Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Blood ; 137(8): 1019-1023, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33167024

RESUMO

Trials assessing first-line, fixed-duration approaches in chronic lymphocytic leukemia (CLL) are yielding promising activity, but few long-term data are available. We report follow-up data from a phase 2 trial (ICLL07 FILO) in previously untreated, medically fit patients (N = 135). Patients underwent obinutuzumab-ibrutinib induction for 9 months; then, following evaluation (N = 130 evaluable), those in complete remission and with bone marrow measurable residual disease (BM MRD) <0.01% (n = 10) received ibrutinib for 6 additional months; those in partial remission and/or with BM MRD ≥0.01%, the majority (n = 120), also received 4 cycles of immunochemotherapy (fludarabine/cyclophosphamide-obinutuzumab). Beyond end of treatment, responses were assessed every 3 month and peripheral blood MRD every 6 months. At median follow-up 36.7 months from treatment start, progression-free and overall survival rates (95% confidence interval) at 3 years were 95.7% (92.0% to 99.5%) and 98% (95.1% to 100%), respectively. Peripheral blood MRD <0.01% rates were 97%, 96%, 90%, 84%, and 89% at months 16, 22, 28, 34, and 40, respectively. No new treatment-related or serious adverse event occurred beyond end of treatment. Thus, in previously untreated, medically fit patients with CLL, a fixed-duration (15 months), MRD-guided approach achieved high survival rates, a persistent MRD benefit beyond the end of treatment, and low long-term toxicity. This trial was registered at www.clinicaltrials.gov as #NCT02666898.


Assuntos
Adenina/análogos & derivados , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Piperidinas/uso terapêutico , Adenina/administração & dosagem , Adenina/efeitos adversos , Adenina/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Ciclofosfamida/uso terapêutico , Seguimentos , Humanos , Leucemia Linfocítica Crônica de Células B/diagnóstico , Neoplasia Residual/diagnóstico , Neoplasia Residual/tratamento farmacológico , Piperidinas/administração & dosagem , Piperidinas/efeitos adversos , Indução de Remissão , Resultado do Tratamento , Vidarabina/administração & dosagem , Vidarabina/efeitos adversos , Vidarabina/uso terapêutico
2.
Haematologica ; 106(12): 3100-3106, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34047178

RESUMO

The aim of this study was to characterize a large series of 154 patients with acute promyelocytic leukemia (median age, 53 years; range, 18-90 years) and evaluate real-life outcome after up-front treatment with arsenic trioxide and all-trans retinoic acid. All patients were included in the prospective NAPOLEON registry (NCT02192619) between 2013 and 2019. The acute promyelocytic leukemia was de novo in 91% (n=140) and therapy-related in 9% (n=14); 13% (n=20) of the patients were older than 70 years. At diagnosis bleeding/hemorrhage was present in 38% and thrombosis in 3%. Complete remission was achieved in 152 patients (99%), whereas two patients (1%) experienced induction death within 18 days after starting therapy. With a median follow-up of 1.99 years (95% confidence interval: 1.61-2.30 years) 1-year and 2-year overall survival rates were 97% (95% confidence interval: 94-100%) and 95% (95% confidence interval: 91-99%), respectively. Age above 70 years was associated with a significantly shorter overall survival (P<0.001) compared to that of younger patients. So far no relapses have been observed. Six patients (4%) died in complete remission at a median of 0.95 years after diagnosis (range, 0.18-2.38 years). Our data confirm the efficiency and durability of arsenic trioxide and all-trans retinoic acid therapy in the primary management of adults with low-/intermediate-risk acute promyelocytic leukemia in the real-life setting, irrespective of age.


Assuntos
Trióxido de Arsênio , Leucemia Promielocítica Aguda , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Trióxido de Arsênio/uso terapêutico , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Pessoa de Meia-Idade , Estudos Prospectivos , Indução de Remissão , Medição de Risco , Resultado do Tratamento , Tretinoína/uso terapêutico , Adulto Jovem
3.
Hum Mutat ; 41(7): 1220-1225, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32227665

RESUMO

Thrombocytopenia-absent radius (TAR) syndrome is characterized by radial defect and neonatal thrombocytopenia. It is caused by biallelic variants of RBM8A gene (1q21.1) with the association of a null allele and a hypomorphic noncoding variant. RBM8A encodes Y14, a core protein of the exon junction complex involved in messenger RNA maturation. To date, only two hypomorphic variants have been identified. We report on a cohort of 26 patients affected with TAR syndrome and carrying biallelic variants in RBM8A. Half patients carried a 1q21.1 deletion and one of the two known hypomorphic variants. Four novel noncoding variants of RBM8A were identified in the remaining patients. We developed experimental models enabling their functional characterization in vitro. Two variants, located respectively in the 5'-untranslated region (5'-UTR) and 3'-UTR regions, are responsible for a diminished expression whereas two intronic variants alter splicing. Our results bring new insights into the molecular knowledge of TAR syndrome and enabled us to propose genetic counseling for patients' families.


Assuntos
Síndrome Congênita de Insuficiência da Medula Óssea/genética , Proteínas de Ligação a RNA/genética , Trombocitopenia/genética , Deformidades Congênitas das Extremidades Superiores/genética , Regiões 5' não Traduzidas , Adolescente , Adulto , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 1 , Estudos de Coortes , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Rádio (Anatomia)/patologia , Adulto Jovem
4.
Blood Adv ; 7(15): 3936-3945, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37026799

RESUMO

In previously untreated, medically fit patients with chronic lymphocytic leukemia (CLL), research is focused on developing fixed-duration strategies to improve long-term outcomes while sparing patients from serious toxicities. The ICLL-07 trial evaluated a fixed-duration (15-month) immunochemotherapy approach in which after obinutuzumab-ibrutinib induction for 9 months, patients (n = 10) in complete remission (CR) with bone marrow (BM) measurable residual disease (MRD) <0.01% continued only ibrutinib 420 mg/day for 6 additional months (I arm), whereas the majority (n = 115) received up to 4 cycles of fludarabine/cyclophosphamide-obinutuzumab 1000 mg alongside the ibrutinib (I-FCG arm). Primary analysis at month 16 showed that 84 of 135 (62.2%) patients enrolled achieved CR with a BM MRD <0.01%. Here, we report follow-up at median 63 months. Peripheral blood (PB) MRD was assessed 6 monthly beyond the end of treatment using a highly sensitive (10-6) flow cytometry technique. In the I-FCG arm, the PB MRD <0.01% rate (low-level positive <0.01% or undetectable with limit of detection ≤10-4) in evaluable patients was still 92.5% (74/80) at month 40 and 80.6% (50/62) at month 64. No differences in the PB MRD status were apparent per to the IGHV mutational status. In the overall population, 4-year progression-free and overall survival rates were 95.5% and 96.2%, respectively. Twelve deaths occurred overall. Fourteen serious adverse events occurred beyond the end of treatment. Thus, our fixed-duration immunochemotherapy approach produced deep and sustained PB MRD responses, high survival rates, and low long-term toxicity. A randomized trial is needed to compare our immunochemotherapy approach with a chemotherapy-free strategy. This trial was registered at www.clinicaltrials.gov as #NCT02666898.


Assuntos
Leucemia Linfocítica Crônica de Células B , Humanos , Leucemia Linfocítica Crônica de Células B/diagnóstico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Rituximab/uso terapêutico , Ciclofosfamida , Medula Óssea , Indução de Remissão , Neoplasia Residual/tratamento farmacológico
5.
Am J Case Rep ; 22: e928369, 2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33667215

RESUMO

BACKGROUND Alpha-fetoprotein-producing gastric carcinoma (AFPGC) is a rare but aggressive cancer with a poor prognosis. Disseminated intravascular coagulation (DIC) is usually associated with several tumors, including gastric cancer, but only a few cases have been reported in patients with AFPGC. This report describes a case of advanced-stage AFPGC associated with DIC in a 50-year-old White man. CASE REPORT A 50-year-old, White, non-smoker man was hospitalized for a recent left hemiparesis associated with anorexia and loss of weigh. Clinically, we had multiple, hard, irregular, subcutaneous nodules, left supraclavicular lymph nodes, and a left, complete hemiparesis. Laboratory tests showed a DIC. A whole-body CT scan documented multiple lymph node, liver, subcutaneous, bone, and muscular metastases, a right femoral venous thrombosis, a left popliteal arterial thrombosis, and splenic and renal infarcts. The patient underwent an excisional biopsy of a subcutaneous lesion. Histology and immunohistochemistry confirmed the diagnosis of a metastasis from a high-grade AFPGC. Before starting any systemic treatment, the patient presented a massive intraventricular brain hemorrhage, quickly leading to his death. CONCLUSIONS We report a case of metastatic AFPGC associated with a DIC and multiple venous and arterial thromboses resulting in a fatal intracerebral hemorrhage. AFPGC is a distinctive and very difficult to diagnose tumor showing aggressive behavior and poor prognosis.


Assuntos
Carcinoma , Coagulação Intravascular Disseminada , Neoplasias Gástricas , Trombose , Coagulação Intravascular Disseminada/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Gástricas/complicações , alfa-Fetoproteínas
6.
Lancet Haematol ; 6(9): e470-e479, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31324600

RESUMO

BACKGROUND: In patients with chronic lymphocytic leukaemia, achievement of a complete response with minimal residual disease of less than 0·01% (ie, <1 chronic lymphocytic leukaemia cell per 10 000 leukocytes) in bone marrow has been associated with improved progression-free survival. We aimed to explore the activity of induction therapy for 9 months with obinutuzumab and ibrutinib, followed up with a minimal residual disease-driven therapeutic strategy for 6 additional months, in previously untreated patients. METHODS: We did a single-arm, phase 2 trial in 27 university hospitals, general hospitals, and specialist cancer centres in France. Eligible patients were at least 18 years old and previously untreated, and had immunophenotypically confirmed B-cell chronic lymphocytic leukaemia; an Eastern Cooperative Oncology Group (ECOG) performance status score of less than 2; a Binet stage C according to IWCLL 2008 criteria or Binet stage A and B with active disease; no 17p deletion or absence of p53 mutation; and were considered medically fit. In the first part of the study (induction phase), all participants received eight intravenous infusions of obinutuzumab 1000 mg over six 4-weekly cycles and oral ibrutinib 420 mg once per day for 9 months. In part 2, after assessment on day 1 of month 9, patients with a complete response and bone marrow minimal residual disease of less than 0·01% received only oral ibrutinib 420 mg once per day for 6 additional months. Patients with a partial response, or with a complete response and bone marrow minimal residual disease of 0·01% or more, received 6 months of four 4-weekly cycles of intravenous fludarabine, cyclophosphamide, and obinutuzumab 1000 mg, alongside continuing ibrutinib 420 mg once per day. The primary endpoint was the proportion of patients achieving a complete response with bone marrow minimal residual disease less than 0·01% on day 1 of month 16 assessed by intention to treat (ITT). This trial is registered with ClinicalTrials.gov (number NCT02666898) and is still open for follow-up. FINDINGS: Between Oct 27, 2015, and May 16, 2017, 135 patients were enrolled. After induction treatment (day 1 of month 9), 130 patients were evaluable, of which ten (8%) achieved a complete response with bone marrow minimal residual disease of less than 0·01% and were assigned to ibrutinib, and 120 (92%) were assigned to ibrutinib plus fludarabine, cyclophosphamide, and obinutuzumab. After minimal residual disease-guided treatment (day 1 of month 16), 84 (62%, 90% CI 55-69) of 135 patients (ITT population) achieved a complete response with bone marrow minimal residual disease of less than 0·01%. The most common haematological adverse event was thrombocytopenia (in 45 [34%] of 133 patients at grade 1-2 in months 1-9 and in 43 [33%] of 130 patients at grade 1-2 in months 9-15). The most common non-haematological adverse events were infusion-related reactions (in 83 [62%] patients at grade 1-2 in months 1-9) and gastrointestinal disorders (in 62 [48%] patients at grades 1 and 2 in months 9-15). 49 serious adverse events occurred, most frequently infections (ten), cardiac events (eight), and haematological events (eight). No treatment-related deaths occurred. INTERPRETATION: Obinutuzumab and ibrutinib induction therapy followed by a minimal residual disease driven strategy is safe and active in patients with previously untreated chronic lymphocytic leukaemia. With longer follow-up, including assessing the evolution of minimal residual disease, if response is maintained, this strategy could be an option in the first-line setting in patients with chronic lymphocytic leukaemia, although randomised evidence is needed. FUNDING: Roche, Janssen.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Adenina/análogos & derivados , Idoso , Anticorpos Monoclonais Humanizados/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Esquema de Medicação , Feminino , Gastroenteropatias/etiologia , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia Linfocítica Crônica de Células B/mortalidade , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasia Residual , Piperidinas , Pirazóis/efeitos adversos , Pirimidinas/efeitos adversos , Taxa de Sobrevida , Trombocitopenia/etiologia , Resultado do Tratamento , Proteína Supressora de Tumor p53/genética
7.
Bull Cancer ; 99(2): 163-71, 2012 Feb 01.
Artigo em Francês | MEDLINE | ID: mdl-21737380

RESUMO

Protein phosphorylation is a fundamental post-translational modification. It regulates a large number of critical cellular processes (differentiation, division, proliferation, apoptosis). Cell division is a process including a series of phases by which a parent cell divides into two daughter cells. The cells enter these stages then progress within the cell division under an accurate control by many proteins. These proteins are activated by phosphorylation. Cyclin-dependent kinases are responsible for this phosphorylation and therefore represent potential therapeutic targets especially in oncology.


Assuntos
Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas/metabolismo , Purinas/farmacologia , Animais , Quinases Ciclina-Dependentes/fisiologia , Humanos , Indóis/farmacologia , Camundongos , Camundongos Knockout , Fosforilação , Proteínas/química , Roscovitina , Relação Estrutura-Atividade
9.
Presse Med ; 39(9): e188-96, 2010 Sep.
Artigo em Francês | MEDLINE | ID: mdl-20400261

RESUMO

BACKGROUND: Pleural and pulmonary manifestations of giant cell arteritis are rare and not well known. They can be associated to more typical signs of the disease and to an inflammatory biological syndrome which are comprised in the multisystemic manifestations of the disease. They can be inaugural, leading to a late management if unrecognized. METHODS: Retrospective and descriptive study of 8 cases over a 10 year period was conducted. Five females and three males with a 67-year-old average age were included according to the American College of Rheumatology criteria. They illustrated the clinical and/or radiological respiratory manifestations of the disease. RESULTS: Pulmonary manifestation was inaugural in six cases over eight. The time to diagnosis range was 15-60 days. Cough was the most frequent symptom (five cases over eight). Dyspnea with orthopnea was described in one case. Pleural and parenchymal radiological manifestations had no specific characteristics: pleurisy, pleural thickening, nodules of variable size, reticular lesions. Temporal artery biopsy was positive in five cases, atypical in one case and negative in two cases. Bronchial and transbronchial biopsies (in two and one cases respectively) did not find any specific lesion. Clinical and radiological signs disappeared quickly after the introduction of glucocorticoid therapy. CONCLUSION: The knowledge of these different respiratory manifestations during giant cell arteritis (persistent cough, nodules, pleural effusion) is useful for the clinician. It helps him in prescribing non invasive investigations or even a presumptive glucocorticoid therapy, in an often old and weakened patient.


Assuntos
Arterite de Células Gigantes/complicações , Transtornos Respiratórios/etiologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Respiratórios/diagnóstico , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA