Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Cancer Discov ; 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39121480

RESUMEN

Current treatments for KRAS-mutant colorectal cancers (CRCs) are often limited by cellular plasticity and rewiring responses. Here we describe a promising therapeutic strategy that simultaneously targets epigenetic and oncogenic signals. Specifically, we show that inhibitors of the histone methyltransferase, EZH2, synergize with various RAS pathway inhibitors and promote dramatic tumor regression in vivo. Together these agents cooperatively suppress WNT-driven transcription and drive CRCs into a more differentiated cell state by inducing the Groucho/TLE corepressor, TLE4, along with a network of WNT pathway inhibitors and intestinal differentiation proteins. However, these agents also induce the pro-apoptotic protein BMF, which subsequently kills these more differentiated cells. Accordingly, cell death can be prevented by activating ß-catenin, blocking differentiation, or by ablating BMF expression. Collectively, these studies reveal a new therapeutic approach for treating KRAS-mutant CRCs and illustrate a critical convergence of EZH2 and RAS on oncogenic WNT signals, intestinal differentiation, and apoptosis.

2.
Cancer Res ; 84(14): 2247-2264, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38657118

RESUMEN

Alveolar soft part sarcoma (ASPS) is a rare mesenchymal malignancy driven by the ASPSCR1::TFE3 fusion. A better understanding of the mechanisms by which this oncogenic transcriptional regulator drives cancer growth is needed to help identify potential therapeutic targets. In this study, we characterized the transcriptional and chromatin landscapes of ASPS tumors and preclinical models, identifying the essential role of ASPSCR1::TFE3 in tumor cell viability by regulating core transcriptional programs involved in cell proliferation, angiogenesis, and mitochondrial biology. ASPSCR1::TFE3 directly interacted with key epigenetic regulators at enhancers and promoters to support ASPS-associated transcription. Among the effector programs driven by ASPSCR1::TFE3, cell proliferation was driven by high levels of cyclin D1 expression. Disruption of cyclin D1/CDK4 signaling led to a loss of ASPS proliferative capacity, and combined inhibition of CDK4/6 and angiogenesis halted tumor growth in xenografts. These results define the ASPS oncogenic program, reveal mechanisms by which ASPSCR1::TFE3 controls tumor biology, and identify a strategy for therapeutically targeting tumor cell-intrinsic vulnerabilities. Significance: The ASPSCR1::TFE3 fusion propels the growth of alveolar soft part sarcoma  by activating transcriptional programs that regulate proliferation, angiogenesis, mitochondrial biogenesis, and differentiation and can be therapeutically targeted to improve treatment.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Proliferación Celular , Proteínas de Fusión Oncogénica , Sarcoma de Parte Blanda Alveolar , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Sarcoma de Parte Blanda Alveolar/genética , Sarcoma de Parte Blanda Alveolar/patología , Sarcoma de Parte Blanda Alveolar/metabolismo , Humanos , Animales , Ratones , Proliferación Celular/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/metabolismo , Péptidos y Proteínas de Señalización Intracelular
3.
Nature ; 628(8007): 424-432, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509359

RESUMEN

Fusobacterium nucleatum (Fn), a bacterium present in the human oral cavity and rarely found in the lower gastrointestinal tract of healthy individuals1, is enriched in human colorectal cancer (CRC) tumours2-5. High intratumoural Fn loads are associated with recurrence, metastases and poorer patient prognosis5-8. Here, to delineate Fn genetic factors facilitating tumour colonization, we generated closed genomes for 135 Fn strains; 80 oral strains from individuals without cancer and 55 unique cancer strains cultured from tumours from 51 patients with CRC. Pangenomic analyses identified 483 CRC-enriched genetic factors. Tumour-isolated strains predominantly belong to Fn subspecies animalis (Fna). However, genomic analyses reveal that Fna, considered a single subspecies, is instead composed of two distinct clades (Fna C1 and Fna C2). Of these, only Fna C2 dominates the CRC tumour niche. Inter-Fna analyses identified 195 Fna C2-associated genetic factors consistent with increased metabolic potential and colonization of the gastrointestinal tract. In support of this, Fna C2-treated mice had an increased number of intestinal adenomas and altered metabolites. Microbiome analysis of human tumour tissue from 116 patients with CRC demonstrated Fna C2 enrichment. Comparison of 62 paired specimens showed that only Fna C2 is tumour enriched compared to normal adjacent tissue. This was further supported by metagenomic analysis of stool samples from 627 patients with CRC and 619 healthy individuals. Collectively, our results identify the Fna clade bifurcation, show that specifically Fna C2 drives the reported Fn enrichment in human CRC and reveal the genetic underpinnings of pathoadaptation of Fna C2 to the CRC niche.


Asunto(s)
Neoplasias Colorrectales , Fusobacterium nucleatum , Animales , Humanos , Ratones , Adenoma/microbiología , Estudios de Casos y Controles , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Heces/microbiología , Fusobacterium nucleatum/clasificación , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/aislamiento & purificación , Fusobacterium nucleatum/patogenicidad , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Genoma Bacteriano/genética , Boca/microbiología , Femenino
4.
Clin Cancer Res ; 30(8): 1669-1684, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38345769

RESUMEN

PURPOSE: ERBB2-amplified colorectal cancer is a distinct molecular subtype with expanding treatments. Implications of concurrent oncogenic RAS/RAF alterations are not known. EXPERIMENTAL DESIGN: Dana-Farber and Foundation Medicine Inc. Colorectal cancer cohorts with genomic profiling were used to identify ERBB2-amplified cases [Dana-Farber, n = 47/2,729 (1.7%); FMI, n = 1857/49,839 (3.7%)]. Outcomes of patients receiving HER2-directed therapies are reported (Dana-Farber, n = 9; Flatiron Health-Foundation Medicine clinicogenomic database, FH-FMI CGDB, n = 38). Multisite HER2 IHC and genomic profiling were performed to understand HER2 intratumoral and interlesional heterogeneity. The impact of concurrent RAS comutations on the effectiveness of HER2-directed therapies were studied in isogenic colorectal cancer cell lines and xenografts. RESULTS: ERBB2 amplifications are enriched in left-sided colorectal cancer. Twenty percent of ERBB2-amplified colorectal cancers have co-occurring oncogenic RAS/RAF alterations. While RAS/RAF WT colorectal cancers typically have clonal ERBB2 amplification, colorectal cancers with co-occurring RAS/RAF alterations have lower level ERRB2 amplification, higher intratumoral heterogeneity, and interlesional ERBB2 discordance. These distinct genomic patterns lead to differential responsiveness and patterns of resistance to HER2-directed therapy. ERBB2-amplified colorectal cancer with RAS/RAF alterations are resistant to trastuzumab-based combinations, such as trastuzumab/tucatinib, but retain sensitivity to trastuzumab deruxtecan in in vitro and murine models. Trastuzumab deruxtecan shows clinical efficacy in cases with high-level ERBB2-amplified RAS/RAF coaltered colorectal cancer. CONCLUSIONS: Co-occurring RAS/RAF alterations define a unique subtype of ERBB2-amplified colorectal cancer that has increased intratumoral heterogeneity, interlesional discordance, and resistance to trastuzumab-based combinations. Further examination of trastuzumab deruxtecan in this previously understudied cohort of ERBB2-amplified colorectal cancer is warranted.


Asunto(s)
Neoplasias Colorrectales , Variaciones en el Número de Copia de ADN , Humanos , Animales , Ratones , Amplificación de Genes , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Resultado del Tratamiento , Mutación
5.
Nutrition ; 120: 112359, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38340390

RESUMEN

OBJECTIVE: The study aimed to identify the main folate sources and examine socio-demographic and lifestyle determinants influencing folate intake among 1410 women aged 18 to 39. METHODS: Data were collected using a self-administered health and lifestyle questionnaire and a 5-d dietary record method. To assess folate intake in relation to the dietary reference intakes, the probability approach was used. Folate intake determinants were identified using multivariate-adjusted logistic regression models; odds ratios (ORs) with 95% confidence intervals (95% CIs). RESULTS: The average total folate intake among women was 311 ± 144 µg/day dietary folate equivalents. Vegetables (30.7%) and cereals (22.6%) were the most important folate sources. Foods fortified with folic acid were consumed by 20.6% of women, dietary supplements by 7.2%. More than half of the participants (55%) had a high probability of inadequate folate intake. The predictors of being in the highest tertile of folate intake (>303 versus <225 µg) were: physical activity (high versus low; OR: 2.97, 95% CI: 1.77-4.97), nutritional knowledge (high versus low; OR: 5.32, 95% CI: 2.82-10.1), following a vegetarian diet (yes versus no; OR: 6.13; 95% CI: 2.79-13.5), daily number of meals (≥5 versus ≤3; OR: 4.17, 95% CI: 2.38-7.32), excluding/including some foods (yes versus no; OR: 2.47; 95% CI: 1.41-4.31) and energy intake (3rd versus 1st tertile; OR:17.4, 95% CI: 11.1-27.4). CONCLUSION: Identifying factors associated with a higher intake of folate may be helpful in shaping public health nutrition policy. It allows the design of effective nutrition education programs to promote increased intake of folate in subgroups at risk of deficiency.


Asunto(s)
Suplementos Dietéticos , Ácido Fólico , Humanos , Femenino , Universidades , Estado Nutricional , Estudiantes
6.
Antioxidants (Basel) ; 12(10)2023 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-37891878

RESUMEN

This systematic review aims to summarise the results of controlled trials on dietary supplements (DS) usage and inflammation, oxidative stress, antioxidant status, and thyroid parameter improvement in hypothyroidism (HT)/Hashimoto's thyroiditis (AIT) patients. The study protocol was registered with PROSPERO (no. CRD42022365149). A comprehensive search of the PubMed, Scopus, and Web of Science databases resulted in the identification of nineteen randomised controlled trials and three non-randomised studies for the review; three studies examined the effect of supplementation with vitamin D, twelve studies-with selenium, and seven studies-with other DS. Based on very limited evidence, the lack of influence of vitamin D supplementation on inflammatory parameters was found, while no studies have examined oxidative stress and antioxidant status parameters, and only one provided results for a single thyroid parameter after an intervention. Some evidence was found proving that selenium supplementation may decrease inflammation and improve thyroid parameters, but reaching a conclusion about its influence on oxidative stress and antioxidant status is not possible because of the insufficient number of studies. Additionally, due to examining other DS (e.g., multicomponent, Nigella sativa, and genistein) only in single studies, conclusions cannot be drawn. Further long-term, high-quality randomised controlled trials are necessary to better understand the influence of DS on inflammation, oxidative stress, and antioxidant status, as well as their potential to improve thyroid gland function in HT/AIT patients.

7.
Sci Adv ; 9(39): eadd9668, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37756410

RESUMEN

Neuroendocrine tumors (NETs) are rare cancers that most often arise in the gastrointestinal tract and pancreas. The fundamental mechanisms driving gastroenteropancreatic (GEP)-NET growth remain incompletely elucidated; however, the heterogeneous clinical behavior of GEP-NETs suggests that both cellular lineage dynamics and tumor microenvironment influence tumor pathophysiology. Here, we investigated the single-cell transcriptomes of tumor and immune cells from patients with gastroenteropancreatic NETs. Malignant GEP-NET cells expressed genes and regulons associated with normal, gastrointestinal endocrine cell differentiation, and fate determination stages. Tumor and lymphoid compartments sparsely expressed immunosuppressive targets commonly investigated in clinical trials, such as the programmed cell death protein-1/programmed death ligand-1 axis. However, infiltrating myeloid cell types within both primary and metastatic GEP-NETs were enriched for genes encoding other immune checkpoints, including VSIR (VISTA), HAVCR2 (TIM3), LGALS9 (Gal-9), and SIGLEC10. Our findings highlight the transcriptomic heterogeneity that distinguishes the cellular landscapes of GEP-NET anatomic subtypes and reveal potential avenues for future precision medicine therapeutics.


Asunto(s)
Neoplasias Intestinales , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Neoplasias Gástricas , Humanos , Tumores Neuroendocrinos/genética , Neoplasias Intestinales/genética , Neoplasias Gástricas/genética , Neoplasias Pancreáticas/genética , Microambiente Tumoral/genética
8.
Nutrients ; 15(9)2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37432245

RESUMEN

The Mediterranean diet (MD) has been previously proven to have various health-related benefits; however, its effect on women's reproductive health over a lifespan is yet to be summarized. This study aimed to summarize the evidence-based knowledge regarding the association between the MD and selected reproductive health outcomes. By searching PubMed, ScienceDirect, and Google Scholar databases, as well as reference lists, 21 studies were included in this narrative review. The available evidence was very limited; however, there is some suggestion that higher adherence to the MD may be positively associated with a lower risk of early age menarche (1 study) and shorter menstrual cycles (1 study), but is unrelated to dysmenorrhea (1 study). Moreover, no study to date has examined the relationship between the MD and the onset age of natural menopause. Considering reproductive health diseases, there was limited evidence that a higher adherence to the MD was associated with a lower risk of premenstrual syndrome (1 study), an improvement in sexual health and a lower prevalence of sexual dysfunction (3 studies), and an improvement in the general condition of with endometriosis and the pain they can experience (1 study). The largest number of studies were found for polycystic ovarian syndrome (PCOS; 7 studies) and infertility (6 studies). Results showed that a higher adherence to the MD was associated with a lower risk of infertility, while results for PCOS were unclear, but mostly indicated a beneficial trend. Further investigations are necessary to establish the role of adherence to the MD in reproductive health maintenance and its possible role in the prevention and treatment of reproductive health diseases in women.


Asunto(s)
Dieta Mediterránea , Infertilidad , Síndrome del Ovario Poliquístico , Humanos , Femenino , Longevidad , Salud Reproductiva , Dismenorrea
9.
Sci Adv ; 9(25): eadg4128, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37352344

RESUMEN

A potential cause of cancer relapse is pretreatment chemoresistant subpopulations. Identifying targetable features of subpopulations that are poorly primed for therapy-induced cell death may improve cancer therapy. Here, we develop and validate real-time BH3 profiling, a live and functional single-cell measurement of pretreatment apoptotic sensitivity that occurs upstream of apoptotic protease activation. On the same single cells, we perform cyclic immunofluorescence, which enables multiplexed immunofluorescence of more than 30 proteins on the same cell. Using cultured cells and rapid ex vivo cultures of colon cancer patient-derived xenograft (PDX) models, we identify Bak as a univariate correlate of apoptotic priming, find that poorly primed subpopulations can correspond to specific stages of the cell cycle, and, in some PDX models, identify increased expression of Bcl-XL, Mcl-1, or Her2 in subpopulations that are poorly primed for apoptosis. Last, we generate and validate mathematical models of single-cell priming that describe how targetable proteins contribute to apoptotic priming.


Asunto(s)
Apoptosis , Neoplasias , Neoplasias/metabolismo , Neoplasias/patología , Proteómica , Humanos , Análisis de la Célula Individual , Línea Celular Tumoral
10.
Cell Rep Med ; 4(4): 101007, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37030295

RESUMEN

Pancreatic ductal adenocarcinomas (PDACs) frequently harbor KRAS mutations. Although MEK inhibitors represent a plausible therapeutic option, most PDACs are innately resistant to these agents. Here, we identify a critical adaptive response that mediates resistance. Specifically, we show that MEK inhibitors upregulate the anti-apoptotic protein Mcl-1 by triggering an association with its deubiquitinase, USP9X, resulting in acute Mcl-1 stabilization and protection from apoptosis. Notably, these findings contrast the canonical positive regulation of Mcl-1 by RAS/ERK. We further show that Mcl-1 inhibitors and cyclin-dependent kinase (CDK) inhibitors, which suppress Mcl-1 transcription, prevent this protective response and induce tumor regression when combined with MEK inhibitors. Finally, we identify USP9X as an additional potential therapeutic target. Together, these studies (1) demonstrate that USP9X regulates a critical mechanism of resistance in PDAC, (2) reveal an unexpected mechanism of Mcl-1 regulation in response to RAS pathway suppression, and (3) provide multiple distinct promising therapeutic strategies for this deadly malignancy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
11.
Nutrients ; 15(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36771431

RESUMEN

Due to the lack of studies comparing the determinants of well-being in omnivores and vegetarians, we examined associations of socio-demographic and lifestyle factors, including adherence to a Mediterranean-style diet, in relation to well-being in omnivorous, vegetarian, and vegan women. Well-being was assessed using a validated WHO-5 Well-Being Index. Adherence to the Mediterranean-style diet was determined using a modified Mediterranean diet score. The study was conducted on 636 women (23.9 ± 5.7 years), of whom 47.3% were omnivores, 33.2% vegetarians, and 19.5% vegans. The good well-being group (WHO-5 Index ≥ 13 points) comprised 30.9% of the omnivores, 46.0% of the vegetarians, and 57.3% of the vegans. The remaining participants were classified as belonging to the poor well-being group (<13 points). Compared to the omnivores, the vegetarians and vegans had a 1.6-fold (95% CI: 1.04-2.42) and a 2.4-fold (95% CI: 1.45-3.99) higher probability of having good well-being, respectively. In omnivores, the predictors of good well-being were adherence to the Mediterranean-style diet (a 1-score increment was associated with a 17% higher probability of good well-being, P-trend = 0.016), higher self-perceived health status, and lower levels of stress. In vegetarians and vegans, it was older age, higher physical activity (≥3 h/week), 7-8 h sleep time, and similarly to omnivores' higher self-perceived health status and lower stress level. Our findings indicate that following a Mediterranean-style diet was associated with better well-being in omnivores. Furthermore, we identified that different determinants were associated with well-being in omnivorous and vegetarian and vegan women.


Asunto(s)
Dieta Mediterránea , Veganos , Humanos , Femenino , Dieta Vegetariana , Vegetarianos , Dieta Vegana , Dieta
12.
Nature ; 611(7937): 810-817, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36385528

RESUMEN

The tumour-associated microbiota is an intrinsic component of the tumour microenvironment across human cancer types1,2. Intratumoral host-microbiota studies have so far largely relied on bulk tissue analysis1-3, which obscures the spatial distribution and localized effect of the microbiota within tumours. Here, by applying in situ spatial-profiling technologies4 and single-cell RNA sequencing5 to oral squamous cell carcinoma and colorectal cancer, we reveal spatial, cellular and molecular host-microbe interactions. We adapted 10x Visium spatial transcriptomics to determine the identity and in situ location of intratumoral microbial communities within patient tissues. Using GeoMx digital spatial profiling6, we show that bacterial communities populate microniches that are less vascularized, highly immuno­suppressive and associated with malignant cells with lower levels of Ki-67 as compared to bacteria-negative tumour regions. We developed a single-cell RNA-sequencing method that we name INVADEseq (invasion-adhesion-directed expression sequencing) and, by applying this to patient tumours, identify cell-associated bacteria and the host cells with which they interact, as well as uncovering alterations in transcriptional pathways that are involved in inflammation, metastasis, cell dormancy and DNA repair. Through functional studies, we show that cancer cells that are infected with bacteria invade their surrounding environment as single cells and recruit myeloid cells to bacterial regions. Collectively, our data reveal that the distribution of the microbiota within a tumour is not random; instead, it is highly organized in microniches with immune and epithelial cell functions that promote cancer progression.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Colorrectales , Interacciones Microbiota-Huesped , Microbiota , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/microbiología , Carcinoma de Células Escamosas/patología , Microbiota/genética , Microbiota/inmunología , Microbiota/fisiología , Neoplasias de la Boca/genética , Neoplasias de la Boca/inmunología , Neoplasias de la Boca/microbiología , Neoplasias de la Boca/patología , Células Mieloides/inmunología , Microambiente Tumoral , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Análisis de Secuencia de ARN , Perfilación de la Expresión Génica , Antígeno Ki-67/metabolismo , Progresión de la Enfermedad
13.
Br J Cancer ; 127(11): 2072-2085, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36175617

RESUMEN

BACKGROUND: Advanced gastrointestinal stromal tumour (GIST) is characterised by genomic perturbations of key cell cycle regulators. Oncogenic activation of CDK4/6 results in RB1 inactivation and cell cycle progression. Given that single-agent CDK4/6 inhibitor therapy failed to show clinical activity in advanced GIST, we evaluated strategies for maximising response to therapeutic CDK4/6 inhibition. METHODS: Targeted next-generation sequencing and multiplexed protein imaging were used to detect cell cycle regulator aberrations in GIST clinical samples. The impact of inhibitors of CDK2, CDK4 and CDK2/4/6 was determined through cell proliferation and protein detection assays. CDK-inhibitor resistance mechanisms were characterised in GIST cell lines after long-term exposure. RESULTS: We identify recurrent genomic aberrations in cell cycle regulators causing co-activation of the CDK2 and CDK4/6 pathways in clinical GIST samples. Therapeutic co-targeting of CDK2 and CDK4/6 is synergistic in GIST cell lines with intact RB1, through inhibition of RB1 hyperphosphorylation and cell proliferation. Moreover, RB1 inactivation and a novel oncogenic cyclin D1 resulting from an intragenic rearrangement (CCND1::chr11.g:70025223) are mechanisms of acquired CDK-inhibitor resistance in GIST. CONCLUSIONS: These studies establish the biological rationale for CDK2 and CDK4/6 co-inhibition as a therapeutic strategy in patients with advanced GIST, including metastatic GIST progressing on tyrosine kinase inhibitors.


Asunto(s)
Neoplasias Gastrointestinales , Tumores del Estroma Gastrointestinal , Humanos , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Quinasa 6 Dependiente de la Ciclina , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética
14.
Nutrients ; 14(13)2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35807925

RESUMEN

The study aimed to examine socio-demographic and lifestyle determinants, including diet quality, of dietary supplement (DS) use among 2545 students who attended public universities in Warsaw. The data was collected using a self-administered health and lifestyle questionnaire and a 5-day dietary record method. Diet quality was assessed using a Mediterranean Diet Score. To determine the covariates of DS use, multivariate-adjusted logistic regression models with an estimation of odds ratios (ORs) and 95% confidence intervals (95% CIs) were used. The results showed that 41% of participants were DS users. The following predictors of DS use were identified: gender (male vs. female; OR:0.62, 95% CI:0.50-0.79), physical activity (high vs. low; OR:1.79, 95% CI:1.45-2.20), BMI (≥25 vs. 18.5-24.9 kg/m2; OR:0.77, 95% CI:0.61-0.98), cigarette smoking (yes vs. no; OR:0.67, 95% CI:0.52-0.86), and presence of chronic diseases (yes vs. no; OR:2.37, 95% CI:1.89-2.98). Moreover, higher nutritional knowledge, special diet usage, eating more meals/day, and fortified food consumption were determinants of DS use. Adherence to the Mediterranean-style diet was positively associated with DS use, a 1-score increment was associated with a 10% (p-trend = 0.011) higher probability of DS usage. Given that the use of vitamins and/or minerals is common among university students and their users are characterized by eating a higher quality diet, investigating the contribution of DS in overall dietary nutrient intake warrants further study.


Asunto(s)
Dieta Mediterránea , Demografía , Suplementos Dietéticos , Femenino , Humanos , Estilo de Vida , Masculino , Estudiantes , Encuestas y Cuestionarios , Universidades
15.
Nat Commun ; 13(1): 3181, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35676246

RESUMEN

The RNF43_p.G659fs mutation occurs frequently in colorectal cancer, but its function remains poorly understood and there are no specific therapies directed against this alteration. In this study, we find that RNF43_p.G659fs promotes cell growth independent of Wnt signaling. We perform a drug repurposing library screen and discover that cells with RNF43_p.G659 mutations are selectively killed by inhibition of PI3K signaling. PI3K/mTOR inhibitors yield promising antitumor activity in RNF43659mut isogenic cell lines and xenograft models, as well as in patient-derived organoids harboring RNF43_p.G659fs mutations. We find that RNF43659mut binds p85 leading to increased PI3K signaling through p85 ubiquitination and degradation. Additionally, RNA-sequencing of RNF43659mut isogenic cells reveals decreased interferon response gene expression, that is reversed by PI3K/mTOR inhibition, suggesting that RNF43659mut may alter tumor immunity. Our findings suggest a therapeutic application for PI3K/mTOR inhibitors in treating RNF43_p.G659fs mutant cancers.


Asunto(s)
Neoplasias Colorrectales , Fosfatidilinositol 3-Quinasas , Serina-Treonina Quinasas TOR , Ubiquitina-Proteína Ligasas , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Humanos , Mutación , Fosfatidilinositol 3-Quinasas/genética , Serina-Treonina Quinasas TOR/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
16.
Cancer Discov ; 12(7): 1804-1823, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35499757

RESUMEN

Gastrointestinal stromal tumor (GIST) is commonly characterized by activating mutations in the receptor tyrosine kinase KIT. Tyrosine kinase inhibitors are the only approved therapy for GIST, and complementary treatment strategies are urgently needed. As GIST lacks oncogene amplification and relies upon an established network of transcription factors, we hypothesized that unique chromatin-modifying enzymes are essential in orchestrating the GIST epigenome. We identified through genome-scale CRISPR screening that MOZ and Menin-MLL chromatin regulatory complexes are cooperative and unique dependencies in GIST. These complexes were enriched at GIST-relevant genes and regulated their transcription. Inhibition of MOZ and Menin-MLL complexes decreased GIST cell proliferation by disrupting interactions with transcriptional/chromatin regulators, such as DOT1L. MOZ and Menin inhibition caused significant reductions in tumor burden in vivo, with superior effects observed with combined Menin and KIT inhibition. These results define unique chromatin regulatory dependencies in GIST and identify potential therapeutic strategies for clinical application. SIGNIFICANCE: Although many malignancies rely on oncogene amplification, GIST instead depends upon epigenetic regulation of KIT and other essential genes. Utilizing genome-scale CRISPR dependency screens, we identified complementary chromatin-modifying complexes essential to GIST and characterize the consequences of their disruption, elucidating a novel therapeutic approach to this disease. This article is highlighted in the In This Issue feature, p. 1599.


Asunto(s)
Neoplasias Gastrointestinales , Tumores del Estroma Gastrointestinal , Histona Acetiltransferasas/metabolismo , Cromatina/genética , Epigénesis Genética , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Humanos , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Tirosina Quinasas Receptoras/genética , Factores de Transcripción/genética
17.
Clin Cancer Res ; 28(11): 2397-2408, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35325095

RESUMEN

PURPOSE: Leiomyosarcoma (LMS) is a neoplasm characterized by smooth muscle differentiation, complex copy-number alterations, tumor suppressor loss, and the absence of recurrent driver mutations. Clinical management for advanced disease relies on the use of empiric cytotoxic chemotherapy with limited activity, and novel targeted therapies supported by preclinical research on LMS biology are urgently needed. A lack of fidelity of established LMS cell lines to their mesenchymal neoplasm of origin has limited translational understanding of this disease, and few other preclinical models have been established. Here, we characterize patient-derived xenograft (PDX) models of LMS, assessing fidelity to their tumors of origin and performing preclinical evaluation of candidate therapies. EXPERIMENTAL DESIGN: We implanted 49 LMS surgical samples into immunocompromised mice. Engrafting tumors were characterized by histology, targeted next-generation sequencing, RNA sequencing, and ultra-low passage whole-genome sequencing. Candidate therapies were selected based on prior evidence of pathway activation or high-throughput dynamic BH3 profiling. RESULTS: We show that LMS PDX maintain the histologic appearance, copy-number alterations, and transcriptional program of their parental tumors across multiple xenograft passages. Transcriptionally, LMS PDX cocluster with paired LMS patient-derived samples and differ primarily in host-related immunologic and microenvironment signatures. We identify susceptibility of LMS PDX to transcriptional cyclin-dependent kinase (CDK) inhibition, which disrupts an E2F-driven oncogenic transcriptional program and inhibits tumor growth. CONCLUSIONS: Our results establish LMS PDX as valuable preclinical models and identify strategies to discover novel vulnerabilities in this disease. These data support the clinical assessment of transcriptional CDK inhibitors as a therapeutic strategy for patients with LMS.


Asunto(s)
Leiomiosarcoma , Animales , Carcinogénesis/patología , Modelos Animales de Enfermedad , Expresión Génica , Xenoinjertos , Humanos , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/genética , Leiomiosarcoma/patología , Ratones , Análisis de Secuencia de ARN , Microambiente Tumoral
18.
Cell ; 184(25): 6119-6137.e26, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34890551

RESUMEN

Prognostically relevant RNA expression states exist in pancreatic ductal adenocarcinoma (PDAC), but our understanding of their drivers, stability, and relationship to therapeutic response is limited. To examine these attributes systematically, we profiled metastatic biopsies and matched organoid models at single-cell resolution. In vivo, we identify a new intermediate PDAC transcriptional cell state and uncover distinct site- and state-specific tumor microenvironments (TMEs). Benchmarking models against this reference map, we reveal strong culture-specific biases in cancer cell transcriptional state representation driven by altered TME signals. We restore expression state heterogeneity by adding back in vivo-relevant factors and show plasticity in culture models. Further, we prove that non-genetic modulation of cell state can strongly influence drug responses, uncovering state-specific vulnerabilities. This work provides a broadly applicable framework for aligning cell states across in vivo and ex vivo settings, identifying drivers of transcriptional plasticity and manipulating cell state to target associated vulnerabilities.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral , Adulto , Anciano , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Análisis de la Célula Individual
19.
Front Nutr ; 8: 753843, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34722613

RESUMEN

The influence of fruit and vegetable consumption on semen quality by reducing oxidative stress is inconsistent. Thus, the association between the consumption of these products, antioxidant status, and semen quality was investigated in 90 men aged 18-40. The consumption of fruit and vegetables was collected using the 3-day food record method. Antioxidant status: total antioxidant capacity in semen (TAC-s) and blood (TAC-b), blood superoxide dismutase (SOD-b), glutathione reductase (GR-b), glutathione peroxidase (GPx-b), catalase (CAT-b) activity, and malondialdehyde concentration in blood (MDA-b) were measured. Sperm concentration, leukocytes in the ejaculate, vitality, motility, and sperm morphology were examined using computer-aided semen analysis (CASA). The consumption of fruit and vegetables was positively correlated with sperm concentration, vitality, motility, TAC-s, TAC-b, and SOD-b activity. The TAC-s and TAC-b were positively related to motility, TAC-s was inversely correlated with sperm tail defects. The SOD-b activity was positively correlated with vitality, motility, sperm morphology, and inversely with sperm tail defects and leukocytes in the ejaculate. Compared to the men in the first quartile of fruit and vegetable consumption (<318 g/day), those in the highest quartile (>734 g/day) had the highest sperm concentration, vitality, motility, TAC-s, TAC-b, GPx-b activity, and the lowest MDA-b concentration (based on multivariate regression models). A high consumption of fruit and vegetables may positively influence selected sperm quality parameters by improving the antioxidant status of semen and blood.

20.
Clin Cancer Res ; 27(6): 1695-1705, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33414136

RESUMEN

PURPOSE: Receptor tyrosine kinase fusions in colorectal cancers are rare, but potentially therapeutically relevant. We describe clinical, molecular, and pathologic attributes of RTK fusion-associated colorectal cancer. EXPERIMENTAL DESIGN: We identified all cases with RTK fusions in patients with colorectal cancer seen at Dana-Farber Cancer Institute (Boston, MA) who underwent OncoPanel testing between 2013 and 2018. Clinical, histologic, and molecular features were extracted from the patient charts and molecular testing results. RESULTS: We identified 12 driver oncogenic fusions in various RTKs. These fusions occurred exclusively in BRAF and RAS wild-type tumors and were enriched in right-sided and mismatch repair-deficient (MMR-D) colorectal cancers. All of the MMR-D colorectal cancers with RTK fusions were found in tumors with acquired MMR-D due to MLH1 promoter hypermethylation and one was associated with a sessile serrated polyp. Molecular profiles of MMR-D colorectal cancer with RTK fusions largely resembled BRAF V600E-mutated MMR-D colorectal cancer, rather than those secondary to Lynch syndrome. We describe two patients with fusion-associated microsatellite stable (MSS) colorectal cancer who derived clinical benefit from therapeutic targeting of their translocation. The first harbored an ALK-CAD fusion and received sequential crizotinib and alectinib therapy for a total of 7.5 months until developing an ALK L1196Q gatekeeper mutation. The second patient, whose tumor contained an ROS1-GOPC fusion, continues to benefit from entrectinib after 9 months of therapy. CONCLUSIONS: RTK fusions in colorectal cancer are a rare, but important disease subgroup that occurs in RAS and BRAF wild-type tumors. Despite enrichment in acquired MMR-D tumors, RTK fusions also occur in MSS colorectal cancer and provide an important therapeutic target.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/patología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/genética , Adulto , Anciano , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA