Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Emotion ; 24(1): 116-129, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37227830

RESUMEN

Individuals differ markedly in how they experience the ebb and flow of emotions. In this study, we used daily experience sampling to examine whether these differences reflect the nature and presence of mood disorders or whether they can better be characterized as distinct dynamic emotion profiles that cut-across diagnostic boundaries. We followed 105 individuals in 2019-2020 with diagnoses of major depression, remitted major depression, bipolar disorder, or no history of disorder, over 14 days (n = 6,543 experience-sampling assessments). We applied group iterative multiple model estimation, using both diagnosis-based and data-driven methods to investigate similarities in unfolding within-person emotion-network time-courses. Results did not support diagnosis-based subgroupings but rather revealed two significant data-driven subgroups based on dynamic emotion patterns. These data-driven subgroups did not significantly differ in terms of clinical features or demographics, but did differ on key emotion metrics-instability, granularity, and inertia. These data-driven subgroupings, agnostic to diagnostic status, provide insights into the nature of idiographic emotion-network dynamics that cut-across clinical diagnostic divisions. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Asunto(s)
Trastorno Bipolar , Evaluación Ecológica Momentánea , Humanos , Emociones , Trastornos del Humor/diagnóstico , Trastornos del Humor/psicología , Afecto , Trastorno Bipolar/diagnóstico , Trastorno Bipolar/psicología
2.
Clin Cancer Res ; 29(8): 1450-1459, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-36705597

RESUMEN

PURPOSE: Preclinical data showed that prophylactic, low-dose temozolomide (TMZ) significantly prevented breast cancer brain metastasis. We present results of a phase I trial combining T-DM1 with TMZ for the prevention of additional brain metastases after previous occurrence and local treatment in patients with HER2+ breast cancer. PATIENTS AND METHODS: Eligible patients had HER2+ breast cancer with brain metastases and were within 12 weeks of whole brain radiation therapy (WBRT), stereotactic radiosurgery, and/or surgery. Standard doses of T-DM1 were administered intravenously every 21 days (3.6 mg/kg) and TMZ was given orally daily in a 3+3 phase I dose escalation design at 30, 40, or 50 mg/m2, continuously. DLT period was one 21-day cycle. Primary endpoint was safety and recommended phase II dose. Symptom questionnaires, brain MRI, and systemic CT scans were performed every 6 weeks. Cell-free DNA sequencing was performed on patients' plasma and CSF. RESULTS: Twelve women enrolled, nine (75%) with prior SRS therapy and three (25%) with prior WBRT. Grade 3 or 4 AEs included thrombocytopenia (1/12), neutropenia (1/12), lymphopenia (6/12), and decreased CD4 (6/12), requiring pentamidine for Pneumocystis jirovecii pneumonia prophylaxis. No DLT was observed. Four patients on the highest TMZ dose underwent dose reductions. At trial entry, 6 of 12 patients had tumor mutations in CSF, indicating ongoing metastatic colonization despite a clear MRI. Median follow-up on study was 9.6 m (2.8-33.9); only 2 patients developed new parenchymal brain metastases. Tumor mutations varied with patient outcome. CONCLUSIONS: Metronomic TMZ in combination with standard dose T-DM1 shows low-grade toxicity and potential activity in secondary prevention of HER2+ brain metastases.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Ácidos Nucleicos Libres de Células , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Temozolomida/uso terapéutico , Prevención Secundaria , Receptor ErbB-2/genética , Receptor ErbB-2/uso terapéutico , Ado-Trastuzumab Emtansina/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario
3.
Bioinformatics ; 38(20): 4677-4686, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36040167

RESUMEN

MOTIVATION: Somatic copy-number alterations (SCNAs) play an important role in cancer development. Systematic noise in sequencing and array data present a significant challenge to the inference of SCNAs for cancer genome analyses. As part of The Cancer Genome Atlas, the Broad Institute Genome Characterization Center developed the Tangent normalization method to generate copy-number profiles using data from single-nucleotide polymorphism (SNP) arrays and whole-exome sequencing (WES) technologies for over 10 000 pairs of tumors and matched normal samples. Here, we describe the Tangent method, which uses a unique linear combination of normal samples as a reference for each tumor sample, to subtract systematic errors that vary across samples. We also describe a modification of Tangent, called Pseudo-Tangent, which enables denoising through comparisons between tumor profiles when few normal samples are available. RESULTS: Tangent normalization substantially increases signal-to-noise ratios (SNRs) compared to conventional normalization methods in both SNP array and WES analyses. Tangent and Pseudo-Tangent normalizations improve the SNR by reducing noise with minimal effect on signal and exceed the contribution of other steps in the analysis such as choice of segmentation algorithm. Tangent and Pseudo-Tangent are broadly applicable and enable more accurate inference of SCNAs from DNA sequencing and array data. AVAILABILITY AND IMPLEMENTATION: Tangent is available at https://github.com/broadinstitute/tangent and as a Docker image (https://hub.docker.com/r/broadinstitute/tangent). Tangent is also the normalization method for the copy-number pipeline in Genome Analysis Toolkit 4 (GATK4). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Neoplasias , Programas Informáticos , Humanos , Algoritmos , Variaciones en el Número de Copia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias/genética
4.
Cancer Immunol Res ; 10(8): 996-1012, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35706413

RESUMEN

Melanoma-derived brain metastases (MBM) represent an unmet clinical need because central nervous system progression is frequently an end stage of the disease. Immune checkpoint inhibitors (ICI) provide a clinical opportunity against MBM; however, the MBM tumor microenvironment (TME) has not been fully elucidated in the context of ICI. To dissect unique elements of the MBM TME and correlates of MBM response to ICI, we collected 32 fresh MBM and performed single-cell RNA sequencing of the MBM TME and T-cell receptor clonotyping on T cells from MBM and matched blood and extracranial lesions. We observed myeloid phenotypic heterogeneity in the MBM TME, most notably multiple distinct neutrophil states, including an IL8-expressing population that correlated with malignant cell epithelial-to-mesenchymal transition. In addition, we observed significant relationships between intracranial T-cell phenotypes and the distribution of T-cell clonotypes intracranially and peripherally. We found that the phenotype, clonotype, and overall number of MBM-infiltrating T cells were associated with response to ICI, suggesting that ICI-responsive MBMs interact with peripheral blood in a manner similar to extracranial lesions. These data identify unique features of the MBM TME that may represent potential targets to improve clinical outcomes for patients with MBM.


Asunto(s)
Neoplasias Encefálicas , Melanoma , Humanos , Inhibidores de Puntos de Control Inmunológico , Microambiente Tumoral
5.
Bioinformatics ; 38(8): 2096-2101, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35176131

RESUMEN

MOTIVATION: Cross-sectional analyses of primary cancer genomes have identified regions of recurrent somatic copy-number alteration, many of which result from positive selection during cancer formation and contain driver genes. However, no effective approach exists for identifying genomic loci under significantly different degrees of selection in cancers of different subtypes, anatomic sites or disease stages. RESULTS: CNGPLD is a new tool for performing case-control somatic copy-number analysis that facilitates the discovery of differentially amplified or deleted copy-number aberrations in a case group of cancer compared with a control group of cancer. This tool uses a Gaussian process statistical framework in order to account for the covariance structure of copy-number data along genomic coordinates and to control the false discovery rate at the region level. AVAILABILITY AND IMPLEMENTATION: CNGPLD is freely available at https://bitbucket.org/djhshih/cngpld as an R package. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genoma , Neoplasias , Humanos , Estudios Transversales , Genómica , Variaciones en el Número de Copia de ADN , Neoplasias/genética , Estudios de Casos y Controles , Programas Informáticos
7.
Nat Commun ; 12(1): 5955, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34642316

RESUMEN

Leptomeningeal disease (LMD) is a devastating complication of solid tumor malignancies, with dire prognosis and no effective systemic treatment options. Over the past decade, the incidence of LMD has steadily increased due to therapeutics that have extended the survival of cancer patients, highlighting the need for new interventions. To examine the efficacy of immune checkpoint inhibitors (ICI) in patients with LMD, we completed two phase II clinical trials. Here, we investigate the cellular and molecular features underpinning observed patient trajectories in these trials by applying single-cell RNA and cell-free DNA profiling to longitudinal cerebrospinal fluid (CSF) draws from enrolled patients. We recover immune and malignant cell types in the CSF, characterize cell behavior changes following ICI, and identify genomic features associated with relevant clinical phenomena. Overall, our study describes the liquid LMD tumor microenvironment prior to and following ICI treatment and demonstrates clinical utility of cell-free and single-cell genomic measurements for LMD research.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Antígeno CTLA-4/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Carcinomatosis Meníngea/tratamiento farmacológico , Neoplasias Meníngeas/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral/efectos de los fármacos , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/secundario , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/genética , Ácidos Nucleicos Libres de Células/genética , Ácidos Nucleicos Libres de Células/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Interferón gamma/genética , Interferón gamma/inmunología , Ipilimumab/uso terapéutico , Masculino , Carcinomatosis Meníngea/inmunología , Carcinomatosis Meníngea/mortalidad , Carcinomatosis Meníngea/patología , Neoplasias Meníngeas/inmunología , Neoplasias Meníngeas/mortalidad , Neoplasias Meníngeas/patología , Persona de Mediana Edad , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Análisis de la Célula Individual , Análisis de Supervivencia , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
8.
Nat Commun ; 12(1): 5954, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34642329

RESUMEN

Leptomeningeal disease (LMD) is a common complication from solid tumor malignancies with a poor prognosis and limited treatment options. We present a single arm Phase II study of 18 patients with LMD receiving combined ipilimumab and nivolumab until progression or unacceptable toxicity (NCT02939300). The primary end point is overall survival at 3 months (OS3). Secondary end points include toxicity, cumulative time-to-progression at 3 months, and progression-free survival. A Simon two-stage design is used to compare a null hypothesis OS3 of 18% against an alternative of 44%. Median follow up based on patients still alive is 8.0 months (range: 0.5 to 15.9 months). The study has met its primary endpoint as 8 of 18 (OS3 0.44; 90% CI: 0.24 to 0.66) patients are alive at three months. One third of patients have experienced one (or more) grade-3 or higher adverse events. Two patients have discontinued protocol treatment due to unacceptable toxicity (hepatitis and colitis, respectively). The most frequent adverse events include fatigue (N = 7), nausea (N = 6), fever (N = 6), anorexia (N = 6) and rash (N = 6). Combined ipilimumab and nivolumab has an acceptable safety profile and demonstrates promising activity in LMD patients. Larger, multicenter clinical trials are needed to validate these results.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Encefálicas/tratamiento farmacológico , Ipilimumab/administración & dosificación , Carcinomatosis Meníngea/tratamiento farmacológico , Neoplasias Meníngeas/tratamiento farmacológico , Nivolumab/administración & dosificación , Adulto , Anciano , Anorexia/inducido químicamente , Anorexia/mortalidad , Anorexia/patología , Antineoplásicos Inmunológicos/efectos adversos , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/secundario , Colitis/inducido químicamente , Colitis/mortalidad , Colitis/patología , Exantema/inducido químicamente , Exantema/mortalidad , Exantema/patología , Fatiga/inducido químicamente , Fatiga/mortalidad , Fatiga/patología , Femenino , Fiebre/inducido químicamente , Fiebre/mortalidad , Fiebre/patología , Hepatitis/etiología , Hepatitis/mortalidad , Hepatitis/patología , Humanos , Ipilimumab/efectos adversos , Masculino , Carcinomatosis Meníngea/mortalidad , Carcinomatosis Meníngea/patología , Neoplasias Meníngeas/mortalidad , Neoplasias Meníngeas/patología , Persona de Mediana Edad , Náusea/inducido químicamente , Náusea/mortalidad , Náusea/patología , Nivolumab/efectos adversos , Análisis de Supervivencia
9.
JAMA Netw Open ; 4(8): e2120040, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34369989

RESUMEN

Importance: Leptomeningeal disease (LMD) is a devastating complication of cancer that is frequently underdiagnosed owing to the low sensitivity of cerebrospinal fluid (CSF) cytologic assessment, the current benchmark diagnostic method. Improving diagnostic sensitivity may lead to improved treatment decisions. Objective: To assess whether cell-free DNA (cfDNA) analysis of CSF may be used to diagnose LMD more accurately than cytologic analysis. Design, Setting, and Participants: This diagnostic study conducted in a neuro-oncology clinic at 2 large, tertiary medical centers assessed the use of genomic sequencing of CSF samples obtained from 30 patients with suspected or confirmed LMD from 2015 through 2018 to identify tumor-derived cfDNA. From the same CSF samples, cytologic analyses were conducted, and the results of the 2 tests were compared. This study consisted of 2 patient populations: 22 patients with cytologically confirmed LMD without parenchymal tumors abutting their CSF and 8 patients with parenchymal brain metastases with no evidence of LMD. Patients were considered positive for the presence of LMD if previous CSF cytologic analysis was positive for malignant cells. The analysis was conducted from 2015 to 2018. Main Outcomes and Measures: The primary outcome was the diagnostic accuracy of cfDNA analysis, defined as the number of tests that resulted in correct diagnoses out of the total number of tests assayed. Hypotheses were formed before data collection. Results: In total, 30 patients (23 women [77%]; median age, 51 years [range, 28-81 years]), primarily presenting with metastatic solid malignant neoplasms, participated in this study. For 48 follow-up samples from patients previously diagnosed via cytologic analysis as having LMD with no parenchymal tumor abutting CSF, cfDNA findings were accurate in the assessment of LMD in 45 samples (94%; 95% CI, 83%-99%), whereas cytologic analysis was accurate in 36 samples (75%; 95% CI, 60%-86%), a significant difference (P = .02). Of 43 LMD-positive samples, CSF cfDNA analysis was sensitive to LMD in 40 samples (93%; 95% CI, 81%-99%), and cytologic analysis was sensitive to LMD in 31 samples (72%; 95% CI, 56%-85%), a significant difference (P = .02). For 3 patients with parenchymal brain metastases abutting the CSF and no suspicion of LMD, cytologic findings were negative for LMD in all 3 patients, whereas cfDNA findings were positive in all 3 patients. Conclusions and Relevance: This diagnostic study found improved sensitivity and accuracy of cfDNA CSF testing vs cytologic assessment for diagnosing LMD with the exception of parenchymal tumors abutting CSF, suggesting improved ability to diagnosis LMD. Consideration of incorporating CSF cfDNA analysis into clinical care is warranted.


Asunto(s)
ADN Tumoral Circulante/líquido cefalorraquídeo , Pruebas Diagnósticas de Rutina , Neoplasias Meníngeas/líquido cefalorraquídeo , Neoplasias Meníngeas/diagnóstico , Neoplasias Meníngeas/genética , Neoplasias/complicaciones , Neoplasias/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/líquido cefalorraquídeo , Neoplasias Primarias Secundarias/diagnóstico , Neoplasias Primarias Secundarias/genética , Valor Predictivo de las Pruebas
10.
Nat Commun ; 12(1): 1334, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637765

RESUMEN

To understand the mechanisms that mediate germline genetic leukemia predisposition, we studied the inherited ribosomopathy Shwachman-Diamond syndrome (SDS), a bone marrow failure disorder with high risk of myeloid malignancies at an early age. To define the mechanistic basis of clonal hematopoiesis in SDS, we investigate somatic mutations acquired by patients with SDS followed longitudinally. Here we report that multiple independent somatic hematopoietic clones arise early in life, most commonly harboring heterozygous mutations in EIF6 or TP53. We show that germline SBDS deficiency establishes a fitness constraint that drives selection of somatic clones via two distinct mechanisms with different clinical consequences. EIF6 inactivation mediates a compensatory pathway with limited leukemic potential by ameliorating the underlying SDS ribosome defect and enhancing clone fitness. TP53 mutations define a maladaptive pathway with enhanced leukemic potential by inactivating tumor suppressor checkpoints without correcting the ribosome defect. Subsequent development of leukemia was associated with acquisition of biallelic TP53 alterations. These results mechanistically link leukemia predisposition to germline genetic constraints on cellular fitness, and provide a rational framework for clinical surveillance strategies.


Asunto(s)
Hematopoyesis Clonal/genética , Hematopoyesis Clonal/fisiología , Síndrome de Shwachman-Diamond/genética , Síndrome de Shwachman-Diamond/metabolismo , Adolescente , Adulto , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/metabolismo , Niño , Preescolar , Factores Eucarióticos de Iniciación/genética , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación , Ribosomas/genética , Proteína p53 Supresora de Tumor/genética , Adulto Joven
11.
Nat Cancer ; 2(5): 498-502, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-35122016

RESUMEN

Recent studies suggest that the cyclin-dependent kinase (CDK) pathway may be a therapeutic target for brain metastases (BM). Here, we present interim analysis of a basket trial evaluating the intracranial efficacy of the CDK inhibitor palbociclib in patients with progressive BM and CDK alterations. Our study met its primary endpoint and provides evidence for performing molecular testing of archival BM tissue, if available, to inform the choice of CNS-penetrant targeted therapy.


Asunto(s)
Neoplasias Encefálicas , Quinasa 6 Dependiente de la Ciclina , Neoplasias Encefálicas/tratamiento farmacológico , Quinasa 4 Dependiente de la Ciclina , Humanos , Piperazinas/uso terapéutico , Piridinas
12.
Sci Rep ; 10(1): 15859, 2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32985541

RESUMEN

Feeding pigs with very-low protein (VLP) diets while supplemented with limiting amino acids (AA) results in decreased growth. The objective of this study was to determine if supplementing VLP diets with branched-chain AA (BCAA) would reverse the negative effects of these diets on growth and whether this is associated with alterations in energy balance, blood metabolomics and fecal microbiota composition. Twenty-four nursery pigs were weight-matched, individually housed and allotted into following treatments (n = 8/group): control (CON), low protein (LP) and LP supplemented with BCAA (LP + BCAA) for 4 weeks. Relative to CON, pigs fed with LP had lower feed intake (FI) and body weight (BW) throughout the study, but those fed with LP + BCAA improved overall FI computed for 4 weeks, tended to increase the overall average daily gain, delayed the FI and BW depression for ~ 2 weeks and had transiently higher energy expenditure. Feeding pigs with LP + BCAA impacted the phenylalanine and protein metabolism and fatty acids synthesis pathways. Compared to CON, the LP + BCAA group had higher abundance of Paludibacteraceae and Synergistaceae and reduced populations of Streptococcaceae, Oxyphotobacteria_unclassified, Pseudomonadaceae and Shewanellaceae in their feces. Thus, supplementing VLP diets with BCAA temporarily annuls the adverse effects of these diets on growth, which is linked with alterations in energy balance and metabolic and gut microbiome profile.


Asunto(s)
Aminoácidos de Cadena Ramificada/análisis , Dieta con Restricción de Proteínas , Suplementos Dietéticos/análisis , Metabolismo Energético/efectos de los fármacos , Heces/microbiología , Metabolómica , Microbiota/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Citocinas/sangre , Ingestión de Alimentos/efectos de los fármacos , Respiración/efectos de los fármacos , Porcinos
13.
Nat Med ; 26(8): 1280-1284, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32483359

RESUMEN

An increasing fraction of patients with metastatic cancer develop leptomeningeal dissemination of disease (LMD), and survival is dismal1-3. We conducted a single-arm, phase 2 study of pembrolizumab in patients with solid tumor malignancies and LMD (NCT02886585). Patients received 200 mg of pembrolizumab intravenously every 3 weeks until definitive progression or unacceptable toxicity. The primary endpoint was rate of overall survival at 3 months (OS3). Secondary objectives included toxicity, response rate and time to intracranial or extracranial disease progression. A Simon two-stage design was used to compare a null hypothesis OS3 of 18% against an alternative of 43%. Twenty patients-17 with breast cancer, two with lung cancer and one with ovarian cancer-were enrolled into the pre-specified evaluation group having received at least one dose of pembrolizumab. The median follow-up of surviving patients was 6.3 months (range, 2.2-12.5 months). The percentage of patients who experienced one (or more) grade 3 or higher adverse events at least possibly related to treatment was 40%, the most frequent being hyperglycemia (n = 6), nausea (n = 7) and vomiting (n = 7). The study met the primary endpoint, as 12 of 20 (OS3, 0.60; 90% confidence interval, 0.39-0.78) patients were alive at 3 months after enrollment. Pembrolizumab is safe and feasible and displays promising activity in patients with LMD. Further investigations are needed to identify which patients with LMD can benefit from pembrolizumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Carcinomatosis Meníngea/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/efectos adversos , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hiperglucemia/inducido químicamente , Hiperglucemia/patología , Neoplasias Pulmonares/patología , Carcinomatosis Meníngea/patología , Náusea/inducido químicamente , Náusea/patología , Metástasis de la Neoplasia , Neoplasias Ováricas/patología , Vómitos/inducido químicamente , Vómitos/patología
15.
Cancer Discov ; 10(9): 1296-1311, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32371478

RESUMEN

The molecular mechanisms leading to resistance to PD-1 blockade are largely unknown. Here, we characterize tumor biopsies from a patient with melanoma who displayed heterogeneous responses to anti-PD-1 therapy. We observe that a resistant tumor exhibited a loss-of-function mutation in the tumor suppressor gene FBXW7, whereas a sensitive tumor from the same patient did not. Consistent with a functional role in immunotherapy response, inactivation of Fbxw7 in murine tumor cell lines caused resistance to anti-PD-1 in immunocompetent animals. Loss of Fbxw7 was associated with altered immune microenvironment, decreased tumor-intrinsic expression of the double-stranded RNA (dsRNA) sensors MDA5 and RIG1, and diminished induction of type I IFN and MHC-I expression. In contrast, restoration of dsRNA sensing in Fbxw7-deficient cells was sufficient to sensitize them to anti-PD-1. Our results thus establish a new role for the commonly inactivated tumor suppressor FBXW7 in viral sensing and sensitivity to immunotherapy. SIGNIFICANCE: Our findings establish a role of the commonly inactivated tumor suppressor FBXW7 as a genomic driver of response to anti-PD-1 therapy. Fbxw7 loss promotes resistance to anti-PD-1 through the downregulation of viral sensing pathways, suggesting that therapeutic reactivation of these pathways could improve clinical responses to checkpoint inhibitors in genomically defined cancer patient populations.This article is highlighted in the In This Issue feature, p. 1241.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Anciano , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral/trasplante , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Modelos Animales de Enfermedad , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Células HeLa , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Mutación con Pérdida de Función , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
16.
Future Oncol ; 16(14): 899-909, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32270710

RESUMEN

Brain metastases occur in up to 25-55% of patients with metastatic HER2-positive breast cancer. Standard treatment has high rates of recurrence or progression, limiting survival and quality of life in most patients. Temozolomide (TMZ) is known to penetrate the blood-brain barrier and is US FDA approved for treatment of glioblastoma. Our group has demonstrated that low doses of TMZ administered in a prophylactic, metronomic fashion can significantly prevent development of brain metastases in murine models of breast cancer. Based on these findings, we initiated a secondary-prevention clinical trial with oral TMZ given to HER2-positive breast cancer patients with brain metastases after recent local treatment in combination with T-DM1 for systemic control of disease. Primary end point is freedom from new brain metastases at 1 year. (NCT03190967).


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/prevención & control , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Telomerasa/metabolismo , Temozolomida/uso terapéutico , Animales , Antineoplásicos Alquilantes/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor , Neoplasias Encefálicas/terapia , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Proyectos de Investigación , Temozolomida/farmacología
17.
Nat Genet ; 52(4): 371-377, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203465

RESUMEN

Brain metastases from lung adenocarcinoma (BM-LUAD) frequently cause patient mortality. To identify genomic alterations that promote brain metastases, we performed whole-exome sequencing of 73 BM-LUAD cases. Using case-control analyses, we discovered candidate drivers of brain metastasis by identifying genes with more frequent copy-number aberrations in BM-LUAD compared to 503 primary LUADs. We identified three regions with significantly higher amplification frequencies in BM-LUAD, including MYC (12 versus 6%), YAP1 (7 versus 0.8%) and MMP13 (10 versus 0.6%), and significantly more frequent deletions in CDKN2A/B (27 versus 13%). We confirmed that the amplification frequencies of MYC, YAP1 and MMP13 were elevated in an independent cohort of 105 patients with BM-LUAD. Functional assessment in patient-derived xenograft mouse models validated the notion that MYC, YAP1 or MMP13 overexpression increased the incidence of brain metastasis. These results demonstrate that somatic alterations contribute to brain metastases and that genomic sequencing of a sufficient number of metastatic tumors can reveal previously unknown metastatic drivers.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Neoplasias Encefálicas/genética , Neoplasias Pulmonares/genética , Metástasis de la Neoplasia/genética , Adenocarcinoma del Pulmón/patología , Animales , Neoplasias Encefálicas/patología , Estudios de Casos y Controles , Línea Celular , Variaciones en el Número de Copia de ADN/genética , Femenino , Genes myc/genética , Genómica/métodos , Células HEK293 , Humanos , Neoplasias Pulmonares/patología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Desnudos , Mutación/genética , Metástasis de la Neoplasia/patología , Factores de Transcripción/genética , Secuenciación del Exoma
18.
Sci Rep ; 10(1): 3318, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094453

RESUMEN

Reducing dietary crude protein (CP) beyond a certain threshold leads to poor growth performance in pigs; however, the underlying mechanisms are not well understood. Following an adaption period, thirty-seven weaned pigs were weight matched (8.41 ± 0.14 kg), housed individually and randomly assigned into three groups with different dietary CP levels: 24% CP (CON; n = 12), 18% CP (n = 12) and 12% CP (n = 13) for 28 days. The body weight was not different between the CON and 18% CP diets, but 12% CP significantly decreased body weight after day 21. Compared to the CON, pigs fed with 12% CP decreased feed intake day 17 onwards. The 12% CP diet increased the energy expenditure during week 1 compared to the CON. The 12% CP influenced starch and sucrose, nitrogen, and branched-chain amino acids metabolism pathways. The feces of pigs fed with 12% CP were less enriched in Prevotella, but had higher relative abundance of Christensenedilaceae, Aligiphilus and Algoriphagus than CON and 18% CP. Overall, reducing dietary CP by 50%, but not by 25%, significantly influenced the physiological responses in nursery pigs. The pigs fed with low or standard protein diets had differential bacterial communities in their feces as well as serum metabolomics profile.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Carbohidratos de la Dieta/efectos adversos , Metabolismo Energético , Microbioma Gastrointestinal , Metabolómica , Porcinos/sangre , Porcinos/microbiología , Animales , Biodiversidad , Peso Corporal , Análisis Discriminante , Heces/microbiología , Conducta Alimentaria , Metaboloma , Filogenia , Análisis de Componente Principal , Porcinos/metabolismo
20.
Clin Cancer Res ; 25(16): 5135-5142, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31164371

RESUMEN

PURPOSE: Leiomyosarcoma and liposarcoma are common subtypes of soft tissue sarcoma (STS). Patients with metastatic leiomyosarcoma or dedifferentiated liposarcoma (DDLPS) typically have worse outcomes compared with localized leiomyosarcoma or well-differentiated liposarcoma (WDLPS). A better understanding of genetic changes between primary/metastatic leiomyosarcoma and between WDLPS/DDLPS may provide insight into their genetic evolution. EXPERIMENTAL DESIGN: We interrogated whole-exome sequencing (WES) from "trios" of normal tissue, primary tumor, and metastatic tumor from individual patients with leiomyosarcoma (n = 9), and trios of normal tissue, well-differentiated tumor, and dedifferentiated tumor from individual patients with liposarcoma (n = 19). Specifically, we performed mutational, copy number, and tumor evolution analyses on these cohorts and compared patterns among leiomyosarcoma and liposarcoma trios. RESULTS: Leiomyosarcoma cases harbored shared drivers through a typical parent/child relationship where the metastatic tumor was derived from the primary tumor. In contrast, while all liposarcoma cases shared the characteristic focal chromosome 12 amplicon, most paired liposarcoma cases did not share additional mutations, suggesting a divergent evolutionary pattern from a common precursor. No highly recurrent genomic alterations from WES were identified that could be implicated as driving the progression of disease in either sarcoma subtype. CONCLUSIONS: From a genomic perspective, leiomyosarcoma metastases contain genetic alterations that are also found in primary tumors. WDLPS and DDLPS, however, appear to divergently evolve from a common precursor harboring 12q amplification, rather than as a transformation to a higher-grade tumor. Further efforts to identify specific drivers of these distinct evolutionary patterns may inform future translational and clinical research in STS.


Asunto(s)
Transformación Celular Neoplásica/genética , Predisposición Genética a la Enfermedad , Genómica , Leiomiosarcoma/genética , Leiomiosarcoma/patología , Liposarcoma/genética , Liposarcoma/patología , Adulto , Anciano , Biopsia , Femenino , Perfilación de la Expresión Génica , Genómica/métodos , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Estudios Retrospectivos , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA