Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 128(4): 1355-1370, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29480816

RESUMEN

Metastatic breast cancers are still incurable. Characterizing the evolutionary landscape of these cancers, including the role of metastatic axillary lymph nodes (ALNs) in seeding distant organ metastasis, can provide a rational basis for effective treatments. Here, we have described the genomic analyses of the primary tumors and metastatic lesions from 99 samples obtained from 20 patients with breast cancer. Our evolutionary analyses revealed diverse spreading and seeding patterns that govern tumor progression. Although linear evolution to successive metastatic sites was common, parallel evolution from the primary tumor to multiple distant sites was also evident. Metastatic spreading was frequently coupled with polyclonal seeding, in which multiple metastatic subclones originated from the primary tumor and/or other distant metastases. Synchronous ALN metastasis, a well-established prognosticator of breast cancer, was not involved in seeding the distant metastasis, suggesting a hematogenous route for cancer dissemination. Clonal evolution coincided frequently with emerging driver alterations and evolving mutational processes, notably an increase in apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like-associated (APOBEC-associated) mutagenesis. Our data provide genomic evidence for a role of ALN metastasis in seeding distant organ metastasis and elucidate the evolving mutational landscape during cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Evolución Molecular , Mutación , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Femenino , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Metástasis de la Neoplasia
2.
BMC Cancer ; 17(1): 802, 2017 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-29187174

RESUMEN

BACKGROUND: Transcriptomic profiling of breast tumors provides opportunity for subtyping and molecular-based patient stratification. In diagnostic applications the specimen profiled should be representative of the expression profile of the whole tumor and ideally capture properties of the most aggressive part of the tumor. However, breast cancers commonly exhibit intra-tumor heterogeneity at molecular, genomic and in phenotypic level, which can arise during tumor evolution. Currently it is not established to what extent a random sampling approach may influence molecular breast cancer diagnostics. METHODS: In this study we applied RNA-sequencing to quantify gene expression in 43 pieces (2-5 pieces per tumor) from 12 breast tumors (Cohort 1). We determined molecular subtype and transcriptomic grade for all tumor pieces and analysed to what extent pieces originating from the same tumors are concordant or discordant with each other. Additionally, we validated our finding in an independent cohort consisting of 19 pieces (2-6 pieces per tumor) from 6 breast tumors (Cohort 2) profiled using microarray technique. Exome sequencing was also performed on this cohort, to investigate the extent of intra-tumor genomic heterogeneity versus the intra-tumor molecular subtype classifications. RESULTS: Molecular subtyping was consistent in 11 out of 12 tumors and transcriptomic grade assignments were consistent in 11 out of 12 tumors as well. Molecular subtype predictions revealed consistent subtypes in four out of six patients in this cohort 2. Interestingly, we observed extensive intra-tumor genomic heterogeneity in these tumor pieces but not in their molecular subtype classifications. CONCLUSIONS: Our results suggest that macroscopic intra-tumoral transcriptomic heterogeneity is limited and unlikely to have an impact on molecular diagnostics for most patients.


Asunto(s)
Neoplasias de la Mama/genética , Perfilación de la Expresión Génica/métodos , Heterogeneidad Genética , Biomarcadores de Tumor/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Análisis de Secuencia de ARN
3.
J Natl Cancer Inst ; 109(3): 1-14, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28376210

RESUMEN

Background: Breast cancer cells with tumor-initiating capabilities (BSCs) are considered to maintain tumor growth and govern metastasis. Hence, targeting BSCs will be crucial to achieve successful treatment of breast cancer. Methods: We characterized mammospheres derived from more than 40 cancer patients and two breast cancer cell lines for the expression of estrogen receptors (ERs) and stem cell markers. Mammosphere formation and proliferation assays were performed on cells from 19 cancer patients and five healthy individuals after incubation with ER-subtype selective ligands. Transcriptional analysis was performed to identify pathways activated in ERß-stimulated mammospheres and verified using in vitro experiments. Xenograft models (n = 4 or 5 per group) were used to study the role of ERs during tumorigenesis. Results: We identified an absence of ERα but upregulation of ERß in BSCs associated with phenotypic stem cell markers and responsible for the proliferative role of estrogens. Knockdown of ERß caused a reduction of mammosphere formation in cell lines and in patient-derived cancer cells (40.7%, 26.8%, and 39.1%, respectively). Gene set enrichment analysis identified glycolysis-related pathways (false discovery rate < 0.001) upregulated in ERß-activated mammospheres. We observed that tamoxifen or fulvestrant alone was insufficient to block proliferation of patient-derived BSCs while this could be accomplished by a selective inhibitor of ERß (PHTPP; 53.7% in luminal and 45.5% in triple-negative breast cancers). Furthermore, PHTPP reduced tumor initiation in two patient-derived xenografts (75.9% and 59.1% reduction in tumor volume, respectively) and potentiated tamoxifen-mediated inhibition of tumor growth in MCF7 xenografts. Conclusion: We identify ERß as a mediator of estrogen action in BSCs and a novel target for endocrine therapy.


Asunto(s)
Carcinoma/genética , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Pirazoles/farmacología , Pirimidinas/farmacología , Esferoides Celulares/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Proliferación Celular/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Femenino , Fulvestrant , Técnicas de Silenciamiento del Gen , Glucólisis , Humanos , Células MCF-7 , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Fenotipo , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Esferoides Celulares/efectos de los fármacos , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral , Regulación hacia Arriba
4.
Cancer Lett ; 367(1): 76-87, 2015 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-26208432

RESUMEN

Breast cancer cells with stem cell characteristics (CSC) are a distinct cell population with phenotypic similarities to mammary stem cells. CSCs are important drivers of tumorigenesis and the metastatic process. Tamoxifen is the most widely used hormonal therapy for estrogen receptor (ER) positive cancers. In our study, tamoxifen was effective in reducing proliferation of ER + adherent cancer cells, but not their CSC population. We isolated, expanded and incubated CSC from seven breast cancers with or without tamoxifen. By genome-wide transcriptional analysis we identified tamoxifen-induced transcriptional pathways associated with ribosomal biogenesis and mRNA translation, both regulated by the mTOR-pathway. We observed induction of the key mTOR downstream targets S6K1, S6RP and 4E-BP1 in-patient derived CSCs by tamoxifen on protein level. Using the mTOR inhibitors rapamycin, everolimus and PF-04691502 (a dual PI3K/mTOR inhibitor) and in combination with tamoxifen, significant reduction in mammosphere formation was observed. Hence, we suggest that the CSC population play a significant role during endocrine resistance through activity of the mTOR pathway. In addition, tamoxifen further stimulates the mTOR-pathway but can be antagonized using mTOR-inhibitors.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Neoplasias de la Mama/enzimología , Antagonistas de Estrógenos/toxicidad , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tamoxifeno/toxicidad , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Separación Celular/métodos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática , Everolimus , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Piridonas/farmacología , Pirimidinas/farmacología , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Sirolimus/farmacología , Esferoides Celulares , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética , Activación Transcripcional , Células Tumorales Cultivadas
5.
Lab Invest ; 94(7): 796-805, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24776644

RESUMEN

Breast cancer is a unique tumor disease in terms of the stringent requirement of predictive biomarker assessments. As recommended by current international guidelines, the established markers consist of estrogen receptor (ER), progesterone receptor, human epidermal growth factor and Ki67, and are primarily analyzed by immunohistochemistry. However, new diagnostic methods based on microarray or next-generation sequencing on DNA and mRNA level are gaining ground. These analyses require fresh-frozen tumor tissue that is generally not available from tumors <10 mm in diameter, comprising almost 25% of all resected breast cancer at our department. We here present a simple and standardized method to generate material from small tumors without risking the histopathological examination. Furthermore, we show that the quality of this material is sufficient for subsequent analysis on mRNA, DNA, and epigenetic level. We were also able to use this method for isolation and expansion of cancer stem cells from the majority of tumors. Consequently, researches can be provided with clinically relevant material for translational studies. In conclusion, this method opens up a new possibility for usage of valuable fresh tumor material for research purposes, biobanking, and next-generation sequencing.


Asunto(s)
Bancos de Muestras Biológicas/normas , Investigación Biomédica/métodos , Neoplasias de la Mama/patología , Manejo de Especímenes/métodos , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Metilación de ADN , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , Femenino , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Patología Molecular/métodos , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN
6.
PLoS One ; 5(12): e15064, 2010 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-21179214

RESUMEN

BACKGROUND: Recognizing specific protein changes in response to drug administration in humans has the potential for the development of personalized medicine. Such changes can be identified by pharmacoproteomics approach based on proteomic technologies. It can also be helpful in matching a particular target-based therapy to a particular marker in a subgroup of patients, in addition to the profile of genetic polymorphism. Warfarin is a commonly prescribed oral anticoagulant in patients with prosthetic valve disease, venous thromboembolism and stroke. METHODS AND FINDING: We used a combined pharmacogenetics and iTRAQ-coupled LC-MS/MS pharmacoproteomics approach to analyze plasma protein profiles of 53 patients, and identified significantly upregulated level of transthyretin precursor in patients receiving low dose of warfarin but not in those on high dose of warfarin. In addition, real-time RT-PCR, western blotting, human IL-6 ELISA assay were done for the results validation. CONCLUSION: This combined pharmacogenomics and pharmacoproteomics approach may be applied for other target-based therapies, in matching a particular marker in a subgroup of patients, in addition to the profile of genetic polymorphism.


Asunto(s)
Anticoagulantes/farmacología , Biomarcadores/metabolismo , Oxigenasas de Función Mixta/genética , Farmacogenética/métodos , Prealbúmina/biosíntesis , Proteómica/métodos , Warfarina/farmacología , Línea Celular Tumoral , Estudios de Cohortes , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Interleucina-6/metabolismo , Hígado/metabolismo , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem/métodos , Vitamina K Epóxido Reductasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...