Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cancer Lett ; 598: 217125, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39084456

RESUMEN

DIAPH1, a member of the formins family and a Rho effector, was found to be involved in thrombocytopoiesis, and the process of MDS in mice with unknown pathogenesis. In this study, we reported a preliminary study about the heterogeneity in the clinical features and outcomes of DIAPH1 mutations in MDS. DIAPH1 frameshift mutations were identified in 20 out of 88 MDS patients, including 11 frameshift mutations locating at 140892588-141000567 (5q31.3), which causes structure changes at FH1 domain. DIAPH1 mutated cases were correlated with lower megakaryocyte dysplasia in lower-risk patients (IPSS-M score <0) at first diagnosis, and higher megakaryocyte counts pre-transplant. The megakaryopoiesis-related genes: GP1BA and SETBP1 mutation were positively and negatively associated with DIAPH1 mutation, respectively. DIAPH1 mutated cases showed superior overall survival of all patients and low-risk cohorts. In conclusion, we found DIAPH1 frameshift mutations are implicated in megakaryopoiesis of MDS and correlated with superior prognosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Forminas , Mutación del Sistema de Lectura , Síndromes Mielodisplásicos , Humanos , Forminas/genética , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Femenino , Masculino , Persona de Mediana Edad , Anciano , Pronóstico , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Megacariocitos/patología , Megacariocitos/metabolismo , Proteínas Nucleares/genética , Proteínas Portadoras/genética , Anciano de 80 o más Años , Mutación
2.
Am J Hematol ; 98(6): 881-889, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36880836

RESUMEN

Conditioning therapy is an essential procedure prior to hematopoietic stem cell transplant (HSCT), imposing a great impact on the outcomes of recipients. We performed a prospective randomized controlled trial to assess the outcome of HSCT recipients with myeloid malignancies after receiving the conditioning therapy consisting of modified BUCY (mBUCY), N-acetyl-L-cysteine (NAC), and decitabine. Enrolled patients were randomly allocated to either Arm A (decitabine, day -12 to -10; NAC, day -9 to +30; mBUCY, day -9 to -2), or Arm B (mBUCY regimen followed by stem cells infusion). Seventy-six patients in Arm A and 78 patients in Arm B were finally evaluated. The results showed platelet recovery accelerate in Arm A, with more patients achieving a platelet count of ≥50 × 109 /L than Arm B at day +30 and +60 (p = .004 and .043, respectively). The cumulative incidence of relapse is 11.8% (95% CI 0.06-0.22) in Arm A, and 24.4% (95% CI 0.16-0.35) in Arm B (p = .048). The estimated 3-year overall survival rate was 86.4% (±4.4%) and 79.9% (±4.7%) in 2 arms, respectively (p = .155). EFS at 3 years was 79.2% (±4.9%) in Arm A and 60.0% (±5.9%) in Arm B (p = .007). Intracellular reactive oxygen species (ROS) level was found to be reversely correlated with platelet recovery, and fewer patients in Arm A displayed excessive ROS within hematopoietic progenitor cells compared to Arm B. In conclusion, the addition of decitabine and NAC to mBUCY is a feasible and promising conditioning therapy for myeloid malignancies patients.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Trastornos Mieloproliferativos , Neoplasias , Humanos , Decitabina , Acetilcisteína/uso terapéutico , Busulfano , Estudios Prospectivos , Especies Reactivas de Oxígeno , Trastornos Mieloproliferativos/complicaciones , Trasplante de Células Madre Hematopoyéticas/métodos , Terapia Conductista , Neoplasias/complicaciones , Acondicionamiento Pretrasplante/efectos adversos , Leucemia Mieloide Aguda/terapia , Enfermedad Injerto contra Huésped/etiología
3.
Clin Epigenetics ; 15(1): 54, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36991512

RESUMEN

BACKGROUND: DNA methylation is a form of epigenetic modification that regulates gene expression. However, there are limited data on the comprehensive analysis of DNA methylation regulated gene mutations (DMRGM) in acute myeloid leukemia (AML) mainly referring to DNA methyltransferase 3α (DNMT3A), isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), and Tet methylcytidine dioxygenase 2 (TET2). RESULTS: A retrospective study of the clinical characteristics and gene mutations in 843 newly diagnosed non-M3 AML patients was conducted between January 2016 and August 2019. 29.7% (250/843) of patients presented with DMRGM. It was characterized by older age, higher white blood cell count, and higher platelet count (P < 0.05). DMRGM frequently coexisted with FLT3-ITD, NPM1, FLT3-TKD, and RUNX1 mutations (P < 0.05). The CR/CRi rate was only 60.3% in DMRGM patients, significantly lower than in non-DMRGM patients (71.0%, P = 0.014). In addition to being associated with poor overall survival (OS), DMRGM was also an independent risk factor for relapse-free survival (RFS) (HR: 1.467, 95% CI: 1.030-2.090, P = 0.034). Furthermore, OS worsened with an increasing burden of DMRGM. Patients with DMRGM may be benefit from hypomethylating drugs, and the unfavorable prognosis of DMRGM can be overcome by hematopoietic stem cell transplantation (HSCT). For external validation, the BeatAML database was downloaded, and a significant association between DMRGM and OS was confirmed (P < 0.05). CONCLUSION: Our study provides an overview of DMRGM in AML patients, which was identified as a risk factor for poor prognosis.


Asunto(s)
Metilación de ADN , Leucemia Mieloide Aguda , Humanos , Pronóstico , Estudios Retrospectivos , Isocitrato Deshidrogenasa/genética , Nucleofosmina , Mutación , Leucemia Mieloide Aguda/genética , Metilasas de Modificación del ADN/genética , Genes Reguladores
4.
J Thromb Haemost ; 20(12): 2998-3010, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36128771

RESUMEN

BACKGROUND AND OBJECTIVES: Platelets are affected by many factors, such as infectious or aseptic inflammation, and different inflammatory states may induce either thrombocytopenia or thrombocytosis. Tumor necrosis factor α (TNFα) is an important inflammatory cytokine that has been shown to affect the activity of hematopoietic stem cells. However, its role in megakaryocyte (MK) development and platelet production remains largely unknown. This study aimed to investigate the effects of TNFα on MK and platelet generation. METHODS AND RESULTS: The ex vivo study with human CD34+ cells demonstrated that TNFα differentially modulated commitment toward the MK lineage. Specifically, a low concentration of 0.5 ng/ml TNFα promoted MK maturation, proplatelet formation, and platelet production, whereas a high concentration of 10 ng/ml or more TNFα exhibited a substantial inhibitory effect on MK and platelet production. The distinct effect of TNFα on MKs was mainly dependent on TNFα receptor 1. TNFα differentially regulated the MAPK-ERK1/2 signaling pathway and the cytoskeletal proteins cofilin and MLC2. The in vivo study with Balb/c mice indicated that low-dose or high-dose TNFα administration differentially affected short-term platelet recovery after bone marrow transplantation. CONCLUSIONS: Our study revealed distinct roles for TNFα in megakaryopoiesis and thrombopoiesis and may provide new insights regarding the treatment for platelet disorders.


Asunto(s)
Trombopoyesis , Factor de Necrosis Tumoral alfa , Ratones , Animales , Humanos , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Megacariocitos/metabolismo , Citocinas/metabolismo , Plaquetas/metabolismo
5.
Transplant Cell Ther ; 28(11): 764.e1-764.e7, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35940529

RESUMEN

Transplantation-associated thrombotic microangiopathy (TA-TMA) is a life-threatening complication for patients undergoing hematopoietic stem cell transplantation (HSCT). N-acetylcysteine (NAC) has recently been considered as a potential treatment for patients with thrombotic thrombocytopenic purpura. To assess the value of NAC for the prevention of TA-TMA, we conducted a prospective study at the First Affiliated Hospital of Soochow University. This open-label, randomized placebo-controlled trial included 160 patients who were scheduled for allogeneic HSCT. Participants were assigned at random 1:1 to either oral NAC (50 mg/kg/day from 9 days before HSCT to 30 days after HSCT) or placebo treatment. The primary outcome was the incidence of TA-TMA. Overall survival (OS) and event-free survival (EFS) were assessed in the NAC and placebo control groups. The incidence of TA-TMA was 9.1% (95% confidence interval [CI], 2% to 16.2%) in the NAC group, compared with 23% (95% CI, 13.2% to 32.8%) in the control group, with a rate ratio of .34 (95% CI, .123 to .911; P = .039). The median time to the onset of TA-TMA was 60 days (interquartile range [IQR], 42 to 129 days) in the NAC group and 36 days (IQR, 30.5 to 51 days) in the control group (P = .063). The 2-year OS rate was 75.4% (95% CI, 28.65% to 73.53%) in the NAC group and 63.0% (95% CI, 50.8% to 73.5%) in the control group, with a hazard ratio (HR) of .622 (95% CI, .334-1.155; P = .132). The EFS rate was 25.8% in the NAC patients and 8.1% in controls (HR, .254; 95% CI, .094 to .692; P = .024). The median time of EFS was 60 days in the NAC group and 38 days in controls. Our findings suggest that NAC may be a potential treatment to reduce the incidence of TA-TMA.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Microangiopatías Trombóticas , Humanos , Acetilcisteína/uso terapéutico , Estudios Prospectivos , Estudios Retrospectivos , Microangiopatías Trombóticas/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos
6.
Br J Haematol ; 199(4): 603-615, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35864790

RESUMEN

Transplantation-associated thrombotic microangiopathy (TA-TMA) is a severe complication of haematopoietic stem cell transplantation (HSCT). Complement activation is involved in the development of TA-TMA. However, the underlying mechanism is unclear. Therefore, 21 samples of TA-TMA and 1:1 matched controls were measured for hypoxia-inducible factor-1α (HIF-1α) and complement protein. The mechanism was investigated both in vitro and in vivo. In this study, we found that levels of HIF-1α were significantly higher in TA-TMA patients than that in non-TA-TMA controls. Upregulation of HIF-1α induced an increase in membrane-bound complement C3 and dysfunction of human umbilical vein endothelial cells (HUVECs) in vitro. Increasing HIF-1α in vivo led to C3 and C5b-9 deposition in the glomerular endothelial capillary complex, thrombocytopenia, anaemia, and increased serum lactate dehydrogenase (LDH) levels in wild-type (WT) but not in C3-/- mice subjected to HSCT. High platelet aggregation in peripheral blood and CD41-positive microthrombi in the kidney were also found in dimethyloxallyl glycine (DMOG)-treated mice, recapitulating the TA-TMA phenotype seen in patients. Comprehensive analysis, including DNA array, luciferase reporter assay, chromatin immunoprecipitation (ChIP)-seq, and quantitative polymerase chain reaction (PCR), revealed that HIF-1α interacted with the promoter of complement factor H (CFH) to inhibit its transcription. Decreased CFH led to complement activation in endothelial cells.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Microangiopatías Trombóticas , Humanos , Ratones , Animales , Regulación hacia Arriba , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Endoteliales , Microangiopatías Trombóticas/etiología , Activación de Complemento , Trasplante de Células Madre Hematopoyéticas/efectos adversos
7.
Ann Hematol ; 101(6): 1295-1309, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35357522

RESUMEN

Allogeneic hematopoietic stem cell transplantation is the only curative therapy for patients with myelodysplastic syndrome. Transplantation-associated thrombotic microangiopathy (TA-TMA) remains a cause of death after transplantation. This study assessed the risk factors of TA-TMA and established a prediction model for this complication. We launched a real-world study from 303 MDS patients after allo-HSCT from Dec 1, 2007, to Jun 1, 2018. Logistic regression was used to analyze risk factors and to establish a nomogram. The accuracy of the model was assessed by C-index and calibration curve. TA-TMA class was associated with an over twofold increase in the risk of death (HR 2.66, 95% CI 1.39-5.09, p = 0.003). Stage III or IV acute graft-versus-host disease (aGVHD) (OR: 6.17, 95% CI: 2.19-17.18, p < 0.001) and occurrence time of aGVHD were the risk factors for TA-TMA. Next, we put these two variants and the other three variants into the prediction model via multivariate Lasso regression. In order to quantify the contribution of each factor, a nomogram was generated and displayed (C index of 0.783). TA-TMA predicts worsened outcomes of overall survival. A cross-validated multivariate score including aGVHD occurrence showed excellent concordance and efficacy of predicting TA-TMA in HSCT patients.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Síndromes Mielodisplásicos , Microangiopatías Trombóticas , Enfermedad Injerto contra Huésped/epidemiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Síndromes Mielodisplásicos/complicaciones , Síndromes Mielodisplásicos/terapia , Estudios Retrospectivos , Factores de Riesgo , Microangiopatías Trombóticas/epidemiología , Microangiopatías Trombóticas/etiología
8.
Transplant Cell Ther ; 28(4): 216.e1-216.e5, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34979328

RESUMEN

Transplant-associated thrombotic microangiopathy (TA-TMA) has a high mortality rate and lacks effective treatments. We searched the GEO database and analyzed RNA-seq data and whole-genome sequencing data from patients' blood samples. We identified N-acetyl-L-cysteine (NAC) as a possible therapeutic target for TA-TMA. In vitro experiments showed that NAC reduced complement activation and VWF multimerization in HUVECs. We also treated a 36-year-old female TA-TMA patient with NAC. Hemoglobin, platelet counts, lactate dehydrogenase levels, and sC5b-9 levels and schistocytes were normalized after using NAC. It shows that NAC may be an effective drug to improve TA-TMA symptoms by inhibiting complement activation.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Microangiopatías Trombóticas , Acetilcisteína/farmacología , Adulto , Activación de Complemento , Femenino , Humanos , Microangiopatías Trombóticas/tratamiento farmacológico , Resultado del Tratamiento
9.
Cancer Med ; 11(5): 1413-1426, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35023630

RESUMEN

BACKGROUND: Previous studies have confirmed the existence of epigenetic regulation of immune responses in acute myeloid leukemia. However, the potential role of RNA N6-methyladenosine (m6A) remodeling in tumor microenvironment (TME) infiltration remains unclear. METHODS AND MATERIALS: m6A patterns of 469 AML patients (420 of which provided survival data) based on 18 m6A regulators were systematically evaluated. Based on the expression of 18 m6A regulators, unsupervised agglomerative cluster analysis was applied to recognize the various m6A modification types and to classify patients. We linked these patterns to TME infiltration characteristics and identified three distinct populations of m6A modifications. RESULTS: These three TME cell infiltration patterns are characterized by a high degree of concordance with the three tumor immunophenotypes, which include immunoinflammatory, immunorejection, and immune inert patterns. We showed that assessment of m6A modification patterns within individually neoplasms can forecast the stage of neoplasmic inflammation, TME basal activity, subtype, hereditary mutation, and clinical patient prognosis. Limited low m6Ascore, featuring increased mutational load and immune activation, indicates an inflammatory phenotype of TME with a 5-year survival rate at 14.4% compared to the high-m6Ascore group (40.9%). CONCLUSIONS: Data from two different cohorts demonstrated that a higher m6Ascore showed a marked therapeutic superiority as well as clinical advantage. Assessing m6A modification patterns in AML patients could improve our knowledge of the TME infiltrative profile as well as directing effective immunotherapeutic approaches.


Asunto(s)
Leucemia Mieloide Aguda , Microambiente Tumoral , Epigénesis Genética , Humanos , Leucemia Mieloide Aguda/genética , Metilación , Pronóstico , Microambiente Tumoral/genética
10.
Hematol Oncol ; 40(1): 63-71, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34606093

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy has shown excellent clinical efficacy in patients with hematologic malignancies. However, severe bleeding after this treatment is a life-threatening complication for most patients. This study evaluated the risk factors associated with bleeding in CAR T treatment and developed a predictive model for this complication. Analysis performed in the First Affiliated Hospital of Suzhou University and external validation launched in Suzhou Hongci Hematology Hospital (Jiangsu, China). We conducted a real-world study incorporating data from 400 patients with hematologic malignancies treated with CAR T between 1 November 2015 and 1 September 2019. Also, 39 patients from another hospital were selected for external validation. Patients with severe bleeding (hazard ratio [HR] 13.04, 95% confidence interval 5.82-29.18; p < 0.001) had a higher risk of death after CAR T. Stage III and IV cytokine release syndrome (CRS) (odds ratio [OR] 6.07, 95% CI 2.35-16.76; p < 0.001) and higher tumor necrosis factor-α (TNF-α) levels (OR 4.00, 95% CI 1.53-11.35; p < 0.001) were independent factors of bleeding in patients after CAR-T treatment. The predictive model developed by Lasso regression, which selected factors such as CRS period, transfusion volume, platelet percentage, platelet count, thrombinogen time, interleukin 6, and TNF-α levels, and showed Nomogram, yielded excellent agreement (C-statistics = 0.905) with the calibration curve, which improved clinical benefit with respect to established bleeding scores such as outpatient bleeding risk index (MOBRI). External validation was performed using 39 patients from another hospital with an AUC of 0.700. Patients with severe bleeding after Car-T therapy had increased the risk of death. A cross-validated bleeding risk score based on CRS stages and TNF-α level show significant prognostic value in patients undergoing CAR-T treatment.


Asunto(s)
Neoplasias Hematológicas/terapia , Hemorragia/patología , Inmunoterapia Adoptiva/efectos adversos , Factor de Necrosis Tumoral alfa/efectos adversos , Adulto , Femenino , Estudios de Seguimiento , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Hemorragia/etiología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo
11.
Expert Rev Hematol ; 14(11): 1041-1048, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34844489

RESUMEN

Thrombocytopenia is a tough complication after hematopoietic stem cell transplantation (HSCT) with elusive pathogenesis and lack of well-established therapies. Thrombopoietin receptor agonists (TPO-RAs) have been used for thrombocytopenia post HSCT in recent years, but the outcomes remain debatable. We conducted this meta-analysis and systematic-review to evaluate the efficacy and safety of TPO-RAs for platelet recovery after HSCT. We searched PubMed, EMBASE, and Cochrane databases for studies on the application of TPO-RAs (eltrombopag and romiplostim) in the settings of primary or secondary thrombocytopenia after HSCT by 17 March 2021. Efficacy outcomes included response rate and survival rate, and adverse events were also evaluated. A total of 19 studies involving 378 patients were included. The pooled response rate was 73% (95%CI: 68-78%), which was significantly higher than recombinant human thrombopoietin (rhTPO) (27.8%). The pooled survival rate was 66% (95%CI: 54-77%), and infection was found to be the main cause of death. In addition, the pooled rate of adverse events was 3% (95%CI: 1-7%), with no severe adverse events reported. TPO-RAs could effectively and safely promote the recovery of platelets in patients after HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Trombocitopenia , Benzoatos/efectos adversos , Plaquetas , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Receptores Fc , Receptores de Trombopoyetina/agonistas , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes , Trombocitopenia/inducido químicamente , Trombocitopenia/etiología
12.
Cell Death Differ ; 28(11): 3009-3021, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33993195

RESUMEN

The liver has recently been identified as a major organ for destruction of desialylated platelets. However, the underlying mechanism remains unclear. Kupffer cells, which are professional phagocytic cells in the liver, comprise the largest population of resident tissue macrophages in the body. Kupffer cells express a C-type lectin receptor, CLEC4F, that recognizes desialylated glycans with an unclear in vivo role in mediating platelet destruction. In this study, we generated a CLEC4F-deficient mouse model (Clec4f-/-) and found that CLEC4F was specifically expressed by Kupffer cells. Using the Clec4f-/- mice and a newly generated platelet-specific reporter mouse line, we revealed a critical role for CLEC4F on Kupffer cells in mediating destruction of desialylated platelets in the liver in vivo. Platelet clearance experiments and ultrastructural analysis revealed that desialylated platelets were phagocytized predominantly by Kupffer cells in a CLEC4F-dependent manner in mice. Collectively, these findings identify CLEC4F as a Kupffer cell receptor important for the destruction of desialylated platelets induced by bacteria-derived neuraminidases, which provide new insights into the pathogenesis of thrombocytopenia in disease conditions such as sepsis.


Asunto(s)
Receptor de Asialoglicoproteína/metabolismo , Plaquetas/metabolismo , Lectinas Tipo C/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones
13.
Ann Hematol ; 100(9): 2351-2361, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33846855

RESUMEN

Severe bleeding is a major cause of death in acute leukemia (AL) patients with graft-versus-host disease (GVHD) after allogene hematopoietic stem-cell transplantation (allo-HSCT). However, the prognostic value and prediction of HSCT-associated severe bleeding in GVHD patients have not been reported in cohort studies. We did a retrospective analysis of 200 AL patients with GVHD after allo-HSCT from Feb 1, 2014, to Dec 1, 2015. Multivariate analysis showed that the severe bleeding class was associated with the risk of death (HR 2.26, 95% CI 1.31-3.92, p<0.001***). In order to predict severe bleeding and figure out the solution to bleeding events, we established a multiple logistic regression model. HLA-DQB1 unmatching, megakaryocyte reconsititution failure, and III or IV GVHD were the independent risk factors for severe bleeding. Among all the variations above, OR of HLA-DQB1 was the highest (OR: 16.02, 95% CI: 11.54-48.68). Adding HLA-DQB1 to other factors improved the reclassification for predicting severe bleeding (NRI=0.195, z=2.634, p=0.008**; IDI=0.289, z=3.249, p<0.001***). Lasso regression was used to select variants. A nomogram of the logistic model was generated and displayed. Calibration curve demonstrated excellent accuracy in estimating severe bleeding (C index of 0.935). HLA-DQB1 showed excellent efficacy of predicting severe bleeding in HSCT patients.


Asunto(s)
Enfermedad Injerto contra Huésped/etiología , Cadenas beta de HLA-DQ/análisis , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Hemorragia/etiología , Leucemia Mieloide Aguda/terapia , Adulto , Femenino , Humanos , Masculino , Estudios Retrospectivos , Factores de Riesgo , Acondicionamiento Pretrasplante , Trasplante Homólogo/efectos adversos
14.
Blood Adv ; 5(5): 1250-1258, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33646303

RESUMEN

Refractory prolonged isolated thrombocytopenia (RPIT) is an intractable complication after allogeneic hematopoietic cell transplantation (HCT), which often leads to poor prognosis. A clinical study was designed to validate the efficacy and safety of low-dose decitabine for RPIT after HCT and explore the related underlying mechanisms. Eligible patients were randomly allocated to receive 1 of 3 interventions: arm A, low-dose decitabine (15 mg/m2 daily IV for 3 consecutive days [days 1-3]) plus recombinant human thrombopoietin (300 U/kg daily); arm B, decitabine alone; or arm C, conventional treatment. The primary end point was the response rate of platelet recovery at day 28 after treatment. Secondary end points included megakaryocyte count 28 days after treatment and survival during additional follow-up of 24 weeks. Among the 91 evaluable patients, response rates were 66.7%, 73.3%, and 19.4% for the 3 arms, respectively (P < .001). One-year survival rates in arms A (64.4% ± 9.1%) and B (73.4% ± 8.8%) were similar (P = .662), and both were superior to that in arm C (41.0% ± 9.8%; P = .025). Megakaryocytes, endothelial cells (ECs), and cytokines relating to megakaryocyte migration and EC damage were improved in patients responding to decitabine. This study showed low-dose decitabine improved platelet recovery as well as overall survival in RPIT patients after transplantation. This trial was registered at www.clinicaltrials.gov as #NCT02487563.


Asunto(s)
Síndromes Mielodisplásicos , Trombocitopenia , Decitabina , Células Endoteliales , Humanos , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología , Trombopoyetina
15.
J Cancer Res Clin Oncol ; 147(9): 2659-2670, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33550446

RESUMEN

PURPOSE: Genetic changes have prognostic significance in cytogenetically normal acute myeloid leukemia (CN-AML). We set out to evaluate the prognostic of 6 gene mutations in CN-AML. METHODS: We performed a mutational analysis and evaluated prognostic findings of six genes (NPM1, CEBPA, DNMT3A, FLT3-ITD, FLT3-TKD, and C-KIT) in 428 CN-AML patients at our center over 10 years. RESULTS: A total of 282 patients (65.9%) had at least one gene mutation, and the mutation frequencies were as follows: 29.7% (NPM1), 24.1% (CEBPA), 20.1% (FLT3-ITD), 4.0% (FLT3-TKD), 11.9% (DNMT3A), and 4.7% (C-KIT). Multivariate analysis indicated that FLT3-ITDmut and CEBPAwt were independent risk factors correlated with poor overall survival (OS) and disease-free survival (DFS) of CN-AML. Compared with patients who received chemotherapy as consolidation, hematopoietic stem cell transplantation (HSCT) significantly improved OS of CN-AML patients. For standard/high risk patients, HSCT improved both OS and DFS. Combined analysis showed that patients with CEBPAmut/FLT3-ITDwt had the best prognosis, and patients with CEBPAwt/FLT3-ITDmut had the worst OS, with 3-year OS of only 44%. In 212 patients who received HSCT, FLT3-ITD/CEBPA mutations and minimal residual disease (MRD) were correlated with OS and DFS in univariate analysis. CONCLUSIONS: We found that HSCT significantly improves the prognosis of standard/high risk CN-AML patients with superior OS and DFS. Molecular marker analyses, especially combined analysis of the FLT3-ITD and CEBPA status revealed a correlation with the prognosis of CN-AML. For patients who have received HSCT, MRD before transplantation was a strong prognostic marker predicting patient outcome.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Trasplante de Células Madre Hematopoyéticas/mortalidad , Leucemia Mieloide Aguda/mortalidad , Mutación , Neoplasia Residual/mortalidad , Secuencias Repetidas en Tándem , Tirosina Quinasa 3 Similar a fms/genética , Adolescente , Adulto , Anciano , Citogenética , Femenino , Estudios de Seguimiento , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Neoplasia Residual/genética , Neoplasia Residual/patología , Neoplasia Residual/terapia , Nucleofosmina , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia , Trasplante Homólogo , Adulto Joven
16.
Platelets ; 32(5): 677-683, 2021 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-32799612

RESUMEN

Early hemorrhagic death remains a major cause of treatment failure in acute promyelocytic leukemia (APL). This study investigated the role of fibrinogen concentrations in early hemorrhage and overall survival (OS) of APL patients. Laboratory and clinical data, including fibrinogen concentrations and other coagulation indexes, bleeding events, and survival data, of 198 patients newly diagnosed with APL from February 2012 to December 2017 were extracted from patient records and retrospectively investigated. Patients with moderate/severe bleeding had significantly lower median fibrinogen concentrations (p = .023), higher Chinese disseminated intravascular coagulation scoring system (CDSS) (p < .001), and were more often female (p = .034) than patients with no such bleeding. Additionally, patients with fibrinogen <1.0 g/L and 1.0-1.6 g/L had significantly higher moderate/severe bleeding rates than those with fibrinogen >1.6 g/L (p = .015; p = .023). However, moderate/severe (p = .088) and severe bleeding rates (p = .063) were comparable for patients with fibrinogen <1.0 g/L and 1.0-1.6 g/L. Multivariate analysis showed that fibrinogen ≤1.6 g/L (p = .036), platelet counts ≤10 × 109/L (p = .037), and CDSS scores ≥5 (p = .023) were independent risk factors for moderate/severe bleeding. Survival analysis indicated that moderate/severe bleeding (p = .018), fibrinogen ≤1.6 g/L combined with prothrombin time >12.8 s (p = .005), age ≥60 years (p = .001), and CDSS ≥5 (p = .044) were independent predictors of 1-year OS. Fibrinogen ≤1.6 g/L may be an independent risk factor for early bleeding in newly treated patients with APL and is associated with a worse 1-year OS. Increasing fibrinogen to >1.6 g/L may help to prevent bleeding.


Asunto(s)
Fibrinógeno/metabolismo , Hemorragia/sangre , Leucemia Promielocítica Aguda/sangre , Adolescente , Adulto , Anciano , Femenino , Hemorragia/complicaciones , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
18.
Crit Rev Oncol Hematol ; 150: 102944, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32247246

RESUMEN

BACKGROUND: Graft-versus-host disease (GVHD) is a leading cause of death in patients after hematopoietic stem-cell transplantation (HSCT). Previous studies have shown different efficacy of GVHD prophylaxis therapies. METHODS: We reviewed 46 randomized controlled trials (including 8050 participants) systematically from Jun 20, 2004 to Aug 20, 2019. These investigations compared the following drugs or their combination at therapeutic dose range for GVHD after HSCT. The main results were based on the proportion of patients who respond to these therapies. RESULTS: Cyclosporine + methotrexate + Anti-T cell globulin (ATG), tacrolimus + methotrexate + ATG, tacrolimus + bortezomib + sirolimus and cyclosporine + marrow mesenchymal stem cells (MMSCs) were significantly more efficacious than corticosteroids alone (OR: 12.15, 6.71, 6.25, 3.73). corticosteroids alone were less efficacious than all the other GVHD prophylaxis therapies tested. CONCLUSION: Cyclosporine + methotrexate + ATG may be the best choice when starting treatment for GVHD.


Asunto(s)
Suero Antilinfocítico/uso terapéutico , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inmunosupresores/uso terapéutico , Bortezomib/uso terapéutico , Ciclosporina/uso terapéutico , Quimioterapia Combinada , Enfermedad Injerto contra Huésped/inmunología , Humanos , Metotrexato/uso terapéutico , Metaanálisis en Red , Ensayos Clínicos Controlados Aleatorios como Asunto , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico , Trasplante Homólogo , Resultado del Tratamiento
19.
Proc Natl Acad Sci U S A ; 116(28): 14105-14112, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31235575

RESUMEN

Preimplantation genetic testing for aneuploidy (PGT-A) with trophectoderm (TE) biopsy is widely applied in in vitro fertilization (IVF) to identify aneuploid embryos. However, potential safety concerns regarding biopsy and restrictions to only those embryos suitable for biopsy pose limitations. In addition, embryo mosaicism gives rise to false positives and false negatives in PGT-A because the inner cell mass (ICM) cells, which give rise to the fetus, are not tested. Here, we report a critical examination of the efficacy of noninvasive preimplantation genetic testing for aneuploidy (niPGT-A) in the spent culture media of human blastocysts by analyzing the cell-free DNA, which reflects ploidy of both the TE and ICM. Fifty-two frozen donated blastocysts with TE biopsy results were thawed; each of their spent culture medium was collected after 24-h culture and analyzed by next-generation sequencing (NGS). niPGT-A and TE-biopsy PGT-A results were compared with the sequencing results of the corresponding embryos, which were taken as true results for aneuploidy reporting. With removal of all corona-cumulus cells, the false-negative rate (FNR) for niPGT-A was found to be zero. By applying an appropriate threshold for mosaicism, both the positive predictive value (PPV) and specificity for niPGT-A were much higher than TE-biopsy PGT-A. Furthermore, the concordance rates for both embryo ploidy and chromosome copy numbers were higher for niPGT-A than TE-biopsy PGT-A. These results suggest that niPGT-A is less prone to errors associated with embryo mosaicism and is more reliable than TE-biopsy PGT-A.


Asunto(s)
Aneuploidia , Blastocisto/patología , Pruebas Genéticas , Cariotipo , Adulto , Biopsia , Blastocisto/metabolismo , Masa Celular Interna del Blastocisto/patología , Ácidos Nucleicos Libres de Células/genética , Medios de Cultivo/análisis , Femenino , Fertilización In Vitro/normas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pruebas Prenatales no Invasivas/normas , Embarazo , Diagnóstico Preimplantación/normas
20.
Crit Rev Oncol Hematol ; 133: 74-83, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30661660

RESUMEN

SF3B1 gene mutations are the most frequent mutations found in myelodysplastic syndromes (MDS), and the prognostic implication of these mutations remains controversial. We conducted a meta-analysis of studies assessing the prognostic impact and clinical feature of SF3B1 mutations in MDS patients. The overall hazard ratio for overall survival (OS) was 0.90 (95% confidence interval 0.60-1.35, P = 0.61) in MDS patients with SF3B1 mutations compared to those without. Lower leukemia-free survival was associated with SF3B1 mutations. Subgroup analyses showed that Asian cohorts and Illumina HiSeq 2000 methods were significantly associated with OS. Furthermore, SF3B1 mutations were significantly correlated with a lower level of blast cells and a high level of platelet counts and bone marrow ring sideroblasts. Thus, the current meta-analysis suggests that SF3B1 mutations have no significant impact on the OS of MDS patients, and the hematologic parameters of SF3B1 mutations identify a distinct subset of MDS patients with homogeneous features.


Asunto(s)
Mutación , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Biomarcadores de Tumor/genética , Humanos , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/terapia , Valor Predictivo de las Pruebas , Pronóstico , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA