Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
NPJ Precis Oncol ; 5(1): 60, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183722

RESUMEN

Despite recent advancements in the treatment of multiple myeloma (MM), nearly all patients ultimately relapse and many become refractory to multiple lines of therapies. Therefore, we not only need the ability to predict which patients are at high risk for disease progression but also a means to understand the mechanisms underlying their risk. Here, we report a transcriptional regulatory network (TRN) for MM inferred from cross-sectional multi-omics data from 881 patients that predicts how 124 chromosomal abnormalities and somatic mutations causally perturb 392 transcription regulators of 8549 genes to manifest in distinct clinical phenotypes and outcomes. We identified 141 genetic programs whose activity profiles stratify patients into 25 distinct transcriptional states and proved to be more predictive of outcomes than did mutations. The coherence of these programs and accuracy of our network-based risk prediction was validated in two independent datasets. We observed subtype-specific vulnerabilities to interventions with existing drugs and revealed plausible mechanisms for relapse, including the establishment of an immunosuppressive microenvironment. Investigation of the t(4;14) clinical subtype using the TRN revealed that 16% of these patients exhibit an extreme-risk combination of genetic programs (median progression-free survival of 5 months) that create a distinct phenotype with targetable genes and pathways.

2.
PLoS Med ; 17(11): e1003323, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33147277

RESUMEN

BACKGROUND: The tumor microenvironment (TME) is increasingly appreciated as an important determinant of cancer outcome, including in multiple myeloma (MM). However, most myeloma microenvironment studies have been based on bone marrow (BM) aspirates, which often do not fully reflect the cellular content of BM tissue itself. To address this limitation in myeloma research, we systematically characterized the whole bone marrow (WBM) microenvironment during premalignant, baseline, on treatment, and post-treatment phases. METHODS AND FINDINGS: Between 2004 and 2019, 998 BM samples were taken from 436 patients with newly diagnosed MM (NDMM) at the University of Arkansas for Medical Sciences in Little Rock, Arkansas, United States of America. These patients were 61% male and 39% female, 89% White, 8% Black, and 3% other/refused, with a mean age of 58 years. Using WBM and matched cluster of differentiation (CD)138-selected tumor gene expression to control for tumor burden, we identified a subgroup of patients with an adverse TME associated with 17 fewer months of progression-free survival (PFS) (95% confidence interval [CI] 5-29, 49-69 versus 70-82 months, χ2 p = 0.001) and 15 fewer months of overall survival (OS; 95% CI -1 to 31, 92-120 versus 113-129 months, χ2 p = 0.036). Using immunohistochemistry-validated computational tools that identify distinct cell types from bulk gene expression, we showed that the adverse outcome was correlated with elevated CD8+ T cell and reduced granulocytic cell proportions. This microenvironment develops during the progression of premalignant to malignant disease and becomes less prevalent after therapy, in which it is associated with improved outcomes. In patients with quantified International Staging System (ISS) stage and 70-gene Prognostic Risk Score (GEP-70) scores, taking the microenvironment into consideration would have identified an additional 40 out of 290 patients (14%, premutation p = 0.001) with significantly worse outcomes (PFS, 95% CI 6-36, 49-73 versus 74-90 months) who were not identified by existing clinical (ISS stage III) and tumor (GEP-70) criteria as high risk. The main limitations of this study are that it relies on computationally identified cell types and that patients were treated with thalidomide rather than current therapies. CONCLUSIONS: In this study, we observe that granulocyte signatures in the MM TME contribute to a more accurate prognosis. This implies that future researchers and clinicians treating patients should quantify TME components, in particular monocytes and granulocytes, which are often ignored in microenvironment studies.


Asunto(s)
Médula Ósea/patología , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Microambiente Tumoral , Adulto , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Pronóstico , Carga Tumoral
3.
Gigascience ; 9(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32696951

RESUMEN

BACKGROUND: Mechanistic models, when combined with pertinent data, can improve our knowledge regarding important molecular and cellular mechanisms found in cancer. These models make the prediction of tissue-level response to drug treatment possible, which can lead to new therapies and improved patient outcomes. Here we present a data-driven multiscale modeling framework to study molecular interactions between cancer, stromal, and immune cells found in the tumor microenvironment. We also develop methods to use molecular data available in The Cancer Genome Atlas to generate sample-specific models of cancer. RESULTS: By combining published models of different cells relevant to pancreatic ductal adenocarcinoma (PDAC), we built an agent-based model of the multicellular pancreatic tumor microenvironment, formally describing cell type-specific molecular interactions and cytokine-mediated cell-cell communications. We used an ensemble-based modeling approach to systematically explore how variations in the tumor microenvironment affect the viability of cancer cells. The results suggest that the autocrine loop involving EGF signaling is a key interaction modulator between pancreatic cancer and stellate cells. EGF is also found to be associated with previously described subtypes of PDAC. Moreover, the model allows a systematic exploration of the effect of possible therapeutic perturbations; our simulations suggest that reducing bFGF secretion by stellate cells will have, on average, a positive impact on cancer apoptosis. CONCLUSIONS: The developed framework allows model-driven hypotheses to be generated regarding therapeutically relevant PDAC states with potential molecular and cellular drivers indicating specific intervention strategies.


Asunto(s)
Algoritmos , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/patología , Susceptibilidad a Enfermedades , Modelos Biológicos , Comunicación Autocrina , Carcinoma Ductal Pancreático/metabolismo , Comunicación Celular/genética , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Especificidad de Órganos , Comunicación Paracrina , Fenotipo
4.
Sci Rep ; 10(1): 1915, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32024856

RESUMEN

Failure to clear antigens causes CD8+ T cells to become increasingly hypo-functional, a state known as exhaustion. We combined manually extracted information from published literature with gene expression data from diverse model systems to infer a set of molecular regulatory interactions that underpin exhaustion. Topological analysis and simulation modeling of the network suggests CD8+ T cells undergo 2 major transitions in state following stimulation. The time cells spend in the earlier pro-memory/proliferative (PP) state is a fixed and inherent property of the network structure. Transition to the second state is necessary for exhaustion. Combining insights from network topology analysis and simulation modeling, we predict the extent to which each node in our network drives cells towards an exhausted state. We demonstrate the utility of our approach by experimentally testing the prediction that drug-induced interference with EZH2 function increases the proportion of pro-memory/proliferative cells in the early days post-activation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Redes Reguladoras de Genes/inmunología , Modelos Inmunológicos , Animales , Linfocitos T CD8-positivos/metabolismo , Simulación por Computador , Conjuntos de Datos como Asunto , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , RNA-Seq , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología
5.
Leukemia ; 34(7): 1866-1874, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32060406

RESUMEN

While the past decade has seen meaningful improvements in clinical outcomes for multiple myeloma patients, a subset of patients does not benefit from current therapeutics for unclear reasons. Many gene expression-based models of risk have been developed, but each model uses a different combination of genes and often involves assaying many genes making them difficult to implement. We organized the Multiple Myeloma DREAM Challenge, a crowdsourced effort to develop models of rapid progression in newly diagnosed myeloma patients and to benchmark these against previously published models. This effort lead to more robust predictors and found that incorporating specific demographic and clinical features improved gene expression-based models of high risk. Furthermore, post-challenge analysis identified a novel expression-based risk marker, PHF19, which has recently been found to have an important biological role in multiple myeloma. Lastly, we show that a simple four feature predictor composed of age, ISS, and expression of PHF19 and MMSET performs similarly to more complex models with many more gene expression features included.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Ensayos Clínicos como Asunto/estadística & datos numéricos , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Modelos Estadísticos , Mieloma Múltiple/patología , Factores de Transcripción/metabolismo , Biomarcadores de Tumor/genética , Ciclo Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Bases de Datos Factuales , Conjuntos de Datos como Asunto , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
7.
Genet Med ; 19(5): 559-567, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27657682

RESUMEN

PURPOSE: Cell-free DNA (cfDNA) prenatal screening tests have been rapidly adopted into clinical practice, due in part to positive insurance coverage. We evaluated the framework payers used in making coverage decisions to describe a process that should be informative for other sequencing tests. METHODS: We analyzed coverage policies from the 19 largest US private payers with publicly available policies through February 2016, building from the University of California San Francisco TRANSPERS Payer Coverage Policy Registry. RESULTS: All payers studied cover cfDNA screening for detection of trisomies 21, 18, and 13 in high-risk, singleton pregnancies, based on robust clinical validity (CV) studies and modeled evidence of clinical utility (CU). Payers typically evaluated the evidence for each chromosomal abnormality separately, although results are offered as part of a panel. Starting in August 2015, 8 of the 19 payers also began covering cfDNA screening in average-risk pregnancies, citing recent CV studies and updated professional guidelines. Most payers attempted, but were unable, to independently assess analytic validity (AV). CONCLUSION: Payers utilized the standard evidentiary framework (AV/CV/CU) when evaluating cfDNA screening but varied in their interpretation of the sufficiency of the evidence. Professional guidelines, large CV studies, and decision analytic models regarding health outcomes appeared highly influential in coverage decisions.Genet Med advance online publication 22 September 2016.


Asunto(s)
Ácidos Nucleicos Libres de Células/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Diagnóstico Prenatal/métodos , Análisis de Secuencia de ADN/métodos , Trisomía/diagnóstico , Toma de Decisiones Clínicas , Toma de Decisiones , Síndrome de Down/diagnóstico , Síndrome de Down/genética , Femenino , Pruebas Genéticas , Humanos , Cobertura del Seguro , Embarazo , Sistema de Registros , Trisomía/genética , Síndrome de la Trisomía 13/diagnóstico , Síndrome de la Trisomía 13/genética , Síndrome de la Trisomía 18/diagnóstico , Síndrome de la Trisomía 18/genética
8.
J Investig Med ; 59(5): 746-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21441830

RESUMEN

Our understanding of human biology has increased tremendously for the last several decades, yet the pace at which these discoveries have translated into new therapies for patients has been frustratingly stagnant. Universities and academic health centers, as the major recipients of public investment in biomedical science, have an obligation to translate new knowledge into applications that confer human benefit. However, translating fundamental discoveries into practical applications is expensive and involves highly regulated steps with which few academic scientists have experience. Challenges in engaging universities and academic health centers in translational research include building the appropriate infrastructures for human investigation, training and stabilizing the careers of young scientists and physicians interested in the requisite work, educating academic investigators about the regulatory requirements inherent in successful therapeutic discovery and development, and finding more efficient ways to match good ideas with adequate funding resources. The purpose of this article is to examine the early-stage drug development process and evaluate the role that academia could play in it. Because interest in early-stage drug development grows among academic investigators, the need for more integrated partnerships among academia, government, and industry has become increasingly apparent.


Asunto(s)
Investigación Biomédica Traslacional/métodos , Investigación Biomédica Traslacional/tendencias , Centros Médicos Académicos , Investigación Biomédica/tendencias , Diseño de Fármacos , Humanos , Industrias , Investigadores , Transferencia de Tecnología , Terapéutica/tendencias , Investigación Biomédica Traslacional/economía , Universidades
9.
AMIA Annu Symp Proc ; : 1065, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17238684

RESUMEN

In a time-motion study conducted in a hospital that recently implemented barcode medication administration (BCMA) technology, we found that the BCMA system did not increase the amount of time nurses spend on medication administration activities, and did not compromise the amount of time nurses spent on direct care of patients. Our results should allay concerns regarding the impact of BCMA on nursing workflow.


Asunto(s)
Procesamiento Automatizado de Datos , Sistemas de Medicación en Hospital , Atención de Enfermería/organización & administración , Humanos , Proceso de Enfermería , Innovación Organizacional , Estudios de Tiempo y Movimiento
10.
Mol Cell Biol ; 23(9): 3043-51, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12697807

RESUMEN

Despite the central role of TATA-binding protein (TBP) in transcription, changes in cellular TBP concentration produce selective effects on gene expression. Moreover, TBP is up-regulated by oncogenic signaling pathways. These findings suggest that TBP could be a nexus in pathways that regulate cell proliferation and that genetic lesions that result in cellular transformation may produce their effects at least in part through TBP. We provide evidence consistent with this hypothesis, demonstrating that increases in TBP expression contribute to cellular transformation. A Ras-mediated increase in TBP expression is required for full Ras transforming activity. TBP overexpression induces cells to grow in an anchorage-independent manner and to form tumors in athymic mice. These effects on cellular transformation require changes in RNA polymerase II-dependent transcription and on the selective recruitment of TBP to promoters via its DNA binding activity. TBP expression is elevated in human colon carcinomas relative to normal colon epithelium. Both Ras-dependent and Ras-independent mechanisms mediate increases in TBP expression in colon carcinoma cell lines. We conclude that TBP may be a critical component in dysregulated signaling that occurs downstream of genetic lesions that cause tumors.


Asunto(s)
Carcinoma/genética , Carcinoma/metabolismo , Transformación Celular Neoplásica , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína de Unión a TATA-Box/metabolismo , Células 3T3/metabolismo , Células 3T3/patología , Animales , Pruebas de Carcinogenicidad , Células Cultivadas , Epitelio/metabolismo , Humanos , Ratones , Ratones Desnudos , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión a TATA-Box/genética , Regulación hacia Arriba , Proteínas ras/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA