Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 977
Filtrar
1.
Mol Genet Genomics ; 297(4): 947-963, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35532795

RESUMEN

Malignancies dependent on hormone homeostasis include breast, ovary, cervical, prostate, testis and uterine tumors. Hormones are involved in signal transduction which orchestrate processes, such as apoptosis, proliferation, cell cycle or cytoskeleton organization. Currently, there is a need for novel biomarkers which would help to diagnose cancers efficiently. In this study, the genes implicated in signaling that is important in hormone-sensitive carcinogenesis were investigated regarding their prognostic significance. Data of seven cancer cohorts were collected from FireBrowse. 54 gene sets implicated in specific pathways were browsed through MSig database. Profiling was assessed via Monocle3, while gene ontology through PANTHER. For confirmation, correlation analysis was performed using WGCNA. Protein-protein networks were visualized via Cytoscape and impact of genes on survival, as well as cell cycle or cytoskeleton-related prognostic signatures, was tested. Several differences in expression profile were identified, some of them allowed to distinguish histology. Functional annotation revealed that various regulation of cell cycle, adhesion, migration, apoptosis and angiogenesis underlie these differences. Clinical traits, such as histological type or cancer staging, were found during evaluation of module-trait relationships. Of modules, the TopHubs (COL6A3, TNR, GTF2A1, NKX3-1) interacted directly with, e.g., PDGFB, ITGA10, SP1 or AKT3. Among TopHubs and interacting proteins, many showed an impact on hazard ratio and affected the cell cycle or cytoskeleton-related prognostic signatures, e.g., COL1A1 or PDGFB. In conclusion, this study laid the foundation for further hormone-sensitive carcinogenesis research through identification of genes which prove that crosstalk between cell cycle and cytoskeleton exists, opening avenues for future therapeutic strategies.


Asunto(s)
Neoplasias Hormono-Dependientes , Neoplasias , Biomarcadores , Carcinogénesis/genética , Ciclo Celular/genética , Biología Computacional , Citoesqueleto/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Hormonas , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias Hormono-Dependientes/genética , Proteínas Proto-Oncogénicas c-sis/genética , Transducción de Señal/genética
2.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35163166

RESUMEN

It is known that estrogen stimulates growth and inhibits apoptosis through estrogen receptor(ER)-mediated mechanisms in many cancer cell types. Interestingly, there is strong evidence that estrogens can also induce apoptosis, activating different ER isoforms in cancer cells. It has been observed that E2/ERα complex activates multiple pathways involved in both cell cycle progression and apoptotic cascade prevention, while E2/ERß complex in many cases directs the cells to apoptosis. However, the exact mechanism of estrogen-induced tumor regression is not completely known. Nevertheless, ERs expression levels of specific splice variants and their cellular localization differentially affect outcome of estrogen-dependent tumors. The goal of this review is to provide a general overview of current knowledge on ERs-mediated apoptosis that occurs in main hormone dependent-cancers. Understanding the molecular mechanisms underlying the induction of ER-mediated cell death will be useful for the development of specific ligands capable of triggering apoptosis to counteract estrogen-dependent tumor growth.


Asunto(s)
Apoptosis , Neoplasias Hormono-Dependientes/patología , Receptores de Estrógenos/metabolismo , Animales , Humanos , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/genética , Transducción de Señal
3.
Genes (Basel) ; 12(8)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34440460

RESUMEN

Endocrine tumors are neoplasms originating from specialized hormone-secreting cells. They can develop as sporadic tumors, caused by somatic mutations, or in the context of familial Mendelian inherited diseases. Congenital forms, manifesting as syndromic or non-syndromic diseases, are caused by germinal heterozygote autosomal dominant mutations in oncogenes or tumor suppressor genes. The genetic defect leads to a loss of cell growth control in target endocrine tissues and to tumor development. In addition to the classical cancer manifestations, some affected patients can manifest alterations of bone and mineral metabolism, presenting both as pathognomonic and/or non-specific skeletal clinical features, which can be either secondary complications of endocrine functioning primary tumors and/or a direct consequence of the gene mutation. Here, we specifically review the current knowledge on possible direct roles of the genes that cause inherited endocrine tumors in the regulation of bone modeling and remodeling by exploring functional in vitro and in vivo studies highlighting how some of these genes participate in the regulation of molecular pathways involved in bone and mineral metabolism homeostasis, and by describing the potential direct effects of gene mutations on the development of skeletal and mineral metabolism clinical features in patients.


Asunto(s)
Neoplasias de las Glándulas Endocrinas/genética , Genes Supresores de Tumor , Neoplasias Hormono-Dependientes/genética , Osteoporosis/genética , Huesos/metabolismo , Huesos/patología , Neoplasias de las Glándulas Endocrinas/complicaciones , Neoplasias de las Glándulas Endocrinas/patología , Heterocigoto , Humanos , Mutación , Neoplasias Hormono-Dependientes/complicaciones , Neoplasias Hormono-Dependientes/patología , Oncogenes/genética , Osteoporosis/complicaciones , Osteoporosis/patología
4.
Cancer Lett ; 520: 48-56, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34229060

RESUMEN

The deregulation of PI3K/Akt signaling is among the most causes in inducing the acquisition of a metastatic phenotype in breast cancer cells, leading to Epithelial-Mesenchymal Transition (EMT). Inhibition of the PI3K/Akt pathway is known to be beneficial in the clinical setting. However, the activation of secondary pathways and toxicity profiles of available inhibitors, hindering optimal therapeutic results. Preliminary studies showed that myo-Inositol inhibits the PI3K/Akt pathway by exerting a pleiotropic anti-tumor action. Herein, we demonstrate that myo-Inositol triggers a prompt and profound remodeling of delineated expression pattern in triple-negative breast cancer cells (MDA-MB-231). Consequently, it inhibits metastasis and tumor progression through miR-125a-5p transcription and the subsequent inhibition of IP6K1. In contrast, hormone-responsive breast cancer cells (MCF-7) are insensitive to myo-Inositol. This is due to the persistence of MDM2 synthesis promoted by estrogen-dependent pathways. Conversely, the counteraction of estrogen effects recovered the sensitivity to myo-Inositol in the hormone-responsive model. Overall, these results identify a novel axis primed by miR-125a-5p to downregulate IP6K1 gene that inhibits metastasis. Thus, administration of myo-Inositol can activate this axis as a molecular target therapy in breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , MicroARNs/genética , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inositol/farmacología , Células MCF-7 , Metástasis de la Neoplasia , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Transducción de Señal/efectos de los fármacos
5.
Int J Mol Sci ; 22(11)2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34199930

RESUMEN

Endometriosis is a common gynecological disorder that has been associated with endometrial, breast and epithelial ovarian cancers in epidemiological studies. Since complex diseases are a result of multiple environmental and genetic factors, we hypothesized that the biological mechanism underlying their comorbidity might be explained, at least in part, by shared genetics. To assess their potential genetic relationship, we performed a two-sample mendelian randomization (2SMR) analysis on results from public genome-wide association studies (GWAS). This analysis confirmed previously reported genetic pleiotropy between endometriosis and endometrial cancer. We present robust evidence supporting a causal genetic association between endometriosis and ovarian cancer, particularly with the clear cell and endometrioid subtypes. Our study also identified genetic variants that could explain those associations, opening the door to further functional experiments. Overall, this work demonstrates the value of genomic analyses to support epidemiological data, and to identify targets of relevance in multiple disorders.


Asunto(s)
Neoplasias Endometriales/epidemiología , Endometriosis/epidemiología , Endometrio/patología , Predisposición Genética a la Enfermedad , Neoplasias Hormono-Dependientes/epidemiología , Neoplasias Ováricas/epidemiología , Polimorfismo de Nucleótido Simple , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Endometriosis/genética , Endometriosis/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Factores de Riesgo , España/epidemiología
6.
Prostate ; 81(9): 572-579, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33955569

RESUMEN

BACKGROUND: Clinical outcomes may differ among patients presenting with primary (de novo) metastatic hormone-sensitive prostate cancer (mHSPC) versus secondary (metachronous) mHSPC occurring after local therapy. It is unknown what molecular features distinguish these potentially distinct presentations. METHODS: A single-center retrospective study of mHSPC patients classified as primary mHSPC (n = 121) or secondary mHSPC (n = 106). A targeted set of genes was analyzed: BRCA2, PTEN, RB1, TP53, SPOP, CDK12, any two out of PTEN/RB1/TP53 alterations, and homologous recombination deficiency mutations. TP53 mutations were categorized as loss-of-function (LOF) versus dominant-negative (DN). The impacts of genetic features on progression-free survival (PFS) and overall survival (OS) were assessed using univariate and multivariate Cox proportional hazards regression. RESULTS: Median PFS was 15 and 30 months for men with primary and secondary mHSPC, respectively (hazard ratio: 0.57, 95% confidence interval: 0.41-0.78; p < .01). OS did not show a significant difference between groups. There were more men with Gleason 8-10 disease in the primary versus secondary mHSPC groups (83% vs. 68%; p < .01). In univariate and multivariate analyses, TP53 DN mutations showed a statistically significant association with OS for the entire mHSPC population. Conversely, SPOP mutations were associated with improved OS. Additionally, TP53 mutations (DN and LOF) were associated with worse OS for secondary mHSPC. A combination of PTEN/RB1/TP53 mutations was associated with worse OS and PFS for secondary mHSPC, while no genomic alteration affected outcomes for primary mHSPC. CONCLUSIONS: TP53 DN mutations, but not all TP53 alterations, were the strongest predictor of negative outcomes in men with mHSPC, while SPOP mutations were associated with improved outcomes. In subgroup analyses, specific alterations were prognostic of outcomes in secondary, but not primary, mHSPC.


Asunto(s)
Genes p53/genética , Neoplasias Hormono-Dependientes , Neoplasias Primarias Secundarias , Proteínas Nucleares/genética , Neoplasias de la Próstata , Proteínas Represoras/genética , Antagonistas de Andrógenos/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Metástasis de la Neoplasia/genética , Estadificación de Neoplasias , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Neoplasias Primarias Secundarias/tratamiento farmacológico , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/patología , Pronóstico , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Estudios Retrospectivos
7.
Anticancer Res ; 41(5): 2669-2680, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33952498

RESUMEN

BACKGROUND/AIM: The histopathological variability of each type of pituitary adenoma (PA) that causes growth hormone (GH) excess influences the phenotype, radiological characteristics and therapy response of acromegaly patients. We correlated the immunohistochemical (IHC) features of GH-secreting PAs with their clinical, laboratory and imaging data. PATIENTS AND METHODS: We included 32 patients with documented acromegaly; tumour specimens were histologically and IHC examined: anterior pituitary hormones, pituitary-specific transcription factor-1 (PIT-1), Ki-67 labelling index were evaluated. RESULTS: Macroadenomas represented 93.75%. Post-surgery disease control negatively correlated with the maximum initial tumour diameter (p=0.04). Ki-67 did not predict remission. No correlation was found between GH serum levels and IHC expression (p=0.45). PIT-1 was positive in all specimens, two had a weak expression. Four were considered PIT-1 positive plurihormonal adenomas and several had unusual IHC combinations. CONCLUSION: PIT-1 accurately classifies GH-secreting PAs. The IHC classification as well as radiological dimensions and extent influence disease control, probably being the best prognosis factors.


Asunto(s)
Acromegalia/sangre , Hormona del Crecimiento/sangre , Neoplasias Hormono-Dependientes/sangre , Neoplasias Hipofisarias/sangre , Acromegalia/complicaciones , Acromegalia/genética , Acromegalia/patología , Adulto , Anciano , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Antígeno Ki-67/sangre , Masculino , Persona de Mediana Edad , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Hormonas Adenohipofisarias/sangre , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Periodo Preoperatorio , Factor de Transcripción Pit-1/sangre
8.
Prostate Cancer Prostatic Dis ; 24(2): 482-491, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33214693

RESUMEN

BACKGROUND: Prostate cancer (PCa) shows racial disparity in clinical and genomic characteristics, and Asian patients with PCa often present with more aggressive phenotypes at diagnosis. The ability of TP53 to serve as a prognostic biomarker of PCa has been well studied in Western populations. However, no studies to date have examined the role of TP53 in the disparities of primary hormone-naïve prostate cancer (HNPC) between Chinese and Western populations. METHODS: We collected prostate tumors and matched normal tissues or blood samples to perform targeted next-generation sequencing of 94 Chinese primary localized HNPC samples, and correlated these genomic profiles with clinical outcomes. The OncoKB knowledge database was used to identify and classify actionable alterations. RESULTS: The aberrations of PTEN, CDK12, and SPOP in Chinese HNPC samples were similar to those in the Western samples. However, we demonstrated an association of a high frequency of TP53 alterations (21/94) with a relatively higher percentage of alterations in the Wnt signaling pathway (15/94) in Chinese HNPC. Additionally, we highlighted alterations of LRP1B as accounting for a high proportion of PCa and found more frequent alterations in CDH1 in Chinese PCa. Of these, only CDH1 alteration was associated with rapid biochemical recurrence (BCR). However, we verified that TP53 status was at the core of the genomic alteration landscape in Chinese HNPC with putative driver mutations because of the strong connections with other signaling pathways. The mutually exclusive relationship between alterations in TP53 and Wnt/CTNNB1 further molecularly characterizes subsets of prostate cancers. Moreover, the alteration of KMT2C was more likely to co-occur with TP53 alteration, indicating a more aggressive phenotype of PCa, which was associated with sensitivity to treatment with poly ADT-ribose polymerase (PARP) inhibitors. CONCLUSIONS: Detection of TP53 alterations has clinical utility for guiding precision cancer therapy for HNPC, especially in the Chinese population.


Asunto(s)
Biomarcadores de Tumor/genética , Mutación , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/patología , Proteína p53 Supresora de Tumor/genética , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , China/epidemiología , Estudios de Seguimiento , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Neoplasias Hormono-Dependientes/epidemiología , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/cirugía , Pronóstico , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/cirugía , Tasa de Supervivencia
9.
J Hum Genet ; 66(3): 307-314, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33046835

RESUMEN

BRCA1/2 pathogenic variant prevalence in Japanese breast cancer is unclear. Here, we analyzed BRCA1/2 pathogenic variant prevalence with a particular focus on age factors, using the Japanese HBOC consortium database. All registered subjects were Japanese individuals who underwent BRCA1/2 genetic testing from January 1996 to July 2017 according to the Japanese HBOC consortium database. Cases were extracted and analyzed for each evaluation item. Overall BRCA1 and BRCA2 pathogenic variant prevalence was 11.2% and 9.0% in the cohort of 2366 proband patients, respectively. The age at onset of breast cancer for patients with BRCA1/2 pathogenic variants was significantly lower than that for patients without a BRCA1/2 pathogenic variant. In both BRCA1/2 patients, ages at onset were not statistically significantly different between two subtype groups (ER-positive vs. TNBC). We analyzed the BRCA1/2 pathogenic variant prevalence among age groups in patients with no family history of breast or ovarian cancer. In the TNBC group, the rate of genetic variants was more frequent among younger patients. Our results demonstrated that early breast cancer onset is associated with a BRCA1/2 pathogenic variant in the Japanese population. Younger TNBC patients were more likely to have a BRCA1/2 pathogenic variant irrespective of a family history of breast or ovarian cancer.


Asunto(s)
Edad de Inicio , Pueblo Asiatico/genética , Neoplasias de la Mama/genética , Genes BRCA1 , Genes BRCA2 , Síndrome de Cáncer de Mama y Ovario Hereditario/genética , Proteína BRCA1 , Proteína BRCA2 , Neoplasias de la Mama/química , Neoplasias de la Mama/epidemiología , Estrógenos , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Mutación de Línea Germinal , Síndrome de Cáncer de Mama y Ovario Hereditario/epidemiología , Síndrome de Cáncer de Mama y Ovario Hereditario/etnología , Humanos , Japón/epidemiología , Neoplasias Hormono-Dependientes/química , Neoplasias Hormono-Dependientes/epidemiología , Neoplasias Hormono-Dependientes/genética , Prevalencia , Progesterona , Receptor ErbB-2/análisis , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Neoplasias de la Mama Triple Negativas/epidemiología , Neoplasias de la Mama Triple Negativas/genética
10.
J Hum Genet ; 66(5): 519-534, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33177704

RESUMEN

Our recent research has revealed that passenger strands of certain microRNAs (miRNAs) function as tumor-suppressive miRNAs in cancer cells, e.g., miR-101-5p, miR-143-5p, miR-144-5p, miR-145-3p, and miR-150-3p. Thus, they are important in cancer pathogenesis. Analysis of the miRNA expression signature of breast cancer (BrCa) showed that the expression levels of two miRNAs derived from pre-miR-99a (miR-99a-5p and miR-99a-3p) were suppressed in cancerous tissues. The aim of this study was to identify oncogenic genes controlled by pre-miR-99a that are closely involved in the molecular pathogenesis of BrCa. A total of 113 genes were identified as targets of pre-miR-99a regulation (19 genes modulated by miR-99a-5p, and 95 genes regulated by miR-99a-3p) in BrCa cells. Notably, FAM64A was targeted by both of the miRNAs. Among these targets, high expression of 16 genes (C5orf22, YOD1, SLBP, F11R, C12orf49, SRPK1, ZNF250, ZNF695, CDK1, DNMT3B, TRIM25, MCM4, CDKN3, PRPS, FAM64A, and DESI2) significantly predicted reduced survival of BrCa patients based upon The Cancer Genome Atlas (TCGA) database. In this study, we focused on FAM64A and investigated the relationship between FAM64A expression and molecular pathogenesis of BrCa subtypes. The upregulation of FAM64A was confirmed in BrCa clinical specimens. Importantly, the expression of FAM64A significantly differed between patients with Luminal-A and Luminal-B subtypes. Our data strongly suggest that the aberrant expression of FAM64A is involved in the malignant transformation of BrCa. Our miRNA-based approaches (identification of tumor-suppressive miRNAs and their controlled targets) will provide novel information regarding the molecular pathogenesis of BrCa.


Asunto(s)
Neoplasias de la Mama/genética , Estrógenos , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor , Péptidos y Proteínas de Señalización Intracelular/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Neoplasias Hormono-Dependientes/genética , Proteínas Nucleares/genética , Oncogenes , Progesterona , ARN Neoplásico/genética , Aminopiridinas/administración & dosificación , Aminopiridinas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bencimidazoles/administración & dosificación , Bencimidazoles/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Genes erbB-2 , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/fisiología , Estimación de Kaplan-Meier , MicroARNs/fisiología , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/fisiología , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/mortalidad , Neoplasias Hormono-Dependientes/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/fisiología , Piperazinas/administración & dosificación , Piperazinas/uso terapéutico , Pronóstico , Supervivencia sin Progresión , Piridinas/administración & dosificación , Piridinas/uso terapéutico , Interferencia de ARN , ARN Neoplásico/fisiología , ARN Interferente Pequeño/genética , Resultado del Tratamiento
11.
Lancet Oncol ; 21(11): 1443-1454, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33152284

RESUMEN

BACKGROUND: Preoperative and perioperative aromatase inhibitor (POAI) therapy has the potential to improve outcomes in women with operable oestrogen receptor-positive primary breast cancer. It has also been suggested that tumour Ki67 values after 2 weeks (Ki672W) of POAI predicts individual patient outcome better than baseline Ki67 (Ki67B). The POETIC trial aimed to test these two hypotheses. METHODS: POETIC was an open-label, multicentre, parallel-group, randomised, phase 3 trial (done in 130 UK hospitals) in which postmenopausal women aged at least 50 years with WHO performance status 0-1 and hormone receptor-positive, operable breast cancer were randomly assigned (2:1) to POAI (letrozole 2·5 mg per day orally or anastrozole 1 mg per day orally) for 14 days before and following surgery or no POAI (control). Adjuvant treatment was given as per UK standard local practice. Randomisation was done centrally by computer-generated permuted block method (variable block size of six or nine) and was stratified by hospital. Treatment allocation was not masked. The primary endpoint was time to recurrence. A key second objective explored association between Ki67 (dichotomised at 10%) and disease outcomes. The primary analysis for clinical endpoints was by modified intention to treat (excluding patients who withdrew consent). For Ki67 biomarker association and endpoint analysis, the evaluable population included all randomly assigned patients who had paired Ki67 values available. This study is registered with ClinicalTrials.gov, NCT02338310; the European Clinical Trials database, EudraCT2007-003877-21; and the ISRCTN registry, ISRCTN63882543. Recruitment is complete and long-term follow-up is ongoing. FINDINGS: Between Oct 13, 2008, and April 16, 2014, 4480 women were recruited and randomly assigned to POAI (n=2976) or control (n=1504). On Feb 6, 2018, median follow-up was 62·9 months (IQR 58·1-74·1). 434 (10%) of 4480 women had a breast cancer recurrence (280 [9%] POAI; 154 [10%] control), hazard ratio 0·92 (95% CI 0·75-1·12); p=0·40 with the proportion free from breast cancer recurrence at 5 years of 91·0% (95% CI 89·9-92·0) for patients in the POAI group and 90·4% (88·7-91·9) in the control group. Within the POAI-treated HER2-negative subpopulation, 5-year recurrence risk in women with low Ki67B and Ki672W (low-low) was 4·3% (95% CI 2·9-6·3), 8·4% (6·8-10·5) with high Ki67B and low Ki672W (high-low) and 21·5% (17·1-27·0) with high Ki67B and Ki672W (high-high). Within the POAI-treated HER2-positive subpopulation, 5-year recurrence risk in the low-low group was 10·1% (95% CI 3·2-31·3), 7·7% (3·4-17·5) in the high-low group, and 15·7% (10·1-24·4) in the high-high group. The most commonly reported grade 3 adverse events were hot flushes (20 [1%] of 2801 patients in the POAI group vs six [<1%] of 1400 in the control group) and musculoskeletal pain (29 [1%] vs 13 [1%]). No treatment-related deaths were reported. INTERPRETATION: POAI has not been shown to improve treatment outcome, but can be used without detriment to help select appropriate adjuvant therapy based on tumour Ki67. Most patients with low Ki67B or low POAI-induced Ki672W do well with adjuvant standard endocrine therapy (giving consideration to clinical-pathological factors), whereas those whose POAI-induced Ki672W remains high might benefit from further adjuvant treatment or trials of new therapies. FUNDING: Cancer Research UK.


Asunto(s)
Inhibidores de la Aromatasa/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Antígeno Ki-67/genética , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Anciano , Inhibidores de la Aromatasa/efectos adversos , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Posmenopausia/efectos de los fármacos , Pronóstico , Receptor ErbB-2/genética
12.
Dis Markers ; 2020: 8884450, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33082888

RESUMEN

The AR signaling pathway plays an important role in initiation and progression of many hormone-related cancers including prostate, bladder, kidney, lung, and breast cancer. However, the potential roles of androgen-responsive long noncoding RNAs (lncRNAs) in hormone-related cancers remained unclear. In the present study, we identified 469 novel androgen-responsive lncRNAs using microarray data. After validating the accuracy of the array data, we constructed a transcriptional network which contained more than 30 transcriptional factors using ChIP-seq data to explore upstream regulators of androgen-responsive lncRNAs. Next, we conducted bioinformatics analysis to identify lncRNA-miRNA-mRNA regulatory network. To explore the potential roles of androgen-responsive lncRNAs in hormone-related cancers, we performed coexpression network and PPI network analyses using TCGA data. GO and KEGG analyses showed these lncRNAs were mainly involved in regulating signal transduction, transcription, development, cell adhesion, immune response, cell differentiation, and MAPK signaling pathway. We also highlight the prognostic value of HPN-AS1, TPTEP1, and LINC00623 in cancer outcomes. Our results suggest that androgen-responsive lncRNAs played important roles in regulating hormone-related cancer progression and could be novel molecular biomarkers.


Asunto(s)
Andrógenos/farmacología , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Neoplasias Hormono-Dependientes/genética , ARN Largo no Codificante/genética , Perfilación de la Expresión Génica , Humanos , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/patología , Pronóstico , Transducción de Señal , Tasa de Supervivencia
13.
Sci Rep ; 10(1): 14071, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32826944

RESUMEN

Stratification of breast cancer (BC) into molecular subtypes by multigene expression assays is of demonstrated clinical utility. In principle, global RNA-sequencing (RNA-seq) should enable reconstructing existing transcriptional classifications of BC samples. Yet, it is not clear whether adaptation to RNA-seq of classifiers originally developed using PCR or microarrays, or reconstruction through machine learning (ML) is preferable. Hence, we focused on robustness and portability of PAM50, a nearest-centroid classifier developed on microarray data to identify five BC "intrinsic subtypes". We found that standard PAM50 is profoundly affected by the composition of the sample cohort used for reference construction, and we propose a strategy, named AWCA, to mitigate this issue, improving classification robustness, with over 90% of concordance, and prognostic ability; we also show that AWCA-based PAM50 can even be applied as single-sample method. Furthermore, we explored five supervised learners to build robust, single-sample intrinsic subtype callers via RNA-seq. From our ML-based survey, regularized multiclass logistic regression (mLR) displayed the best performance, further increased by ad-hoc gene selection on the global transcriptome. On external test sets, mLR classifications reached 90% concordance with PAM50-based calls, without need of reference sample; mLR proven robustness and prognostic ability make it an equally valuable single-sample method to strengthen BC subtyping.


Asunto(s)
Neoplasias de la Mama/clasificación , Carcinoma/clasificación , Aprendizaje Automático , Análisis de Secuencia de ARN , Biomarcadores de Tumor , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Carcinoma/química , Carcinoma/genética , Conjuntos de Datos como Asunto , Estrógenos , Femenino , Humanos , Modelos Logísticos , Neoplasias Hormono-Dependientes/química , Neoplasias Hormono-Dependientes/genética , Pronóstico , Receptores de Estrógenos/análisis , Recurrencia
14.
FASEB J ; 34(9): 11511-11528, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32713076

RESUMEN

The androgen receptor (AR) is a major driver of prostate cancer (PCa) and a key therapeutic target for AR inhibitors (ie, Enzalutamide). However, Enzalutamide only inhibits androgen-dependent AR signaling, enabling intrinsic AR activation via androgen-independent pathways, leading to aggressive castration-resistant PCa (CRPC). We investigated the ability of novel anti-cancer agents, Dp44mT and DpC, to overcome androgen resistance. The effect of Dp44mT and DpC on androgen-dependent and independent AR signaling was assessed in androgen-dependent and -independent PCa cells using 2D- and 3D-tissue culture. The clinically trialed DpC was then examined in vivo and compared to Enzalutamide. These agents uniquely promote AR proteasomal degradation and inhibit AR transcription in PCa cells via the upregulation of c-Jun, potently reducing the AR target, prostate-specific antigen (PSA). These agents also inhibited the activation of key molecules in both androgen-dependent and independent AR signaling (ie, EGFR, MAPK, PI3K), which promote CRPC. The clinically trialed DpC also significantly inhibited PCa tumor growth, AR, and PSA expression in vivo, being more potent than Enzalutamide. DpC is a promising candidate for a unique, structurally distinct generation of AR inhibitors that simultaneously target both androgen-dependent and independent arms of AR signaling. No other therapies exhibit such comprehensive and potent AR suppression, which is critical for overcoming the development of androgen resistance.


Asunto(s)
Andrógenos/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Tiosemicarbazonas/farmacología , Andrógenos/farmacología , Animales , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/genética , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
15.
Breast Cancer Res Treat ; 183(1): 95-105, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32601968

RESUMEN

PURPOSE: The development of multi-gene signatures has led to improvements in identification of breast cancer patients at high risk of recurrence. The prognostic power of commercially available gene signatures is mostly restricted to estrogen receptor (ER)-positive breast cancer. On the contrary, immune-related gene signatures predict prognosis only in ER-negative breast cancer. This study aimed to develop a better prognostic signature for breast cancer. METHODS: The expressions of long non-coding RNA (lncRNA) genes from 30 independent microarray datasets with a total of 4813 samples were analyzed. A prognostic lncRNA signature was developed based on likelihood-ratio Cox regression analysis. Survival analysis was used to compare the prognostic efficiencies of our signature and 10 previously reported prognostic gene signatures. RESULTS: Cox regression analysis on 30 independent datasets showed that the 6-lncRNA signature identified in this study performed as well as five commercially available signatures in recurrence prediction for ER-positive breast cancer. In ER-negative breast cancer, this lncRNA signature was as prognostic as three immune-related gene signatures. Moreover, our lncRNA signature also demonstrated a good capacity to predict recurrence risk for triple-negative breast cancer. Function analysis showed that several lncRNAs in this signature were probably involved in cell proliferation and immune processes. CONCLUSIONS: A six-LncRNA signature was identified that is prognostic for ER-positive, ER-negative, and triple-negative breast cancers and thus deserves further validation in prospective studies.


Asunto(s)
Neoplasias de la Mama/genética , Estrógenos , Neoplasias Hormono-Dependientes/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Transcriptoma , Biomarcadores de Tumor , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , División Celular , Conjuntos de Datos como Asunto , Femenino , Humanos , Funciones de Verosimilitud , Análisis Multivariante , Proteínas de Neoplasias/análisis , Neoplasias Hormono-Dependientes/inmunología , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/biosíntesis , Receptores de Estrógenos/análisis , Recurrencia , Análisis de Matrices Tisulares , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad
16.
Breast Cancer Res Treat ; 183(1): 41-48, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32572714

RESUMEN

BACKGROUND: Multi-gene expression assays have been developed with the aim of predicting late recurrence in patients with estrogen receptor (ER)-positive breast cancer. However, establishment of alternative markers based on immunohistochemistry is also important for achieving practical use. Based on our previous study, forkhead box A1 (FOXA1) protein was tested as a potentially useful predictive marker for late recurrence. METHODS: 117 patients with ER-positive HER2-negative invasive breast cancer who developed distant metastasis following curative surgery were retrospectively investigated. We also evaluated responsiveness to endocrine therapy according to FOXA1 expression. Furthermore, publicly available mRNA microarray data were analyzed to examine patterns of metastasis according to FOXA1 mRNA expression, employing the Kaplan-Meier plotter. RESULTS: High expression of FOXA1 was an independent factor predicting long disease-free survival (DFS), along with small tumor size (p = 0.010 and 0.016, respectively). Discrimination of DFS was improved by combining these two factors, i.e., patients with FOXA1-high small tumors had the longest DFS while those with FOXA1-low large tumors had the shortest DFS. Moreover, we revealed that risk of distant metastasis started to increase after the completion of adjuvant endocrine therapy in patients with FOXA1-high tumors. CONCLUSION: Among patients who developed distant metastasis, those with FOXA1-high tumors had significantly longer DFS. We believe our data to raise the possibility of FOXA1 being a useful predictive marker for late recurrence and to provide new insights into the biology of FOXA1-high breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estrógenos , Factor Nuclear 3-alfa del Hepatocito/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias Hormono-Dependientes/metabolismo , Adulto , Anciano , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/fisiología , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias Hormono-Dependientes/química , Neoplasias Hormono-Dependientes/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Receptor ErbB-2/análisis , Receptores de Estrógenos/análisis , Recurrencia , Factores de Tiempo
17.
Breast Cancer Res Treat ; 183(1): 1-8, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32572717

RESUMEN

OBJECTIVES: The NR5A2 and RYR2 genes are important players in steroid metabolism and play an important role in cancer research. In this research, we want to evaluate the effect of NR5A2 and RYR2 polymorphisms on breast cancer (BC). METHODS: Four single nucleotide polymorphisms on NR5A2 and RYR2 were selected to genotype by Agena MassARRAY in 379 BC patients and 407 healthy controls. Using the PLINK software to calculate the Odds ratio (OR) and 95% confidence intervals (CIs) via the logistic regression analysis to evaluate the risk for BC. RESULTS: We found that NR5A2 rs2246209 significantly decreased the risk of BC with the AA genotype (OR 0.58, 95%CI 0.34-0.99, p = 0.049), and recessive model (OR 0.59, 95%CI 0.35-0.99, p = 0.046); rs12594 in the RYR2 gene significantly decreased the risk of BC in the GG genotype (OR 0.44, 95%CI 0.22-0.88, p = 0.020), and recessive model (OR 0.43, 95%CI 0.21-0.85, p = 0.016). Further stratification analysis showed that NR5A2 rs2246209 was related to a lower incidence of BC affected by age, lymph nodes metastasis, and tumor stage; RYR2 rs12594 was related to a decreased BC risk restricted by age, estrogen receptor (ER), progesterone receptor (PR), menopausal status, tumor size, and tumor stage. Rs12594 in the RyR2 gene remained significant on the genetic susceptibility of PR-positive BC after Bonferroni correction (p < 0.0125). CONCLUSIONS: This study provides an evidence that NR5A2 rs2246209 and RYR2 rs12594 decreased the risk of breast cancer.


Asunto(s)
Pueblo Asiatico/genética , Neoplasias de la Mama/genética , Etnicidad/genética , Polimorfismo de Nucleótido Simple , Receptores Citoplasmáticos y Nucleares/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Adulto , Anciano , Neoplasias de la Mama/química , Neoplasias de la Mama/etnología , Neoplasias de la Mama/patología , Estudios de Casos y Controles , China/epidemiología , Resistencia a la Enfermedad/genética , Estrógenos , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Incidencia , Persona de Mediana Edad , Modelos Genéticos , Metástasis de la Neoplasia , Proteínas de Neoplasias/análisis , Estadificación de Neoplasias , Neoplasias Hormono-Dependientes/etnología , Neoplasias Hormono-Dependientes/genética , Oportunidad Relativa , Progesterona , Receptor ErbB-2/análisis , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Riesgo , Canal Liberador de Calcio Receptor de Rianodina/fisiología
18.
Strahlenther Onkol ; 196(11): 963-972, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32430662

RESUMEN

PURPOSE: For many decades, endometrial cancer (EC) has been considered as a homogenous tumor entity with good prognosis. The currently valid risk stratification considers clinical and pathological factors. Treatment recommendations differ considerably from country to country. MATERIALS AND METHODS: The Cancer Genome Atlas (TCGA) Research Network has shown that ECs should be reclassified into four novel molecular prognostic groups, with the potential of changing adjuvant management of EC patients: ultra-mutated, hyper-mutated, copy-number low, and copy-number high. Clinical examples are shown, and the available literature has been highlighted. The European Society of Gynaecological Oncology (ESGO) guideline for endometrial cancer takes the new classification system into consideration for adjuvant treatment decisions and will be published this year. RESULTS: In the near future, we expect new treatment recommendations that may differ considerably from the clinicopathologically driven recommendations on the basis of our deeper insight and better understanding of molecular markers in endometrial cancer. The PORTEC 4a study is the only recruiting study which randomizes patients to adjuvant or no adjuvant treatment on the basis of the aforementioned new classification system. CONCLUSION: The aim of the new classification is a more personalized adjuvant radio(chemo)therapy decision and better oncologic outcomes or avoidance of overtreatment.


Asunto(s)
Carcinoma/radioterapia , Neoplasias Endometriales/radioterapia , Radioterapia Adyuvante , Biomarcadores de Tumor , Carcinoma/clasificación , Carcinoma/genética , Carcinoma/patología , Carcinoma Endometrioide/clasificación , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patología , Carcinoma Endometrioide/radioterapia , Toma de Decisiones Clínicas , Neoplasias Endometriales/clasificación , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Estrógenos , Femenino , Predicción , Dosificación de Gen , Genes Relacionados con las Neoplasias , Humanos , Inestabilidad de Microsatélites , Mutación , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/radioterapia , Medicina de Precisión/métodos , Progesterona , Pronóstico , Riesgo , Transducción de Señal/genética
19.
Oncogene ; 39(22): 4436-4449, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32350443

RESUMEN

Activator protein (AP)-1 transcription factors are essential elements of the pro-oncogenic functions of transforming growth factor-ß (TGFß)-SMAD signaling. Here we show that in multiple HER2+ and/or EGFR+ breast cancer cell lines these AP-1-dependent tumorigenic properties of TGFß critically rely on epidermal growth factor receptor (EGFR) activation and expression of the ΔN isoform of transcriptional regulator p63. EGFR and ΔNp63 enabled and/or potentiated the activation of a subset of TGFß-inducible invasion/migration-associated genes, e.g., ITGA2, LAMB3, and WNT7A/B, and enhanced the recruitment of SMAD2/3 to these genes. The TGFß- and EGF-induced binding of SMAD2/3 and JUNB to these gene loci was accompanied by p63-SMAD2/3 and p63-JUNB complex formation. p63 and EGFR were also found to strongly potentiate TGFß induction of AP-1 proteins and, in particular, FOS family members. Ectopic overexpression of FOS could counteract the decrease in TGFß-induced gene activation after p63 depletion. p63 is also involved in the transcriptional regulation of heparin binding (HB)-EGF and EGFR genes, thereby establishing a self-amplification loop that facilitates and empowers the pro-invasive functions of TGFß. These cooperative pro-oncogenic functions of EGFR, AP-1, p63, and TGFß were efficiently inhibited by clinically relevant chemical inhibitors. Our findings may, therefore, be of importance for therapy of patients with breast cancers with an activated EGFR-RAS-RAF pathway.


Asunto(s)
Neoplasias de la Mama/patología , Factor de Crecimiento Epidérmico/fisiología , Regulación Neoplásica de la Expresión Génica , Invasividad Neoplásica/genética , Proteínas de Neoplasias/fisiología , Transducción de Señal , Factor de Transcripción AP-1/genética , Factores de Transcripción/genética , Transcripción Genética , Factor de Crecimiento Transformador beta1/fisiología , Proteínas Supresoras de Tumor/genética , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Receptores ErbB/fisiología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/genética , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Receptor ErbB-2/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta/fisiología , Proteínas Smad/fisiología
20.
BMC Cancer ; 20(1): 347, 2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32326897

RESUMEN

INTRODUCTION: The relationships among PIK3CA mutations, medication use and tumor progression remains poorly understood. Aspirin use post-diagnosis may modify components of the PI3K pathway, including AKT and mTOR, and has been associated with lower risk of breast cancer recurrence and mortality. We assessed time to metastasis (TTM) and survival with respect to aspirin use and tumor PIK3CA mutations among women with metastatic breast cancer. METHODS: Patients with hormone receptor positive, HER2 negative (HR+/HER2-) metastatic breast cancer treated in 2009-2016 who received tumor genotyping were included. Aspirin use between primary and metastatic diagnosis was extracted from electronic medical records. TTM and survival were estimated using Cox proportional hazards regression. RESULTS: Among 267 women with metastatic breast cancer, women with PIK3CA mutated tumors had longer TTM than women with PIK3CA wildtype tumors (7.1 vs. 4.7 years, p = 0.008). There was a significant interaction between PIK3CA mutations and aspirin use on TTM (p = 0.006) and survival (p = 0.026). PIK3CA mutations were associated with longer TTM among aspirin non-users (HR = 0.60 95% CI:0.44-0.82 p = 0.001) but not among aspirin users (HR = 1.57 0.86-2.84 p = 0.139). Similarly, PIK3CA mutations were associated with reduced mortality among aspirin non-users (HR = 0.70 95% CI:0.48-1.02 p = 0.066) but not among aspirin users (HR = 1.75 95% CI:0.88-3.49 p = 0.110). CONCLUSIONS: Among women who develop metastatic breast cancer, tumor PIK3CA mutations are associated with slower time to progression and mortality only among aspirin non-users. Larger studies are needed to confirm this finding and examine the relationship among aspirin use, tumor mutation profile, and the overall risk of breast cancer progression.


Asunto(s)
Aspirina/administración & dosificación , Biomarcadores de Tumor/genética , Neoplasias de la Mama/mortalidad , Fosfatidilinositol 3-Quinasa Clase I/genética , Mutación , Recurrencia Local de Neoplasia/mortalidad , Neoplasias Hormono-Dependientes/mortalidad , Adulto , Antiinflamatorios no Esteroideos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/secundario , Pronóstico , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Estudios Retrospectivos , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...