Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
bioRxiv ; 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38798673

RESUMEN

Tumors frequently harbor isogenic yet epigenetically distinct subpopulations of multi-potent cells with high tumor-initiating potential-often called Cancer Stem-Like Cells (CSLCs). These can display preferential resistance to standard-of-care chemotherapy. Single-cell analyses can help elucidate Master Regulator (MR) proteins responsible for governing the transcriptional state of these cells, thus revealing complementary dependencies that may be leveraged via combination therapy. Interrogation of single-cell RNA sequencing profiles from seven metastatic breast cancer patients, using perturbational profiles of clinically relevant drugs, identified drugs predicted to invert the activity of MR proteins governing the transcriptional state of chemoresistant CSLCs, which were then validated by CROP-seq assays. The top drug, the anthelmintic albendazole, depleted this subpopulation in vivo without noticeable cytotoxicity. Moreover, sequential cycles of albendazole and paclitaxel-a commonly used chemotherapeutic -displayed significant synergy in a patient-derived xenograft (PDX) from a TNBC patient, suggesting that network-based approaches can help develop mechanism-based combinatorial therapies targeting complementary subpopulations.

2.
bioRxiv ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37873470

RESUMEN

The Mechanism of Action (MoA) of a drug is generally represented as a small, non-tissue-specific repertoire of high-affinity binding targets. Yet, drug activity and polypharmacology are increasingly associated with a broad range of off-target and tissue-specific effector proteins. To address this challenge, we have implemented an efficient integrative experimental and computational framework leveraging the systematic generation and analysis of drug perturbational profiles representing >700 FDA-approved and experimental oncology drugs, in cell lines selected as high-fidelity models of 23 aggressive tumor subtypes. Protein activity-based analyses revealed highly reproducible, drug-mediated modulation of tissue-specific targets, leading to generation of a proteome-wide polypharmacology map, characterization of MoA-related drug clusters and off-target effects, and identification and experimental validation of novel, tissue-specific inhibitors of undruggable oncoproteins. The proposed framework, which is easily extended to elucidating the MoA of novel small-molecule libraries, could help support more systematic and quantitative approaches to precision oncology.

3.
Cancer Discov ; 13(6): 1386-1407, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37061969

RESUMEN

Predicting in vivo response to antineoplastics remains an elusive challenge. We performed a first-of-kind evaluation of two transcriptome-based precision cancer medicine methodologies to predict tumor sensitivity to a comprehensive repertoire of clinically relevant oncology drugs, whose mechanism of action we experimentally assessed in cognate cell lines. We enrolled patients with histologically distinct, poor-prognosis malignancies who had progressed on multiple therapies, and developed low-passage, patient-derived xenograft models that were used to validate 35 patient-specific drug predictions. Both OncoTarget, which identifies high-affinity inhibitors of individual master regulator (MR) proteins, and OncoTreat, which identifies drugs that invert the transcriptional activity of hyperconnected MR modules, produced highly significant 30-day disease control rates (68% and 91%, respectively). Moreover, of 18 OncoTreat-predicted drugs, 15 induced the predicted MR-module activity inversion in vivo. Predicted drugs significantly outperformed antineoplastic drugs selected as unpredicted controls, suggesting these methods may substantively complement existing precision cancer medicine approaches, as also illustrated by a case study. SIGNIFICANCE: Complementary precision cancer medicine paradigms are needed to broaden the clinical benefit realized through genetic profiling and immunotherapy. In this first-in-class application, we introduce two transcriptome-based tumor-agnostic systems biology tools to predict drug response in vivo. OncoTarget and OncoTreat are scalable for the design of basket and umbrella clinical trials. This article is highlighted in the In This Issue feature, p. 1275.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Transcriptoma , Medicina de Precisión/métodos , Oncología Médica/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
4.
Med ; 3(11): 774-791.e7, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36195086

RESUMEN

BACKGROUND: Malignant rhabdoid tumors (MRTs) and Wilms' tumors (WTs) are rare and aggressive renal tumors of infants and young children comprising ∼5% of all pediatric cancers. MRTs are among the most genomically stable cancers, and although WTs are genomically heterogeneous, both generally lack therapeutically targetable genetic mutations. METHODS: Comparative protein activity analysis of MRTs (n = 68) and WTs (n = 132) across TCGA and TARGET cohorts, using metaVIPER, revealed elevated exportin 1 (XPO1) inferred activity. In vitro studies were performed on a panel of MRT and WT cell lines to evaluate effects on proliferation and cell-cycle progression following treatment with the selective XPO1 inhibitor selinexor. In vivo anti-tumor activity was assessed in patient-derived xenograft (PDX) models of MRTs and WTs. FINDINGS: metaVIPER analysis identified markedly aberrant activation of XPO1 in MRTs and WTs compared with other tumor types. All MRT and most WT cell lines demonstrated baseline, aberrant XPO1 activity with in vitro sensitivity to selinexor via cell-cycle arrest and induction of apoptosis. In vivo, XPO1 inhibitors significantly abrogated tumor growth in PDX models, inducing effective disease control with sustained treatment. Corroborating human relevance, we present a case report of a child with multiply relapsed WTs with prolonged disease control on selinexor. CONCLUSIONS: We report on a novel systems-biology-based comparative framework to identify non-genetically encoded vulnerabilities in genomically quiescent pediatric cancers. These results have provided preclinical rationale for investigation of XPO1 inhibitors in an upcoming investigator-initiated clinical trial of selinexor in children with MRTs and WTs and offer opportunities for exploration of inferred XPO1 activity as a potential predictive biomarker for response. FUNDING: This work was funded by CureSearch for Children's Cancer, Alan B. Slifka Foundation, NIH (U01 CA217858, S10 OD012351, and S10 OD021764), Michael's Miracle Cure, Hyundai Hope on Wheels, Cannonball Kids Cancer, Conquer Cancer the ASCO Foundation, Cycle for Survival, Paulie Strong Foundation, and the Grayson Fund.


Asunto(s)
Neoplasias Renales , Niño , Humanos , Preescolar , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Renales/tratamiento farmacológico , Proteína Exportina 1
6.
Nat Genet ; 50(7): 979-989, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29915428

RESUMEN

We introduce and validate a new precision oncology framework for the systematic prioritization of drugs targeting mechanistic tumor dependencies in individual patients. Compounds are prioritized on the basis of their ability to invert the concerted activity of master regulator proteins that mechanistically regulate tumor cell state, as assessed from systematic drug perturbation assays. We validated the approach on a cohort of 212 gastroenteropancreatic neuroendocrine tumors (GEP-NETs), a rare malignancy originating in the pancreas and gastrointestinal tract. The analysis identified several master regulator proteins, including key regulators of neuroendocrine lineage progenitor state and immunoevasion, whose role as critical tumor dependencies was experimentally confirmed. Transcriptome analysis of GEP-NET-derived cells, perturbed with a library of 107 compounds, identified the HDAC class I inhibitor entinostat as a potent inhibitor of master regulator activity for 42% of metastatic GEP-NET patients, abrogating tumor growth in vivo. This approach may thus complement current efforts in precision oncology.


Asunto(s)
Antineoplásicos/farmacología , Tumores Neuroendocrinos/tratamiento farmacológico , Benzamidas/farmacología , Línea Celular Tumoral , Estudios de Cohortes , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Medicina de Precisión/métodos , Piridinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética
7.
Nat Genet ; 43(9): 830-7, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804550

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common form of human lymphoma. Although a number of structural alterations have been associated with the pathogenesis of this malignancy, the full spectrum of genetic lesions that are present in the DLBCL genome, and therefore the identity of dysregulated cellular pathways, remains unknown. By combining next-generation sequencing and copy number analysis, we show that the DLBCL coding genome contains, on average, more than 30 clonally represented gene alterations per case. This analysis also revealed mutations in genes not previously implicated in DLBCL pathogenesis, including those regulating chromatin methylation (MLL2; 24% of samples) and immune recognition by T cells. These results provide initial data on the complexity of the DLBCL coding genome and identify novel dysregulated pathways underlying its pathogenesis.


Asunto(s)
Dosificación de Gen , Regulación Leucémica de la Expresión Génica , Linfoma de Células B Grandes Difuso/genética , Cromatina/metabolismo , Análisis Mutacional de ADN , Diploidia , Genoma Humano , Centro Germinal/inmunología , Humanos , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Metilación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Mutación Puntual , Polimorfismo de Nucleótido Simple , Linfocitos T/inmunología
8.
J Exp Med ; 208(7): 1389-401, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21670202

RESUMEN

The pathogenesis of chronic lymphocytic leukemia (CLL), the most common leukemia in adults, is still largely unknown. The full spectrum of genetic lesions that are present in the CLL genome, and therefore the number and identity of dysregulated cellular pathways, have not been identified. By combining next-generation sequencing and copy number analysis, we show here that the typical CLL coding genome contains <20 clonally represented gene alterations/case, including predominantly nonsilent mutations, and fewer copy number aberrations. These analyses led to the discovery of several genes not previously known to be altered in CLL. Although most of these genes were affected at low frequency in an expanded CLL screening cohort, mutational activation of NOTCH1, observed in 8.3% of CLL at diagnosis, was detected at significantly higher frequency during disease progression toward Richter transformation (31.0%), as well as in chemorefractory CLL (20.8%). Consistent with the association of NOTCH1 mutations with clinically aggressive forms of the disease, NOTCH1 activation at CLL diagnosis emerged as an independent predictor of poor survival. These results provide initial data on the complexity of the CLL coding genome and identify a dysregulated pathway of diagnostic and therapeutic relevance.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/genética , Mutación , Receptor Notch1/genética , Adulto , Anciano , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Genoma Humano , Humanos , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Pronóstico , Insuficiencia del Tratamiento
9.
Nature ; 471(7337): 189-95, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21390126

RESUMEN

B-cell non-Hodgkin's lymphoma comprises biologically and clinically distinct diseases the pathogenesis of which is associated with genetic lesions affecting oncogenes and tumour-suppressor genes. We report here that the two most common types--follicular lymphoma and diffuse large B-cell lymphoma--harbour frequent structural alterations inactivating CREBBP and, more rarely, EP300, two highly related histone and non-histone acetyltransferases (HATs) that act as transcriptional co-activators in multiple signalling pathways. Overall, about 39% of diffuse large B-cell lymphoma and 41% of follicular lymphoma cases display genomic deletions and/or somatic mutations that remove or inactivate the HAT coding domain of these two genes. These lesions usually affect one allele, suggesting that reduction in HAT dosage is important for lymphomagenesis. We demonstrate specific defects in acetylation-mediated inactivation of the BCL6 oncoprotein and activation of the p53 tumour suppressor. These results identify CREBBP/EP300 mutations as a major pathogenetic mechanism shared by common forms of B-cell non-Hodgkin's lymphoma, with direct implications for the use of drugs targeting acetylation/deacetylation mechanisms.


Asunto(s)
Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Proteína de Unión a CREB/genética , Proteína p300 Asociada a E1A/genética , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Mutación/genética , Acetilcoenzima A/metabolismo , Acetilación , Acetiltransferasas/química , Acetiltransferasas/deficiencia , Animales , Secuencia de Bases , Proteína de Unión a CREB/química , Proteína de Unión a CREB/deficiencia , Proteína de Unión a CREB/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Proteína p300 Asociada a E1A/química , Proteína p300 Asociada a E1A/deficiencia , Proteína p300 Asociada a E1A/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Histona Acetiltransferasas/química , Histona Acetiltransferasas/deficiencia , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Linfoma de Células B/patología , Linfoma Folicular/enzimología , Linfoma Folicular/genética , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Mutación Missense/genética , Polimorfismo de Nucleótido Simple/genética , Unión Proteica , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas c-bcl-6 , Recurrencia , Eliminación de Secuencia/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
Nature ; 459(7247): 717-21, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19412164

RESUMEN

Diffuse large B-cell lymphoma (DLBCL), the most common form of lymphoma in adulthood, comprises multiple biologically and clinically distinct subtypes including germinal centre B-cell-like (GCB) and activated B-cell-like (ABC) DLBCL. Gene expression profile studies have shown that its most aggressive subtype, ABC-DLBCL, is associated with constitutive activation of the NF-kappaB transcription complex. However, except for a small fraction of cases, it remains unclear whether NF-kappaB activation in these tumours represents an intrinsic program of the tumour cell of origin or a pathogenetic event. Here we show that >50% of ABC-DLBCL and a smaller fraction of GCB-DLBCL carry somatic mutations in multiple genes, including negative (TNFAIP3, also called A20) and positive (CARD11, TRAF2, TRAF5, MAP3K7 (TAK1) and TNFRSF11A (RANK)) regulators of NF-kappaB. Of these, the A20 gene, which encodes a ubiquitin-modifying enzyme involved in termination of NF-kappaB responses, is most commonly affected, with approximately 30% of patients displaying biallelic inactivation by mutations and/or deletions. When reintroduced in cell lines carrying biallelic inactivation of the gene, A20 induced apoptosis and cell growth arrest, indicating a tumour suppressor role. Less frequently, missense mutations of TRAF2 and CARD11 produce molecules with significantly enhanced ability to activate NF-kappaB. Thus, our results demonstrate that NF-kappaB activation in DLBCL is caused by genetic lesions affecting multiple genes, the loss or activation of which may promote lymphomagenesis by leading to abnormally prolonged NF-kappaB responses.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes/genética , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/fisiopatología , Mutación/genética , FN-kappa B/metabolismo , Apoptosis , Línea Celular Tumoral , Proteínas de Unión al ADN , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Proteínas Nucleares/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
11.
J Exp Med ; 203(2): 311-7, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16492805

RESUMEN

PR domain containing 1 with zinc finger domain (PRDM1)/B lymphocyte-induced maturation protein 1 (BLIMP1) is a transcriptional repressor expressed in a subset of germinal center (GC) B cells and in all plasma cells, and required for terminal B cell differentiation. The BLIMP1 locus lies on chromosome 6q21-q22.1, a region frequently deleted in B cell lymphomas, suggesting that it may harbor a tumor suppressor gene. We report here that the BLIMP1 gene is inactivated by structural alterations in 24% (8 out of 34) activated B cell-like diffuse large cell lymphoma (ABC-DLBCL), but not in GC B cell-like (n = 0/37) or unclassified (n = 0/21) DLBCL. BLIMP1 alterations included gene truncations, nonsense mutations, frameshift deletions, and splice site mutations that generate aberrant transcripts encoding truncated BLIMP1 proteins. In all cases studied, both BLIMP1 alleles were inactivated by deletions or mutations. Furthermore, most non-GC type DLBCL cases (n = 20/26, 77%) lack BLIMP1 protein expression, despite the presence of BLIMP1 mRNA. These results indicate that a sizable fraction of ABC-DLBCL carry an inactive BLIMP1 gene, and suggest that the same gene is inactivated by epigenetic mechanisms in an additional large number of cases. These findings point to a role for BLIMP1 as a tumor suppressor gene, whose inactivation may contribute to lymphomagenesis by blocking post-GC differentiation of B cells toward plasma cells.


Asunto(s)
Silenciador del Gen , Linfoma de Células B/genética , Linfoma de Células B Grandes Difuso/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Linfocitos B/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Codón sin Sentido , Mutación del Sistema de Lectura , Genes Supresores de Tumor , Humanos , Linfoma de Células B/clasificación , Linfoma de Células B/metabolismo , Linfoma de Células B Grandes Difuso/clasificación , Linfoma de Células B Grandes Difuso/metabolismo , Células Plasmáticas/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Sitios de Empalme de ARN/genética , Proteínas Represoras/biosíntesis , Factores de Transcripción/biosíntesis
12.
Am J Hum Genet ; 73(4): 886-97, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-13680528

RESUMEN

We previously reported a genomewide scan to identify autism-susceptibility loci in 110 multiplex families, showing suggestive evidence (P <.01) for linkage to autism-spectrum disorders (ASD) on chromosomes 5, 8, 16, 19, and X and showing nominal evidence (P <.05) on several additional chromosomes (2, 3, 4, 10, 11, 12, 15, 18, and 20). In this follow-up analysis we have increased the sample size threefold, while holding the study design constant, so that we now report 345 multiplex families, each with at least two siblings affected with autism or ASD phenotype. Along with 235 new multiplex families, 73 new microsatellite markers were also added in 10 regions, thereby increasing the marker density at these strategic locations from 10 cM to approximately 2 cM and bringing the total number of markers to 408 over the entire genome. Multipoint maximum LOD scores (MLS) obtained from affected-sib-pair analysis of all 345 families yielded suggestive evidence for linkage on chromosomes 17, 5, 11, 4, and 8 (listed in order by MLS) (P <.01). The most significant findings were an MLS of 2.83 (P =.00029) on chromosome 17q, near the serotonin transporter (5-hydroxytryptamine transporter [5-HTT]), and an MLS of 2.54 (P =.00059) on 5p. The present follow-up genome scan, which used a consistent research design across studies and examined the largest ASD sample collection reported to date, gave either equivalent or marginally increased evidence for linkage at several chromosomal regions implicated in our previous scan but eliminated evidence for linkage at other regions.


Asunto(s)
Trastorno Autístico/genética , Mapeo Cromosómico , Predisposición Genética a la Enfermedad , Trastorno Autístico/diagnóstico , Familia , Frecuencia de los Genes , Pruebas Genéticas/métodos , Genoma Humano , Genotipo , Humanos , Modelos Genéticos , Reproducibilidad de los Resultados
13.
Am J Hum Genet ; 70(1): 51-9, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11715113

RESUMEN

Celiac disease (CD), or gluten-sensitive enteropathy, is a common multifactorial disorder resulting from intolerance to cereal prolamins. The only established genetic susceptibility factor is HLA-DQ, which appears to explain only part of the overall genetic risk. We performed a genomewide scan of CD in 60 Finnish families. In addition to strong evidence for linkage to the HLA region at 6p21.3 (Z(max)>5), suggestive evidence for linkage was found for six other chromosomal regions--1p36, 4p15, 5q31, 7q21, 9p21-23, and 16q12. We further analyzed the three most convincing regions--4p15, 5q31, and 7q21--by evaluation of dense marker arrays across each region and by analysis of an additional 38 families. Although multipoint analysis with dense markers provided supportive evidence (multipoint LOD scores 3.25 at 4p15, 1.49 at 5q31, and 1.04 at 7q21) for the initial findings, the additional 38 families did not strengthen evidence for linkage. The role that HLA-DQ plays was studied in more detail by analysis of DQB1 alleles in all 98 families. All but one patient carried one or two HLA-DQ risk alleles, and 65% of HLA-DQ2 carriers were affected. Our study indicates that the HLA region harbors a predominant CD-susceptibility locus in these Finnish families.


Asunto(s)
Enfermedad Celíaca/genética , Ligamiento Genético/genética , Predisposición Genética a la Enfermedad/genética , Adulto , Alelos , Mapeo Cromosómico , Cromosomas Humanos/genética , Finlandia , Frecuencia de los Genes/genética , Marcadores Genéticos/genética , Genoma Humano , Antígenos HLA-DQ/genética , Cadenas beta de HLA-DQ , Humanos , Escala de Lod
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...