Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 553(7687): 222-227, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29323298

RESUMEN

Chromosomal translocations that generate in-frame oncogenic gene fusions are notable examples of the success of targeted cancer therapies. We have previously described gene fusions of FGFR3-TACC3 (F3-T3) in 3% of human glioblastoma cases. Subsequent studies have reported similar frequencies of F3-T3 in many other cancers, indicating that F3-T3 is a commonly occuring fusion across all tumour types. F3-T3 fusions are potent oncogenes that confer sensitivity to FGFR inhibitors, but the downstream oncogenic signalling pathways remain unknown. Here we show that human tumours with F3-T3 fusions cluster within transcriptional subgroups that are characterized by the activation of mitochondrial functions. F3-T3 activates oxidative phosphorylation and mitochondrial biogenesis and induces sensitivity to inhibitors of oxidative metabolism. Phosphorylation of the phosphopeptide PIN4 is an intermediate step in the signalling pathway of the activation of mitochondrial metabolism. The F3-T3-PIN4 axis triggers the biogenesis of peroxisomes and the synthesis of new proteins. The anabolic response converges on the PGC1α coactivator through the production of intracellular reactive oxygen species, which enables mitochondrial respiration and tumour growth. These data illustrate the oncogenic circuit engaged by F3-T3 and show that F3-T3-positive tumours rely on mitochondrial respiration, highlighting this pathway as a therapeutic opportunity for the treatment of tumours with F3-T3 fusions. We also provide insights into the genetic alterations that initiate the chain of metabolic responses that drive mitochondrial metabolism in cancer.


Asunto(s)
Respiración de la Célula , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Peptidilprolil Isomerasa de Interacción con NIMA/química , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Biogénesis de Organelos , Fosforilación Oxidativa/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Peroxisomas/efectos de los fármacos , Peroxisomas/metabolismo , Fosforilación , Biosíntesis de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/metabolismo , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Clin Lab Anal ; 37(7): e24884, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37088872

RESUMEN

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a severe complication of heparin therapy associated with thrombosis that requires a quick diagnosis. Therefore, laboratory assays must provide an accurate and swift answer. This work aims to evaluate the performances of an ELISA assay, especially when combined with 4T risk score, and a functional assay. METHODS: Data were collected for 894 patients treated by heparin who underwent anticoagulant switch because of HIT suspicion and were examined by a multidisciplinary expert team who confirmed or ruled out HIT diagnosis. All patients were tested for anti-PF4 IgG with Asserachrom HPIA IgG (ELISA), and 307 were tested with a platelet aggregation test done on platelet-rich plasma (PRP-PAT). The 4T risk score was available for 607 of them. RESULTS: HIT was diagnosed in 232 patients. 4T risk score had a 94.2% negative predictive value (NPV) for risk scores ≤3 and 77.3% for risk scores ≤5. The sensitivity of ELISA was 90.9%, its specificity 79.0%, and its NPV 96.1%. When combined with 4T risk score, its NPV reached 100% and 97% for risk scores ≤3 and ≤5, respectively. PRP-PAT sensitivity was 70.4%, and its specificity was 92.3%. Combination of ELISA and PRP-PAT had a 0.7% false-negative rate. CONCLUSION: This study shows that ELISA can rule out HIT with an excellent NPV, especially when combined with the 4T risk score. Nonetheless, it has low specificity; hence, it needs to be associated with a functional assay.


Asunto(s)
Factor Plaquetario 4 , Trombocitopenia , Humanos , Estudios Retrospectivos , Factor Plaquetario 4/efectos adversos , Trombocitopenia/inducido químicamente , Trombocitopenia/diagnóstico , Heparina/efectos adversos , Anticoagulantes/efectos adversos , Ensayo de Inmunoadsorción Enzimática , Pruebas de Función Plaquetaria , Inmunoglobulina G
3.
Proc Natl Acad Sci U S A ; 114(25): 6581-6586, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28607076

RESUMEN

Identification of biomarkers and therapeutic targets is a critical goal of precision medicine. N-glycoproteins are a particularly attractive class of proteins that constitute potential cancer biomarkers and therapeutic targets for small molecules, antibodies, and cellular therapies. Using mass spectrometry (MS), we generated a compendium of 1,091 N-glycoproteins (from 40 human primary lymphomas and cell lines). Hierarchical clustering revealed distinct subtype signatures that included several subtype-specific biomarkers. Orthogonal immunological studies in 671 primary lymphoma tissue biopsies and 32 lymphoma-derived cell lines corroborated MS data. In anaplastic lymphoma kinase-positive (ALK+) anaplastic large cell lymphoma (ALCL), integration of N-glycoproteomics and transcriptome sequencing revealed an ALK-regulated cytokine/receptor signaling network, including vulnerabilities corroborated by a genome-wide clustered regularly interspaced short palindromic screen. Functional targeting of IL-31 receptor ß, an ALCL-enriched and ALK-regulated N-glycoprotein in this network, abrogated ALK+ALCL growth in vitro and in vivo. Our results highlight the utility of functional proteogenomic approaches for discovery of cancer biomarkers and therapeutic targets.


Asunto(s)
Biomarcadores de Tumor/genética , Linfoma/genética , Transcriptoma/genética , Línea Celular Tumoral , Humanos , Proteogenómica/métodos , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética
4.
Blood ; 124(25): 3768-71, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349176

RESUMEN

The spectrum of cutaneous CD30-positive lymphoproliferative disorders (LPDs) includes lymphomatoid papulosis and primary cutaneous anaplastic large cell lymphoma. Chromosomal translocations targeting tyrosine kinases in CD30-positive LPDs have not been described. Using whole-transcriptome sequencing, we identified a chimeric fusion involving NPM1 (5q35) and TYK2 (19p13) that encodes an NPM1-TYK2 protein containing the oligomerization domain of NPM1 and an intact catalytic domain in TYK2. Fluorescence in situ hybridization revealed NPM1-TYK2 fusions in 2 of 47 (4%) primary cases of CD30-positive LPDs and was absent in other mature T-cell neoplasms (n = 151). Functionally, NPM1-TYK2 induced constitutive TYK2, signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5 activation. Conversely, a kinase-defective NPM1-TYK2 mutant abrogated STAT1/3/5 signaling. Finally, short hairpin RNA-mediated silencing of TYK2 abrogated lymphoma cell growth. This is the first report of recurrent translocations involving TYK2, and it highlights the novel therapeutic opportunities in the treatment of CD30-positive LPDs with TYK2 translocations.


Asunto(s)
Antígeno Ki-1/genética , Linfoma Anaplásico Cutáneo Primario de Células Grandes/genética , Papulosis Linfomatoide/genética , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias Cutáneas/genética , TYK2 Quinasa/genética , Western Blotting , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Hibridación Fluorescente in Situ , Antígeno Ki-1/metabolismo , Linfoma Anaplásico Cutáneo Primario de Células Grandes/metabolismo , Linfoma Anaplásico Cutáneo Primario de Células Grandes/patología , Papulosis Linfomatoide/metabolismo , Papulosis Linfomatoide/patología , Mutación , Proteínas Nucleares/metabolismo , Nucleofosmina , Fusión de Oncogenes , Proteínas de Fusión Oncogénica/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , TYK2 Quinasa/metabolismo , Transcriptoma/genética
5.
Blood ; 124(9): 1460-72, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-24825865

RESUMEN

The comprehensive genetic alterations underlying the pathogenesis of T-cell prolymphocytic leukemia (T-PLL) are unknown. To address this, we performed whole-genome sequencing (WGS), whole-exome sequencing (WES), high-resolution copy-number analysis, and Sanger resequencing of a large cohort of T-PLL. WGS and WES identified novel mutations in recurrently altered genes not previously implicated in T-PLL including EZH2, FBXW10, and CHEK2. Strikingly, WGS and/or WES showed largely mutually exclusive mutations affecting IL2RG, JAK1, JAK3, or STAT5B in 38 of 50 T-PLL genomes (76.0%). Notably, gain-of-function IL2RG mutations are novel and have not been reported in any form of cancer. Further, high-frequency mutations in STAT5B have not been previously reported in T-PLL. Functionally, IL2RG-JAK1-JAK3-STAT5B mutations led to signal transducer and activator of transcription 5 (STAT5) hyperactivation, transformed Ba/F3 cells resulting in cytokine-independent growth, and/or enhanced colony formation in Jurkat T cells. Importantly, primary T-PLL cells exhibited constitutive activation of STAT5, and targeted pharmacologic inhibition of STAT5 with pimozide induced apoptosis in primary T-PLL cells. These results for the first time provide a portrait of the mutational landscape of T-PLL and implicate deregulation of DNA repair and epigenetic modulators as well as high-frequency mutational activation of the IL2RG-JAK1-JAK3-STAT5B axis in the pathogenesis of T-PLL. These findings offer opportunities for novel targeted therapies in this aggressive leukemia.


Asunto(s)
Leucemia Prolinfocítica de Células T/genética , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Secuencia de Bases , Muerte Celular/efectos de los fármacos , Estudios de Cohortes , Simulación por Computador , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Exoma , Femenino , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Janus Quinasa 1/genética , Janus Quinasa 3/química , Janus Quinasa 3/genética , Leucemia Prolinfocítica de Células T/tratamiento farmacológico , Leucemia Prolinfocítica de Células T/metabolismo , Masculino , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Pimozida/farmacología , Conformación Proteica , Factor de Transcripción STAT5/antagonistas & inhibidores , Factor de Transcripción STAT5/química , Factor de Transcripción STAT5/genética , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
6.
Am J Pathol ; 184(5): 1331-42, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24667141

RESUMEN

Deregulation of signaling pathways controlled by protein phosphorylation underlies the pathogenesis of hematological malignancies; however, the extent to which deregulated phosphorylation may be involved in B-cell non-Hodgkin lymphoma (B-NHL) pathogenesis is largely unknown. To identify phosphorylation events important in B-NHLs, we performed mass spectrometry-based, label-free, semiquantitative phosphoproteomic profiling of 11 cell lines derived from three B-NHL categories: Burkitt lymphoma, follicular lymphoma, and mantle-cell lymphoma. In all, 6579 unique phosphopeptides, corresponding to 1701 unique phosphorylated proteins, were identified and quantified. The data are available via ProteomeXchange with identifier PXD000658. Hierarchical clustering highlighted distinct phosphoproteomic signatures associated with each lymphoma subtype. Interestingly, germinal center-derived B-NHL cell lines were characterized by phosphorylation of proteins involved in the B-cell receptor signaling. Of these proteins, phosphoprotein associated with glycosphingolipid-enriched microdomains 1 (PAG1) was identified with the most phosphorylated tyrosine peptides in Burkitt lymphoma and follicular lymphoma. PAG1 knockdown resulted in perturbation of the tyrosine phosphosignature of B-cell receptor signaling components. Significantly, PAG1 knockdown increased cell proliferation and response to antigen stimulation of these germinal center-derived B-NHLs. These data provide a detailed annotation of phosphorylated proteins in human lymphoid cancer. Overall, our study revealed the utility of unbiased phosphoproteome interrogation in characterizing signaling networks that may provide insights into pathogenesis mechanisms in B-cell lymphomas.


Asunto(s)
Linfoma de Células B/metabolismo , Linfoma no Hodgkin/metabolismo , Fosfoproteínas/metabolismo , Proteómica/métodos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Proliferación Celular , Análisis por Conglomerados , Técnicas de Silenciamiento del Gen , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Linfoma de Células B/diagnóstico , Linfoma no Hodgkin/diagnóstico , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Fosfopéptidos/metabolismo , Fosforilación , Transducción de Señal , Familia-src Quinasas/metabolismo
7.
Blood ; 122(6): 958-68, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23814019

RESUMEN

The mechanisms underlying the pathogenesis of the constitutively active tyrosine kinase nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) expressing anaplastic large cell lymphoma are not completely understood. Here we show using an integrated phosphoproteomic and metabolomic strategy that NPM-ALK induces a metabolic shift toward aerobic glycolysis, increased lactate production, and biomass production. The metabolic shift is mediated through the anaplastic lymphoma kinase (ALK) phosphorylation of the tumor-specific isoform of pyruvate kinase (PKM2) at Y105, resulting in decreased enzymatic activity. Small molecule activation of PKM2 or expression of Y105F PKM2 mutant leads to reversal of the metabolic switch with increased oxidative phosphorylation and reduced lactate production coincident with increased cell death, decreased colony formation, and reduced tumor growth in an in vivo xenograft model. This study provides comprehensive profiling of the phosphoproteomic and metabolomic consequences of NPM-ALK expression and reveals a novel role of ALK in the regulation of multiple components of cellular metabolism. Our studies show that PKM2 is a novel substrate of ALK and plays a critical role in mediating the metabolic shift toward biomass production and tumorigenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Linfoma Anaplásico de Células Grandes/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Humanos , Metabolómica , Ratones , Ratones SCID , Trasplante de Neoplasias , Fosforilación , Proteómica , Especificidad por Sustrato , Proteínas de Unión a Hormona Tiroide
8.
Mol Cell Proteomics ; 12(10): 2714-23, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836920

RESUMEN

Chromosomal translocations encoding chimeric fusion proteins constitute one of the most common mechanisms underlying oncogenic transformation in human cancer. Fusion peptides resulting from such oncogenic chimeric fusions, though unique to specific cancer subtypes, are unexplored as cancer biomarkers. Here we show, using an approach termed fusion peptide multiple reaction monitoring mass spectrometry, the direct identification of different cancer-specific fusion peptides arising from protein chimeras that are generated from the juxtaposition of heterologous genes fused by recurrent chromosomal translocations. Using fusion peptide multiple reaction monitoring mass spectrometry in a clinically relevant scenario, we demonstrate the specific, sensitive, and unambiguous detection of a specific diagnostic fusion peptide in clinical samples of anaplastic large cell lymphoma, but not in a diverse array of benign lymph nodes or other forms of primary malignant lymphomas and cancer-derived cell lines. Our studies highlight the utility of fusion peptides as cancer biomarkers and carry broad implications for the use of protein biomarkers in cancer detection and monitoring.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Péptidos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Humanos , Linfoma Anaplásico de Células Grandes/genética , Espectrometría de Masas/métodos , Proteínas de Fusión Oncogénica/genética , Péptidos/genética , Proteínas Tirosina Quinasas/genética
9.
Lancet Oncol ; 14(6): 472-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23598171

RESUMEN

BACKGROUND: Various human cancers have ALK gene translocations, amplifications, or oncogenic mutations, such as anaplastic large-cell lymphoma, inflammatory myofibroblastic tumours, non-small-cell lung cancer (NSCLC), and neuroblastoma. Therefore, ALK inhibition could be a useful therapeutic strategy in children. We aimed to determine the safety, recommended phase 2 dose, and antitumour activity of crizotinib in children with refractory solid tumours and anaplastic large-cell lymphoma. METHODS: In this open-label, phase 1 dose-escalation trial, patients older than 12 months and younger than 22 years with measurable or evaluable solid or CNS tumours, or anaplastic large-cell lymphoma, refractory to therapy and for whom there was no known curative treatment were eligible. Crizotinib was given twice daily without interruption. Six dose levels (100, 130, 165, 215, 280, 365 mg/m(2) per dose) were assessed in the dose-finding phase of the study (part A1), which is now completed. The primary endpoint was to estimate the maximum tolerated dose, to define the toxic effects of crizotinib, and to characterise the pharmacokinetics of crizotinib in children with refractory cancer. Additionally, patients with confirmed ALK translocations, mutations, or amplification (part A2 of the study) or neuroblastoma (part A3) could enrol at one dose level lower than was currently given in part A1. We assessed ALK genomic status in tumour tissue and used quantitative RT-PCR to measure NPM-ALK fusion transcript in bone marrow and blood samples of patients with anaplastic large-cell lymphoma. All patients who received at least one dose of crizotinib were evaluable for response; patients completing at least one cycle of therapy or experiencing dose limiting toxicity before that were considered fully evaluable for toxicity. This study is registered with ClinicalTrials.gov, NCT00939770. FINDINGS: 79 patients were enrolled in the study from Oct 2, 2009, to May 31, 2012. The median age was 10.1 years (range 1.1-21.4); 43 patients were included in the dose escalation phase (A1), 25 patients in part A2, and 11 patients in part A3. Crizotinib was well tolerated with a recommended phase 2 dose of 280 mg/m(2) twice daily. Grade 4 adverse events in cycle 1 were neutropenia (two) and liver enzyme elevation (one). Grade 3 adverse events that occurred in more than one patient in cycle 1 were lymphopenia (two), and neutropenia (eight). The mean steady state peak concentration of crizotinib was 630 ng/mL and the time to reach this peak was 4 h (range 1-6). Objective tumour responses were documented in 14 of 79 patients (nine complete responses, five partial responses); and the anti-tumour activity was enriched in patients with known activating ALK aberrations (eight of nine with anaplastic large-cell lymphoma, one of 11 with neuroblastoma, three of seven with inflammatory myofibroblastic tumour, and one of two with NSCLC). INTERPRETATION: The findings suggest that a targeted inhibitor of ALK has antitumour activity in childhood malignancies harbouring ALK translocations, particularly anaplastic large-cell lymphoma and inflammatory myofibroblastic tumours, and that further investigation in the subset of neuroblastoma harbouring known ALK oncogenic mutations is warranted. FUNDING: Pfizer and National Cancer Institute grant to the Children's Oncology Group.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Adolescente , Factores de Edad , Quinasa de Linfoma Anaplásico , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Niño , Preescolar , Crizotinib , Progresión de la Enfermedad , Esquema de Medicación , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Lactante , Linfoma Anaplásico de Células Grandes/enzimología , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/patología , Masculino , Dosis Máxima Tolerada , Terapia Molecular Dirigida , Mutación , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/efectos adversos , Pirazoles/farmacocinética , Piridinas/efectos adversos , Piridinas/farmacocinética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
10.
J Gynecol Obstet Hum Reprod ; 51(8): 102434, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35768063

RESUMEN

OBJECTIVE: To implement a vaginal delivery of the second twin simulation program for obstetric and gynecology residents, to reduce maternal-fetal mortality in the management of twin pregnancies in the delivery room. DESIGN: A prospective education program. The session consisted of a theoretical part, a practical part on a mannequin and an evaluation. The model was designed in a simple and reproducible way. The simulation evaluation was done at several levels according to the validated Kirkpatrick model. SETTING: A tertiary level university maternity hospital. PARTICIPANTS: Ten obstetric and gynecology residents participated. RESULTS: The resident's personal feelings about their ability to perform the maneuvers increased significantly after participation in the simulation session. Their technical skills in performing the maneuvers were assessed in a second step. The majority of the participants considered the model realistic and the session useful. All agreed that the simulation was an essential part of their learning process. CONCLUSION: A simulated vaginal delivery of the second twin session allows residents to be safely trained in these obstetrical maneuvers, which can be difficult to teach and perform.


Asunto(s)
Ginecología , Internado y Residencia , Obstetricia , Competencia Clínica , Parto Obstétrico/educación , Femenino , Ginecología/educación , Maternidades , Humanos , Obstetricia/educación , Embarazo , Estudios Prospectivos , Universidades
11.
J Gynecol Obstet Hum Reprod ; 50(7): 102107, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33705991

RESUMEN

INTRODUCTION: Initially dispensed in specialized simulation centers, simulation training has recently begun to take place directly in healthcare facilities, that is, in situ. The objective of this study is to assess the effect of training by in situ simulation in obstetrics. MATERIAL AND METHODS: The training program, dispensed over a 2-day period, took place in maternity units of the members of the Pays de la Loire perinatal network, Réseau Sécurité Naissance (Network Safety Birth). All participants received a learner satisfaction questionnaire to complete (5-point Likert-like scales). Then, at least 6 months later, each maternity ward received a general questionnaire to assess the effect of the training, as well as a second questionnaire specific to each institution, about the areas for improvement proposed by the teaching team after debriefings. RESULTS: The 14 establishments included in our study returned 375 satisfaction questionnaires. In all, 91.1 % were very satisfied and reported that the training met their expectations, and 99.7 % thought the program would have an impact on their professional practice. More than 94 % of the learners wanted more simulation sessions. Among the 14 facilities, 9 (64.3 %) returned their evaluation questionnaires. In 44.4 % of cases, they reported improvement in team cohesion and in team communication, while the others reported these elements remained stable. All maternity units reported that the training had a positive impact on their team, and that they would be interested in new training program with in situ simulation. DISCUSSION: Most participants clearly appreciated this training. In situ simulation training also led to the identification of areas for improvements, many of them accomplished, through the drafting of protocols or material modifications aimed at improving staff practices and therefore global patient care. There are many ways by which these training programs can be made sustainable, including the development of a new training program of in situ simulation or the creation of onsite simulation sessions on demand or by the professionals at each institution. CONCLUSION: This survey demonstrated the enthusiasm of healthcare professionals about in situ simulation. Moreover, overall improvement in team communication and cohesion was reported in the medium term (evaluation at more than 6 months). The interest of continuing these training sessions appears undeniable.


Asunto(s)
Personal de Salud/educación , Obstetricia/educación , Entrenamiento Simulado/normas , Enseñanza/normas , Adulto , Competencia Clínica/normas , Competencia Clínica/estadística & datos numéricos , Educación Profesional/métodos , Educación Profesional/normas , Educación Profesional/estadística & datos numéricos , Femenino , Humanos , Masculino , Entrenamiento Simulado/métodos , Entrenamiento Simulado/estadística & datos numéricos , Encuestas y Cuestionarios , Enseñanza/estadística & datos numéricos
12.
Cancer Res ; 80(12): 2498-2511, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32350066

RESUMEN

Mature B-cell neoplasms are the fifth most common neoplasm. Due to significant heterogeneity at the clinical and genetic levels, current therapies for these cancers fail to provide long-term cures. The clinical success of proteasome inhibition for the treatment of multiple myeloma and B-cell lymphomas has made the ubiquitin pathway an important emerging therapeutic target. In this study, we assessed the role of the E3 ligase FBXW7 in mature B-cell neoplasms. FBXW7 targeted the frequently inactivated tumor suppressor KMT2D for protein degradation, subsequently regulating gene expression signatures related to oxidative phosphorylation (OxPhos). Loss of FBXW7 inhibited diffuse large B-cell lymphoma cell growth and further sensitized cells to OxPhos inhibition. These data elucidate a novel mechanism of regulation of KMT2D levels by the ubiquitin pathway and uncover a role of FBXW7 in regulating oxidative phosphorylation in B-cell malignancies. SIGNIFICANCE: These findings characterize FBXW7 as a prosurvival factor in B-cell lymphoma via degradation of the chromatin modifier KMT2D.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación Neoplásica de la Expresión Génica , Linfoma de Células B Grandes Difuso/genética , Proteínas de Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Linfoma de Células B Grandes Difuso/patología , Ratones , Proteínas de Neoplasias/genética , Fosforilación Oxidativa , Proteolisis , ARN Interferente Pequeño/metabolismo , Transducción de Señal/genética , Ubiquitina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Clin Cancer Res ; 26(3): 690-703, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31636099

RESUMEN

PURPOSE: Peripheral T-cell lymphomas are clinically aggressive and usually fatal, as few complete or durable remissions are achieved with currently available therapies. Recent evidence supports a critical role for lymphoma-associated macrophages during T-cell lymphoma progression, but the specific signals involved in the cross-talk between malignant T cells and their microenvironment are poorly understood. Colony-stimulator factor 1 receptor (CSF1R, CD115) is required for the homeostatic survival of tissue-resident macrophages. Interestingly, its aberrant expression has been reported in a subset of tumors. In this article, we evaluated its expression and oncogenic role in T-cell lymphomas. EXPERIMENTAL DESIGN: Loss-of-function studies, including pharmacologic inhibition with a clinically available tyrosine kinase inhibitor, pexidartinib, were performed in multiple in vitro and in vivo models. In addition, proteomic and genomic screenings were performed to discover signaling pathways that are activated downstream of CSF1R signaling. RESULTS: We observed that CSF1R is aberrantly expressed in many T-cell lymphomas, including a significant number of peripheral and cutaneous T-cell lymphomas. Colony-stimulating factor 1 (CSF1), in an autocrine or paracrine-dependent manner, leads to CSF1R autophosphorylation and activation in malignant T cells. Furthermore, CSF1R signaling was associated with significant changes in gene expression and in the phosphoproteome, implicating PI3K/AKT/mTOR in CSF1R-mediated T-cell lymphoma growth. We also demonstrated that inhibition of CSF1R in vivo and in vitro models is associated with decreased T-cell lymphoma growth. CONCLUSIONS: Collectively, these findings implicate CSF1R in T-cell lymphomagenesis and have significant therapeutic implications.


Asunto(s)
Aminopiridinas/farmacología , Linfoma de Células T Periférico/patología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirroles/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Humanos , Linfoma de Células T Periférico/tratamiento farmacológico , Linfoma de Células T Periférico/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Oncogene ; 39(10): 2103-2117, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31804622

RESUMEN

Rational new strategies are needed to treat tumors resistant to kinase inhibitors. Mechanistic studies of resistance provide fertile ground for development of new approaches. Cancer drug addiction is a paradoxical resistance phenomenon, well-described in MEK-ERK-driven solid tumors, in which drug-target overexpression promotes resistance but a toxic overdose of signaling if the inhibitor is withdrawn. This can permit prolonged control of tumors through intermittent dosing. We and others showed previously that cancer drug addiction arises also in the hematologic malignancy ALK-positive anaplastic large-cell lymphoma (ALCL) resistant to ALK-specific tyrosine kinase inhibitors (TKIs). This is driven by the overexpression of the fusion kinase NPM1-ALK, but the mechanism by which ALK overactivity drives toxicity upon TKI withdrawal remained obscure. Here we reveal the mechanism of ALK-TKI addiction in ALCL. We interrogated the well-described mechanism of MEK/ERK pathway inhibitor addiction in solid tumors and found it does not apply to ALCL. Instead, phosphoproteomics and confirmatory functional studies revealed that the STAT1 overactivation is the key mechanism of ALK-TKI addiction in ALCL. The withdrawal of TKI from addicted tumors in vitro and in vivo leads to overwhelming phospho-STAT1 activation, turning on its tumor-suppressive gene-expression program and turning off STAT3's oncogenic program. Moreover, a novel NPM1-ALK-positive ALCL PDX model showed a significant survival benefit from intermittent compared with continuous TKI dosing. In sum, we reveal for the first time the mechanism of cancer drug addiction in ALK-positive ALCL and the benefit of scheduled intermittent dosing in high-risk patient-derived tumors in vivo.


Asunto(s)
Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Resistencia a Antineoplásicos , Linfoma Anaplásico de Células Grandes/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Quinasa de Linfoma Anaplásico/genética , Quinasa de Linfoma Anaplásico/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma Anaplásico de Células Grandes/enzimología , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/metabolismo , Nucleofosmina , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteómica , Factor de Transcripción STAT3/genética
15.
Semin Hematol ; 56(1): 52-57, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30573045

RESUMEN

Mass spectrometry-based techniques now enable the unbiased identification of proteins in complex mixtures including proteins isolated from cells and tissues. These powerful tools permit near-complete annotation of proteins expressed in cells, tissues or organs. Further, these techniques permit the interrogation of the numerous posttranslational modifications that govern cell-specific responses to signaling cues and underlie the functional heterogeneity of cellular composition and contribute to biological complexity. Parallel developments in technologies such as mass cytometry and multicolor ion-beam imaging which permit multi-parameter detection of numerous proteins at the single-cell and in situ level respectively, are poised to radically impact our understanding of the functional and translational importance of proteins in hematologic conditions. Importantly, the field of proteomics is poised to realize the immensely powerful opportunities in integration with genomic information that is being discovered at an unprecedented pace for many hematologic conditions.


Asunto(s)
Genómica/métodos , Hematología/métodos , Espectrometría de Masas/métodos , Proteómica/métodos , Humanos
17.
Cancer Chemother Pharmacol ; 61(5): 855-63, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17639395

RESUMEN

INTRODUCTION: The cytotoxic activity of the farnesyltranseferase inhibitor R115777 was evaluated in cell lines representative of mantle cell lymphoma (MCL). METHODS: Cell growth, proliferation, and apoptosis were analyzed in four human MCL cell lines (Granta, NCEB, REC, and UPN1) in presence of R115777, alone or in combination with vincristin, doxorubicin, bortezomib, cisplatin and cytarabine. Inhibition of farnesylation was determined by the appearance of prelamin A. The antitumor activity of R115777, administered p.o. at 100, 250 and 500 mg/kg, was determined in vivo in nude mice xenografted with UPN1 cells. RESULTS: R115777 inhibited the growth of MCL cell lines in vitro with inhibitory concentrations ranging between 2 and 15 nM. A fifty percent decrease of cell viability was observed at concentrations comprised between 0.08 and 17 microM. Apoptosis, evaluated by annexin V and activated caspase 3 staining, was induced in all cell lines, in 40 to 71% of the cells depending on the cell lines. In addition, R115777 significantly increased the cytotoxic effect of vincristine, doxorubicin, bortezomib, cisplatin and cytarabine (p=0.001, p=0.016, p=0.006, p=0.014 and p=0.007 respectively). Exposure of MCL cell lines to R115777 during 72 hours resulted in inhibition of protein farnesylation. R115777 administered p.o. twice daily for 8 consecutive days to mice bearing established s.c. UPN1 xenograft displayed cytostatic activity at the 500 mg/kg dosage. CONCLUSION: We have demonstrated that inhibition of farnesyltransferase by R115777 was associated with growth inhibition and apoptosis of MCL cell lines in vitro and tumor xenograft stability in vivo.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Linfoma de Células del Manto/tratamiento farmacológico , Quinolonas/farmacología , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Farnesiltransferasa/antagonistas & inhibidores , Femenino , Humanos , Lamina Tipo A , Ratones , Ratones Desnudos , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/metabolismo , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Quinolonas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Clin Oncol ; 35(28): 3215-3221, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28787259

RESUMEN

Purpose Fusions involving the ALK gene are the predominant genetic lesion underlying pediatric anaplastic large cell lymphomas (ALCL) and inflammatory myofibroblastic tumors (IMTs). We assessed the activity of the ALK inhibitor crizotinib in patients who had no known curative treatment options at diagnosis or with relapsed/recurrent disease. Methods In this study, 26 patients with relapsed/refractory ALK-positive ALCL and 14 patients with metastatic or inoperable ALK-positive IMT received crizotinib orally twice daily. Study objectives were measurement of efficacy and safety. Correlative studies evaluated the serial detection of NPM-ALK fusion transcripts in patients with ALCL. Results The overall response rates for patients with ALCL treated at doses of 165 (ALCL165) and 280 (ALCL280) mg/m2 were 83% and 90%, respectively. The overall response rate for patients with IMT (treated at 100, 165, and 280 mg/m2/dose) was 86%. A complete response was observed in 83% (five of six) of ALCL165, 80% (16 of 20) of ALCL280, and 36% (five of 14) of patients with IMT. Partial response rates were 0% (none of six), 10% (two of 20), and 50% (seven of 14), respectively. The median duration of therapy was 2.79, 0.4, and 1.63 years, respectively, with 12 patients ceasing protocol therapy to proceed to transplantation. The most common drug-related adverse event was decrease in neutrophil count in 33% and 70% of the ALCL165 and ALCL280 groups, respectively, and in 43% of patients with IMT. Levels of NPM-ALK decreased during therapy in most patients with ALCL. Conclusion The robust and sustained clinical responses to crizotinib therapy in patients with relapsed ALCL and metastatic or unresectable IMT highlight the importance of the ALK pathway in these diseases.


Asunto(s)
Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Neoplasias de Tejido Muscular/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Piridinas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Adolescente , Quinasa de Linfoma Anaplásico , Niño , Preescolar , Crizotinib , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Linfoma Anaplásico de Células Grandes/enzimología , Masculino , Terapia Molecular Dirigida , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/enzimología , Neoplasias de Tejido Muscular/enzimología , Inhibidores de Proteínas Quinasas/efectos adversos , Pirazoles/efectos adversos , Piridinas/efectos adversos , Proteínas Tirosina Quinasas Receptoras/metabolismo
20.
Oncotarget ; 8(69): 113895-113909, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29371955

RESUMEN

We recently demonstrated that obinutuzumab (GA101), a novel glycoengineered type II CD20 Ab compared to rituximab (RTX) mediates significantly enhanced antibody-dependent cell cytotoxicity (ADCC) in vitro and increased overall survival in a Burkitt lymphoma (BL) xenograft non-obese diabetic severe combined immunodeficiency gamma (NSG) model. In this study we compared the phosphoproteomic changes by pathway analysis following obinutuzumab vs RTX against RTX-sensitive (Raji) and -resistant BL (Raji4RH). Phosphoproteomic analyses were performed by mass-spectrometry (MS)-based label-free quantitative phosphoproteomic profiling. We demonstrated that 418 proteins in Raji and 377 proteins in Raji 4RH, were differentially phosphorylated (>1.5-fold) after obinutuzumab vs. RTX. Proteins that were significantly differentially phosphorylated included the B cell antigen receptor (BCR) (PLCG2, BTK and GSK3B), Fc gamma phagocytosis (FCRG2B, MAPK1, PLCG2 and RAF1), and natural killer cell-mediated cytotoxicity (MAPK1, RAF1, PLCG2 and MAPK3) signaling pathways. Differential phosphorylation of BCR or cytotoxicity pathway proteins revealed significant up-regulation of BTK, PLCY2 and ERK1/RAF1 after obinutuzumab compared to RTX. Silencing of PLCG2 in the BCR and MAPK1 in the cytotoxicity pathway significantly increased BL proliferation and decreased BL cytotoxicity after obinutuzumab compared to RTX. These results in combination with our previous results demonstrating a significant improvement in in vitro BL cytotoxicity and in vivo BL survival by obinutuzumab compared to RTX may in part be due to differential effects on selected BL protein signaling pathways.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA