 expression is broadly correlated with Rac-mediated signal transduction in AML patients. Together, these data reveal a previously unappreciated mechanistic link between RASSF2, Hippo kinases, and Rac activity with potentially broad functional consequences in leukemia.


Asunto(s)
Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/prevención & control , Proteínas de Fusión Oncogénica/metabolismo , Translocación Genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Biomarcadores de Tumor/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Fusión Oncogénica/genética , ARN Largo no Codificante , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP rac/genética
12.
Science ; 367(6477): 586-590, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32001657

RESUMEN

The initiating mutations that contribute to cancer development are sometimes present in premalignant cells. Whether therapies targeting these mutations can eradicate premalignant cells is unclear. Acute myeloid leukemia (AML) is an attractive system for investigating the effect of preventative treatment because this disease is often preceded by a premalignant state (clonal hematopoiesis or myelodysplastic syndrome). In Npm1c/Dnmt3a mutant knock-in mice, a model of AML development, leukemia is preceded by a period of extended myeloid progenitor cell proliferation and self-renewal. We found that this self-renewal can be reversed by oral administration of a small molecule (VTP-50469) that targets the MLL1-Menin chromatin complex. These preclinical results support the hypothesis that individuals at high risk of developing AML might benefit from targeted epigenetic therapy in a preventative setting.


Asunto(s)
Terapia Genética/métodos , Leucemia Experimental/prevención & control , Leucemia Mieloide Aguda/prevención & control , Proteínas Nucleares/genética , Preleucemia/terapia , Animales , Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Técnicas de Sustitución del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Experimental/genética , Leucemia Mieloide Aguda/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Células Progenitoras Mieloides/patología , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Nucleofosmina , Preleucemia/genética , Preleucemia/patología , Proteínas Proto-Oncogénicas/metabolismo
13.
Curr Oncol Rep ; 22(1): 4, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974774

RESUMEN

PURPOSE OF REVIEW: To understand how myelodysplastic syndromes (MDS) transform to AML and to describe how transformation can be predicted and prevented. RECENT FINDINGS: Recent genomic analyses have shown that MDS progression to AML is associated with clonal expansion and clonal evolution. Mutation profiles of MDS change during progression and new mutations in signaling genes and transcription factors emerge. AML transformation can be predicted by several parameters including International Prognostic Scoring System IPSS risk category and transfusion requirements. The prognostic relevance of the acquisition of some gene mutations (i.e., IDH1 and 2, CBL, FT3, RAS, NPM1, TP53, and ASXL1) has to be prospectively validated. The most effective preventive therapy for AML transformation is allogeneic stem cell transplantation. Hypomethylating agents have been associated with prolonged time to AML transformation even in patients who did not achieve an objective response. The recent progress in the understanding of the molecular events leading to transformation and the event of new effective therapies open new avenues for a better prediction and prevention of AML transformation in patients with MDS.


Asunto(s)
Transformación Celular Neoplásica/patología , Leucemia Mieloide Aguda/prevención & control , Síndromes Mielodisplásicos/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Metilación de ADN , Progresión de la Enfermedad , Humanos , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/patología , Mutación , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/metabolismo , Nucleofosmina , Factores de Riesgo
14.
Blood ; 135(3): 167-180, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31805184

RESUMEN

NF-κB is a key regulator of inflammation and cancer progression, with an important role in leukemogenesis. Despite its therapeutic potential, targeting NF-κB using pharmacologic inhibitors has proven challenging. Here, we describe a myeloid cell-selective NF-κB inhibitor using an miR-146a mimic oligonucleotide conjugated to a scavenger receptor/Toll-like receptor 9 agonist (C-miR146a). Unlike an unconjugated miR146a, C-miR146a was rapidly internalized and delivered to the cytoplasm of target myeloid cells and leukemic cells. C-miR146a reduced expression of classic miR-146a targets (IRAK1 and TRAF6), thereby blocking activation of NF-κB in target cells. IV injections of C-miR146a mimic to miR-146a-deficient mice prevented excessive NF-κB activation in myeloid cells, and thus alleviated myeloproliferation and mice hypersensitivity to bacterial challenge. Importantly, C-miR146a showed efficacy in dampening severe inflammation in clinically relevant models of chimeric antigen receptor (CAR) T-cell-induced cytokine release syndrome. Systemic administration of C-miR146a oligonucleotide alleviated human monocyte-dependent release of IL-1 and IL-6 in a xenotransplanted B-cell lymphoma model without affecting CD19-specific CAR T-cell antitumor activity. Beyond anti-inflammatory functions, miR-146a is a known tumor suppressor commonly deleted or expressed at reduced levels in human myeloid leukemia. Using The Cancer Genome Atlas acute myeloid leukemia data set, we found an inverse correlation of miR-146a levels with NF-κB-related genes and with patient survival. Correspondingly, C-miR146a induced cytotoxic effects in human MDSL, HL-60, and MV4-11 leukemia cells in vitro. The repeated IV administration of C-miR146a inhibited expression of NF-κB target genes and thereby thwarted progression of disseminated HL-60 leukemia. Our results show the potential of using myeloid cell-targeted miR-146a mimics for the treatment of inflammatory and myeloproliferative disorders.


Asunto(s)
Síndrome de Liberación de Citoquinas/prevención & control , Inflamación/prevención & control , Leucemia Mieloide Aguda/prevención & control , MicroARNs/genética , Células Progenitoras Mieloides/patología , FN-kappa B/metabolismo , Animales , Apoptosis , Proliferación Celular , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Células Progenitoras Mieloides/metabolismo , FN-kappa B/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Blood ; 135(1): 56-70, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31697837

RESUMEN

Lineage-defining transcription factors (TFs) are compelling targets for leukemia therapy, yet they are among the most challenging proteins to modulate directly with small molecules. We previously used CRISPR screening to identify a salt-inducible kinase 3 (SIK3) requirement for the growth of acute myeloid leukemia (AML) cell lines that overexpress the lineage TF myocyte enhancer factor (MEF2C). In this context, SIK3 maintains MEF2C function by directly phosphorylating histone deacetylase 4 (HDAC4), a repressive cofactor of MEF2C. In this study, we evaluated whether inhibition of SIK3 with the tool compound YKL-05-099 can suppress MEF2C function and attenuate disease progression in animal models of AML. Genetic targeting of SIK3 or MEF2C selectively suppressed the growth of transformed hematopoietic cells under in vitro and in vivo conditions. Similar phenotypes were obtained when cells were exposed to YKL-05-099, which caused cell-cycle arrest and apoptosis in MEF2C-expressing AML cell lines. An epigenomic analysis revealed that YKL-05-099 rapidly suppressed MEF2C function by altering the phosphorylation state and nuclear localization of HDAC4. Using a gatekeeper allele of SIK3, we found that the antiproliferative effects of YKL-05-099 occurred through on-target inhibition of SIK3 kinase activity. Based on these findings, we treated 2 different mouse models of MLL-AF9 AML with YKL-05-099, which attenuated disease progression in vivo and extended animal survival at well-tolerated doses. These findings validate SIK3 as a therapeutic target in MEF2C-addicted AML and provide a rationale for developing druglike inhibitors of SIK3 for definitive preclinical investigation and for studies in human patients.


Asunto(s)
Compuestos de Anilina/farmacología , Leucemia Mieloide Aguda/prevención & control , Factores de Transcripción MEF2/metabolismo , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis , Ciclo Celular , Proliferación Celular , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Factores de Transcripción MEF2/genética , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Semin Oncol Nurs ; 35(6): 150958, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31759817

RESUMEN

OBJECTIVES: Shared decision-making (SDM) is the gold standard approach to cancer treatment decision-making in the 21st century, but it is frequently misunderstood, and many clinicians do not know how to operationalize the SDM framework in their busy practices. Here we review the principles behind SDM, discuss unique aspects of acute myeloid leukemia (AML) that complicate the decision-making process, and provide one recommended framework for how to implement SDM into practice. DATA SOURCES: Published literature and clinical experiences. CONCLUSION: AML poses unique challenges to treatment decision-making. These challenges can be effectively addressed by following the SDM framework in practice. IMPLICATIONS FOR NURSING PRACTICE: Nurses can play an important role in the AML treatment decision-making process. Being on the front lines of care, working most directly with patients and families, nurses are best positioned to assess understanding after treatment discussions take place, detect emotional distress, and provide empathic support as part of the SDM process.


Asunto(s)
Vías Clínicas/organización & administración , Leucemia Mieloide Aguda/prevención & control , Participación del Paciente/métodos , Atención Dirigida al Paciente/métodos , Humanos , Leucemia Mieloide Aguda/terapia , Oncología Médica/normas
18.
EMBO J ; 38(20): e102096, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31483066

RESUMEN

Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53-/- mice. Surprisingly, stabilization of p53R178E in Mdm2-/- mice nevertheless triggers extensive apoptosis, indicative of residual wild-type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E ;Mdm2-/- embryos, it proves insufficient for suppression of spontaneous and oncogene-driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53-/- mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53-/- ones, resulting in enhanced eradication of p53-mutated tumor cells. Together, this provides genetic proof-of-principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia Mieloide Aguda/prevención & control , Linfoma/prevención & control , Mutación , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinogénesis/patología , Ciclo Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Linfoma/genética , Linfoma/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...