Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(18)2022 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-36139590

RESUMO

BACKGROUND: Increasing knowledge of cancer biology and an expanding spectrum of molecularly targeted therapies provide the basis for precision oncology. Despite extensive gene diagnostics, previous reports indicate that less than 10% of patients benefit from this concept. METHODS: We retrospectively analyzed all patients referred to our center's Molecular Tumor Board (MTB) from 2018 to 2021. Molecular testing by next-generation sequencing (NGS) included a 67-gene panel for the detection of short-sequence variants and copy-number alterations, a 53- or 137-gene fusion panel and an ultra-low-coverage whole-genome sequencing for the detection of additional copy-number alterations outside the panel's target regions. Immunohistochemistry for microsatellite instability and PD-L1 expression complemented NGS. RESULTS: A total of 109 patients were referred to the MTB. In all, 78 patients received therapeutic proposals (70 based on NGS) and 33 were treated accordingly. Evaluable patients treated with MTB-recommended therapy (n = 30) had significantly longer progression-free survival than patients treated with other therapies (n = 17) (4.3 vs. 1.9 months, p = 0.0094). Seven patients treated with off-label regimens experienced major clinical benefits. CONCLUSION: The combined focused sequencing assays detected targetable alterations in the majority of patients. Patient benefits appeared to lie in the same range as with large-scale sequencing approaches.

2.
Hemasphere ; 6(5): e0708, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35519003

RESUMO

Resistance to cytarabine is a key problem in the treatment of acute myeloid leukemia (AML). To understand the molecular biology of resistance to cytarabine, a viability-based chemosensitizer screen was utilized. We screened synthetic lethal targets using 437 different small interfering RNAs (siRNAs) directed against factors involved in DNA repair mechanisms and cytarabine as the chemical compound. Three hits were identified: CUL4A, TP73, and RFC2. We show here that the ubiquitin ligase CULLIN 4A (CUL4A) and the tumor-suppressive transcription factor p73 contribute to drug resistance by modulating DNA damage response. P73 confers resistance to cytarabine therapy by transactivation of REV3L, encoding the catalytic subunit of translesion DNA polymerase ζ, and CUL4A probably by influencing proliferating cell nuclear antigen (PCNA) and the polymerase switch towards error-prone translesion DNA polymerases. Abrogation of the polymerase ζ by siRNA causes identical effects as siRNAs against CUL4A or TP73 and resensitizes cells towards cytarabine therapy in vitro. As CUL4A needs to be activated by neddylation to facilitate the degradation of several proteins including PCNA, we propose a novel explanation for the synergism between cytarabine and the neddylation inhibitor pevonedistat by inhibition of translesion synthesis. In keeping with this, in AML patients treated with cytarabine, we found high expression of CUL4A and TP73 to be associated with poor prognosis.

3.
Blood Adv ; 6(14): 4147-4156, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35413116

RESUMO

The phase 3 MIRROS (MDM2 antagonist Idasanutlin in Relapsed or Refractory acute myeloid leukemia [AML] for Overall Survival) trial (NCT02545283) evaluated the efficacy and safety of the small-molecule MDM2 antagonist idasanutlin plus cytarabine in patients with relapsed/refractory (R/R) AML. Adults (n = 447) with R/R AML whose disease relapsed or was refractory after ≤2 prior induction regimens as initial treatment or following salvage chemotherapy regimen, with Eastern Cooperative Oncology Group performance status ≤2 were enrolled regardless of TP53 mutation status and randomly assigned 2:1 to idasanutlin 300 mg or placebo orally twice daily plus cytarabine 1 g/m2 IV on days 1 to 5 of 28-day cycles. At primary analysis (cutoff, November 2019), 436 patients were enrolled, including 355 in the TP53 wild-type intention-to-treat (TP53WT-ITT) population. The primary endpoint, overall survival in the TP53WT-ITT population, was not met (median, 8.3 vs 9.1 months with idasanutlin-cytarabine vs placebo-cytarabine; stratified hazard ratio [HR], 1.08; 95% confidence interval [CI], 0.81-1.45; P = .58). The complete remission (CR) rate, a key secondary endpoint, was 20.3% vs 17.1% (odds ratio [OR], 1.23; 95% CI, 0.70-2.18). The overall response rate (ORR) was 38.8% vs 22.0% (OR, 2.25; 95% CI, 1.36-3.72). Common any-grade adverse events (≥10% incidence in any arm) were diarrhea (87.0% vs 32.9%), febrile neutropenia (52.8% vs 49.3%), and nausea (52.5% vs 31.5%). In summary, despite improved ORR, adding idasanutlin to cytarabine did not improve overall survival or CR rates in patients with R/R AML.


Assuntos
Citarabina , Leucemia Mieloide Aguda , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Humanos , Pirrolidinas , para-Aminobenzoatos/uso terapêutico
4.
Ultrasound Med Biol ; 46(8): 1865-1871, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32499194

RESUMO

Veno-occlusive disease (VOD) is a severe complication of allogeneic stem cell transplantation (allo-SCT). Its diagnosis is difficult, and ultrasound (US) has not been proven to be of additional diagnostic value. In the work described here, we prospectively analyzed the hepatic contrast-enhanced ultrasound (CEUS) pattern before and after allo-SCT and correlated these results with the pre-allo-SCT VOD risk factors, clinical VOD findings and conventional US findings. Thirty consecutive patients who underwent allo-SCT and had at least one VOD risk factor were studied. B-Mode US and CEUS were longitudinally performed at defined time points before and after allo-SCT. The 1-min hepatic enhancement (OMHE) in CEUS was standardized to splenic enhancement and quantified using optical density (OD) measurement software. A hypo-OMHE was arbitrarily defined as pathologic (pOMHE) if the OD of the liver was less than 90% compared with the mean splenic OD. Twenty-one patients (70%) had a pOMHE, the occurrence of which peaked at day 10 after allo-SCT. The number of pre-treatment VOD risk factors significantly differed between the pathologic and non-pathologic OD groups. Together, patients undergoing allo-SCT frequently exhibit a pathologic hepatic CEUS pattern, which can be quantified by OD measurement and is suggestive of pre-clinical VOD or other hepatic pathologies.


Assuntos
Fígado/diagnóstico por imagem , Transplante de Células-Tronco , Ultrassonografia/métodos , Adulto , Idoso , Meios de Contraste , Feminino , Hepatopatia Veno-Oclusiva/diagnóstico , Hepatopatia Veno-Oclusiva/diagnóstico por imagem , Hepatopatia Veno-Oclusiva/etiologia , Humanos , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/métodos
5.
Nat Commun ; 5: 3981, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24889111

RESUMO

Tumours are heterogeneous cell populations that undergo clonal evolution during tumour progression, metastasis and response to therapy. Short hairpin RNAs (shRNAs) generate stable loss-of-function phenotypes and are versatile experimental tools to explore the contribution of individual genetic alterations to clonal evolution. In these experiments tumour cells carrying shRNAs are commonly tracked with fluorescent reporters. While this works well for cell culture studies and leukaemia mouse models, fluorescent reporters are poorly suited for animals with solid tumours--the most common tumour types in cancer patients. Here we develop a toolkit that uses secreted luciferases to track the fate of two different shRNA-expressing tumour cell clones competitively, both in vitro and in vivo. We demonstrate that secreted luciferase activities can be measured robustly in the blood stream of tumour-bearing mice to accurately quantify, in a minimally invasive manner, the dynamic evolution of two genetically distinct tumour subclones in preclinical mouse models of tumour development, metastasis and therapy.


Assuntos
Evolução Clonal/genética , Vetores Genéticos , Luciferases , Neoplasias/genética , RNA Interferente Pequeno/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Células HCT116 , Humanos , Técnicas In Vitro , Camundongos , Microscopia de Fluorescência , Neoplasias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...