Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Community Ment Health J ; 60(1): 115-123, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38105337

RESUMEN

This study sought to evaluate the impact of telepsychiatry during the COVID-19 pandemic among patients discharged from psychiatric inpatient units in the New York City Health and Hospitals Corporation system. We compared patients discharged to telepsychiatry (April 2020, n = 739) and in-person follow-up (May 2019, n = 527); we collected number, timing and attendance for follow-up appointments and number and timing of emergency room (ER) visits and readmissions. We used logistic regression to evaluate the odds of having these encounters and Kaplan-Meier analyses to compare time to these encounters. Patients discharged in 2020 were more likely to have a follow-up (29.4 vs. 19.9%, p < 0.001) and an ER visit or readmission (40.5 vs. 28.7%, p < 0.001). Kaplan-Meier analyses showed shorter time to first follow-up (chi-square = 14.69, d.f.=1, p < 0.0001, follow-ups = 322) and ER visit or readmission (chi-square = 19.57, d.f.=1, p < 0.0001, ER visits or admissions = 450) in the 2020 cohort. In multivariable analyses, patients discharged in 2020 were more likely to have a follow-up visit (adjusted OR 1.85, 95% confidence interval 1.40, 2.45, p < 0.0001). We found an increase in psychiatric service utilization during the pandemic, with an increase in and shorter time until outpatient visits and ER visits or readmissions. Although increased use of psychiatric services during the height of the COVID-19 pandemic is encouraging, it also points to the depth of the crisis among vulnerable populations; this pattern warrants further exploration and intervention.


Asunto(s)
COVID-19 , Psiquiatría , Telemedicina , Humanos , COVID-19/epidemiología , Pandemias , Ciudad de Nueva York/epidemiología , Estudios Retrospectivos
3.
Cancer Res ; 83(15): 2471-2479, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37289018

RESUMEN

The emergence of resistance to targeted therapies restrains their efficacy. The development of rationally guided drug combinations could overcome this currently insurmountable clinical challenge. However, our limited understanding of the trajectories that drive the outgrowth of resistant clones in cancer cell populations precludes design of drug combinations to forestall resistance. Here, we propose an iterative treatment strategy coupled with genomic profiling and genome-wide CRISPR activation screening to systematically extract and define preexisting resistant subpopulations in an EGFR-driven lung cancer cell line. Integrating these modalities identifies several resistance mechanisms, including activation of YAP/TAZ signaling by WWTR1 amplification, and estimates the associated cellular fitness for mathematical population modeling. These observations led to the development of a combination therapy that eradicated resistant clones in large cancer cell line populations by exhausting the spectrum of genomic resistance mechanisms. However, a small fraction of cancer cells was able to enter a reversible nonproliferative state of drug tolerance. This subpopulation exhibited mesenchymal properties, NRF2 target gene expression, and sensitivity to ferroptotic cell death. Exploiting this induced collateral sensitivity by GPX4 inhibition clears drug-tolerant populations and leads to tumor cell eradication. Overall, this experimental in vitro data and theoretical modeling demonstrate why targeted mono- and dual therapies will likely fail in sufficiently large cancer cell populations to limit long-term efficacy. Our approach is not tied to a particular driver mechanism and can be used to systematically assess and ideally exhaust the resistance landscape for different cancer types to rationally design combination therapies. SIGNIFICANCE: Unraveling the trajectories of preexisting resistant and drug-tolerant persister cells facilitates the rational design of multidrug combination or sequential therapies, presenting an approach to explore for treating EGFR-mutant lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Transducción de Señal , Receptores ErbB/metabolismo , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/farmacología , Mutación
4.
J Med Chem ; 65(9): 6643-6655, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35486541

RESUMEN

Despite the clinical efficacy of epidermal growth factor receptor (EGFR) inhibitors, a subset of patients with non-small cell lung cancer displays insertion mutations in exon20 in EGFR and Her2 with limited treatment options. Here, we present the development and characterization of the novel covalent inhibitors LDC8201 and LDC0496 based on a 1H-pyrrolo[2,3-b]pyridine scaffold. They exhibited intense inhibitory potency toward EGFR and Her2 exon20 insertion mutations as well as selectivity over wild type EGFR and within the kinome. Complex crystal structures with the inhibitors and biochemical and cellular on-target activity document their favorable binding characteristics. Ultimately, we observed tumor shrinkage in mice engrafted with patient-derived EGFR-H773_V774insNPH mutant cells during treatment with LDC8201. Together, these results highlight the potential of covalent pyrrolopyridines as inhibitors to target exon20 insertion mutations.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Mutagénesis Insercional , Mutación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
5.
Mol Cancer Ther ; 21(5): 821-830, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35247925

RESUMEN

NRG1 fusions are recurrent somatic genome alterations occurring across several tumor types, including invasive mucinous lung adenocarcinomas and pancreatic ductal adenocarcinomas and are potentially actionable genetic alterations in these cancers. We initially discovered CD74-NRG1 as the first NRG1 fusion in lung adenocarcinomas, and many additional fusion partners have since been identified. Here, we present the first CD74-NRG1 transgenic mouse model and provide evidence that ubiquitous expression of the CD74-NRG1 fusion protein in vivo leads to tumor development at high frequency. Furthermore, we show that ERBB2:ERBB3 heterodimerization is a mechanistic event in transformation by CD74-NRG1 binding physically to ERBB3 and that CD74-NRG1-expressing cells proliferate independent of supplemented NRG1 ligand. Thus, NRG1 gene fusions are recurrent driver oncogenes that cause oncogene dependency. Consistent with these findings, patients with NRG1 fusion-positive cancers respond to therapy targeting the ERBB2:ERBB3 receptors.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Animales , Carcinogénesis/genética , Humanos , Ratones , Neurregulina-1/genética , Oncogenes , Receptor ErbB-2/genética , Receptor ErbB-3/genética
6.
Nat Commun ; 12(1): 5505, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34535668

RESUMEN

Kinase inhibitors suppress the growth of oncogene driven cancer but also enforce the selection of treatment resistant cells that are thought to promote tumor relapse in patients. Here, we report transcriptomic and functional genomics analyses of cells and tumors within their microenvironment across different genotypes that persist during kinase inhibitor treatment. We uncover a conserved, MAPK/IRF1-mediated inflammatory response in tumors that undergo stemness- and senescence-associated reprogramming. In these tumor cells, activation of the innate immunity sensor RIG-I via its agonist IVT4, triggers an interferon and a pro-apoptotic response that synergize with concomitant kinase inhibition. In humanized lung cancer xenografts and a syngeneic Egfr-driven lung cancer model these effects translate into reduction of exhausted CD8+ T cells and robust tumor shrinkage. Overall, the mechanistic understanding of MAPK/IRF1-mediated intratumoral reprogramming may ultimately prolong the efficacy of targeted drugs in genetically defined cancer patients.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Inmunidad Innata , Inflamación/patología , Sistema de Señalización de MAP Quinasas , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Receptores Inmunológicos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citocinas/metabolismo , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Evasión Inmune/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Factor 1 Regulador del Interferón/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Oncogenes , Transducción de Señal/efectos de los fármacos
7.
Nat Commun ; 10(1): 3485, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31375684

RESUMEN

MYC paralogs are frequently activated in small cell lung cancer (SCLC) but represent poor drug targets. Thus, a detailed mapping of MYC-paralog-specific vulnerabilities may help to develop effective therapies for SCLC patients. Using a unique cellular CRISPR activation model, we uncover that, in contrast to MYCN and MYCL, MYC represses BCL2 transcription via interaction with MIZ1 and DNMT3a. The resulting lack of BCL2 expression promotes sensitivity to cell cycle control inhibition and dependency on MCL1. Furthermore, MYC activation leads to heightened apoptotic priming, intrinsic genotoxic stress and susceptibility to DNA damage checkpoint inhibitors. Finally, combined AURK and CHK1 inhibition substantially prolongs the survival of mice bearing MYC-driven SCLC beyond that of combination chemotherapy. These analyses uncover MYC-paralog-specific regulation of the apoptotic machinery with implications for genotype-based selection of targeted therapeutics in SCLC patients.


Asunto(s)
Apoptosis/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Carcinoma Pulmonar de Células Pequeñas/genética , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Terapia Molecular Dirigida/métodos , Proteínas Proto-Oncogénicas c-myc/genética , ARN Interferente Pequeño/metabolismo , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico
8.
Clin Cancer Res ; 24(6): 1337-1343, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29284707

RESUMEN

Purpose: We sought to investigate the clinical response to MET inhibition in patients diagnosed with structural MET alterations and to characterize their functional relevance in cellular models.Experimental Design: Patients were selected for treatment with crizotinib upon results of hybrid capture-based next-generation sequencing. To confirm the clinical observations, we analyzed cellular models that express these MET kinase alterations.Results: Three individual patients were identified to harbor alterations within the MET receptor. Two patients showed genomic rearrangements, leading to a gene fusion of KIF5B or STARD3NL and MET One patient diagnosed with an EML4-ALK rearrangement developed a MET kinase domain duplication as a resistance mechanism to ceritinib. All 3 patients showed a partial response to crizotinib that effectively inhibits MET and ALK among other kinases. The results were further confirmed using orthogonal cellular models.Conclusions: Crizotinib leads to a clinical response in patients with MET rearrangements. Our functional analyses together with the clinical data suggest that these structural alterations may represent actionable targets in lung cancer patients. Clin Cancer Res; 24(6); 1337-43. ©2017 AACR.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Adenocarcinoma del Pulmón/diagnóstico , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/metabolismo , Adulto , Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Crizotinib/farmacología , Crizotinib/uso terapéutico , Femenino , Duplicación de Gen , Reordenamiento Génico , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Estadificación de Neoplasias , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/química , Tomografía Computarizada por Rayos X
9.
Cell Rep ; 20(12): 2833-2845, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930680

RESUMEN

Kinase inhibitors represent the backbone of targeted cancer therapy, yet only a limited number of oncogenic drivers are directly druggable. By interrogating the activity of 1,505 kinase inhibitors, we found that BRD4-NUT-rearranged NUT midline carcinoma (NMC) cells are specifically killed by CDK9 inhibition (CDK9i) and depend on CDK9 and Cyclin-T1 expression. We show that CDK9i leads to robust induction of apoptosis and of markers of DNA damage response in NMC cells. While both CDK9i and bromodomain inhibition over time result in reduced Myc protein expression, only bromodomain inhibition induces cell differentiation and a p21-induced cell-cycle arrest in these cells. Finally, RNA-seq and ChIP-based analyses reveal a BRD4-NUT-specific CDK9i-induced perturbation of transcriptional elongation. Thus, our data provide a mechanistic basis for the genotype-dependent vulnerability of NMC cells to CDK9i that may be of relevance for the development of targeted therapies for NMC patients.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Ciclina T/metabolismo , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Inhibidores de Proteínas Quinasas/química , ARN Polimerasa II/metabolismo , Elongación de la Transcripción Genética/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
10.
Sci Transl Med ; 9(394)2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615362

RESUMEN

Oncogenic fusion events have been identified in a broad range of tumors. Among them, RET rearrangements represent distinct and potentially druggable targets that are recurrently found in lung adenocarcinomas. We provide further evidence that current anti-RET drugs may not be potent enough to induce durable responses in such tumors. We report that potent inhibitors, such as AD80 or ponatinib, that stably bind in the DFG-out conformation of RET may overcome these limitations and selectively kill RET-rearranged tumors. Using chemical genomics in conjunction with phosphoproteomic analyses in RET-rearranged cells, we identify the CCDC6-RETI788N mutation and drug-induced mitogen-activated protein kinase pathway reactivation as possible mechanisms by which tumors may escape the activity of RET inhibitors. Our data provide mechanistic insight into the druggability of RET kinase fusions that may be of help for the development of effective therapies targeting such tumors.


Asunto(s)
Adenocarcinoma/metabolismo , Reordenamiento Génico/genética , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-ret/genética , Adenocarcinoma del Pulmón , Animales , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Resistencia a Antineoplásicos/genética , Reordenamiento Génico/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Imidazoles/farmacología , Ratones , Mutación , Células 3T3 NIH , Piridazinas/farmacología
11.
J Affect Disord ; 198: 148-57, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27016658

RESUMEN

BACKGROUND: DGKH is a replicated risk gene of bipolar disorder (BD). However, the pathophysiological role of the coded protein, diacylglycerol kinase eta, remains elusive. METHODS: In this proof-of-concept study we isolated mRNA from peripheral blood and fibroblasts of heterozygote DGKH risk variants carriers (risk haplotype rs994856/rs9525580/rs9525584 GAT) with bipolar disorder and non-risk variant carriers with and without bipolar disorder. Gene expression of DGKH1, DGKH2, INPP5E, PI4K2B, PIK4CA, PLCG2, PRKCA, PRKCD, PRKCE and PRKCH was analysed by qRT PCR. RESULTS: DGKH1 expression was increased in peripheral blood of risk variant carriers (p=0.027). In fibroblast cells, PRKCD expression was significantly increased in DGKH GAT carriers (p=0.037). Patients with a current depressive episode had lower PRKCD levels and lithium treatment was associated with increased PRKCA expression (p=0.005, and p=0.033). LIMITATIONS: No homozygote risk variant carriers and no healthy risk variant carriers were included due to their infrequency. Bipolar patients carrying the GAT haplotype were older with marginal significance, as age had also an influence on DGKH expression levels but not on PRKCD levels, replication with better age-matched samples and also bigger samples are needed. CONCLUSIONS: The results add evidence for the role of fibroblast cells and peripheral blood as useful tools in the functional characterisation of risk gene variants. Also a combination of genotyping and peripheral gene expression analysis could proof useful in the search of biomarkers for endophenotypes. Furthermore, we could confirm the role of the inositol-1,4,5-triphosphate second messenger pathway and protein kinase C in the pathogenesis of BD.


Asunto(s)
Trastorno Bipolar/genética , Diacilglicerol Quinasa/genética , Expresión Génica/genética , Adulto , Anciano , Trastorno Bipolar/sangre , Diacilglicerol Quinasa/sangre , Femenino , Variación Genética/genética , Genotipo , Haplotipos/genética , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...