Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Blood ; 142(20): 1697-1707, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37595359

RESUMO

Addition of gemtuzumab ozogamicin (GO) to induction chemotherapy improves outcomes in older patients with acute myeloid leukemia (AML), but it is uncertain whether a fractionated schedule provides additional benefit to a single dose. We randomized 852 older adults (median age, 68-years) with AML/high-risk myelodysplasia to GO on day 1 (GO1) or on days 1 and 4 (GO2) of course 1 induction. The median follow-up period was 50.2 months. Although complete remission (CR) rates after course 1 did not significantly differ between arms (GO2, 63%; GO1, 57%; odds ratio [OR], 0.78; P = .08), there were significantly more patients who achieved CR with a measurable residual disease (MRD)<0.1% (50% vs 41%; OR, 0.72; P = .027). This differential MRD reduction with GO2 varied across molecular subtypes, being greatest for IDH mutations. The 5-year overall survival (OS) was 29% for patients in the GO2 arm and 24% for those in the GO1 arm (hazard ratio [HR], 0.89; P = .14). In a sensitivity analysis excluding patients found to have adverse cytogenetics or TP53 mutations, the 5-year OS was 33% for GO2 and 26% for GO1 (HR, 0.83; P = .045). In total, 228 (27%) patients received an allogeneic transplantation in first remission. Posttransplant OS was superior in the GO2 arm (HR, 0.67; P = .033); furthermore, the survival advantage from GO2 in the sensitivity analysis was lost when data of patients were censored at transplantation. In conclusion, GO2 was associated with a greater reduction in MRD and improved survival in older adults with nonadverse risk genetics. This benefit from GO2 was dependent on allogeneic transplantation to translate the better leukemia clearance into improved survival. This trial was registered at www.isrctn.com as #ISRCTN 31682779.


Assuntos
Daunorrubicina , Leucemia Mieloide Aguda , Humanos , Idoso , Gemtuzumab/uso terapêutico , Anticorpos Monoclonais Humanizados , Citarabina , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Reino Unido , Aminoglicosídeos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos
2.
JPRAS Open ; 27: 119-128, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33490356

RESUMO

INTRODUCTION: The establishment and success of new treatments are significantly influenced by patient satisfaction. Post-operative scarring is an important outcome for patients, and subsequently influences overall satisfaction with treatment. The objective was to measure post-treatment scarring satisfaction using a novel scale, the FACE-Q Skin Cancer Module, to compare electrochemotherapy (ECT) to traditional surgical excision (SE) to demonstrate equivalence of ECT and SE regarding outcome and survivorship. METHODS AND MATERIALS: This was a multicentre first-time appraisal study of the efficacy of ECT. All patients with facial BCCs treated with either ECT or SE were deemed eligible and subsequently recruited from either a previous clinical trial or outpatient clinics, respectively. Of the 40 participants invited, 25 responses were received. Patient information recorded included age, gender, location and size of BCCs, and time since treatment. Patient outcomes were measured using the FACE-Q Skin Cancer Module. RESULTS: The ECT and SE groups consisted of 14 and 11 patients, respectively. Mean age was 68 years (M:F = 16:9), while mean time since treatment was 4.98 years (range 0.3-9.58 years). Appraisal of scars was significantly higher in the ECT cohort versus SE (p = 0.034). Cancer worry was equivalent across both cohorts (p = 0.804). According to treatment type, no correlation was detected between time since treatment and both appraisal of scars (ECT p = 0.466 and SE p = 0.214) and adverse effects (ECT p = 0.924 and SE p = 0.139). CONCLUSION: Based on this study, ECT has superior scar outcomes and overall equivalence to SE. This demonstrates high patient satisfaction for those treated with ECT without any additional cancer worry.

3.
Nat Commun ; 11(1): 1407, 2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32179751

RESUMO

Leukaemogenic mutations commonly disrupt cellular differentiation and/or enhance proliferation, thus perturbing the regulatory programs that control self-renewal and differentiation of stem and progenitor cells. Translocations involving the Mll1 (Kmt2a) gene generate powerful oncogenic fusion proteins, predominantly affecting infant and paediatric AML and ALL patients. The early stages of leukaemogenic transformation are typically inaccessible from human patients and conventional mouse models. Here, we take advantage of cells conditionally blocked at the multipotent haematopoietic progenitor stage to develop a MLL-r model capturing early cellular and molecular consequences of MLL-ENL expression based on a clear clonal relationship between parental and leukaemic cells. Through a combination of scRNA-seq, ATAC-seq and genome-scale CRISPR-Cas9 screening, we identify pathways and genes likely to drive the early phases of leukaemogenesis. Finally, we demonstrate the broad utility of using matched parental and transformed cells for small molecule inhibitor studies by validating both previously known and other potential therapeutic targets.


Assuntos
Transformação Celular Neoplásica , Histona-Lisina N-Metiltransferase/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Feminino , Células-Tronco Hematopoéticas/metabolismo , Histona-Lisina N-Metiltransferase/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Cancer Cell ; 36(2): 123-138.e10, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31303423

RESUMO

Myeloid leukemia in Down syndrome (ML-DS) clonally evolves from transient abnormal myelopoiesis (TAM), a preleukemic condition in DS newborns. To define mechanisms of leukemic transformation, we combined exome and targeted resequencing of 111 TAM and 141 ML-DS samples with functional analyses. TAM requires trisomy 21 and truncating mutations in GATA1; additional TAM variants are usually not pathogenic. By contrast, in ML-DS, clonal and subclonal variants are functionally required. We identified a recurrent and oncogenic hotspot gain-of-function mutation in myeloid cytokine receptor CSF2RB. By a multiplex CRISPR/Cas9 screen in an in vivo murine TAM model, we tested loss-of-function of 22 recurrently mutated ML-DS genes. Loss of 18 different genes produced leukemias that phenotypically, genetically, and transcriptionally mirrored ML-DS.


Assuntos
Biomarcadores Tumorais/genética , Transformação Celular Neoplásica/genética , Cromossomos Humanos Par 21 , Subunidade beta Comum dos Receptores de Citocinas/genética , Síndrome de Down/genética , Fator de Transcrição GATA1/genética , Leucemia Mieloide/genética , Reação Leucemoide/genética , Mutação , Animais , Modelos Animais de Doenças , Progressão da Doença , Síndrome de Down/diagnóstico , Fator de Transcrição GATA1/metabolismo , Regulação Leucêmica da Expressão Gênica , Predisposição Genética para Doença , Células HEK293 , Humanos , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/patologia , Reação Leucemoide/diagnóstico , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Fenótipo , Transcrição Gênica
6.
Nat Med ; 24(8): 1167-1177, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30013198

RESUMO

Mutations in the gene encoding isocitrate dehydrogenase 2 (IDH2) occur in several types of cancer, including acute myeloid leukemia (AML). In model systems, mutant IDH2 causes hematopoietic differentiation arrest. Enasidenib, a selective small-molecule inhibitor of mutant IDH2, produces a clinical response in 40% of treated patients with relapsed/refractory AML by promoting leukemic cell differentiation. Here, we studied the clonal basis of response and acquired resistance to enasidenib treatment. Using sequential patient samples, we determined the clonal structure of hematopoietic cell populations at different stages of differentiation. Before therapy, IDH2-mutant clones showed variable differentiation arrest. Enasidenib treatment promoted hematopoietic differentiation from either terminal or ancestral mutant clones; less frequently, treatment promoted differentiation of nonmutant cells. Analysis of paired diagnosis/relapse samples did not identify second-site mutations in IDH2 at relapse. Instead, relapse arose by clonal evolution or selection of terminal or ancestral clones, thus highlighting multiple bypass pathways that could potentially be targeted to restore differentiation arrest. These results show how mapping of clonal structure in cell populations at different stages of differentiation can reveal the response and evolution of clones during treatment response and relapse.


Assuntos
Aminopiridinas/uso terapêutico , Isocitrato Desidrogenase/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Triazinas/uso terapêutico , Aminopiridinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Clonais , Estudos de Coortes , Hematopoese , Humanos , Imunofenotipagem , Isocitrato Desidrogenase/metabolismo , Mutação/genética , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Triazinas/farmacologia
7.
Clin Cancer Res ; 23(21): 6430-6440, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28765326

RESUMO

Purpose: Azacitidine (AZA) is a novel therapeutic option in older patients with acute myeloid leukemia (AML), but its rational utilization is compromised by the fact that neither the determinants of clinical response nor its mechanism of action are defined. Co-administration of histone deacetylase inhibitors, such as vorinostat (VOR), is reported to improve the clinical activity of AZA, but this has not been prospectively studied in patients with AML.Experimental Design: We compared outcomes in 259 adults with AML (n = 217) and MDS (n = 42) randomized to receive either AZA monotherapy (75 mg/m2 × 7 days every 28 days) or AZA combined with VOR 300 mg twice a day on days 3 to 9 orally. Next-generation sequencing was performed in 250 patients on 41 genes commonly mutated in AML. Serial immunophenotyping of progenitor cells was performed in 47 patients.Results: Co-administration of VOR did not increase the overall response rate (P = 0.84) or overall survival (OS; P = 0.32). Specifically, no benefit was identified in either de novo or relapsed AML. Mutations in the genes CDKN2A (P = 0.0001), IDH1 (P = 0.004), and TP53 (P = 0.003) were associated with reduced OS. Lymphoid multipotential progenitor populations were greatly expanded at diagnosis and although reduced in size in responding patients remained detectable throughout treatment.Conclusions: This study demonstrates no benefit of concurrent administration of VOR with AZA but identifies a mutational signature predictive of outcome after AZA-based therapy. The correlation between heterozygous loss of function CDKN2A mutations and decreased OS implicates induction of cell-cycle arrest as a mechanism by which AZA exerts its clinical activity. Clin Cancer Res; 23(21); 6430-40. ©2017 AACR.


Assuntos
Azacitidina/administração & dosagem , Inibidor de Quinase Dependente de Ciclina p18/genética , Isocitrato Desidrogenase/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Proteína Supressora de Tumor p53/genética , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Azacitidina/efeitos adversos , Terapia Combinada , Inibidor p16 de Quinase Dependente de Ciclina , Intervalo Livre de Doença , Esquema de Medicação , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Inibidores de Histona Desacetilases/efeitos adversos , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/efeitos adversos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Resultado do Tratamento , Vorinostat
8.
J Exp Med ; 213(8): 1513-35, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27377587

RESUMO

Our understanding of the perturbation of normal cellular differentiation hierarchies to create tumor-propagating stem cell populations is incomplete. In human acute myeloid leukemia (AML), current models suggest transformation creates leukemic stem cell (LSC) populations arrested at a progenitor-like stage expressing cell surface CD34. We show that in ∼25% of AML, with a distinct genetic mutation pattern where >98% of cells are CD34(-), there are multiple, nonhierarchically arranged CD34(+) and CD34(-) LSC populations. Within CD34(-) and CD34(+) LSC-containing populations, LSC frequencies are similar; there are shared clonal structures and near-identical transcriptional signatures. CD34(-) LSCs have disordered global transcription profiles, but these profiles are enriched for transcriptional signatures of normal CD34(-) mature granulocyte-macrophage precursors, downstream of progenitors. But unlike mature precursors, LSCs express multiple normal stem cell transcriptional regulators previously implicated in LSC function. This suggests a new refined model of the relationship between LSCs and normal hemopoiesis in which the nature of genetic/epigenetic changes determines the disordered transcriptional program, resulting in LSC differentiation arrest at stages that are most like either progenitor or precursor stages of hemopoiesis.


Assuntos
Antígenos CD34/genética , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Leucemia Mieloide Aguda , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/metabolismo , Animais , Antígenos CD34/metabolismo , Células Progenitoras de Granulócitos e Macrófagos/patologia , Xenoenxertos , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia
9.
Blood Adv ; 1(3): 193-204, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-29296935

RESUMO

Disease relapse is the major cause of treatment failure after allogeneic stem cell transplantation (allo-SCT) in acute myeloid leukemia (AML). To identify AML-associated genes prognostic of AML relapse post-allo-SCT, we resequenced 35 genes in 113 adults at diagnosis, 49 of whom relapsed. Two hundred sixty-two mutations were detected in 102/113 (90%) patients. An increased risk of relapse was observed in patients with mutations in WT1 (P = .018), DNMT3A (P = .045), FLT3 ITD (P = .071), and TP53 (P = .06), whereas mutations in IDH1 were associated with a reduced risk of disease relapse (P = .018). In 29 patients, we additionally compared mutational profiles in bone marrow at diagnosis and relapse to study changes in clonal structure at relapse. In 13/29 patients, mutational profiles altered at relapse. In 9 patients, mutations present at relapse were not detected at diagnosis. In 15 patients, additional available pre-allo-SCT samples demonstrated that mutations identified posttransplant but not at diagnosis were detectable immediately prior to transplant in 2 of 15 patients. Taken together, these observations, if confirmed in larger studies, have the potential to inform the design of novel strategies to reduce posttransplant relapse highlighting the potential importance of post-allo-SCT interventions with a broad antitumor specificity in contrast to targeted therapies based on mutational profile at diagnosis.

10.
World J Gastroenterol ; 21(18): 5751-4, 2015 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-25987804

RESUMO

Children on exclusive jejunal feeding may be at risk of iron deficiency due to the feeds bypassing the duodenum, which is the primary site for iron absorption. We describe the biochemical and hematological features of six children on exclusive jejunal feeding who did not receive iron supplementation. At a mean (standard deviation) period of 11 (6.5) mo after commencing jejunal feeds, there was a significant reduction in both serum iron (18.5 g/L vs 9.8 g/L, P = 0.01) and transferrin saturation levels (23.1% vs 13.7%, P = 0.02), suggesting iron deficiency. However, there was no significant change in ferritin, hemoglobin and mean corpuscular volume levels post-commencement of jejunal feeds. This may be the result of small bowel adaptation in response to early iron deficiency. Larger and longer term prospective studies are required to investigate if children on jejunal feeds are at risk of developing iron deficiency.


Assuntos
Derivação Gástrica , Jejuno/patologia , Fatores de Transcrição/metabolismo , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA